I Hf. 313 ‘3 Em C; (a LIBRARY Michigan State University 1 ' A This is to certify that the dissertation entitled CHARACTERIZATION OF NUCLEAR RECEPTORS OF THE ECDYSONE GENE REGULATORY HIERARCHY DURING MOSQUITO VITELLOGENESIS presented by Chao Li has been accepted towards fulfillment of the requirements for phD 638mm Genetics fl! /42/%é/ Major professor 6-24-2001 Date MSU it an Affirmative Action/Equal Opportunity lamination 0 - 12771 PLACE IN RETURN Box to remove this checkout from your record. To AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 8/01 cJClFiC/DateDuepss-sz CHARACTERIZATION OF NUCLEAR RECEPTORS OF THE ECDYSONE GENE REGULATORY HIERARCHY DURING MOSQUITO VITELLOGENESIS By Chao Li A DISSERTATION Submitted to Michigan State University In partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Genetics Program 2001 Tilt :I. Q . ICELIJIIM I & V tepmdudm that this 5 tanst‘ripnu knouicdgc meal is red hamn and precursors ( ABSTRACT CHARACTERIZATION OF NUCLEAR RECEPTORS OF THE ECDYSONE GENE REGULATORY HIERARCHY DURING MOSQUITO VITELLOGENESIS By Chao Li The insect steroid hormone, 20-hydroxyecdysone (20E), plays a critical role in regulating widely diverse physiological functions of development, metamorphosis and reproduction. The mode of 20E action is well understood in Drosophila development, where this steroid hormone regulates genes via a complex hierarchy containing several transcription factors belonging to the nuclear receptor superfamily. In contrast, our knowledge of 20E action in insect reproduction has been limited. In mosquitoes, a blood meal is required for egg development and as a consequence, they are vectors of numerous human and domestic animal diseases. The process of production of yolk protein precursors or vitellogenesis is central to egg development and is regulated by 20E. The goal of my thesis has been characterization of nuclear receptors of the 20E gene regulatory hierarchy involved in the mosquito vitellogenesis. The functional receptor for 2013, which is at the top of the hierarchy, is a heterodimer of two nuclear receptors, the ecdysone receptor (EcR) and Ultraspiracle (USP). There are two isoforms for both EcR and USP in mosquitoes, which are differentially expressed in a stage- and tissue-specific manner during vitellogenesis. In vitro fat body culture experiments suggest that the ligand, 20E itself, differentially regulates expression of AaEcR and AaUSP isoforms. In Drosophila, the action of the EcR/USP heterodimer is mediated by the product of an early gene E75, which is an orphan nuclear receptor. Analysis of the E75 gene has Show Ii xitc‘llogct‘ 53:35. \I i' guitars. .i ctgiohm‘. SCESIIIW It 10 directl} . pm me! {tugs competence expressed h are Iou. but bod} cultur Nth-31rd“ 3.1 ii‘Suils in St demonstrat: Ric-rem cr, 1 List 'fl v .Oprnc: AHR‘sihfi AHR‘S mi It] ECd'v 50178 F“ ' g Chao Li Shown that the ZOE-regulated hierarchy is used reiteratively during mosquito vitellogenesis. In the fat body, AaE 75 mRNA is not present during the previtellogenic stage, while AaE 75 is highly expressed during the vitellogenic stage. In in vitro fat body culture, AaE 75 is rapidly induced by 205 and this induction is not blocked by cycloheximide. Dose response experiments have revealed that AaE 75 is ten fold more sensitive to induction by 20E than the yolk protein gene, Vg. Furthermore, AaE75 is able to directly activate the Vg promoter in cell transfection assays, suggesting its role as an early gene. The mosquito becomes competent for a vitellogenic response to 20E after previtellogenic development. The nuclear orphan receptor AaFTZ-F 1 serves as a competence factor for the stage-specific response to 20E. AaFTZ-FI transcript is expressed highly in the fat body during pre- and post-vitellogenic periods when 20E titers are low, but it disappears in mid-vitellogenesis when 20E titers are high. In in vitro fat body culture, AaFTZ-FI expression is inhibited by 20B and super-activated by its withdrawal. Following in vitro AaFTZ-FI super-activation, a secondary 20E challenge results in super-induction of the early AaE 75 gene and the late target VCP gene, demonstrating involvement of AaF 72-F1 in developing additional 20E responsiveness. Moreover, the onset of ZOE-response competence in the mosquito fat body is correlated with the appearance of the functional AaFTZ-Fl protein at the end of previtellogenic development. Finally, I have identified another mosquito nuclear orphan receptor AHR78 that is expressed at a high level only in the ovary during the vitellogenic period. AHR78 inhibits the DNA binding and transactivation activities of AaEcR/AaUSP on the ecdysone response element of the Vg promoter. lslnt‘crc continua disciplira. acadcmit enhanced Dar id Ar: Third I \k; ACKNOWLEDGMENTS I sincerely appreciate my major professor, Dr. Alexander Raikhel, for his guidance and continued support throughout my graduate studies. His thoughtful, patient and disciplined mentoring provided me invaluable guidance, while allowing sufficient academic freedom and thus my interest and understanding of biomedical research were enhanced. Second, I would like to thank my guidance committee, Drs. Donald Jump, David Arnosti, and Ke Dong, for their help and suggestions during my five-year studies. Third, I wish to thank the friends I made in the Raikhel lab and the Genetics Program for their friendship and advice. Especially, I greatly appreciated Dr. Ken Miura for his detailed teaching of molecular biology techniques at the beginning of my studies, Dr. Sheng-fu Wang for his helpful discussions during my experiment difficulties, and Mr. Alan Hays for culturing mosquitoes and other technical helps. I also thank people who have been doing excellent collaborative work with me including Drs. Marianna Kapitskaya, S-F. Wang, Jinsong Zhu, K. Miura, Avraham Biran, and Kook-Ho Cho. In addition, I would like to thank Drs. William Pierceall and William Segraves at Yale University for cloning the AaE 75 gene and editing my manuscripts. I am also very grateful to Michel Trail for editing a manuscript, and S-F. Wang and Geoffrey Attardo for editing my dissertation. Finally, I would like to thank my parents and brothers for their love, support and encouragement from the beginning to the end of my graduate studies. list oil _ list of I. List of A Chapter 1 GAP-[CI 2. 7"." Hi »_ ‘riz‘r .1 TABLE OF CONTENTS List of Tables vii List of Figures viii List of Abbreviations xii Chapter 1. Literature Review 1 Molecular mechanism of nuclear receptor action 2 Nuclear receptors in the insect ecdysone gene regulatory hierarchy --------- 1 1 Nuclear receptors in mosquito vitellogenesis 19 Rationale for current studies 23 Chapter 2. Characterization of two Isoforms of the Mosquito Ecdysone Receptor (EcR) and Ultraspiracle (USP) 27 Abstract 28 Introduction 29 Materials and Methods 32 Results 38 Discussion 58 Chapter 3. Characterization of the Mosquito Early Gene E75 68 Abstract i 69 Introduction 70 Materials and Methods 73 Results 79 Discussion 99 106 Chapter 4. Cloning and Characterization of the Mosquito Early-late Gene HR3 ------- Chapter 5 Chaiter 6. Re" Abstract Introduction Materials and Methods Results Discussion Chapter 5. Cloning and Characterization of the Mosquito F 72-F1 Gene Abstract Introduction Materials and Methods Results Discussion Chapter 6. Cloning and Characterization of the Mosquito HR 78 Gene Abstract Introduction Materials and Methods Results Discussion Chapter 7. Summary and Future Research Perspectives References vi 107 108 112 116 128 133 134 135 139 145 163 168 169 170 174 178 191 195 204 Chapter Table 1.( Chapter I] ~- “ 1 1.10:; l. . Chapter: Table l. .\ LIST OF TABLES Chapter 1 Table 1. Conservation of nuclear receptors and cofactors between insects and vertebrates l8 Chapter 2 Table 1. The equilibrium dissociation constants (Kd) of different DNA sequences binding to AaEcR/USP-A and AaEcR/USP-B 57 Chapter 5 Table 1. Sequences of the PCR primers used in experiments 142 vii C haptcr 2 Fit; 1. Ev Fig. 2. D... ~ 0 (I .4 S7. -1 r; LIST OF FIGURES Chapter 1 Fig. 1. Functional domains of nuclear receptors Fig. 2. Activation and repression of a gene by nuclear receptors through coactivators or corepressors 10 Fig. 3. Structures of major insect hormones and their vertebrate analogs Fig. 4. Summary of events during the first cycle of vitellogenesis in the anautogenous mosquito, Aedes aegypti Chapter 2 Fig. 1. Expression profiles of USP and EcR mRNA during vitellogenesis Fig. 2. Dose response of USP-B, EcR and VCP to 20E in in vitro fat body 12 20 40 43 organ culture Fig. 3. Effect of 20-hydroxyecdysone and cycloheximide on the transcription of USP-A, USP-B, EcR and VCP in the previtellogenic fat bodies Fig. 4. Developmental profiles of USP proteins revealed by Western blot Fig. 5. EcR/USP binding to a direct repeat DR-4 Fig. 6. EcR/USP binding to DR-12 Fig. 7. Direct measurement of equilibrium dissociation constants (Kd) Fig. 8. EcR/USP-A transactivated a reporter gene with either IR-l or DR-4 EcRES in CV-l cells 47 50 50 50 53 55 Fig. 9. AaEcR/USP-B mediated more efficient transactivation than EcR/USP-A ------- 57 Fig. 10. Characteristics of the AaEcR-A isoform viii 66 Chapter 3 Fig. 1. Nucleotide and amino acid sequences of AaE75 81-83 Fig. 2. Genomic organization of AaE75 86 Fig. 3. Comparison of A edes aegypti and D. melanogaster E75 amino acid sequences 86 Fig. 4. AaE 75 transcripts in the mosquito ovary 88 Fig. 5. AaE75 transcripts in the mosquito fat body 88 Fig. 6. Accumulation of AaE75 mRNA in the mosquito fat body 90 Fig. 7. Expression of AaE75 RNA isoforms in the mosquito ovary during the first vitellogenic cycle 92 Fig. 8. Accumulation of AaE75 mRNA in the mosquito ovaries 92 Fig. 9. AaE75 response to 20-hydroxyecdysone in in vitro fat body organ culture ------ 94 F ig.10. Effect of cycloheximide on the activation of the AaE 75 gene 94 F ig.1 1. Time course of AaE 75 induction in in vitro fat body organ culture --------------- 95 F ig.12. Action of E75 and EcR/USP on the Vg Promoter 98 Chapter 4 Fig. 1. Alignment of the deduced amino acid sequence of the mosquito AHR3 cDNA with its insect homologues 118 Fig. 2. Domain comparison of the mosquito HR3 with selected members of the ROR/HR3 subfamily of nuclear receptors 120 Fig. 3. AHR3 transcripts in the mosquito A edes aegypti 122 ChapterS is l. 8141 Lilli! \itc .lJ (It (is -1ni a. .' .7 (":4 0‘ E.‘.~ ‘ . s: 1W in n Fifi” (’7 m (1. r in ll:- Fig. 4. Expression of AHR3 in the mosquito ovary during the first vitellogenic cycle 123 Fig. 5. Expression of AHR3 in the mosquito fat body during the first vitellogenic cycle 123 Fig. 6. AHR3 response to 20-hydoxyecdysone in in vitro fat body organ culture---- 125 Fig. 7. Time course of AHR3 induction by 20E in in vitro fat body organ culture---- 125 Fig. 8. Effect of cycloheximide on the AHR3 induction by 20E in in vitro fat body organ culture — 127 Chapter 5 Fig. 1. Summary of events during the first cycle of vitellogenesis in the anautogenous mosquito, Aedes aegypti 137 Fig. 2. Northern blot analysis of stage-specific induction of the vitellogenin and vitellogenic carboxypeptidase transcripts by the hormone 20E 146 Fig. 3. Alignment of the deduced amino acid sequence of the mosquito AaFTZ-FI cDNA with its insect homologs 149 Fig. 4. AaFTZ-FI expression profiles in the female mosquito fat body and pupae-- 152 Fig. 5. In vitro repression of AaF TZ-F I expression by the hormone 20E, and super- induction of AaF YZ-F 1 by withdrawal of 20E in fat body organ culture ---- 155 Fig. 6. Effects of cycloheximide on the 20E regulation of AaF TZ—F I in in vitro fat body organ culture 157 Fig. 7. Secondary responses to 20E of the ecdysone-inducible genes in in vitro fat body culture 159 Fig. 8. Identification of the functional AaFTZ-F 1 protein in fat body nuclear extracts of previtellogenic female mosquitoes 161 Chapter 6 Fig. 1. Alignment of the deduced amino acid sequence of the mosquito AHR78 cDNA with its insect homologs 179-180 Fig. 2. Domain comparison of the mosquito AHR78 with selected members of the HR78/TR2 subfamily nuclear receptors 18] Fig. 3. AHR 78 expression profiles in the female mosquito fat body and ovary ------ 183 Fig. 4. Induction of AHR78 mRNA by 20E in the ovary but not the fat body-------- 185 Fig. 5. Inhibition of EcR/USP binding to the natural EcREs by AHR78 ------------- 187 Fig. 6. AHR78 inhibition of ZOE-stimulated transactivation by AaEcR/AaUSP in 82 cells 189-190 xi ME. 21'- A31 .lt..‘ AR: ant: B-Cul. [ Bmzsfih. Bp;bu\c CAltd1 cDNAio lhnzlhvn DRidnect ECRICCd}; EMS-‘11 Cl; E‘REZCCQ Emiethg ER3 851 10 3 LIST OF ABBREVIATIONS 20E: 20-hydroxyecdysone Aa: Aedes aegypti AR: androgen receptor B-Gal: B—galactosidase Bm: Silkworm Bombyx mori Bp: base pair CAT: chloramphenicol aceyltransferase cDNA: complementary DNA Dm: Drosophila melanogaster DNA: Deoxyrbonucleic acid DR: direct repeat EcR: ecdysteroid receptor EMSA: electrophoretic mobility shift assay EcRE: ecdysteroid response element Eip: ecdysone inducible protein ER: estrogen receptor GR: glucocorticoid receptor GST: glutathione S—transferase HRE: hormone response element Hsp: heat Shock protein 27 IR: inverted repeat III: juvenile hormone xii Kb: kilobase pair Kd: equilibrium dissociation constant LBD: ligand binding domain Luc: luciferase MR: mineralocorticoid receptor mRNA: messenger RNA MurA: muristerone A NR: nuclear hormone receptor PAGE: polyacrylamide gel electrophoresis PR: progesterone receptor PBM: post blood meal PCR: polymerase chain reaction RAR: retinoid acid receptor RNA: ribonucleic acid RT: reverse transcription RXR: retinoid X receptor 82: Drosoplzila Schneider cell line 2 SDS: sodium dodecyl sulfate Tm: yellow mealworm Tenebrio moliter TR: thyroid hormone receptor USP: Ultraspiracle VCP: vitellogenic carboxypeptidase VCB: vitellogenic cathepsin-B xiii l'g: tit; VDR: \' \gR: it: IPP: )t Vg: vitellogenin VDR: vitamin D receptor VgR: vitellogenin receptor YPP: yolk protein precursor xiv Chapter 1 Literature Review piece»; receptor hindirtu: J . t._ mt'iCkUi’t these liga and mind titamm D MOLECULAR MECHANISM OF NUCLEAR RECEPTOR ACTION General structures and functions of nuclear receptors Steroid/Thyroid hormones play critical roles in a wide array of biological processes such as development, reproduction and metabolism. Classical nuclear hormone receptors are ligand-activated transcription factors that activate gene expression upon binding to their respective ligands. The ligands for nuclear receptors are small lipophilic molecules such as steroids, retinoids, vitamin D, and thyroid hormone. The receptors for these ligands are: the sex steroid receptors including estrogen (ER), progesterone (PR), and androgen (AR) receptors; the adrenal steroid receptors including glucocorticoid (GR) and mineralocorticoid (MR) receptors; and others including thyroid hormone (TR), vitamin D (VDR), retinoic acid (RAR) and 9-cis retinoic acid (RXR), and ecdysone (EcR) receptors. In addition, a large number of so-called nuclear orphan receptors have been identified. While these receptors have similar structures to nuclear hormone receptors, their ligands are unknown. The nuclear receptor superfamily is comprised of both nuclear hormone receptors and orphan receptors (Tsai and O’Malley, 1994; Mangelsdorf et al., 1995). One striking structural characteristic of the nuclear receptor superfamily is that most members contain five to six evolutionarily conserved functional domains, which can be divided into five regions starting from the N-terminal: A/B, C, D, E and F (Fig. 1). The A/B domain iS most variable among family members and contains the ligand- independent transactivation function (AF 1). The C domain is also named the DNA binding domain (DBD). The D domain acts as a hinge region between the C and E domains. It contains a nuclear localization Signal, as well as corepressor binding sites on .fl x20: 00 :BoE om? E 0:00 NA? 05. .qu 0% EEE» 5:05: 5:038: “5:53??me— 05 8 80:0: Nd? .:o:0:£ 5050383505 603302720? 0% 2 £80: E0820 m2 05 E TUE dougcommgb Em :ESEBELoEESmS acumNtBE: 5083080: 305 E88: 0—005 80: mo 226:3 3:256: mm: Amy E050: wEuEn Bum: 051 $8300: 320:: 088 E wEvEn 838380 :50 5:03:02 80—05: :8 60:an 80:03:00 068 0:5:00 ADV :Ew0: 0wE: 0f. .>__Eat0qsm 05 E 5:023:00 00:03:00 52%:— 05 mm: ADV E060: wEwEn <75 05. .EEEESQS 05 w:oEm 00:0:c0m DEER “Em 0E0 E ES, mEmEoc m “0:0 m2 d2 :0 REES“- Z 05 E3 0:080:00 05 E :32? .mEmEov 3:288: _80>0m («0 “0008800 0:0 EEEESQE 003000: 50—03: 0:: .«0 30980:: 05. 6.003000.— ..a0_0=: he 055:3. _a:e:0:=m .—.w_.m m L 1 _ :0=a>:0am:u..._. _ L W L 5:09.85 0803.00-00 a _ £50335 300050.700 _ _ w=_0=£-PUI $-193g I amour} meadda l . mtduni r \ tfixcplvr. be used as targets. The two most common half-Site arrangements are the symmetric repeat and the direct repeat with different inter-half-Site spacers. The steroid receptors for glucocorticoids, mineralocorticoids, androgens, and progesterone, bind to 5’- AGAACA-3’ half Sites; the estrogen receptor (ER), RXR, TR, VDR, RAR, PPAR, COUP-TF, and NGFI-B preferentially bind to consensus 5’-AGGTCA-3’ half sites (Klock et al., 1987; Umenoso et al., 1991). Three critical residues in the DBDS, called the P-box, predispose receptors to preferentially bind the GR versus the ER elements. Receptors recognizing the AGAACA half sites contain a GSXXV in their P-box, while those that recognize AGGTCA half-sites typically contain EGXXG residues at the corresponding positions. Thus, the P-box is critical for half-Site discrimination (Mader et al., 1989). The LBD of nuclear receptors is a large domain that sometimes encompasses almost the entire C terminus of these proteins. The LBD has been credited with having the additional functions of hormone-dependent dimerization and transactivation. Nuclear localization and heat Shock protein association functions also require the LED of some receptors. The binding of hormone results in receptor transformation and the initiation of a series of events by which receptors activate the expression of target genes (Kumar et al., 1987; Dobson et al., 1989; Allan et al., 1992). Interaction with a molecular chaperone-containing heterocomplex (MCH) is a pre-requisite for activation of steroid receptors. This interaction includes the molecular chaperones (Hsp90 and Hsc70), molecular chaperone interacting proteins (Hop, Hip, and p23), and peptidyl- prolyl isomerases (F KBP51, F KBP52, and Cyp40). In the case of PR and GR, the MCH is required for maturation to the aporeceptor state (DeFranco et al., I‘ll'O t, B' contort“ ehpICHZL .111 Egttngq basal trail. Ohl'n (“Ck 1998). Association of hsp90 is required for steroid binding to GR both in vitro and in vivo (Bresnick et al., 1989; Picard et al., 1990), and the binding of hsp90 to GR causes a conformational change in the GR LBD (Stancato et al., 1996). Roles of cofactors in gene activation and silencing by nuclear receptors Upon hormone binding, receptors bind to DNA and activate target gene expression. Early studies have demonstrated that PR, ER and GR activate transcription by enhancing the formation of stable preinitiation complexes at their target promoters by either direct interaction with components of the transcription machinery, or through an intermediate factor such as a coactivator (Klein-Hitpass et al., 1990; Tsai et al., 1990; Elliston et al., 1990). In the past few years, numerous nuclear receptor coactivators have been identified. They interact directly with the activation domain of a nuclear receptor in an agonist-dependent manner, and also interact with other factors and components of the basal transcription machinery but do not enhance the basal transcription activity on their own (Mckenna et al., 1999; Robyr et al., 2000; and Glass and Rosenfeld 2000). The first cloned nuclear receptor coactivator, steroid receptor coactivator-1 (SRC- l), represents one of the well-documented coactivators for nuclear receptors (Onate etal., 1995). Many other subsequently identified coactivators with Structures and functions Similar to SRC-1 comprise the SRC coactivator family. The SRC family can be divided into three subfamilies: SRC-l/NCoA-l, SRC-2/GRIPl/TIF2/NcoA-2, and SRC- 3/p/CIP/RAC3/ACTR/AIB-l/TRAM-1. SRC coactivators Share an ability to stimulate ligand-dependent transactivation by a rather large number of nuclear receptors in transient overexpression experiments. These coactivators also exhibit structure shnflar boxcs.n 1.81% t' ehccs" mcconu dantp"t this in.“I limit-err; till (0ng receptors. hamsteras addition, I 1X11 e! d!” MI, 3132‘)”. “é‘l—i‘il‘ix't Similarities. A distinct feature is the presence of multiple LXXLL Signature motifs or NR boxes, which comprise of determinants for direct interactions with the nuclear receptor LBDS (Mckenna et al., 1999). Several LBD crystal structures have established that upon ligand binding, the a helices in the LBD, including the AF2 core (helix 12), undergo a major reorientation in the context of the overall LBD structure. This reorientation forms part of a “charged clamp” that accommodates SRC coactivators within a hydrophobic cleft of the LBD. This interaction appears to occur through direct contacts with the LXXLL motif. Furthermore, SRC coactivators interact with cointegrators CBP/p300 and together they can coactivate synergistically, while C BP/p300 itself interacts directly with nuclear receptors. SRC and CBP/p300 coactivators both possess intrinsic histone acetyl transferase (HAT) activity and therefore can act in concert to remodel chromatin. In addition, p/CIP and P/CAF coactivators are recruited to nuclear receptors in response to hormone binding as part of a growing, HAT-containing, chromatin-remodeling complex (Xu et al., 1999; Freedman 1999). Moreover, nuclear receptors can repress transcription under various circumstances. Repression occurs mostly in the absence of a ligand or when an antagonist is bound to the receptor. It has been established that the unliganded TR is able to bind DNA acting as a repressor. In the past several years, three active repressors have been identified: SMRT (silencing mediator for retinoid and thyroid hormone receptor) (Chen and Evans, 1995), N-COR (nuclear receptor corepressor) (Kurokawa et al., 1995; Horlein et al., 1995) and SUN-COR (small ubiquitous nuclear corepressor) (Zamir et al 1997). These corepressors are able to interact with the unliganded TR and RAR Fig. 2. Activation and repression of a gene by nuclear receptors through coactivators or corepressors. The nuclear hormone receptor (NR) is associated with a corepressor (N-COR, SMRT), which in turn recruits a histone deacctylase (HDAC) through its interaction with Sin3. Deacctylation of histonc tails leads to transcriptional repression. Addition of the ligand disrupts this repression complex in favor of the association of a coactivator complex (SRC-l, P/CAF, p300/CBP, pC IP, and others). These proteins possess a histone acetyltransferase activity that allows chromatin decompaction through histone modifications. The interaction between the nuclear receptor AF -2 domain and the coactivation complex occurs through the LXXLL motif in many coactivators. The coactivator and corepressor complexes are represented with dashed lines since their exact composition in vivo is not determined. Modified from Robyr et al. (2000). Histone deacetylatlon \. LiganN Co-actlvators recruitment \) Co-re ressors re ease Hlstone acetylation domah uponlz, dentun~ rerunr ofSNIR HDAC- factor Til Kills 1. nuclear hl Remittr iRklb‘fl' t): associated with RXR on DNA. The C terminus of N-CoR interacts with the hinge domain and a portion of the LBD in TR and RAR. The silencing activity is abolished upon ligand binding by the release of corepressors from the receptor. Further studies have demonstrated that SMRT and N-CoR mediate transcription repression through recruitment of a histone deacetylase complex. The N terminus repression domain (SD-1) of SMRT interacts with Sin3A, which in turn associates with a histone deacetylase HDAC-l through one of its two Silencing domains. Thus, chromatin modification through deacetylation plays an active role in nuclear receptor mediated gene Silencing. In addition, TR silencing is mediated by its direct interaction with the general transcription factor TFIIB. This interaction is interrupted by thyroid hormone binding (Robyr et al., 2000). As a general model (Fig. 2), association of coactivators or corepressors with a nuclear hormone receptor is a ligand-dependent process. Coactivators and corepressors recruit HATS and HDACS, respectively, that finally result in gene activation or repression (Robyr et al., 2000). NUCLEAR RECEPTORS IN THE INSECT ECDYSONE GENE REGULATORY HIERARCHY In insects, two classes of lipophilic hormones control many events in metamorphosis, development and reproduction. One class is the steroid hormones known as ecdysteroids. The active form of ecdysteroid appears to be 20-hydroxyecdysone (20E). The other class is the scsquiterpenoids also known as juvenile hormones (Fig. 3) (Riddiford, 1985; Bownes, 1986; Dhadialla and Raikhel, 1994). During the last decade, . I DJ." :3: a" J ‘i"."..- i ll- . ' ”Pk U‘lgllnhi’l’l'i ' l 7‘ " 'I‘ki'i in I 'w ’I’nctiq ' if” :1 0 l1» 1 1“: h"... «an "" .1 ‘ I- -. ,. :.-. ,;. ‘1 .. -‘.. ..o 1".3‘ be. u.“- 1“. ‘3‘ .". .,E 0‘: 7‘ n.- to~ 58‘,» 16 ' :...l . t . l w- ir‘hdu‘" \‘1 t: ‘01 "‘ ”v ' . r . .L; . ‘i .3. Lil .rr ' "u r. -,.H I-f I “hug-trio ‘fi'lr..m""'“ 1:.1i..t..m.r.\-" "F...” nu ‘ I . '. _ u owl fl r“"._‘ it‘“ I I 20E Estradiol 0 COOH Retinoic acid Fig. 3. Structures of major insect hormones and their vertebrate analogs. 20-hydroxy-ecdysone (20B) and juvenile hormone 111 (JH III) are insect hormones; Estradiol and 9-cis retinoic acid (retinoic acid) are two vertebrate hormones. The arrow on 20E indicates the difference between ecdysone and its active form (ZOE). l .1 ,, moxie. iNRlll BUIII Ex I domains ECR 15011 .i. , . 31:931le UHdcrth; 111611111100: the molecular mechanism of 20E action has been studied in detail (Thummel, 1995; Raikhel et al., 1999; Kozlova and Thummel, 2000; Riddiford et al., 2001), while the molecular basis underlying juvenile hormone action remains obscure (Riddiford, 1996). The functional receptor for 20-hydroxyecdysone is the heterodimer of EcR (NRlHl) and Ultraspiracle (USP; NR 284) (Koelle et al., 1991; Yao et al., 1992, 1993). Both EcR and USP have multiple isoforms that differ only in their N-terminal A/B domains in many insects. In Drosophila, there exists one EcR gene, which encodes three EcR isoforms: EcR-A, ~Bl and-82. EcR-A and EcR-B result from utilization of alternative promoters while EcR-Bl and EcR-BZ are products of alternative Splicing under the same promoter (Koelle etal., 1991; Talbot et al., 1993). The EcR isoforms identified in other insects are named similarly to the Drosophila EcR isoforms. In Drosophila, only one isoform of USP encoded by a Single gene is known (Oro et al., 1990; Henrich er al., 1990; Shea et al., 1990). Yet two different molecular weight proteins are found, possible due to the use of an alternative translational start site (Henrich et al., 1994). By contrast, two USP isoforms have been found in three other insects including, A edes aegypti (Kapitskaya et al., 1996), Manduca (Jindra et al., 1997), and Chironomus tentans (Vogtli et al., 1999). It is unclear thus far whether these isoforms result from alternative splicing of a single gene, or from different genes. The A/B domains of Aedes USP-A, Chironomus USP-1, and Manduca USP-1 exhibit the highest Similarity, particularly at the N-terminus, to that of the single USPS of Drosophila, the Silkworm Bombyx mori (Tzertzinis et al., 1994), and Spruce budworm Choristoneurafumiferana (Perera et al., 1999). respon ,~ ~ x x l l . .. . . . MW. NH 1. I. ll . u a W a e 1... n1 run iu fit. .1 . r n n .4 Pl. r d H1 \ L \0 P a .\. i .L 10 flu l. J W .U 1 . . r . . C a: . 1h. a C . h Oil Ill hit 351 tV c n I 9‘ 45 1 it It. \\ iL «N “u Fl «\_ U n n “I ‘1 0 that H; N. MN on U; D. . a fit it... - r I. - .C t .\ .0 F A. «Au ydrU Q Alfis r I “L; iu AL ..l._ A\ . I .4 i“ . . .. 1W1. Di , 1 It. Up n... 3 R S 0: run 0.: ml. .\ h. «A. .Q Both EcR and USP are required for 20E binding and binding to ecdysone responsive elements (EcRE). The first EcRE was identified in the promoter of the heat shock protein-27 gene in Drosophila (Riddihough and Pelman; 1987). It is an imperfect palindrome with only a 1-bp spacer (hsp27). Several other EcREs have also been identified in the regulatory regions of four Drosoplu'la genes: Eip28/29 (Cherbas et al. 1991), Fbp-l (Antoniewski er al., 1994), Sgs-4 (Lehmann and Korge, 1995), and Lsp-Z (Antoniewski et al., 1995), each of which contains an imperfect inverted repeat with a 1- bp spacer (IR-1). These findings suggest that natural Drosophila EcREs are predominantly IR-ls. Nonetheless, artificial direct repeats (DR) with different spacers appear to be functional as well (Homer et al., 1995). In addition, a direct repeat EcRE with a 12-nucleotide spacer exists in the Drosophila 3C/ng (D’Avino etal., 1995). In invertebrates, perhaps the most surprising discovery in the nuclear receptor superfamily is the large number of genes that are regulated by ecdysone. Most of these genes are nuclear orphan receptors, which are either induced or repressed by this hormone at the level of transcription. In Drosophila, the discovery of most of these genes arose from efforts to understand how ecdysone directs early stages of metamorphosis. Early studies from polytene chromosome puffing patterns in larval salivary glands have revealed that ecdysone signaling is transduced and amplified via a two-step regulatory hierarchy (Ashbumer et al., 1974), which is widely cited as the Ashbumer model. At the ends of larval and prepupal development, pulses of ecdysone directly induce a small number of “early genes”, at least three of which encode transcription factors: E74 (Burtis et al., 1990; Thummel et al., 1990), and two nuclear receptors: E75 (Segraves and Hogness, 1990) and BR-C (DiBello etal., 1991). These mdyg S€CO ‘. HOE'IIC‘» d‘élll‘iCl . first Zin, “Nhthc pl Unity“ 1:35 a ‘i93e\ early genes both repress their own expression and induce more than 100 “late genes” as secondarily responsive genes. Interestingly, the early genes are expressed widely, while many late genes are expressed in a tissue-specific manner such as the Sgs-4 glue and L7] genes (Thummel, 1996). E75 is a complex ecdysone—inducible early gene that encodes three isoforms, E75A, E758 and E75C as a result of alternative splicing (F eigl et al., 1989; Segraves and Hogness, 1990). All three E75 isoforms are orphan receptors. E75A and E75C have distinct A/B domains joined to the identical DBD and LBD. In contrast, E75C lacks the first zinc finger of the DBD, suggesting that it is incapable of binding DNA. Consistent with the Ashbumer model, several EcR/USP binding sites have been found in the E75 promoter (Talbot, 1993), and E75A protein is found to be bound to both early and late gene loci (Hill et al., 1993). Moreover, recent studies have revealed that E75 is also required for development of egg chambers. This function was determined in E75 germline mutants in which nurse cells degenerate (Buszczak et al., 1999). There are some nuclear orphan receptors showing delayed early responses to ecdysone. HR3 is one of those, and therefore is called an “early-late” gene in the ecdysone gene regulatory hierarchy (Koelle et al., 1992). Drosophila HR3 (DHR3) comprises a complex locus, encoding at least three different-sized classes of mRNAs. The Drosophila “early-late” genes are induced by ecdysone, but differ from the early genes in that they require ecdysone-induced protein for maximal levels of gene expression (Stone and Thummel, 1993; Homer et al., 1995). Functional studies have revealed that DHR3 represses the ecdysone induction of early genes and induces BFTZ- 15 subseu. tempur- l994l. . in mid,” in in 11;. cud} 50ft; hierarcha Specific I‘ 319903;") lL ecdysone ln appear lo '- ! ‘CCSPIOB (Zcrth {If The {Wm :15th DH ECR ESP widens I 1995a: H FU. l J U? . Fl, an orphan nuclear receptor that is essential for the appropriate response to the subsequent prepupal pulse of ecdysone (White et al., 1997). The orphan receptor Drosop/zila BFTZ-F 1 has been identified as providing a temporal link between late larval and prepupal ecdysone responses (Woodard et al., 1994). Drosophila BFTZ-Fl is expressed during the brief interval of low ecdysone titer in midprepupae. Consistent with its expression profile, 20E represses BFTZ-F 1 transcript in in vitro cultured salivary glands. Ectopic expression of BFTZ-Fl in flies enhances ecdysone induction of the early genes; in BFTZ mutants the ecdysone-triggered genetic hierarchy is severely attenuated. Thus, BFTZ-F 1 serves as a competence factor for stage- specific transcription responses to ecdysone, and its expression in the midprepupae appears to reset the system to allow reinduction of the early and late genetic responses to ecdysone (Woodard et al., 1994; Broadus et al., I999). In addition, it has been established that some insect nuclear orphan receptors appear to be involved in negative regulation of ecdysone responses. Three orphan receptors with this function have been identified, DHR38 (Sutherland et al., 1995), Svp (Zelhof et al., 1995a), DHR78 (Fisk and Thummel, 1995, 1998; Zelhof et al., 1995b). The formation of inactive heterodimers between DHR38 and USP, SVP and EcR, as well as the DHR78 homodimer as a competitor, inhibit DNA binding and activation of EcR/USP on the ecdysone responsive elements, providing different levels at which ecdysone responses can be negatively controlled (Sutherland et al., 1995; Zelhof et al., 1995a; Fisk and Thummel, 1995, 1998; Zelhofet al., 1995b). Furthermore, recent studies on the molecular mechanism of EcR/USP action have demonstrated that cofactors are involved in activation or repression of ecdysone gene regulation. Two Drosophila co-repressors have been characterized to interact with unliganded EcR/U SP: Alien (Dressel et'al., 1999) and SMRTER (silencing mediator for RXR and TR-related ecdysteroid receptor interacting factor) (Tsai et al., 1999). These proteins can interact directly with EcR, but not USP, in the absence of ligand. SMRTER has a similar function but different structure relative to the vertebrate corepressors SMRT and N-CoR. Antibody staining of salivary gland polytene chromosomes has displayed that SMRTER co-localizes with EcR/USP, suggesting these proteins function together to repress target gene expression. Like vertebrate SMRT and N-CoR, Drosophila SMRTER mediates repression by interaction with Sin3A, a repressor to form a complex with HDAC (Tsai et al., 1999). Drosophila Alien is another co-repressor for EcR/USP, belonging to a novel class of co-repressors that are unrelated to SMRT and N-CoR. Alien proteins are highly conserved in evolution, showing 90% identity between human and fly. Vertebrate Alien interacts with TR only in the absence of hormone; Drosophila Alien shows similar activity, mediating repression through the Sin3A pathway (Dressel et aL,1999) It was believed that there was no SRC family coactivator in Drosophila and yeast (Freedman, 1999); however, a recent discovery has demonstrated that Drosophila TAI is an EcR/USP coactivator, the homolog of A181 (SRC3) (Bai et al., 2000). Tai protein has the conserved sequence and overall domain structure of a SRC family member, containing bHLH, PAS and LXXLL motifs. TAI also shows a functional similarity with SRC proteins. TAI directly interacts with EcR in a ligand dependent manner, and co- localizes with EcR/USP in egg chambers and salivary gland polytene chromosomes. Furthermore, TAI augments transcriptional activation by EcR/USP in cultured cells (Bai tumult int enci‘ Simildfi. and th‘l'. tertehr; \ lnsect _\ et al., 2000). These findings provide the first evidence that nuclear receptor coactivators exist in both vertebrates and invertebrate flies. Based on findings to date, most insect nuclear receptors have vertebrate homologs, exhibiting high levels of evolutionary conservation between vertebrates and invertebrates in the nuclear receptor superfamily. These conservations include similarities of protein structures and general molecular mechanisms of nuclear receptors and their cofactors (Table 1). Table 1. Conservation of nuclear receptors and cofactors between insects and vertebrates Insect Nuclear Receptor Vertebrate Homolog % DBD/LBD Identity EcR BAR (FXR) 81/34 USP RXR 86/49 E75 Rev-Erb 79/35 HR3 RORa 75/20 FTZ-Fl SFl 88/33 HR78 TR2 74/26 HR38 NGFI-B 88/54 SVP COUP-TF 94/93 HNF4 HNF 4 90/67 Insect Co-activator Vertebrate Homolog Domain Similarity TAI AIBl bHLH, PAS, LXXLL Insect Co—repressor Vertebrate Homolog % Sequence identity SMRTER SMRT/N-CoR High in SNOR, SANT, ITS, LSD, and GSI motifs Alien Alien 90 18 Faster» anauttg female : resuitzn: llPP ,1 ;; Raikhel. WCIOL \ km?- PR Control H“;~"C‘L1Li Immed‘. “10$qu NUCLEAR RECEPTORS IN MOSQUITO VITELLOGENESIS Mosquitoes serve as vectors for many devastating human diseases (Collins and Paskewitz, 1995; Butler et al., 1997; Beier, 1998) due to their ability to transmit pathogens during blood feeding, which is required for their egg development. In the anautogenous mosquitoes, vitellogenesis and egg maturation are initiated only after a female mosquito ingests vertebrate blood. A blood meal triggers a hormonal cascade resulting in production of 20E, the terminal signal, which activates yolk protein precursor (YPP) genes in the fat body (Hagedom, 1983; 1985; Raikhel, 1992; Dhadialla and Raikhel, 1994; Deitsch etal., 1995a; 1995b). In the anautogenous mosquito Aedes aegypti, which is being studied as a model vector, vitellogenesis proceeds through two periods (Fig. 4). The approximately 3-day- long previtellogenic development of both the ovary and fat body proceeds under the control of juvenile hormone (111) Ill (Gwartz and Spielman, 1973; F lannagan and Hagedom, 1977). JH titers are high during the previtellogenic period and drop immediately following the onset of vitellogenesis, which is initiated when the female mosquito takes a blood meal (Shapiro et al., 1986). The fat body then produces YPP, which are internalized by the developing oocytes (Raikhel, 1992). In addition to vitellogenin (Vg), the major YPP, the mosquito fat body synthesizes three other YPPs, vitellogenic carboxypeptidase (VCP), vitellogenic cathepsin B (VCB) (Cho et al., 1991; 1999), and lipophorin (Sun et al., 2000). YPP synthesis reaches its maximal levels at 24 hr post-blood meal (PBM) and then proceeds until 30-32 hr PBM, when it is rapidly terminated (Raikhel, 1992). 20! "I ate 0e ‘: \l'l‘ Fig. 4. S mosquit female. 519358un ICCUT.\'(\1| Sl'nthesi/ lB\1l, 15 351d ccd} Hagt‘dun 1110,qu ‘s‘itcllogc; 1991). .\ E 1 A 88M :1 s 1,2 1L6 17,0214 2832 31643 433514 6109673 Hormones 10*: A W m mnflfllflMMmL W Late Genes Vs VC P E 1 «3 33M ‘1 {5 12 1.6202512832364248 5JL6LQ'6 72 Days Hours Fig. 4. Summary of events during the first cycle of vitellogenesis in the anautogenous mosquito, Aedes aegypti. The previtellogenic period begins at eclosion (E) of the adult female. During first ~2-3 days of post-eclosion life, the fat body becomes competent for subsequent vitellogenesis. The female then enters a state-of-arrest; yolk protein precursors, vitellogenin (Vg) and vitellogenic carboxypeptidase (VCP) are not synthesized during this previtellogenic period. Only when the female takes a blood meal (BM), is vitellogenesis initiated. Hormones: hormonal titers of juvenile hormone (JH) and ecdysteroids (20E) in A. aegypti females (Hagedom et al., 1975; Shapiro and Hagedom, 1982). Transcript profiles of yolk protein precursors (Late Genes) in the mosquito fat body were determined by Northern blot and RT-PCR/Southem blot. Vg, vitellogenin (Cho and Raikhel, 1992); VCP, vitellogenic carboxypeptidase (Cho et al., 1991). Modified from Li et al., 2000. 20 rate of C1331; 16\ 615 .; role of. shown 1 WP 11) C}'C10ilc Ecdysteroid titers in the female mosquito hemolymph positively correlate with the rate of YPP synthesis in the fat body (Fig. 4). While the 20E titers are only slightly elevated at 4 hr PBM, they begin to rise sharply at 6-8 hr PBM, and reach their maximum levels at 18-24 hr PBM (Hagedom et al., 1975). Consistent with the initially proposed role of 20E in activating vitellogenesis in anautogenous mosquitoes (Hagedom etal., 1973; Hagedom and Fallon, 1973), experiments using an in vitro fat body culture have shown that physiological doses of 20E (10'7 - 10'6 M) activate two YPP genes, Vg and VCP (Deitsch el al., 1995a; 1995b). Utilization of the protein synthesis inhibitor, cycloheximide (Chx), has demonstrated that the activation of YPP genes in the mosquito fat body requires protein synthesis (Deitsch et al., 1995a). Thus, it is likely that a regulatory cascade similar to that seen in Drosophila development mediates the action of 20E in the mosquito fat body. The functional ecdysteroid receptor is a heterodimer of the ecdysone receptor (EcR) and a retinoid X receptor (RXR) homolog, Ultraspiracle (USP) (Yao et al., 1992; 1993). In the mosquito A edes aegypti, one isoform of ecdysone receptor (AaEcR) and two USP isoforms, AaUSP-A and AaUSP-B have been cloned (Cho et al., 1995; Kapitskaya et al., 1996). The mosquito EcR/USP heterodimer has been shown to bind to various EcREs to modulate ecdysone regulation of target genes (Wang et al., 1998). Two classes of EcREs, inverted repeats (IR) and directed repeats (DR), with different spacers are able to be bound by AaEcR/AaUSP. The IR-l (IR with a one nucleotide spacer) and DR-4 (DR with a four nucleotide spacer) display optimal binding affinities in each class of EcRE. Changing the half sites of the Drosop/zi/a natural hsp27 EcRE from imperfect repeats to perfect repeats AGGTCA increases the DNA binding affinity of to IRS. I pmmot. may 6. mm; :1; : located . the \‘g 1i . l 111 1058 0' AaEcR/AaUSP 10 fold. The spacer-length requirement for DRs is less stringent relative to IRs. These findings suggest that although EcREs identified in some Drosophilrz gene promoters are predominantly le, some other native EcREs with different configurations may exist in other insects (Wang et al., 1998). Indeed, most recent studies on the mosquito Vg gene promoter have revealed that an imperfect DR-l EcRE (Vg EcRE) is located at the 348 bp upstream of the Vg promoter, and AaEcR/AaUSP is able to bind to the Vg EcRE. Moreover, deletion of the Vg EcRE from the Vg promoter-reporter results in loss of activation by AaEcR/AaUSP in cultured cells (Martin et al., 2001). AaUSP-A and AaUSP-B are the first two USP isoforms identified in insects (Kapitskaya et al., 1996). AaUSP-A and AaUSP-B are identical proteins except at the N- terminal regions of the A/B domains: the first 31 aa in AaUSP—A different from the first 6 aa in AaUSP-B. Both mosquito USP isoforms can form heterodimers with AaEcR and bind to EcREs. In the fat body, AaUSP-A is expressed highly at the previtellogenic stage and post-vitellogenic stage while AaUSP-B is expressed at relative high levels in the vitellogenic stage (Kapitskaya et al., 1996). These data suggest that the two isoforms may play different roles during mosquito vitellogenesis. Previtellogenic, preparatory development is required for mosquito fat bodies to attain both the responsiveness to 20E and competence for massive yolk protein synthesis and secretion. Following this stage, mosquitoes enter a previtellogenic developmental arrest, namely the state-of—arrest, which prevents the activation of YPP genes in the previtellogenic female fat body prior to blood feeding (Raikhel and Lea, 1990). Recent studies have demonstrated that AHR3 8, a mosquito homolog of DHR38 and vertebrate NGFI-B, plays a crucial role in the state of arrest (Zhu et al., 2000). AHR38 is able to 22 “10] I disruptE Ech_I | SW It" heterot: EcR L r Ech5> EXISA~ P081V11c 10 ecd} 5( I Ii ("D a t ._ 1.. Lutdl‘ f disrupt binding of EcR/USP to EcREs and inhibit transactivation of a reporter gene by EcR/U SP in cultured cells. GST pulldown and co-immunoprecipitation experiments have revealed that AaUSP, the obligatory heterodimer partner for AaEcR, forms a heterodimer with AHR38 at the state-of-arrest, preventing the formation of a functional EcR/U SP complex (Zhu et al., 2000). Consistent with these findings, binding of the EcR/USP complex to EcREs in the fat body nuclear extracts is barely detectable by EMSAs at the state-of-arrest; in contrast, a large amount of binding is visible in the 1hr PBM nuclear extracts (Miura et al., 1999). Another nuclear orphan receptor AaSVP, a homolog of Drosophila SVP and vertebrate COUP-TF, has been cloned recently in our lab. AaSVP is highly conserved with its homologs in term of amino acid sequence. AaSVP interacts with AaUSP in the post vitellogenic period, suggesting its involvement in terminating vitellogenic responses to ecdysone in mosquito fat bodies (Miura, Zhu, and Raikhel, unpublished data). Finally, it is known that the mosquito Vg gene is activated in a tissue specific manner. The nuclear orphan receptor HNF-4, a member of the Hepatocyte Nuclear Factor 4 (HNF-4) family, is possibly implicated in controlling tissue specificity. Three HN F -4 isoforms are expressed in mosquito fat bodies with different developmental profiles during vitellogenesis, suggesting distinct functions for each isoform at different stages of vitellogenesis (Kapitskaya et al., 1998). RATIONALE FOR CURRENT STUDIES One of the main research interests in Dr. Raikhel’s lab is to understand the molecular mechanism underlying mosquito vitellogenesis. In his lab, several yolk 23 sex-5.: which prox id . C001T3> dCI'EiOp EESCCIIK protein precursor genes have been cloned, which are expressed in a stage-, tissue- and sex-specific manner. These genes are activated in female fat bodies by the hormone 20E, which is triggered by a blood meal. This system of stringently controlled gene regulation provides an ideal model to study ecdysone gene regulation during insect reproduction. In contrast to detailed studies on the ecdysone regulatory cascade in Drosophila development and metamorphosis, however, the ecdysone gene regulatory hierarchy in insect reproduction was poorly understood. In mosquitoes, one isoform of EcR and two isoforms of USP have been cloned in our lab, but some primary questions still need to be addressed. Three EcR isoforms have been identified in Drosophila; similarly, two or three isoforms have been found in many other insects. It has been demonstrated that the different EcR isoforms are expressed in different tissues and mediate functionally different ecdysone responses. Therefore, in mosquitoes, one of the key questions was whether any other EcR isoforms exist, and if they do, whether they are involved in vitellogenesis. Secondly, two USP isoforms have been identified in mosquitoes and they are expressed differentially during vitellogenesis. AaUSP-A mRNAs in fat bodies are at high levels during previtellogenic and postvitellogenic periods, but at low levels during the vitellogenic period. In contrast, AaUSP-B is expressed at high levels during the vitellogenic stage. Correlated with AaUSP mRN A levels, the two hormones, JHIII and 20E, display opposite titer dynamics during pre- and vitellogenic stages. JHIII titers are high during the previtellogenic stage and decline immediately following the onset of vitellogenesis. 20E remains at low levels during the previtellogenic stage and rises sharply after a blood meal. A recent study suggests that USP may be a candidate receptor for JH (Jones and Sharp, 1997). 24 Therefore, other more critical questions addressed in the first part of my research were how the mosquito USP gene(s) are regulated during vitellogenesis and whether AaUSP-A and AaUSP-B isoform expressions are under the control of JHIII or 20E. The ecdysone gene regulatory hierarchy has been well documented in Drosophila metamorphosis. This regulatory cascade contains many transcription factors, most of which are nuclear orphan receptors. In mosquitoes, utilization of the protein synthesis inhibitor, cycloheximide (Chx), has demonstrated that the activation of YPP genes in the mosquito fat body requires protein synthesis (Deitsch et al., 19953). Thus, YPP genes serve as the ecdysone regulated late target genes. It is likely that a regulatory cascade similar to that seen in Drosophila development mediates the action of 20E in the mosquito fat body. Among the components of the ecdysone gene regulatory hierarchy in Drosophila, the early gene E75 and the early-late gene HR3 are two key ones. Therefore, in the second and third parts of my studies, mosquito homologs of E75 and HR3 were cloned and characterized as a beginning to elucidating the ecdysone regulation cascade in mosquito vitellogenesis. A preparatory developmental previtellogenic period is required for the mosquito fat body to attain competence for ecdysone responsiveness (Sappington and Raikhel, 1999). F lannagan and Hagedom (1977) have demonstrated that the acquisition of competence for the ecdysone response in the mosquito fat body is under the control of juvenile hormone 111. However, the molecular mechanism underlying the change in sensitivity to ecdysone in the mosquito fat body remains obscure. In the fourth part of my research, I addressed the question of whether competence for the ecdysone response is controlled at the transcriptional level or translational level. Furthermore, the mosquito 25 ‘3 1“ 1" '. mosq- COHIIL' 111013.111. DIE/Sop homolog of BFTZ-Fl, a competence factor in the Drosophila ecdysone regulatory network, was cloned. I then designed experiments to address the question of what role mosquito FTZ-Fl plays during vitellogenesis, and whether the molecular mechanisms controlling the stage-specific ecdysone responses are conserved during Drosophila metamorphosis and mosquito vitellogenesis. Finally, in the fifth part of my research, I cloned the mosquito homolog of Drosophila HR 78, which is involved in negative regulation of ecdysone responses. I designed experiments and answered the questions of whether mosquito HR78 represses the DNA binding and transactivation functions of AaEcR/AaUSP, and what role it plays during vitellogenesis. 26 I . . g ‘4‘ 6 o a. '_3” ’T -vl ‘. .3 .u . .1 1 -.., 7.! '.‘C 1.. _':_t ., ,‘4. ." -1. - u‘r . v t .. .1 O «- oft t. 11031 r951 Chapter 2* Characterization of Two Isoforms of the Mosquito Ecdysone Receptor (EcR) and Ultraspiracle (USP) *Most results have originally been published in Dev.Biol. Ref: Wang, s.-E.,Lj,_g,”,2hu,1.,Miura, K., Miksicek, R. 1., Raikhel, A. s. (2000). Dev. Biol. 218, 99-113. # Underlined two authors contributed equally to this work. 27 flprcss' EcR L's them 11 1131154“ ”1938 r, ABSTRACT Ultraspiracle (USP), the insect homologue of the vertebrate retinoid X receptor (RXR) is an obligatory dimerization partner for the ecdysteroid receptor (EcR). Two USP isoforms, USP-A and USP—B, with distinct N-terrnini occur in the mosquito, Aedes aegypti. In the fat body and ovary, USP-A mRNA is highly expressed during the pre— and late-vitellogenic stages, corresponding to a period of low ecdysteroid titer, while USP-B mRNA exhibits its highest levels during the vitellogenic period, correlating with a high ecdysteroid titer. Remarkably, 20-hydroxyecdysone (20E) has opposite effects on USP isoform transcripts in in vitro fat body culture. This steroid hormone up-regulates USP-B transcription and its presence is required to sustain a high level of USP-B expression. In contrast, 20E inhibits activation of USP-A transcription. Although EcR/USP-A recognizes the same ecdysteroid responsive elements, EcR/USP-B binds them with an affinity two—fold higher than that of EcR/USP-A. Likewise, EcR/USP-B transactivates a reporter gene in CV-l cells two-fold more strongly than EcR/USP-A. These results suggest that USP-B functions as a major heterodimerization partner for EcR during the vitellogenic response to 20E in the mosquito. 28 was funk... domai" «r r) tOlilLr" WHEEL. members 1 INTRODUCTION In insects, as in higher metazoans, steroid-regulated gene expression is mediated by a family of intracellular receptors, which belong to the superfamily of nuclear receptors. These receptors have a highly conserved structure, including a DNA—binding domain (DBD) and a multifunctional ligand-binding domain (LBD). In addition to conferring ligand binding specificity, the LBD is responsible for dimerization, transactivation, co-activator/co—repressor interactions, and in the case of vertebrate steroid receptors, heat shock protein binding (O’Malley and Conneely, 1992; Mangelsdorf et al., 1995; Onate et al., 1995). Nuclear receptors are often encoded by several genes and each gene may produce several subtypes called isoforms due to utilization of alternative promoters, splicing or polyadenylation signals (Keightley, 1998). The major differences between isoforms appear to be centered in the N-terminal A/B domain. This least conserved region of nuclear receptors contains the activation function AF-l, which has been demonstrated to provide cell- and promoter-specific transactivation properties (Kastner et al., 1990; Kuiper et al., 1996; Paech et al., 1997). The major insect steroid hormones, ecdysteroids, are involved in regulating development, molting, metamorphosis and reproduction (Riddiford, 1993b; Raikhel, 1992). DNA binding, ligand binding and transfection assays indicate that the functional ecdysteroid receptor complex consists of two subunits, the ecdysteroid receptor itself (EcR) and the retinoid X receptor homologue Ultraspiracle (USP), both of which are members of the nuclear receptor superfamily (Koelle et al., 1991; Yao et al., 1992; 1993; Thomas et al., 1993). The role of USP as a partner in the functional ecdysteroid receptor was further substantiated by mutational analysis. Fly usp mutants show multiple defects 29 EUR 15 meni: 199'L; Um I); dmnmr spficzng {Esme-a ”c301“ 31.199. dfgcncr DWI/1'), 1‘ in ecdysteroid-regulated gene expression at the larval-prepupal transition, indicating that USP is required for ecdysteroid receptor activity in vivo (Hall and Thummel, 1998). The pleiotropic effects of ecdysteroids are reflected by the existence of multiple EcR isoforms. EcR isoforms were first identified in Drosophila (Talbot et al., 1993) and then in other insects including Bombyx mori (Swever et al., 1995; Kamimura et al., 1996; 1997) and Manduca sexta (Fujiwara et al., 1995; Jindra et al., 1996a). The three Drosophila EcR isoforms (EcR-A, EcR-Bl and EcR-B2) differ in the N-terminal A/B domain, as a result of utilization of different transcription start sites and alternative splicing. Distinct functions of DmEcR isoforms were first evidenced by their different tissue- and stage-specific expression profiles (Talbot et al., 1993). In the Drosophila nervous system, EcR-A and EcR-Bl isoforms have clearly distinct functions (Truman et al., 1994). A high level of EcR-A expression in the ventral CNS correlates with rapid degeneration of neurons after adult emergence (Robinow et al., 1993). Moreover, Drosophila EcR mutations confirm that functional differences exist among receptor isoforms (Bender et al., 1997). In particular, the EcR-Bl mutants can not survive through metamorphosis, and EcR-Bl and EcR-B2 mutants fail to prune back larval- specific dendrites to initiate larval neuron remodeling (Schubiger et al., 1998). Unlike Drosophila USP for which a single form of mRNA has been identified (Shea et al., 1990; Henrich et al., 1990, Oro et al., 1990), two USP isoforms are reported in the mosquito Aedes aegypti (USP-A and USP-B, Kapitskaya et al., 1996) and in the moth, M. sexta (USP-l and USP-2, Jindra et al., 1997). In both insects, USP isoforms exhibit distinct N-terminal A/B domains similar to isoform variation in the EcR isoforms. In A. aegypti, the N-terminal 31 aa in USP—A are different from the first 6 aa in USP-B. 30 50kt? 11101111 ' Bonn? 111 Dit- Houcx [SPtsr 1116! \1}; RRLS P1017101: 31.1991 distinCt \il'c‘llgg‘ €\pressi Comm“: Moreover, their 5’ untranslated regions (UTRs) are different, suggesting that these isoforms are most likely derived from the same gene via utilization of alternative promoters (Kapitskaya, et al., 1996). In sharp contrast to the large body of information defining the roles of EcR isoforms, little is known about the properties of USP isoforms. The lack of USP isoforms in Drosophila makes it difficult to perform USP isoform-specific mutant analysis. However, identification of two forms of USP from M. sexta has permitted the study of USP isoform profiles during development (Jindra, 1997; Asahina et al., 1997; Hiruma et al., 1999; Lan et al., 1999). The level of USP-2 mRNA correlates with the ecdysteroid titer whereas USP-1 transcription is complementary to that of USP-2. However, it is the EcR/USP-l complex, not the complex containing EcR/USP-2, that activates the MHR-3 promoter although MHR-3 mRNA appears following a rise of ecdysteroid levels (Lan et al., 1999). Likewise, the two Aedes USP isoforms enabled us to investigate the roles of distinct USP isoforms in the adult insect during reproduction. Indeed, mosquito vitellogenesis has provided a perfect model to study ecdysteroid-regulated gene expression during reproduction, as the endocrine release of ecdysteroids is tightly controlled by a blood meal (Fig. l-A). The DNA binding properties of the Aedes EcR/USP-B complex have been studied in detail: it binds to various ecdysteroid responsive elements (EcRE) with the AGGTCA consensus half site arranged either as direct repeats or inverted repeats. A l-bp spacer is optimal for inverted repeats, while a 4- bp spacer is optimal for direct repeats. DNA binding activity is correlated with transactivation as demonstrated by transfection assays in CV-l cells (Wang et al., 1998). Only the heterodimeric complex containing the mosquito EcR and one of the mosquito 31 Cal USP isoforms (either USP-A or USP-B) represents a functional receptor capable of binding the EcRE or the ecdysteroid ligand (Kapitskaya et al., 1996). Here, we report the developmental profiles of the mosquito USP isoforms, their differential responses to 20—hydroxyecdysone (20E) as well as functional differences with respect to their DNA binding and transactivation abilities. Taken together, these findings suggest that USP isoforms clearly perform distinct functions during mosquito vitellogenesis. Furthermore, these data suggest that it is USP-B that serves as a major partner for EcR during the mosquito vitellogenic response to ecdysteroid. MATERIALS AND METHODS Animals Mosquitoes, A. aegypti, were reared as described by Hays and Raikhel (1990). Vitellogenesis was initiated in 3-5 day old adult female mosquitoes by blood feeding them on rats. RNA Isolation and RT -PCR/Southern Blot Analysis Fat bodies and ovaries were dissected from female mosquitoes at different time points ranging from 0- to 5-day after eclosion at the previtellogenic stage, or from vitellogenic females ranging from 1 to 60 hours post-blood-meal (PBM). The temporal profiles of transcript abundance for the mosquito EcR, USP-A and USP-B in the mosquito fat body and ovary were examined using RT-PCR followed by Southern blotting with specific radioactive probes. Total RNA was prepared from fat body throughout the first vitellogenic cycle, using the guanidine isothiocyanate method as 32 described previously (Bose and Raikhel, 1988) with the modification that all isopropanol precipitation steps were done without low-temperature incubation to avoid co- precipitation of glycogen and salts. Alternatively, total RNA was extracted with TRIZOL Reagent (Gibco-BRL). RNA yields were determined spectrophotometrically, absorbency ratios A260/A280 and A260/A230 of RNA preparations were consistently above 1.7 and 2.0, respectively. Five microgram aliquots of each RNA preparation were reverse- transcribed by the SuperscriptII reverse transcriptase (Gibco-BRL) using random hexamer primers (Promega) in a reaction volume of 20 ul. The reverse transcription product was diluted to 40 p] with TE buffer (10 mM Tris-Cl, 1 mM EDTA, pH 8.0) and stored at -200C as a cDNA pool until use. For developmental profile analyses, 0.025 fat body-equivalents of cDNA pools were used as PCR templates in the case of mosquito fat body tissues whereas an equal amount of total RNA was used for reverse transcription in the case of ovarian tissues. A 522bp USP—A specific fragment was amplified with the primer pair: forward, 5’-TCATATCGTTCCGGAGATGTGG-3’ and reverse, 5’- CCAATCCTGCCAGAGGTAGTG-3’. A 400bp USP-B specific fragment was amplified with the primer pair, forward, 5’-CTTCTCACAAGAGGTGCTGAGO-3’and reverse, 5’- TGGTATCCAACTGGAACTTGCG-3’. A 314bp AaEcR specific fragment was amplified with the primer pair, forward, 5’-GAGGAAGATCAACATGACGTGC-3’ and reverse, 5’-ACCGTGAGGGAGAACATCTGC-3’. A 463-bp VCP (vitellogenic carboxypeptidase, Cho et al, 1991) specific fragment was amplified with the primer pair, forward, 5’-AGCGCCCATTCTTGTTTGG-3’ and reverse, S’-CAGCTCATACAGGTATTCTCC-3 ’. 33 tor 3 preps: [SP-.1. t‘ragm: lllCorp. 550C lf‘ Thermal cycling conditions were as follows: the reaction was incubated at 94°C for 3 min followed by 17 cycles of 94°C for 30 sec, 600C for 30 sec and 720C for 40 sec. As a reference to the developmental changes of the fat body, the same cDNA was subjected to 10 or 15 cycles of RT-PCR followed by Southern blotting with a primer pair specific to the mosquito VCP. After PCR amplification, 10 pl each out of the total 25p] reaction was fractionated on a 2% agarose gel, and transferred onto Hybond N+ membrane (Amersham) under alkaline conditions. Radioactive cDNA probes were prepared from 15 ng each of the PCR fragments generated by the primer pairs of EcR, USP-A, USP-B, or VCP with the corresponding plasmids as templates. These amplified fragments were labeled by a random-primers DNA labeling system (Gibco BRL) to incorporate [a-32P]-dATP (NEN). The membrane was hybridized with the probe at 650C in a solution of 5 x SSC, 5 x Denhardt' solution, 0.5%SDS, 25mM NaHzPO4, 25mM NazHPO4, 200 ug/ml of denatured salmon sperm DNA (Sigma) following prehybridization. The membrane was washed twice at 650C in 2 x SSC, 0.1%SDS for 15 min, twice in 0.1 x SSC, 0.1%SDS for 15 min, then autoradiographed. RT- PCR/Southem blotting of the total RNA preparations without the reverse transcriptase did not result in any appreciable signals, indicating that contamination of RNA preparations with genomic DNA fragments was virtually negligible (not shown). The radioactive blots were quantified with a phosphorimager (Molecular Dynamics). Examination of the EcR isoforms was performed in the same method as that in the USP isoforms. When conducting this experiment, AaEcR is named as AaEcR-B, and the new isoform is named AaEcR-A. The primers for AaEcR-A are: Forward, 5’- TGACGGCCATTCCGGCTTC; Reverse, GAGACGAGTAGCCATTGGAC. 34 In vitro Fat Body Culture To examine the transcriptional inducibility of mosquito EcR, USP-A and USP-B by ecdysteroid, the abdominal walls with adhering fat body (hereafter referred to as the fat body) were dissected from 3~5-day-old previtellogenic females and incubated in an organ culture system as previously described (Deitsch et al., 1995a; Raikhel et al., 1997). The fat bodies were incubated either with culture media in the absence of ecdysteroid or in the presence of 10’6 M 20E (unless otherwise indicated). At every 4-hour interval after incubation, 9 fat bodies of each time point were collected and immediately stored in liquid nitrogen until RNA isolation. For the hormone withdrawal experiments, fat bodies were washed 3 times with culture media after incubating with 10'6 M 20B for 4 hours and then further incubated in hormone-free media. Total RNA extraction and RT- PCR/Southem blot analyses were performed as described above. To confirm that the transcriptional response was due to the fat body and not the epidermis of the abdominal wall, the epidermis-free fat bodies were obtained by gently vortexing the dissected abdominal walls. The epidermis-free fat body was incubated in culture medium supplemented with 10'6 M 20E for 3-hr. The fat body adhering to the abdominal wall was incubated under the same conditions. Similar amounts of fat body tissue from both preparations were examined for the presence of two ZOE-induced genes, an early gene E 75 and the late gene VCP (Deitsch et al., 1995a; Pierceall et al., 1999). The levels of induction of both genes were similar in fat bodies of both preparations. However, the epidermis-free fat body could be kept alive for only a short incubation time (data not shown). 35 1T3115. with . C0110; 1111th desert gencrm acre ex 6131, 1 7.53, 5 mcmbf‘; monocj To test the effect of the protein synthesis inhibitor cycloheximide (Chx) on transcription, dissected 3-5 day old female mosquito fat bodies were pretreated for l-hr with culture media containing lmM Chx, then further incubated with the same concentration of Chx either without hormone or with 10'6 M 20E. Chx/hormone withdrawal experiments were conducted as for the hormone withdrawal experiment described above except that lmM Chx was added to all culture media. Antibodies and Western Blot Analysis The monoclonal antibody ABll against DmUSP (Christianson et al., 1992) was a generous gift from Dr. F .C. Kafatos (EMBL, Heidelberg, Germany). Nuclear proteins were extracted from the fat bodies of female mosquitoes as previously described (Miura et al., 1999). Protein extracts corresponding to 50 mosquito equivalents were resolved on 7.5% SDS-polyacrylamide gels, followed by electro-blotting to polyvinylidene difluoride membranes (Amersham). These membranes were probed with the anti-DmUSP monoclonal antibody (1:200 dilution) and then incubated with a secondary anti-mouse lgG antibodies (1:5,000 dilution) conjugated to horseradish peroxidase (Cappel). Immune complexes were visualized by addition of SuperSignal Ultra Chemiluminescent Substrate (Pierce) and autoradiography. In vitro Protein Synthesis and Electrophoretie Mobility Shift Assay (EMSA) Nuclear receptors were synthesized by the coupled in vitro transcription- translation (TNT) system (Promega). The EcoRI fragment of mosquito USP-A cDNA in pBluescript was subcloned into the EcoRI site of pGEM7Z (Promega). The same vector 36 W35 1. M197; 1996. which 11131111? £00511; ecdysr Sf'nthcs Pill} ltCr measm 133.1113“; Smith: was used to subclone DmEcR and DmUSP cDNA fragments (Wang, et al., 1998) whereas mosquito EcR and USP-B cDNAs were cloned into pGEM3Z (Kapitskaya et al., 1996). All these expression vectors utilized the SP6 promoter to synthesize proteins, which were first confirmed by 35S—Methionine labeling and SDS-PAGE following the manufacturer’s instruction. EMSA and measurement of equilibrium dissociation constants were conducted as described before (Wang et al., 1998). In brief, four ecdysteroid response elements (EcREs): IR,” -1 (agagacaagGGTTCAaTGCACTtgtccaat), IRper-l (agcttcaaAGGTCAaTGACCTtgtccatcg), DR-4 (aagcgaaAGGTCAaggaAGGTCAggaaaat) and DR12 (aagcgaaAGGTCAagaggccaaagaAGGTCAggaaaat) were end-labeled with y’ 3'ZP-ATP (DuPont NEN) with T4 polynucleotide kinase (Gibco BRL). Unless indicated, 50 fmol of labeled EcRE was incubated with 1 pl each of the indicated in vitro synthesized nuclear receptor proteins. Binding reactions were resolved using a 6% native polyacrylamide gel, which was then vacuum dried at 800C and exposed to X-ray film. To measure the equilibrium dissociation constants, bound and free EcRE probes were quantified with a phosphorimager, permitting the construction of saturation curves and Scatchard analysis (Wang et al., 1998). Construction of Reporter and Expression Plasmids and Cell T ransaetivation Assay Transactivation assays were conducted with the green African monkey kidney cell line CV-l (ATCC). The EcoRI fragment of USP-A was subcloned into the mammalian 37 expr: l'SP-i the re: )ll\'- 81 al.. 11611 p' follow 1 1 pt) 1‘." ether-t cells 11. galdt‘lt ~ am It}. C11 a, teen 1 :1} M,” and L‘SP- pnmtrs f I l I expression vector pCDNA3. l/Zeo (+), which was also used to express mosquito EcR and USP-B proteins (Wang et al., 1998). CMV-LacZ was used as a co-reporter to normalize the reporter gene activities of the two reporter plasmids AMTV-SxIRhSP-l—CAT and A MTV-3xDR4-CAT. Transfection assays were performed as described previously (Wang et al., 1998). In brief, CV-l cells were maintained in DMEM with 10% calf serum. Six- well plates were seeded at a density of 2x105 cells/well the day before transfection. Following the manufacturer’s instruction, cells were transfected for 5 hours with LipofectAMlNETM (Gibco BRL) and then added fresh medium together with either ethanol vehicle or 1 pM Muristerone A. After a hormone treatment of 36 or 48 hours, cells were harvested for CAT (chloramphenicol acetyl transferase) activity and [3- galactosidase activity. CAT activity was normalized with B-galactosidase activity. CAT activity was expressed as the percent of substrate converted to a product, and lunit of CAT activity was defined as 1% of the chloramphenicol substrate converted to its acetylated forms. RESULTS Tissue- and Stage-Specific mRNA Expression of the Mosquito USP Isoforms It has been previously reported that in vitellogenic tissues, the fat body and ovary, there are several transcripts corresponding to two Aedes aegypti USP isoforms (USP-A and USP-B) (Kapitskaya et al., 1996). Here, more detailed analysis was undertaken for the developmental changes in transcripts of the two USP isoforms in the fat body and ovary during vitellogenesis. RT-PCR was performed utilizing USP isoform-specific primers followed by Southem-Blot hybridization with isoform-specific probes (Fig. 1). 38 Fig.1. Expression profiles of USP and EcR mRN A during vitellogenesis. (A) Hormone and Yolk protein mRNA profiles. The whole body ecdysteroid titer at various time points (hours) in post vitellogenic female mosquito is converted to 20E equivalent (Hagedom et al., 1975). JH (juvenile hormone) titer is from Shapiro et al., 1986. The mRNA expression profiles of yolk protein Vg (Vitellogenin) and VCP (Vitellogenic carboxypeptidase) are from Deitsch et al., 1995b. The titers are measured at various days’ post-eclosion in previtellogenic mosquito and various hours’ post—blood meal in vitellogenic mosquitoes. Other abbreviations: E, eclosion; BM: blood meal. (B) USP and EcR mRNA profiles in the fat body. Total RNA was isolated from fat body dissected from previtellogenic female mosquitoes at O-ld, 1-2d, or 3-5d after eclosion (columns 1- 3), or from vitellogenic mosquitoes 1h, 3h, 4h, 6h, 15h, 24h, 36h or 48h PBM (columns 4-1 1). (C). USP and EcR profiles in the ovary. Total RNA was isolated from ovaries dissected from previtellogenic female mosquitoes, O-ld, 2—3d or 4-5d (columns 1-3) or vitellogenic mosquitoes 1h, 6h, 12h, 24h, 36h, or 48h PBM (column 4-9). RNA samples from each time point was subjected to reverse transcription and PCR amplification using either one pair of primers specific to AaEcR or two pairs of primers specific to USP-A and USP-B, respectively. Reverse transcribed products from equal number of fat bodies (fat body profile) or equal amount 5 pg total RNA (ovary profile) were used for PCR amplification. The PCR products were resolved by agarose gel electrophoresis, followed by Southern blotting and autoradiography. 39 A Days llours l’ost-liclosion Post-Blood-Meal 1% l ‘ 3 11M 4 X 12 16 20 24 28 32 36 42 4X 54 (IU OI) 72 b L Hormones 2015 III Yolk Protein mRNA Vs VC P B Day: Post-Ecloslon Hours Post-blood Meal 0-11-2 3-511 3 4 6 15 2436 48 3::3 ”23-3.1-3” ECR p ---—-----m Lane1234567891011 C Days Post-Ecloslon Hours Post-blood Meal 0.1 2-3 4-5 11 6 12 24 36 4a USP-Ab ‘. C p I- -u. USP-B> v~m~-~..., g. EcR> w..-“ Lane12 3 4 5 6 7 8 9 40 L A A l A I. I L 1 J 4 #1 .L.-_,-»__, Expr’ expel mat; l for t“! SSIIlC blot (1' (its. 1 5 PI? 'lIL’ mmed iFIS- 11 Expression of mosquito EcR was monitored in the same tissues as well. To minimize the experimental variability between PCR reactions, pooled RNA samples from fat body or ovary tissues were used in addition to low cycle numbers (17 cycles) for amplification. For control purposes, Northern blot analysis performed in parallel on samples from the same fat body or ovary tissues revealed identical mRNA profiles as RT-PCR/Southem blot analysis (data not shown). In the fat body, USP-A mRNA was highly expressed at 0-1 day after emergence (Fig. 18, lane 1), however its levels gradually decreased over the next 5 days of the previtellogenic period (Fig 18, lanes 2 and 3). The level of USP-A mRNA declined even more dramatically immediately after a blood meal; it dropped at 3-hr PBM (Fig. 1B, lane 5) and remained low during the entire active vitellogenic period until 24-hr PBM (Fig 18, lanes 6-9). The level of USP-A mRNA increased again in the fat body at the termination of the vitellogenic period, 36-hr PBM (Fig. 18, lane 10). The expression pattern of USP- B in the fat body was significantly different from that of USP-A. The level of the USP-B transcript was relatively constant throughout the pre- and early vitellogenic periods, but exhibited an obvious enhancement by 24- hr PBM (Fig 1B, lane 9). However, this transcript declined at termination of vitellogenesis at 36-hr PBM (Fig. 13, lane 10). The USP-A mRNA was abundant in ovaries of newly emerged female mosquitoes (Fig 1C, lane 1). Its level increased during the second day of previtellogenic development (Fig. 1C, lane 2), but decreased slightly in 4-5 day old previtellogenic females (Fig. 1C, lane 3). As in the fat body, a blood meal resulted in a dramatic drop in the USP-A mRNA level (Fig. 1C, lane 4) and the decline persisted throughout vitellogenesis (Fig. 1C, lanes 5-7). At 36-hr PBM, a small increase of USP-A mRNA was observed (Fig. 1C, 41 lane 8). Ovaries of newly emerged female mosquitoes also contained a detectable level of USP-B mRNA (Fig. 1C, lane 1) which was enhanced in 2- to 3-day old ovaries (Fig 1C, lanes 2-3). A blood meal triggered a considerable increase in the USP-B mRNA within one hour, and the level of USP-B transcript remained high during the first half of the vitellogenic period (Fig.1C, lanes 4-5). Thus, our results clearly demonstrated that in the adult mosquito female, the expression patterns of the two USP isoforms during the first vitellogenic cycle were dramatically different in both the fat body and ovaries. Levels of the EcR transcript were monitored in the same tissues and times as those of the USP isoforms. The levels of EcR transcript were relatively constant in the female fat body, showing elevations at the beginning of the pre-vitellogenic phase (Fig. 1B, lane 1), early vitellogenesis (Fig. 1B, lane 5) and post-vitellogenesis (Fig. 1B, lane 10). In the ovary, the EcR transcript level also exhibited three increases at approximately similar times as in the fat body (Fig. 1C). In Vitro E fleet of 2041 ydroxyecdysone on USP-A and USP-B Transcripts The dramatic differences in the expression patterns of mosquito USP isoform transcripts observed in vitellogenic tissues suggested that mosquito USP isoforms were differentially regulated during vitellogenesis. I began studying the control of their expression by examining the effect of 20E on these USP transcripts. These experiments were conducted using an in vitro fat body culture system, which has been well- established and is more suitable for these studies than small mosquito ovaries (Raikhel et al., 1997). In addition, unlike ovaries containing several cell types, the fat body is a relatively homogeneous tissue consisting of predominantly one cell type, trophocytes 42 mRNA (Fold-Induction. EcR 8- USP-B) 5 120 DEcR 4.5 T 2 4 'IVCP a? 8 1 0 .5 3.5 >_ g A... 80 g u: 3 .§ .5 E 3 25 60 b S 3 C 2 C 5 l t L9. 1.5 4° 9. § g a: 1 I! E , 20 E 0.5 0 - L- 0 10-8 10-7 10-6 10-5 205 (M) Fig. 2. Dose response of USP-B, EcR and. VCP to 20E in in vitro fat body organ culture. Fat bodies dissected from nine previtellogenic female mosquitoes were cultured for four hours in vitro either in the absence of hormone or in the presence of increasing concentrations of 20E ranging from 10’9M-10'S M. Total RNA was isolated from these fat bodies and subjected to RT-PCR with specific primers to EcR, USP-B and VCP, respectively. The PCR products were resolved by the agarose gel electrophoresis, followed by Southern blotting and autoradiography, and were quantified by the phosphorimaging analysis. The levels of EcR and USP-B mRNA from fat bodies cultured without hormone were defined as 1 unit. The maximum level of VCP mRNA from fat bodies incubated with 10’6M 20E was defined as 100, because VCP mRNA level was below detection in the previtellogenic fat bodies before the hormone treatment. 43 inctix incub sou: lhctf contr. isoforr‘ tuhurc afierin express: COEIhlLt 1311:11 Squares) 1 (Raikhel, 1992), making interpretation of results easier. Abdominal walls with adhering fat bodies, isolated from previtellogenic female mosquitoes (3-5 days after eclosion) were incubated in culture medium either in the absence of hormone or in the presence of increasing amounts of 20E ranging from 10'9 M-lO'5 M (Fig. 2). After a 4-hour incubation, these fat bodies were subjected to RNA isolation, RT-PCR and Southern blotting analyses. USP-B and EcR were activated by 20E in a dose-dependent manner. The mRNA levels of both EcR and USP-B were maximal at 10'6M 20E (Fig. 2). In contrast, USP-A mRNA was not induced by any concentration of 20E (data not shown). As a positive control, I monitored the 20E dose response of vitellogenic carboxypeptidase (VCP) mRNA, one of the late target genes in the mosquito fat body (Cho et al., 1991; Deitsch et al., 1995a,b). This ecdysteroid-responsive gene exhibited a peak induction level at 10'6 M 20E (Fig. 2). Next, I investigated the time course of 20E effects on the expression of USP isoforms. In this experiment, dissected previtellogenic fat bodies were incubated in culture media in the presence or absence of 10’6M 20E (Fig. 3, A-D). At 4-hour intervals after incubation, USP transcripts were analyzed as described above. The effect of 20E on expression of the two USP isoforms in cultured previtellogenic fat bodies was opposite: continuous presence of 20E in the culture medium failed to induce the expression of USP-A (Fig. 3A, open squires), while it activated USP-B expression (Fig. 3B, open squares). USP-B transcript levels steadily increased with 5-, 7- and 10-fold enhancement at 8-, 12- and 16-hr of incubation, respectively (Fig. 3B, open squares). When fat bodies were incubated for four hours in the presence of 20B and then transferred into a hormone- free medium, the level of the USP-A transcript rose dramatically (Fig. 3A, open 44 bUd. triangles). In contrast, the USP-B transcript declined to its background level after an initial rise at 4-hr (Fig. 38, open triangles). The effect of 20E on the expression of AaEcR in in vitro cultured fat bodies was different from that of both USP isoforms. Incubation in hormone-free medium for l6-hr resulted in a steady increase from the high basal level of the EcR transcript present in the previtellogenic fat body (Fig. 3C, open circles). Continuous incubation of the fat body in 20E-containing culture medium first led to a rapid increase of the AaEcR transcript level by 4-hr incubation and then a considerably slower rise until 12-hr after incubation when the EcR level reached a plateau (Fig. 3C , open squares). Interestingly, withdrawal of the hormone after 4-hr of incubation caused the EcR transcript level to rise to a two-fold higher level (Fig. 3C, open triangles). As a positive control, I monitored the effect of 20E on VCP. Previtellogenic fat bodies incubated in hormone-free media did not show any discernible level of VCP expression (Fig. 3D). This is consistent with results reported before (Deitsch et al., 1995a). Continuous incubation in the presence of 20E resulted in the robust activation of VCP gene expression: the level of VCP mRNA doubled every 4 hours during the 12-hour incubation period and then continued to increase to the end of incubation (Fig. 3D, open squares). Hormone withdrawal after a 4-hr induction period reversed VCP gene activation, and the VCP mRNA levels retumed to an undetectable level by l6-hr of incubation (Fig. 3D, open triangles). 45 a... 1.2.1.; . . .uw. '1. 1. . l . ._ . .......... ......:....4.1. 4.. .1. .fiu.....$ if .99.... .. .4 .. . .. .? ......... _ . ,.._ a”, .. . :2... f... ...:.u.u...1..... . .11 Fig. 3. Effect of 20-hydroxyecdysone and cycloheximide on the transcription of USP-A (A and E, respectively), USP-B (B and F), EcR (C and G) and VCP (D and H) in the previtellogenic fat bodies. Previtellogenic fat bodies dissected from female mosquitoes (3-5 day after eclosion) were incubated in culture media only (open circles), with a 4-hour pulse treatment of 10'6M 20E (open triangles), with continuous 20E treatment (open squares), with lmM Chx (closed circles), with lmM Chx and a 4-hour pulse 20E treatment (closed triangles) or with lmM Chx and continuous 20E treatment (closed squares) for 16 hours. At every 4-hour interval, a group of 9 fat bodies were collected and subjected to RNA isolation, RT-PCR amplification and Southern blot analysis. The intensity of bands from Southern blots was quantified by phosphorimaging. For EcR, USP-A and USP-B, the level of amplified transcript detected from previtellogenic fat bodies prior to any treatment was defined as 1 unit. For the VCP mRNA, the level of amplified transcript detected from previtellogenic fat body treated only with 20E for 16 hours was defined as 100 units. For comparison, the mRNA profiles of fat bodies treated with 20E continuously were plotted in both, left and right, panels (dotted curve with open squares). 46 20E/Chx 20E --_ .. __.__q a .P _S "U __ m h h 4 em. mp. mmm CCC. 8 6 4 2 O E USP-A ——-a—— 20E-4h - o - -2OE ——-o-—CM 00000000 4208642 4| 22635.28. - - .3 ' ‘4 Lane12345678910 Fig. 4. Developmental profiles of USP proteins revealed by Western blot. Fat body nuclear extracts were isolated from female mosquitoes at previtellogenic stages 0—0.5 day, l-2 day, 3-5 day (lanes 3-5), or post blood meal stages PBM 3-6h, l8-24h, 36h, 48h, 60h (lanes 6-10). Protein extracts, equivalent to 50 fat bodies per lane, were loaded in each lane for SDS-PAGE and Western blot analysis with the anti-DmUSP monoclonal antibody. In parallel, in vitro TNT expressed USP-A and USP-B proteins (lanes 1 and 2) were analyzed as positive controls. Arrowheads indicate the sizes of major bands. 32p DR-4 2 1!: «fl ; 9; :z T— L‘ % A % if. j {g If TNT Lys-te 2 S S S 5‘ _§ 3 DmUSP Ab - s ‘+—'. + - 4» - - U Lane 1 2 3 4 5 6 7 8 9 10 1112 13 14 Fig. 5. EcR/USP binding to a direct repeat DR-4. 32P labeled DR-4 EcRE was incubated with in vitro synthesized AaEcR (lane 1), USP-B (lanes 2 and 3), AaEcR and USP-B (lanes 4 and 5), DmEcR (lane 6), DmUSP (lanes 7 and 8), DmEcR and DmUSP (lanes 9 and 10), USP-A (lanes 13 and 14) or AaEcR and USP-A (lanes 1 l and 12) and subjected to EMSA. Anti-DmUSP antibody was used for supershift assays to verify binding specificity (lanes 4, 9 and l 1). TNT Lycato AaEcR A-EcR DmEcR AaUSP-A AaUSP—B DmUSP DmUSP Ab '-—¥_ fi‘ 'ifi'v’ ‘— " “‘ ' PO. -‘~ . w l 4.1.1, - 1 2 3 4 5 6 Lana Fig. 6. EcR/USP binding to DR-12. ”P-labeled DR12 was incubated with in vitro synthesized AaEcR and USP-A (Lanes l-2), AaEcR and USP-B (lanes 3-4) or DmEcR and DmUSP (Lane 5-6) and subjected to EMSA. Supershift assays were conducted with anti-DmUSP antibody (Lanes 2, 4 and 6). 50 lei'1 lexc deel CUM] “35 1 tr“) M , r “1‘54 level of nuclear USP-A protein in the fat body drastically decreased (Fig. 4, lane 7). The level of USP-A increased again at the late vitellogenic stage, 36-48 hr PBM, before declining again at 60-hr PBM. In contrast to USP-A, the USP-B protein appeared in the fat body of previtellogenic, 1-2 day old mosquitoes and reached a high level in 3-5 day old females. Moreover, the level of USP-B protein remained without any significant changes throughout the entire vitellogenic and postvitellogenic periods (Fig. 4). DNA Binding Properties of the E cR/USP-A Heterodimer: the E cR/USP-A complex is Capable of Binding to Various EcREs It has been shown before that both heterodimers, EcR/USP-A and EcR/USP-B, bind to the hsp27 response element IR”“”-l (Kapitskaya et al., 1996). More recently, it has been demonstrated that the EcR/USP—B heterodimer displays promiscuous binding and transactivation properties towards DNA elements derived from the AGGTCA half- site, arranged either as inverted repeats or as direct repeats (Wang et al., 1998). Here, it was investigated whether the EcR/USP-A heterodimer exhibited DNA binding and transactivation properties similar to those of the EcR/USP-B heterodimer. First, EMSA was conducted with the inverted repeats containing the perfect consensus sequence AGGTCA separated by a single nucleotide spacer (IRpcr-l). These experiments showed that the EcR/USP-A heterodimer bound efficiently to IRp°'-1 (data not shown). Next, a direct repeat of the AGGTCA half-site with a four-nucleotide spacer (DR- 4 element) was tested. This sequence has been shown to have an optimal spacer among 51 lm 0’1 direct repeats for mosquito EcR/USP-B and Drosophila EcR/USP (Wang et al., 1998; Antoniewski et al., 1996; Vogtli et al., 1998). TNT—expressed AaEcR, USP-A, USP-B, DmEcR or DmUSP proteins alone did not exhibit any binding to DR-4 (Fig. 5, lanes 1-3, 6-8, 13 and 14). A combination of mosquito AaEcR and USP-B lysates yielded efficient binding, whose specificity was confirmed by supershifts using the anti-DmUSP monoclonal antibody (Fig. 5, lanes 4 and 5). Similar binding activity was detected for the DmEcR/DmUSP heterodimer (Fig. 5, lanes 9 and 10). Likewise, the AaEcR/USP-A heterodimer displayed similar specific binding to the DR-4 element as confirmed by a supershift assay using anti-DmUSP monoclonal antibody (Fig 5, lanes 11 and 12). Then it was investigated whether AaEcR/USP-A could bind to a direct repeat of the AGGTCA half-site with a 12-nucleotide spacer (DR-12), which displays robust binding activity with the AaEcR/USP-B and DmEcR/DmUSP heterodimers (D’Avino et al., 1995; Wang et al., 1998). EMSA revealed that the AaEcR/USP-A bound the DR-12 element efficiently, and the specificity of binding was demonstrated by a supershift with anti-DmUSP antibody (Fig 6). Taken together, these experiments indicate that the AaEcR/USP-A complex, like its AaEcR/USP-B complex counterpart, is capable of binding to various EcREs including inverted and direct repeats as well as widely spaced repeats such as DR-12. Estimation of Equilibrium Dissociation Constant (K4): AaEcR/USP-B Bound on EcRE Twice as Strongly as AaEcR/USP-A The similar binding properties of the AaEcR/USP-A and AaEcR/USP-B heterodimers prompted us to investigate whether they might possess differential binding 52 AProbe Concen.(nM) 20 8.0 4.0 2.0 1.0 0.4 ”a. Q's" :1 La ne B Bound (nl'l) 0.2 Boumvru-r 0 I) \ V o ' 0 ()_N’ 002‘» 0020 I ”HIS - 0.05 omn ””mn nm or on o: lhmmi (n3!) 0 E A 0 5 10 15 20 25 EeRE(thsp-1) (an) Fig.7, Direct measurement of equilibrium dissociation constants (Kd). Increasing amounts of 32F labeled IRhSp-l EcRE probe ranging from 0.4nM-20nM was incubated with in vitro synthesized AaEcR and USP-A proteins and subjected to EMSA. Bound and free probes were quantified by phosphorimager analysis. Saturation curve (B) and Scatchard plot (inset in panel B) were prepared to calculate the Kd. This experiment was repeated three times and the mean value taken as the Kd. 53 all: billt. DR- \\ he a {‘11 7 affinities for EcREs. The AaEcR/USP-B heterodimer binds various EcREs with different binding affinities. The equilibrium dissociation constants (de) for IRhSp—l, IRper-l and DR-4 are 3.73 nM, 0.326 nM and 2.21nM, respectively (Wang et al., 1998). To evaluate whether combinations of EcR with different USP isoforms exhibit different binding affinities for these DNA elements, the Kd of AaEcR/USP-A to each [Rpm-1, IRhSp-land DR-4 was estimated. The AaEcR/U SP heterodimer was incubated in the presence of 5x10'7 M 20E, with increasing concentrations of radioactive probes, IRhSp-l or DR—4. Saturation binding analyses and Scatchard plots were used to estimate Kd values for the heterodimeric pairs bound to thSp-l (Fig. 7) or DR-4 (not shown). These analyses indicated that like the AaEcR/USP-B heterodimer, the AaEcR/USP-A bound to various EcREs with different affinities. The differences in Kd values for these EcREs showed that the binding affinity of EcR/USP-A to a DR-4 was about 2-fold higher than to an IRhSp-l (Table 1), yet ten times weaker than to the IRper-l (not shown). More importantly, these results demonstrated the AaEcR/USP-A complex bound to each EcRE with a 2-fold lower affinity than the corresponding AaEcR/USP-B complex, suggesting that USP-B is a more potent heterodimerization partner for EcR (Table 1). USP-B is a More Potent EcR Partner than USP-A for Transactivation via EcREs in C V-I Cells Transactivation properties of two mosquito heterodimers were next compared using the CV-l cell line. This mammalian cell line has no endogenous EcR and contains very low endogenous level of RXR. It has been used to study transactivation of EcR/USP from several arthropod species including Drosophila and mosquito (Thomas 54 A a .m DEtOH 3 Themes g. 61 8 < 5 '8 E ‘* é 3 ’5 2“ : 5 o F‘- rtl :- ’5xtR”"’-1 ._.. ........................ .+.. ...................... ;. ........................ ; ................ 4. ........ EcR - - + - + 39§ET§ ‘ ............... T ...................... T. ......................... f ...................... f ...... B 09 ,_ 0.3 ,5 g 0.71 0.6. 3 :5, 0.5 3 g o... E’ 03 E . 3 0.2- '— 01 o Y . .. EtOH MurA Fig.8. EcR/USP-A transactivated a reporter gene with either IR-l or DR-4 EcREs in CV-l cells. A. The AMTV-leRhsP-l-CAT reporter gene (1.2ug) and 0.4ug CMV- LacZ were transfected into CV-l cells with 0.4ug of either AaEcR or USP-A plasmid or 0.4ug each of the two expression vectors. The expression vector pCDNA3.1/zeo (+) was used as carrier DNA to equalize the total amount of plasmid so that each well received 2.4ug DNA. Cells receiving no exogenous DNA were used as a mock control. B. The AMTV—3xDR-4-CAT reporter gene (1.2ug) and 0.4ug CMV-LacZ were transfected into CV-l cells with 0.4ug of AaEcR and USP-A expression vectors. After transfection cells were incubated with either vehicle (ethanol) or luM MurA at 37°C for 36 hours and harvested for CAT activity and B-galactosidase activity, and CAT activity was normalized with B-galactosidase activity. The experiments were repeated more than three times. 55 et al., 1993; Yao et al., 1992; 1993; Wang et al., 1998). It has been shown previously that transfection of AaEcR/USP—B into CV-l cells rendered them ecdysteroid responsive. Furthermore, it has been shown that the levels of reporter transactivation by AaEcR/USP- B correlated with the DNA binding affinities of this heterodimer when three distinct three functional EcREs were compared (Wang et al., 1998). The transactivation potential of the AaEcR/USP-A heterodimer was assessed with the AMTV-leRhSp-l-C AT reporter plasmid, which contains five tandem repeats of IR”— 1. Transfection of CV-l cells with the reporter alone resulted in a very low basal level of CAT activity (Fig. 8A). Co-transfection of the reporter with USP-A alone did not confer MurA responsiveness. Weak response to MurA was detected in cells transfected with the reporter gene and AaEcR expression vector, suggesting that AaEcR can heterodimerize with endogenous RXR to activate this reporter gene. However, strong activation of the reporter gene by 1 uM MurA (50-fold) was noted only in cells co-transfected with AaEcR and USP-A expression vectors (Fig. 8A). This indicated that the AaEcR/USP-A heterodimer formed a functional hormone-dependent transactivator in CV—l cells. Next, to test whether the AaEcR/USP-A heterodimer could also activate a reporter gene containing the DR-4 as an EcRE, the AMTV-3xDR-4-CAT reporter plasmid containing three tandem repeats of DR-4 was transfected into CV—l cells. Co- transfection of this reporter construct with either the AaEcR or the USP-A alone did not render CV-l cells ecdysteroid-responsive (data not shown). However, co-transfecting the reporter construct with both AaEcR and USP-A expression vectors rendered CV-l cells highly responsive to the ecdysteroid MurA giving 6-fold induction (Fig. 8B). 56 Table 1. The equilibrium dissociation constants (Kd) of different DNA sequences binding to AaEcR/USP-A and AaEcR/USP-B. EcRE IR'””-l(nM)* DR-4 (nM)* AaEcR/USP-A 7.36i0.81 4.06:0.05 AaEcR/USP-B 3.73:0.85 2.21:0.36 To measure the Kd values, in vitro synthesized AaEcR and USP-A were incubated with , increasing concentrations of 32F labeled IR,” -1 or DR-4 as described in Fig. 6. EMSA and quantification for each element were performed a minimum of three times and the mean values were taken as the Kd. For comparison, Kd values for EcR/USP-B to the same elements are also listed (Wang et al., 1998. with permission). * Mean of three determinations : SE CAT Activity (Percent of Acetylatlon) 0 I 1 IT I l T I l l USP (no) 0 (55 bis (1‘? .133 (go Kgo (19° b9o 19° Fig. 9. AaEcR/USP-B mediated more efficient transactivation than EcR/USP-A. Reporter plasmid (0.6ug), CMV-LacZ (0.2ug) and AaEcR plasmid (0.2ug) were transfected into CV-l cells with increasing amounts of either USP-A or USP-B expression vector ranging from 3.1ng-800ng. The expression vector pCDNA3.1/zeo (+) was used as carrier DNA to equalize the total amount of plasmid so that each well received 2.0ug DNA. Cells receiving no exogenous DNA were used as a mock control. After transfection, cells were incubated with luM MurA at 370C for 48 hours and harvested for CAT activity and B-galactosidase activity. CAT activity was normalized to B-galactosidase activity. The experiments were repeated three times. 57 The transactivation potentials of the two heterodimers were then compared. In this experiment (Fig. 9), a fixed amount of reporter plasmid (AMTV-SxIRhSPl-CAT, 600ng) and an AaEcR expression vector (200mg) were co-transfected into CV-l cells along with increasing amounts of either USP-A or USP-B expression vectors, ranging from 3 ng to 800 ng. Cells transfected with a reporter plasmid and the AaEcR expression vector alone displayed less than 0.5 units of CAT activity (not shown). Co-transfecting these cells with the USP-A plasmid increased the Mur A-dependant CAT activity in proportion to the amount of USP plasmid added. The activation reached saturation with 200 ng of USP-A plasmid (Fig. 9). Co-transfecting the cells with USP—B instead of USP- A resulted in higher Mur A-dependant activation of the reporter at lower levels of input plasmid. Notably, the USP-B mediated transactivation reached its peak with about half the amount of input plasmid compared to that of USP-A (lOOng versus 200 ng). In a separate experiment, transactivation potentials of the two heterodimers were compared using the AMTV-3xDR-4—CAT reporter plasmid. Co-transfection of this reporter with the AaEcR and AaUSP-A expression plasmids consistently led to a lower level of reporter induction than with the AaEcR and AaUSP-B plasmids (data not shown). Taken together, these results further suggested that USP-B is a more potent partner than USP-A for EcR mediated transactivation in CV-l cells. DISCUSSION In this work, the expression profiles of USP isoforms were characterized during the first vitellogenic cycle in the female mosquito A. aegypti. The transcriptional responses of USP isoforms to 20E were investigated, and the DNA-binding and 58 transactivation properties of the EcR/USP-A and EcR/USP-B heterodimers were compared by employing EMSA and transfection assays. Importantly, here two mosquito USP isoforms, USP-A and USP-B, displayed distinct stage-specific expression in two vitellogenic tissues, the fat body and ovary. USP-A mRNA was highly expressed in previtellogenic and late vitellogenic fat bodies and ovaries, coinciding with a period of low ecdysteroid titer, whereas USP-B mRNA was enriched at the vitellogenic stage when the ecdysteroid titer in the female mosquito is high. Significantly, USP-B, but not USP- A mRNA levels, correlated with the level of EcR in both tissues. The mRNA profiles of the USP isoforms strongly suggest that ecdysteroids be involved in differential regulation of their respective promoters. Indeed, when I utilized the fat body in vitro organ culture to investigate the effect of 20E on the transcription of USP isoforms, these experiments unequivocally established ecdysteroid engagement in controlling the transcription of both USP-A and USP-B. Remarkably, the effect of 20E on USP-A expression was opposite to that of USP-B expression. Continuous exposure to 20E abrogated the transcription of USP-A. However, a 4-hour pulse treatment of fat bodies with 20E followed by continued incubation in hormone-free medium resulted in a drastic augmentation of USP-A transcription. Interestingly, these USP-A mRNA responses to 20E in vitro are similar to those of AaFTZ-F 1. According to our observations, AaFTZ-Fl is also suppressed by 20E, but is super-activated under conditions of 20E withdrawal (Li C., Kapitskaya, M.Z. and Raikhel, A.S., unpublished observation). FTZ-F 1 , an insect homologue of the steroidogenic factor 1 (SF-1), has been implicated in regulating a stage-specific response to ecdysteroid (Woodard et al., 1994). This similarity in the effect of 2013 suggests that, as in the case of FTZ-Fl, the 59 nuclear regulatory receptor, HR3 (Hormone Receptor 3) may be involved in mediating the ZOE effect on USP-A. HR3 is involved in mediating 20E responsiveness in DmSOphila (Lam et al., 1997; White et al., 1997). Moreover, the HR3 homologue has been identified in the mosquito and has been found to respond positively to 20E in an in vitro fat body culture system (Kapitskaya, M.Z., Li, C. and Raikhel, A.S., unpublished results). The observation that Chx completely inhibited USP-A super-induction following 20E withdrawal further substantiated the involvement of HR3 or other labile factors in activation of USP-A expression. In contrast to USP-A, the USP-B transcript was highly elevated by the continuous presence of 2013 in an in vitro fat body assay. Furthermore, continuous exposure of the fat body to 2013 was required for maximal activation of USP-B transcription. In the presence of Chx, 20E super-induced USP-B transcription, suggesting that it represents an early gene of the ecdysteroid gene regulatory hierarchy. One critical criterion to differentiate early and late genes is the effect of a protein synthesis inhibitor such as Chx on their response to hormone (Ashbumer et al., 1974; Huet et al., 1995). Vg and VCP genes, which are transcriptionally induced by ecdysteroid but inhibited by Chx, are typical late genes in the mosquito (Deitsch et al., 19953). In contrast, addition of cycloheximide into the culture media super-induces AaE75, a mosquito early gene (Pierceall et al., 1999). Of note, the effects of 20E on EcR in the mosquito fat body, as well as the EcR mRNA response to Chx were generally similar to those of USP-B. However, as in the case of EcR expression, a more complex pattern of response to 20E compared to that of USP-B was observed (Fig. 3). This may reflect the presence of more than one isoform of EcR as has been shown for M. sexta (Jindra et al., 1996a). In 60 r}. lSU llo contrast to USP, only a single EcR isoform corresponding to Drosophila EcR—B1 has been identified so far in the mosquito (Cho et al., 1995). However, our recent studies suggest that there is at least one additional EcR isoform present in the mosquito during vitellogenesis (Wang, S.F., Li, C., Zhu, 1.8. and Raikhel, A.S. unpublished data). Interestingly, USP isoforms have been found to be differentially expressed during the Manduca epidermis during larval molts (J indra et al., 1997). When the ecdysteroid titer is high, the Manduca USPl transcript (the counterpart of USP-A) disappears and the levels of the USP2 transcript (the counterpart of USP-B) are elevated, resembling the USP isoform expression profiles in the adult female mosquito during vitellogenesis. Moreover, in vitro exposure of the day-2 fourth instar larval epidermis to 20E led to the decrease of Manduca USP-1 mRNA, whereas the effect of 20E on USP-2 mRNA was opposite causing its levels to rise (Hiruma et al, 1999). A differential effect of 2013 has also been reported for Manduca EcR isoforms (Jindra et al., 1996b). In vertebrates, ligands can cause different or even opposite responses of various receptor isoforms (Syvala et al., 1996; Paech et al., 1997; Keightley, 1998). In the fat body of newly eclosed female mosquitoes, transcripts of both USP isoforms and EcR are present with the USP-A transcript being particularly abundant. However, our immunoblot analysis showed that the corresponding proteins were not detectable at this stage in the fat body. A high mobility 43-kDa protein, recognized by the USP monoclonal antibody, was observed in the newly eclosed female fat body. During the extraction of the fat body nuclear proteins, a cocktail of proteinase inhibitors was added and all samples were processed under identical conditions. Therefore, it is unlikely that the 43-kDa protein is a degradation product of USP-A or USP-B. Our previous 61 Northern blot analyses have demonstrated the existence of four USP transcripts of different sizes in both the fat body and ovary (Kapitskaya et al., 1996). Utilization of isoform—specific probes derived from the 5’- and 3’- untranslated regions of USP-A and USP-B revealed that the largest 3-kb transcript corresponds to USP-A, while two smaller transcripts of 2.5 and 2.2 kb correspond to USP-B (Kapitskaya et al., 1996). The identity of the smallest 1.8-kb transcript remains unknown. It is possible that a newly identified 43—kDa USP-related polypeptide is a product of the 1.8-kb transcript. This suggestion, however, requires further study. Traces of the USP-B protein appeared in 1-2 day old mosquitoes and reached high levels in 3-5 day-old mosquitoes. By contrast, only a low level of the USP-A protein was detectable in 3-5 day-old mosquitoes. EMSA utilizing nuclear extracts from different stages of previtellogenic fat bodies also showed the appearance of functional AaEcR/U SP heterodimers only in fat bodies of previtellogenic females 3-5 days after eclosion (Zhu, J .S. and Raikhel, A.S., unpublished data). Thus, since the USP-B protein is the most abundant USP isoform in the fat body of 3-5 day old previtellogenic females, an increase in the USP-B protein along with EcR protein, which is also appeared at this stage, may promote competence for the vitellogenic response to a blood meal (Zhu, J .S., Wang, S.F., Li, C. and Raikhel, A.S., unpublished data). During the previtellogenic phase which covers the period from adult emergence to 3-5 days after eclosion, the female mosquito becomes competent for vitellogenic events (reviewed by Hagedom, 1989; Raikhel, 1992). The previtellogenic phase is accompanied by a rise in the juvenile hormone (JH) titer, which reaches its peak two days after eclosion and then declines (Fig. l-A; Shapiro et al., 1986). Acquisition of competency to 62 Elia vitellogenic events and responsiveness to ecdysteroid have been shown to be controlled by JH (reviewed in Raikhel, 1992; Dhadialla and Raikhel, 1994). In the ovary, the development of the receptor-mediated complex responsible for yolk protein uptake is controlled by JH (Raikhel and Lea, 1985). In the fat body, JH-dependent events include an increase in ploidy and proliferation of ribosomes (Dittmann et al., 1989; Raikhel and Lea, 1990). However, the molecular mechanism of JH action in the mosquito is unknown. In view of our observations on the delayed appearance of USP protein during previtellogenic development, it is possible that .lH acts at the translational level regulating coordinated production of a set of transcription factors required for rapid activation of target genes in response to blood-meal induced hormones. Verification of this hypothesis requires further study. The level of the USP-A protein increased dramatically in the fat body in response to a blood meal. However, at the active phase of vitellogenesis, 18-24 hr PBM, the USP- A protein dropped to quite a low level, consistent with the decrease in USP-A mRNA level, which was also absent at this time. In contrast to the fluctuation of USP-A mRN A and protein levels during the vitellogenic stage, USP-B mRNA and protein levels are expressed evenly high throughout this stage, suggesting once again that USP-B may be the major partner for EcR function during vitellogenesis. In order to provide additional insights into the possible roles of USP isoforms in the mosquito, we have compared the DNA-binding and transactivation properties of the EcR/USP-A heterodimer with those of the EcR/USP-B heterodimer which we have characterized previously (Wang et al., 1998). Although the extent of our present analysis has not been as detailed as for USP-B (Wang et al., 1998), it appears that USP-A can 63 effectively dimerize with EcR, and the resulting heterodimer exhibits a wide specificity of EcRE recognition similar to that of the AaEcR/USP-B heterodimer. The EcR/USP-A heterodimer bound inverted repeats (thSp-l and IRper-l), direct repeats (DR-4) and widely spaced EcREs such as DR-12. However, by measuring binding affinities of both heterodimers we have clearly shown that the EcR/USP-B consistently displayed higher binding affinity than EcR/USP-A to either direct or inverted repeats. Moreover, although EcR/USP-A mediates ligand-induced transactivation in CV-l cells from either IRhsP-l or DR-4 EcREs, once again the EcR/USP-B heterodimer appears to be the more effective transactivator. The functional difference between USP-A and USP-B in mosquito cells could be more profound than observed in CV-l cells. The Manduca EcR heterodimerized with either USP-1 or USP-2 bind IR,” -1 and another imperfect palindrome (EcREl) from the MHR3 promoter. Yet only EcR-USP], but not EcR/USP—Z, efficiently transactivated the MHR3 promoter in Manduca GV-l cells (Lan et al., 1999). Thus, these findings have demonstrated that throughout the vitellogenic cycle, the mosquito USP isoforms are differentially regulated at both the transcriptional and translational levels. They exhibit strikingly different responses to 20E in fat body cultured in vitro. Our functional studies of the DNA binding and transactivation properties indicate that, although both isoforms form functional heterodimers with EcR capable of recognizing a wide range of EcREs, USP-B forms a more efficient functional ecdysteroid receptor than USP-A. Taken together, our findings strongly suggest that USP-B is the major heterodimeric partner of EcR responsible for mediating the ecdysteroid response during vitellogenesis in the mosquito. Elucidation of the possible role of USP-A in the adult female mosquito requires further investigation. 64 Sequence comparison of the AaEcR with Drosophila EcR isoforms revealed that the AaEcR shows high similarity with Drosophila EcR-B. Since many insect EcR isoforms have been identified, it was likely to exist more than one EcR isoform in mosquitoes. Indeed, most recently, the second AaEcR isoform has been identified in mosquitoes by means of PCR amplification using degenerated primers based on highly conserved regions in the EcR-A A/B domain between Drosophila and other insects (Fig. 10A; Wang, S-F, Li, C. and Raikhel, A., unpublished). Subsequently, the PCR generated DNA fragment was used to screen the mosquito vitellogenic cDNA library. However, extensive library screening failed to identify a cDNA clone in this library (Li, C. and Raikhel, A., unpublished). When I performed PCR using a primer located on the PCR- generated AaEcR-A fragment and a second primer on the 3’UTR of the AaEcR common region, I amplified the entire nucleotide sequence of AaEcR-A from the vitellogenic- staged fat body cDNA. This result further has confirmed existence of AaEcR-A in mosquito fat bodies (now shown). Next, I used AaEcR isoform-specific primers to examine the expression levels of AaEcR-A in both fat bodies and ovaries (Fig. 10B). In fat bodies, AaEcR-A is barely detectable during the previtellogenic stage; whereas, its levels increase after a blood meal with a peak level at 24hr PBM followed by a rapid decline to the previtellogenic level at 48hr PBM. In ovaries, AaEcR-A is expressed at relatively high levels throughout the previtellogenic and vitellogenic stages with elevated levels after a blood meal (Fig. 108). Thus, the AaEcR-A expression levels appear to correlate with 20E levels during vitellogenesis. 65 A. E F A/B C D EcR-A m1 f _J:JH°mone EcR-B l f H___.l.__lormone Fat body Hours After a Blood Meal PV 3 4 6 15 24 36 48 EcR-A W G - .. .... EcR-B HQQQ~~-M Ovary Hours After a Blood Meal PV 6 12 18 24 30 36 42 48 EcR-A use- cm.-- 9 EcR-B ,, . ,- ,- .. an-..‘ ”O C, o 4 s 12 16 (hr) Culture EcR-A - - m. 1“ fl EcR-B . -‘ “1.“ Fig.10. Characteristics of the AaEcR-A isoform. (A). Schematic domain structures of the mosquito EcR isoforms. (B). Expression profiles of AaEcR-A in fat bodies and ovaries by RT-PCR/Southern analysis. (C). Induction of AaEcR-A mRNA in the fat body culture. 66 The correlation of the expression pattern of AaEcR-A in the fat body with 20E levels suggests that AaEcR-A expression may be under the control of 20E. To test this hypothesis, previtellogenic fat bodies were incubated with 20E for 16hr, and the mRNA levels of AaEcR-A were monitored during this period of time. Indeed, I detected that the AaEcR-A mRNA levels rose gradually during the 16hr culture with a maximal level at 16hr, indicating that AaEcR-A is more inducible by 20E than the AaEcR-B (Fig. 10C). In the fat body, the substantial low level of AaEcR-A expression during the previtellogenic stage, and relatively high level expression only within a limited time frame during the vitellogenic stage, suggest that AaEcR-A may not be a major receptor for the ecdysone response during vitellogenesis. Elucidation of the possible role of AaEcR-A in mosquito vitellogenesis requires further investigation. ACKNOWLEDGMENTS I thank Sheng-fu Wang for DNA binding and cell transfection assays and initiation of the AaEcR isoform project, J insong Zhu for Western blot analysis and Ken Miura for some RT-PCR experiments. I also thank Alan R. Hays’ assistance in mosquito rearing and Dr. F. C. Kafatos for providing us with the Drosophila USP monoclonal antibody. This work was supported by NIH grant Al-36959. 67 Chapter 3* Characterization of the Mosquito Early Gene E 75 *Most results have originally been published in Mol. Cell. Endocrinol. Ref: Pierceall, W: E., Li, Cf, Biran, A., Miura, K., Raikhel, A. S., and Segraves, W. A. (1999). Mol. Cell Endocrinol. 150, 73-89. " Underlined two authors contributed equally to this work. 68 d: 111,.) L. Inc ABSTRACT The steroid hormone ecdysone# controls genetic regulatory hierarchies underlying insect molting, metamorphosis and, in some insects, reproduction. Cytogenetic and molecular analysis of ecdysone response in Drosophila larval salivary glands has revealed regulatory hierarchies including early genes which encode transcription factors controlling late ecdysone response. In order to determine whether similar hierarchies control reproductive ecdysone response, ecdysone-regulated gene expression in vitellogenic mosquito ovaries and fat bodies has been investigated. Here, the homologue of the Drosophila E 75 early ecdysone inducible gene has been identified in the yellow fever mosquito Aedes aegypti, and show that, as in Drosophila, the mosquito homologue, AaE 75, consists of three overlapping transcription units with three mRNA isoforms, AaE75A, AaE7SB, and AaE75C, originating as a result of alternative splicing. All three A dB 75 isoforms are induced at the onset of vitellogenesis by a blood meal-activated hormonal cascade, and highly expressed in the mosquito ovary and fat body, suggesting their involvement in the regulation of oogenesis and vitellogenesis, respectively. In vitro fat body culture experiments demonstrate that AaE 75 isoforms are induced by 20- hydroxyecdysone, an active ecdysteroid in the mosquito, independently of protein synthesis. Furthermore, AaE75 is able to activate the Vg promoter in cell transfection assays, suggesting its role as an early vitellogenic gene. These findings suggest that related ecdysone-triggered regulatory hierarchies may be used reiteratively during developmental and reproductive ecdysone response. # Following the recommendation of Cherbas et al. (1984), ecdysone is used as the generic term for hormones with the appropriate biological activities. The active hormone in the Drosophila salivary gland hierarchies and mosquito vitellogenesis and oogenesis has been shown to be 20-hydroxyecdysone. 69 INTRODUCTION The insect steroid hormone ecdysone controls developmental processes including larval molting, metamorphosis and, in some insects, reproduction. In Drosophila, gene regulation by ecdysone is best understood in the late larval and prepupal salivary gland, where hierarchies of gene expression are reflected in the activities of polytene chromosome puffs. Extensive characterization of puffing pattems has revealed that ecdysone acts first to activate a small number of early ecdysone inducible genes, and that their products control subsequent progression of the genetic response to ecdysone (Ashbumer et al., 1974). Molecular characterization of this hierarchy has led to the identification of the ecdysone receptor as a heterodimer of the EcR (Ecdysone Receptor) and USP (Ultraspiracle) proteins, and has shown that the early genes are direct targets of the ecdysone-receptor complex (Cherbas et al., 1991; Koelle et al., 1991; Yao et al., 1992; Yao et al., 1993). Consistent with their proposed regulatory role, the early genes, BR-C, E74 and E 75, each encode transcription factors which have been implicated in the regulation of early and late gene expression (Burtis et al., 1990; Segraves and Hogness, 1990; Cherbas et al., 1991; Guay and Guild, 1991; Koelle et al., 1991; DiBello et al., 1992; Hill et al., 1992; Yao et al., 1992, 1993; Fletcher and Thummel, 1995). The expression of these genes in other tissues and at other stages of development is consistent with the idea that these genes are also involved in controlling spatially and temporally parallel regulatory hierarchies mediating ecdysone response in other tissues and stages (Segraves, 1988; Thummel et al., 1990; Huet et al., 1993). Among likely ecdysone- regulated processes in Drosophila, two of the least understood are the progression of oogenesis and vitellogenesis. The ecdysone-inducibility of yolk proteins (Bownes, 1986) 70 and the female sterility of mutations affecting ecdysone synthesis and ecdysone response are suggestive of a role for ecdysone in female reproduction, but little is known of the specific role of ecdysone in these processes (Garen et al., 1977; Oro et al., 1992). In contrast, the role of ecdysone in female reproduction is well established in anautogenous mosquitoes, especially the yellow fever mosquito Aedes aegypti. In this mosquito, initiation of egg maturation and vitellogenesis requires a blood meal, which stimulates neuroendocrine production of the ecdysiotropic neuropeptide EDNH (egg development neurosecretory hormone) or OEH (ovarian ecdysteroidogenic hormone) (Lea, 1967, 1972; Matsumoto et al., 1989). This hormone stimulates ovarian ecdysone production, which in turn controls the induction and progression of vitellogenesis and egg development. In A. aegypti, ecdysone levels are low during the first 8-10 hr post blood meal (PBM), with only a small peak at 4 hr. Thereafter, the ecdysone level increases dramatically to a maximum at 16-20 hr PBM, then rapidly declines to‘previtellogenic levels in preparation for the termination of vitellogenesis at 30-32h PBM (Hagedom, 1983, 1985; Hays and Raikhel, 1990; Raikhel, 1992). Aspects of the oogenic and vitellogenic responses can be induced by exogenous 20-hydroxyecdysone administration (Hagedom, 1983, 1985; Raikhel, 1992; Dhadialla and Raikhel, 1994), but the action of 20-hydroxyecdysone (20E) on the mosquito yolk protein genes, vitellogenin (Vg) and vitellogenic carboxypeptidase (V CP) requires protein synthesis (Deitsch et al., 1995a). This indicates that, as in the Drosophila salivary gland, ecdysone response occurs via a cascade of regulatory events. Other functions associated with ecdysone during mosquito egg maturation include the formation of vitelline envelope (Raikhel and Lea, 1991; Lin et al., 1993) and the separation of the secondary follicle in the ovary (Beckemeyer and Lea, 71 1980). Thus, the mosquito provides an ideal opportunity to investigate hierarchical control of ecdysone-induced gene expression during reproduction. Moreover, the critical role of anautogenous mosquitoes as disease vectors amplifies the importance of understanding this process, a potential target in the development of pest control strategies. Consistent with the proposed role of ecdysone in controlling oogenesis and vitellogenesis, EcR and USP transcripts can be detected in pre- and post-vitellogenic ovaries and fat bodies. Three size classes of transcripts for AaEcR, the A. aegypti homologue of Drosophila EcR, are expressed in pre- and post-vitellogenic ovaries and fat bodies (Cho et al., 1995). The other critical component, USP, is also expressed in these tissues with one predominating in the previtellogenic stage and the other in the vitellogenic stage (Kapitskaya et al., 1996). Here, the A. aegypti homologue of the Drosophila E 75 gene has been identified and characterized, a putative representative of the next level in the ecdysone response hierarchy. As one of the classical early genes, E 75 has been proposed to play a role in controlling ecdysone response in the late third instar salivary gland. E 75 transcripts are induced during all developmental ecdysone pulses, and genetic analysis suggests that E 75 is required for all critical developmental ecdysone responses, during embryogenesis, larval development and metamorphosis (W.A. Segraves, P. Jenik, C. F ichtenberg and C. Hughes, unpublished). It is shown here that A. aegypti E 75 is expressed in ovary and fat body following a blood meal, and that AaE 75 transcripts are ecdysone-inducible in isolated fat body in vitro. These findings suggest that AaE 75 plays an important role in mediating ovarian and fat body ecdysone response, and that ecdysone-triggered 72 regulatory hierarchies such as those implicated in the initiation of metamorphosis may be reiteratively utilized in the control of reproductive ecdysone response. MATERIALS AND METHODS Degenerate PCR/In verse PCR amplifications Several conserved regions with relatively low codon degeneracy, within the DNA and hormone binding domains of Manduca sexta (Segraves and Woldin, 1993) and D. melanogaster E 75 were used to design degenerate oligonucleotides for PCR amplification of mosquito E 75 sequences (Figs. 1 and 2). Oligonucleotides used for amplification of the 421 bp hormone binding domain fragment were E2F, GNCTNATHTGYATGTTYGAY, annealing to common region nucleotides 560-579 and E4R, CCACATYTGYTGNCKNAG, annealing to common region nucleotides 981-964 (Fig. 1). Each PCR amplification reaction contained ~lOO ng of genomic DNA, 100 ng of each primer, 250 mM each dNTP, 2 mM MgC12, 33.5 mM Tris pH 8.8, 16.6 mM ammonium sulfate, 10 mM b-mercaptoethanol, and 1.0 unit of Taq polymerase (Amersham) in 32 ul final reaction volume, and was conducted for 35 cycles of 94°C for 1 min, 46°C for l min, 72°C for 1.5 min. If no product was observed after the initial amplification, 1 ul of the initial reaction was diluted into fresh reaction mix and re- amplified for 20 to 35 cycles. Where possible, re-amplifications were performed using nested primer sets. For PCR amplifications using non-degenerate primers, annealing temperatures were increased to 50-52°C and the extension cycle was lengthened to 3 min. For mapping of genomic clones using PCR amplification, 10 ng of template DNA was used in each amplification reaction. 73 Co Ra lle ill“: Plt Inverse PCR templates were prepared by digestion with the appropriate restriction enzyme, followed by ethanol precipitation in the presence of ammonium acetate, dilution to a volume of 1 ml and self-ligation overnight at 18°C. The template DNA was then re- precipitated and 250 ng was used in each PCR reaction. Oligonucleotides used for the specific amplification of exon A0 sequences were GTCGAATTCDATRKNAGYTC, annealing to exons A0 and Al from nucleotide 8 of exon A1 to nucleotide 5 88 of exon A0, and 1N3, GTGTGTGCGGTGATAAAGC, annealing to nucleotides 28-46 of exon A1. cDNA and genomic libraries An A. aegypti cDNA library prepared from poly (A+) selected mRNA from fat body 0-48 hr post blood meal (PBM) (Cho et al., 1991) and an A. aegypti genomic library constructed from Sau3A-digested DNA cloned into the AF ix II vector (Deitsch and Raikhel, 1993) have been described previously. For size-selected genomic DNA libraries, fragments of the desired size were excised from low melting point agarose gels, digested with b-agarase (NEB), phenol/chloroform extracted, ethanol precipitated and ligated into EcoRl-digested phage arms of the replacement vector 1607 (Davis et al., 1980). Phage were packaged with Gigapak 11 XL packaging extract (Stratagene) and propagated on E. coli C600 hflA to select for recombinant phage, which are cI-. DNA sequencing Vitellogenic fat body cDNA clones in XZAPII were propagated in Bluescript KS- following in vivo excision of the recombinant Bluescript phagemid in the presence of a 74 helper filamentous phage (Stratagene). Sequence analysis of cDNA clones, genomic clones and PCR products was performed by thermal cycle sequencing using a modified Taq polymerase and chain terminating dideoxynucleotides (Perkin-Elmer). All sequence data obtained from PCR-derived templates was confirmed by comparison to unamplified genomic or cDNA sequence. Some preliminary and confirmatory sequence analysis was generated on the IE] automated sequencing system at the Keck Biotechnology Center (Yale University School of Medicine). Sequence data was assembled and analyzed using Geneworks sequence analysis software. Animals Mosquitoes (Aedes aegypti) were reared as described (Hays and Raikhel, 1990). Vitellogenesis was initiated in adult females 3-5 days after eclosion by allowing them to feed on rats. Northern blot analyses Total RNA was prepared from ovaries or fat bodies dissected from female mosquitoes 3-5 days old (previtellogenic), or at 6 hr intervals post blood meal (PBM), using the guanidine isothiocyanate method (Cho et al., 1991). For analysis of transcription events early after the onset of vitellogenesis, ovaries and fat bodies were also dissected l, 3, and 4 hr PBM. Northern blot analyses were performed with the same amounts of mosquito equivalents of total RNA (6-48 hr PBM ovaries) loaded on 1% formaldehyde-agarose gels. Isoforrn-specific probes (designated A625 for AaE75A, B660 for AaE75B and C350 or C796 for AaE75C) were derived from isoform-specific 75 exons. A625 contains all of the indicated exon A0 sequences. B660 extends from the EcoRI site (nt 337) to the PstI site (nt 1007) of exon B 1. C796 was obtained by amplification of exon CO sequences using oligonucleotides CF 1 and CR1 (Fig. 1). C350 is derived from an AaE75C cDNA clone, and extends from nucleotide 504 of exon C0 to the EcoRI site formed at the junction of exons CO and Al (Fig. 1). The probe common to all isoforms extends from an EcoRI site (nt 862) to the end of the indicated exon 4/5 sequences in the AaE7SB cDNA 2-3A6 (Fig. 1). All probes were used for hybridization under high stringency conditions. The following cDNA fragments were used as probes to control for stage- and tissue-specificity: mosquito vitellogenic carboxypeptidase (VCP), the fat body-specific vitellogenic gene expressed exclusively during vitellogenesis (Cho et al., 1991); the vitellogenin receptor (VgR), an ovary-specific gene expressed in the germ-line cells, i.e., the oocyte and nurse cells (Sappington et al., 1996); the vitelline envelope gene 15a-1, an ovary-specific gene expressed in the follicular epithelium (Lin et al., 1993). In addition, a cDNA probe for A edes aegpti cytoplasmic actin (Deitsch et al., 1995a,b) served as a control for a constitutively expressed gene. Developmental RT-PCR analyses The temporal changing profiles of the transcript abundance of the three E 75 isoforms were examined by using RT-PCR followed by Southern blotting with specific radioactive probes. Total RNA was prepared from fat body or ovaries throughout the first vitellogenic cycle as described above (Cho et al., 1991) with some modification in which all isopropanol precipitation steps were done without low-temperature incubation to avoid the co-precipitation of glycogen and salts. RNA preparations were determined 76 spectrophotometrically. A260/A280 and A260/A230 of RNA preparations were always above 1.7 and 2.0, respectively and quite consistent irrespective of developmental stages of the mosquitoes. Five microgram each of the RNA preparation was reverse-transcribed by SuperscriptII reverse transcriptase (Gibco-BRL) and random hexamer (Promega) in a reaction volume of 20 ul, diluted to 40 pl with TE buffer (10 mM Tris-Cl, pH 8.0, containing 1 mM EDTA) and stored at -20°C as a cDNA pool until use. In the case of mosquito tissues, 0.025 fat body- or ovary-equivalent cDNA pools ranging from 0.3 ul to 1.4 ul were used as PCR templates because of drastic changes in size of the tissue during vitellogenesis. PCR-amplification of three isoform-specific DNA fragments was done either simultaneously in a single tube with 25 ul of reaction mixture containing three isoform-specific sense primers (75A-F, 7SB-F, and 75C-F) and one common antisense primer (75com-R), or separately using only one set of primers. For the details of the primers, see below. Thermal cycling conditions were as follows: the reaction was incubated at 940C for 3 min; this was followed by 18 (for fat body samples) or 16 (for ovarian samples) cycles of 940C for 30 sec, 600C for 30 sec and 720C for 30 sec. As a reference of the development in the fat body and ovary, the same cDNA was subjected to the 10 cycles of RT-PCR followed by Southern blotting with a primer pair specific to the mosquito VCP (Cho et al., 1991), and vitellogenin receptor (Sappington et al., 1996), respectively. After PCR, 10 ul each of the reaction was fractionated 2.5 % agarose gel, transferred onto Hybond N+ membrane (Amersham) under alkaline conditions. Radioactive cDNA probe was prepared from 15 mg each of three PCR fragments generated by the primer pairs of 75A-F/75com-R (generating 517 bp fragment), 7SB-F/75com-R (394 bp) and 75C- 77 F/7SCom-R (280 bp), respectively, by random-priming labeling with [a32P]-dATP (NEN), and the membrane hybridized with the probe at 65°C in a solution of 5 x SSPE, 5 x Denhardt' solution, 200 ug/ml of denatured salmon sperm DNA (Sigma) following prehybridization. The membrane was washed twice at 65°C in 2 x SSPE for 15 min, once in 1 x SSPE for 15 min and finally twice in 0.1 x SSPE for 15 min, all containing 0.1% SDS, then autoradiographed. PCR/Southem blotting of the total RNA preparations without reverse transcription did not result in any appreciable signal on autoradiogram, indicating that the contamination of genomic DNA fragments into the total RNA fraction was virtually negligible (not shown). The radioactive blots were quantified with a phosphorimager (Molecular Dynamics). Primers used for the developmental RT-PCR: 75A-F, TAGTGCAATCAACGTATACCAATC; 7SB-F, CGTGGAAGAAGACCACGATCG; 75C-F, AACATAACCACGTGACCTCAATG; 75com-R, CAAGGGCGATACTGAATTTTCTG. In vitro fat body culture To examine the ecdysone inducibility of the mosquito AaE 75 homologue, the abdominal walls with adhering fat body (hereafter referred to as the fat body) were dissected from 3 to 5-day-old previtellogenic females and incubated in an organ culture system for 2 to 4 hr with increasing doses of 20E as previously described (Deitsch et al., l995a,b; Raikhel et al., 1997). Cycloheximide (Chx) (Calbiochem) was dissolved in water immediately before use (Deitsch et al., 1995). Total RNA was extracted as indicated above (Cho et al., 1991). As a control, RNA was extracted from fat bodies 78 incubated without the hormone or Chx. Northern blot or RT-PCR analyses were utilized as described above. Hybridization using a probe for the ecdysone-inducible VCP gene provided a positive control, while a cDNA probe for mosquito actin served as a control for a constitutively expressed gene (Deitsch et al., 1995a,b). Cell culture and transient transfection assay Cell culture and transient transfection assay were performed basically as previously described (Wang et al., 2000b). The pAc5-AaEcR and pAc5-AaUSPb constructs contained the full length AaEcR and AaUSPb (Wang et al., 2000b). The Vg- promoter-luc is the 2.1kb Vg promoter driving a luciferase reporter gene in the pGL3- basic vector. The full-length of AaE75A was subcloned into the pAc5 NotI and ApaI sites for transfection. RESULTS Cloning of an E 75 homologue from Aedes aegypti cDNA and genomic libraries To identify the A. aegypti homologue of the Drosophila E 75 early ecdysone inducible gene, a PCR-based strategy was utilized to take advantage of scattered highly conserved regions in the DNA-binding domain and the E domain of the D. melanogaster and Manduca sexta E75 proteins; the latter domain has strong similarity to the hormone binding domain of receptors in the nuclear hormone receptor superfamily (Segraves and Hogness, 1990; Segraves and Woldin, 1993). Several combinations of degenerate primers within the hormone-binding domain amplified genomic DNA fragments of the predicted size (see Materials and Methods). The largest of these, a 421 bp fragment, was cloned and sequenced. It encodes a pollypeptide with ~90% amino acid identity to the 79 corresponding region of Drosophila E75. Thus the gene from which these sequences had been amplified was tentatively named A. aegypti E 75 or AaE 75. Using the internal sequence of the 421-bp fragment, a second set of oligonucleotide primers was designed, directed away from one another, for use in inverse PCR reactions. DNA prepared from a fat body cDNA library was cleaved with EcoRI, recircularized and subjected to inverse PCR, yielding a 2.2 kb fragment. Direct sequence analysis indicated that this fragment contained sequences extending 5' into the D region which links the DNA and hormone binding domains and 3' through the carboxyl terminal domain to the 3' untranslated region. Further PCR analysis of the cDNA library, using degenerate primers for the DNA binding domain and primers derived from the sequence of the 2.2 kb fragment, indicated that the fat body cDNA library contained cDNAs extending at least into the DNA binding domain. To obtain vitellogenic fat body cDNA clones, the 2.2-kb inverse PCR fragment was used as a probe to screen approximately 2 x 10° recombinants. The cDNA clone extending farthest in the 5' direction, 2-3A6, was sequenced in its entirety. Analysis of this sequence indicated that this was a full length Aedes aegypti E 753 cDNA (Fig. 1, exon BI and common region). A second type of cDNA clone contained a region closely resembling the E 75 A1 exon, which encodes the first finger of the DNA binding domain, preceded by sequences closely resembling the E75C-specific sequences of Drosophila. However, this clone contained only 350 bp of E75C-specific sequence and appeared to be truncated at its 5' end. Three sequential rounds of inverse PCR "walking" gave rise to an additional 2 kb of AaE75C-specific genomic sequence. PCR of vitellogenic fat body cDNA using 80 mt. (’1 D TAWTACTCICICGCTCGGICMAGCTAGTGICGGTGIGCIMTIMUTGWTM(CWTWGUIWUWGWCWUCGCTMH 120 IQCLNKLTQDElDSlEI 1mmGTMTCGGTGATMCCMCWTMTUAWGCWGTMCCTWUGIWGTMUAWUGCCAGIGGKICMICAGGCGCCWGIWUTCC 240 RTFVIGDIQRIIYSPIIVOCKKIVKLEIASGLIQAPISSS ATGGTMTUMTGGCCMC1106WWI(WWWMTMWCUCWCGIGCWGIGGCGCUHCWW 360 IVTSIGHHQLGQDRARIQIIAKPSIKPCENVAPVQAQEVII CICATICGGUTWCWUKQIWWUIKMUKMQTWWCWCTTCCCATCGTCCGGT‘CIGICCICCTCICCMACICTMCGGT 4” VIIDGRFYDLQIISIIASPSHHIHILQEPSIIIPVI.SSSPISIC CI MTCATMCCMTGCCGGCACT AGI GMTWACCGGT MICCTCCMGCCWGCCCCCACT GCACGICCI CT CT CGCCMCACAATCATAGT 601m (CT ((601 EU LIITNAGSSIIDTGIPPSIQPPPINHPVSTITIIVOAPPII 7501‘ —r TCGWHCWWWKCCCGCUCCWWGCTMIUICWWIWTWIWGGGWIMEUCUWUWG 720 SAVSSSK011LPPPPATIIIVQQQHQHQHQQQQQHINVTSI r——nr 06CUCGCGCCKCCCUCWMKCWAUATWGGTMTWGCC IGTGGCWMWWIWGCGTCCMTICWGCCCWCIAG M1 QPPPPPPPVCAGSIGGIGGAGIIAKIKSAHEEPSSSIIPDI. runnnwmttummrmnnwmcnummurmmmmmmmmnmnuncm 120 HtSDISYQQSVIKEITIIVVQVQKCSQSQI ISA-f CWTMMCWWWWGGTCCCWKGCGWWCWICWCGGCCGCWWAGIWTCMCGTAMCA 240 PVISYIAETGEPCIPISTTV‘RIEEVIPPKEFIVVQSIYT § ATGICCTMMTTATWATWIATTGTWWCICCACCWCTAIGCTGGCTGG‘GITIAICIACCMCTUTUTCTCGCWGMCGC 360 llllPKI'i‘lSRKNIVLESSPPHKPILAGDYIPAHHlHQQQQI mmutmmummmumammtmmumm ((GGIATIMTCCMCGTCTTCTCC“(16601010111010 480 QlAlllll0FSKGPVLLVPKTEDCVIYSHPTPSSPTGHHNIF (“T“CI'ICICCMCTCWTGCKGCUTUGGCICCTCCTGTGGATAIMCCCCGGIGTIAIWTMCAGCGIIGCQWUMWMCWCICMWAG 6“ SVASPAPIHHHQAAPVDIKPPVIIDISVASSSKGEPDLNI MAI MITCGACGGMCCAU.‘GIACIWCGTCICIGC‘CIWMMC‘ITCCGGI'ITCGCIKGGTCTGCKTCCIGCWGGCTGCMG 89 FDGT'TVL(RVCCDKASGFNYGVHSCEGCK Fig. 1. Nucleotide and amino acid sequences of AaE75. Sequences for the transcription unit-specific exons, C0, A0, A1 and Bl, and for the common region, shared by all three AaE75 transcription units, are shown. Oligonucleotides used for RT-PCR analysis and other probe generation are indicated above the nucleotide sequence. Introns separating common region exons 2, 3 and 4/5 are indicated by vertical arrows. 81 0101131 CMTTCGGCACCACATCCT CCCCCCT C61 C61 MACCTT CT (CT CGCCCT CMCCMACCCCCT (GT CGCCCITT MGCT CTAGCT ACT GCGT CGT CAGT GCCT CT CAGCCACATATACC TCMCAAMMATMCCMCATCCMTCCMGGATCTCTGCCAMGTGTCCAMCTCCGTATCMTCMGCGGCMGTCATCCCCCAGCMTACACACAGCTGTAUACTTATCACCMG TGCMTACATCGGGACT GMGMCGMGGCCGGCACTI CACAGCT ACMMMGACTT CAAGT CCATTCATCCMGT ACT CCAACAGT CCMCI CT CMTTCCCCT CAAMTCCCATGT C 11 G C CAGTGCACCACGAGCCCGCCACIAACCAGMATCCICGGCCGCGGCGCICAACACTCACMMCTCIGMAG'ITCCMGCAGTCACTCTAGTITMACMTACTMTCATACCACCMCA AVQQEPASIEKSSAAALKTEKSLKVPSSQCSLK111110115! GTACCCCIAGCACCAGCACCMCACCCTTMCMGGCCCACAGIGCACIAGITAAMTTCTTCMTCCGCTCCCCTGMTACTTCMATTTCATCACACTCTACMCGCMCTTCCGCTG SSCSSSSASLKKAHSAIVKIlESAPlNSSItlTLYNGSSC GCACT CGTTCIACT ACCACT ACT MTAGT ACT ACT ACT CMAGT MICMGT ACT ACCT CCT MGGAGGAMCCMMTCACI GT GIT AGCAAGT AGIT CCAGT CAMCCCT CCMCATC CSGSSSSSNSSSSQSNQVVAPKEETKISVLASSSSQTVED m-r ‘0 CTCACGCCCGCMATICAMACMCCCCCMCATCGATTTCIGCCCGTGCMCMCACCACCATCCCCAMCAGTCCATTAGTG CATGCCAACCGGGCTCGTCTCCCTCAGTAGATGATA PNACKLKTTPKIOFCPIKKITIAKEIISACERGSSASVDD ACMTCMCCT CACCAGCACCMCMCMCCCGT CACCCAGT 6T GCCAIACT ACACCMCACT CCCACAMCACCCAACCGCACCATCCCGGAGI CGCCACT CCT C GCACACTT CCCATC KIQPQQQQEECVSQCAIlOEQSHKQPTCCCRSRESSQSSD ACT CGT CCCT GT CCGCATCT CCCCAGCCCACCACCCATCACACCT GCT GCAGCMTACTT GCAGTT CCAAIGGMCCAGT AGCCACI CCICTTCC CACTATCI CATCAAIGATCT CT GCA 115$LSASPCPTSODSCCSNSCSSNCSSSDCCSDYLINDLC mcmwcmrrcwmm 1112 KQFEEILCEOH cm region ‘ CGTTT CTT CACGCGCT CCATCCAGCACAMATICACT ATCGCCCTT CT ACT MAMCCAGCAGT CT ACT ATACT CCGMTAMCCCTMTCGCT GCCAGT ATTCT GAMMA CFFRR SIIQQK QYRPCTKAQQC lRIIRNR CQYC°IGIGCTKKTCc I ATCCCCGTTGGAATGACI’CGCGACGCGGICACATTCGGTCGTGTGCCGMCCGCCMAAGGCCCGCATCCTCGCGCCCATGCAGCAAACCACCCAGM IA VGISADAVRFGRVPKAEKARI lAAIQQSTquccmgwccamccum GCCACCCAGCTCCACCATCAGCCCCCTCTCCTCGCGCCCCTCCTCCCACCCCACATGCACACGTGTCAGTTCACGCGTCAGMGGTCACTTCCATGCGGCMCCT ATE DQPIII.LAAVLllAllltlllCEliTllEKVT$11thR°ATCACGCCTCCCIST I ACCIACTCCATCCCTACTTTCCCATCTCCACTCMCCCCCCCCCACACCIACAATEACACCAGCAGTTCAGCCAACGCTTCCCCCAIGTMTTCGTGGCGICATTCACTTT YSSIP'I'I.ACP1.IIPAPELQEQEl’lellFAlilllIlGVIDliGAmgnAG ATCCCCGCCTTCCACATGCTCACCCAEGACGATMGITCACCCICCTCMCCCCGGCCTGTTCCATGCGCTCTTCCIGCGTCTGATTTGCATCTTCGACTCGCACATAMCICCATCATC FlQI TDQD HLLKAGLFDALFVRLICIFDSQIN II TGCCTGAATCCCCAGGTCATCCCCCGCCATACCATTCACMCCCTGCCMCGCACGTTTCCTGCTCGATICGACGTTCAACTTTGCCCMCCMTCMCTCGATCMTCTGACCCACCCC C ICQVIIRRDTIQACAIARFLVDSTFIFAER1111 SAN LT DA 240 1080 240 720 WATCGCTCT CIT CT CT CCCATTCT CTT GATCACGCCCGACCCACCGCCACT GCGCMTCT GCAGIT CATCCMCCCATCT ACT CCMCCT CMCGCTT GCCT CCACT CGATCCT ATCC m EIC1FCAIVlITPORPCLRNVELIERIYSKLKAClQSIVS Fig. 1 (cont’d). 82 CMMCCCGCCCHTMCCCACMTTCATCCACCMCTGCTCCGMCACTACCGGATCTGCGTACGCTCAGCACCTTGCACACGGAGMGCTCGTAGTGTTCCCMCGCAGCATAMGAA QIAPDKPEFIQELLRTLPOLRTLSTLHIEKLVVFRTEHKE CT CCT ACCCCACCACATCT CCMCCCT CMCMCATCT CGCAAACACT CCCACCT CCMCACCT GCACCT CCCACGGCMTMTGT CCAGCATGT AGCCMCAGI CCCATGAGCT C661 A I.RQIIAIIAEEDLAKSPSSITISCDGINVEDVAKSPASSV TCCAGT ACT CAST CCGCCGMACMCCT CCCACI ACT CGCATATTCCTT CCT CCTT GACCGCTT CGCCGCCT CI GCT GGCCCCT ACT CT GI CCGGT CMTGCCCT ATCCGC CATCGGCCA SSIESAETTSDYSNIPSSLSASAPLLAATLSGQCPIRHRA AGCACCGGTTCGTCCGCACAACATCACATCATTCGTGGIACSGCCCATCTCGCACAGAACCCATTGACCATCACACCCCITATCCCATCGGTTGGMCACATCACGTCCGGTACCGGAM SSCSSAEDDIICGTAHLAQNCLTITPVIRSVGTHHVRYRK CTACACTCACCCACCGATTCC CGTATTGMTCTGGMACCMAMCACGAC CACAMCCAGTCAGCAGTGGATCGTCIICTTGCTCMGTCCACCATCTTCCCTACACCAICAATCCGAC lOSPTDSCIESGIEKHDHKPVSSCSSSCSSPRSSLEDQSD CACMCCCTCACATCGTGCMMCAICCCCCICCTCAACCCGGTCTTGCAGGCACCTCCCCTGTACCATACCAACACCCTGATGCACGMCCCTACMACCCCACAACAMTTCCGTGCA DKRNIVEAHPVLKRVLQAPPLYDTNSLIDEAYKPHKKFRA ATGCCT CACACCCAMCTCMGCCCAGCCT CCT CCMGTT CCT (CT (CT CT ACAATCACT CT CT CATCACATTCGC CCMACCATCCT CCAGCT CTT CCCCATCTT CCCTGGT ATCGT CA 11 SEA EPAPS SSSSTITCSSASPKP SSSSSPSSVVSS CACACCACCACTATGCCTCMTCACCTCCCCACCMCCCCTTTCCCACCITCATATCCATTTMCCCCACCGTI’CMTCACTCACCACACCMACCCTTCACCACCACCATCATCAICAC 11555116 CQSPPQQPLSQCHIHLTRPLAQSPHQSLHllllllllllll (ACT CGGGMCT CCT CCCGCT AGCCATCATCAGI CCT CTT CCCT ATCCAGCACCCATTCCGT GIT CGCCMATCCCT CATGCAGCAACCCACGATCACT CCGCAACAGATCMACCTT CC HSGTPPAS llSllQS $lSSTll SVLAKS EQPTATPEQIKRS CACATCATCCACAACTACATCATCCCACAGTCCCACCMTCTCACCACCATCCCCMGCGCACAGTTCGTACCGCAGTGGTCTCCTACTTTCCAGCCCACCATCCTCATCGGTCTCGCCA DIINNYIIRESQQSQHNPCAQSSYRSCLLVCSPRSSSVSP CT GACCT CCACCAGT AGCACT ACCAGT MCT CATCCT CCT CCGGCAGTGCTI GT CCATTCT CACCCMTAGCAGCAGCAGCAGCI CCCACACI GCACCCACMCCGI'T ATCACCACCACC lTSSSSSSSISSSSCSCCPFSANSSSSRIQSGPTSVITTT CCCCACCAMCT CCTT CT CCATCCCT CMTTCCACCCGMGT GCT CATACTT CCT CCI CTT CCAGT ACT GCCAGCACCAGT ACT ACCATCCCCT ACTTCCAGT CACCACATTCCACATCG RQQTPSPSLISTCSADTSSSSSTASSSTSIIYFQSPIISTS ATCT CCCCACCGCT GGCCCT CATCCCTCMCMGCCACCAGCACMCCACCGCATCCATTGCCT CCCCI (ACT (CST 6111’ CACGGCACCCT CTCCCT CC CC'I CCCCT CATCCACCTT CM ISPPVAVIAQQASSTTTCSICSPQSVVTAPSPSPRLIElQ CIGCACATCGCTCCCGCTTCCACTTCCTCCCGCACCACCCATCCCCCCCTCMCCTCTCCAMMAICCCCCTCCCCTTCCCCACCICCCCACCMTCMCCTCCTCCTCCTCCACAGCC VDIACGSTSSGTTDAPLNLSKKSPSPSPARHQSISSSSIA ACCATCCATAAGGT CCT ACT CfigAfiCGT MGCCACCCCCACCACCATTACMCMCMCMCMCT ACT ACT ACT ACT MGT CATCGACAAAMTCMCI ACT ACT AMCAMACAAGCC T I 11 K V I. 1. A CITTMACACGCAGATCAGTTTACACMCTGTACAMCTITMCATAGTMCACTAGTCACTATTGCTACGCGCTACCCATTTACMTTTTATAMTCACAMCCCCACCCCCCCCCCAC GCICCCCICACGCCCACTCCTCCCGAATTC 2790 Fig. 1 (cont’d). 83 ton 1.200 1320 1440 1920 2160 2640 2760 oligonucleotides based on this sequence indicated that the cDNA library contained sequences extending nearly to the start of the AaE75C open reading frame, but screening of an additional 2 X 10° cDNAs did not reveal any full length E75C cDNAs. Therefore inverse PCR on EcoRI-digested recircularized templates was utilized to isolate the C- specific region from the rare or truncated cDNA or cDNAs responsible for this signal. At the 3' end of the isolated sequences, the presence of an EcoRI site derived from the exon CO-exon Al splice (see Fig. l) confirms that these sequences derive from spliced RNA and not from genomic DNA. At the 5' end, sequence analysis indicates that these sequences extend to within 30 bp of a putative initiator Met closely preceded by an in- frame stop codon (Fig. 1). RT-PCR analysis indicates, however, that the 5' end of AaE75C likely lies significantly upstream of this region (data not shown). No AaE75A-specific cDNAs were identified in the screens described above. In order to isolate the AaE75A-specific coding region from the fat body library, primer sets were designed to amplify specifically AaE75A sequences. First, primers annealing within the A1 exon were used for an initial round of inverse PCR amplification, then a predicted exon AO-specific degenerate primer was used to re-amplify these products (see Materials and Methods). This yielded 183 bp of sequence closely matching predicted AaE75A-specific sequences. Primers derived from this sequence were used in a second inverse PCR reaction on recircularized EcoRI-digested fat body cDNA library material, giving rise to a fragment extending beyond the 5' end of the AaE75A-specific ORF (Fig. l). 84 Defining the Aedes aegypti E 75 genomic structure To elucidate the genomic structure of the AaE 75 gene, the cDNA fragments described above were used as probes to identify genomic clones from size-selected and complete A. aegvpti genomic libraries, yielding a collection of overlapping genomic clones within each region. Exons were localized by restriction mapping, hybridization and PCR using exon-specific and vector primers (Fig. 2). The intron between exons A0 and A1 is approximately 15.6 kb in length and the intron between the AaE7SB-specific region and exon 2 is 150-200 bp in length. The failure of clones A8A and 810 to cross-hybridize indicated that there are additional intron sequences separating these two clones. Genomic clones A8A and 810 were attempted to use to define the length of this intron through genomic walking. Three rounds of chromosomal walking from the A8A side yielded an additional 20+ kb of genomic DNA before the presence of repetitive sequences prevented further walking. Screening with a 5' terminal fragment of clone B 10 yielded no additional clones, indicating a potential gap in the genomic library. While the complete length of the intron between the A1 and B1 exons thus cannot be determined, these findings suggest that it is in excess of 30 kb. Within the common region, restriction mapping, hybridization, PCR and direct sequence analysis showed that there is no intron separating the hormone binding domain and the carboxyl terminal F domain, but that there is an intron of approximately 3.8 kb separating exon 3 from exon 4/5. To determine the length of the intron separating exon 2 from exon 3, a 5' terminal fragment of clone E7 was used to isolate a clone, L13, which also hybridized with the 810 clone. Further restriction mapping and Southern blot 85 co A0 A1 312 345 0111575 0 0111575 A DmElS B co ‘° “ 312 3 us me “7% “a“ l—‘—“-‘-* A8 F- J. 810 : . . :57; in U, C13 L13 Hull—1 5: l I I l l J 1 . g I 13. 10k!) Fig. 2. Genomic organization of AaE75. The intron-exon structure and partial restriction map of genomic clones are shown. The DNA binding domain is encoded within exon Al and exon 2. The region of the steroid receptor hormone binding domain is located within the first ~200 amino acids of exon 4/5. The intron-exon map of D. melanogaster E 75 (DmE 75) is shown for comparison. EcoRI sites are indicated on the genomic clones. lsoform-specific domains AaE75A shared common region domains AaE75C AaE7SB Fig. 3. Comparison of Aedes aegypti and D. melanogaster E75 amino acid sequences. Schematic representation of sequence identity within structural domains of the A. aegypti E75 A, E75B, and E75C-specific regions as well as the DNA binding domain (C), linking region (D), hormone binding domain (E), and repressor region (F). Overall percentage of amino acid identity between A. aegypti and D. melanogaster E75 domains are presented below each region. Sub- regions showing high levels of identity within less conserved domains are indicated as dark stippled bars. 86 analysis allowed alignment of these clones and showed that the intron separating exons 2 and 3 is approximately 17 kb in length (Fig. 2). Evolutionary conservation of the dipteran E 75 proteins The predicted amino acid sequences of the AaE75A, AaE7SB and AaE75C proteins are shown above the corresponding nucleic acid sequences (Fig. 1). Fig. 3 schematically depicts the comparison between these sequences and those of E75 A, B and C from D. melanogaster. The DNA binding, linking, and hormone binding regions display high levels of identity with Drosophila E75: 100%, 91% and 93%, respectively, at the amino acid level. The terminal domains are substantially more substantially divergent, but show a similar organization and contain several highly conserved subregions. More significantly, the amino terminal domains specific to the AaE75A, AaE7SB and AaE75C isoforms are clearly conserved with each showing ~30% identity to the amino terminal domain of the corresponding Drosophila protein. AaE 75 transcript analysis The transcript size of the AaE 75 gene was examined by the Northern blot analysis of vitellogenic tissues of female mosquitoes during the first cycle of vitellogenesis. Mosquito ovarian RNA 24-30 hours PBM was hybridized with isoform-specific probes (A625 for AaE75A, B660 for AaE75B and C350 or C796 for AaE75C; see Materials and Methods), revealing two transcripts per isoform (Fig. 4): 6.2 and 5, 6.9 and 5.6, and 6.8 and 5.4 kb, for isoforms A, B, and C, respectively. When a probe to the region common to all three AaE 75 isoforms was utilized, 5 bands were observed (Fig. 4). These findings 87 Probe A B C Com ‘L‘A “on .3. 9.5—— 7'5 _6_2 -6.9 —6.8 ‘3' 4.4— l --5.0 —5-5 —5.4 - 2.4— 1.4— Fig. 4. AaE 75 transcripts in the mosquito ovary. Total RNA was extracted from ovaries dissected from vitellogenic mosquitoes 24-30 hr after a blood meal. Northern blots were hybridized with AaE75 isoform-specific probes. (Lane A, AaE75A-specific probe; Lane B, AaE7SB-specific probe; Lane C, AaE75C-specific probe; Com, common region AaE75 probe). A 1" ( u _ 'l— '1; 9 5 " ‘ _— - t 7.5— ; i _7.2 . -. _-, ’ —6.1 4.4—— y; .. ,3 2.4—— " 1.4— ;. Fig. 5. AaE75 transcripts in the mosquito fat body. Total RNA was extracted from fat bodies dissected from vitellogenic mosquitoes 24-30 hr after a blood meal. Northern blots were hybridized with AaE75 isoform-specific probes. (Lane A, AaE75A-specifrc probe; Lane B, AaE75B-specific probe; Lane C, AaE75C-specific probe). 88 are consistent with the presence of several different 3' ends in the cDNA clones analyzed (data not shown), suggesting that these RNA size-classes arise from utilization of alternative polyadenylation signals, as in the case of Drosophila E 75 (Segraves and Hogness, 1990). In the fat body, transcript size was also established using Northern blot hybridization of RNA from the vitellogenic fat bodies 24-30 hr PBM with isoform- specific AaE 75 probes. As in the ovary, two transcripts are clearly observed for the AaE75A and AaE75C isoforms. Similarly, it is likely that two transcripts existed for AaE75B (Fig. 5). Overall, the level of E 75 RNA in the fat body was considerably lower than in the ovary, making the Northern blot analysis a relatively undesirable alternative for further analysis in this tissue. Time course of AaE 75 expression in the fat body and ovary during vitellogen esis To determine the temporal expression pattern of AaE 75 mRN A levels in the vitellogenic tissues, the fat body and ovary, RT-PCR analysis was primarily used. Complementary DNA was reverse transcribed from fat body total RNA and then amplified in PCR reactions using isoform-specific primers (see Fig. 1 and Materials and Methods). In both vitellogenic tissues, AaE 75 expression was induced at the onset of vitellogenesis by a blood meal-activated hormonal cascade, and AaE 75 isoforms were highly expressed in the mosquito ovary and fat body, suggesting an involvement in the regulation of oogenesis and vitellogenesis, respectively. In the fat body (Figs. 6a and 6b), E 75 transcripts exhibit a sharp rise immediately after the onset of vitellogenesis reaching 89 (a). Days after Eclosion Hours after Blood Meal o-11-23-sl1 3 4 6 15 24 3648 E75A—\ ..... a.-- E7SB— "' --.8 ~a-“e- . 517 bp 394 bp 137st F280 bp VCP ]—456 bp (b). Em 3 E“ E PV in 30 411 611 15h 24h 36h 4811 Time After a Blood Meal Fig. 6. Accumulation of AaE75 mRNA in the mosquito fat body. This experiment was performed by Southern blotted RT-PCR (see Materials and Methods). The results are expressed normalized units of the highest readings per fat body and represent means of three determinations :t SE. Shown also are the results of a RT-PCR with VCP, which serves to illustrate the course of vitellogenesis in the fat body. A. a representative Southern blot of the RT-PCR; B. quantitative analysis of the data. 90 a peak at 3-4 hr PBM when ecdysone titer shows a first small peak (Hagedom et al., 1975). After the initial drop, levels of AaE75A and AaE7SB transcripts gradually increased again reaching a second peak at the 24 hr PBM when ecdysone levels just have passed their major peak, at 18-20 hr PBM (Hagedom et al., 1975). The second gradual increase in AaE 75 transcripts closely correlated with the accumulation of RNA for YPPs in the fat body as demonstrated by the VCP transcript kinetics. The second peak of isoforms A and C transcripts was not as pronounced as that of isoform B. After the second peak, the amounts of A01? 75 transcripts decreased to very low levels at the end of vitellogenesis, 42 and 48 h PBM (Figs. 6a and 6b). The temporal expression of the AaE 75 gene in the ovary was first examined by Northern blot analyses utilizing total ovarian RNA for time points spanning the first vitellogenic cycle from 0 to 48 hr PBM. For each time point, the same number of ovary- equivalents of RNA was loaded, and blots were probed with the three isoform-specific A aE 75 probes. To provide a reference with follicle development, two ovary-specific probes were utilized: vitellogenin receptor VgR cDNA, (Sappington et al., 1996), which is specific to the germ-line cells (i.e., the oocyte and nurse cells); and the VE gene 15a-1 (Lin et al., 1993), which is specifically expressed in the follicular epithelium surrounding the germ cells in the mosquito follicle. These Northern blots showed that transcripts for all three A015 75 isoforms exhibited similar patterns of accumulation and disappearance in the mosquito ovary (Fig. 7). The AaE 75 transcripts accumulated slowly during the first 10-12 hr PBM; however AaE75A and Aa7SB transcripts exhibited dramatic accumulation during the next 12 hr, reaching a peak by 24 hr PBM. The amount of these AaE 75 transcripts declined between 24 and 36 hr PBM, the time coinciding with the 91 hours PBM E758 E75C V9 Receptor . 6 , Envelope Fig. 7. Expression of AaE75 RNA isoforms in the mosquito ovary during the first vitellogenic cycle. Total RNA was extracted from the same amount of ovary equivalents dissected from previtellogenic mosquitoes 3-5 days after eclosion and from vitellogenic mosquitoes at the indicated hours after a blood meal. Northern blots were hybridized with AaE75 isoform-specific probes (see Materials and Methods), and with Vitellogenin Receptor (VgR) and Vitelline Envelope (VE) cDNA clones. Developmental Profile of E75 in Mosquito Ovary 140 I ““1 12 l +E7SA 120]l /§\I l-l-E753 10 a: 76100 4 ' \ ‘\ l— ‘5750 9 - 380i / / J_\\\L__Y_9_E_6 EE’ “'3 sol / / ~- % < LU l 4 g 2 401; l o CE 2 20 4 “L 2 o l- 0 PV 6h 12h 18h 24h 30h 36h 42h 48h Tlme after a Blood Meal Fig. 8. Accumulation of AaE75 mRNA in the mosquito ovaries, quantified by phosphorimager analysis of Southern blotted RT-PCR products (see Materials and Methods). The results represent means of three determinations i S.E. Shown also are the results of a RT-PCR with VgR, which serves to illustrate the course of ovarian development. 92 termination of vitellogenesis and cessation of yolk accumulation by the ovary, and was followed by a more gradual decline over the next 12 hr, reaching low levels by 48 hr PBM. The AaE75C showed only a modest increase with a small peak at 24 hr PBM (Fig. 7). The onset of high AaE 75 expression in the vitellogenic ovary closely paralleled that of the ecdysone-inducible VE genes. However, the VE gene transcripts remain in the ovary at peak levels well beyond the peak of AaE 75 expression (Fig.7). The overall expression profiles of AaE 75 transcripts resembled even more closely the vitellogenic profile of VgR expression (Sappington et al., 1996), which increased to a peak at 24 hr PBM, and then declined during the latter portion of vitellogenesis (Fig. 7). In contrast to AaE 75, however, VgR was also expressed at a low level in previtellogenic ovaries (not shown). The quantitative analysis of the temporal expression of AaE 75 mRNA levels in the vitellogenic ovary, performed using the RT-PCR analysis as described for the fat body, is in agreement with the Northern blot analysis (Fig. 8). Response of AaE 75 to ecdysone in vitro The pattern of AaE 75 transcript kinetics in the mosquito ovary and fat body following a blood meal suggests that ecdysone produced in response to a blood meal is likely responsible for the induction of the AaE 75 gene. To determine whether AaE 75 induction following a blood meal is ecdysone-mediated, we have tested the in vitro effect of 20-hydroxyecdysone on the appearance of AaE 75 mRNA in isolated previtellogenic female fat body, under conditions previously demonstrated in vitro induction of the VCP gene (Deitsch et al., l995a) (Fig. 9). As it has been previously established (Deitsch et al., 1995a), the levels of the V CP mRNA increased in response to increasing concentration of 93 C.) 01 800 700 CI E75A - so 7; a BE758 3 E 600 BE75C . 25% g 500 IActin % m .ch 20 > ."2 on 1.1.1 400 c 'g 15 '8 .5 300 < ‘6 1: E 10 .315 g 200 E 100 . 5 z 0 _-_.=n J—Eh: 49m , 0 PV 0 10‘ 10‘ 208 (M) Fig. 9. AaE75 response to 20-hydroxyecdysone in in vitro fat body organ culture. The transcriptional response of the mosquito E75 homologue to 20-hydroxyecdysone was examined in an in vitro culture system of female previtellogenic fat body. Total RNA also was extracted from fat bodies dissected from previtellogenic (PV) mosquitoes to provide a control for the in vitro system. RT-PCR and Southern hybridization were performed with isoform-specific probes for AaE75A, AaE7SB, or AaE75C (see Materials and Methods, and Fig. 6). RT-PCR for VCP provided a positive control, while Actin was used as control for loading. 700 35 __ 600 DE75A 30 2 135758 .5 e: 0 _l 500 . SE75C . 25 < g IActin 06 '6 3‘, 400 Ich 20 8 e): in > -‘ '0 300 15 g ‘2‘ £3 .5 a: '5 3 g 200 10 E o O _ . z - - + + 20 E (10‘ M) - + — + CHX(10'°M) Fig. 10. Effect of cycloheximide (Chx) on the activation of the AaE 75 gene. The previtellogenic fat bodies were first pre-incubated in the culture medium for 1 hr in the absence or presence of 10'5 M Chx, and then continued incubating with the same dose of Chx but with the addition of 10'6 M 20E for 2 hr. RT-PCR and Southern hybridization were performed with isoform-specific probes for AaE75A, AaE75B, or AaE75C (see Materials and Methods, and Fig. 6). 94 3w a 600 . 3 500 . .. 400 ‘ E 300 < O z 200 1 100 o . - PV 1 2 4 a (a) Culture Time (h) 1200 +E7sA '°°° ‘ +5758 + E75C -_______l-"‘W o 4 -—-—'"—""—' ‘ ' . e: . 4L . :1 PV 1 2 4 6 (b) Culture Tlme (h) Fig. 11. Time course of AME 75 induction in in vitro fat body organ culture. Total RNA was extracted from previtellogenic (PV) fat bodies or fat bodies incubated in the presence of ZO-hydroxyecdysone for the indicated times. RT-PCR and southern blotting were performed with isoform-specific probes for AaE75A, AaE7SB or AaE75C (see Section 2, Fig. 6). VCP was included as a positive control. The results represent the means of three independent experiments +SE: (a). RNA accumulation of E75A, E7SB, E75C and VCP during exposure to 10 '7 M ZO-hydroxyecdysone; (b) RNA accumulation of E75A, E753, E75C and VCP during exposure to 10 '7 M ZO-hydroxyecdysone and 10 '5 M cycloheximide 95 20-hydroxyecdysone, reaching maximum at 10’6 to 10'5 M of the hormone in the medium (not shown). The levels of actin mRNA remained relatively constant. The mRNA levels of all three AaE 75 isoforms increased in a dose-dependent manner in response to 20- hydoxyecdysone. The levels of AaE75A and AaE7SB were elevated significantly, while only small increase was observed for AaE75C mRNA. Importantly, the AaE 75 transcripts reached their maximal levels at the concentration of 20-hydroxyecdysone in the culture medium at least 10-fold lower (10'7 M) than that for the VCP and Vg genes (Fig. 9). To determine the requirement of protein synthesis for activation of the E 75 early gene in the mosquito fat body by 20E, the previtellogenic fat bodies were incubated with cycloheximide (Chx), a protein synthesis inhibitor which reversibly represses protein synthesis in this tissue in a dose-dependent manner (Deitsch et al., l995a). The fat bodies were first pre-incubated in the culture medium for 1 hr in the absence or presence of 10'5 M Chx, and then continued incubating with the same dose of Chx but with the addition of 10'6 M 20B for 2 hr. Transcripts for all three AaE 75 isoforms appeared in response to 2013 in the presence of Chx, in contrast to the V CP gene, which was inhibited by Chx (Fig 10). Incubation of the non-stimulated, previtellogenic fat bodies in the presence of Chx alone activated neither the AaE 75 gene nor VCP (not shown). The time course of response also differentiated between AaE75 and VCP. AaE75 transcripts are induced rapidly and reach a peak by 2—4 h before beginning to regress. In contrast, VCP transcripts continue to accumulate to 6 h and beyond in response to hormonal stimulation. When the accumulation of early gene protein products is blocked by cycloheximide, both 96 the markcq‘ blocked (F A (‘I_ On been idem: unpublishe promoter, ( AaETSA w containing activated 11 increased i (Fig-l2). 1 30E USlnt:y Ne the largct EXPTCSSjO With the ‘ Signll‘ica, EcR,-US} amouM ( present \ the marked regression of early gene expression and the increase in VCP expression are blocked (Fig. 1 1). Activation of the Vg promoter by E 75 in 52 cells On the Vg promoter, a number of potential transcription factor binding sites have been identified including E75 and EcR/U SP (Martin et al., 2001; Martin and Raikhel, unpublished). To examine whether E75 is capable of inducing activation of the Vg promoter, cell transfection assays were performed. The expression plasmid carrying AaE75A was transfected into Drosophila 82 cells along with the reporter plasmid containing the Vg-promoter-luciferase. Either in the presence or absence of 20E, E75A activated the reporter gene to more than two fold (Fig. 12, lOOng), and the activation increased in a dose-dependent manner in response to increasing amounts of E75A (F ig.12). Interestingly, higher levels of activation were detected without 20E than with 20E using this 2.1kb Vg promoter construct (Fig. 12, -ZOE and +20E). Next, to investigate whether E75 affects the EcR/USP-induced transactivation of the target Vg gene, which has been reported previously (Martin et al., 2001), the expression plasmids carrying EcR/U SP and E75A were co-transfected into SZ cells along with the Vg-luciferase plasmid. In the presence of 20E, the reporter gene activities were significantly enhanced when both EcR/USP and E75A were present, compared with EcR/U SP or E75A alone (Fig. 12). This synergistic activation was dependant on the amount of E75A, exhibiting stronger activation when higher amounts of E75A were present with constant amount of EcR/USP (l lng) (Fig. 12). 97 200 O 5 4| 0 0 1 33:3. .03.. 0 5 O 0.4 1.2 3.7 1 1 33 100 E75A amount (ng) 20E F’ Fig.12. Action of E75 and EcR/U SP on the Vg Promoter. Increasing amounts of the expression plasmid for AaE75A were cotransfected with the 2.1kb-Vg promoter-luciferase construct and a constant amount of AaEcR/AaUSP plasmids (+EcRUSP) into Drosophila 82 cells in the presence (+20E) or absence of 20E (-20E). Reporter gene activities were measured after 1 day culture (see Materials and Methods). Vg Promoter 98 vitellocm Tl molecular‘ control of 1992 t. L': 09mm hi identificc transcri p These results indicate that the early gene E 75 may be involved in the activation of the Vg gene and be part of the ecdysone gene regulatory hierarchy during mosquito vitellogenesis. DISCUSSION The yellow fever mosquito Aedes aegypti is an ideal system for investigating the molecular basis of ecdysone action in insect reproduction, because of the stringent control of egg maturation and vitellogenic events triggered by a blood meal (Raikhel, 1992). Using PCR-based strategies to isolate genomic and cDNA clones, the Aedes aegypti homologue of the Drosophila early ecdysone-inducible gene E75 has been identified. Its genomic organization, including the structure of the three overlapping transcription units AaE75A, AaE7SB, and AaE75C, is highly conserved, with the exon- intron structure of all three transcription units closely resembling that seen in Drosophila (F ig.1). By contrast, in lepidoptera there is an altered structure of the E758 transcription unit, in which the E7SB-specific region is fused to exon 2, and E75C transcripts have never been observed (Zhou et al., 1998; Segraves and Woldin, 1993; Jindra et al., 1994a). These differences are paralleled by a striking difference in the length of both the E75A and E758 transcript units. In Drosophila and mosquito, the E75A transcript units, are, e.g. 50 and >6Skb, respectively, comparing to ~16 kb in Manduca (Zhou et al., 1998) A similar relationship is seen for the E75 proteins, in which the conservation of E75 sequences among species is consistent with their phylogenetic relatedness. For example, within the largest block of conserved sequence, the E region, a comparison of AaE75 to E75 from Drosophila, another dipteran, shows approximately half as much 99 dit'ergen proteins t fitmlferw each shox family of Ti conscrx'at resulting i consemm Conserved Smaller an 1993; Jind and AaE7: Proteins b Clean), C0: These Obs lSofOrmS Z fUnQnOnaj Ea OfuleuoE eCdFSOnQ Critical to divergence (93% identity) as the comparison of either diperan E75 protein to E75 proteins of lepidopteran Manduca Sexta, Galleria mellonella and Chortstoneura fumiferana (87% identity). The mosquito, Drosophila and lepidopteran E75 E regions each show approximately 36—37% identity within the E region to the Rev—erb/RVR family of mammalian E75 homologues (Miyajima et al., 1989; Lazar et al., 1990). The comparison between AaE75 and Drosophila E75 also shows clear conservation of each of the three independent isoforms; the AaE75A, AaE7SB and AaE75C-specific amino termini each show several clusters of conserved amino acids, resulting in overall identity of 31, 30 and 33% respectively. While evolutionary conservation of E75A- and E7SB-specific sequences has been seen in Manduca, the conserved regions consisted of only a small number of amino acids within the much smaller amino terminal domains of the lepidopteran E75 proteins (Segraves and Woldin, 1993; Jindra et al., 1994a; Palli et al., 1997; Zhou et al., 1998). In contrast, the AaE75A- and AaE7SB-specific regions much more closely resemble those of the Drosophila proteins both in size and amino acid sequence. The AaE75C-specific region is also clearly conserved, and represents the first E75C isoform seen outside of Drosophila. These observations extend our knowledge of the evolutionary divergence of E75 isoforms and support the hypothesis that these represent evolutionarily conserved functionally independent forms of the E75 protein. Each of the three AaE 75 transcription units is expressed in' the fat body and ovary of vitellogenic female mosquitoes, and in vitro culture experiments indicate that AaE 75 is ecdysone-inducible in isolated fat body. These findings suggest that E 75, which plays a critical role in the control of ecdysone response during Drosophila development, is also 100 involved gene ma} signaling be expres- ecdysone govemed In onset of \' Peak (Ha: increase (1 gradually 20E lCVel; Hou‘eycr‘ 3CCUmula in the ind b69n fOUI involved in the control of reproductive ecdysone response in the mosquito. The AaE 75 gene may be one of the key genes responsible for controlling downstream ecdysone signaling in the regulation of vitellogenic genes in the mosquito. Insofar, E 75 appears to be expressed and required for all ecdysone responses; it may thus be an integral part of an ecdysone response cascade used reiteratively in developmental and reproductive events governed by this hormone. In the fat body (Fig. 6), E 75 transcripts exhibit a sharp rise immediately after the onset of vitellogenesis reaching a peak at 3 hr PBM when ZOE titer shows a first small peak (Hagedom etal., 1975). In contrast, the YPP transcripts show only a small gradual increase during first 4 hr PBM. After the initial drop, levels of AaE 75 transcripts gradually increase again reaching second peak of their expression at the 24 hr PBM when 20E levels have passed their major peak, at 18 hr PBM (Hagedom et al., 1975). However, the second gradual increase in AaE 75 transcripts closely correlates with the accumulation of RNA for YPPs in the fat body, suggesting that AaE 75 may be involved in the indirect regulation of these genes by 20E (Fig. 6). Indeed, E75 binding sites have been found in the regulatory regions of mosquito YPP genes, Vg and VCP (D. Martin and A. Raikhel, unpublished observation). While the transient early induction of AaE 75 may readily be explained in the context of E 75's role in Dr050phila developmental ecdysone response hierarchies, the sustained later response during the period of peak vitellogenesis is less easily understood. Elucidating the conditions, which allow this sustained response of the AaE 75 gene will no doubt be critical to our understanding of the hormonal regulation of developmental events which require prolong expression of genes at a high level, such as vitellogenic IO] genes in 1 high levc ecdysone vitellogei I I; correspon induction that AaE 7 tissues in al,1997;! 1996‘. Zhu mOSquito ImPOrtant VitellOng ‘M \‘8.10" participm le\'€l of C fiI‘SI AUE are harui. Mean}? 2 Die/0,20% requiri‘m Oflhe 1a] genes in the mosquito fat body. Interestingly, AaE7SB reinduction reaches a particularly high level. Recent studies of White et al. (1997), showing the role of 13758 in attenuating ecdysone response, suggest that AaE7SB may be acting here to help terminate some vitellogenic events in the mosquito fat body. In vitro organ culture experiments have demonstrated that 20E induces transcripts corresponding to all three AaE 75 isoforms in the previtellogenic fat body. The 20E induction of the AaE 75 gene was independent of protein synthesis, thus demonstrating that AaE 75 acts as an early gene in ecdysone hierarchy, as previously seen for larval tissues in other insects (Segraves and Hogness, 1990; Jindra and Riddiford, 1996; Palli et al., 1997; Zhou et al., 1998). Unlike Galleria and Manduca E 75 (Jindra and Riddiford, 1996; Zhou et al., 1998), in which E75A exhibited higher sensitivity to 20E than E7SB, mosquito E 75 isoform transcripts show similar sensitivity to this hormone in vitro. Importantly, however, AaE 75 is ten-fold more sensitive to 20E induction than vitellogenic genes, VCP and Vg (Fig. 10). AaE 75 transcripts were induced by 20E at 10' 7M vs. 10'6M required for in vitro induction of VCP and Vg, suggesting that AaE 75 may participate in activation of genes important for vitellogenic events preceding the high level of expression of YPP genes. This suggestion is agreement with the early time of the first AaE 75 expression peak in the fat body in vivo, when the transcripts for YPP genes are hardly detectable. Previously, a similar concentration-dependent pattern of activation of early and late chromosome puffs and genes by 2013 has been observed in Drosophila melanogaster (Karim and Thummel, 1992; Russell and Ashbumer, 1996), reflecting the requirement for a threshold level of early gene activation required for maximal induction of the late genes. [02 by 24 hr of .405 75 response transcript and AaE— considera body, wh‘. the ovary rthiireme “19 semi-i exprCSSiQr Th abOut its l"- SallVar}. ‘1 (- (I decline as unUSualh, plaUSibic . As in the fat body, the highest level of AaE 75 expression in the ovary was reached by 24 hr PBM. More sensitive RT-PCR analysis indicates a much earlier detectable level of AaE 75, as early as 1 hr PBM, in conjunction with the initial onset of ecdysone response. However, in contrast to the fat body, there was no evident early AaE 75 transcript peak in the ovary soon after the onset of vitellogenesis. In addition, AaE75A and AaE7SB transcripts were present in the ovary at the high levels until 42 hr PBM, considerably longer than in the fat body. It is important to remember that unlike the fat body, which is relatively homogeneous tissue consisting predominantly of one cell type, the ovary contains both somatic and germ-line tissues which may have distinct AaE 75 requirements. Indeed, the kinetics of expression of the ecdysone-inducible genes, VgR the germ-line cells and VE in the follicular epithelium, are different. Thus, the expression patterns of AaE 75 transcripts in these ovarian tissues may differ as well. The timing of AaE 75 expression within the ovary raises interesting questions about its position within the reproductive ecdysone response hierarchy. In Drosophila salivary glands, E 75 transcripts peak within approximately 2-4 hr, and then begin to decline as a result of negative autoregulation. One factor proposed to influence the timing of ecdysone response in Drosophila is the time required to transcribe the unusually long early genes, E74, BR-C and E 75 (Thummel, 1992). It also seems plausible that the full progression of vitellogenic ecdysone response may be most closely analogous to the series of ecdysone responses in the Drosophila third instar larva and prepupa, in which each ecdysone pulse results in the synthesis of factors required to set the stage for next (Hagedom et al., 1975), rather than to a single discrete ecdysone response. l03 levels oi'I acting as during ti: equivale: broader t autoregul Suggests gene isoft difference \vithin cm active dur Ofecdygo during 00 lWham [Q relam'e u. have dl lTC' While these factors may contribute to the relatively slow appearance of high levels of AaE 75 transcripts, the broad, sustained response suggests that AaE 75 may be acting as an early-late in this system, such that it is further induced rather than repressed during the late ecdysone response. Hierarchically, the early and early-late genes are equivalent (Woodard et al., 1994); however, the early-late genes are expressed over a broader time frame than the transiently expressed early genes which are subject to autoregulatory repression. Comparison between different tissues in the Drosophila larva suggests that there can be substantial differences in the kinetics with which various early gene isoform levels rise and fall (Huet et al., 1993; I995); presumably, these reflect differences in the temporal requirements of these genes for the regulation of target genes within each tissue. Comparison of AaE 75 expression to the expression of other genes active during the peak of vitellogenesis suggests that it may be required for the regulation of ecdysone induced genes, such as vitelline envelope protein, for a sustained period during oogenesis. The specific AaE 75 isoforms which might be involved in this process remain to be determined; although AaE75C expression is low in the ovary as a whole relative to fat body, the ovary consists of both somatic and germ line tissues which may have different AaE 75 requirements. Despite these intriguing differences, the present findings suggest that elements of 20E-triggered regulatory hierarchies are used reiteratively during development and reproduction. Indeed, E 75 is thus far expressed and required for all characterized 20E responses; it may be an integral part of all or nearly all 20E-responsive regulatory hierarchies. Parallel genetic studies in Drosophila support the suggestion that reproductive and developmental ZOE responses are controlled by closely related primary l04 response BR-C C; et al.. i9 mid-00g in mosqt reproduc strengths possible i ofinsect CVolved. ’7- genOmic Brad D, Slipping ArizOn: Support WAS responses. Of the three transcription factor-encoding classical early genes, E 75, E74 and BR-C, each is involved in the regulation of oogenesis as well as development (Buszczak etal., 1999). Remarkably, E 75 and E74 appear to be involved in progression through a mid-oogenesis checkpoint, which may be analogous to the previtellogenic ovarian arrest in mosquitoes, suggesting that the fundamental mechanisms underlying the control of reproductive 20E response may be highly conserved. By combining the complementary strengths of A. aegipti physiological studies and Drosophila genetics, it should be possible to dissect the mechanisms of through which 20E and E 75 control critical aspects of insect reproduction, and to understand the ways in which these mechanisms have evolved. ACKNOWLEDGMENTS I thank William Pierceall and William Segraves at Yale University for cDNA and genomic cloning of AaE 75, Avraham Biran for Northern blot. I also thank Judy Cole and Brad Dunlap for assistance in the isolation and analysis of genomic clones, Dr. Tom Sappington for critical reading of the manuscript, and Dr. H. Hagedom (University of Arizona) for the gift of a mosquito vitelline envelope gene probe. This work was supported by a grant from the National Institutes of Health (A136959) to A.S.R. and W.A.S. W.E.P was supported in part by the Seessel Anonymous Fund. 105 Chapter 4* Cloning and Characterization of the Mosquito Early-late Gene HR3 *Originally published in Mol. Cell. Endocrinol. Ref: Kapitskaya, m, 2., Li, g“, Miura, 1c, Segraves, w., and Raikhel, A. s. (2000). Mol. Cell. Endocrinol. 160, 25-37. # Underlined two authors contributed equally to this work. 106 controllii metamon‘_ understai deveIOpn‘ important molecular meal-actii hierarchy eXpressioi been prev regUlaIOQ representc ”‘6 reprod COnsenan regulating AHR3, a h inducible 3 female mC ec9).-”Stem, culture ex; acme ecdv ABSTRACT The insect steroid hormone, 20-hydroxyecdysone (20E), is a key factor controlling critical developmental events of embryogenesis, larval molting, metamorphosis, and, in some insects, reproduction. Our lab is interested in understanding the molecular basis of the steroid hormone ecdysone action in insect egg development. The yellow fever mosquito, Aedes aegypti, in addition to being an important vector of human diseases, represents an outstanding model for studying molecular mechanisms underlying egg maturation due to stringently controlled, blood meal-activated reproductive events in this insect. To elucidate the genetic regulatory hierarchy controlling the reproductive ecdysone response, ecdysone-regulated gene expression in vitellogenic mosquito ovaries and fat bodies has been investigated. It has been previously demonstrated the conservation of a primary ecdysone-triggered regulatory hierarchy, implicated in development of immature stages of Drosophila, represented by the ecdysone receptor/Ultraspiracle complex and an early gene E 75 during the reproductive ecdysone response (Chapter 2, 3). The present study demonstrates that conservation of the factors involved in the ecdysone-responsive genetic hierarchy regulating female reproduction extends beyond the early genes. Here, it is a report on AHR3, a highly conserved homologue of the Drosophila HR3 early-late ecdysone- inducible gene in the mosquito. AHR3 is expressed in both vitellogenic tissues of the female mosquito, the fat body and the ovary. The expression of AHR3 correlates with the ecdysteroid titer, reaching a peak at 24 hr after a blood meal. Moreover, in vitro fat body culture experiments demonstrate that the kinetics and dose response of AHR3 to 20E, an active ecdysteroid in the mosquito, is similar to those of the late vitellogenic genes rather l07 than the activatit inhibitor regulatir‘ T dCVClOpn some insl Analysis larval ant Small nUl‘ large Hun hierarcm hcterodir. the reunc FunhL’rm Sindeed IranSCn-p Segraves 01., 1991 UE[\,\-Ork ”D the m than the early E 75 gene. However, as shown for other early and early-late genes, the 20E activation of AHR3 is not inhibited by the presence of cycloheximide, a protein synthesis inhibitor. Taken together, these findings strongly suggest AHR3 involvement in regulating the vitellogenic response to ecdysone in the adult mosquito. INTRODUCTION The insect steroid hormone 20—hydroxyeedysone (20E) regulates multiple developmental processes including embryogenesis, larval molting, metamorphosis and, in some insects, reproduction (Bownes, 1986; Riddiford, 1993a,b; Thummel, 1995). Analysis of the ecdysone effect on polytene chromosome puffing patterns in the late larval and prepupal salivary gland of Drosophila suggested that the initial activation of a small number of early ecdysone-inducible genes leads to subsequent inducement of a large number of late target genes (Ashbumer et al., 1974). Elucidation of this genetic hierarchy at the molecular level has led to the identification of the ecdysone receptor as a heterodimer of two nuclear receptors, the ecdysone receptor (EcR) and the homologue of the retinoid X receptor, Ultraspiracle (USP) (Koelle et al., 1991; Yao et al., 1992; 1993). Furthermore, these studies have shown that the action of the ecdysone-receptor complex is indeed mediated by early genes such as the BR-C, E74 and E 75 genes encoding transcription factors involved in regulation of late gene expression (Burtis et al., 1990; Segraves and Hogness, 1990; Thummel et al., 1990; Guay and Guild, 1991; DiBello et al., 1991; Fletcher and Thummel, 1995). The ecdysone-mediated genetic regulatory network is further refined by the presence of a set of genes which are involved in setting up the timing and stage—specificity of gene activation by this genetic hierarchy 108 (Thummel, 1997). The orphan nuclear receptor DHR3, an early-late gene with induction characteristics including properties of both early and late genes, is one of the key genes in ' the ecdysone-mediated genetic regulatory network. Recent studies have revealed that during insect metamorphosis DHR3 has a dual role in repressing the early genes while activating another orphan nuclear receptor, flF TZ—F 1, which in turn provides competence for the stage-specific late prepupal response to 20E (Woodard et al, 1994; Lam et al, 1997; White et al, 1997; Broadus et al., 1999). DHR3 homologues cloned from several Lepidopteran insects are highly conserved and their expression suggests HR3 involvement in mediating ecdysone response during Lepidopteran development and metamorphosis (Palli et al., 1992; I996; Jindra et al., 1994b). Recent analysis of Drosophila DHR3 mutants has revealed that DHR3 function is, in addition, required to complete embryogenesis (Carney et al., 1997), prepupal—pupal transition and differentiation of adult structures during Drosophila metamorphosis (Lam et al., 1999). In contrast to the wealth of information concerning the molecular basis of the ecdysone-mediated genetic hierarchy during development and metamorphosis, comparatively little is known about the ecdysone-regulated aspects of insect female reproduction, oogenesis and vitellogenesis. In Drosophila, ecdysone involvement in female reproduction has been suggested by the female sterility of mutations affecting ecdysone synthesis and ecdysone response (Garen et al., 1977; Oro et al., 1992), by the ecdysone-inducibility of yolk protein genes (Bownes, 1986) and most recently by studies implicating the ecdysone response hierarchy in egg chamber maturation (Buszczak et al., 1999). The role of ecdysone in female reproduction is best documented in anautogenous mosquitoes, especially the yellow fever mosquito, Aedes aegypti. In anautogenous 109 mosquitoes, a blood meal is required for egg maturation. Ingestion of blood stimulates the brain to secrete the ecdysiotropic neuropeptide, egg development neurosecretory hormone or ovarian ecdysteroidogenic hormone (Lea, 1967; 1972; Matsumoto et al., 1989; Brown et al., 1998). These hormones activate the ovary to produce ecdysone, which controls the synchronous progression of egg development in the ovary and the synthesis of yolk protein precursors by the fat body. In A. aegypti, ecdysteroid levels are relatively low during the first 8 hr post blood meal (PBM), with only a small peak at 4 hr. Later, the ecdysone level rapidly increases to the maximum at 18-24 hr PBM, then abruptly declines to the previtellogenic level in preparation for the termination of vitellogenesis at 30-32 hr PBM (Hagedom, 1983; 1985; Raikhel, 1992; Dhadialla and Raikhel, 1994). Action of 20-hydroxyecdysone on the mosquito yolk protein genes, vitellogenin (Vg) and vitellogenic carboxypeptidase (VCP), requires protein synthesis, suggesting indirect control by the genetic regulatory cascade involving early genes (Deitsch et al., 1995a,b). In the mosquito ovary, established functions associated with ecdysone include the formation of the vitelline envelope (Raikhel and Lea, 1991; Lin et al., 1993) and the separation of the secondary follicle in the ovary (Beckemeyer and Lea, 1980). Thus, the mosquito provides an ideal system to investigate control of ecdysone- induced gene expression during female reproduction. Furthermore, the critical role of anautogenous mosquitoes as disease vectors increases the significance of elucidating the ecdysone-mediated gene regulatory hierarchy, a potential target in the development of novel strategies for vector control. In full agreement with the suggested role of ecdysone in regulating female reproduction, ecdysone receptor transcripts can be detected in pre- and vitellogenic llO ovaries and fat bodies (Cho et al., 1995). Transcripts encoding two different USP isoforms are differentially expressed in these tissues (Kapitskaya et al., 1996; Wang et al., 2000a). The mosquito EcR/U SP heterodimer has been shown to bind to various ecdysone response elements (EcREs) to modulate ecdysone regulation of target genes (Wang et al., 1998). Recently, the A. aegypti homologue of the Drosophila E 75 gene, a putative representative of the next level in the ecdysone response hierarchy, has been identified and characterized. A. aegypti E 75, AaE 75, is expressed in ovary and fat body following a blood meal. AaE 75 transcripts, corresponding to three E75 isoforms, are ecdysone-inducible in isolated fat body cultured in vitro. These findings suggest that AaE 75 mediates ovary and fat body ecdysone responses, and that ecdysone-triggered regulatory hierarchies such as those implicated in the initiation of metamorphosis may be reiteratively utilized in the control of the reproductive ecdysone response (Pierceall et al., 1999). The present study demonstrates that conservation of the factors involved in the ecdysone genetic hierarchy regulating female reproduction extends beyond the early genes. It is shown here that a highly conserved homologue of DHR3 is expressed in the vitellogenic tissues of the female mosquito, the fat body and the ovary. The expression of the mosquito homologue, which is named AHR3, correlates with the titer of 20E, peaking at 24 hr PBM. Furthermore, 20E activates the AHR3 transcript in the previtellogenic fat body in vitro; this activation is not inhibited by cycloheximide, a protein inhibitor, suggesting that AHR3 is acting as an early-late gene in this system. Taken together, these findings strongly suggest AHR3 involvement in regulating the vitellogenic response to ecdysone in the adult mosquito. Ill MATERIALS AND METHODS Animals Mosquitoes, A.aegypti, were reared according to Hays and Raikhel (1990). Larvae were fed on a standard diet as described before (Lea, 1964). Vitellogenesis was initiated by allowing females 3-5 days after eclosion to feed on an anesthetized white rat. Materials The RNA ladder was purchased form Life Technologies, Inc.; Sequenase was from U. S. Biochemical Corp.; restriction enzymes were from Boehringer Mannheim. All primers used in PCR were ordered from Gibco-BRL. Superscript II reverse transcriptase and Taq DNA polymerase were from Gibco-BRL; random hexamer primers from Promega. Perkin-Elmer was the source of reagents for the polymerase chain reaction (PCR). MSI CO supplied nitrocellulose-blotting membranes. Radionucleotides for labeling of probes and DNA sequencing were from NEN life Science Products. All other reagents were of analytical grade from Sigma or Baker. Cloning, sequencing, and 5 ’RA CE of cDNA The cDNA fragment of AHR3 was first obtained by PCR, for which degenerate primers were designed based on the sequences of its homologues in other insect species. Amplification was achieved in a Perkin Elmer thermal cycler using the template cDNA reverse-transcribed from 20 pg of total RNA prepared from the fat bodies of vitellogenic female mosquitoes. The PCR-generated fragment was used as a probe to screen a 112 XZAPII cDNA library, which was prepared from the fat bodies of vitellogenic female mosquitoes 6-48 hr post-blood meal (PBM) as previously reported (Cho and Raikhel, 1992). Several positive cDNA clones were subsequently isolated and sequenced in W.M. Keck Foundation Biotechnology Resource Laboratory at Yale University. Analyses of nucleotide and deduced amino acid sequences were performed using the software of GCG (Genetics Computer Group, University of Wisconsin, Madison). 5’ RACE (Rapid Amplification of cDNA Ends) was performed with a 5’ RACE kit from GIBCO/BRL using the manufacturer’s protocol. The first strand of cDNA for the mosquito HR3 gene was prepared with the gene-specific primer HR3-SP1: ATTTGCGCTAACATGCTATCG and total RNA from post blood meal 24 hr fat bodies. After the first round PCR, the gene-specific primer HR3-SP2: CAGCCATTTCAAGTTCACTACG for HR3 was used for the second round nested PCR. The PCR fragments positively confirmed by Southern Blot hybridization were then subcloned into pGEM-T-easy vectors (Promega) and sequenced. Northern blot hybridization Total RNA was isolated from mosquitoes of different stages and tissues using the guanidine isothiocyanate method as described previously (Bose and Raikhel, 1988). Polyadenylated mRNA was isolated using Niomag oligo (dT)20 magnetic beads and the manufacturer’s protocols (PerSeptive Diagnostics, Inc.). For Northern blot analysis, total or poly(A)+ RNA was separated by electrophoresis in 1.2% agarose/formaldehyde gels in MOPS buffer, blotted to a nitrocellulose membrane, and hybridized with 32P-labed DNA 113 probes under high stringency conditions. Autoradiography was conducted at -80°C using intensifying screens. R T-PCR/Southern blot analyses The temporal profile of transcript abundance of HR3 in the mosquito fat body was examined by RT-PCR followed by Southern blotting with the gene-specific radioactive probe. Total RNA was prepared from fat bodies throughout the first vitellogenic cycle, as described in Northern Blot above, with the modification that all isopropanol precipitation steps were done without low-temperature incubation to avoid co- precipitation of glycogen and salt. RNA concentration was determined spectrophotometrically; Absorbance ratios, A260/A280 and A260/A230, in RNA preparations were always above 1.7 and 2.0, respectively, quite consistent irrespective of the developmental stages of mosquitoes. Five microgram aliquots of each RNA preparation were reverse-transcribed by SuperscriptII reverse transcriptase (Gibco-BRL) and random hexamer (Promega) in a reaction volume of 20 pl. After the reaction, the volume of each sample was brought to 40 pl with TE buffer (10 mM Tris-Cl, lmM EDTA, pH 8.0). cDNA samples were stored at -200C until use. In the case of mosquito tissues, 0.025 fat body- or pupae-equivalent cDNA pools were used as PCR templates. See Fig. l for the details ofthe primer design. Thermal cycling conditions were as follows: the samples were incubated at 940C for 3 min, followed by 18 cycles of 940C for 30 sec, 60°C for 30 sec and 720C for 30 see. As a reference to the developmental change of the fat body, the samples of the same cDNAs were subjected to the 10 cycles of PCR with a primer pair specific to the 114 mosquito vitellogenic carboxypeptidase; the reaction was followed by Southern blotting hybridization using a V CP specific probe generated by PCR using DNA from a VCP cDNA clone as a template (Cho et al., 1991). The primers for V CP were: forward, 5’- AGCGCCCATTCTTGTTTGG-3 ’ and reverse, 5 ’-CAGCTCATACAGGTATTCTCC-3 ’. RT-PCR/Southem blotting of the total RNA preparations without reverse transcriptase did not result in any appreciable signal, indicating that contamination of RNA preparations with genomic DNA fragments was virtually negligible (not shown). The radioactive blots were quantified with a phosphorimager (Molecular Dynamics). In vitro fat body culture To examine the effect of ecdysone on expression of the mosquito HR3 gene, the abdominal walls with adhering fat bodies (hereafter referred to as the fat body) were dissected from 3-5-day-old previtellogenic females and incubated in an organ culture system as previously described (Raikhel et al., 1997). The fat bodies were incubated with 20E either for 16 hr, or after 4hr with 20E followed by washing with the hormone- free medium 3 times and 12hr continuing incubation in the hormone-free medium. At every 4 hr interval, a sample of 9 fat bodies was collected and stored in liquid nitrogen until use. Total RNA was extracted as described in RT-PCR above. As a control, RNA was extracted from fat bodies incubated in the hormone-free medium. RT-PCR/Southem blot analyses were utilized as described above. Hybridization using probes for the ecdysone-inducible VCP gene was provided as a positive control. To test the effect of the protein synthesis inhibitor cycloheximide (Chx) on transcription, dissected 3-5 day old female mosquito fat bodies were pretreated with 115 culture media containing lmM Chx for 1 hr then further incubated with the same concentration of Chx either without or with 106 M 20E. The Chx/hormone withdrawal experiment was conducted as described for the hormone withdrawal experiment above except that lmM Chx was added to all culture media. RESULTS Isolation and characterization of a cDNA clone encoding the mosquito AHR3 nuclear receptor To obtain a mosquito HR3 clone, degenerate primers were designed, based on the amino acid comparison of its insect homologues. The sense primer Pr-l conformed to the conserved sequence QIEIIPCK adjacent to the C (DNA-binding) domain; the antisense primer Pr—2 corresponded to the sequence EKVEDEVR from the conserved part of the D domain of these nuclear receptors (Fig. 1). After a mosquito PCR-fragment of the expected size, 285 bp, was sequenced and confirmed to encode a DHR3-related polypeptide, it was used as a probe to screen the mosquito fat body cDNA library (see Materials and Methods). The cDNA clone with the largest insert of 2.56 kb (designated as AHR3 clone) was fully sequenced. This cDNA clone contained an open reading frame (ORF) of 1.42-kb and 3’-untranslated region (3’-UTR) of 1.14 kb. It did not have in- frame stop-codons at the 5’-end, indicating the ORF could potentially be truncated at the 5’-terminus; it also lacked a canonical site of polyadenylation and poly(A)-tail at the 3’- end. In the isolated AHR3 cDNA clone, there were 20 base pairs without any in-frame termination codon located upstream of potential translation initiation site ATG. In order 116 Fig.1. Alignment of the deduced amino acid sequence of the mosquito AHR3 cDNA with its insect homologues: DHR3 from Drosophila (Koelle et al, 1992), MHR3 from Manduca (Palli et a1, 1992), and GHR3 from Galleria (Jindra et al, 1994b). Asterisks or hats indicate amino acids, which are identical or similar, respectively. The borders of DNA-binding domain (DBD) and putative ligand-binding domain (LBD) are marked with square braces. The P-box in DBD is in bold. The carboxy-terminal extension (CTE) of DBD, defined according to Giguere et. al. (1994), is double-underlined. The conserved 8-amino acid stretch at the C-termini of LBD is in bold. Only part of A/B- domains, adjacent to DBDs, are shown for DHR3, MHR3, and GHR3 (notice a high percentage of identical amino acids between lepidopteran MHR3 and GHR3). The vertical arrowhead marks a position from which a high similarity between different HR3 receptors starts. Horizontal double arrows indicate sequences corresponding to the degenerate sense primer (Pr-1) and antisense primer (Pr-2) used for AHR3 cloning by PCR. An arrow shows the sequence-specific antisense PCR-primer Pr-3, and two black dots stand for two Xhol restriction sites of the AHR3 cDNA, which were utilized to prepare a single-stranded DNA hybridization probe (see Fig. 3, 4). Two arrows indicate the sequence-specific sense (Pr-4) and antisense (Pr-5) primers used for RT-PCR analysis (see Fig. 5, 6, 7, 8). The sequence of the mosquito AHR3 has been submitted to the GenBank (http;//www.ncbi.nlm.nih.gov1; its accession number is AF230281. 117 [——— DBD Pr-l P154 P'box 1 MLRDAPNRSELEMAVSSTVFDSMLAQIEIIPCKVCGDKSSGVHYGVITCIGCKGFFRR 58 19 HANNLGQSNVQSPAGQNNSSGSIKAQIEIIPCKVCGDKSSGVHYGVITCIGCKGFFRR 77 73 SSEGMFGP-ISGMFMDKKAANSIRAQIEIIPCKVCGDKSSGVHYGVITCIGCKGFFRR 130 76 SSEGMFGSSISGMFMDKKAANSIRAQIEIIPCKVCGDKSSGVHYGVITCEGCKGFFRR 134 * ********************************** Pr-S Pr-2 DEBS iiii iiii iiii iiii iiii iiii SQSSVVNYQCPRNKQCVVDRVNRNRCQYCRLQKCLKLGMSRDAVKFGRMSKKQREKVEDEVRFH SQSSVVNYQCPRNKQCVVDRVNRNRCQYCRLQKCLKLGMSRDAVKFGRMSKKQREKVEDEVRFH SQSTVVNYQCPRNKACVVDRVNRNRCQYCRLQKCLKLGMSRDAVKFGRMSKKQREKVEDEVRYH SQSTVVNYQCPRNKACVVDRVNRNRCQYCRLQKCLKLGMSRDAVKFGRMSKKQREKVEDEVRYH ***A********** ***********************************************A* o RAQMRAQSDAAPDSSVFDTQ--TPSSSDQLHHGGYNGY-A--Y-NNEV---GYGSP---YGYST RAQMRAQSDAAPDSSVYDTQ--TPSSSDQLHHNNYNSY-SGGYSNNEV---GYGSP---YGYSA KAQMRAQNDAAPDS-VYDAQQQTPSSSDQFH-GHYNGY-P ----------- GYASPLSSYGYNN RAQMRAQTDAAPDS-VYDAQQQTPSSSDQFH—GHYNGYPG--Y-RSPLSSYGYGGN---AGPAL A******A****** *A*A* *******A* A **A* **AA * S----VTPQ -------- QTMGYDISADYVDSTTTYEPRST--IIDSDFISGHTEGDINDVLIKT s—--—VTPQ -------- QTMQYDISADYVDSTT-YEPRST--IIDPEFIS-HADGDINDVLIKT AGPPLTSNMSSIQAQPQAQQPY---—DYADSTTTYEPKQPG-YLDADFI-GQVEGDISKVLVKS TSNMNIQPQAP ------ QQQPYDVSADYVDSTTAYEPKQTGGFLDADFI-GHAEGDISKVLVKS A **A**** *it““ A* A** ***A **A*A [— LBD LAEAHANTNHKLEIVHDMFRKSQDVTRIMYYKNMSQEELWLDCAEKLTAMIQQIIEFAKLIPGF LAEAHANTNTKLEAVHDMFRKQPDVSRILYYKNLGQEELWLDCAEKLTQMIQNIIEFAKLIPGF LAERHANTNPKLEYINEMFSKPQDVSKLLFYNSLTYEEMWLDCADKLTQMIQNIIEFAKLIPGF LAEAHANTNPKLEYVHEMFRKPPDVSKLLFYNSMTYEEMWLDCADKLTGMIQNIIEFAKLIPGF *** ***** *** AAA** * *iAAAAA*AAAA **A*****A*** ***A*********** MRLSQDDQILLLKTGSFELAIVRMSRLMDLSTNSVLYGDIMLP-QEVFYTSDSFEMKLVACIFE MRLSQDDQILLLKTGSFELAIVRMSRLLDLSQNAVLYGDVMLP-QEAFYTSDSEEMRLVSRIFQ MKLTQDDQILLLKSGSFELAIVRLSRLIDVNREQVLYGDVVLPIRECVHARDPRDMALVSGIFE MKLSQDDQILLLKSGSFELAIVRLSRLIDVNREQVLYGDVVLPIRECVHARDPRDVTLVVGIFD *A*A*********A*********A***A*AA AA*****AA** A* AA * AA **A **A Pr-3 TAKSIAELKLTETELALYQ LVLLWPERNGVRGNTEIQRLFEMSMSAIRQEIEANHAPLKGDVT TAKSIAELKLTETELALYQSLVLLWPERNGVRGNTEIQRLFNLSMNAIRQELETNHAPLKGDVT AAKSIARLKLTESELALYQSLVLLWPERHGVMGNTEIRCLFNMSMSAMRHEIEANHAPLKGDVT AAKTIERLKLTETELALYQSLVLLWPERHGVRGNPEIQCLFNMSMAAMRHEIESNHAPLKGDVT A**A* A*****A***************A** ** *** **AA**A*A*A*A*A********** o VLEILLNKIPTFRELSIMHMEALQKFKQDHPQYVFPALYKILFSIDSQQDLMT VLDTLLNNIPNFRDISILHMESLSKFKLQHPNVVFPALYKILFSIDSQQDLT VLDTLLAKIPTFRELSLMHLGALSRFKMTHPHHVFPALYKELFSLDSVLDYTHG VLDTLLAKIPTFRELSLMHLEALCRFKAAHPHHVFPALYKILFSLDSVLDYTHG *iA ** A**A**AA*AA*A A* A** **A ***********A** i 468 487 548 557 118 122 141 194 198 174 199 245 257 224 244 302 311 288 308 366 375 351 371 430 439 415 435 494 503 to identify the missing 5’-end of AHR3 cDNA, S’RACE PCR was carried out using total RNA from the fat body 24 hr post blood meal (PBM) as a template (see Materials and Methods). A 200-bp fragment was the major amplification product (data not shown). To verify the absence of genomic contamination, 5’RACE was performed under the same conditions except without superscript II reverse transcriptase. This control did not result in any amplification product, indicating that the 200-bp PCR fragment originated from mRNA. This fragment was subcloned into an pGEM-T-easy vector (Promega) and sequenced. It shared an expected 36-bp sequence overlapping with the S’end of the AHR3 cDNA clone. This observation confirmed that the 200-bp fragment was indeed a portion of the mosquito HR3 cDNA. There were two in-frame termination codons, TAG and TGA, located at the 45 bp and 42 bp upstream of the initiation site ATG, suggesting that the 5’ end of the AHR3 ORF was complete. Conceptual translation of the mosquito HR3 clone showed that it exhibited a structural organization similar to that of other members in the HR3-subfamily of nuclear receptors (F i gs.l and 2). The most conserved regions were the DNA-binding domain (DBD) and so—called carboxy-terminal extension (CTE) of the DNA-binding domain (Figsl and 2). As illustrated in Fig.1 and Fig.2, the mosquito AHR3 had the P-box which shared 100%, 97% and 76% identical amino acids in the DBD, and 100%, 91% and 74% identical amino acids in CTE with Drosophila HR3, Chorz'stoneura HR3 and human homologue RORa, respectively. This indicated that the AHR3 and other HR3 receptors could bind to similar target DNA-sites. A putative LBD of AHR3 did not show high similarity with those of the nematode CeHR3 and human RORa counterparts, though it was still significantly conserved within 119 HI<——>I<——>I< > AHR3 DBD! E H 31 66 23 114 234 DHR3 300%] g 33% SO 66 23 115 233 MHR3 {2% g 61% 103 66 23 120 236 CHR3 -7%‘ g 48% 101 66 23 121 235 CeHR3 591% g 127 66 23 115 222 RON“ Fig. 2. Domain comparison of the mosquito HR3 with selected members of the ROR/HR3 subfamily of nuclear receptors. Shown are the HR3 nuclear receptors: AHR3 from Aedes aegypti (this study), DHR3 from Drosophila (Koelle et al, 1992), MHR3 from Manduca (Palli et al, 1992), CHR3 from Chortstoneura (Palli et al, 1996), CeHR3 from nematode Caenorhabditis elegans (Kostrouch et al, 1995), and human RORal (Giguere et al, 1994). The letters above the mosquito HR3 protein denote functional domains. The D-domain is divided into the CTE (carboxy-terminal extension) and H (hinge) subdomains. Numbers below proteins indicate the number of amino acids in each subdomain. The percentages of identities between corresponding domains of AHR3 and other homologues are indicated within each domain; the ratios were calculated as a number of amino acids identical between comparative sequences divided by the total amino acid number of the shorter sequence. 120 insect LBDs (Fig. 2). Notably, there were 8 conserved amino acids in all members of the HR3 subfamily at the C-terminus of LBD (shown in bold in Fig. 1). They contained a stretch LYKELF similar to the AF-2 core motif (Beato et al, 1995), which strongly suggested the functional significance of this sequence. The high similarity between HR38 started in A/B-domains from the 7th amino acid preceding DBDS; however, the rest of the HR3 A/B domain was poorly conserved in all members of the insect HR3. The AHR3 transcripts and the time course of AHR3 expression in the mosquito ovary and fat body during the first vitellogenic cycle To determine the size of AHR3 transcripts, Northern analysis was conducted for poly (A) RNA of the ovary and fat body isolated from females in the middle of the vitellogenic stage (24 hr PBM), when female mosquitoes were exposed to a high concentration of the steroid hormone 20E. Analysis of the ovarian RNA revealed the presence of a major 7.0 kb transcript, and two minor transcripts of 9.5 kb and 5.9 kb in size. In the vitellogenic fat body, the same three AHR3 transcripts were observed. In contrast to the ovary, however, all three transcripts were expressed evenly in the fat body. Interestingly, in pupae only the major 7.0 kb and minor 9.5 kb transcripts were present (Fig. 3). Northern blot analysis of AHR3 RNA in the ovary from female mosquitoes taken at different stages of vitellogenesis showed a transcriptional pattern of AHR3 with a prominent peak at 24-30 hr PBM, soon after a peak of 20E titer in the mosquito hemolymph (Fig. 4). Due to the relatively low abundance of AHR3 mRNA, the RT-PCR analysis was used for elucidation of the AHR3 mRNA kinetics in the female mosquito fat body during 121 0v FB P _ 8.5 kb 9.5 kb — 7.0 kb — a _ " N 7.0 kb — 5.9 kb it 5.9 kb '— Fig. 3. AHR3 transcripts in the mosquito Aedes aegypti. Northern blot analysis of poly(A) RNA extracted from ovaries (CV) and fat bodies (F B) of female mosquitoes, and whole body pupae (P). A 0.82 kb XhoI-fragment of the AHR3 cDNA (marked by two black dots in Fig. 1) was a template for a sequence-specific anti-sense primer Pr-3 (shown in Fig. l) to prepare a hybridization probe by PCR. The resulting single-stranded DNA-probe of 0.68 kb corresponded to the C-terminal part of the hinge-subdomain and the N-terminal part of the LBD of the mosquito HR3. 122 PBM (h) PV 6 1 2 1 8 24 30 36 42 9.” _ - - “Ir 7.0%.! -i n 5..“ ‘1 tubuacautidfivo. Fig. 4. Expression of AHR3 in the mosquito ovary during the first vitellogenic cycle. Northern blot analysis of total RNA extracted from ovaries of female mosquitoes dissected before (PV) or after a blood meal (PBM) at the indicated times. Hybridization was performed as described in Fig. 3. The bottom lanes present ribosomal RNA in the same gel stained by ethidium bromide prior to blotting. PV ((1) PBM Q1) 0-1 1-2 3-5 1 3 4 6 15 24 36 48 (A) .. C.- .... Fig. 5. Expression of AHR3 in the mosquito fat body during the first vitellogenic cycle. Total RNA, extracted from mosquito fat bodies, was reverse transcribed and cDNA equivalents to 0.025 mosquito fat body were subjected to PCR, using primers specific to either AHR3 or VCP. The resulting PCR-fragments were hybridized with 32P- labeled gene-specific DNA-probes. (A) RT-PCR/Southem analysis of the AHR3 expression. The 0.22 kb PCR-fragment was generated with the AHR3-specific primers Pr-4 and Pr—S (shown in Fig. 1). (B) RT-PCR/Southem analysis of the VCP expression. VCP is a late gene regulated by 20E in the mosquito fat body (Deitsch et al., 1995a). (B) 123 the first vitellogenic cycle. AHR3 mRNA was absent during the previtellogenic period, the transcript could be detected only after the onset of vitellogenesis triggered by a blood meal. A peak of AHR3 expression was clearly seen at 24 hr PBM. Determination of the VCP mRNA levels in the same samples clearly showed that this expression of AHR3 mRNA coincided with the peak expression of yolk protein genes (Fig. 5). In vitro 20E induction of AHR3 mRNA in the female mosquito fat body To determine whether the positive correlation between the AHR3 expression and the peak of ecdysteroids observed in vivo reflected the involvement of 20E in regulation of expression of the AHR3 gene, I investigated the patterns of AHR3 transcript accumulation in in vitro cultured fat bodies in response to 20E. For these experiments, previtellogenic fat bodies of 3-5 days after eclosion were incubated in the culture medium in the presence or absence of the hormone. The RT-PCR analysis was used to monitor the transcript levels in the cultured tissues (see Materials and Methods). In the preliminary experiments it was found that the AHR3 transcript, which was absent in the previtellogenic fat body, appeared after exposure of the tissue to 20E (not shown). In the next series of experiments, I determined the 20E concentration required for maximal activation of AHR3 in the fat body by culturing this tissue with increasing concentrations of the hormone. Fat bodies from previtellogenic 3-5 day-old females were used to determine the background levels of transcripts for an early gene E 75, a late gene V CP, a housekeeping gene actin, and the early-late gene under investigation, AHR3. Actin was expressed in detectable amounts in previtellogenic fat bodies. Culturing in the medium without the hormone did not result in any measurable increase in mRNA of all these genes (Fig. 6). The levels of actin mRNA remained relatively constant in the presence of 124 140 IE75A, 120 1 135753; 53400 IHRS ; “ V IVCP . 33 s ' s 2 § . \‘s - °°* \ Se ss E ‘° R \\ R 20 \ s \ o, . It s s s PV 0 10* 10'7 10‘ 10“ ZOE (M) Fig. 6. AHR3 response to 20-hydoxyecdysone (20E) in in vitro fat body organ culture. The transcriptional response of AHR3 to 20E was examined in an in vitro culture system of female previtellogenic fat bodies (Deitsch et al., 1995a; Raikhel etal., 1997). Total RNA was extracted from untreated previtellogenic (PV) fat bodies, or from fat bodies incubated for 2 hr at the indicated 20E concentrations. RT-PCR/Southem blot hybridization was performed using probes specific for AHR3 or each other gene (see Materials and Methods, and Fig. 1). AaE 75A and AaE 75B provided controls for an early gene, VCP for a late 20E-responsive gene. The results represent the average of three independent experiments : S.D. (a) Culture Time (hr) (b) Culture Tlme (hr) Fig.7. Time course of AHR3 induction by 20E in in vitro fat body organ culture. Accumulation of AHR3 mRNA (A) and VCP mRN A (B) durin culturing in the hormone-free medium (CM), in the continuous presence of 10' M 20E (20E), or 4 hr exposure to 20E followed by culturing in the hormone-free medium (20E-4hr). RT- PCR/Southem blot hybridization was performed using probes specific for either AHR3, or VCP as a late 20E-responsive gene. The results represent the average of three independent experiments i SD. 125 every tested concentration of 20E (not shown). In contrast, E 75, HR3 and VCP genes responded to increasing concentrations of 20E in the culture medium by dramatic rise in their mRNA levels. As it has been previously established, the VCP mRNA reached its maximal levels at 10'6 M of 20E (Deitsch et al., l995a), while E 75A and E 758 mRNAs at 10'7 M of 20E (Pierceall et al., 1999). The mRNA levels of AHR3 increased in a dose- dependent manner in response to 20E. The AHR3 transcript reached its maximal level at the 20E concentration of 10'6 M (Fig. 6). Culturing the previtellogenic fat bodies in the hormone-free medium up to 16 hr did not induce AHR3 and VCP mRNAs (Fig. 7). In contrast, AHR3 mRNA appeared rapidly in the fat bodies cultured in the medium containing 10'6 M 20E. The levels of AHR3 mRN A then gradually increased over the l6-hr culturing period and did not show any regression (Fig. 7A). The VCP mRNA induction profile was similar to that of AHR3, the levels of this transcript increased continuously throughout the entire 16 hr incubation with 10'6 M 20E (Fig. 7B). In the next experiment, the fat bodies were cultured in this hormone-containing medium for only 4 hrs, washed three times with the hormone—free medium and continued incubation in the hormone-free medium for 12 more hrs. Both AHR3 and V CP mRNA levels increased after first the 4 hr of incubation, however, their levels decreased following withdrawal of the hormone from the medium. Interestingly, the AHR3 mRNA level dropped more rapidly that that of VCP (Fig.7A and 7B). To determine the requirement of protein synthesis for activation and maintenance by 20E of the HR3 early-late gene in the mosquito fat body, the previtellogenic fat bodies were incubated with cycloheximide (Chx), a protein synthesis inhibitor which reversibly represses protein synthesis in this tissue in a dose-dependent manner (Deitsch eta1., 126 "-l—o— Chx/205.411 + Chx/20E —a— Chx --.--20E Relative mRNA Level (a) Culture Time (hr) -—O——Chx/20E-4h +ChXIZOE —o—Chx - - .- - -20E 120 '15 100 . I g vcp < 3° ‘ . ' ' 2 p' e .. .. o '. . 4o « .' e ,- g 20 " ' ' I ' _ _ . I o .W 0 4 8 12 16 (b) Culture Time (hr) Fig. 8. Effect of cycloheximide on the AHR3 induction by 20E in in vitro fat body organ culture. Accumulation of AHR3 mRNA (A) and VCP mRNA (B) during culturing with 10'3 M Chx in the hormone-free medium (Chx), in the continuous presence of 10’6 M 20E (Chx/20E), or in the 4 hr exposure to 20E followed by culturing in the hormone- free medium (Chx/20E-4hr). Dashed lines in (A) and (B), marked (20E), are controls from the previous experiment (Fig. 7). RT-PCR/Southem blot hybridization was performed using probes specific for either AHR3, or VCP as a late 20E-responsive gene. The results represent the average of three independent experiments j; SD. 127 l995a,b). The fat bodies were first pre-incubated in the culture medium for 1 hr in the presence of 10'3 M Chx, and then continued incubation with the same dose of Chx but with the addition of 10'6 M 20B for various times (Fig. 8). Incubation of the previtellogenic fat bodies in the presence of Chx in the hormone-free medium induced neither the AHR3 gene nor VCP. Addition of 10’6 M 20B to the Chx-containing medium activated the AHR3 gene in contrast to the VCP gene, which was inhibited by Chx. Furthermore, the AHR3 gene was induced to a higher level by Chx/20E than by 20E alone. Likewise, withdrawal of 20E from the Chx—containing medium after the first 4 hrs of incubation, caused a slower decline in the AHR3 mRNA level in fat bodies (Fig. 8A) than withdrawal of 20E from the Chx-free medium (Fig. 7A). DISCUSSION In this work, cloning and characterization of the mosquito nuclear receptor HR3 (AHR3) is reported. Analysis of the AHR3 deduced amino acid sequence showed that it was highly similar to its insect counterparts. The most conserved were the DNA-binding domain (DBD) and so-called carboxy-terminal extension (CTE) of the DNA-binding domain (F igs.l and 2). Giguere et al. (1994, 1995) have performed a detailed analysis of RORa nuclear receptor and have shown that this human HR3 courterpart binds DNA as a monomer, recognizing an asymmetric response element (RORE) composed of two distinct half-sites, a 6 bp AT-rich sequence at the 5’-end and a PuGGTCA core half-site motif at the 3’-end (Giguere et al, 1994). The specificity of RORa binding to the RORE is provided by amino acids from the P-box and the CTE: the P-box recognizes the core half-site motif, and a group of amino acids from the CTE recognizes the AT-rich half—site (Giguere et al, 1995). Subsequently, the presence of similar DNA-sites has been shown 128 for insect HR3s from Drosophila (Lam et al, 1997; Homer et al, 1995) and Choristoneura (Palli et al, 1996). The mosquito AHR3 had a highly conserved P-box and CTE, suggesting that it likely recognized a similar target DNA-site as a monomer. The ROR/HR3 transcription factors are classified as orphan nuclear receptors due to the lack of any known ligand (Mangelsdorf et al, 1995). The lack of ligand-dependent transactivation could lead to the relaxation in conservation of the orphan receptors’ LBDS, which in nuclear receptors are generally responsible for dimerization and ligand- binding functions. This may be one reason why putative LBD of AHR3 did not show a high similarity with those of nematode CeHR3 and human RORa counterparts (Giguere et al, 1994; Kostrouch et al, 1995), though it was still significantly conserved among HR3 receptors (Fig. 2). The high similarity between HR3s started in A/B-domains from the 7th amino acid preceding DBDs. Interestingly, an intron-exon junction (marked by arrowhead in Fig. 1) has been determined at this position after the structure of DHR3 and human RORa genes were characterized (Koelle et al, 1992; Giguere et al, 1994). The rest of the mosquito A/B domain was poorly conserved, as is consistent with the generally low conservation of highly variable nuclear receptor A/B domains. However, in the case of the ROR/HR3 subfamily, analysis of the A/B-domains merits special attention. In fact, cloning of three human RORa isoforms, which differ only in their A/B-domains, has revealed that the distinct A/B-domains modulate the DNA-binding specificity of each RORor isoform (Giguere et al, 1994; McBroom et al 1995). So far no similar HR3 isoform variation has been demonstrated in insects. In this context, it is interesting to note that in lepidopteran HR3 receptors from Manduca (Palli et al, 1992), Galleria 129 (Jindra et al, 1994b), and Choristoneura (Palli et al, 1996), isolated from larval stages of these insects, the A/B-domains are not only similar in size but also possess a high percentage of identical amino acids (Palli et al, 1996). The mosquito HR3 is the first HR3 isolated from an adult insect. It had a distinct A/B-domain and might represent a distinct adult-specific HR3 isoform. Although, HR3 was one of the first insect nuclear receptors to be cloned (Palli et al., 1992; Koelle et al., 1992), understanding of its role in regulating the ecdysone- mediated genetic network has been achieved only recently (Lam et al, 1997; White et al, 1997). These studies have demonstrated that the orphan nuclear receptor DHR3 is one of the key genes in the ecdysone-mediated genetic regulatory network. During Drosophila metamorphosis, DHR3 has a dual role in repressing the early genes while activating another orphan nuclear receptor, flFIZ—Fl , which has been implicated in providing competence for stage-specific responses to ecdysone (Lam et al, 1997; White et al, 1997). DHR3 homologues cloned from several Lepidopteran insects are highly conserved, and their expressions suggest HR3 involvement in mediating ecdysone response during Lepidopteran development and metamorphosis (Palli et al., 1992; 1996; Jindra et al., 1994b). Moreover, recent analysis of Drosophila DHR3 mutants revealed that DHR3 function is required to complete embryogenesis (Carney et al., 1997), prepupal-pupal transition and differentiation of adult structures during Drosophila metamorphosis (Lam et al., 1999). In this study, the novel evidence provided strongly suggests that the involvement of HR3 in the ecdysone gene regulatory hierarchy during female reproduction. 130 In insects, transcription of the HR3 gene is under the regulation of 20E (Homer et al, 1995; Palli et al, 1996;1indra et al, 1994b; Palli et al, 1992). Expression analysis of the mosquito HR3 concurs with the studies in other insects, which have previously shown the hallmarks of early-late gene expression, direct positive regulation of the HR3 expression by 20E followed by a sustained expression during the late ecdysteroid response. The in vivo developmental profiles of AHR3 in both fat body and ovary showed that the peak of the AHR3 transcript coincided with a. high titer of 20E in hemolymph. In in vitro cultured fat bodies, in contrast to the early gene AaE 75, which showed rapid peak activation by 20E at 2~4 hr of culturing (Pierceall et al., 1999), the AHR3 induction slowly and continuously increase in its mRNA level up to 16 hr of culturing. This pattern of AHR3 activation by 20E was consistent with observations on the successive puffing expression of the corresponding loci 7SB (encoding E 75) and 46F (encoding DHR3) in the polytene chromosomes (Ashbumer, 1972; Walker and Ashbumer, 1981), and with findings in cultured Drosophila organs (Homer et al., 1995) and salivary glands (Huet et al., 1995). Likewise, in the cultured silk glands of Galleria mellonella, 20E induction of GHR3 mRNA has been observed 2 hr after the peak of E 75A mRNA (Jindra and Riddiford, 1996). Our in vitro study suggested that delayed induction of the early-late HR3 gene was due to the relatively low sensitivity of its maximal response to 20E, which was more similar to late genes rather than to early genes. It seems likely that the high level of 20E needed for maximal response is required to trigger the expression of factors that contribute to the sustained induction, presumably through elements in addition to the EcREs responsible for early expression. However, 131 like other early-late genes, AHR3 is also directly regulated by 20E; in contrast to the late VCP gene, AHR can be activated by 20E in the absence of new protein synthesis. Previously, I have described on the expression and function of the ecdysone receptor genes AaEcR and AaUSP and the induction of the early ecdysone-inducible gene AaE 75 in conjunction with the initiation of vitellogenic ecdysteroid response. Here, this study demonstrates that conservation of ecdysone responsive genes, originally elucidated in development of immature stages of Drosophila, extends to the next level in this hierarchy, to the early-late genes involved in the transition from the early to the late ecdysone response. The expression pattern of the early-late gene AHR3 suggests that it may play a central role in mediating this transition during the vitellogenic response to ecdysone in the adult mosquito. ACKNOWLEDGMENTS I thank Marianna Kapitskaya for partial cloning of AHR3 and Northern analysis, Ken Miura for RT-PCR analysis, and William Segraves for critical reading. This work was supported by a grant from the National Institutes of Health (A136959). 132 Chapter 5* Cloning and Characterization of the Mosquito FTZ-FI Gene *Modified from the original publication in Dev. Biol. Ref: Li, C., Kapitskaya, M. Z., Zhu, 1., Miura, K., Segraves, W., Raikhel, A. S. (2000). Dev. Biol. 224, 96-110 133 ABSTRACT In the mosquito Aedes aegypti, the adult female becomes competent for a vitellogenic response to ecdysone after previtellogenic development. Here, BFTZ-F 1, the nuclear receptor implicated as a competence factor for stage-specific responses to ecdysone during Drosophila metamorphosis, serves a similar function during mosquito vitellogenesis. AaFTZ-Fl is expressed highly in the mosquito fat body during pre- and post-vitellogenic periods when ecdysteroid titers are low. The mosquito AaFIZ-FI transcript nearly disappears in mid-vitellogenesis when ecdysteroid titers are high. An expression peak of HR3, a nuclear receptor implicated in the activation of flF IZ-F I in Drosophila, precedes each rise in mosquito F IZ-F 1 expression. In in vitro fat body culture, AaFTZ-FI expression is inhibited by 20-hydroxyecdysone (20E) and super- activated by its withdrawal. Following in vitro AaFTZ—FI super-activation, a secondary 20E challenge results in super-induction of the early AaE 75 gene and the late target VCP gene. Electrophoretic mobility shift assays show that the onset of ecdysone—response competence in the mosquito fat body is correlated with the appearance of the functional AaFTZ-Fl protein at the end of the previtellogenic development. These findings suggest that a conserved molecular mechanism for controlling stage specificity is reiteratively used during metamorphic and reproductive responses to ecdysone. 134 INTRODUCTION Competence to respond to specific developmental signals allows the spatially and temporally restricted utilization of global signals within a multicellular organism. A signal molecule, which is spread throughout an organism via the circulatory system, elicits a specific response only from competent cells and tissues, leaving other parts of an organism unaffected. Recent studies have given rise to considerable insight into the molecular mechanisms underlying competence. One of the general means for accomplishing competence is the time- and cell-specific expression of a receptor or receptor cofactor needed to render a cell responsive to a corresponding hormonal signal (Shi et al., 1996; Puigserver et al., 1998). Regulation of competence can also be achieved through chromatin remodeling via involvement of linker histories, which repress transcription (Steinbach et al., 1997; Lee and Archer, 1998). Finally, the stage- specificity of a response to a hormonal signal can be determined by a specialized competence factor. Drosophila metamorphosis provides one of the best-studied examples of how competence affects the specificity of response to a hormonal signal. In this organism, the BFTZ-Fl orphan nuclear receptor functions as a competence factor which directs the developmental transition from larval to pupal-specific gene expression in response to the insect steroid hormone ecdysone (Woodard et al., 1994; Broadus et al., 1999) Mosquitoes serve as vectors for many harmful human diseases (Collins and Paskewitz, 1995; Butler et al., 1997; Beier, 1998) because they require blood feeding for their egg development. In the so-called anautogenous mosquitoes, vitellogenesis and egg maturation are initiated only after a female mosquito ingests vertebrate blood. A blood 135 meal triggers a hormonal cascade resulting in production of 20-hydroxyecdysone (20E), the terminal signal, which activates yolk protein precursor (YPP) genes in the fat body (Hagedom, 1983; 1985; Raikhel, 1992; Dhadialla and Raikhel, 1994; Deitsch et al., l995a;1995b) In the anautogenous mosquito Aedes aegipti, which is being used as a model vector, vitellogenesis proceeds through two periods (Fig. 1). The approximately 3-day- long previtellogenic development of both the ovary and fat body proceeds under the control of juvenile hormone (.1 H) 111 (Gwartz and Spielman, 1973; Flannagan and Hagedom, 1977). The JH titers are high during the previtellogenic period and drop immediately following the onset of vitellogenesis, which is initiated when the female mosquito takes a blood meal (Shapiro et al., 1986). The fat body then produces yolk protein precursors (YPP), which are internalized by the developing oocytes (Raikhel, 1992). In addition to vitellogenin (Vg), the major YPP, the mosquito fat body synthesizes two other YPPs, vitellogenic carboxypeptidase (VCP) and vitellogenic cathepsin B (VCB) (Cho et al., 1991; 1999). YPP synthesis reaches its maximal levels at 24 hr post-blood meal (PBM) and then proceeds until 30-32 hr PBM, when it is rapidly terminated (Raikhel, 1992). The titers of ecdysteroid in the mosquito female hemolymph are correlated with the rate of YPP synthesis in the fat body (Fig. 1). While the 20E titers are only slightly elevated at 4 hr PBM, they begin to rise sharply at 6—8 hr PBM, and reach their maximum levels at 18-24 hr PBM (Hagedom et al., 1975). Consistent with the initially proposed role of 20E in activating vitellogenesis in anautogenous mosquitoes (Hagedom et al., 1973; Hagedom and Fallon, 1973), experiments using an in vitro fat body culture have 136 rm N l—bJ 03 Z J; l-oo 12 6 g) 4 218312 3642 4854 go 6672 L J J Homiones 20F. 4— W 1" WIDE W Nuclear Receptors AaEcR M— A-USP- _— ,— A— AalTSPb AHR3 _ A L Late Genes Vs VCP er .2 3.81M {31.21.6302518323942485.46.06.672 Days Hours Fig. 1. Summary of events during the first cycle of vitellogenesis in the anautogenous mosquito, Aedes aegvpti. The previtellogenic period begins at eclosion (E) of the adult female. During first ~2-3 days of post-eclosion life, the fat body becomes competent for subsequent vitellogenesis. The female then enters a state-of-arrest; yolk protein precursors, vitellogenin (Vg) and vitellogenic carboxypeptidase (VCP) are not synthesized during this previtellogenic period. Only when the female takes a blood meal (BM), vitellogenesis is initiated. Hormones: hormonal titers of juvenile hormone (JH) and ecdysteroids (20E) in A. aeg’pti females (Hagedom et al., 1975; Shapiro and Hagedom, 1982). Transcript profiles of nuclear receptors (Nuclear Receptors) in the mosquito fat body were determined by RT-PCR/Southem blot; Transcript profiles of yolk protein precursors (Late Genes) in the mosquito fat body were determined by Northern blot and RT-PCR/Southem blot. AaEcR, ecdysone receptor; AaUSPa, Ultraspiracle isoform A; AaUSPb, Ultraspiracle isoform B (Wang et al., 2000); AaE75A, early gene AaE 75 isoform A (Pierceall et al., 1999); AHR3, early-late gene AHR3 (Kapitskaya et al., 2000); Vg, vitellogenin (Cho and Raikhel, 1992); VCP, vitellogenic carboxypeptidase (Cho et al., 1991). 137 shown that physiological doses of 20B (10’7 - 10'6 M) activate two YPP genes, Vg and VCP (Deitsch et al., 19953; 1995b). Utilization of the protein synthesis inhibitor, cycloheximide (Chx), has demonstrated that the activation of YPP genes in the mosquito fat body requires protein synthesis (Deitsch et al., l995a). Thus, it is likely that a regulatory cascade similar to that seen in Drosophila development mediates the action of 20E in the mosquito fat body. The functional ecdysteroid receptor is a heterodimer of the ecdysone receptor (EcR) and a retinoid X receptor (RXR) homolog, Ultraspiracle (USP) (Yao et al., 1992; 1993). In the mosquito Aedes aegypti, one isoform of ecdysone receptor (AaEcR) and two USP isoforms, AaUSP-A and AaUSP—B have been cloned (Cho et al., 1995; Kapitskaya et al., 1996). The mosquito EcR/U SP heterodimer has been shown to bind to various ecdysone response elements (EcREs) to modulate ecdysone regulation of target genes (Wang et al., 1998). A mosquito homolog of the Drosophila early gene E 75 is involved in mediating the ecdysteroid response during vitellogenesis (Pierceall et al., 1999). Furthermore, a highly conserved homolog of the DHR3 orphan nuclear receptor previously implicated in activation of Drosophila flFTZ-Fl is expressed in vitellogenic tissues, the fat body and ovary (Kapitskaya et al., 2000). A preparatory, developmental, previtellogenic period is required for the mosquito fat body to attain competence for ecdysone responsiveness (Sappington and Raikhel, 1999). F lannagan and Hagedom (1977) have demonstrated that the acquisition of competence for ecdysone response in the mosquito fat body is under the control of juvenile hormone 111. However, the molecular mechanisms underlying the acquisition of sensitivity to ecdysone in the mosquito fat body remain obscure. 138 Here this chapter reports that BFTZ-F l , the orphan nuclear factor implicated as a competence factor for stage-specific responses to ecdysone during Dr050phila metamorphosis, likely serves a similar function during mosquito vitellogenesis. A homolog of Drosophila MTZ—FI is expressed relatively highly in the mosquito fat body during pre- and post-vitellogenic periods when ecdysteroid titers are low. In in vitro fat body culture, AaFTZ—FI expression is inhibited by 20B and super-activated by its withdrawal. Moreover, afier the in vitro super-activation of AaF 72-F1 expression in the previtellogenic mosquito fat body, a secondary 20E challenge results in super-induction of the early AaE 75 gene and the late target VCP gene. Although AaFIZ—FI mRNA is abundant even in newly emerged mosquitoes, electrophoretic mobility shift assays show that it is the appearance of functional AaFTZ-F 1 protein that correlates with the onset of ecdysone-response competence in the mosquito fat body. Our findings suggest that the regulation and function of F TZ-F 1 during mosquito reproduction closely resemble those shown at the onset of Drosophila metamorphosis, and that FTZ-Fl may therefore be part of a conserved and broadly utilized molecular mechanism controlling the stage specificity of ecdysone responses. MATERIALS AND METHODS Animals Mosquitoes, Aedes aegypti, were reared according to the method of Hays and Raikhel (1990). Larvae were fed on a standard diet as previously described (Lea, 1964). Vitellogenesis was initiated by allowing females, 3-5 days after eclosion, to feed on an anesthetized white rat. 139 Materials The RNA ladder was purchased from Gibco—BRL; Sequenase, from U. S. Biochemical Corp.; restriction enzymes, from Boehringer Mannheim. All primers used in PCR were ordered from Gibco-BRL. Superscript II reverse transcriptase and Taq DNA polymerase were from Gibco-BRL; random hexamer primers, from Promega. Perkin-Elmer was the source of reagents for the polymerase chain reaction (PCR). MSI CO and Amersham Pharrnacia Biotech supplied nitrocellulose and Hybond-N+ blotting membranes, respectively. Radionucleotides for labeling probes and DNA sequencing were from NEN Life Science Products. All other reagents were of an analytical grade from Sigma or Baker. cDNA cloning Degenerate primers based on the sequences of F TZ—F 1 insect homologs were used for PCR to obtain a mosquito homolog cDNA fragment. Amplification was carried out in a Perkin Elmer thermal cycler using template cDNA produced by reverse transcription from total RNA prepared from the fat bodies of vitellogenic female mosquitoes. The PCR-generated fragment was used as a probe to screen a XZAPII cDNA library prepared from the fat bodies of vitellogenic female mosquitoes during 6-48 hr post-blood meal (PBM) as previously reported (Cho and Raikhel, 1992). Several positive cDNA clones were subsequently isolated and sequenced at W.M. Keck Foundation Biotechnology Resource Laboratory, Yale University. Analyses of nucleotide and deduced amino acid 140 sequences were performed using the software of GCG (Genetics Computer Group, University of Wisconsin, Madison). Northern blot analysis Total RNA was isolated by using the guanidine isothiocyanate method as described previously (Bose and Raikhel, 1988), or using TRIZOL Reagent (Gibcol- BRL). Polyadenylated mRNA was isolated using Niomag oligo (dT)20 magnetic beads and the manufacturer’s protocols (PerSeptive Diagnostics, Inc.). For Northern blot analysis, total or poly(A) RNA was separated by electrophoresis in 1.2% agarose/formaldehyde gels in MOPS buffer, blotted to nitrocellulose membranes, and hybridized with 32P-labeled DNA probes (Random Primers DNA Labeling Systems, Gibco-BRL) under high stringency conditions. Autoradiography was conducted at -80°C with intensifying screens. RT -PCR/Southern blot RT-PCR/Southem analyses were performed basically as previously described (Pierceall et al., 1999). Total RNA was prepared from fat bodies throughout the first vitellogenic cycle, as described in the Northern blot procedure above, with the modification that all isopropanol precipitation steps were done without low-temperature incubation, to avoid co-precipitation of glycogen and salt. RNA concentration was determined spectrophotometrically; absorbance ratios, A260/A280 and A260/A230, in RNA preparations were always above 1.7 and 2.0, respectively. Five microgram aliquots of each RNA preparation were reverse-transcribed by Superscript II reverse transcriptase 141 (Gibco-BRL) and random hexamer (Promega) in a reaction volume of 20 it]. After this reaction, the volume of each sample was brought to 40 ul with TE buffer (10 mM Tris- Cl, lmM EDTA, pH 8.0). cDNA samples were stored at -200C until use. In the analyses of developmental profiles, 0.025 fat body- or pupa-equivalent cDNA pool was used as a PC R template for each point. Thermal cycling conditions were as follows: 940C for 3 min, followed by 18 cycles of 940C for 30 sec, 600C for 30 sec and 720C for 30 see. As a reference to the developmental change of the fat body, the samples of the same cDNAs were subjected to the 13 cycles of PCR with a primer pair specific to the mosquito vitellogenic carboxypeptidase (V CP). The reaction was followed by Southern blot hybridization using a VCP specific probe, which was generated by PCR using cloned VCP cDNA as a template (Cho et al., 1991). The PCR primers for each gene examined in this paper were listed in Table 1. Table 1 Primers Forward Reverse AaE75A TAGTGCAATCAACGTATACCAATC GGCACACGACCGAATCTGAC AaE7SB CGTGGAAGAAGACCACGATCG GGCACACGACCGAATCTGAC AHR3 GTGTGCGGCGACAAGTCGTC TCGACCGAATTTGACCGCGTC AaFTZ-Fl TGAAGGTGGACGACCAGATGAA TGTTCTGCAGCTCGTTGAAGTG VCP AGCGCCCATTCTTGTTTGG C AGCTCATACAGGTATTCTCC V g TAC TTGAAGAC AAGATGC TAGC G CGTTCTTGTAACTGTAGCCTTG 142 RT-PCR/ Southern blotting of total RNA preparations without reverse transcriptase did not result in any appreciable signal, indicating that contamination of RNA preparations with genomic DNA fragments was virtually negligible (not shown). As a quantitative control for the RT-PCR, each cDNA was diluted to 1/2, 1/4, 1/8, and 1/16, and PCR amplifications of the cDNA dilutions were monitored by different PCR cycles ranging from 13 to 22 cycles followed by Southern blot quantification. The analysis of normalized results showed that RT-PCR amplifications were linear (not shown). To further test whether results of the RT-PCR/Southem analyses correlate with those from the Northern blot analysis, both of these techniques were used in parallel to measure the VCP mRNA levels in fat bodies during the vitellogenic cycle as well as over the course of in vitro treatments with 20-hydroxyecdysone. VCP mRNA levels were analyzed by either Northern blot or RT-PCR/Southern blot analyses. In both cases, the results were quantified using a phosphorimager (Molecular Dynamics). The Northern blot and RT-PCR/Southem blot analyses yielded very similar results (data not shown). In vitro organ culture The abdominal walls with adhering fat bodies (hereafter referred to as the fat body) were dissected from 0~0.S-day or 3~5-day-old previtellogenic females and incubated in an organ culture system as previously described (Raikhel et al., 1997). For the stage-specific responses to 20E, the dissected fat bodies were exposed to 10'6 M 20B for 6 hr; for all other cultures, the fat bodies were incubated with 20E either for 16 hr, or for 4 hr with 20E followed by washing three times with hormone-free medium and an additional 12 hr incubation in hormone-free medium. At every 4 hr interval, a sample of 143 nine fat bodies was collected and stored in liquid nitrogen until use. As a control, the fat bodies were incubated in hormone-free medium. RT-PCR/Southern blot analyses were utilized as described above. Hybridization using probes for the ecdysone-inducible VCP gene was provided as a positive control. To test the effect of the protein synthesis inhibitor cycloheximide (Chx) on the expression of each gene, dissected fat bodies from 3-5 day old female mosquitoes were pre-treated with culture medium containing 10'4 M Chx for 1 hr, then further incubated with the same concentration of Chx either with or without 10'6 M 20E. The C hx/hormone withdrawal experiment was conducted as described previously in the hormone withdrawal experiment above except that 104 M Chx was added to all culture media. The experiments of secondary responses to 20E or Chx/20E in cultured fat bodies were an extension of the 20E-withdrawal experiments described above. After the initial 4 hr exposure to 20E followed by 12 hr culturing without 20E, the fat bodies were exposed again to 20E, or pre-treated with Chx for 1 hr and then exposed to 20E/Chx for an additional 4 or 16 hr. In addition to these two experiments, as the third experiment, after pre-treatment with Chx for 1 hr and the initial 4 hr exposure to 20E/Chx followed by 12 hr culturing with Chx only, the fat bodies were exposed again to 20E/Chx for an additional 4 or 16 hr. RT-PCR/Southem blot were utilized to analyze the responses of each gene under investigation. 144 Electrophoretic mobility shift assays Electrophoretic mobility shift assays (EMSA) were carried out as described previously (Wang et al., 1998). The nucleotide sequence of the FIRE used in EMSA is 5’GCAGCACCGTCTCAAGGTCGCCGAGTAGGAGAA3’ (Ohno et al., 1994). The FIRE oligo was purified before EMSA. The in vitro synthesized AaFTZ-FI protein was made by the TNT system (Promega). Fat body nuclear extracts (FBNE) were prepared as previously described (Miura et al., 1999). The DmFTZ-Fl antibody was a generous gift from Dr. Carl Thummel (University of Utah). RESULTS E cdysone-responsive vitellogenic competence in the mosquito fat body Previously, Flanagan and Hagedom (1977) have shown that the fat body of newly emerged female mosquitoes is not responsive to 20E in vitro with respect to vitellogenin protein synthesis. However, responsiveness to this hormone can be detected in the fat body as early as 12 hr post-eclosion. Furthermore, the levels of the 20E-activated Vg synthesis in the fat body in vitro reach its maximum in 36-48-hr-old mosquitoes. Here, I utilized the fat body from mosquitoes of different post-eclosion ages and monitored the appearance of mRNAs for two YPP genes, Vg and VCP, in response to 20E in vitro. These experiments confirmed that the fat body of a newly emerged mosquito was not competent to respond to hormonal stimulation and that the YPP genes were not activated (Fig. 2). In contrast, both Vg and VCP genes were expressed in the fat body of 3-5 day old females treated with 10‘6 M 20B in vitro (Fig. 2). Moreover, the RT-PCR analysis of both Vg and VCP mRNAs showed that the ability of the YPP genes to respond to 145 0-0.5d 3-5d + - + - 20E 6.5kb « urn V9 1.5kb - ' , VCP _..... Fig. 2. Northern blot analysis of stage-specific induction of the vitellogenin (Vg) and vitellogenic carboxypeptidase (VCP) transcripts by the hormone 20E. Nine previtellogenic fat bodies of different stages were cultured in the presence (+) or absence (-) of 10‘6 M 20B for 6 hr. Total RNA from 3 fat bodies (~10 ug) was loaded in each lane. Hybridization was performed with the 32P- labeled Vg or VCP probe (see Materials and Methods). Ribosomal RNA (rRNA) stained with ethidium bromide is shown as a loading control. 0-0.5d: fat bodies from newly eclosed previtellogenic females, 0-12 hr post-eclosion; 3-5d: fat bodies from previtellogenic females, 3—5 day post-eclosion. 146 hormonal activation increased gradually over the previtellogenic period from 1 to 3 day post-eclosion (not shown). These data indicated that the ecdysone-responsive vitellogenic competence in the mosquito fat body was under transcriptional control. Isolation and characterization of a cDNA clone encoding the mosquito AaF T Z- F1 nuclear receptor To investigate whether the mosquito homolog of the competence factor BFTZ-Fl could be involved in the control of competence in the mosquito fat body, a mosquito homolog of Drosophila BFTZ-F l was cloned. The initial isolation of a cDNA fragment encoding the mosquito F IZ-F I homolog was achieved through a PCR-based approach. A pair of degenerate PCR-primers used to amplify cDNA of the mosquito F IZ—F 1 homolog is shown in Fig. 3. After a mosquito PCR-fragment of the appropriate size, 266 bp, was sequenced and confirmed to encode a polypeptide of the expected sequence, it was used as a probe to screen a vitellogenic mosquito fat body cDNA library. Several positive clones were identified, the largest of which contained an insert of 4.6 kb. The first methionine residue at the N-terminal region of the ORF (811 aa) was located at 1.4 kb downstream of this cDNA clone. Before the first Met, there were several in-frame stop codons in the 5’UTR, suggesting that the first Met was the translation start site. There was a poly (A) tail at the 3’UTR end of this clone. The conceptual translation of AaFTZ-Fl amino acid sequence revealed distinctly high similarity with its insect counterparts of Drosophila and Bombyx (Lavorgna et al., 1991; 1993; Sun et al., 1994), particularly in the DBD and the adjacent stretch of 29 amino acids known as the F TZ-Fl box (Fig. 3). According to recent data, members of 147 Fig. 3. Alignment of the deduced amino acid sequence of the mosquito AaFTZ-FI cDNA with its insect homologs: DmBFTZ-Fl from Drosophila (Lavorgna et al., 1993) and BmFTZ-F I from Bombyx (Sun et al., 1994). Asterisks or carets indicate amino acids which are identical or similar, respectively. The borders of DNA-binding domain (DBD) and putative ligand-binding domain (LBD) are marked with square braces. The P-box in the DBD is in bold. The F TZ-F 1 -box is double-underlined. Two vertical arrowheads flank the non-conserved insertions in the hinge-regions of insect FTZ-F ls. Horizontal arrows indicate sequences corresponding to the degenerate sense primer (Pr-l) and antisense primer (Pr-2) used for AaFTZ—Fl cDNA cloning. A black dot marks an EcoRV restriction site. Two other arrows represent the regions of the sequence-specific sense (Pr-3) and antisense (Pr-4) primers utilized in RT-PCR analysis of AaF TZ-F I . The Pr-4 was also used to generate a single-stranded DNA-probe for Northern blot (see Fig. 4A). Most part of A/B domains in FTZ-Fls are not shown due to very low similarity. The complete sequence of mosquito AaFTZ—Fl has been submitted to the GenBank (http://wwwngbi.nlm.nih.govl; its accession number is AF274870. 148 m2 -r1 mane 4'1 m2 4'1 AaFTZ-FI mflnz -F1 BmFTZ-FI MHZ-F1 mflnz -F1 BmFTZ-Ffi m2 -1"1 magma-F1 BmFTZ-Fi amz 4'1 MHz-r1 amnz -r'1 m2 -r'1 mflnz -F1 mz-n MHZ-F1 mgr-1'2 -1r1 BmFTZ-Ffl mz-n mBHz-m BmFTZ-Ffl m2 -r1 mama-F1 m2 -r'1 m2 -r1 magma-r1 mam-r1 l—* DBD Pr-l I P-box 335 ELCPVCGDKVSGYHYGLLTCESCKGFFKRTVQ 281 ELCPVCGDKVSGYHYGLLTCESCKGFFKRTVQ 87 ELCPVCGDKVSGYHYGLLTCESCKGFFKRTVQ ******************************** 4—1 NKKVYTCVAERQCHIDKTQRKRCPYCRFQKCLEVGMKLEAVRADRMRGGRNKFGPMYKRDR NKKVYTCVAERSCHIDKTQRKRCPYCRFQKCLEVGMKLEAVRADRMRGGRNKFGPMYKRDR NKKVYTCVAERACHIDKTQRKRCPFCRFQKCLDVGMKLEAVRADRMRGGRNKFGPMYKRDR ***********A************A******ir"**************************** ARKLQIMRQRQLAIQALRGSIG-GGVGIGQLGSDGASAQLQGIDYHQPYSNMHIKQEIQIP ARKLQVMRQRQLALQALRNSMGPDIKPTPI--SPG --------- YQQAYPNMNIKQEIQIP ARKLQMMRQRQIAVQTLRGSLGDGGLVLG-FGSP -------------- YTAVSVKQEIQIP *****A*****A*A*A**A*A* * * ******* — V' o QVSSLTSSPDSSPSPIAIALGQVNPQTTISLSSGVGGNGSSGGGSGGGGSNNSSGAGTGGS QVSSLTQSPDSSPSPIAIALGQVNASTGGVIATPMNAGTGGSGGGGLNGPSSVGNGNSSNG QVSSLTSSPESSPGP---ALLRAQPQPPQPPPPPTHDKWEAH ------------------- ******A**A***A* ** V NSTSTAIQNQLSESKLWMSANSTTA ---------------------------- SPHSLSPK SSNGNNNSSTGNGTSGGGGGNNAGGGGGGTNSNDGLHRNGGNDSSSCHEAGIGSLQNTADS ————————————————————————————————————————————————————— SPHSASPD *AAA AFSFDSGTN—---PTSTADSGNPADTLRVSPMIRDFVQSIDDREWQTSLFSLLQSQSYNQC KLCFDSGTH----PSSTADA--LIEPLRVSPMIREFVQSIDDREWQTQLFALLQKQTYNQV AFTFDTQSNTAATPSSTAEA-TSTETLRVSPMIREFVQTVDDREWQNALFGLLQSQTYNQC *A***A’\ A *‘k‘k‘k****A***AA****‘k'kAA‘k'kA'k‘kir/‘i/‘ir'k‘k r——> LBD “'3 EVDLFEL-MCKVLDQNLFSQVDWARNTIFFKDLKVDDQMKLLQNSWSDMLVLDHIHQRLH EVDLFELLMCKVLDQNLFSQVDWARNTVFFKDLKVDDQMKLLQHSWSDMLVLDHLHHRIH EVDLFEL-MCKVLDQNLFSQVDWARNTVFFKYLKVDDQMKLLQDSWSVMLVLDHLHQRMH ******* *******************A*** *‘k*********"*** ******A*A*A* Pr-4 NGLPDETTLHNGQKFDLLGLGLLGVPSLABHFNELQNKLQELKFDVGDYICMKFLLLLNP NGLPDETQLNNGQVFNLMSLGLLGVPQPGDYFNELQNKLQDLKFDMGDYVCMKFLILLNP NGLPDETTLHNGQKFDLLCLGLLGVPSLADHFNELQNKLAELKFDVPDYICVKFMLLLNP ******* *A'k‘kir *A*’\ *******A AAA‘k‘ki'k'k'k‘k'ki'A‘k‘k'k‘kA **A*A**AA**** A **A AA VEVRGITNRKTVVEGYENVQAALLDYTLTCYPSVPDKFSKLLSIIPEIHAMATRGEEHLY —SVRGIVNRKTVSEGHDNVQAALLDYTLTCYPSVNDKFRGLVNILPEIHAMAVRGEDHLI -EVRGIVNVKCVREGYQTVQAALLDYPYL-LSTIQDKFGKLVMVVPEIHALRL-GEKSTC ****A* * * **AAA******** AA *** *A AA*****A ** IKHCAGSAPSQTLLMEMLHAKRK 840 TCTPSTVPAVRPPKRCSWRCCTPSARDRGRENVTRNT 816 TSGIVQARHLPRLFSWKCCTQNANLEVPVTNKVEELRSAKPRRHHNK 555 149 366 312 118 Pr—2 427 373 179 487 423 225 548 484 264 581 545 272 638 600 332 697 660 391 757 720 451 817 779 508 the FTZ-F l subfamily bind as a monomer to a 5’-PyCAAGGPyCPu-3’ DNA response element. The P-box from the DBD recognizes the core motif 5’-AGGPyCPu-3’, and the F TZ-F 1 -box recognizes the first three base pairs 5’—PyCA-3’ of the target DNA-element (Ueda and Hirose, 1991; Wilson et al., 1993; Ueda et al., 1992). The mosquito AaFTZ- F 1 had a P-box with conserved CfiCKfi amino acids, which were characteristic for members of the FTZ-F 1 subfamily. The DBD and FTZ-F 1 box of AaFTZ-Fl were highly conserved relative to the corresponding sequences in insect homologs, and also shared a high similarity with those in vertebrate FTZ-F l homologs, exhibiting 83% identity with mouse LRH-l (Tugwood et al., 1991) and 85% identity with bovine Ad4BP (Honda et al., 1993). In the hinge subdomain, which connects the F TZ-Fl box and the putative LBD, significant conservation was retained only among insect FTZ-Fls. The conserved sequences were interrupted by a non-conserved spacer sequence (marked with vertical arrowheads in Fig. 3) in each insect receptor. The A/B-domains of the insect FTZ-Fl receptors were quite divergent (not shown). Comparison of the mosquito LBD with the LBDs of insect and non-insect FTZ-Fl homologs showed that there was limited identity in LBDs between mosquito FTZ-F 1 and its vertebrate counterparts, exhibiting only 46% identity with mouse LRH-l and 42% with bovine Ad4BP. However, a high level of identity was retained in the LBDs of insect FTZ-F ls, showing 70% with DmFTZ-F l and 66% with BmFTZ-Fl. 150 Temporal expression of A aF T Z-F I in the mosquito fat body during the first vitellogenic cycle To determine the pattern of AaF 72-F1 transcripts in the female fat body, Northern blot was performed for analysis of poly(A) RNA isolated from 100 fat bodies at each time point. Three critical time points during mosquito vitellogenesis were chosen: the early previtellogenic stage at 1 day post emergence (Fig. 4A, PV), the late vitellogenic stage at 36 hr PBM (Fig. 4A, PBM 36h) when the 20E titers are low, and the middle vitellogenic stage at 18 hr PBM (Fig.4A, PBM 18h) when 20E is at the peak level. The 10 kb and 5.2 kb AaFTZ-FI mRNAs were detected in the fat body of 1d PV and 36h PBM mosquitoes; in contrast, the fat bodies at 18 hr PBM contained no detectable AaFTZ-Fl message (Fig. 4A, PBM 18h). In order to examine the AaFIZ-FI developmental profile in this vitellogenic tissue in more details, a more sensitive RT-PCR analysis was utilized. First, RT-PCR was conducted by two pairs of primers located in the 5’UTR or between the hinge region and LBD, and it appeared that the expression patterns of AaFTZ-F 1 during vitellogenesis were the same (not shown). In the later experiments, the primer pair in the hinge region and LBD was chosen. As demonstrated in Fig. 4B, the level of the AaF 72-F1 transcript was high in the fat bodies of newly eclosed females, and its level declined slightly over the course of previtellogenic development. After the onset of vitellogenesis, triggered by a blood meal, the AaF 72-F1 transcript level dropped dramatically during the first 4 hr PBM and then stayed at background levels until 24 hr PBM. Then, it rose again, reaching the previtellogenic levels at 36 hr PBM, and stayed at a high level for the next 12 hr of post-vitellogenesis. Previously, it has been demonstrated that the AHR3 gene is 151 A __EEM_. PV 18h 35h 1on1: d R 5.2kb —‘ “e. 8 PV (d) PBM (h) 0-1 1-2 3-5 1 3 4 6 15 24 33 48 b n - “ — ~— “-flj FTZ-F1 i c l . «an. - VCP C 0 6 12 1 8 24 30 36 (h) "-1 * m... Fig. 4. AaFTZ-FI expression profiles in the female mosquito fat body and pupae. (A) Northern blot analysis of AaF TZ—F 1 in fat bodies. Poly(A) RNA was extracted from fat bodies of 100 female mosquitoes at each point, PV: previtellogenic stage 0-1 day post-eclosion; PBM 18h or 36h: post blood meal 18 or 36 hours. The region between EcoRV site and Pr-4 (Fig.3) of AaFTZ-Fl was used as a probe for hybridization. (B) Temporal expression of AaF IZ-F I in the fat body during the first vitellogenic cycle by RT-PCR/Southem analyses. A. The early-late gene AHR3 expression profile. The 0.22 kb PCR product was generated with the AHR3-specific primers and hybridized with a 32P-labeled AHR3 specific DNA probe (see Methods and Materials). B. AaFTZ-Fl expression profile. The 0.2 kb PCR-fragment was generated with the AaFTZ-FI-specific primers Pr-3 and Pr-4 and hybridized with a 32P AaFIZ-FI specific probe (shown in Fig. 3). C. Expression profile of the mosquito vitellogenic carboxypeptidase (VCP) (Cho et al., 1991). VCP is shown as an independent control for RT-PCR/Southem analyses of an ecdysone-inducible late gene in the mosquito fat body. (C) Temporal expression of AaF TZ—F 1 during pupa development analyzed by RT-PCR/Southem analyses. Expression profiles of AHR3 (HR3) and AaFTZ—FI (F TZ-F l) are shown. Each lane contained the same mosquito pupa equivalents of total RNA subjected to RT-PCR/Southem analyses. 152 expressed in a narrow window correlated with the high levels of ecdysteroid at 18-24 hr PBM in the mosquito fat body (Kapitskaya et al., 2000). The post-vitellogenic rise in AaFTZ-FI mRNA levels occurred immediately after the AHR3 mRNA peak and in correlation with a drop in the ecdysteroid titer (Fig. 4B). The high level of the AaFIZ-FI transcript in the fat body of the early previtellogenic females suggested that it might be activated prior to emergence by a falling ecdysteroid titer in late pupae. To test this hypothesis, RT-PCR was performed for analysis of the levels of A aF TZ-F 1 and AHR3 transcripts during development of female mosquito pupae. Developing female pupae were collected at 6 hr intervals, from 0 hr (newly-pupated) to the final collection of late pupae at 36 hr post-pupation (adult mosquitoes normally emerge within the next 6 hr). RT-PCR analysis was then performed using RNA collected at different intervals of pupal development. AHR3 transcript levels increased during early and mid-pupal stages, reaching its peak in 24 hr old pupae. By 30 hr, the AHR3 transcript level decreased dramatically and remained low in late pupae. The A aF TZ-F 1 transcript level exhibited a sharp increase in 18-24 hr old pupae and remained at the elevated levels until the end of pupal development (Fig. 4C). Dual effects of 20E on AaF T Z—F 1 mRNA in cultured mosquito fat bodies To determine whether the negative correlation between AaFTZ-FI expression and the levels of ecdysteroids observed in vivo reflected the involvement of 20E in regulation of the A aF IZ-F 1 gene, I investigated the patterns of AaF TZ—F 1 transcript accumulation in in vitro cultured fat bodies. Previtellogenic fat bodies, 3-5 days after eclosion, were incubated for up to 16 hr in culture medium either in the presence or absence of 10'6 M 153 20E. RT-PCR analysis was used to monitor the transcript levels of AaF IZ-F 1 in the cultured organs (see Material and Methods). Fat bodies from previtellogenic 3-5 day-old females were used to determine the background levels of transcripts for each ecdysone- regulated gene including two isoforms of an early gene E 75 (AaE 75A and AaE75B), an early—late gene AHR3, and a late gene VCP. Culturing the previtellogenic fat bodies in hormone-free medium up to 16 hr did not induce AaE 75, AHR3, or VCP mRNAs (Fig. 5). The AaF 72-F1 mRNA level, which was relatively high in the previtellogenic fat body, was not effected by the 16 hr incubation in hormone-free medium (Fig. 5, FTZ-F 1). In contrast to A aE 75, AHR3, and VCP genes, which were induced by the presence of 20E in culture medium, the AaFTZ-FI mRNA dropped to an undetectable level after a 12 hr incubation with 10'6M 20E (Fig. 5, 2013). In the next experiment, fat bodies were cultured in hormone-containing medium for only 4 hr, then washed three times in hormone-free medium and incubated in hormone-free medium for an additional 12 hr. Both AaE 75A and AaE75B levels dramatically increased after a 4 hr incubation in 20E-containing medium, and then dropped sharply following the withdrawal of the hormone from medium (Fig. 5, E75A, E75B). AHR3 and VCP mRNA levels, which rose moderately during 4 hr incubation with 20E, also declined (Fig. 5, HR3, VCP). In contrast, the AaFIZ-FI mRNA exhibited a considerable elevation of its level following the withdrawal of 20E from the incubating medium (Fig. 5, FTZ-Fl). To determine the requirement of protein synthesis for 20E-dependent activation or repression of the AaF 72-F1 gene in the mosquito fat body, the previtellogenic fat bodies were incubated with cycloheximide (Chx), a protein synthesis inhibitor which reversibly 154 120 100« E75A E 80 % +205.“ g 60 +205 5 +0117! 2 40 82 20 ° - =- — L‘543—J o 4 120 o 12 16 < z o: — —o—205.4n g +205 :5 p—o—cm 33 100. g so 5 41—20541 g 60 +205 é —a—CM 40 a? 20« 0 120 g 100 FTZ-F1 a: 801 E 60 i 40 39 20. 0 0 4 a 12 1a 120 100 g VCP {E 5° +205.“ 2 60 +205 g —o—CM 5 40. BE 20 o .. er - a o 4 a 12 16 Culture Tlme (hr) Fig. 5. In vitro repression of AaF TZ—F 1 expression by the hormone 20E, and super- induction of AaF T Z-F I by withdrawal of 20E in fat body organ culture. RT- PCR/Southem analyses were performed for each ecdysone-regulated gene in mosquitoes as follow: early gene AaE 75A (E75A) and AaE75B (E75B) isoforms (Pierceall et al., 1999); early-late gene AHR3 (HR3) (Kapitskaya et al., 2000); AaFIZ-FI (this paper); late gene VCP (VCP) (Deitsch et al., l995a; l995b). For the in vitro fat body culture and RT-PCR/Southem analyses, see Methods and Materials. Closed squares represent a continuous treatment with 20E (20E); open squares, a 4hr treatment with 20E followed by an additional culture in 20E-free medium (20E-4h); open triangles, a control medium without 20E (CM). Each point represents an average of three independent experiments : SD. 155 represses YPP protein synthesis in this tissue in a dose-dependent manner (Deitsch et al., l995a; 1995b). The fat bodies were first pre-incubated in hormone-free culture medium for 1 hr in the presence of 104M Chx, and then incubated with the same dose of Chx but with the addition of 10'6M 20E for various times (Fig. 6). While incubation of the previtellogenic fat bodies in the presence of Chx in the hormone-free medium did not induce AaE 75, AHR3, and VCP mRNAs, the AaFIZ-FI transcript levels were considerably elevated (Fig. 6, Chx). Addition of 10‘6 M 20E to Chx-containing medium activated A 0E 75 and AHR3 genes to the levels much higher than those attained with 20E alone; the levels of AaF TZ-F l mRN A were reduced compared with those in hormone- free, Chx-containing medium (Fig. 6, Chx/20E). Withdrawal of 20E from Chx- containing medium after 4-hr incubation caused a slower decline in the AaE 75A, AaE75B and AHR3 mRNA levels than 20E withdrawal from Chx-free medium (Fig. 5, 20E-4h; Fig. 6, Chx/2OE-4h). Interestingly, the AaFTZ—F! gene was super-induced under the same conditions (Fig. 6, FTZ-Fl). The V CP gene was not responsive to any stimulation in the presence of Chx (Fig. 6, VCP). In vitro enhancement of ecdysone-responsive vitellogenic competence following over-expression of AaF T Z-F I Withdrawal of 20E from the fat body culture after an initial 4 hr incubation in the presence of 20E resulted in a considerable elevation of AaF TZ—F 1 mRNA (Fig. 5, FTZ- Fl, 20E-4h). If AaFTZ-F l is a competence factor for 20E sensitivity, one might expect that these conditions should lead to enhanced responsiveness of the fat body to 20E. To test this hypothesis, I re-introduced the fat body, after treatment for 16 hr as in Fig. 5 156 Relative RNA Level ——o-— Chx/20E-4h —.—— Chx/206 Relative RNA Level -—-O— Chx/2054b + Chx/ZOE —o— Chx - - O - -ZOE Relative RNA Level Relative RNA Level —-o-—Chx/205-4h +0hx/206 —o—-Chx - - D - -20E --.'°2OE —o— Chx/206411 + Chx/205 —o— Chx E VCP —’ 801 .5. 1" CE 50 .‘ O .' E. 40« 8 -' Ct 20 .' 0‘... O -" - ‘ m 0 4 8 12 Culture Tlme (hr) Fig. 6. Effects of cycloheximide (Chx) on the 20E regulation of AaF TZ—F 1 in in vitro fat body organ culture. The mRNA levels of each AaE 75A (E75A), AaE7SB (E75B), AHR3 (HR3), AaFIZ-FI (FTZ-Fl) or VCP (VCP) gene were examined during culturing with 104 M Chx in hormone-free medium (Chx), in the continuous presence of 104 M Chx and 10'6 M 20B (Chx/20E), or in the 4 hr exposure to 10'4 M Chx and 10'6 M 2013 followed by culturing witth“1 M Chx in hormone-free medium (Chx/20E-4h). Dashed lines (20B) are from the previous experiment (Fig. 5, 20E), and all other data are relative to these values. RT-PCR/Southem blot hybridization were performed using a probe specific for each 20E-responsive gene. Each point represents an average of three independent experiments i SD. 157 16 —o—- Chx/2064b —-.-— Chx/20E -——o—— Chx --..-.205 (20E-4h) with the elevated level of AaF IZ-F 1 mRNA, to a 20E-containing medium and examined its capacity for a secondary 20E-activation of the early E 75, early-late HR3, and late VCP genes (Fig. 7). In the first experiment (Fig. 7A), the tissue was incubated in the presence of 10'6 M 20E for 4 hr or for 16 hr. The 4-hr secondary incubation resulted in super-induction of the early AaE 75 gene, with the level of AaE 75A mRNA more than three-fold higher than that seen in the 4-hr primary 20E-incubation (Fig. 7A). The HR3 and VCP transcript levels were also increased compared to the primary incubation. However, their levels increased even further after a 16-hr secondary incubation. In particular, this secondary incubation resulted in a 2-fold induction of the late VCP gene compared to that in the primary 20E incubation (Fig. 7A). Thus, this test suggests that the fat body with the experimentally increased level of AaF 72-F1 mRN A was competent to activate the early, early-late, and late 20E-responsive genes to a much higher level. Next, I modified the second part of the experiment. As in the first experiment, the previtellogenic female fat bodies were first incubated in 20E-containing medium for 4 hr, followed by 12 hr incubation in the hormone-free medium. However, the secondary 20E incubations were performed in the presence of 10‘4 M Chx (Fig. 78). Under these conditions, A aE 75A showed even higher induction after 4-hr incubation (8-fold). Furthermore, AaE 75A transcript continued accumulation reaching the l6-fold level of increase after 16-hr incubation. Likewise, the early-late HR3 gene transcript showed continuing accumulation. In contrast, the secondary activation of the late VCP gene was inhibited (Fig.7B). 158 A 400 } i-576A' g a°° ' l-HRS lDVCP E 100 l m 0 , L :h - Oh 4 h 16 h + Oh + 4n +16h 1st Responoe (20E) 2nd Response (ZOE) a 2400 2000 - - 3 1.575% g 1600 l-HR3 1200 lDYCP . ‘ 800 400 O e M Oh 4 h 16 h + on + 411 +16h 1st Response (ZOE) 2nd Response (20E+Chx) 1000 “-5752; l i-HRS , 19.1.1931 NormalzedRNAlevelo e§§§§ on 4 h 16 h + on + 4n + 16h 1st Response (20E+Chx) 2nd Response (205+Chx) Fig. 7. Secondary responses to 20E of the ecdysone-inducible genes in in vitro fat body culture. The primary response experiments were performed as previously described (Fig. 5, 20B, or Fig. 6, Chx/20E) and presented for comparison with the secondary responses. The secondary response experiments were performed following the 20E withdrawal experiment: the previtellogenic competent fat body was first incubated with 10'6 M 20B for 4 hr and then in hormone-free medium for 12 hr in the absence or presence of 10" M Chx (Fig. 5, 20E-4h, or Fig. 6, Chx/20E-4h, respectively). The secondary responses were evaluated in such a fat body after additional incubation with 20E for 4 hr or 16 hr in the absence (20E) or presence of Chx (20E + Chx). RT-PCR/Southern analyses were conducted for the cultured fat bodies. (A) Super-induction of ecdysone-inducible genes (E75A, HR3 and VCP) in the secondary responses to 20E. Both the primary and secondary response experiments were performed in the presence of 10" 20B. (B) Effects of Chx on the secondary responses of 20E-responsive genes. The primary response experiment was performed as in Panel A. The 20E withdrawal prior to the secondary incubation was performed in the presence of 10‘ M 20E only (as in Fig. 5, 20E-4h), while the secondary response experiment was in the presence of 10" M Chx and 10'6 M 20B. (C) Abolishment of the secondary responses of 20E-inducible genes by Chx present in the initial 20E withdrawal incubation. In the primary responses, E 75 and HR3 genes were super-induced by 20E in the presence of 10" M Chx. However, after the 20E-withdrawal experiment performed in the presence of the same concentration of Chx, as in Fig. 6, Chx/20E-4h, the secondary induction of E 75 and HR3 genes was inhibited. For each 20E-inducible gene, data were normalized based on the I6-hr 20E induction in the primary response, which was referred to as 100. Each point represents an average of three independent experiments : SD. 159 In the final experiment, I conducted both the initial (Fig. 6, F TZ-Fl, Chx/20E-4h) and secondary incubations in the presence of 10“t M Chx. Under these conditions, activation of all 20E-responsive genes during the secondary response was abolished (Fig. 7C). This suggests that the AaFTZ-F 1 protein synthesis resulted from the initial incubation without Chx was required for the secondary activation of the 20E-responsive genes. The onset of ecdysone-response competence was correlated with the appearance of functional AaF T Z-F 1 protein The fat body of a newly emerged female mosquito was not competent to respond to 20E, and the competence was acquired by this tissue as a result of the previtellogenic development (Fig. 2). However, the AaFTZ-FI gene was highly expressed in this tissue throughout the previtellogenic development (Fig. 4B, FTZ-Fl). To resolve this apparent contradiction, investigation was conducted on whether the AaFTZ-Fl protein was present at the first day after eclosion, or the appearance of the functional AaFTZ-Fl protein could correlate with the acquisition of competence for 20E-vitellogenic response by this tissue. To test this hypothesis, EMSA was performed for nuclear extracts from previtellogenic fat bodies and the FTZ-Fl recognition element (FIRE) from in the upstream regulatory region of Drosophilafushi tarazu (ftz) gene. This 9-bp binding sequence for FTZ-F 1 (5’- YCAAGGYCR-B’) was identified by an in vitro DNase I footprinting assay (Ohno et al., 1994) First, it was evaluated whether the in vitro translated AaFTZ-F 1 protein was able to bind to the FIRE (Fig. 8A, lane 2). The specificity of binding was confirmed by 160 A B TNT-beta FTZ-Fl - + + + + + ----- 3—5d PV FBNE ------ + + + + + TNT-beta FTZ-Fl - + - - unlabeledFlRE --+----+--- 0-0.5dPVFBNE -'+- nonspecificDNA ---+----+-- 3-5dPV FBNE ---+ anti-DmFTZ—Fl ----+----+- prc-immunc scrum - + 123456789101 1234 Fig. 8. Identification of the functional AaFTZ-Fl protein in fat body nuclear extracts (F BNE) of previtellogenic female mosquitoes. (A) EMSA analysis of the AaFTZ-Fl protein translated in vitro (TNT-beta FTZ-Fl) or in the FBNE from mosquitoes, 3-5 days after eclosion. The protein samples were incubated with the 32P- labeled FTZ-F 1 response element, F 1RE. The specificity of the interaction between the FIRE and AaFTZ-F l was examined by the addition of SO-fold molar excess of the unlabeled FIRE (lanes 3 and 8) or a double-stranded nonspecific DNA (lanes 4 and 9). In addition, anti-DmFTZ-F 1 antibodies (lanes 5 and 10) and pre-immune serum (lanes 6 and 1 l) were added to the reactions. (B) EMSA analysis of the AaFTZ-F 1 protein in F ENE at different stages of the previtellogenic mosquitoes: lane 1, negative control; lane 2, TNT—AaFTZ-F 1; lane 3, FBNE from newly eclosed previtellogenic females, 0-12 hr post-eclosion; lane 4, FBNE from previtellogenic females, 3-5 days post-eclosion. l6] competition with an excess amount of the specific unlabeled F 1RE (Fig. 8A, lane 3) and by co-incubation with anti-DmFTZ-Fl antibodies (Fig. 8A, lane 5). Both of these treatments eliminated the binding complex. The EMSA analyses were further carried out by incubating the fat body nuclear extracts (F BNE) of the 3-5-day old female mosquitoes with 32P-labeled F 1 RE. Two retarded DNA-protein complexes were observed (Fig. 8A, lane 7), one of which was similar in mobility to the complex formed by F 1RE and in vitro translated AaFTZ-Fl protein. Both bands formed by the nuclear extract were removed by competition with an excess amount of the specific unlabeled F 1RE (Fig. 8A, lane 8) or by co-incubation with anti-DmFTZ-Fl antibodies (Fig. 8A, lane 10), indicating that these complexes were the specific binding of AaFTZ-F 1 to F 1RE. Next, the DNA binding activity of AaFTZ-F l in the FBNE of the female mosquitoes at O-O.5 day after eclosion was tested. No AaFTZ-Fl-containing complex was detected in the F BNE of the newly-eclosed females (Fig. 8B, lane 3). In contrast, the AaFTZ-Fl binding was clearly seen in the 3-5 day-old FBNE used as a positive control in the same experiment (Fig. SB, lane 4). Testing of the FBNE from mosquitoes of intermediate previtellogenic ages, 1 and 2 day-old, was inconclusive due to variability in development and limitation in the sensitivity of the assays. Interestingly, when the FBNE from the vitellogenic mosquitoes were used in EMSA, the results showed that the intensity of the AaFTZ-Fl-containing complexes decreased sharply after a blood meal and disappeared completely by the peak of vitellogenesis. The AaFTZ-Fl-containing complexes appeared again in the FBNE from the post-vitellogenic mosquitoes (data not shown). Thus, these analyses confirmed that the acquisition of competence for 20E- 162 vitellogenic response by the mosquito fat body correlated with the appearance of the functional AaFTZ-Fl protein. DISCUSSION The Drosophila F TZ-F 1 gene, located within the 75CD mid-prepupal salivary gland polytene chromosome puff, includes two overlapping transcription units encoding (1F TZ-Fl and BFTZ-F 1, two orphan nuclear receptor isoforms differing only in the N- terminal part of their A/B domains (Ueda et al., 1990; Lavorgna et al., 1991; 1993). BFTZ-Fl has been shown to play a central role in the acquisition of competence for the late prepupal ecdysone response at the onset of Drosophila metamorphosis (Woodard et al., 1994; Broadus et al., 1999). This nuclear receptor is repressed by ecdysone and expressed during the low hormone titer interval in mid-prepupae (Lavorgna et al., 1993; Woodard et al., 1994). lmmunocytochemical staining of polytene chromosomes with anti-BFTZ-F 1 antibodies shows that BFTZ-F 1 protein is bound to several sites corresponding to ecdysone-regulated genes, including the early genes, E 75 and E 74, suggesting its direct involvement in regulation of these genes (Lavorgna et al., 1993). Indeed, ectopic over-expression of BFTZ-Fl significantly increases the ecdysone induction of the early genes BR-C, E 74A and E 75A (Woodard et al., 1994). Finally, a recent study has shown that the ecdysone-triggered hierarchy, which directs the stage- specific metamorphic transition, is attenuated in BFTZ-Fl mutants (Broadus et al., 1999). These observations support the initial suggestion made from studying ecdysone responses of puffs of Drosophila salivary gland polytene chromosomes, which has showed that competence is acquired during the mid-prepupal period for re-induction of the BR-C, 163 E 74A and E 75A early puffs (Richards, 1976a; 1976b; 1976c), and clearly define BFTZ-Fl as a key competence factor for stage—specific ecdysone responses in Drosophila metamorphosis. Ecdysone plays a critical role in controlling oogenesis and vitellogenesis in flies and mosquitoes (Bownes, 1986; Hagedom, 1985; Raikhel, 1992; Buszczak et al., 1999; Raikhel et al., 1999); however, limited information has been available concerning the molecular basis of these ecdysone-mediated events. In particular, an important question to be answered is how the same hormone, which regulates larval molts and pupal metamorphosis, activates entirely different sets of target genes in the reproductive tissues of an adult insect. In the case of mosquito, it is shown here that AaFTZ-F I likely serves as a competence factor for the stage-specific responses to ecdysone in the fat body during vitellogenesis. A homolog of Drosophila flFTZ-FI is expressed at relatively high levels in the mosquito fat body during pre- and post-vitellogenic periods when ecdysteroid titers are low. However, the AaFTZ-FI transcript nearly disappears during the vitellogenic period with high ecdysteroid levels. In Drosophila, another orphan nuclear receptor (DHR3) is implicated in regulation of flF TZ—F 1 gene transcription. During Drosophila metamorphosis, DHR3 has dual roles in both repressing the early genes and activating flFYZ—FI transcription (Lam et al., 1997; White et al., 1997). The mosquito HR3 homolog, AHR3, has been cloned (Kapitskaya et al., 2000), and its expression analysis concurs with the studies on Drosophila metamorphosis, suggesting its role in AaFTZ-FI gene activation. The in vivo developmental profiles of AHR3 in both the fat body and ovary have shown that the peak of the AHR3 transcript coincides with a high ecdysteroid titer in the hemolymph 164 (Kapitskaya et al., 2000). In this study, the developmental profile of AHR3 in pupae was also examined. Remarkably, the peak expression of AHR3 preceded expression of the AaFTZ-FI gene in pupae and in the adult vitellogenic tissues, the fat body (this study) and ovary (Kapitskaya, M., Li, C., and Raikhel, A., unpublished observation). The sequential expression of AHR3 and AaFTZ-FI in the late pupa-early adult suggests that these factors are involved in setting up stage-specific responses to ecdysone in the previtellogenic fat body for the first vitellogenic cycle. Likewise, expression of AHR3 and AaFYZ-FI during the late vitellogenic period is likely preparing the fat body for a second vitellogenic cycle. Experiments with in vitro organ culture strongly supported the hypothesis that AaFTZ-F 1 expression is involved in the acquisition of competence for ecdysone response in the adult mosquito fat body. In the previtellogenic fat body, AaFTZ—FI expression was inhibited by 20E and strongly activated by its withdrawal. Similarly, in the silkworm Bombyx mori, BmFTZ-Fl is inducible by a decline in the ecdysteroid titer (Sun et al., 1994). Addition of Chx into the medium in withdrawal experiments also led to super- induction of the mosquito F TZ-F 1 , suggesting that AaFTZ-FI expression is repressed by one or more unstable repressors. Importantly, after in vitro over-expression of AaF TZ-F 1 in the previtellogenic mosquito fat body, a secondary 20E challenge resulted in super- induction of the early AaE 75 gene and the late V CP gene. These experiments are consistent with in vitro organ culture experiments in Drosophila which have shown that ,6FTZ—Fl is repressed by ecdysone, and that in vivo ectopic expression of ,677 TZ-F 1 in late-third instar larvae enhances early gene induction (Woodard et al., 1994). The acquisition of competence for ecdysone responsiveness by the mosquito fat body occurs 165 over the first two days of the previtellogenic development (Flanagan and Hagedom, 1977; this study). Although the newly emerged mosquitoes were not competent for ecdysone response, AaFTZ-F] mRNA was abundant in their fat bodies. The explanation for this apparent contradiction was revealed by EMSA analysis of AaFTZ-F 1 DNA binding activity, which showed that the appearance of functional AaFTZ-F 1 protein, rather than its mRNA, was correlated with the ecdysone-response competence in the mosquito fat body. The previtellogenic development of the mosquito fat body, which involves an increase in ploidy (Dittmann et al., 1989) and ribosome proliferation (Raikhel and Lea, 1991), is controlled by J H III. Flanagan and Hagedom (1977) have shown that the fat body responsiveness to ecdysone is also under JH regulation. Further studies are required to elucidate whether JH is involved in the post-transcriptional control of AaFTZ- F1 activity. In conclusion, our findings indicate that a conserved molecular mechanism for the stage specificity may be used during insect metamorphic and reproductive responses to ecdysone. However, the unique feature of mosquito vitellogenesis is the apparent involvement of post-transcriptional control of AaFTZ-F 1 in the acquisition of the ecdysone-response competence. Elucidation of the molecular mechanism underlying this unique characteristic of mosquito vitellogenesis represents an exciting and challenging task for future research. ACKNOWLEDGMENTS I thank Marianna Kapitskaya for partial cloning of AaFTZ-F 1, J insong Zhu for EMSA analysis and Ken Miura for RT/PCR analysis. I also thank William Segraves for 166 critical reading, Alan Hays for excellent technical assistance, Michel Trail for editing the manuscript, and Carl Thummel for his generous gift of Drosophila FTZ-F 1 antibodies. This grant was supported by NIH grant AI-36959. 167 Chapter 6 Cloning and Characterization of the Mosquito HR 78 Gene 168 ABSTRACT The ecdysone gene regulatory hierarchy is mediated by a number of transcription factors belonging to the nuclear receptor superfamily. Here, AHR 78 has been identified, which is the mosquito homolog of Drosophila hormone receptor 78 (DHR78) and its vertebrate counterpart, testicular receptor 2 (TR2). A full-length cDNA clone was obtained from a mosquito vitellogenic fat body cDNA library. The complete ORF of AHR78, consisting of 630 amino acids, exhibits the highest homology to DHR78 and TR2. The AHR78 DBD displays 95% identity (98% similarity) with the DHR78 DBD and 79% identity (89% similarity) with the TR2 DBD. During the previtellogenic period, AHR 78 is expressed at extremely low levels in the fat body and previtellogenic ovary, however, the AHR78 transcript is considerably elevated in the ovary during the vitellogenic period, reaching its peak level at 24 hr post blood meal, which correlates with the expression peak of the vitellogenin gene (Vg), a target of 20E in the fat body. In in vitro culture, AHR 78 mRNA is induced by 20E in the ovary but not in the fat body. Furthermore, AHR78 protein is capable of inhibiting the DNA binding and transactivation activities of AaEcR/AaUSP on various ecdysone response elements (EcRE), including Drosophila hsp27- and Eip28/29—EcREs, and the Aedes Vg-EcRE. These results suggest that AHR78 may be involved in inhibition of the Vg gene expression in the ovary during vitellogenesis. 169 INTRODUCTION The major insect steroid hormone, 20E, is involved in regulating multiple developmental processes including embryogenesis, larval molting, metamorphosis and reproduction (Bownes, 1986; Raikhel, 1992; Riddiford, 1993a,b; Thummel, 1995). The functional ecdysteroid receptor is a heterodimer of the ecdysone receptor (EcR) and a retinoid X receptor (RXR) homolog, Ultraspiracle (USP) (Koelle et al., 1991; Yao et al., 1992; 1993). EcR and USP play the key roles in the ecdysone gene regulatory hierarchies during Drosoplzila metamorphosis (Bender et al., 1997; Hall and Thummel 1998). In addition, the formation of inactive heterodimers between DHR38 and USP, SVP and EcR, as well as the DHR78 homodimer as a competitor, inhibit DNA binding and activation by EcR/USP on the ecdysone responsive elements, providing different levels at which ecdysone responses can be negatively controlled (Sutherland et al., 1995; Zelhofet al., 1995a; Fisk and Thummel, 1995, 1998; Zelhof et al., 1995b). In contrast to the large body of information concerning the molecular basis of the ecdysone-mediated gene regulation during development and metamorphosis, comparatively little is known about the ecdysone-regulated aspects of insect female reproduction, especially the negative regulation of ecdysone responses. The mosquito is an ideal model animal to study ecdysone-mediated gene regulation during reproduction. This is due to the fact that mosquito reproduction is triggered by blood feeding, which is a requirement for egg development. In addition, mosquitoes act as vectors for many devastating human diseases, which increases the significance of current research (Collins and Paskewitz, 1995; Butler etal., 1997; Beier, 1998). In the anautogenous mosquito Aedes aegypti, the process of vitellogenesis is the cornerstone of egg maturation. 170 Vitellogenin is initiated only after a female mosquito ingests vertebrate blood. A blood meal triggers a hormonal cascade which results in production of 20E, the terminal signal that activates yolk protein precursor (YPP) genes in the fat body (Hagedom, 1983; 1985; Raikhel, 1992; Dhadialla and Raikhel, 1994; Deitsch et al., 1995a; 1995b). Vitellogenin (Vg) is one of the major YPPs. In addition, the mosquito fat body synthesizes several other YPPs, vitellogenic carboxypeptidase (VCP), vitellogenic cathepsin B (VCB) and lipophorin (Cho et al., 1991, 1999; Sun et al., 2000). YPP synthesis reaches maximal levels at 24 hr post-blood meal (PBM) and then proceeds until 30-32 hr PBM, when it is rapidly terminated (Raikhel, 1992). YPP genes are activated in the fat body and their protein products are translocated through the hemolymph and finally internalized into the developing oocytes (Raikhel, 1992). The titers of ecdysteroid in the female mosquito hemolymph positively correlate with the rate of YPP synthesis in the fat body. While 20E titers are only slightly elevated at 4 hr PBM, they begin to rise sharply at 6-8 hr PBM, and reach their maximum levels at 18-24 hr PBM (Hagedom et al., 1975). Consistent with the initially proposed role of 20E in activating vitellogenesis in anautogenous mosquitoes (Hagedom et al., 1973; Hagedom and Fallon, 1973), experiments using an in vitro fat body culture have shown that physiological doses of 20E (10'7 - 10’6 M) activate two YPP genes, Vg and VCP (Deitsch et al., l995a; 1995b). Utilization of the protein synthesis inhibitor, cycloheximide (Chx), has demonstrated that the activation of YPP genes in the mosquito fat body requires protein synthesis (Deitsch et al., 1995a). Thus, it is likely that a regulatory cascade similar to that seen in Drosophila development mediates the action of 20E in the mosquito fat body. 171 In A. aegypti, two isoforms of both the ecdysone receptor (AaEcR-A and AaEcR- B) and Ultraspiracle (AaUSP-A and AaUSP-B). have been identified. Also AaEcR-B and AaUSP-B function as major heterodimerization partners during vitellogenesis (Cho et al., 1995; Kapitskaya et al., 1996; Wang et al., 2000a; Wang, Li, and Raikhel, unpublished data). Mosquito EcR and USP are expressed in pre- and vitellogenic fat bodies and ovaries (Cho et al., 1995; Wang et al., 2000a). The AaEcR/AaUSP heterodimer has been shown to bind to various ecdysone response elements (EcREs) to modulate ecdysone regulation of target genes (Wang et al., 1998). Most recently, we have demonstrated that the Vg S’- regulatory region contains a functional EcRE that is necessary to confer its responsiveness to 20E. AaEcR/AaUSP binds directly to the Vg EcRE, and activates the Vg promoter in the presence of 20E in Drosophila 82 cells (Martin et al., 2001). Furthermore, studies in the previous chapters have demonstrated that conservation of the ecdysone-responsive gene regulatory hierarchy extends beyond EcR and USP. A mosquito homolog of the Drosophila early gene E 75 is involved in mediating the ecdysteroid responses during vitellogenesis (Pierceall et al., 1999). Mosquito AaFTZ-F 1 serves as a competence factor for the vitellogenic response to ecdysone, which is implicated in activation of AaE75 and YPP genes (Li et al., 2000). The orphan nuclear receptor AHR3, a mosquito homolog of DHR3, has also been cloned, and is thought to be involved in the activation of AaF TZ-F 1 during vitellogenesis (Kapitskaya et al., 2000; Li et al., 2000). In addition, AHR3 8, the mosquito homolog of DHR38 and vertebrate NGFI-B, inhibits the ecdysone response in the mosquito fat body during the previtellogenic stage by strongly interacting with the AaUSP protein and preventing the formation of a functional ecdysteroid receptor (Zhu et al., 2000). Thus, 172 despite the functional diversity of genes targeted by ecdysone-mediated gene regulation, some molecular mechanisms of the ecdysone gene regulatory hierarchy appear to be conserved between mosquito vitellogenesis and Drosophila metamorphosis. In this study, I report the cloninglof mosquito HR78, a homolog of Drosophila DHR78 (Zelhof et al., I995a; Fisk and Thummel, 1995) and vertebrate TR2 (Chang and Kokontis, 1988). DHR78 is an orphan receptor, which can inhibit ecdysone signaling and is required for ecdysone-regulated gene expression in mid-third instar larvae in Drosophila (Fisk and Thummel, 1995, 1998; Zelhof et al., 1995b). While the mosquito AHR78 DBD is highly conserved relative to the DHR78 DBD, the LBDs in this subfamily show less similarity relative to the other nuclear receptor subfamilies, suggesting that the AHR78 subfamily has evolved rapidly, particular in the E domain. AHR78 is expressed at extremely low levels in the fat body. In the ovary, AHR78 mRNA levels are relatively low during the previtellogenic stage; however, its levels increase dramatically during the vitellogenic stage with a peak level at 24 hr post blood meal. Moreover, AHR78 inhibits binding by EcR/USP to natural EcREs and inhibits the transactivation of EchU SP in cell transfection assays. Most significantly, AHR78 inhibits DNA binding'and transactivation of EcR/USP on the Vg promoter, suggesting that AHR78 may be involved in inhibition of Vg gene activation in the ovary during vitellogenesis. 173 MATERIALS AND METHODS Animals Mosquitoes, A edes aegypti, were reared according to the method of Hays and Raikhel (1990). Larvae were fed on a standard diet as previously described (Lea, 1964). Vitellogenesis was initiated by allowing females, 3-5 days afier eclosion, to feed on an anesthetized white rat. cDNA cloning Degenerate primers based on the specific-conserved sequences in DBDS between DHR 78 and mTRZ were designed for PCR to obtain a mosquito homolog HR78 cDNA fragment. Two forward degenerate primers, AHR78-F1 (GCN WSI GGI MGI CAY TAY GII G) and AHR78-F 2 (TGY GAR GGI TGY AAR GGN TTY TT), and one reversed primer, AHR78-R (TGR CAI CKR TTI CKR TGR TGY TT) were utilized for nested PCR. Amplification was carried out in a Perkin Elmer thermal cycler using a template of cDNA produced by reverse transcription from total RNA prepared from the fat bodies of the vitellogenic female mosquitoes. The PCR—generated fragment was used as a probe to screen a kZAPII cDNA library prepared from the fat bodies of vitellogenic female mosquitoes during 6-48 hr post-blood meal (PBM) as previously reported (Cho and Raikhel, 1992). Several positive cDNA clones were subsequently isolated and the one with longest cDNA insert was sequenced at W.M. Keck Foundation Biotechnology Resource Laboratory, Yale University. Analyses of nucleotide and deduced amino acid sequences were performed using the software of GCG (Genetics Computer Group, University of Wisconsin, Madison). 174 RT-PCR/Southern blot RT-PCR/Southem analyses were performed as previously described (Li et al., 2000). Total RNA was prepared from fat bodies throughout the first vitellogenic cycle by TRIZOL Reagent (Gibco BRL). After reverse-transcription by Superscript II reverse transcriptase (Gibco BRL), 0.025 fat body equivalent of the cDNA pool was used as a PCR template for each point for the analyses of developmental profiles. Thermal cycling conditions were as follows: 940C for 3 min, followed by 18 cycles of 94°C for 30 sec, 600C for 30 sec and 720C for 30 sec. The PCR primers for AHR78 are AHR78-F 33 (GCT TC C CAA CTA CCT GAG TGC G) and AHR78-R73 (GCC AGC TGG CCG CTA CTC T), both of which are located in the ligand binding domain of AHR78. As a reference to the developmental change of the fat body and the ovary, the samples of the same cDNAs were subjected to the 13 cycles of PCR with each primer pair specific to the mosquito vitellogenic carboxypeptidase (VCP) or vitellogenin receptor (VgR). Southern blot hybridization was performed under high stringency conditions using each gene- specific probe, which was generated by PCR using each cloned cDNA as a template. In vitro fat body and ovary culture Mosquito fat body culture experiments were performed as previously described (Li et al., 2000). The ovary culture experiments were performed according to the method by Sappington et a1. (1997). After culturing, RT-PCR/Southem blot were utilized to analyze the mRNA levels. 175 Electrophoretic mobility shift assays Electrophoretic mobility shift assays (EMSA) were carried out as described previously (Wang et al., 2000a). Nuclear receptor proteins were synthesized in vitro using a coupled transcription-translation (TNT) kit from Promega. AaEcR and AaUSPb proteins were expressed with the SP6 promoter from the vectors pGEM3Z-AaEcR and pGEM7Z-AaUSPb, respectively. AHR78 protein was expressed from pBluescriptSK- AHR78 with the T3 promoter. The double strands of EcREs are: Vg-EcRE, agctgthGACCTagcgggAGGCCAaTGGTCGagt; Hsp27, agagacaagGGTTCAaTGCACTtgtccaat; Eip28/29, ttaaAGGATCtTGACCCcaaT GAACthat. Five pmols of each EcRE was end-labeled with 7-321) ATP (Dupont NEN) by T4 polynucleotide kinase (Gibco BRL). In each binding reaction, typically 1 pl TNT protein and 100 fmol labeled EcRE were incubated in total 20ul of binding buffer containing 20mM HEPES (pH7.5), 2mM DTI‘, lOOmM KCl, 7.5% Glycerol, 0.1% NP- 40, 2 pg of poly (dI-dC)o poly (dI-dC) (Pharrnacia Biotech). In some experiments, the binding buffer also contained 2 pg of non-specific single stranded DNA to eliminate non- specific binding (Wang et al., 1998). The binding reaction was conducted for 30 min at room temperature. Cell culture and transient transfection assay Cell culture and transient transfection assay were performed as previously described (Wang et al., 2000b). The Drosophila S2 cell line (Invitrogen) was maintained at 22-24 °C in Schneider Drosophila medium supplemented with 10% heat- 176 inactivated fetal bovine serum, penicillin (100U/ml), and streptomycin (IOOug/ml) (Gibco BRL). Transfection was conducted with Cellfectin (Gibco BRL). Typically, lOOng of the luciferase reporter plasmid and 12.5 ng of the reporter pAc5-LacZ plasmid as well as a total 180ng of all receptors and pAc5 plasmids, were mixed in a 48-well plate with 2 ug of Cellfectin in a total volume of ZOul, and then incubated at room temperature for 30min. The transfection cocktail was overlaid with 200 pl of 82 cells (2x106 cells/ml) in Drosophila serum-free medium. Transfection was terminated 12 hr later by adding 200ul 20% fetal bovine serum media containing either 2><10'6 M 20E or an equal amount of vehicle ethanol. After treatment with the hormone 20E for 27hr, cells were collected and lysed by 100 pl lxreporter lysis buffer (Promega) with one cycle of freezing and thawing. Reporter gene assays were conducted as described by the Promega firefly luciferase reporter and B-galactosidase systems. A luminometer (Turner Designs model TD20e) was used to detect luciferase activity with 10 sec pre-delay and 30 sec integration time. Luciferase activities were normalized by B-galactosidase activities in each experiment. The reporter plasmids AMTV-Eip28/29-luc and AMTV-hsp27-luc were kind gifts from M. Mckeown (Salk Institute, San Diego, CA). The AMTV-Eip28/29-luc construct contains four copies of Eip28/29 EcRE and the AMTV-hsp27—luc has five copies of hsp27 EcRE. The pAc5-AaEcR and pAc5-AaUSPb constructs contained the full length AaEcR and AaUSPb (Wang et al., 2000b). The pAc5-AHR78 plasmid was constructed by digesting pBluesript SK-AHR78, which was directly from the mosquito lZaplI cDNA library, with XbaI and ApaI and subcloning the ORF into the pAcS XbaI/Apal sites. The Vg-luc plasmid was the Vg promoter (-348 to +115bp) subcloned in the pGL3—basic vector (Promega). 177 RESULTS Cloning and characterization of AHR 78 cDNA Three degenerate primers were designed (see Materials and Methods), based on specific-conserved regions of the DNA binding domain (DBD) in the DHR78/TR2 family. After the first round of PCR amplification (using the fat body cDNA as a template) by the primer pair AHR78-Fl and AHR78-R, and purification of the amplified DNA fragment, the second round of nested PCR amplification was conducted using the primers AHR78-F2 and AHR78-R. The final PCR amplified DNA fragment (~110 bp) was then subcloned into a pGEM-T vector and sequenced. The sequencing data indicated that this DNA fragment highly resembled the DHR78/TR2 family, and the deduced amino acid sequence was more similar to that of DHR78 than that of any other nuclear receptors in the GeneBank. Subsequently, this DNA fragment was used as a probe to screen the mosquito fat body cDNA library that was generated from fat bodies of 6-48hr PBM mosquitoes by the poly (T) method (Cho and Raikhel, 1992). Several positive clones were identified by screening of the cDNA library. The clone with the largest insert was completely sequenced on both strands. This clone of a 2.8 kb insert contained an open reading frame (ORF) of 630 amino acids. There were 3 stop codons before the first putative start codon (ATG), and polyadenylation signaling (AATAAA) after the ORF, suggesting the ORF is complete (the GeneBank accession number is ......... ). The conceptual translation of AHR78 amino acid sequence revealed distinctly high similarity with its insect counterparts of Drosophila and Tenebrio molitor (Zelhof 178 AHR78 TmHR78 DHR78 AHR78 TmHR78 DHR78 AHR78 TmHR78 DHR78 AHR78 TmHR78 DHR78 AHR78 TmHR78 DHR78 AHR78 TmHR78 DHR78 AHR78 TmHR78 DHR78 AHR78 TmHR78 DHR78 AHR78 TmHR78 DHR78 AHR78 TmHR78 DHR78 AHR78 TmHR78 DHR78 l ~~~MEPHNFD ~~~~~~~~~~ MDGVKVETFI 5 DBD ELCLVCGDRA ELCVVCEDRA ELCLVCGDRA 101 HRNRCQYCRL HRNRCQYCRL HRNRCQFCRL 151 NPNSKYNPHR NIGNNYNSNS NGSSTYLSGK 201 ELAVANLSKR DLGLQFLNKR GLNFAELTQT 251 PAD.SIAALM SMD.S ..... SSTLCLQLLA 301 H.8LPMETTD D.ALIM.... HKGLQILQPI 351 ....NGRDDD ....AA.... FECGGNRSGG 401 ETGSRLLFAT ESGSRLLFLS ETGSRIIFLT 451 LAFNTVMLAL LSLSTILSAL LSVPTIIGQF 501 TDQEFAYMRL DEHEYAILKA TDMEFGLLRL IKPNLQQMKL ~~~~~~~mI KSEENRAMPL SGRHYGAISC SGRHYGAISC SGRHYGAISC 4—1 QKCLACGMRS QKCLACGMRS QKCLBSGMRS SGYQQGRGKG A..SKSPPAP IGNSGSSDLP MATQQQQQ QLGPTAAAAV SNSMEKNLIC ..ARDKQLIS QNQLERN.GN RDDDCLNYDY .DEKC..YEY SDEAINEAVF VHWMKKNHLF IHWTRNIPAF IHTLRKVPVF IQYMKNLILN ISHLHTLIAQ IQSTRQLADI LCIFNPDNIL ITLFSADQ.. ILLFNP.... EPGTQQFGSS z...asrxsz recesasccr EGCKGFFKRS EGCKGFFKRS EGCKGFFKRS DSVQHERKPI DSVQHERKPI DSVQHERKPI ...KEYHNEQ ...KIF..IR HSVKAESAPR sanwsssrwr ranspsgasr QQQQHQQSGS NMSGLGPPPQ TNTGAG.... LNFNAGEVPT NSLEFIQNLE KALDTMARVQ stxpzcnsz GSLELSDNCI EGPILQEQHI EQDLLTDVQC sunsnsrosz QYLTTETQIT EQLEAHTQVK KKXGSDVINY DKMSATKVKQ DKIEPLKISK QDNVKNQHLA PDILLRKHVE ...TLFQHRK 179 SLDDKGPSLQ ILSDX...IR PLPGGGVGMG IRKQLGYQCR IRKQLGYQCR IRKQLGYQCR IDKKDG.... IDKKDY.... VDRKEGIIAA KSNTTVTPSY KDLSTDSPGL LQCTARQQRA SSNPFSSLHS YSPDIPKADP QPPPSLEPPE ......... E ALPTTSTMGL HELNNSVNNN C.LNGT.... AEDSGTEDAV SFDIQVPNVL SFNLQIPGPV AFHVQPPTLV LMRQTWPELF LLRGCWBELF LLRGVWPALM LTKYILLIQE VSDHIVKLQD MANLTRTLHD KIQDMVLSSF KLQEKSFQAL E ...... RSL 50 HLSSLNSMSI NNSNNLCLTV AGA8.AILSV 100 GSMNCEVTKH GSKNCEVTKH GAMNCEVTKH 150 ...... SGGP ...... 8N.. AGSSSTSGGG 200 LNLFQGFNLA LP..APFNPC FNLNAEYIPM 250 FKSQDDRQPT ...EDD...V EDDEDDSMDN 300 LPSKPTPPSS L ........ 8 IQSSLDMRVI 350 YGIKSELN.. ..DL88LT.. DAELEHMELD 400 PNYLSAHYVC ppvnnrurrc asrnnrarvc ‘-’*Lae 450 MIGLAQSSGQ TLGLAQCSQT AIALAQCQGQ 500 FVSDVQKLNL YANTMNRLNV FVQELQSLDV 550 RDYYKQKHSR KT! ....... RGYVRRVQLY 551 600 AHR78 LMBS.RSEEN IRGGQDDDDD YYESSHHQQQ RQQRHNLEQR LVTILMKLPT TmHR78 .......... VHNSFPDDTD ................... R FPRLLLRLPP DHR78 ALSSLRRQGG IGGG ....................... ERR FNVLVARLLP 601 650 AHR78 LRALNSKKDL EDLFFSTLIG QVQIESVEM! ILQTNDG.AT SFSNL.VRNY TmHR78 LRGLEPLV.L EELFEAGLIG QVQIDSVIPY ILRMGNGMPT PTSNRHVKSE DHR78 LSSLDA.EAMIEELFFANLVG QMQMDALIPF IEMTSNTSGL ~~~~~~~~~~ 651 668 AHR78 arsarpvesc cameo!) TmHR78 gunmen“ ~~~~~~~~ Fig. 1. Alignment of the deduced amino acid sequence of the mosquito AHR78 cDNA with its insect homologs: DHR78 from Drosophila (Fisk and Thummel, 1995; Zelhof, et al., 1995b) and TmHR78 from T enebrio molitor (Mouillet et al., 1999). The borders of DNA-binding domain (DBD) and putative ligand-binding domain (LBD) are marked with square braces. The complete nucleotide sequence of AHR 78 has been submitted to the GenBank (http;//www,ngbi,nlm.nih.gov1; its accession number is .......... 180 .8538 8:05 2: mo .382: Boa 05:8 .38 05 .3 e336 32.0368 033.8988 Boston 32:52 mesa 05:8 .8 confine a ma coon—=28 803 move.— ofi EEEop some 555 @8865 2a mezwoBEo: 550 one why—*2 mo mEmEoo w:_e=oqmetoo 5053 Gotta—EV. .3 85:82 .8 mowfieooeom of. .3382 :08 E memos oEEm Mo 0382. 05 8865 2:205 323 £3832 .mEmEoe _mcouoee 828p 5805 mum: 8:588 05 goes Betc— ef. Ammo— .mzeoxov. can mess“: NM... 8308 one .332 do 3 uoEoN ”32 ._oEE=nH EB 02“: SEQSEQ Eob mummo .2635 £23 agave 83V 80¢ mummies ”£9308 320:: mum: 05 2a :Bozm .3358.— ..335: gags—.4. «MP—.35:— 2: we 22.52: 138.3. :33 why—E 33.08:. 2: no eemtanfieo 5355 .N .wE am _ 3...: $3 2...: Se _ _ ”DE: :3 _ _ ”Ema. 181 et al., 1995a; Fisk and Thummel, 1995; Mouillet et al., 1999) (Fig. 1). AHR78 contains the classic nuclear receptor functional domains: A/B domain, DNA binding domain (DBD) or C domain, hinge region or D domain, ligand binding domain (LBD) or E domain, and F domain. Comparison of AHR78 with Drosophila DHR78 and its vertebrate counterpart, human testicular receptor 2 (TR2) showed that these proteins were of similar length in regards to their amino acid sequences: AHR78, 630aa; DHR78 601aa; mTR2, 590aa. Moreover, the AHR78 DBD is highly conserved in relation to the DBDs in DHR 78 and TR2, exhibiting 95% identity (98% similarity) and 79% identity (89% similarity), respectively. The LBD of AHR78 exhibits 33% identity (47% similarity) with DHR78 and 30% identity (41% similarity) with mTRZ. The other domains of this family are relatively less conserved (Fig. 2). AHR 78 is highly expressed in the vitellogenic ovary, but not the fat body To examine the AHR78 expression profiles, RT-PCR was utilized to analyze the mRNA levels during mosquito vitellogenesis. The two critical vitellogenic tissues, the fat body and ovary, were chosen for examination of AHR78 expression levels during the first vitellogenic cycle. In the fat body, traces of AHR78 were expressed at the relatively constant levels throughout the previtellogenic and vitellogenic stages. Vitellogenic carboxypeptidase (VCP), one of the major YPPs (Cho et al, 1991), was used as a positive expression control in the fat body. In the ovary, AHR78 mRNA levels were considerably higher than those in the fat body, with dramatic increases after a blood meal peaking at 18—24h PBM, followed by a gradual decline after 30hr PBM (Fig. 3). Interestingly, expression dynamics of AHR78 in the ovary correlates with 20E level changes during 182 (A) W! PV ((1) Post Blood Meal (hr) 0-1 1-2 3-5 1 3 6 15 24 36 48 VCP (B) 91313! Post Blood Meal (hr) PV 612 18 24 30 36 42 48 “M--- HR73 Fig. 3. AHR 78 expression profiles in the female mosquito fat body and ovary. (A) Temporal expression of AHR 78 in the fat body during the first vitellogenic cycle examined by RT-PCR/Southern analyses. The expression profiles of AHR 78 (upper panel) and vitellogenic carboxypeptidase (VCP) as a control (lower panel) were examined (see Materials and Methods). (B) Temporal expression of AaHR 78 in the ovary during the first vitellogenic cycle. The expression profiles of AHR 78 (upper panel) and vitellogenin receptor (VgR) as a control (lower panel) were examined (see Materials and Methods). Each lane contained the same mosquito equivalents of total RNA subjected to RT-PCR/Southem analyses. PV: previtellogenic stage. 183 this period as well as the accumulation of yolk protein precursors by the oocytes. As a positive control, the expression profile for vitellogenic receptor (VgR) (Sappington et al 1996) was provided for the ovary. In addition, AHR78 expression patterns and levels in both the fat body and ovary were monitored by using different pairs of PCR primers, which were located in either the 5’UTR or LBD. These results were the same, suggesting that the expression profiles of AHR78 detected by the RT-PCR method were specific to the AHR78 gene (not shown). AHR 78 is inducible by 20E in the ovary but not in the fat body To investigate whether the high levels of AHR78 mRNA in the ovary during the vitellogenic stage are under the control of 20E, ovaries of previtellogenic 3-5d-old or 2h PBM were incubated with 20E for 6h followed by RT-PCR/Southem analysis. The AHR78 mRNA levels increased significantly after the 20E treatment in both previtellogenic and 2h PBM ovaries (Fig. 48). For control, treatment of ovaries with ethanol, which is solvent for 20E, did not show any effect on AHR78 mRNA levels (Fig. 48). Next, fat bodies were tested in a parallel experiment. After previtellogenic 3-5d- old fat bodies were cultured with 10'6 M 20E for 4b and 8h, AHR78 did not exhibit detectable changes in mRNA levels (Fig. 4A). In the control, VCP mRNA was highly induced by 20E in the same experiment. 184 A Fat Body Culture 205 - 4h 8h AHR78 B PV Ovary 2hPBM Ovary EtOH - + + - + + 205 - - + - - + n- “mat?!“ we. . I: AHR78 3; _ __,____ _____3 Fig. 4. Induction of AHR78 mRNA by 20E in the ovary but not the fat body. A). The AHR78 gene was not responsive to 20E in the fat body. Previtellogenic 3-5d-old female fat bodies were incubated with 10'6 M 20E for 4 or 8 hr followed by RT- PCR/Southern analysis. The VCP gene was used as a positive control. B). Induction of AHR78 mRNA in the ovary. Previtellogenic 3-5d-old female ovaries (PV Ovary) or 2h- PBM ovaries were treated with 20E or 20E solvent ethanol (EtOH) for 6hr, and then analyzed by RT—PCR/Southem blot. 185 AHR 78 inhibits the DNA binding of EcR/USP to EcREs To test the effect of AHR78 on EcR/USP DNA binding, EMSA was performed using two well characterized EcREs, hsp27 and Eip28/29. In the absence of AHR78, AaEcR/AaUSPb bound to hsp27 (F ig.5A, lane 4, 7) as previously reported (Wang et al., 1998). Addition of increasing amounts of AHR78 to the binding reactions dramatically decreased EcR/USP binding to EcRE (lane 8, 9). In the control, addition of another nuclear orphan receptor AaE75A did not have any effect on DNA binding of mosquito EcR/U SP (Fig. 5A, lane 10, 1 l). Unexpectedly, AHR78 alone was not able to bind to hsp27 (Fig. 5A, lane 12). When testing the effect of AHR78 on EcR/U SP DNA binding by using the Eip28/29 EcRE, results similar to those in the hsp27 experiment were obtained: addition of AHR78 dramatically decreased the DNA binding of EcR/U SP (Fig. SB, lane 8, 9). Furthermore, I examined whether AHR78 had a similar effect on EcR/U SP binding to Vg-EcRE, an EcRE identified in the Vg promoter (Martin, et al., 2001), and found that AHR78 inhibited the binding of EcR/USP to the Vg-EcRE (Fig. 5C, lane 2, 3). Moreover, I tested whether 20E had any effect on the AHR78 inhibiting action. As shown in Fig. 5C, Lane 7, in the presence of 20E, inhibition of AHR78 on the EcR/USP DNA-binding was similar to that seen in the absence of 20E (Fig. 5C, lane 3). As a control, addition of increasing amounts of another nuclear orphan receptor AaFTZ-Fl protein into the binding reactions did not show any detectable effect in the absence of 20E (Fig. 5C, lane 4, 5). Likewise, in the presence of 20E (Fig. 5C, lane 8), AaFTZ-F 1 protein did not affect the DNA binding of EcR/USP. 186 (A) EcR - + - + + + + + + + + - USP - - + + + + + + + + + - HR78 - - - - - - 0 l 2 - — + E75 - - - — - - - .. .. 1 2 _ Ab-DmUSP - - - - + - - - .. - ' - _ Competitor - - - — - + — _ - - - .. Hsp27-EcRE + + + + + + + + + + + + —'9 u—e e—o H H Lane 1 2 3 4 5 6 7 8 9 10 ll 12 Hsp27 EcRE: agagacaagma I GCACTtgtccaat (C) (B) EcR + + + + + + + + SCSI; + J i i i ' : : : USP + + + + + + + + was .' _ L 0 I 2 HR78 . 1 2 . - - 2 + Ab-DmUSP . . . + - - - . , FrZ-Fl - - - 1 2 - - 2 Competitor - - - - + . . - . 20E - - - - - + + + Eip28/29 EcRE + + + + + + + + + VgEcRE + + + + + + + + H ‘ : Lane 1 2 3 4 5 6 7 8 9 Lane 1 2 3 4 5 6 7 8 Eip28/29 EcRE: ttaaAGGATS ITGAC CC caaTGAAC that Vg EcRE: agctgthGACCTagcgggAQQCCAaTgiQngagt Fig. 5. Inhibition of EcR/USP binding to the natural EcREs by AHR78. In vitro-translated AaEcR and AaUSP proteins alone or along with in vitro translated AHR78 protein were incubated with 32P-labeled different natural EcREs. The reaction mixtures were subjected to EMSA and autoradiography. (A) Inhibition of EcR/USP binding to the hsp27-EcRE by AHR78. In addition to 1 ul of each TNT-AaEcR and AaUSP (lane 4 and 7), 1 ul or 2 ul of TNT-AHR78 (lane 8 and 9) or AaE75A (lane 10 and 11) were added to the reactions. For controls, antibody against Drosophila USP supershified the EcR/USP binding (lane 5); 50 times molar excess of cold hsp27 EcRE was added as competition (lane 6). All reactions were in the absence of 20E. The arrow indicates the specific binding. (B) Inhibition of EcR/USP binding to the Eip28/29-ECRE by AHR78. This was a similar experiment to that in the panel A except that Eip28/29- EcRE was used as 32P-labeled or cold oligonucleotides. (C) Inhibition of EcR/USP binding to the mosquito Vg-ECRE by AHR78. This was a similar experiment to that in the panel A except that Vg-EcRE was used as 32P-labeled oligonucleotides, and some experiments in the presence of 5x 1045M 20E (lane 6, 7 and 8) as well as AaFTZ-F 1 protein as controls (lane 4 and S). 187 AHR 78 inhibits 20E-stimulated transactivation by E cR/USP To investigate whether AHR78 affects the EcR/USP-induced transactivation of target genes, a cell transfection assay was utilized. The reporter plasmid Eip-luc containing the Drosophila Eip28/29-EcRE, which has been previously shown to be activated by the mosquito EcR/U SP (Wang et al., 2000b), was transfected into Drosophila 32 cells alone or along with the expression plasmids carrying AaEcR, AaUSP or AHR 78 cDNAs. After transfection, cells were incubated either in the absence of hormone or in the presence of 20E at 10'6M for 27 hr. When no EcR/ USP plasmid was transfected, cells receiving Eip-luc alone exhibited a low level of 20E-stimulated reporter gene activities, which was presumably due to the presence of endogenous Drosophila EcR/U SP in $2 cells (Fig. 6A, column I). Transfection of both mosquito EcR and USP plasmids did not affect this baseline activity in the absence of 20E (Fig. 6A, column 2); however, addition of 20E resulted in 2.5 fold induction in reporter gene activities (F i g.6A, column 2). When increasing amounts of AHR78 plasmid was co-transfected with EcR/ USP plasmids, the induction was reduced continuously with virtually no induction at 8-fold molar excess of the AHR78 plasmid (Fig. 6A, column 5). Interestingly, in the absence of 20E, AHR78 also reduced the basal reporter activities by up to 50% (Fig. 6A, column 5). Similarly, using another hsp27-EcRE construct in the AMTV promoter driving a luciferase reporter, co-transfection of AHR 78 plasmids along with EcR/USP plasmids also reduced 20E-stimulated activation. The effect of AHR78 was less dramatic in the hsp27-luc construct than that in the Eip28/29-luc construct, presumably due to the high 188 (A) 300 200 ~ D(-)ZOE .(+)20E 100 4 Normalized Luc. Unit r—F A MTV-Eip28/29 Luc. Fig 6. AHR78 inhibition of 20E-stimulated transactivation by AaEcR/AaUSP in 82 cells. (A) AHR78 inhibition of 20E-stimulated transactivation of the Eip-luc by AaEcR/AaUSP in 82 cells. The reporter plasmids (Eip-luc,) were transfected into Drosophila 82 cells with no expression plasmid (column 1), or with AaEcR/AaUSP expression plasmids (lane 2) and increasing molar excesses of AHR78 expression plasmids (lane 3, 4 and 5) (see Materials and Methods). Afier transfection, cells were incubated either in the absence or in the presence of hormone 20E at 10'6M for 27 hr and harvested for B-galactosidase and luciferase activity assays. Luciferase activity was normalized with B-galactosidase activity. 189 (B) 500 400 - 300 - El (-)20E I (+)ZOE 200 ~ 100 - Normalized Luc. Unit Vg ECRE 350 Luc. Fig. 6. (Cont’d). B) AHR78 inhibition of 20E-stimulated transactivation of the Vg-luc by AaEcR/AaUSP in 82 cells. This experiment was performed similarly to the panel A except that the Vg-luc reporter plasmid was used in this experiment. Each value represents the mean :SD (n=3). 190 affinity of hsp27-EcRE with EcR/U SP and, as a result of it, high background levels of reporter gene activities in this experiment (not shown). In addition to examining the effect of AHR78 on the two Drosophila EcREs, I investigated the effect of AHR78 on the Vg-EcRE, which has been demonstrated to be responsive to EcR/USP (Martin et al., 2001). The reporter activities, driven by a 348bp upstream region of the Vg promoter, increased by 3-fold in responsive to EcR/U SP in the presence of 20E (Fig. 68, columns 1 and 2.). The activation by 20E was not affected by a small amount of AHR78 (Fig. 68, column 3); whereas it was decreased significantly upon co-transfection of increasing amounts of AHR78 (Fig. 63, columns 4 and 5). Other promoter-reporter constructs consisting of either 2kb, lkb or 600bp of the Vg promoter yielded less conclusive results (not shown). This is likely due to the complexity of the Vg promoter and interfering factors in the 82 cells, making the effect of AHR78 invisible. Taken together, these results suggest that AHR78 inhibits the EcR/U SP transactivation on various EcREs, most importantly on the native Vg EcRE. DISCUSSION In this study, I report the cloning and characterization of AHR78, the mosquito homolog of HR78 in other insects and vertebrate TR2. A search of the gene data bank using the full AHR78 amino acid sequence revealed that AHR78 exhibits the highest similarity with the members of this HR78/TR2 subfamily, rather than other subfamilies of the nuclear receptor superfamily (not shown). AHR78 consists of 5 classical functional domains of the nuclear receptors (Fig. 2). While DBDS of this subfamily are highly conserved, LBDS are less similar relative to other members of the nuclear receptor 191 superfamily. Although the AHR78 LBD shows 33% identity with DHR78, this is the closest homology it shares among nuclear receptors. Similarly, T enebrio molitor TmHR78 shows only 31% identity with DHR78 LBD (Mouillet et al., 1999). These results suggest that members of the HR78/TR2 subfamily of nuclear receptors have evolved rapidly, particularly in this domain. Analysis of expression profiles of HR 785 during insect development reveals the similar temporal correlations with ecdysteroid levels. In the mosquito fat body, AHR78 exhibited only negligible expression levels throughout the previtellogenic and vitellogenic stages. In contrast, in the ovary, AHR78 levels rose dramatically after a blood meal with peak expression occurring at 18-24 hr PBM correlating positively with the ecdysteroid peak in the mosquito (Fig. 3). During Drosophila metamorphosis, DHR78 is expressed throughout development with peaks of expression in third instar larvae and prepupae that is correlated with known ecdysteroid pulses (Fisk and Thummel, 1995; Zelhof et al., 1995b). Similar correlation between expression levels and ecdysteroid variations has been reported for TmHR78 (Mouillet et al., 1999). The correlations between expression levels of HR78 and ecdysteroid levels further support that AHR78 is a member of the HR78 family, despite low conservation levels of the LBD amino acid sequences. In Drosophila, it has been demonstrated by DHR78 mutants that DHR78 functions at the top of the ecdysteroid regulatory hierarchies. DHR78 mutations block ecdysteroid-regulated gene expression in mid-third instar larvae (Fisk and Thummel, 1998). In addition, DHR78 protein binds as a homodimer to a subset of EcR/U SP binding sites in vitro; therefore, it has been proposed that DHR78 can function to regulate 192 ecdysone responses negatively (Thummel, 1995). However, in mosquitoes direct binding of AHR78 to EcREs was not observed. This was probably due to sensitivity of the experiment, or the evolutionary divergence among insect HR78, particularly in LBDS. One of the LBD functions in a classical nuclear receptor is to form a receptor homodimer. The less conserved LBDS in the HR78 subfamily may explain why AHR78 did not bind to EcREs directly. In mosquitoes, the direct interaction of AHR78 and EcR/USP was observed in vitro in the GST-pulldown assays (Li, C. and Raikhel, A, unpublished). This result suggested that AHR78 negatively regulated the ecdysone responses in a different way. The direct interaction of AHR78 and EcR/USP may prevent the AaEcR/AaUSP binding to EcREs. Although it has not been reported in other insects that HR78 proteins can interact directly with EcR or USP, DHR78 protein is bound to many ecdysteroid- regulated puff loci, suggesting that DHR78 may interact directly with EcR or USP (Fisk and Thummel, 1998). In addition, consistent with findings in Dros0phila DHR78, AHR78 inhibits the 20E-induced activation of target genes in cell transfection assays (Fisk and Thummel, 1995; Zelhof et al., 1995b; this paper). One unique characteristic of AHR78 is that it is highly expressed in the ovary during the vitellogenic stage (Fig. 3B). YPP genes are silent in the ovary despite the presence of both EcR and USP (Wang et al., 2000a). Taken together with the role of AHR78 in inhibition of the Vg promoter, it suggests that AHR78 may be responsible for the tissue-specific expression of the YPP genes, such as the inactivity of the Vg gene in the ovary. Further elucidation of the functions of AHR78 may provide new insights in our understanding the nature of ecdysone gene regulatory hierarchies in general as well as ecdysteroid regulation of mosquito vitellogenesis in particular. 193 ACKNOWLEDGMENTS This work was supported by a grant (A136959) from the National Institutes of Health. I thank Dr. Kook-Ho Cho for performing the ovary culture experiment, Mr. Alan Hays for his excellent assistance in maintaining the mosquito culture, Dr. M. Ken for contributive help at the initiation of this project, Dr. F. Kafatos for the generous gift of anti-DmUSP antibodies, Dr. M. McKeown for providing reporter constructs, and Dr. D. Martin for providing Vg-luc constructs. 194 Chapter 7 Summary and Future Research Perspectives 195 Summary and Future Research Perspectives Ecdysone gene regulation is one of best systems for studying eukaryotic transcription regulation. In Drosophila, this system has been studied for several decades, and recent discoveries have made significant advances in the molecular mechanism of the ecdysone gene regulatory hierarchies during metamorphosis. In contrast, in other insects, though ecdysone is one of the key hormones and regulates many events including development, metamorphosis and reproduction, the molecular basis of the ecdysone gene regulation hierarchy was poorly understood. This is particularly so in ecdysone gene regulation of insect reproduction. This insufficiency has made the general fundamental molecular mechanism of ecdysone gene regulation in insects unclear. In this dissertation, I have reported the extensive investigation of the molecular basis of ecdysone gene regulation in female mosquito reproduction. Characterizations of many elements of the ecdysone gene regulation cascade have demonstrated that the general molecular mechanism underlying ecdysone gene regulation appears to be conserved between mosquito vitellogenesis and fly metamorphosis. These suggest that despite the diversities of physiological functions of ecdysone in insects, the fundamental molecular features are considerably similar. In the future research, I propose to study the following aspects: Regulatory regions of the Aa USP gene(s). As described in Chapter 2, mosquito USP displays interesting characteristics: 1). The presence of two isoforms with distinct N-termini, 2). Differential expression and regulation by 20E of the two mosquito USP isoforms during vitellogenesis. 3). Direct 196 regulation of AaUSP-B by 20E and indirect regulation of AaUSP-A by 20E. The AaUSP-A expression profile negatively correlates with ecdysteroid titers while the AaUSP-B isoform expression shows positive correlation with ecdysteroid titers during mosquito vitellogenesis. In vitro organ culture experiments have suggested that the differential expression of mosquito USP isoforms is controlled by 20E. AaUSP-A expression is upregulated by the falling 20E titer while AaUSP-B is upregulated by the rising 20E titer. Moreover, the upregulation of AaUSP-B is enhanced in the presence of cycloheximide (Chx), a protein synthesis inhibitor; whereas the upregulation of AaUSP- A is abolished in the presence of Chx. These interesting differential regulation features lead us to ask the questions: what is the molecular basis of remarkable differential regulation of the two USP isoforms? How does the gene structure of USP facilitate this regulation? Does it result from alternative splicing of a single gene or from two separated genes? In insects, most known nuclear receptor isoforms are generated by alternative splicing and/or differential promoter utilization, though in vertebrates many nuclear receptor isoforms result from different genes. Examination of the mosquito USP gene will definitely provide more information about gene structure and regulation in general as well as those specifically involved in vitellogenesis. The clear answers to the questions above will first rely on genomic cloning of the mosquito USP gene. Using the two Aa USP cDNA clones to screen the genomic library can be the method to obtain the genomic structure of AaUSP. Genomic Southern blot can also be utilized to perform hybridization with probes generated by different S’UTR or N-termini of AaUSP cDNAs. In this way, it will be known whether the two AaUSP isoforms are derived from different genes or from different promoters of a single gene. 197 Moreover, further identification of the transcriptional units of Aa USP is necessary, which can be done by primer extension or RNase protection assays. The next step will be to clone the promoter regions of the two AaUSP isoforms and to functionally study the differential regulation of AaUSP by 20E. Once the two promoters are identified, I propose to construct a promoter-reporter plasmid and co-transfect them with EcR/USP into the Drosophila 82 cell line. We will then be able to examine how the two USP promoters respond to the presence or absence of 20E. Furthermore, a series of deletions of the AaUSP promoters will be constructed to map the differential responsive regions on the different promoters. EMSAs can be performed to test whether there is a direct binding site of EcR/U SP on the AaUSP-B promoter. Through this procedure, we will be able to obtain the answer concerning the ecdysone responsive regions of two AaUSP isoforms. The early gene (E 75) regulation in the ecdysone genetic regulatory hierarchy. As described in Chapter 3, the mosquito homolog of Drosophila E 75 has been cloned, which is a crucial early gene involved in Drosophila metamorphosis. E75 belongs to the nuclear receptor superfamily and has a counterpart in vertebrates, Rev-erb. Previous data has demonstrated that E 75 is directly induced and responsive earlier than Vg in the mosquito fat body by the steroid hormone 20E, suggesting that E 75 is involved in the activation and maintenance of the mosquito vitellogenic response to ecdysone. Next, I propose to use the biochemical and genetic approaches to investigate how E 75 is regulated and how it controls its target genes during mosquito vitellogenesis. 198 Three cDNA isoforms and the genomic DNA of mosquito E 75 have been cloned. I propose to extend these studies by identifying the transcription start site of each transcriptional unit, and further to identify the promoter of mosquito E 75. As shown in Chapter 3, E 75 gene expression is induced by 20E. After identifying the E 75 promoter, we can scan the promoter sequence for putative EcR/U SP binding sites using the consensus binding site of EcR/U SP as a reference. These putative binding sites will be tested using in vitro DNA binding assays to identify the EcR/U SP binding sites on the E 75 promoter. Furthermore, I propose to test whether these binding sites are functional in vivo by cell transfection assays, using reporter genes driven by the different E 75 promoter deletions, co-transfected with EcR/USP expression plasmids. Moreover, we can study the molecular basis of the auto-inhibition of the E75 gene. It has been demonstrated that the induction by 20E of E 75 is significantly increased in the presence of cycloheximide, a protein synthesis inhibitor, suggesting that the E75 protein likely represses its own gene promoter. I propose to utilize the same strategy used above to identify potential binding sites of E75 on its own gene promoter, and test the action of the E75 protein on its own promoter by cell transfection assays. Finally, we can characterize the interaction between E75 and EcR/U SP on the E 75 promoter, and investigate how activation by EcR/U SP and repression by E75 on the E 75 gene promoter lead to sustained expression of E 75. The induction of the E 75 gene positively correlates with that of vitellogenin gene both in vitro and in vivo. The putative E75 binding sites have been found on the Vg promoter (Martin and Raikhel, unpublished). Furthermore, it has been shown in cell transfection assays using the Vg promoter carrying a reporter gene that E75 up-regulates 199 the reporter expression (Chapter 3). Next, I propose to identify the functional binding sites of E75 on the Vg promoter, and then compare the differences between each E75 responsive element on the E 75 and Vg promoters. Additionally, we can test whether the different responsive elements result in different E75 activities. As shown in Chapter 3, EcR/U SP and E75 synergistically activated the Vg promoter in Drosophila S2 cells. Next, I propose to characterize the potential different molecular mechanisms of the action of both EcR/U SP and E75 on different promoters (Vg and E 75). Furthermore, we can use Drosophila to further characterize mosquito E 75, by crossing the Drosophila E 75 mutant or the ectopically inducible E 75 Drosophila with the mosquito-Vg-promoter- transgenic Drosophila, both of which are available. In the future, if possible, we can make a transgenic mosquito to characterize the action of E75 and EcR/U SP on the Vg promoter, so that we can obtain a much better understanding of the molecular mechanism of indirect ecdysone control of vitellogenic genes. Molecular basis of competence and its regulation for vitellogenic responses to ecdysone. Results from AaFTZ-Fl studies (Chapter 5) indicate that AaFTZ-Fl serves as a competence factor for controlling vitellogenic responses to ecdysone during the previtellogenic period. This is based on two lines of evidence: one is that in in vitro fat body culture, following the high level of AaFTZ-F lexpression, induction of E75A and VCP by 20E increases significantly; the second lines of evidence is that binding of nuclear extracts from newly enclosed, incompetent female fat bodies to the F TZ-Fl responsive element is undetectable, while binding is clearly detected in nuclear extracts 200 from 3~5-day-old competent fat bodies. These observations lead us to believe that competence of mosquito vitellogenic responses to ecdysone correlates with the appearance of functional AaFTZ-F 1 protein. Since AaFTZ-Fl mRNA is abundant throughout the entire previtellogenic stage, regulation of AaFTZ-Fl binding activities could be by either translational and/or post-translational control. Preliminary Western blot analysis with Drosophila F TZ-Fl antibodies indicated that AaFTZ-F 1 protein was present during the entire previtellogenic stage. This result needs to be confirmed by Western blot using antibodies specific to AaFTZ-Fl. If the possibility of translational control is ruled out, we can focus on examining post-translational control. A common post-translation modification is phosphorylation. We will first determine if this regulating system is being mediated by phosphorylation. Interestingly, steroidogenic factor 1 (SF -1), a vertebrate homolog of the insect FTZ-F l , has been demonstrated to be post-translationally regulated. Activation of SF -1 mediated transcription via interaction with nuclear receptor coactivators depends on the phosphorylation of a single serine residue in the AF -1 domain of SF -1 (Hammer et al., 1999). Moreover, the silkworm BmFTZ-F 1 activation function is mediated by the coactivator MBFI, which stabilizes the BmFTZ-F 1 -DNA complex (Li et al., 1994). To test whether phosphorylation of AaFTZ- F1 contributes to vitellogenic competence, we can introduce protein kinases or their inhibitors into EMSA experiments; or alternatively, in organ culture experiments, we can enhance/ inhibit the phosphorylation in the fat body by kinases or their inhibitors. Furthermore, another interesting question is whether the acquisition of competence via AaFTZ-Fl is under hormonal control. Early studies have established that the acquisition of competence by the mosquito fat body for the vitellogenic response to 201 Pm ecdysone is controlled by juvenile hormone (JH) (F lannagan and Hagedom, 1977). Treatment of previtellogenic incompetent fat bodies with JH renders this tissue capable of synthesizing vitellogenin protein in response to 20E (F lannagan and Hagedom, 1977; Ma etal., 1988). Our preliminary experiments have revealed that after incubation of newly eclosed incompetent fat bodies with 10'6 M JHIII for 24 hr followed by addition of 10'6 M 20E, the induction of VCP mRNA was significantly increased, as shown by RT- PCR/Southem blot (Li C, and Raikhel, A., unpublished). Therefore, one interesting question is whether JH-controlled competence and AaFTZ-F 1 -involved competence are in the same pathway. To answer this question, we can perform EMSAs to examine the DNA binding activity of nuclear extracts from newly eclosed, 24hr JH III-treated female fat bodies. This experiment will provide direct evidence of whether the active DNA- binding AaFTZ-Fl protein results from JH action in the fat body. In vertebrates, recent discoveries have established that two ligands, oxysterols and 25-Hydroxycholesterol, are capable of stimulating SF -1 dependant transcription. This suggests that SF -1 may be a nuclear hormone receptor rather than an orphan receptor (Bertherat, 1998). Therefore, a more interesting question for AaFTZ-F 1 will be whether it is a J H receptor. A simple experiment can be performed by cell transfection assay, using a promoter-reporter construct containing FTZ-F l responsive elements. Co- transfection of both this reporter construct and AaFTZ-F 1 expression plasmids into Drosophila 82 cells (and/or CV-l cells) in the presence or absence of JHIII or its analog methoprene should show if FTZ-Fl is responsive to JH or its analogs. 202 AHR 78 function in vitellogenic ovaries In Chapter 6, AHR78 studies have demonstrated that AHR78 is expressed at very low levels in the fat body and the previtellogenic ovary, but expressed at considerably high levels in the vitellogenic ovary. Functional analysis has established that AHR78 acts as a repressor, inhibiting DNA binding and transactivation of EcR/U SP. These results suggest that AHR78 may be involved in inhibiting activation of YPP genes in the ovary. To further support this hypothesis, first, we should specify the localization of AHR78 expression within the ovary due to the fact that the mosquito ovary contains several different tissues including oocytes, nurse cells and follicle cells. The main challenge of this experiment lies in the relatively low levels of expression of AHR78 mRNA. An AHR78 anti-sense mRN A probe can be used in in situ hybridization experiments. Antibodies against the AHR78 can also be raised to examine the AHR78 protein levels in the ovary. Second, further EMSA analysis of AHR78 inhibition of EcR/USP binding to EcREs will be conducted by nuclear extracts from both fat bodies and ovaries. Previous studies in our lab have successfully detected specific binding to EcREs of EcR/U SP from nuclear extracts in both fat bodies and ovaries (Miura et al., 1999). In this proposed study, in vitro produced AHR78 protein will be added into the binding reactions to test whether it will abolish the native EcR/USP proteins binding to EcREs. Furthermore, preliminary GST pull-down experiments indicate that AHR78 interacts with EcR and USP. Next, co-immunoprecipitation experiments will be designed to examine whether the interactions of AHR78 with EcR/USP occur in vivo. These studies will provide more valuable information regarding the potential inhibitory role of AHR78 on EcR/USP action. ' 203 REFERENCES 204 REFERENCES Allan, G.F., Leng, X., Tsai, S.Y., Weigel, N.L., Edwards, D.P., Tsai, M.J., and O'Malley, B.W. (1992). Hormone and antihorrnone induce distinct conformational changes which are central to steroid receptor activation. J. Biol. Chem. 267, 19513-19520. Antoniewski, C., Laval, M., Dahan, A., and Lepesant, J .A. (1994). The ecdysone response enhancer of the Fbpl gene of Drosophila melanogaster is a direct target for the EcR/USP nuclear receptor. Mol. Cell. Biol. 14, 4465-4474. Antoniewski, C., O'Grady, M.S., Edmondson, R.G., Lassieur, S.M., and Benes, H. (1995). Characterization of an EcR/U SP heterodimer target site that mediates ecdysone responsiveness of the Drosophila Lsp-2 gene. Mol. Gen. Genet. 249, 545-556. Antoniewski, C., Mugat, B., Delbac, F. and Lepesant, J.-A. (1996). Direct repeats bind the EcR/USP receptor and mediate ecdysteroid responses in Drosophila melanogaster. Mol. Cell. Biol. 16, 2977-2986. Asahina, M., Jindra, M., and Riddiford, L. M. (1997). Developmental expression of Ultraspiracle proteins in the epidermis of the tobacco homworm, Manduca sexta, during larval life and the onset of metamorphosis. Dev. Genes Evol. 207, 381- 388. Ashbumer, M. (1972). Puffing patterns in melanogaster and related species. Results Probl. Cell. Difler. 4, 101-151. Review. Ashbumer, M., Chihara, C., Meltzer, P., and Richards, G. (1974). Temporal control of puffing activity in polytene chromosomes. Cold Spring Harb. Symp. Quant. Biol. 38, 655-662. Bai, 1., Uehara, Y., and Montell, DJ. (2000). Regulation of invasive cell behavior by taiman, a Drosophila protein related to AIB l , a steroid receptor coactivator amplified in breast cancer. Cell 103, 1047-1058. Beato, M., Herrlich, P., and Schutz, G. (1995). Steroid hormone receptors: many actors in search of a plot. Cell. 83,851-7. Review. Beckemeyer, E. and Lea, A. (1980). Induction of follicle separation in the mosquito by physiological amounts of ecdysterone. Science 209, 819-821. Beier, J. C. (1998). Malaria parasite development in mosquitoes. Annu. Rev. Entomol. 43, 519-543. Bender, M., Imam, F. B., Talbot, W.S., Ganetzky, B., and Hogness, D. S. (1997). Drosophila ecdysone receptor mutations reveal functional differences among receptor isoforms. Cell 91, 777-788. 205 Bertherat, J. (1998). The nuclear receptor SF-l (steroidogenic factor-1) is no longer an orphan. Eur. J. Endocrinol. 138, 32-33. Review. Bose, S. G., and Raikhel, A. S. (1988). Mosquito vitellogenin subunits originate from a common precursor. Biochem. Biophys. Res. Commun. 155, 436-42. Bownes, M. (1986). Expression of genes coding for vitellogenin (yolk protein). Ann. Rev. Entomol. 31, 507-531. Bresnick, E.H., Dalman, F.C., Sanchez, E.R., and Pratt, W.B. (1989). Evidence that the 90-kDa heat shock protein is necessary for the steroid binding conformation of the L cell glucocorticoid receptor. J. Biol. Chem. 264, 4992-4997. Broadus, J ., McCabe, J. R., Endrizzi, B., Thummel, C. S, and Woodard, C. T. (1999). The Drosophila beta F TZ-Fl orphan nuclear receptor provides competence for stage- specific responses to the steroid hormone ecdysone. Mol. Cell 3, 143-149. Brown, M.R., Graf, R., Swiderek, K.M., Fendley, D., Stracker, T.H., Champagne, DE, and Lea, AD. (1998). Identification of a steroidogenic neurohormone in female mosquitoes. J. Biol. Chem. 273, 3967-3971. Burtis, K. C., Thummel, C. S., Jones, C. W., Karim, F. D., and Hogness, D. S. (1990). The Drosophila 74EF puff contains E74, a complex ecdysone-inducible gene that encodes two ets-related proteins. Cell 61, 85-99. Buszczak, M., Freeman, M., Carlson, J ., Cooley, L., and Segraves, W. A. (1999). Ecdysone response genes govern egg chamber development during mid-oogenesis in Drosophila. Development 26, 4581-4589. Butler, D., Maurice, J ., and O'Brien, C. (1997). Vaccines: a roller-coaster of hopes. Nature 386, 537-538. Carney, G.E., Wade, A.A., Sapra, R., Goldstein, ES, and Bender, M. (1997). DHR3, an ecdysone-inducible early-late gene encoding a Drosophila nuclear receptor, is required for embryogenesis. Proc. Natl. Acad. Sci. USA 94, 12024-12029. Chang C. and Kokontis J. (1988). Identification of a new member of the steroid receptor super-family by cloning and sequence analysis. Biochem. Biophys. Res. Commun. 155, 971-977. Chen, J .D. and Evans, RM. (1995). A transcriptional co-repressor that interacts with nuclear hormone receptors. Nature 377, 454-457. Cherbas, L., Fristrom, J .W., and O'Connor, J .D. (1984). The action of ecdysone in imaginal discs and Kc cells of Drosophila melanogaster. In: Biosynthesis, 206 metabolism and mode of action of invertebrate hormones (Hoffman, J and Porchet, M., eds.) pp. 305-322. Springer-Verlag, Berlin. Cherbas, L., Lee, K., and Cherbas, P. (1991). Identification of ecdysone response elements by analysis of the Eip28/29 gene. Genes Dev. 5, 120-131. Cho, W. L, Tsao, S. M., Hays, A. R., Walter, R., Chen, J. S., Snigirevskaya, E. S., and Raikhel, A. S. (1999). Mosquito cathepsin B-like protease involved in embryonic degradation of vitellin is produced as a latent extraovarian precursor. J. Biol. Chem. 274, 13311-13321. Cho, W. -L., Kapitskaya, M. Z., and Raikhel, A. S. (1995). Mosquito ecdysteroid receptor: analysis of the cDNA and expression during vitellogenesis. Insect Biochem. Mol. Biol. 25, 19-27. Cho, W.-L., and Raikhel, A. S. (1992). Cloning of cDNA for mosquito lysosomal aspartic protease. J. Biol. Chem. 267, 21823-21829. Cho, W.-L., Deitsch, K. W., and Raikhel, A. S. (1991). Extraovarian protein accumulated in mosquito oocytes is a serine carboxypeptidase activated in embryos. Proc. Natl. Acad. Sci. USA 88, 10821-10824. Christianson, A.M. K., King, D.L., Hatzivassiliou, E., Casas, J .E., Hallenbeck, P.L., Nikodem, V.M., Mitsialis, S.A., and Kafatos, F .C. (1992). DNA binding and heteromerization of the Drosophila transcription factor chorion factor l/ultraspiracle. Proc. Natl. Acad. Sci. USA. 89, 11503-11507. Collins, F. H., and Paskewitz, S. M. (1995). Malaria: current and future prospects for control. Annu. Rev. Entomol. 40, 195-219. D'Avino, P. P., Crispi, S., Cherbas, L., Cherbas, P., and Furia, M. (1995). The moulting hormone ecdysone is able to recognize target elements composed of direct repeats. Mol. Cell. Endocrinol. 113, 1-9. Davis, R.W., Botstein, D., and Roth, J .R. (1980). Advanced Bacterial Genetics. Cold Spring Harbor Laboratory, Cold Spring Harbor. DeFranco, D.B., Ramakrishnan, C. and Tang, Y. (1998). Molecular chaperones and subcellular trafficking of steroid receptors. J. Steroid Biochem. Mol. Biol. 65, 51- 58. Review. Deitsch, K. W., Chen, J .-S., and Raikhel, A. S. (l995a). Indirect control of yolk protein genes by 20-hydrocyecdysone in the fat body of the mosquito, Aedes aegypti. Insect Biochem. Mol. Biol. 25, 449-454. 207 Deitsch, K. W., Dittmer, N., Kapitskaya, M. Z., Chen, J .-S., Cho, W.-L., and Raikhel, A. S. (1995b). Regulation of gene expression by 20-hydroxyecdysone in the fat body of Aedes aegxpti (Diptera: Culicidae). Eur. J. Entomol. 92, 237-244. Deitsch, K.W. and Raikhel, AS. (1993). Cloning and analysis of the locus for mosquito vitellogenic carboxypeptidase. Insect Bioch. Molec. Biol. 2, 205-213. Dhadialla, T. S., and Raikhel, A. S. (1994). Endocrinology of mosquito vitellogenesis. In “Perspectives in Comparative Endocrinology” (K. G. Davey, R. E. Peter, and S. S. Tobe, Eds.), pp. 275-281. National Research Council, Canada. DiBello, P. R., Withers, D.A., Bayer, C. A., Fristrom, J. W., Guild, G. M. (1991). The Drosophila Broad-Complex encodes a family of related, zinc finger-containing proteins. Genetics 129, 385-397. Dittmann, F., Kogan, P. H., and Hagedom, H. H., (1989). Ploidy levels and DNA synthesis in fat-body cells of the adult mosquito, A edes Aegypti — the role of Juvenile-Hormone. Arch. Insect Biochem. 12, 133-143. Dobson, A.D., Conneely, O.M., Beattie, W., Maxwell, B.L., Mak, P., Tsai, M.J., Schrader, W.T. and O'Malley, B.W. (1989). Mutational analysis of the chicken progesterone receptor. J. Biol. Chem. 264, 4207-4211. Dressel, U., Thormeyer, D., Altincicek, B., Paululat, A., Eggert, M., Schneider, 8., Tenbaum, S.P., Renkawitz, R. and Baniahmad, A. (1999). Alien, a highly conserved protein with characteristics of a corepressor for members of the nuclear hormone receptor superfamily. Mol. Cell. Biol. 19, 3383-3394. Elliston, J .F ., Fawell, S.E., Klein-Hitpass, L., Tsai, S.Y., Tsai, M.J., Parker, MG. and O'Malley, B.W. (1990). Mechanism of estrogen receptor-dependent transcription in a cell-free system. Mol. Cell. Biol. 10, 6607-6612. F eigl, G., Gram, M., and Pongs, O. (1989). A member of the steroid hormone receptor gene family is expressed in the 20-OH-ecdysone inducible puff 75B in Drosophila melanogaster. Nucleic Acids Res. 17, 7167-7178. Fisk, G. J. and Thummel, C. S. (1995). Isolation, regulation, and DNA-binding properties of three Drosophila nuclear hormone receptor superfamily members. Proc. Natl. Acad. Sci. USA 92, 10604-8. Fisk, G. J. and Thummel, C. S. (1998). The DHR78 nuclear receptor is required for ecdysteroid signaling during the onset of Drosophila metamorphosis. Cell 93, 543-55. Flanagan, T. R., and Hagedom, H. H. (1977). Vitellogenin synthesis in the mosquito: the role of juvenile hormone in the development of responsiveness to ecdysone. 208 Physiol. Entomol. 2, 173-178. Fletcher, J .C. and Thummel, CS. (1995). The Drosophila E 74 gene is required for proper stage- and tissue-specific transcription of ecdysone-regulated genes at the onset of metamorphosis. Development 121, 1411-1421. Freedman LP. (1999). Increasing the complexity of coactivation in nuclear receptor signaling. Cell 97, 5-8. Review. Fujiwara, H., Jindra, M., Newitt, R., Palli, S. R., Hiruma, K., and Riddiford, L. M. (1995). Cloning of an ecdysone receptor homolog from Manduca sexta and the developmental profile of its mRNA in wings. Insect Biochem. Mol. Biol. 25, 845- 4 856. Garen, A.L., Kauvar, L., and Lepesant, J .-A. (1977). Roles of ecdysone in Drosophila development. Proc. Natl. Acad. Sci. USA 74, 5099-5103. Giguere, V., McBroom, L.D., and Flock, G. (1995). Determinants of target gene specificity for RORal: monomeric DNA binding by an orphan nuclear receptor. Mol. Cell. Biol. 15, 2517-2526. Giguere, V., Tini, M., Flock, G., Ong, E., Evans, R.M., Otulakowski, G. (1994). Isoforrn- specific amino-terminal domains dictate DNA-binding properties of ROR alpha, a novel family of orphan hormone nuclear receptors. Genes Dev. 8, 538-553. Glass, GK. and Rosenfeld, MG. (2000). The coregulator exchange in transcriptional functions of nuclear receptors. Genes Dev. 14, 121-141. Review. ~ Guay, P. and Guild, GM. (1991). The ecdysone-induced puffing cascade in Drosophila salivary glands: a Broad-Complex early gene regulates interrnolt and late gene transcription. Genetics 129, 169-175. Gwardz, R.W., and Spielman, A. (1973). Corpus allatum control of ovarian development in Aedes aegypti. J. Insect Physiol. 19, 1441-1448. Hagedom, H. H. (1989). Physiological roles of hemolymph ecdysteroids in adult insect. In “ Ecdysone, From Chemistry to Mode of Action”. (J. Koolman, Eds.), pp.279- 289. Thieme Medical Publishers, Inc., New York. Hagedom, H. H., and Fallon, A. M. (1973). Ovarian control of vitellogenin synthesis by the fat body in Aedes aegypti. Nature 244, 103-105. Hagedom, H. H., Fallon, A. M., and Laufer, H. (1973). Vitellogenin synthesis by the fat body of the mosquito Aedes aegypti: evidence of transcriptional control. Dev. Biol. 31, 285-294. 209 Hagedom, H., O'Connor, J ., Fuchs, M., Sage, B., Schlaeger, D., and Bohm, M. (1975). The ovary as a source of a-ecdysone in an adult insect. Proc. Natl. Acad. Sci. USA 72, 3255-3259. Hagedom, H.H. (1983). The role of ecdysteroids in the adult insect. In: Endocrinology of Insects (Downer, R. and Laufer, H., eds.) pp. 271-304. Liss, New York. Hagedom, H.H. (1985). The role of ecdysteroids in reproduction. In: Comprehensive Insect Physiology, Biochemistry and Pharmacology, Vol. 8 (Kerkut, G. and Gilbert, L., eds.) pp. 205-261. Pergamon, Oxford. Hall, B. L., and Thummel, C. S. (1998). The RXR homolog ultraspiracle is an essential component of the Drosophila ecdysone receptor. Development 125, 4709-4717. Hammer, G.D., Krylova, 1., Zhang, Y., Darimont, B.D., Simpson, K., Weigel, N.L., and Ingraham, HA. (1999). Phosphorylation of the nuclear receptor SF-l modulates cofactor recruitment: integration of hormone signaling in reproduction and stress. Mol. Cell. 3, 521-526. Hard, T., Kellenbach, E., Boelens, R., Maler, B.A., Dahlman, K., Freedman, L.P., Carlstedt-Duke, J ., Yamamoto, K.R., Gustafsson, J .A. and Kaptein, R. (1990). Solution structure of the glucocorticoid receptor DNA-binding domain. Science 249, 157-160. Hays, A. R. and Raikhel, A. S. (1990). A novel protein produced by the vitellogenic fat body and accumulated in mosquito oocytes. Roux ’s Arch. Dev. Biol. 199, 114- 121. Henrich, V. C., Sliter, T. J., Lubahn, D. B., Maclntyre, A., and Gilbert, L. I. (1990). A steroid/thyroid hormone receptor superfamily member in Drosophila melanogaster that shares extensive sequence similarity with a mammalian homologue. Nucleic Acids Res. 18, 4143-8. Henrich, V.C., Szekely, A.A., Kim, S.J., Brown, N.E., Antoniewski, C., Hayden, M.A., Lepesant, J .A., and Gilbert, L1. (1994). Expression and function of the ultraspiracle (usp) gene during development of Drosophila melanogaster. Dev. Biol. 165, 38-52. Hill, R.J., Segraves, W.A., Choi, D., Underwood, P.A., and Macavoy, E. (1993). The reaction with polytene chromosomes of antibodies raised against Drosophila E75A protein. Insect Biochem. Molec. Biol. 23, 99-104. Hiruma, K., Shinoda, T., Malone, F., and Riddiford, L. M. (1999). Juvenile hormone modulates 20-hydroxyecdysone—inducible ecdysone receptor and ultraspiracle gene expression in the tobacco hornworrn, Manduca sexta. Dev. Genes Evol. 209, 18-30. 210 ‘ Honda, 8., Morohashi, K., Nomura, M., Takeya, H., Kitajima, M., and Omura, T. (1993). Ad4BP regulating steroidogenic P-450 gene is a member of steroid hormone receptor superfamily. J. Biol. Chem. 268, 7494-502. Horlein, A.J., Naar, A.M., Heinzel, T., Torchia, J., Gloss, B., Kurokawa, R., Ryan, A., Kamei, Y., Soderstrom, M., Glass, GK. and Rosenfeld, MG. (1995). Ligand- independent repression by the thyroid hormone receptor mediated by a nuclear receptor co-repressor. Nature 377, 397-404. Homer, M.A., Chen, T., Thummel, CS. (1995). Ecdysteroid regulation and DNA binding properties of Drosophila nuclear receptor superfamily members. Dev. Biol. 168, 490-502. Huet, F ., Ruiz, C., and Richards, G. (1993). Puffs and PCR: the in vivo dynamics of early gene expression during ecdysone responses in Drosophila. Development 118, 613-627. Huet, F ., Ruiz, C., and Richards, G. (1995). Sequential gene activation by ecdysone in Drosophila melanogaster: the hierarchical equivalence of early and early late genes. Development 121, 1 195-1204. Jindra, M., Huang, J. Y., Malone, F ., Asahina, M., and Riddiford, L. M. (1997). Identification and mRNA developmental profiles of two ultraspiracle isoforms in the epidermis and wings of Manduca sexta. Insect Mol. Biol. 6, 41-53. J indra, M., Malone, F ., Hiruma, K., and Riddiford, L. M. (1996). Developmental profiles and ecdysteroid regulation of the mRNAs for two ecdysone receptor isoforms in the epidermis and wings of the tobacco homworrn, Manduca sexta. Dev. Biol. 180, 258-272. Jindra, M. and Riddiford, L.M. (1996). Expression of ecdysteroid regulated transcripts in the silk gland of the wax moth, Galleria mellonella. Dev. Gen. Evol. 206, 305- 314. J indra, M., Sehnal, F ., and Riddiford, L.M. (1994a). Isolation, characterization and developmental expression of the ecdysone-induced E75 gene of the wax moth Galleria mellonella. Eur. J. Biochem. 221, 665-675. Jindra, M., Sehnal, F ., and Riddifird, L.M. (1994b). Isolation and developmental expression of the ecdysteroid-induced GHR3 gene of the wax moth Galleria mellonella. Insect Biochem. Mol. Biol. 24, 763-773. Jones, G. and Sharp, RA. (1997). Ultraspiracle: an invertebrate nuclear receptor for juvenile hormones. Proc. Natl. Acad. Sci. USA. 94, 13499-13503. 211 Kamimura, M., Tomita, S., and Fujiwara, H. (1996). Molecular cloning of an ecdysone receptor (BI isoform) homologue from the silkworm, Bombyx mori, and its mRNA expression during wing disc development. Comp. Biochem. Physiol. B Biochem. Mol. Biol., 113, 341-347. Kamimura, M., Tomita, S., Kiuchi, M., and Fujiwara, H. (1997). Tissue-specific and stage-specific expression of two silkworm ecdysone receptor isoforms -- ecdysteroid-dependent transcription in cultured anterior silk glands. Eur. J. Biochem. 248, 786-793. Kapitskaya, M. Z., Li, C., Miura, K., Segraves, W., and Raikhel, A. S. (2000). Expression of the early-late gene encoding the nuclear receptor HR3 suggests its involvement in regulating the vitellogenic response to ecdysone in the adult mosquito. Mol. Cell. Endocrinol. 160, 25-37. Kapitskaya, M., Wang, S. -F., Cress, D. E., Dhadialla, T. S., and Raikhel, A. S. (1996). The mosquito ultraspiracle homologue, a partner of ecdysteroid receptor heterodimer: cloning and characterization of isoforms expressed during vitellogenesis. Mol. Cell. Endocrinol. 121, 119-132. Kapitskaya, M.Z., Dittmer, N.T., Deitsch, K.W., Cho, W.L., Taylor, D.G., Leff, T., and Raikhel, AS. (1998). Three isoforms of a hepatocyte nuclear factor-4 transcription factor with tissue- and stage-specific expression in the adult mosquito. J. Biol. Chem. 273, 29801-29810. Karim, F. D. and Thummel, CS. (1992). Temporal coordination of regulatory gene expression by the steroid hormone ecdysone. EMBO J. 11, 4083-4093. Kastner, P., Krust, A., Turcotte, B., Stropp, U., Tora, L., Gronemeyer. H., and Chambon, P. (1990). Two distinct estrogen-regulated promoters generate transcripts encoding the two functionally different human progesterone receptor forms A and B. EMBO J. 9, 1603-14. Keightley, M. C. (1998) Steroid receptor isoforms: exception or rule? Mol. Cell Endocrinol. 137, 1-5 Klein-Hitpass, L., Tsai, S.Y., Weigel, N.L., Allan, G.F., Riley, D., Rodriguez, R., Schrader, W.T., Tsai, M.J. and O'Malley, B.W. (1990). The progesterone receptor stimulates cell-free transcription by enhancing the formation of a stable preinitiation complex. Cell 60, 247-257. Klock, G., Strahle, U., and Schutz, G. (1987). Oestrogen and glucocorticoid responsive elements are closely related but distinct. Nature. 329, 734-736. Koelle, M. R., Talbot, W. S., Segraves, W..A., Bender, M. T., Cherbas, P., and Hogness, D.S. (1991). The Drosophila EcR gene encodes an ecdysone receptor, a new member of the steroid receptor superfamily. Cell 67, 59-77 212 Koelle, M.R., Segraves, W.A., and Hogness, D.S. (1992). DHR3: a Drosophila steroid receptor homolog. Proc. Natl. Acad. Sci. USA 89, 6167-6171. Kostrouch, Z., Kostrouchova, M., and Rall, J .E. (1995). Steroid/thyroid hormone receptor genes in Caenorhabditis elegans. Proc. Natl. Acad. Sci. USA 92, 156-159. Kozlova, T. and Thummel, C. S. (2000). Steroid regulation of postembryonic development and reproduction in Drosophila.Trends Endocrinol. Metab. 11, 276- 280. Review. Kuiper, G. G., Enmark, E., Pelto-Huikko, M., Nilsson, S., and Gustafsson, J. A. (1996). Cloning of a novel receptor expressed in rat prostate and ovary. Proc. Natl. Acad. Sci. USA 93, 5925-30. Kumar, V., Green, S., Stack, G., Berry, M., Jin, J.R., and Chambon, P. (1987). Functional domains of the human estrogen receptor. Cell 51, 941-951. Kurokawa, R., Soderstrom, M., Horlein, A., Halachmi, S., Brown, M., Rosenfeld, M.G., and Glass, CK. (1995). Polarity-specific activities of retinoic acid receptors determined by a co-repressor. Nature 377, 451-454. Lam, G., Hall, B.L., Bender, M., and Thummel, CS. (1999). DHR3 is required for the prepupal-pupal transition and differentiation of adult structures during Drosophila metamorphosis. Dev. Biol. 212, 204-16. Lam, G.T., J iang, C., and Thummel, CS. (1997). Coordination of larval and prepupal gene expression by the DHR3 orphan receptor during Drosophila metamorphosis. Development 124, 1757-1769. Lan, Q., Hiruma, K., Hu, X., Jindra, M., and Riddiford, L. M. (1999). Activation of a delayed-early gene encoding MHR3 by the ecdysone receptor heterodimer EcR- Bl-USP-l but not by EcR-Bl-USP-Z. Mol. Cell Biol. 19, 4897-4906. Lavorgna, G., Karim, F. D., Thummel, C. S., and Wu, C. (1993). Potential role for a FTZ-Fl steroid receptor superfamily member in the control of Drosophila metamorphosis. Proc. Natl. Acad. Sci. USA 90, 3004-3008. Lavorgna, G., Ueda, H., Clos, J ., and Wu, C. (1991). FTZ-Fl, a steroid hormone receptor-like protein implicated in the activation of fushi tarazu. Science 252, 848-851. Lazar, M.A., Hodin, R.A., Cardona, G., and Chin, W.W. (1990). Gene expression from the c-erbAa/Rev-erbAa genomic locus. J. Biol. Chem. 265, 12859-12863. 213 Lea, A. (1967). The medial neurosecretory cells and egg maturation in mosquitoes. J. Insect Phyio]. 13, 419-429. Lea, A. O. (1964). Studies on the dietary and endocrine regulation of autogenous Aedes taeniorhynchus. J. Med. Entomol. 1, 40-44. Lea, AD. (1967). The medial neurosecretory cells and egg maturation in mosquitoes. J. Insect. Phyiol. 13, 419-429. Lea, AD. (1972). Regulation of egg maturation in the mosquito by the neurosecretory system: the role of the corpus cardiacum. Gen. Comp. Endocrinol. Suppl. 3, 602- 608. Lee, H. L, and Archer, T. K. (1998). Prolonged glucocorticoid exposure dephosphorylates histone H1 and inactivates the MMTV promoter. EMBO J. 17, 1454-1466. Lehmann, M. and Korge, G. (1995). Ecdysone regulation of the Drosophila Sgs-4 gene is mediated by the synergistic action of ecdysone receptor and SEBP 3. EMBO J. 14, 716-726. Li, C., Kapitskaya, M. Z., Zhu, J ., Miura, K., Segraves, W., Raikhel, A. S. (2000). Conserved molecular mechanism for the stage specificity of the mosquito vitellogenic response to ecdysone. Dev. Biol. 224, 96-110. Li, F. Q., Ueda, H., and Hirose, S. (1994). Mediators of activation of fushi tarazu gene transcription by BmFTZ-F1.Mol. Cell. Biol. 14, 3013-3021. Lin, Y., Hamblin, M.T., Edwards, M.J., Barillas-Mury, C., Kanost, M.R., Knipple, D.C., Wolfner, M.F., and Hagedom, H.H. (1993). Structure, expression, and hormonal control of genes from the mosquito, Aedes aegypti, which encode proteins similar to the vitellin membrane proteins of Drosophila melanogaster. Dev. Biol. 155, 558-568. Luisi, B.F., Xu, W.X., Otwinowski, Z., Freedman, L.P., Yamamoto, K.R., and Sigler, RB. (1991). Crystallographic analysis of the interaction of the glucocorticoid receptor with DNA. Nature 352, 497-505. Ma, M., Zhang, J.-Z., Gong, H., and Gwardz, R. (1988). Permissive action of juvenile hormone on vitellogenin production by the mosquito, A edes aegypti. J. Insect Physiol. 34, 593-596. Mader, S., Kumar, V., dc Vemeuil, H., and Chambon, P. (1989). Three amino acids of the oestrogen receptor are essential to its ability to distinguish an oestrogen from a glucocorticoid-responsive element. Nature 338, 271-274. 214 Mangelsdorf, D. J., Thummel, C., Beato, M., Herrlich, P., Schutz, G., Umesono, K., Blumberg, B., Kastner, R, Mark, M., Chambon, P., and Evans, R. M. (1995). The nuclear receptor superfamily: the second decade. Cell 83, 835-839. Martin, D., Wang, S., and Raikhel, A. S. (2001). The vitellogenin gene of the mosquito Aedes aegypti is a direct target of ecdysteroid receptor. Mol. Cell. Endocrinol. 173, 75-86. Matsumoto, S., Brown, M., Suzuki, A., and Lea, A. (1989). Isolation and characterization of ovarian ecdysteroidogenic hormones from the mosquito, Aedes aegypti. Insect Biochem. 19, 651-656. McBroom, L.D., Flock, G., and Giguere, V. (1995). The non-conserved hinge region and distinct amino-terminal domains of the RORa orphan nuclear receptor isoforms are required for proper DNA bending and RORa-DNA interactions. Mol. Cell. Biol. 15, 796-808. McKenna, N.J., Lanz, RB, and O'Malley, B.W. (1999). Nuclear receptor coregulators: cellular and molecular biology. Endocr. Rev. 20, 321—44. Review. Miura, K., Wang, S.-F., and Raikhel, A. R. (1999). Two distinct subpopulations of ecdysone receptor complex in the female mosquito during vetellogenesis. Mol. Cell. Endocrinol. 156, 111-120. Miyajima, N., Horiuchi, R., Shibuya, Y., F ukushige, S., Matsubara, K., Toyoshima, K., and Yamamoto, T. (1989). Two erbA homologs encoding proteins with different T3 binding capacities are transcribed from opposite DNA strands of the same genetic locus. Cell 57, 31-39. Mouillet, J. F., Bousquet, F ., Sedano, N., Alabouvette, J ., Nicolai, M., Zelus, D., Laudet, V. and Delachambre, J. (1999). Cloning and characterization of new orphan nuclear receptors and their developmental profiles during Tenebrio metamorphosis. Eur. J. Biochem. 265, 972-981. Ohno, C. K., Ueda, H., and Petkovich, M. (1994). The Drosophila nuclear receptors FTZ-Fl alpha and FTZ-Fl beta compete as monomers for binding to a site in the fushi tarazu gene. Mol. Cell. Biol. 14, 3166-3175. O'Malley, B. W. and Conneely, O. M. (1992). Orphan receptors: in search of a unifying hypothesis for activation. Mol. Endocrinol. 6, 1359-1361. Onate, S. A., Tsai, S. Y., Tsai, M. J., and O'Malley, B. W. (1995). Sequence and characterization of a coactivator for the steroid hormone receptor superfamily. Science 270, 1354-1357. 215 Oro, A. E., McKeown, M., and Evans, R. M. (1990). Relationship between the product of the Drosophila ultraspiracle locus and the vertebrate retinoid X receptor. Nature 347, 298-301. Oro, A.E., McKeown, M., Evans, RM. (1992). The Drosophila retinoid X receptor homolog ultraspiracle functions in both female reproduction and eye morphogenesis. Development 115, 449-462. Paech, K., Webb, P., Kuiper, G. G., Nilsson, S., Gustafsson, J ., Kushner, P.J., and Scanlan, TS. (1997). Differential ligand activation of estrogen receptors ER-or and ER-B at AP] sites. Science 277, 1508-1510. Palli, S.R., Hiruma, K., Riddiford, L.M. (1992). An ecdysteroid-inducible Manduca gene similar to the Drosophila DHR3 gene, a member of the steroid hormone receptor superfamily. Dev. Biol. 150, 306-318. Palli, S.R., Ladd, T.R., Ricci, A.R., Sohi, SS, and Retnakaran, A. (1997). Cloning and developmental expression of Choristonerura hormone receptor 75: A homolog of the Drosophila E75A gene. Dev. Genet. 20, 36-46. Palli, S.R., Ladd, T.R., Sohi, S.S., Cook, B.J., and Retnakaran, A. (1996). Cloning and developmental expression of Chortstoneura hormone receptor 3, an ecdysone- inducible gene and a member of the steroid hormone receptor superfamily. Insect Biochem. Mol. Biol. 26, 485-499. Perera, S.C., Ladd, T.R., Dhadialla, T.S., Krell, P.J., Sohi, S.S., Retnakaran, A., and Palli, SR. (1999). Studies on two ecdysone receptor isoforms of the spruce budworm, Choristoneurafumiferana. Mol. Cell. Endocrinol. 152, 73-84. Perlmann, T., Rangarajan, P.N., Umesono, K., and Evans, RM. (1993). Determinants for selective RAR and TR recognition of direct repeat HREs. Genes Dev. 7, 1411- 1422. Picard, D., Khursheed, B., Garabedian, M.J., F ortin, M.G., Lindquist, S., and Yamamoto, KR. (1990). Reduced levels of hsp90 compromise steroid receptor action in vivo. Nature 348, 166-168. Pierceall, W. E., Li, C., Biran, A., Miura, K., Raikhel, A. S., and Segraves, W. A. (1999). E75 expression in mosquito ovary and fat body suggests reiterative use of ecdysone-regulated hierarchies in development and reproduction. Mol. Cell Endocrinol. 150, 73-89. Puigserver, P., Wu, Z., Park, C. W., Graves, R., Wright, M., and Spiegelman, B. M. (1998). A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell 92, 829-839. 216 Raikhel, A. S. (1992). Vitellogenesis in mosquitoes. Advance Disease Vector Res. 9, l- 39. Raikhel, A. S., and Lea, A. O. (1985). Hormone-mediated formation of the endocytic complex in mosquito oocytes. Gen. Comp. Endocrinol. 57, 422-433. Raikhel, A. S., and Lea, A. O. (1990). Juvenile hormone controls previtellogenic proliferation of ribosomal RNA in the mosquito fat body. Gen. Comp. Endocrinol. 77, 423-434. Raikhel, A. S., Deitsch, K. W., and Sappington, T. W. (1997). Culture and analysis of the insect fat body. In “The Molecular Biology of Insect Disease Vectors, A Methods Manual”. (J. M. Crampton, C. B. Beard, and C. Louis, Eds.), pp 507- 522. Chapman & Hall, London, UK. Raikhel, A.S., Deitsch, K.W., and Sappington, T.W. (1997). Culture and analysis of the insect fat body. In: The molecular biology of insect disease vectors. A methods manual (Crampton, J .M., Beard, CB. and Louis, C., eds.) pp. 507-522. Chapman and Hall, London. Raikhel, AS. and Lea, AD. (1991). Control of follicular epithelium development and vitelline envelope formation in the mosquito; role of juvenile hormone and 20- hydroxyecdysone. Tissue Cell 23, 577-591. Raikhel, A.S., Miura, K., and Segraves, W.A.(l999). Nuclear receptors in mosquito vitellogenesis. Amer. Zoo]. 39, 722-735. Rastinejad, F. (1998). Structure and function of the steroid and nuclear receptor DNA binding domain. In “Molecular Biology of Steroid and Nuclear Hormone receptors” (Leonard P. Freedman, Eds), pp. 105-132. Birkhauser Boston, Cambridge. Richards, G. (19763). The control of prepupal puffing patterns in vitro; implications for prepupal ecdysone titers in Drosophilia melanogaster. Dev. Biol. 48, 191-195. Richards, G. (1976b). Sequential gene activation by ecdysone in polytene chromosomes of Drosophila melanogaster. IV. The mid prepupal period. Dev. Biol. 54, 256- 263. Richards, G. (1976c). Sequential gene activation by ecdysone in polytene chromosomes of Drosophila melanogaster. V. The late prepupal puffs. Dev. Biol. 54, 264-275. Riddiford, L. M. (1985). Hormone action at the cellular level. In: “Comprehensive insect physiology, biochemistry, and pharmacology” (G.A. Kerkut and LI. Gibert, Eds), Vol. 8, pp. 37-84. Plenum, New York. 217 Riddiford, L. M. (1993a). Hormone and Drosophila development in “The Development of Drosophila melanogaster”, (M. Bate, and A. Martinez Eds.), Vol. 2, pp.899-939. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York. Riddiford, L.M. (1993b). Hormone receptors and the regulation of insect metamorphosis. Receptor 3, 203-209. Review. Riddiford, L.M. (1996). Juvenile hormone: The status of its “Status Quo” action. Arch. Insect Biochem. Physiol. 32, 271-286. Riddiford, L.M., Cherbas, P., and Truman, J.W. (2001). Ecdysone receptors and their biological actions. Vitam. Harm. 60,1-73. Review. Riddihough, G. and Pelham, H.R.B. (1987). An ecdysone responsive element in the Drosophila hsp27 promoter. EMBO J. 6, 3729-3 734. Robinow, S., Talbot, W. S., Hogness, D. S., and Truman, J. W. (1993). Programmed cell death in the Drosophila CNS is ecdysone-regulated and coupled with a specific ecdysone receptor isoform. Development 119, 1251-1259. Robyr, D., Wolffe, AP. and Wahli W. (2000). Nuclear hormone receptor coregulators in action: diversity for shared tasks. Mol. Endocrinol. 14, 329-347. Review. Russell, S. and Ashbumer, M. (1996). Ecdysone-regulated chromosome puffing in Drosophila melanogaster. IN: Metamorphosis (eds. Gilbert, L.I., Tata, J .R., and Atkinson, B.G.), pp. 109-144, Academic Press. Sappington, T. W. and Raikhel, A. S. (1999). Aedes aegypti. In “Encyclopedia of Reproduction” (E. Knobil and J. D. Neill, Eds), Vol. 1, pp.61-77. Academic Press, San Diego. Sappington, T. W., Brown M. R. and Raikhel, A. S. (1997). Culture and analysis of insect ovaries. In “Molecular Biology of Insect Disease Vectors: A methods manual” (J .M. Crampton, C.B. Beard and C.Louis, Eds), Chapter 42, pp.523-535. Chapman & Hall. Sappington, T.W., Kokoza, V.A., Cho, W.-L., and Raikhel, AS. (1996). Molecular characterization of the mosquito vitellogenin receptor reveals unexpected high homology to the Drosophila yolk protein receptor. Proc. Natl. Acad. Sci. USA 93, 8934-8939. Schubiger, M., Wade, A. A., Carney, G. E., Truman, J. W., and Bender, M. (1998). Drosophila EcR-B ecdysone receptor isoforms are required for larval molting and for neuron remodeling during metamorphosis. Development 125, 2053-2062. 218 Segraves, W.A. (1988). Molecular and genetic analysis of the E75 ecdysone-responsive gene of Drosophila melanogaster. Ph.D. thesis, Stanford University, Stanford, California. Segraves, W.A. and Hogness, D.S. (1990). The E75 ecdysone-inducible gene responsible for the 75B early puff in Drosophila encodes two new members of the steroid receptor superfamily. Genes Dev. 4, 204-219. Segraves, W.A. and Woldin, CW. (1993). The E75 gene of Manduca sexta and comparison with its Drosophila homolog. Insect Biochem. Molec. Biol. 23, 91-97. Shapiro, A.B., Wheelock, G. D., Hagedom, H. H., Baker, F. C. Tsai, L. W., and Schooley, D. H. (1986). Juvenile hormone and the juvenile hormone esterase in adult female of the mosquito Aedes aegypti. J. Insect Physiol. 32, 867-877. Shapiro, J. P. and Hagedom, H. H. ( 1982). Juvenile hormone and the development of ovarian responsiveness to a brain hormone in the mosquito, Aedes aegipti. Gen. Comp. Endocrinol. 46, 176-183. Shea, M. J., King, D. L., Conboy, M. J., Mariani, B. D., and Kafatos, F. C. (1990). Proteins that bind to Drosophila chorion cis-regulatory elements: a new C2H2 zinc finger protein and a C2C2 steroid receptor-like component. Genes Dev. 4, 1 128-1 140. Shi, Y. B., Wong, J ., Puzianowska-Kuznicka, M., and Stolow, M. A. (1996). Tadpole competence and tissue-specific temporal regulation of amphibian metamorphosis: roles of thyroid hormone and its receptors. Bioessays 18, 391-399. Stancato, L.F., Silverstein, A.M., Gitler, C., Groner, B., and Pratt, W.B. (1996). Use of the thiol-specific derivatizing agent N-iodoacetyl-3-[1251]iodotyrosine to demonstrate conformational differences between the unbound and hsp90—bound glucocorticoid receptor hormone binding domain. J. Biol. Chem. 271, 8831-8836. Steinbach, O. C., Wolffe, A. P., and Rupp, R. A. (1997). Somatic linker histones cause loss of mesodermal competence in Xenopus. Nature 389, 395-399. Stone, BL. and Thummel, CS. (1993). The Drosophila 78C early late puff contains E78, an ecdysone-inducible gene that encodes a novel member of the nuclear hormone receptor superfamily. Cell 75, 307—320. Sun, J ., Hiraoka, T., Dittmer, N.T., Cho, K.H., and Raikhel, AS. (2000). Lipophorin as a yolk protein precursor in the mosquito, Aedes aegypti. Insect Biochem. Mol. Biol. 30,1161-1171. 219 Sun, G. C., Hirose, S., and Ueda, H. (1994). Intermittent expression of BmFTZ-F l, a member of the nuclear hormone receptor superfamily during development of the silkworm Bombyx mori. Dev. Biol. 162, 426-437. Sutherland, J .D., Kozlova, T., Tzertzinis, G., Kafatos, F .C. (1995). Drosophila hormone receptor 38: a second partner for Drosophila USP suggests an unexpected role for nuclear receptors of the nerve growth factor-induced protein B type. Proc. Natl. Acad. Sci. USA 92, 7966-70. Swevers, L., Drevet, J. R., Junke, M. D., and latrou, K. (1995). The silkmoth homolog of the Drosophila ecdysone receptor (B1 isoform): cloning and analysis of expression during follicular cell differentiation. Insect Biochem. Mo]. Bio]. 25, 857-866. Syvala, H., Pekki, A., Blauer, M., Pasanen, S., Makinen, E., Ylikomi, T., and Tuohimaa, P. (1996). Hormone-dependent changes in A and B forms of progesterone receptor. J. Steroid. Biochem. Mol. Biol. 58, 517-524. Talbot, W. S., Swyryd EA, and Hogness D.S. (1993). Drosophila tissues with different metamorphic responses to ecdysone express different ecdysone receptor isoforms. Cell 73, 1323-1337. Thomas, H. E., Stunnenberg, H. G., and Stewart, A. F. (1993). Heterodimerization of the Drosophila ecdysone receptor with retinoid X receptor and ultraspiracle. Nature 362, 471-475. Thummel, CS. (1992). Mechanisms of transcriptional timing in Drosophila. Science 255, 39-40. Thummel, CS. (1995). From embryogenesis to metamorphosis: the regulation and function of Drosophila nuclear receptor superfamily members. Cell 83, 871-877. Review. Thummel, CS. (1996). Files on steroids--Drosophila metamorphosis and the mechanisms of steroid hormone action. Trends Genet. 12, 306-310. Review. Thummel, CS. (1997). Dueling orphans--interacting nuclear receptors coordinate Drosophila metamorphosis. Bioessays 19, 669-672. Review. Thummel, C.S., Burtis, K. C., and Hogness, D.S. (1990). Spatial and temporal patterns of E 74 transcription during Drosophila development. Cell 61, 101-111. Truman, J. W., Talbot, W. S., Fahrbach, S. E., and Hogness, D. S. (1994). Ecdysone receptor expression in the CNS correlates with stage-specific responses to ecdysteroids during Drosophila and Manduca development. Development 120, 219-234. 220 Tsai, C.C., Kao, H.Y., Yao, T.P., McKeown, M., and Evans, R.M. (I999). SMRTER, a Drosophila nuclear receptor coregulator, reveals that EcR-mediated repression is critical for development. Mol. Cell. 4, 175-186. Tsai, M.J. and O'Malley, B.W. (1994). Molecular mechanisms of action of steroid/thyroid receptor superfamily members. Annu. Rev. Biochem. 63, 451-486. Review. Tsai, S.Y., Srinivasan, G., Allan, G.F., Thompson, E.B., O'Malley, B.W., and Tsai, M.J. (1990). Recombinant human glucocorticoid receptor induces transcription of hormone response genes in vitro. J. Biol. Chem. 265, 17055-17061. Tugwood, J. D., Issemann, I., and Green, S. (1991). Liver receptor homologue protein. GenBank DNA Database Accession No. M81385. Tzertzinis, G., Malecki, A., and Kafatos, F.C. (1994). BmCFl, a Bombyx mori RXR-type receptor related to the Drosophila ultraspiracle. J. Mol. Bio]. 238, 479-486. Ueda, H. and Hirose, S. (1991). Defining the sequence recognized with BmFTZ-Fl, a sequence specific DNA binding factor in the silkworm, Bombyx mori, as revealed by direct sequencing of bound oligonucleotides and gel mobility shifi competition analysis. Nucleic Acids Res. 19, 3689-3693. Ueda, H., Sonoda, S., Brown, J. L., Scott, M. P., and Wu, C. (1990). A sequence-specific DNA-binding protein that activates fushi tarazu segmentation gene expression. Genes Dev. 4, 624-635. Ueda, H., Sun, G. C., Murata, T., and Hirose, S. (1992). A novel DNA-binding motif abuts the zinc finger domain of insect nuclear hormone receptor FTZ-Fl and mouse embryonal long terminal repeat-binding protein. Mol. Cell. Biol. 12, 5667- 5672. Umesono, K., Murakami, K.K., Thompson, CC, and Evans, R.M. (1991). Direct repeats as selective response elements for the thyroid hormone, retinoic acid, and vitamin D3 receptors. Cell 65, 1255-1266. Vogtli, M., Elke, C., Imhof,.M.O., and Lezzi, M. (1998). High level transactivation by the ecdysone receptor complex at the core recognition motif. Nucleic Acids Res. 26, 2407-2414. Vogtli, M., Imhof, M.G., Brown, N.E., Rauch, P., Spindler-Barth, M., Lezzi, M., and Henrich, V.C. (1999). Functional characterization of two Ultraspiracle forms (CtUSP-l and CtUSP-Z) from Chironomus tentans. Insect Biochem. Mol. Bio]. 29, 931-942. 221 Walker, V.K. and Ashbumer, M. (1981). The control of ecdysterone-regulated puffs in Drosophila salivary glands. Cell 61, 101-111. Wang, S.-F., Li, C., Zhu, J., Miura, K., Miksicek, R. J ., and Raikhel, A. S. (20003). Differential expression and regulation by 20-hydroxyecdysone of mosquito ultraspiracle isoforms. Dev. Biol. 218, 99-113. Wang, S. F ., Ayer, S., Segraves, W. A., Williams, D. R., and Raikhel, A. S. (2000b). Molecular determinants of differential ligand sensitivities of insect ecdysteroid receptors. Mol. Cell. Biol. 20, 3870-3879. Wang, S.-F., Miura, K., Miksicek, R. J ., Segraves, W. A., and Raikhel, A. S (1998). DNA binding and transactivation characteristics of the mosquito ecdysone receptor- ultraspiracle complex. J. Biol. Chem. 273, 27531-27540. White, K.P., Hurban, P., Watanabe, T., and Hogness, D.S. (1997). Coordination of Drosophila metamorphosis by two ecdysone-induced nuclear receptors. Science 276,114-117. Wilson, T. E., F ahmer, T. J ., and Milbrandt, J. (1993). The orphan receptors NGFI-B and steroidogenic factor I establish monomer binding as a third paradigm of nuclear receptor-DNA interaction. Mo]. Cell. Biol. 13, 5794-5804. Woodard, C. T., Baehrecke, E. H., and Thummel, C. S. (1994). A molecular mechanism for the stage specificity of the Drosophila prepupal genetic response to ecdysone. Cell 79, 607-615. Xu, L., Glass, C.K., and Rosenfeld, MG. (1999). Coactivator and corepressor complexes in nuclear receptor function. Curr. Opin. Genet Dev. 9, 140-147. Yao, T.-P., Forman, B.M., Jiang, Z., Cherbas, L, Chen, J.-D., McKewon, M., Cherbas, P., and Evans, R.M. (1993). Functional ecdysone receptor is the product of EcR and Ultraspiracle genes. Nature 336, 476-479. Yao, T.-P., Segraves, W.A., Oro, A.E., McKeown, M., and Evans, R.M. (1992). Drosophila ultraspiracle modulates ecdysone receptor function via heterodimer formation. Cell 71, 63-72. Zamir, 1., Dawson, J., Lavinsky, R.M., Glass, C.K., Rosenfeld, M.G., and Lazar, MA. (1997). Cloning and characterization of a corepressor and potential component of the nuclear hormone receptor repression complex. Proc. Natl. Acad. Sci. USA. 94, 14400- 14405. Zechel, C., Shen, X.Q., Chen, J .Y., Chen, Z.P., Chambon, P., and Gronemeyer, H. (1994). The dimerization interfaces formed between the DNA binding domains of RXR, RAR and TR determine the binding specificity and polarity of the full- length receptors to direct repeats. EMBO J. 13, 1425-1433. 222 Zelhof, A. C., Yao, T. P., Chen, J. D., Evans, R. M., and McKeown, M. (1995a). Seven- up inhibits ultraspiracle-based signaling pathways in vitro and in vivo. Mol. Cell. Biol. 15, 6736-45. Zelhof, A. C., Yao, T. P., Evans, R. M., and McKeown, M. (l995b). Identification and characterization of a Drosophila nuclear receptor with the ability to inhibit the ecdysone response. Proc. Natl. Acad. Sci. USA 92, 10477-81. Zhou, B., Hiruma, K., Jindra, M., Shinoda, T., Segraves, W.A., Malone, F ., and Riddiford, L.M. (1998). Regulation of the transcription factor E75 by 20- hydroxyecdysone and juvenile hormone in the epidermis of the tobacco homworm, Manduca sexta, during larval molting and metamorphosis. Dev. Biol. 193, 127-138. Zhu, J ., Miura, K., Chen, L., and Raikhel, A. S. (2000). AHR38, a homolog of NGFI-B, inhibits formation of the functional ecdysteroid receptor in the mosquito Aedes aegypti. EMBO J. 19, 253-62. 223