IIIIIIIIIIIII THESIS 7L 20Gb LIBRARY Michigan State University This is to certify that the dissertation entitled The Regulation of the Mixed-Lineage Kinase SPRK/MLK-3 by the Small GTPase Cdc42 presented by Barbara Carola Bock has been accepted towards fulfillment of the requirements for Ph .D. degree in Physiology jam“ ,4 . «SW’ Major professor Date $4101) MS U it an Affirmative Action/Equal Opportunity Institution 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 W 1i 2002 OCT M. 2.. iNI 11/00 macros-unwed“ THEREGL SPRl THE REGULATION OF THE MD(ED-LINEAGE KINASE SPRK/MLK-3 BY THE SMALL GTPase Cdc42 By Barbara Carola Bock A DISSERTATION Submitted to Michigan State University In partial fulfillment of the requirements For the degree of DOCTOR OF PHILOSOPHY Department of Physiology 2000 m REGLUTK Sr: m: A i" 4 Q . ' tum Irma-mart ovcexprtssm In F [Mime 2mm dim ' In to Its ma Hfil e mm the I'r Kan. TM we first Bork erv SPRK arm-m. Comte g\.\lfl\r it? F. t“- 3PM «_ ”\ufifi . ~ (I 82“» :59): I‘m “4t rt! “WA:- ' ‘I‘ it I 4E \— It???“ ABSTRACT THE REGULATION OF THE MIXED-LINEAGE KIN ASE SPRK/MLK-3 BY THE SMALL GTPase Cdc42 By Barbara Carola Bock Src homology 3 domain containing proline-rich protein kinase (SPRK), also called mixed-lineage kinase (MLK)-3 is a serine/threonine kinase which upon overexpression in mammalian cells activates the c-Jun NHz-terminal kinase (JNK)/stress activated kinase (SAPK) pathway. The 95 kDa protein bears, in addition to its kinase domain, various motifs mediating protein-protein interaction. However the mechanism by which SPRK activity is regulated still remains unclear. This thesis work examines the role of the small GTPase Cdc42 in the regulation of SPRK activity. Coexpression of SPRK with Cdc42 in 293 cells results in an increase in SPRK's catalytic activity. Mutational analysis of the Cdc42/Rac interactive-binding (CRIB) motif, a consensus motif required for binding of small GTPases, shows that SPRK contains a functional CRIB motif, since mutations within this motif abrogate association of SPRK with Cdc42, and thus Cdc42-induced SPRK activation. Further experiments using a SPRK variant which lacks the zipper region/basic stretch suggest that not only the CRIB motif is important for Cdc42 binding, but that also the zipper/basic stretch contributes to Cdc42 binding. Interestingly, SPRK activation by Cdc42 cannot be recapitulated in an in vitro system using recombinant, purified proteins. This suggests that Cdc42-induced activation of SPRK requires the cellular environment. Comparative two-dimensional in vivo phosphopeptide mapping indicated that coexpression of Cdc42 changes SPRK's in vivo phosphorylation pattern, supporting the idea that Cdc42 changes the in viva phosphorylation of SPRK. Analysis of the subcellular distribution of SPRK showed that the majority of SPRK upon overexpression in COS-7 cells colocalizes with the Golgi apparatus. Coexpression of SPRK and Cdc42 induces targeting of SPRK to a plasma membrane-enriched subcellular fraction. The targeting requires prenylation of Cdc42, suggesting the requirement for Cdc42 in this process. The present study demonstrates that Cdc42 is an activator of SPRK and provides important insight into the structural requirements for Cdc42-induced SPRK activation. The observation that Cdc42 changes SPRK's subcellular distribution suggests a role for Cdc42 in membrane targeting, although it was not possible to correlate the subcellular targeting of SPRK with an increase in kinase activity. iv To my family ACKNOWELEDGEMENTS I would like to thank my committee members Drs. Kathleen Gallo, Susan Conrad, Laura McCabe, Richard Miksicek, and William Spielman. I would especially like to thank my mentor Dr. Kathleen Gallo for the constant encouragement, guidance, and support throughout my graduate career. Furthermore I would like to thank Drs. Susan Conrad and Walter Esselman for the useful discussions during the Friday afternoon meetings. I would also like to thank Drs. Joanne Whallon and Shirley Owens for the help with confocal imaging at the Laser Scanning Microscope Laboratory at Michigan State University. I would also like to take this opportunity to thank my friends in the lab, especially Panayiotis Vacratsis, Erion Qamirani, Ritesh Agrawal, Hua Zhang, Mary Chao and David Allton. I am grateful to Panayiotis Vacratsis for his support during this project and constructive discussions. I would like to sincerely thank the office staff for all the help rendered to me. LN of Tables ....... 5.9‘!" W'JM TABLE OF CONTENTS Page List of Tables .............................................................................................................. viii List of Figures .............................................................................................................. .ix Key to Abbreviations .................................................................................................. .xii I. Introduction ............................................................................................................... .1 11. Literature Review ...................................................................................................... 4 1. The Regulation of Protein Kinases ................................................... 5 1.1 Regulation of protein kinases by activators and inhibitors............... . . . . . . . .5 1.2 Regulation of protein kinases by phosphorylation ..................................... 7 1.3 Regulation of protein kinases by proteolytic cleavage.................. . ..........7 1.4 Regulation of protein kinases by subcellular distribution .......................... 8 2. Small GTPases of the Rho family .................................................................. .14 3. The MAPK Pathways ..................................................................................... .22 4. The Physiological Importance of the IN K Pathway ...................................... .30 4.1 JN K signaling in Drosophila melanogaster ........................................... .30 4.2 INK signaling in mammals ...................................................................... 32 5. The Mixed-Lineage Kinase Family ................................................................. 37 III. Cdc42-Induced Activation of the Mixed-Lineage Kinase SPRK in Vivo: Requirement of the Cdc42/Rac Interactive Binding Motif and Changes in Phosphorylation .......................................................................... .46 1. Abstract ........................................................................................................... 46 2. Introduction ..................................................................................................... 47 3. Materials and Methods ................................................................................... .49 3.1 Construction of mammalian expression vectors and mutagenesis.... . . . . . .49 3.2 Expression and purification of recombinant SPRK. ............................... . 51 3.3 Cell lines and transfections ........................................................................ 52 3.4 Cell lysis and immunoprecipitations ....................................................... .52 3.5 Gel electrophoresis and Western blot analysis ........................................ .53 3.6 In vitro kinase assays ............................................................................... .54 3.7 In vivo phosphopeptide mapping ............................................................. .55 vi 4. Rcsuits ...... H ANY; SPRK .' 4.2 SPRK are for! 4.3 fffgix 2;, r; 4.4 SPRK" . 4.5 Acting- 4.6 CU: . 5- DECEM‘?! IV. (at: 0 n 3,; KL,“\\ lined-Laces . 1' ARIA”. , 3~ Miler-115$ 31 COD? I view 4. Results ............................................................................................................ .57 4.1 Association of activated Cdc42 with SPRK does not require SPRK kinase activity ............................................................................... .57 4.2 SPRK's CRIB motif is necessary for association with Cdc42 and for Cdc42-induced activation of SPRK ............................................. 60 4.3 Effects of deleting the COOH-terminal portion of SPRK's zipper domain ........................................................................................... 61 4. 4 SPRK“? fails to activate INK ............................................................... .63 4. 5 Activated Cdc42 fails to activate SPRK in vitro ..................................... .64 4. 6 Cdc42 alters the in vivo phosphorylation pattern of SPRK .................... .65 5. Discussion ........................................................................................................ 84 IV. Cdc42 Changes the Subcellular Distribution of the Mixed—Lineage Kinase SPRK. .............................................................................. 91 1. Abstract ........................................................................................................... 91 2. Introduction ..................................................................................................... 92 3. Materials and Methods ................................................................................... .95 3.1 Construction of mammalian expression vectors and site-directed mutagenesis ..................................................................... .95 3.2 Cell lines and transfections ...................................................................... 96 3.3 Cell lysis, membrane fractionation, and irnmunoprecipitation................ 96 3.4 Gel electrophoresis and Western blot analysis ........................................ 98 3.5 In vitro kinase assays and in vivo phosphopeptide mapping ................... 99 3.6 Immunofluorescence and microscopy .................................................... 100 3.7 Protein Markers ..................................................................................... 101 4. Results ........................................................................................................... 102 4.1 Analysis of cellular fractions ................................................................. .102 4.2 Activated Cdc42 targets SPRK to cellular membranes .......................... 104 4.3 Analysis of the subcellular distribution of SPRK by confocal microscopy .......................................................................... 105 4.4 Analysis of SPRK distribution by biochemical fractionation ................. 109 4.5 Effect of membrane targeting on SPRK kinase activity ..................... 113 5. Discussmnl38 V. Summary and Conclusnon 145 VI. List of References 150 vii Iablc ’4 1 . ARA s15 nj bitumirzi- frailhfi} if. F Estlm",34. a Aun,~\‘ U. aflmg My o .'0 .' . Exifmfg . H . LIST OF TABLES Table Page 1. Analysis of subcellular fractions by marker proteins and biotynilation .......................................................................... 1 l7 2. Estimated distribution of total cellular non-nuclear protein among biochemical fractions ...................................................... 127 3. Estimated distribution of SPRK protein among biochemical fractions in presence and absence of Cdc42VI ................................. 128 viii LIST OF FIGURES Figure Page 1. MAPK scaffold complexes in Saccharomyces cervisiae ..................................... .12 2. Mammalian scaffold complexes ........................................................................... 13 3. Regulation of small GTPases ................................................................................ 21 4. MAPK pathway module ....................................................................................... .23 5. Parallel mammalian MAPK pathways .................................................................. 29 6. Domain arrangement of SPRK ............................................................................. .45 7. Schematic of SPRK ............................................................................................. .68 8. Alignment of CRIB motifs and SPRK variants .................................................... 69 9. Coimmunoprecipitation of SPRK with wild type Cdc42 or Cdc42V'2 ................. 7O 10. Coimmunoprecipitation of SPRKK|44A with activated Cdc42V12 ......................... .71 11. In vitro kinase assays of SPRK and SPRKKIM ................................................... 72 12. Effects of mutations in the CRIB motif on association of SPRK with Cdc42Vl2 .............................................................................................................. . 73 13. Effects of mutations in the CRIB motif on SPRK in vitro kinase activity........... 74 14. Quantitation of in vitro kinase assays of SPRK and SPRK‘492A'S493A .................. 75 15. Effects of partial deletion of the zipper/basic stretch on association with Ccia42Vlz ...................................................................................................... . 76 16. Effects of partial deletion of the zipper/basic stretch on SPRK in vitro kinase activity ........................................................................................................ 77 ix 17. Quantum of 18.135526 of SPRE 19. Qamitarssrt of . , .0. in mm» time - ‘ I .l. PmsFI‘t-"IY"" ‘3- . ‘ r \IU\ . ‘ \ 1 pmxprtorx Lat-:3. Vt *1 .~ . .... games: or ‘- Q as .3. mahxis of \ ‘w 1 3.31! ‘ ‘s - S‘JIY°I" ‘ . \.4‘- ‘1: "K ‘ l7. Quantitation of in vitra kinase assays of SPRK and SPRK“ip ............................ 78 18. Effects of SPRKAzip on JNK activity ................................................................... . 79 19. Quantitation of IN K in vitra kinase assay ........................................................... . 80 20. In vitra kinase assay using purified Cdc42 .......................................................... . 81 21. Phosphopeptide mapping of tryptic peptides derived from in viva phosphorylated SPRK. ......................................................................................... .82 22. Alignment of various Cdc42 variants .................................................. 118 23. Analysis of subcellular fractions ........................................................ 119 24. Subcellular distribution of SPRK ....................................................... 120 25. Subcellular localization of SPRK ....................................................... 121 26. Time course of SPRK and Cdc42 expression ....................................... 122 27. Subcellular distribution of coexpressed SPRK and Cdc42V12 ...................... 123 28. Subcellular distribution of Cdc42V12 and Cdc42V'2‘3'885 ............................ 124 29. Subcellular localization of coexpressed SPRK and Cdc42“20888 ................ 125 30. SPRK's subcellular distribution u n coexpression with Cdc42w2'C1885 and Cdc42“2088 ”’40 ............................................... 126 31. Coirnmunoprecipitation of SPRK and Cdc42 variants ............................... 129 32. Subcellular distribution of SPRKWA and SPRKM" ................................ 130 33. Subcellular distribution of endogenous SPRK ........................................ 131 34. Electrophoretic mobility shift of SPRK ............................................... 132 35. Phosphopeptide mapping of in viva phosphorylated SPRK ........................ 133 36. SPRK in vitra kinase activity upon coexpression with Cdc42V12 or ct1c42‘“2‘C1888 .............................................................. 134 x 37. SPRKLri 31.71" t c142“- or c. 38. Kinase mu :2\ 39.1,: 'i'lt'm kit-ENC _ 37. SPRK in vitra kinase activitIyRupon coexpression with 4Q Cdc42V12 or Cdc42V'2c'885' ....................................................... 135 38. Kinase activity of membrane-targeted SPRK ......................................... 136 39. In vitra kinase assay of endogenous, membrane-targeted SPRK .................. 137 xi ACK AIDS ATP cAMP GAP GDP GEF cGMP GST GTP GTPyS DLK ES ERK HPK IKK IKKK JN K LZK MAPK MAPKK MAPKKK MEK MLK NAK Nef NIK p38/RK PAGE PAK PBS PCR PKA PKC SPRK KEY TO ABBREVIATIONS Activated Cdc42HS-associated kinase Acquired immune deficiency syndrome Adenosine 5' -triphosphate Adenosine 3' -5' cyclic monophosphate GTPase activating protein Guanosine 5' -diphosphate Guanine nucleotide exchange factor Guanosine 3' -5' cyclic monophosphate Glutathione S-transferase Guanosine 5' -triphosphate Guanosine 5' -3-0-(thio)triphosphosphate Cdc42/Rac interactive binding Dual leucine zipper-bearing kinase Embryonic stem cell Extracellular signal-regulated protein kinase Human immune deficiency virus Human hematopoietic kinase IKB-kinase IKB-kinase kinase Interleukin Interferon JN K interacting protein c-Jun NHz-terminal kinase Leucine zipper bearing kinase Mitogen-activated kinase Mitogen-activated kinase kinase Mitogen-activated kinase kinase kinase MAPK kinase Mixed lineage-kinase Nef associated kinase Negative factor NCK interacting kinase p38/reactivating kinase Polyacrylamide gel electrophoresis p21-activated kinase Phosphate-buffered saline Polymerase chain reaction Protein kinase A Protein kinase C Ste-homology 3 domain-containing proline-rich protein kinase xii SRF TCF lGF TLC TNT WASP WI Serum response factor Ternary complex factor Transforming growth factor Thin layer chromatography Thin layer electrophoresis Tumor necrosis factor Thymocyte helper cell Wiskott Aldrich syndrome protein Wild type xiii Pmiczn l: | trmxnpim to i I PIOCCVCS ll 15 CK" | Badmz of arm; ‘ su'tttzztzs tags, SIC-Ira: u 1 53.463 taxed-11:32.. .PTM {ENC (' baffle it: sit?“ G 6.4.... mics. oni} sent. | 9:41.. 19941. SPF: I. Introduction Protein kinases regulate a variety of cellular processes, ranging from transcription to long term memory. Since these enzymes control so many different processes it is clear that protein kinases themselves have to be highly regulated. Binding of activating or inhibitory molecules, posttranslational modifications, and subcellular targeting are mechanisms that regulate protein kinases. Src-homology 3 domain-containing proline-rich protein kinase (SPRK), also called mixed-lineage kinase (MLK)-3 (Ing et al., 1994), or protein tyrosine kinase (PTK)-1 (Ezoe et al., 1994), is a member of the mixed-lineage kinase family. Despite the similarity of SPRK’s kinase domain to both serine/threonine and tyrosine kinases, only serine/threonine kinase activity has been demonstrated in vitra (Gallo et al., 1994). SPRK, with a predicted molecular weight of 95 kDa, contains several domains that may mediate protein-protein interactions, including a Src-homology (SH) 3 domain, a closely, spaced leucine/isoleucine zipper motif, a Cdc42/Rac interactive binding (CRIB) motif, and a proline/serine/threonine-rich COOH- terminal region. The CRIB motif is a sequence of 14-16 amino acids, which is required for binding of Rho family GTPases (Burbelo et al., 1995). SPRK shares six of the eight consensus residues, and a fragment of SPRK containing the CRIB motif has been demonstrated to associate with Cdc42 and Rac in filter binding assays (Burbelo et al., 1995). SPRK and Cdc42 have been identified as upstream activators of the c-Jun NHz-terminal kinase (INK). Although evidence exists that the small GTPase Cdc42 as timed-image 11.7 I Hermann b) \i an largely unkno- Strutwral require: GTPase CdtJZ. .. actuation b} C3“; This them IIIL'OduCtion and s“ and fitment l. _ . {Its-’51- WOlClIl kinaf {c stated protein W, .taml} {Ocuxcs on 0 i {the literature r . l 1L ’3. . u... GTPase Cdc42 associates with SPRK and may be an upstream regulator of this mixed-lineage kinase (Teramoto et al., 1996), the structural requirements and mechanisms by which this small GTPase binds to SPRK and modulates its activity are largely unknown. Accordingly the major aims of this thesis are: (1) to define the structural requirements for activation of the protein kinase SPRK by the small GTPase Cdc42, and (2) to investigate the role of subcellular targeting for SPRK activation by Cdc42. This thesis is essentially divided into five chapters. Chapter I includes a short introduction and states the major aims and hypotheses. A review of the relevant past and current literature in Chapter U will summarize our current understanding of protein kinase regulation and of Rho-family GTPases, as well as of mitogen- activated protein kinase (MAPK) signaling. The summary of MAPK signaling mainly focuses on the INK pathway and its physiological importance. The last part of the literature review will summarize the current knowledge about the mixed- lineage kinase family. Chapter 111 describes the research project which focuses on the regulation of SPRK by Cdc42. This research project is designed to test the hypotheses that (1) that the small GTPase Cdc42 binds to the protein kinase SPRK and increases its catalytic activity, (2) that SPRK's CRIB motif is required for the Cdc42-.induced activation of SPRK, (3) that the CRIB motif is necessary, but not sufficient to mediate Cdc42-induced SPRK activation, and (4) that Cdc42-induced activation of SPRK changes SPRK's in viva phosphorylation. To further address the mechanism by which Cdc42 activates the protein kinase SPRK, the effects of Cdc42 on SPRK's subcellular distribution was examined, testing the hypotheses (1) that Cdc42 changes the subcellular distribution of SPRK and (2) that plasma membrane targeting of SPRK is required for SPRK activation by Cdc42. This research project is described in Chapter IV. The final summary and the conclusion of this work are summarized in Chapter V. Understanding the structural requirements and molecular mechanism of SPRK regulation by Cdc42 may provide insight into the regulation and function of these two proteins. This knowledge may contribute to a better understanding of the INK signaling cascade. Furthermore, these studies may shed light on how small GTPases regulate the activity of other protein kinases. 11. Literature Review 1. Regulation of Protein Kinases Protein kinases catalyze the transfer of the y-phosphate of ATP to their protein substrates. This important physiological process is rendered reversible by. the action of protein phosphatases. The first protein kinase discovered was glycogen phosphorylase kinase in 1955, which activates glycogen phosphorylase (Fischer and Krebs, 1955; Sutherland and Wosilait, 1955). At this point, it was believed that phosphorylation of proteins was restricted to the amino acid residue serine and that phosphorylation was characteristic for proteins involved in glycogen metabolism This point of view was changed in 1968 with the discovery of the serine/threonine kinase cyclic AMP-dependent kinase (PKA) (Walsh et al., 1968). In 1980 v-Src, the gene product encoded by the Rous sarcoma virus src-gene was demonstrated to be a tyrosine kinase (Hunter and Sefton, 1980). Phosphorylation of serine and threonine residues by so called serine/threonine kinases and tyrosine phosphorylation by tyrosine kinases are the most common modifications, although histidine kinases, which phosphorylate histidines, have been identified in prokaryotes, fungi, molds, and plants (Grebe and Stock, 1999; Pea and Saier, 1997). Additionally, there are so- called dual specific kinases, which are able to phosphorylate both serine and threonine, as well as tyrosine residues. Protein kinases represent one of the largest protein superfamilies and it is estimated that the human genome may encode over ww- III} .rterim patch it." | mitt 10:21 i of '12," | iii. sowed casiyit‘ ' cam minim r. m? prank input 3"“; 3R ftg‘l‘aft‘d. Prater. times are Ewifi‘ml aCiillIit‘~' cm} 'hfic ptocem J Ward and. com.“ 1' - 011ml of ‘3’» hilly ‘ ' , m ”Mums: 2000 different protein kinases (Hunter, 1987b). In 1998, the crystal structures of the catalytic domains of 12 protein kinases have been solved and all share a similar fold, with conserved catalytic residues (Johnson et al., 1998), supporting the idea that their catalytic mechanisms are similar. Understanding the regulation of a protein kinase may provide important cues about the mechanism by which protein kinases, in general, are regulated. Protein kinases are involved in a variety of cellular processes ranging from transcriptional activation, to muscle contraction, to long-term memory. In order to control these processes properly it is clear that kinases themselves must be highly regulated and controlled. The regulation of protein kinases is achieved by a number of different mechanisms such as binding of an activator or release of an inhibitor, posttranslational modification by phosphorylation or proteolysis, as well as subcellular targeting. 1.1 Regulation of protein kinases by activators and inhibitors A well-characterized mechanism of protein kinase regulation is by allosteric ligand binding. Receptor tyrosine kinases, such as epidermal growth factor receptor, platelet derived growth factor receptor, and the insulin receptor are activated upon extracellular binding of epidermal growth factor, platelet-derived growth factor, and insulin, respectively. Binding of the ligand dimerizes the receptor tyrosine kinase and subsequently induces a conformational change. In response to the conformational change the receptor undergoes autophosphorylation within its ill-— mslih' Jinan Tm imam citrus - llama. 1987'. Rail i Hanger. 1 997: 1. Bede: limits is.“ K'vl’ttlltt sword rm :. . .WMM.MLIEBNT cm hfi'lvtl‘im { Tilt er al., 1W. 1 34‘1“}. 1 “1 If lt-rerqrm. ti; .. . Ru‘hflm dillk‘ra; Mi. ' “I R'S’ut‘unitt (mhkimfinar' Al intracellular domain. The activation of the receptor triggers a variety of downstream phosphorylation events and subsequently mediates changes in gene expression (Carpenter, 1987; Fantl et al., 1993; Schlessinger and Ullrich, 1992; Ullrich and Schlessinger, 1990). Besides hormonal activation of receptor tyrosine kinases, protein kinases can be activated by second messengers, such as the cyclic nucleotides cAMP and cGMP. For instance, in the absence of cAMP, PKA is maintained in an inactive state as a tetrameric holoenzyme consisting of two regulatory and two catalytic subunits (Taylor et al., 1990). Upon binding of four CAMP molecules to the regulatory subunit, the holoenzyme enzyme undergoes a conformational change and dissociates in an R—subunit dimer and two active monomeric catalytic subunits (Taylor et al., 1990). The R-subunits are considered physiological inhibitors of PKA since they keep this kinase in an inactive state (Corbin et al., 1975; Hofmann et al., 1975; Rosen et al., 1975). Dimerization or oligomerization is a further mechanism of regulating protein kinase activity. A well-established example of activation by dirnerization is the activation of receptor tyrosine kinases. Upon binding of the ligand the receptor dirnerizes and undergoes intermolecular autophosphorylation. In addition, many intracellular protein kinases contain leucine zipper-like motifs that may mediate oligomerization. 1.2 Regulation of protein kinases by phosphorylation Many protein kinases are themselves regulated by phosphorylation. Activation of the mitogen—activated protein kinase (MAPK) cascade involves sequential phosphorylation, whereby a MAPK kinase kinase (MAPKKK) phosphorylates a MAPK kinase (MAPKK), which in turn phosphorylates a MAPK For example, the dual-specific kinase MKK4, an activator of cJun NHz-terminal kinase (INK), requires phophorylation by a MAPKKK on Ser 254 and Thr 258 for activation (Yan et al., 1994; Zheng and Guan, 1994). The non-receptor tyrosine kinase Src is regulated by both activating and inhibitory phosphorylation events (Hunter, 1987a). The autophosphorylation of tyrosine 416, which resides within the activation loop of the kinase domain, has a stimulatory effect on kinase activity (Ferracini and Brugge, 1990), whereas phosphorylation of tyrosine 527 by c-Src kinase represses Src kinase activity (Nada et al., 1991). Crystal structures of Src show that the SH2 domain of Src binds to its own phosphorylated tail to exert autoinhibition (Gonfloni et al., 1999; Williams et al., 1997; Xu et al., 1997). 1.3 Regulation by proteolytic cleavage The onset of apoptosis is correlated with the proteolytic cleavage of nmny proteins, including y-p21-activated kinase (PAK) by caspase CPP32 (caspase 3) (Rudel and Bokoch, 1997). The cleavage by CPP32 produces two peptides, one containing the majority of the regulatory domain, and the other one bearing the entire mic ions 1)) «its? risks i111 1011311 s: has: 402 in the . main of {it lime: ism phages '. .. ‘i ' 1 earned t1) x-ra} m lI'. ACZQQ 6! al., lWIS k 1 Wait .‘kids a cane; “m bi €1.42 .11.. ~ imam}- 1W1. In addllion [0 P.\K {ls-I ' . ‘4 It atli‘s'aied in Cw £1903 . catalytic domain (Walter et al., 1998). In contrast cleavage of y-PAK by caspase 3 results in a lO-fold stimulation (Walter et al., 1998). Upon caspase cleavage, threonine 402 in the catalytic domain is autophosphorylated resulting in the stimulation of the kinase activity (Walter et al., 1998). This threonine corresponds to a regulatory phosphorylation site in the activation loop of many protein kinases as determined by x-ray crystallography (Hanks and Quinn, 1991; Jeffrey et al., 1995; Johnson et al., 1996; Knighton et al., 1991). It has been suggested that y-PAK cleavage yields a constitutive active PAK, which induces cell death, whereas the activation by Cdc42 allows a cycling of the kinase between an active and an inactive form (Lee et al., 1997). In addition to PAK, MEKKI, protein kinase N, and protein kinase C (PKC) can also be activated by caspase 3 (Deak et al., 1998; Pongracz et al., 1999; Takahashi et al., 1998). Pas-induced proteolytic cleavage of MEKKl leads to apoptosis (Deak et al., 1998). It is believed that the cleavage renders a soluble kinase complex that subsequently triggers apoptotic events. In contrast, uncleaved, activated MEKKI renders an insoluble kinase complex, which may activate NF-xB and trigger a survival response. This is an example of how the differential regulation of a kinase may have opposite physiological outcomes. 1.4 Regulation by subcellular localization Recently the regulation of protein kinases by subcellular targeting has received a lot of attention. Compartmentalization may restrict a kinase to the vicinity of its effector and result in the activation of the kinase or enhance its activity by increasing accessibility to the substrate. Specific subcellular targeting may allow the differential regulation of a kinase in one cell, enabling one enzyme to regulate various physiological processes. The serine/threonine kinase Raf, for example, requires membrane targeting for full activation. In viva this takes place by binding to the GTP-bound form of Ras, which recruits Raf to the plasma membrane. Appending a membrane-targeting signal triggers Rafactivation in absence of Ras (Leevers et al., 1994; Stokoe et al., 1994), suggesting that not Res-binding, but rather membrane-targeting is the key activating step. PKC is a well-studied example of a kinase whose activity depends upon proper subcellular localization. The PKC protein kinase family consists of at least 11 family members (Newton, 1995). Most cells express more than one isoform, and all of these isoforms have a very similar ligand-binding and substrate specificity. Therefore proper subcellular localization is important to achieve specificity. PKCa, for example is predominately found at the endoplasmic reticulum and at the cell margin, PKCBII is associated with structures of the actin cytoskeleton, PKCy is mainly targeted to the Golgi apparatus, whereas PKCe is associated with nuclear membranes (Goodnight et al., 1995). Multiple isozymes of PKA are expressed in mammalian cells and many hormones use parallel pathways to activate PKA. Therefore a mechanism must exist to ensure that the correct PKA isoform is activated at the right place at the right time P3581111. 39351 A: an it: so £43643 4" is 1 PM is fom' ;~ gang Emit» 03 SEC-- ,1. - .-.dc: submit area‘s tr 3:55.311 :0 carom" 2.3mm mesh? 1‘“: ° I :8. WU er a}, 19‘" 13493:. Interim to and)” ELL-kw": ‘ 1' > Mai halt/aim ‘tcalAaFl, I \ 31E ‘Iqlizn Y " 7V1“ y é k‘)llh’ &"S;l“[l~t;f\ I .. ~ §«‘\al cficcts .1 Mr . 1‘ ‘ 4- a.» k xanuld pints" Set? SCa‘I‘t' “LII protein \ £21.01 ' We Simian-m am 3‘ "d SPVdguc. Sifiii (Harper et al., 1985). Appropriate subcellular localization is ensured by association of PKA with so called, A-kinase anchoring proteins (AKAPs). For example, 75% of type H PKA is found in association with AKAPs (Rubin, 1994). AKAPs are a growing family of signaling molecules that target PKA to specific locations and enhance substrate accessibility and specificity. Various AKAPs have been shown to be targeted to centrosomes, endoplasmic reticulum, Golgi apparatus, microtubules, mitochondria, membranes, nuclear matrix, and secretory granules (Coghlan et al., 1994; Keryer et al., 1993; Lin et al., 1995b; McCartney et al., 1995; Ndubuka et al., 1993). In contrast to anchoring proteins which localize signaling molecules to the proper subcellular localization, scaffold proteins bind an array of signaling molecules to create a signaling complex. In yeast the same MAPKKK is used to elicit different physiological effects depending upon the scaffold protein it is bound to. Two MAPK scaffold proteins have been identified in Saccharamyces cervisiae. The Ste5p scaffold protein coordinates the module, which induces mating in response to pheromone stimulation (Choi et al., 1994; Kranz et al., 1994; Marcus et al., 1994; Printen and Sprague, 1994), whereas the Pszp scaffold protein arranges the signaling proteins, which induce glycerol synthesis in response to high osmolarity (Posas and Saito, 1997). In both cases the first activated kinase is Stel 1p, but when bound to different scaffolds it activates different downstream targets, and in turn elicits different physiological outcomes (Fig. 1). In mammalian cells such scaffold proteins have also been identified. INK-interacting protein (JIP)1 and JIP2 arrange 10 miles of‘h: Ni p; III-Pam (\lpil if .59. '3, .‘ ' I I") " 315....“ 315.41'rCILM am: that 1h: dc. austngofllikkl ‘ 1311: NM 3 Sic-rt“ A Di. fldl,I Iw“I‘ \ modules of the JNK pathway (Whitmarsh et al., 1998; Yasuda et al., 1999). The MEK-Partner (MP)1 scaffold complex coordinates the kinase MEK] and extracellular signal-regulated kinase (ERK)1 (Schaeffer et al., 1998). There is also evidence that the dual-specific kinase MKK4 organizes a signaling module consisting of MEKKl, MKK4, and INK (Xia et al., 1998), and that ch-interacting kinase (NIK), a Ste-related kinase, forms a complex with MEKKI, MKK4/7, and INK (Su et al., 1997), similar to a Pszp-like complex (Fig.2). ll Pherom Stesr ,tP. lib “APR-K \hl Pheromone High Osmolarity l Mating Response Glycerol Synthesis Fig. 1. MAPK scaffold complexes in Saccharonryces cervisiae. The mating response is coordinated by the scaffold protein Ste5p, which binds the MAPKKK Stel 1p, the MAPKK Ste7p, and the MAPK Fus3p. Increased glycerol synthesis in response to increased osmolarity is controlled by the scaffold protein Pbs2p, which acts as a scaffold protein and a MAPKK. 12 MEKK1 scaffold complex MP1 scaffold complex Fig. 2. Mammalian scaffold complexes. The scaffold proteins HP] and JIP2 coordinate components of the JNK signaling cascade in a SteSp-like complex. MEKKI arranges a Pbs2p-like scaffold complex. The MP1 scaffold protein coordinates kinases of the ERK signaling cascade. 2. Small GTPase“ Sail GII’RS hit: its. $36113 51:": &5773}-1W~l. REV 01 r: as uriccsln [hf GTP-512:“ . . 81.13: 3:4 f. .. i o. it GDP up. 1'51 0‘- mu; " t“ "is it in - Cum atria-.1»: $.37} -- . GTPases exert a "A. 43’. «Y GIT) h:‘d."0i‘1 \; \lf a Unmet 1937, 2. Small GTPases of the Rho-family Small GTPases have been implicated in a variety of cellular processes, ranging fiom intracellular signaling to synaptic transmission (Van Aelst and D'Souza- Schorey, 1997). These small proteins, with molecular weights between 20 and 30 kDa, act as molecular switches, cycling between an inactive GDP-bound and an active GTP-bound state (Fig. 3). The activity of small GTPases is determined by the ratio of the GDP/GTP—bound state. This ratio is regulated by so-called guanine nucleotide exchange factors (GEFs), which enhance the exchange of GDP to GTP, and GTPase activating proteins (GAPS) (Boguski and McCormick, 1993). Since small GTPases exert a very low intrinsic GTPase activity GAPS are required to increase GTP hydrolysis and convert the GTPase into its inactive state (T rahey and McCormick, 1987). The Ras superfamily of small GTPases is divided into four different subfamilies (Macara et al., 1996). The best-characterized family is the Ras family, which consists of H-Ras, K-Ras, and N-Ras. The Ras family gained a lot of attention in the early 1980s for its transforming activity. Indeed, in 10-20% of all human cancers mutations in the ras-gene are found (Barbacid, 1987; B05, 1988). The ARF and Rab families play important roles in the assembly and targeting of vesicles to the appropriate cellular compartment. They are essential for the recruitment of coatamer proteins and are important for proper docking and fusion of vesicles (Macara et al., 1996). 14 The Ran family of small GTPases is associated with the nuclear import machinery and is essential for nuclear import processes (Macara etal., 1996). The Rho-family of small GTPases was first identified in yeast in the context of cytoskeletal rearrangements. The mammalian Rho-family consists of at least 10 members, including Rho A, B, C, D, and E, Rac 1, 2, and E, and Cdc42 and TClO (Van Aelst and D'Souza-Schorey, 1997). The members of the Rho-GTPase family are key regulators linking surface receptors to the organization of the actin cytoskeleton. Rho is implicated in the assembly of stress fibers and focal adhesion (Ridley and Hall, 1992). Rac induces a meshwork of actin filaments at the cell periphery to produce lamellipodia and membrane ruffles (Nobes er al., 1995; Ridley et al., 1992). Cdc42 finally triggers actin rich surface protrusions, so called filopodia (Kozma et al., 1995; Nobes et al., 1995). In Swiss 3T3 cells cross talk between the Rho family members has been demonstrated (Chant and Stowers, 1995; Nobes and Hall, 1995). Besides their effects on the morphological changes of the cytoskeleton (Hall, 1998) Rho family members also have been implicated in apoptosis (Jimenez et al., 1995), cell cycle progression (Olson et al., 1995), cellular transformation (Khosravi- Far et al., 1995; Qiu et al., 1997; Qiu er al., 1995a; Qiu et al., 1995b; Tang et al., 1999; Whitehead et al., 1998; Wu et al., 1998), and the regulation of MAPK signaling pathways (Coso et al., 1995; Minden et al., 1995). Cdc42 has been first identified in S. cervisiae as an important component in the organization of the actin cytoskeleton during the cell cycle (Adams et al., 1990). In 15 {h was. species S g '11: till Johnson mil Cit-'1 also him is 1933111 hum: 5... Sims 3 iii: degree 0. CICJISllST‘QMi ’ Neill Rahal“. but i 5.. . | ‘57}:th lnLlU 4 [flux l “CL—Yin both yeast species, S. pambe and S. cervisiae, Cdc42 is essential for the viability of the cell (Johnson and Pringle, 1990; Miller and Johnson, 1994). The mammalian Cdc42, also known as 625K, was first purified from bovine brain (Waldo et al., 1987) and human placental membranes (Evans et al., 1986; Polakis et al., 1989) and shows a high degree of similarity to yeast Cdc42 (Johnson and Pringle, 1990; Polakis et al., 1989). Studies of embryonic stem (ES) cells in which the gene encoding Cdc42 is disrupted, show that in mammalian cells the small GTPase is not required for cell viability, but confirmed its role in formation of the actin cytoskeleton, since Cdc42-deficient cells show an abnormal cytokeletal arrangement (Chen et al., 2000). In mammals Cdc42 appears to play a pivotal role in embryonic development. Cdc42-deficient mice die very early during embryonic development. Histological analysis of in utera embryos shows that the embryos are totally disorganized and lack the primary ectoderm (Chen et al., 2000), supporting Cdc42's role in cytoskeletal organization. Cdc42 contains a so-called CAAX motif at the COOH-terminus, which is required for geranylgeranylation of the small GTPase (Maltese and Sheridan, 1990). This posttranslational modification allows the GTPase to associate with cellular membranes, including the plasma membrane (Hart et al., 1990) and the Golgi complex (Erickson et al., 1996). Interestingly, in yeast 3 Cdc42 variant which cannot undergo prenylation, due to a change of the cysteine to a serine within the CAAX motif, rescues the lethal phenotype induced by constitutively active Cdc42 (Ziman et al., 1991). Studies in yeast also show that Cdc42 is distributed l6 1 l .41 [I ' " a; ' . )‘.Qy.- 1 yea I m.s.stl.h:lt" Us... -4- C x naked cell grim: 71 I “ ‘\,} :' ‘ ~30 a “ We; ‘4‘“. C&’ n... - et~' . .’r.1r.‘t‘: studies St. ' ' Am §x 5.1m'\vm prov . I. »CJ‘. 1 v E {07171. 53.1 d'JC [Si-310 e: di. EV‘ scrim fetuses I his}; I ~L"~~D-§'CRIB'] 3‘1"" hm ‘ “5 mm The Sim. CL 9‘ ~ I'm: been CV;- differentially during the cell cycle and that proper localization is important for polarized cell growth (Ziman et al., 1993). There is also evidence in mammalian cells that Cdc42 may be required for the subcellular distribution of effector proteins. In viva studies suggest that Cdc42 localizes the structural protein Wiskott Aldrich Syndrome protein (WASP) to the plasma membrane, where a multiprotein complex is formed, subsequently inducing actin polymerization and filopodium formation (Castellano et al., 1999). A very well studied downstream effector of Cdc42 is the serine/threonine kinase PAK, which is activated by Rho-family GTPases (Manser et al., 1994). The interaction between Cdc42 and PAK requires the so-called Cdc42/Rae interactive binding (CRIB) motif (Burbelo et al., 1995), a short, and 14-16 amino acid residue long motif. The structural requirements and the activation mechanism of PAK by Cdc42 have been extensively studied (Gatti et al., 1999; Knaus et al., 1998; Manser et al., 1997; Manser et al., 1994; Zenke et al., 1999; Zhao et al., 1998). Activation of PAK by Cdc42 can be demonstrated in vitra using recombinant purified proteins (Martin et al., 1995). This observation supports a stoichiometric model in which Cdc42 directly increases PAK autophosphorylation activity, without the requirement for additional effectors. In several studies it has been reported that membrane localization of PAK may contribute to PAK activation (Daniels et al., 1998; Lu et al., 1997a; Lu and Mayer, 1999). However, there is no direct evidence that the interaction of Cdc42 with PAK is required for membrane translocation. In contrast a variant of Ste20p, the PAK yeast homologue, which fails to bind Cdc42, shows 17 morasmlc llkd...’.;. fa." 91.1.71‘ I--9,:‘ L's talksLLsé 0 -,.5- Besides the Pkk "Eager ex 123-. l'w' faxed Cat-4:11;». \EKKI (Finger 6! t it CRIB notif in a ‘s a CRIB- 3318111in rem .;. Kai. Cd“: has 1'»: V” l: 51'” Comma, (”Strain . .. Cl} 3kil\c oi» 7d 5': cytoplasmic localization (Peter et al., 1996), suggesting that Cdc42 may be important for targeting of Ste20p to emerging buds. Besides the PAKs, Cdc42 interacts with various other kinases, including MEKKs (Fanger et al., 1997; Gerwins et al., 1997) or the non-receptor tyrosine kinase activated Cdc42HS-associated kinase (ACK) (Manser et al., 1993). Interestingly, MEKKl (Fanger et al., 1997) and the ribosomal S6 kinase (Chou and Blenis, 1996) bind to Cdc42 despite the fact that they lack a CRIB motif. In addition, deletion of the CRIB motif in MEKK4 only partially diminishes binding of Cdc42 and Rac, suggesting a CRIB-independent GTPase binding determinant. Thus, the role of CRIB motifs and the mechanism by which small GTPases promote the activation of protein kinase remains largely unexplained. Cdc42 has been implicated in cell cycle progression, but its effect on this process is still controversial. Olson and coworkers reported that microinjection of constitutively active Cdc42 into Swiss-3 T3 cells increases bromodeoxyuridine incorporation into DNA, whereas the dominant negative Cdc42 blocks the incorporation (Olson et al., 1995), suggesting that Cdc42 may contribute to cell cycle progression. In contrast, microinjection of wild type Cdc42 into synchronized NIH 3T3 cells caused cell cycle arrest, which was mediated by the p3 8/reactivating kinase (p38/RK) pathway (Molnar et al., 1997). Besides its effects on the cell cycle, activated variants of Cdc42 have transforming activity. There is evidence that Cdc42 is required for Ras-mediated transformation and that stable expression of constitutively active Cdc42 in Rat] fibroblasts increases anchorage-independent 18 grown in soft as 1 notes 111.7111 :1 '1'? u,- Ctls‘w': «and Rat! mi regime a vane.) 02 its»? L'apstiv-r'o“ . s . u... i" oxisie'leui reams; A WT} lineman: Cit.” ' . m “statue M 1' “Ik‘azhhu \» - \. r>n 101.", growth in soft agar (Qiu et al., 1997). Injection of activated Cdc42 into nude mice promotes tumor formation (Qiu et al., 1997). Taken together these data suggest that Cdc42 (and Rac) indeed induce cellular transformation. Although Cdc42 appears to regulate a variety of different cellular processes, including cell cycle progression, cellular transformation, and invasiveness, these processes are all dependent on cytoskeletal rearrangements. A very fascinating field, which has recently drawn more attention, is the role of Cdc42 in negative factor (NeD-dependent human immune deficiency virus (HIV) replication. Nef of human and simian immune deficiency virus (HIV -1, HIV -2 and SVI) is a myristylated plasma membrane-associated protein (Allan et al., 1987; Yu and Felsted, 1992). Nef plays an important role in viral pathogenesis and is responsible for high levels of viremia and for the progression of an HIV infection towards acquired immune deficiency syndrome (AIDS) (Kestler et al., 1991). Nef coimmunoprecipitates with the mammalian PAK-like serine/threonine kinase Nef- associated kinase (NAK) (Nunn and Marsh, 1996; Sawai et al., 1994; Sawai et al., 1995). It has been demonstrated that NAK kinase activity is increased upon cotransfection with active Cdc42 and that dominant-negative Cdc42 blocks binding of Nef to NAK and subsequent activation of the kinase. Moreover dominant- negative Cdc42 and Rac decrease HIV-1 production (Lu et al., 1996). Thus these data suggest a role for Cdc42 in the progression of an HIV infection towards AIDS. Therefore understanding the regulation of Cdc42 and its effector proteins may shed l9 further light on the progression of HIV infections and lead to the identification of potential therapeutically useful targets. Since Cdc42 regulates so many different proteins and thus, multiple physiological processes, it is of great interest to learn more about the effector proteins of this small GTPase. Understanding the Cdc42—mediated regulation of proteins may provide important knowledge of how this small GTPase can exert so many different effects and help to decipher the specific roles of the effector proteins. 20 cap, inacflve {are GEFs acfive Fig. 3. Regulation of small GTPases. Small GTPase cycle between a GDP-bound inactive, and a GTP-bound active form. Guanine nucleotide exchange factors promote the exchange of GDP to GTP. GTPase-activating proteins (GAPS) enhance the hydrolysis of GTP to GDP. 21 3. The 3UP Ki For prOPG' ,a '1 out. I\ .e'). e bt'ra:11a:)~azc “ at.- k strontium} st?“- .eaA £721.11? trunnion is ”£333 34'? I‘Lfik Ci fl“‘H' -~ A “it“ (11 fUI tile-“1:9 g1.“- ‘ ~ 3 . Uta-ll IFLFEXUCiHJII 3. The MAP Kinase Pathways For proper development and survival, eukaryotic cells must be able to communicate with their surroundings and to respond to various extracellular and environmental stimuli. Exposure to external stimuli induces a variety of cellular responses, such as proliferation, cell division, differentiation, or apoptosis. One crucial question is how these external signals, which are sensed by cell surface receptors, are processed within the cell to induce the changes in gene expression that account for changes in cell phenotype and cell morphology. Some of the major signal transduction pathways in eukaryotic cells are the protein kinase cascades, which lead to the activation of so called mitogen-activated kinases (Cano and Mahadevan, 1995; Marshall, 1994; Waskiewicz and Cooper, 1995). Although different MAP kinase pathways are activated by different external stimuli they all follow a common conserved module of activation (Fig. 4). The MAPK is activated by phosphorylation on threonine and tyrosine residues within a TXY motif, by a dual-specific MAPKK The MAPKKS are a highly conserved group of protein kinases, which are activated by phosphorylation of serine and threonine residues by MAPKKKS. MAPKKKS receive their signal from cell surface receptor through various proteins, including small GTPases, protein kinases, and/or protein phosphatases. In the different MAPK pathways the MAPKS, MAPKKS and MAPKKKS are highly conserved within their catalytic domain, but can differ significantly in their regulatory domains and substrate specificity. 22 .. 4. MAPK pa ‘v‘se palhua) i ph‘irphonlaiion. Fig. 4. MAPK pathway module. The core module of the mitogen-activated protein kinase pathway is composed of three kinases that are sequentially activated by phosphorylation. 23 artisans of IN? 15431;; retires}. ilkrijxd er a.'.. '_ MAPK the p38 RI- sirplified 01cm: The INK path omit”); I v;-- Raves .1- cag-i - ~ We 15 actlxatt at. 1W4) a:‘d in. (33411 e1 (11. ice-- on- 1“ snail (i 11:: I-lCated in RX 1~ =.- 499‘ H11 UI I 9! a! ‘ C0 i. - mniuthelx ‘1'. ' i‘hn)’ \ The first identified mammalian Signal transduction pathway was the ERK cascade. Later a second MAP kinase pathway was discovered which induces the activation of IN K. This pathway is also called stress-activated protein kinase (SAPK) pathway, since its activation has been linked to various cellular stresses (Derijard et al., 1994; Hibi et al., 1993; Kyriakis et al., 1994). Recently a third MAPK the p38/RK pathway was identified (Han et al., 1994; Rouse et al., 1994). A simplified overview of all three mammalian MAPK pathways is shown in Fig. 5. The INK pathway is mainly known for its activation by cellular stresses, such as osmotic stresses and UV-radiation (Derijard et al., 1994). Additionally the JNK cascade is activated by cytokines such as tumor necrosis factor-a (TNF-ct) (Sluss et al., 1994) and interleukin] (1L1) (Chaudhary and Avioli, 1998; Finch et al., 1997; Guan et al., 1996). Furthermore, evidence exists that the JNK pathway can be activated by growth factors (Bast et al., 1997). Whereas the ERK pathway is activated by members of the Ras family of small GTPases, small GTPases of the Rho-family, especially Racl, Rac2, and Cdc42 are implicated in INK activation (Bagrodia et al., 1995a; Brown et al., 1996; C050 et al., 1995; Hill et al., 1995; Minden et al., 1995; Zhang et al., 1995). Overexpression of constitutively active Cdc42 in COS-7 cells, for example, increases in vitra INK activity 5-10 fold, but Ins only little effect on ERK activity (Coso et al., 1995; Teramoto et al., 1996). Similar results were observed in COS-1 (Bagrodia et al., 1995a), HeLa, NIH-3T3, Ratl (Minden et al., 1995) and 293 cells (Teramoto et al., 1996). Recent studies Showed that expression of dominant negative variants of 24 €ch; Rx. and '1 INT-Q. or hyper 6.1.4:- - ES Cells \ '5 not Stroked it 05ka mix 3:10: {that-[1n ma), 6‘ MLKKS are ‘I ihi’i e1 CL. 1 Hi. mom ' ‘ {.4 M‘ ‘R\T" ' .VT" '~ 1:le hm: hm Lh‘ée‘caner e! A Cdct‘ ‘~ Nor R; i‘lhrt' as“? n b} C0 h 01 I - (if~ . 1.0““ er [no rea— be aUIOHL Cdc42, Rae, and Ras block IN K1 activation, induced by hepatocyte growth factor, TNF-a, or hypcrosmotic glucose (Auer et al., 1998). Interestingly, analysis of cdc42-/- ES cells Showed that JNK and p38/RK activity induced by cellular stresses is not impaired (Chen et al., 2000), suggesting that a Cdc42~independent mechanism of JNK activation or that redundancy among Rho family members in terms of JNK activation may exists. MEKKS are MEKKKS that have been Shown to activate the ERK pathway (Blank et al., 1996; Lange—Carter et al., 1993), but preferentially activate the JNK pathway (Kyriakis et al., 1994; Minden et al., 1994; Yan et al., 1994). Four different MEKKS have been cloned, MEKKl-4 (Blank et al., 1996; Gerwins et al., 1997; Lange-Carter et al., 1993) among which MEKKl and 4 appear to be regulated by Cdc42 and/or Rac.(Fanger et al., 1997). Kinase-inactive MEKK4 blocks INK activation by constitutively activated Cdc42 and Rac (Gerwins et al., 1997), but no direct activation of MEKK4 by these two small GTPases could be demonstrated yet. In addition to Rho-GTPases, MEKKl also binds to the activated form of Ras (Russell et al., 1995). The protein kinase PAK has been shown to activate the JNK pathway (Bagrodia et al., 1995a; Brown et al., 1996). Four different PAK isoforms, PAKl, PAK2, PAK3 and PAK4 have been identified (Abo et al., 1998; Bagrodia et al., 1995b; Brown et al., 1996; Jakobi et al., 1996; Manser et al., 1994) and PAKl-3 increase autophosphorylation in the presence of Cdc42 (Martin et al., 1995). 25 Although PAKs have been implicated in IN K activation, their physiological role in the JNK pathway still remains unclear. Members of the mixed lineage kinase family MLK2, SPRK/MLKB, and dual leucine zipper-bearing kinase (DLK) have been identified as activators of the JNK pathway. (Fan et al., 1996; Hirai et al., 1997; Nagata et al., 1998; Teramoto et al., 1996). MLK2 and SPRK, both associate with the small GTPases Cdc42 and Rac in a GTP-dependent manner (Burbelo et al., 1995; Nagata et al., 1998; Teramoto et al., 1996) and coexpression of kinase-inactive SPRK with activated Cdc42 or Racl blocks Cdc42-induced or Rae-induced IN K activation suggesting that SPRK/MLK3 acts downstream of Cdc42 and Rac (Teramoto et al., 1996). Furthermore it has been demonstrated that hematopoietic protein kinase (HPK)1 associates with SPRK/MLK3 and phosphorylates a kinase-inactive mutant of SPRK/MLK3. Thus HPKI may be a potential activator of SPRK, although activation of SPRK/MLK3 by HPKl has not been demonstrated (Kiefer er al., 1996). SPRK/MLK3 activates the JNK pathway via the dual-specific kinases MKK4 (Rana et al., 1996) and MKK7 (Whitmarsh et al., 1998). SPRK/MLK3 activates MKK4 by phosphorylation of a serine 254 and threonine 258 in the activation loop of the catalytic domain (Yan et al., 1994; Zheng and Guan, 1994). For activation INK has to be phosphorylated on threonine 183 and tyrosine 185 within the TXY-motif of the catalytic domain (Derijard et al., 1995). Two dual- specific kinases, MKK4 (Derijard et al., 1995; Lin et al., 1995a) and MKK7 (Moriguchi et al., 1997; Tournier et al., 1997; Wu et al., 1997; Yao et al., 1997) 26 ”I“ have been shown to phosphorylate INK. MKK7 has been found to associate with the two scaffold proteins HP] and IIP2, which both mediate activation of the INK pathway, presumably by assembling the individual signaling components, including I-IPKl, DLK, MLK2, SPRK, MKK7, and INK into a multi-enzyme complex (Whitmarsh et al., 1995; Yasuda etal., 1999). Upon phosphorylation by a dual-specific kinase INK translocates into the nucleus where it phosphorylates a variety of transcription factors. Originally INK was identified by its ability to phosphorylate the transcription factor c-Iun on serine 63 and serine 73 (Derijard et al., 1994). These activating phosphorylations induce transcriptional activity of c-Iun (Pulverer et al., 1991; Smeal et al., 1992; Smeal et al., 1991). The INK family includes three different gene products, INK] (Derijard et al., 1994), INK2 (Sluss et al., 1994) and INK3, which together due to alternative splicing encode for 10 different INK isoforms (Gupta et al., 1996). INKl and INK2 are ubiquitously expressed (Kallunki et al., 1994), whereas INK3 is restricted to neuronal cells (Gupta et al., 1996). The three INK isoforms differ in their ability to phosphorylate c-Jun. INK2, for example, has a much higher affinity for c-Iun than INKl. Due to the lower Km for c-Iun and higher me INK2 is estimated to be 25 times more efiicient in cJun phosphorylation than INKl (Kallunki et al., 1994). The c-Iun transcription factor is involved in a variety of cellular events, including proliferation, differentiation, and cellular transformation. c-Iun is a member of the AP-l activator, which is composed of leucine zipper-bearing DNA binding proteins including c-Iun, F05, and ATFZ (Sassone-Corsi et al., 1988). INK 27 32:11:; J'L,‘ 01 “K also phosphorylates and activates the transcription factor ATFZ, which is able to dimerize with c-Iun and can induce transcription of c-Iun (Gupta et al., 1995; Livingstone et al., 1995; van Dam et al., 1995). INK also regulates cFos expression, which is mediated by the serum response factor (SRF) and the ternary complex factor (T CF) (Treisman, 1992; Treisman, 1994). One well-characterized TCF is Elk- 1, which is phosphorylated by INK (Cavigelli et al., 1995; Gille et al., 1995; Whitmarsh et al., 1995; Zinck et al., 1995). Interestingly, INK phosphorylates the same activating sites as does ERK (Whitmarsh et al., 1995) suggesting an integration of the ERK and the INK pathways. 28 GTPa MAP! “AF MA GTPases Rae Rae, Cdc42 Rae, Cdc42 PAK / HPK-1 / 7 l? l _. . MAPKKK Raf MEKKt MLK2 SPRK MEKK4 DLK 3:- . , m; MAPKK MEK 1, 2 MKK4 MKK3, MKK7 MKKB MAPK ERK JNK p38IRK Fig. 5. Parallel mammalian MAPK pathways. Three MAPK pathways have been identified in mammalian cells, the ERK, INK, and p3 8/RK pathway. 29 4. The physiological importance of the JNK pathway The INK pathway has been mainly characterized as a MAPK cascade which plays an important role in the induction of apoptosis in response to cellular stresses, including UV and y-irradiation, protein synthesis inhibitors, osmotic stress, toxins, ischemia/reperfusion injury in heart, ceramides, inflammatory cytokines, such as TNF-a, or cytokine deprivation (Cuvillier et al., 1996; Verheij et al., 1996; Xia et al., 1995; Zanke et al., 1996). Most studies showing the apoptotic effect were performed in in vitro cell culture. Analysis of in vivo models finally demonstrated that the INK pathway is not only a stress-activated pathway, but also an important player in the inflammatory and humoral immune response and in developmental processes. Drosophila melanogaster, as a genetic model, and genetically engineered mice rendered much of the knowledge about the physiological function of the INK signaling cascade. Following is a summary of knowledge obtained about the role of INK signaling using Drosophila and knockout mice as a model system. 4.1 JNK signaling in Drosophila melanogaster Many components of the mammalian INK signaling pathway are conserved in mammals and insects (Glise et al., 1995; Holland et al., 1997; Lu et al., 1997b; Moriguchi et al., 1997; Riesgo-Escovar et al., 1996; Sluss et al., 1996; Tournier et al., 1997). The Drosophila INK homologue, also termed DINK or BSK, shares about 80% sequence identity with the mammalian counterparts and contains the INK-specific TPY regulatory motif (Riesgo-Escovar et al., 1996; Sluss et al., 1996). 30 Also a homologue of MKK7, the MAPKK, called DMKK7 or HEP (Holland et al., 1997), and DPAK (Harden et al., 1996) have been cloned. In addition to protein kinases of the INK pathway, the upstream activators, small GTPases of the Rho family, termed D-Racl, D-Cdc42 (Luo et al., 1994) have been identified. Analogous to the nnmmalian system DIN K phosphorylates Djun, the Drosophila homologue of c-Iun, in vitro (Riesgo-Escovar et al., 1996; Sluss et al., 1996) and in viva (Hou et al., 1997; Kockel et al., 1997; Riesgo-Escovar and Hafen, 1997). Studies in Drosophila demonstrated that DINK is important for developmental processes such as tissue polarity, dorsal closure, neural development, as well as the insect immune response. Dorsal closure occurs at mid-embryogenesis in Drosophila. It is a morphogenic process, which requires a shape change of the epidermal cells and the movement of these cells as a sheet, which surrounds the embryo (Martinez-Arias, 1993). Two groups of proteins are required for dorsal closure, structural proteins that control cell shape and signaling molecules that coordinate the morphogenic process. The two signaling pathways, which contribute to dorsal closure, are the INK pathway (Riesgo-Escovar et al., 1996; Sluss et al., 1996) and a transforming growth factor (TGF)-B-like pathway (Glise and Noselli, 1997; Hou et al., 1997; Kockel et al., 1997; Sluss and Davis, 1997). Loss of fimction mutations in hep, bsk and Djun cause severe defects in dorsal closure (Glise et al., 1995; Glise and Noselli, 1997; Hou et al., 1997; Kockel et al., 1997; Riesgo-Escovar and Hafen, 1997; Riesgo- Escovar et al., 1996; Sluss and Davis, 1997; Sluss et al., 1996). In addition loss of 31 firmion m 332'. Pk}; ( 31K‘ '5. 199"}. er. r-y—q filnction mutations of the upstream components of the INK pathway, Racl, Cdc42, and PAK cause defective dorsal closure (Harden et al., 1996). Interestingly, mouse MKK7 is able to rescue hep mutants when expressed in Drosophila (Holland et al., 1997), emphasizing the conservation of this pathway among invertebrates and vertebrates. Taken together theses results strongly suggest an important role for INK during fly development. It also has been proposed that the INK cascade is necessary in the insect immune response. In order to fight microbial infection insects possess signal transduction pathways (1p and Levine, 1994; Sluss et al., 1996). These signaling pathways, which are conserved in fly and mammals, are involved in activation of the transcription factors NF-KB and AP-l . Biochemical studies show that the Drosophila immune response is initiated by bacterial lipopolysaccharides, which induce the activation of the MAPKK HEP (Sluss et al., 1996). 4.2 The JNK Pathway in mammab Although the investigation of INK activity in Drosophila rendered much information about the developmental role of this pathway in the fruit fly the generation of genetically engineered mice exerts great impact on the importance of INK signaling in mammals. Homozygous mice with a targeted disruption of the gene for the dual specific kinase MKK4, also called SEKl, are embryonic lethal between embryonic day (E)10.5 and E125 (Nishina et al., 1999; Yang et al., 1997a). The heterozygotes 32 show no phenotype compared with wild type mice (Ganiatsas etal., 1998; Nishina et al., 1999; Yang et al., 1997a). ES cells, which lack the MKK4 gene, are no longer able to activate the INK pathway in response to environmental stresses, such as anisomycin treatment or heat shock demonstrating that MKK4 activates INK in viva. Results from biochemical studies suggest that MKK4 activates both, INK and p38/RK (Derijard et al., 1995; Lin et al., 1995a). Analysis of the ES cells with a loss of fimction for MKK4, showed that only the activation of the INK pathway is impaired and not activation of the p38/RK pathway, suggesting that MKK4 is a specific INK activator in viva (Ganiatsas et al., 1998; Yang et al., 1997a). Analysis of the phenotype of mkk4-l- mice showed that these animals have severe defects in liver formation and hepatocyte development (Ganiatsas et al., 1998; Nishina et al., 1999). The embryos still form the primordial liver anlage but the hepatocytes undergo massive apoptosis and disappear, suggesting a role for MKK4 as an anti- apoptotic survival signal during liver organogenesis (Ganiatsas et al., 1998; Nishina er al., 1999). Interestingly mice lacking the transcription factor c-Iun also display a similar defect in liver development and die between E13.5 and E14.5 (Hilberg et al., 1993; Johnson et al., 1993). Taken together these results suggest that liver development is dependent on c-Iun activation by the MKK4-INK signaling cascade. Since the disruption of the MKK4 gene is embryonic lethal, it is not possible to eXamine the physiological role of MKK4 in adult knockout animals. However, a Complementation assay using ragZ-I- blastocytes rendered chimeric animals with Mkk4-l- T and B cells (Nishina et al., 1997). In B cells the MKK4-INK signaling 33 l t . C(‘A‘Ctdt 1) i171"; R“ f. me: my. Atl. 5 “‘93: fig“?“_ ‘ “L - xi) ' ‘ ‘ h. 4n .. 3.. K‘“-\T>’: cascade is activated in response to CD40 crosslinking (Berberich et al., 1996; Sakata et al., 1995). INK activation also regulates 1L2 production in T cells (Su et al., 1994). Animals with MKK4 null B cells show a partial block in B cell maturation, but peripheral B cells show no defect demonstrating tlmt MKK4 may be important in early B cell differentiation, but that it is not necessary for proliferation and Ig secretion during the humoral immune response (N ishina et al., 1997). The chimeric mice have smaller thymi, but normal numbers of T cells. This suggests that MKK4 is important for IL2-dependent T cell proliferation. Further analysis of these chimeric mice showed that mkk4-l- mice failed to induce the expression of the death suppressor Bel-XL in response to antigen receptor activation, suggesting that MKK4 transduces cellular survival signals during T cell stimulation (N ishina et al., 1998). In contrast to MKK4 deficient mice, knockouts of the various INK isoforms are viable (Dong et al., 1998; Yang et al., 1998; Yang et al., 1997b). The INKl and INK2 isoforms are ubiquitously expressed in the body, whereas INK3 is mainly restricted to the brain, with a lower expression in heart and testis (Gupta et al., 1996). Analysis of jnkI-l- and jnkZ-l— mice showed that they have defects in T cell differentiation (Dong et al., 1998). Two types of T lymphocytes cytotoxic T cells and helper T cells (T it) exist. The TH cells stimulate the immune response of other cells such as macrophages and B cells by secreting lymphokines, interleukins, and cytokines. TH cells differentiate in two subclasses, TH] and TH2 cells, depending upon the difl‘erentiation signal. Mice deficient for INKl produce a high level of THZ cytokines and therefore differentiation into TH2 cells was enhanced (Dong et al., 34 I - .11 {on diff-'31 .I'tc C {.71 p. 1998) resulting in increased IL4 secretion. In jnk2-/- mice the differentiation into TH] cells is also impaired, but this is due to a decrease in interferon (INF )y secretion, which is required for differentiation into TH] cells (Sabapathy et al., 1999; Yang et al., 1998). These results show that both INK] and INK2 are required for the proper differentiation of T51 cells, but that they exert a different mechanism of regulation. The My secretion, which requires INK, directly controls TH] proliferation and also enhances the activation of cytotoxic T cells, whereas INK] controls IL4 secretion to drive cells into TH] differentiation away from TH2 differentiation. The compound knockout of INK] and INK2 is embryonic lethal between E1] and E12, whereas compound knockouts of INK1/INK3 and INK2/INK3 are viable and show no phenotype (Kuan et al., 1999). Mice deficient for INK] and INK2 have severe a dysregulation of apoptosis in the brain. Interestingly they show a reduction of apoptosis in the lateral edges of the hindbrain prior to neural tube closure and an increase of apoptosis in the forebrain with precarious degeneration, suggesting that INK] and IN K2 have apoptotic as well as anti-apoptotic effects. Furthermore this study shows that INK] and INK2 are absolutely required for proper neural development but that their effects are redundant, since the single knockouts show no neuro-developmental impairment. Mice lacking the INK3 gene showed no abnormalities in the structural organization and cellular composition of the brain, but they show a different phenotype to wild type litterrnates when injected with kainic acid (Yang et al., 1997b). Kainic acid mimics the effect of excitatory amino acids, which cause acute 35 membrane depolarization, seizures, and latent cellular toxicity. The cellular toxicity induces apoptosis and plays an important role in the onset of neurodegenerative diseases (Lipton and Rosenberg, 1994; Rothman and Olney, 1986). Kainic acid causes seizures by direct stimulation of the glutamate receptor and indirectly by increasing the release of excitatory amino acids firom nerve terminals (Ben-Ari, 1985; Ferkany et al., 1984). When treated with an epileptogenic dose of kainic acid (30mg/kg body weight) wild type mice and heterozygous mice showed seizures of progressive severity. The homozygous jnk-/- mice in contrast had much milder symptoms and recovered faster after administration of kainic acid. The neural damage caused by kainic acid is predominant in the hippocampus (Ben-Ari, 1985). Analysis of hippocampi of systemically treated mice showed no signs of cell destruction in the homozygous knockout animals, when compared to wild type or heterozygous littermates. In knockout mice the phosphorylation of c-Iun and the transcriptional activity of AP-l is decreased, suggesting that the protection against kainic acid-induced symptoms is due to the blocking of the INK3 signaling cascade. This effect is not seen in the INK] and INK2 knockout animals suggesting a very Specific role for INK3 in mediating glutamate receptor signaling. Thus, INK3 may be a potential target for the treatment of neurodegenerative diseases. 36 5. The Mixed-Lineage Kinase Family The mixed-lineage kinase family of serine/threonine kinases consists of five family members, MLK], MLK2, SPRK/MLK3, DLK, and leucine zipper-bearing kinase (LZK). The three MLKs are very closely related, whereas DLK and LZK compromise a more distant subfamily of the mixed-lineage kinases. This protein kinase family is characterized by a kinase domain, which resembles in sequence both tyrosine kinases and serine/threonine kinases but only serine/threonine kinase activity has been demonstrated. The mixed-lineage kinases contain several potential protein-protein interaction domains, including a leucine/isoleucine zipper and a COOH-terminus, which is rich in serine, threonine, and proline. The bana fide MLKs contain an NHz-terminal SH3 domain, whereas DLK and LZK do not. These are the only reported serine/threonine kinases that contain an SH3 domain in the absence of an SH2 domain. All of the MLKs were identified by a PCR approach using degenerate oligonucleotides corresponding to conserved regions in the catalytic domains of protein kinases. The full-length clone of MLK1 has not been reported yet. MLK] mRNA has been detected in epithelial cell lines of breast, colonic and esophageal origin (Dorow et al., 1993) and shows differential mRN A expression in pancreatic B- cell lines at different stages of maturation and embryonic pancreas development (DeAizpurua et al., 1997). The protein kinase is primarily expressed in pancreatic duct-like structures during mouse embryogenesis, whereas it is not detected in late 37 gestation or early adulthood suggesting a role of MLK1 in the early inductive phase of pancreatic development. According to Northern blot analysis MLK2 is rminly expressed in brain and skeletal muscle, with a lower level of expression in the pancreas (Dorow et al., 1993). Additionally, several tumor cell lines show high levels of MLK2 expression (Katoh et al., 1995). Overexpression of MLK2 in COS-1 cells increases INK activity 6 to 10-fold. Only modest induction of p38/RK (LS-fold) and ERK (2 to 3-fold) (Hirai et al., 1997; Nagata et al., 1998) was detected. INK activation is presumably mediated by MKK4 and MKK7, since these two dual-specific kinases are activated upon overexpression of MLK2 in COS-1 and human epithelial KB cells (Cuenda and Dorow, 1998; Hirai et al., 1997; Hirai et al., 1998). Interestingly, it appears that MKK7 is the preferred substrate since MLK2 overexpression in these cell types resulted in a higher MKK7 than MKK4 activity (Cuenda and Dorow, 1998; Hirai et al., 1998). The same result is obtained in in vitra kinase assays using a truncated, recombinant MLK2 and purified GST-MKK4 or GST-MKK7 as a substrate (Hirai et al., 1998). Evidence exists that transiently expressed MLK2 is able to activate MKK3 and MKK6, the upstream activators of p38/RK (Cuenda and Dorow, 1998), but the physiological importance of this activation remains unclear, since overexpression of MLK2 does not elicit a prominent increase in p38/RK activity (Cuenda and Dorow, 1998). 38 It Examim: pm‘maze d; pTISTI‘A‘T) 1c: precipitant KIRK. RIF L'arswn ca: .2995! “OCT: ‘he vesicle 3r Addmmgh 0]?“ dm microtubuics Nemesis. 1 Examination of the subcellular distribution of MLK2 in Swiss 3T3 showed a punctuate distribution of microinjected MLK2 along microtubules, together with phosphorylated INK (Nagata et al., 1998). Two-hybrid analysis and coimmuno- precipitation experiments demonstrated tlmt MLK2 associates with KAP3 and KIF3X. KIF3 is a member of the kinesin superfamily of motor proteins, which transport cargo vesicles along microtubules towards the plus end (Brady and Sperry, 1995), whereas KAP3 is a cargo target molecule that serves as an adaptor between the vesicle and the cargo molecule (Yamazaki et al., 1996). It appears that MLK2 can associate with both the motor protein itself and the cargo target molecule. This may suggest that MLK2 may be involved in vesicular transport processes. Additionally it has been reported that the SH3 domain of MLK2 can bind the GTPase dynamin (Rasmussen et al., 1998). Dynarnin is a GTPase which binds to microtubules (Cook et al., 1996) and which is involved in vesicle formation during endocytosis, synaptic transmission, and secretion (Urrutia et al., 1997). Since the study was performed only using the SH3 domain of MLK2 the specificity and physiological significance of this interaction requires further investigation. Studies of rat and mouse development suggest a role for MLK2 in testis development. Whereas MLK2 expression is temporally and spatially restricted in the rat testis, MKK4 is found at all stages and all cell types (Phelan et al., 1999) suggesting that the INK pathway is employed in various cell types and has multiple functions in rat testis, but that INK activation by MLK2 is temporally and spatially restricted. 39 The mixed-lineage kinase MLK3 (Ing et al., 1994), also called SPRK (Gallo et al., 1994) or protein tyrosine kinase-1 (PTK-l) (Ezoe et al., 1994), is a 95 kDa protein consisting of 847 amino acid residues (Gallo et al., 1994). Despite the similarity of SPRK's kinase domain to both, serine/threonine kinases and tyrosine kinases only serine/threonine kinase activity has been demonstrated in vitra (Gallo et al., 1994). SPRK contains an NHz-terminal glycine-rich region, an SH3 domain followed by a catalytic domain, a leucine/isoleucine zipper motif, a CRIB motif, and a proline/serine/threonine rich COOH-terminal region (Fig. 6). SPRK mRN A expression is detected in various tissues, with lower expression in heart and brain (Gallo et al., 1994). The MLK3 gene is localized to human chromosome 11 ql3.1-l3.3 (Ing et al., 1994). Amplifications of this region have been observed in several human malignancies, including melanonm, bladder, breast, lung, esophagus, and head and neck carcinomas (Lammie and Peters, 1991). Additionally a putative tumor suppressor gene has been mapped to 11q13 (Bale et al., 1989; Larsson et al., 1988). Overexpression of SPRK induces cellular transformation in NIH 3T3 fibroblasts, rendering them competent to grow in soft agar (Hartkamp et al., 1999). Moreover, SPRK induces tumor formation in nude mice (unpublished observation). However, very little is known about the physiological fimction of this protein kinase in cellular transformation and its potential role in human cancers. Recently there is accumulating evidence that SPRK may be involved in T cell receptor signaling. It has been demonstrated that overexpression of SPRK in T-cells 4o induces ILZ pro: a ten ic; subunit c BCIIX It} i. IO ID'CTGE: 323‘ m 9:: MEKKI a induces INK-mediated TNF-a promotor activity (Hoffmeyer et al., 1999) as well as 1L2 promotor activity (Hoffmeyer et al., 1998). Upon overexpression in HeLa cells, a cervical adenocarcinoma cell line, SPRK induces nuclear translocation of the p65 subunit of NF-ch, accompanied by an increase in NF-KB-dependent transcriptional activity (Hehner et al., 2000). In vitra kinase assays demonstrate that SPRK is able to increase Ich-kinase (IKK)a and IKKB activity, supporting the idea that SPRK may mediate NF-xB-dependent transcription. However, one has to consider that MEKK] and NTK can also act as IKKKs under these conditions, still leaving open the identity of the physiological effector of this process. Electrophoretic mobility shifts suggest that a combination of certain T cell activating stimuli decrease SPRK's electrophoretic mobility in Iurkat cells supporting the idea that T cell activation induces a change in SPRK phosphorylation. Upon overexpression of a kinase- defective SPRK variant in Iurkat cells NF-ch-dependent transcription, which is induced by these stimuli is blocked. The kinase-defective SPRK variant also inhibits Cdc42 or Rac-induced NF-ch-dependent transcription. However, this effect may be due to sequestration of the GTPases by the SPRK variant. Another study shows that a kinase-defective SPRK variant blocks Vav-induced IL-2 transcription (Hehner et al., 2000). These studies suggest a role for SPRK in T cell activation. Vav is a guaninine-nucleotide exchange factor for the small GTPase Rac. SPRK contains several domains predicted to mediate protein-protein interactions. Two-hybrid analysis and coirnmunoprecipitation experiments, for example, showed that HPKl binds to SPRK's SH3 domain (Kiefer et al., 1996). HPK] phosphorylates 41 kinase-inactive SPRK in an in vitra kinase assay (Kiefer et al., 1996), suggesting a role for HPK] in SPRK regulation, although an effect of HPKl on SPRK activity has not been demonstrated. Furthermore, kinase-defective SPRK variants and a non- phosphorylatable MKK4 mutant block HPKl-induced INK activation placing SPRK and MKK4 downstream of HPK] in the INK signaling cascade, at least upon overexpression in COS-1 cells (Kiefer et al., 1996). The leucine/isoleucine zipper is a potential motif for heterologous or/and homologous oligomerization. Experiments using a SPRK variant bearing a point mutation in the zipper motif indeed show that the zipper is required for SPRK oligomerization. The study also shows that the zipper domain is important for basal phosphorylation activity of SPRK and INK activation. Interestingly, the zipper is not required for Cdc42-induced increase in SPRK autophosphorylation activity, but is pivotal for phosphorylation of MKK4. This suggests that oligomerization by the zipper is not required for SPRK activation per se but for interaction and phosphorylation of MKK4 and subsequent INK activation (V acratsis and Gallo, 2000). The CRIB motif is a short amino acid sequence that has been identified as the binding motif for the GTPases Rae and Cdc42 (Burbelo et al., 1995). Filter binding assays show that a CRIB motif-containing fragment of SPRK binds to GTP-bound Rae and Cdc42, with a strong preference for activated Cdc42 (Burbelo et al., 1995). The result of the filter-binding assay also could be recapitulated by a two-hybrid analysis (Nagata et al., 1998), suggesting that the two proteins indeed associate in 42 3c33312.. viva. Upon coexpression of SPRK with Cdc42 or Rac in COS-7 cells SPRK autophosphorylation is increased, suggesting a role for the small GTPases in SPRK activation (Teramoto et al., 1996). SPRK contains only six of the eight conserved residues of the consensus CRIB motif. Whether SPRK contains a fiinctional CRIB motif, which mediates Cdc42 binding and perhaps Cdc42-induced, activation of SPRK has not been determined. SPRK has been identified as an upstream activator of the INK and p38/RK pathways upon overexpression in COS-1 cells (Tibbles et al., 1996). However, no activation of p38/RK could be demonstrated in 293 cells (Teramoto et al., 1996). INK activation by SPRK is mediated by the dual specific kinases MKK4 (Rana et al., 1996) and MKK7 (Whitmarsh et al., 1998). SPRK, MKK7, and INK bind to the scaffold proteins ITP] (Whitmarsh et al., 1998) and IIP2 (Yasuda et al., 1999), which subsequently induce INK activation. Stable expression of SPRK in NIH 3T3 cells induces transformation and colony formation in soft agar (Hartkamp et al., 1999). Interestingly, SPRK-expressing NIH 3T3 fibroblasts show increased MEK] phophorylation, which is not accompanied by ERK activation. In contrast c-Iun is highly phosphorylated in these cells, supporting the idea that SPRK upon overexpression constitutively activates the INK pathway. This is also consistent with our observation that Rat] fibroblasts, which stably express SPRK, have a 30- 40-fold increased INK activity compared to a vector control. Upon overexpression of SPRK in HEK 293 cells an increase in ERK activation is detected (Hartkamp et 43 al., 1999), but whether there is a physiological role for MEK] and/or ERK activation in NIH 3T3 cells is open for further investigations. Although SPRK-induced INK activation has been demonstrated in a variety of studies the upstream activators of SPRK still remain unclear. Since this protein kinase contains several potential domains mediating protein-protein interaction, a broad range of activators my exist. The small GTPase Cdc42 has been shown to bind a CRIB-containing fi'agment of SPRK in a filter-binding assay (Burbelo et al., 1995). Both, Cdc42 and SPRK have been identified as activators of the INK pathway (Bagrodia et al., 1995a; Minden et al., 1995; Tibbles et al., 1996). However, the effect of Cdc42 on SPRK's kinase activity still remains unclear. This thesis work is mainly concerned with the structural requirements and molecular mechanism of Cdc42-induced activation of SPRK. Understanding the mechanism by which the small GTPase Cdc42 regulates and activates the protein kinase SPRK may provide valuable infornntion about Cdc42-SPRK-induced INK activation. It may provide more insight into the specific physiological role of SPRK and perhaps help to identify the extracellular stimulus for SPRK activation. In addition, understanding the molecular mechanism of SPRK regulation by Cdc42 may help to understand the regulation of protein kinases by small GTPases in general. Fig 6. I) leucine ixIi] high?!) and l Fig. 6. Domain arrangement of SPRK. SPRK consists of an NHz-termina] glycine-rich region, followed by an SH3 domain, a kinase domain and a leucine/isoleucine zipper region. Adjacent to the CRIB motif SPRK contains a basic region and a COOH-terminal serine/threonine/proline rich region. 45 [I]. C do] viva: Re III. Cdc42-Induced Activation of the Mixed-Lineage Kinase SPRK in viva: Requirement of the Cdc42/Rae Interactive Binding Motif and Changes in Phosphorylation 1. Abstract SPRK/MLK-3 is a serine/threonine kinase tint upon overexpression in mammalian cells activates the INK pathway. The mechanisms by which SPRK activity is regulated are not well understood. The small Rho-family GTPases, Rae and Cdc42, have been shown to bind and to modulate the activities of signaling proteins, including SPRK, which contain Cdc42/Rae interactive binding motifs. Coexpression of SPRK and activated Cdc42 increases SPRK’s activity. SPRK’s CRIB-like motif contains six of the eight consensus residues. Using a site-directed mutagenesis approach, we show that SPRK contains a fimctional CRIB motif that is required for SPRK’s association with and activation by Cdc42. However, experiments using a SPRK variant that lacks the COOH-terminal zipper region/basic stretch suggest that this region may also contribute to Cdc42 binding. Unlike the PAK family of protein kinases, we find that the activation of SPRK by Cdc42 cannot be recapitulated in an in vitra system using purified, recombinant proteins. Comparative phosphopeptide mapping demonstrates that coexpression of activated Cdc42 with SPRK alters the in viva phosphorylation pattern of SPRK suggesting that the mechanism by which Cdc42 increases SPRK’s catalytic activity involves a change 46 in {he in r first dcrr Pikrsp'mr} mm 1 lntrod in the in viva phosphorylation of SPRK. This is, to the best of our knowledge, the first demonstrated example of a Cdc42-mediated change in the in viva phosphorylation of a protein kinase. These studies suggest the requirement for an additional component or the cellular environment for SPRK activation by Cdc42. 2. Introduction The serine/threonine kinase SPRK is a member of the mixed-lineage kinase Emily. SPRK contains various domains mediating protein-protein interaction, including an NHz-terminal SH3 domain, a leucine/isoleucine zipper motif, a CRIB motif, and a large COOH-terminal region that is rich in serine, threonine, and proline residues (Fig. 7). The CRIB motif is a short amino acid sequence consisting of 14-16 amino acid residues, and has been identified as the minimal binding sequence for small GTPases of the Rho-Emily (Burbelo et al., 1995). SPRK shares six of the eight conserved amino acid residues within this motif. In filter binding assays it has been demonstrated that SPRK associates with Rae and Cdc42, with a strong preference for Cdc42 (Burbelo et al., 1995). Cdc42 belongs to the Rho Emily of small GTPases. This protein Emily, which includes Rho, Rae, and Cdc42, plays crucial roles in diverse cellular processes (Hall, 1998; Van Aelst and D'Souza-Schorey, 1997), including cytoskeletal rearrangements (Kozrna er al., 1995; Nobes and Hall, 1995; Ridley and Hall, 1992), cell cycle progression (Olson et al., 1995), cellular transformation (Khosravi-Far et 47 01.. 1995; ' Tang e! u.’ (Bagrodia Minden er . his ac: al., 1995; Qiu et al., 1997; Qiu et al., 1995a; Qiu et al., 1995b; Tang et al., 1997; Tang et al., 1999; Whitehead et al., 1998; Wu et al., 1998), and nuclear signaling (Bagrodia et al., 1995a; Brown et al., 1996; C050 et al., 1995; Hill et al., 1995; Minden et al., 1995; Zhang et al., 1995). These proteins can also function as protein kinase activators. One well-characterized target of Cdc42 and Rac is the serine/tineonine kinase PAK (Bagrodia et al., 1995b; Manser et al., 1994; Martin et al., 1995). The interaction between Cdc42 and the serine/threonine kinase PAK requires the CRIB motif (Burbelo et al., 1995). The structural determinants required for GTPase binding and the mechanism of activation of multiple PAK isoforms have been extensively investigated (Gatti et al., 1999; Knaus et al., 1998; Manser et al., 1997; Manser et al., 1994; Zenke et al., 1999; Zhao et al., 1998). CRIB-dependent interaction of PAK with GTP-bound Rac/Cdc42 induces PAK autophosphorylation and activation both in vitro and in viva. Diverse proteins, including the tyrosine kinase, ACK (Manser et al., 1993), and the non kinase, WASP (Aspenstrom et al., 1996; Kolluri et al., 1996), also contain CRIB motifs, suggesting that mechanistically diverse regulatory pathways may share this common structural motif. Likewise, not all protein kinases which interactwitthc42andRacdo sothroughaCRIB motif. For instance, boththe 70 kDa ribosomal S6 kinase (Chou and Blenis, 1996) and MEKK] (Fanger et al., 1997), which lack CRIB motifs, have been shown to interact with GTP-bound Cdc42 and Rac. Furthermore, MEKK4 contains a modified CRIB motif whose deletion only 48 lik 10 Cd Lin Poi inc; C0:- 0P. SP] partially diminishes binding to Cdc42 and Rac, indicative of a CRIB-independent GTPase binding determinant (Fanger eta1., 1997). Thus the role of CRIB motifs and the mechanisms by which many protein kinases are activated by small GTPases remain largely unexplored. Whether activation by Cdc42 of the distantly related PAK and SPRK involves mechanistically analogous processes is unknown. SPRK is Enctionally homologous to Raf, and thus mechanistic aspects of Cdc42-SPRK and Ras-Raf activation may be shared. Here we show that SPRK contains a Enctiona] CRIB motif that is absolutely required for Cdc42 binding to and activation of SPRK. The zipper region and adjacent basic sequences of SPRK may also contribute to Cdc42 binding. Binding of Cdc42 to SPRK does not require SPRK catalytic activity. Interestingly, GTP-bound Cdc42 has no effect on the activity of purified, catalytically active SPRK, suggesting that an additional cellular component is required for kinase activation. These studies point to an important distinction between PAK and SPRK in the mode of GTPase- induced kinase activation. Comparative phosphopeptide mapping revealed that coexpression of activated Cdc42 with SPRK alters the in viva phosphorylation sites on SPRK. This change in serine/threonine phosphorylation correlates with increased SPRK activity. These studies represent the first case where a Cdc42-mediated change in the in viva phosphorylation sites of a protein kinase has been documented and provide evidence for the involvement of in viva phosphorylation of SPRK for Cdc42- 49 induced a; from the rr 3. Mater 3.] Constr mutagEn. induced activation. Thus, the Cdc42-mediated activation of SPRK is clearly distinct fi‘om the mechanism previously described for Cdc42-induced activation of PAK. 3. Materials and Methods 3.1 Construction of mammalian expression vectors and mutagenesis Construction of the cytomegalovirus-based expression vectors carrying the cDNAs for wild type SPRK (pRKS-sprk), and for the kinase-defective variant of SPRK (pRKS-sprkm‘“) has been described elsewhere (Gallo et al., 1994). Expression plasmids encoding NIB-terminal Flag epitope-tagged wild type Cdc42 (pRKS-Nflag.cdc42) and the constitutively active variant (pRKS-Nflag.cdc42v‘2) of Cdc42 were kindly provided by Avi Ashkenazi (Genentech, Inc., So. San Francisco, CA). Variants of SPRK containing point mutations in the CRIB motif were constructed by a modified recombinant polymerase chain reaction (PCR) method described in detail elsewhere (Higuchi, 1990). For each mutation two different PCRs were performed, one containing a left mutagenesis primer and a left outside primer, and one containing a right mutagenesis primer and a right outside primer. The left and right outside primers used for all mutations were 5’-GATG- AGTCATCTGAATCCAGG-3 ’ and 5’-CTGTGGCCTATGGCGTAGCTG-3’, respectively. To obtain the three difierent CRIB mutants the following left and right mutagenesis primers were used, respectively: (SPRKmsA), 5’-GGTGCTTGGCGT- 50 CGAGTGGCAT-3’ and 5’-ACTCGACGCCAAGCACCGCATC-3’; (SPRK"5°°A)' 5’-CTTCAAGGCCCGCATCACCGT-3’ and 5’-GGTGATGCGGGCCTTGAAG- TCG-3’; (SPRKWW93A), 5’-GAAGTCGAGTGGCATGGCGGCACGCT-CG-3’ and 5’-CGAGCGTGCCGCCATGCCACTCGAC-3’. The presence of the desired mutation was confirmed by DNA sequencing using the Sanger method, and the absence of PCR-introduced errors was verified by automated sequencing. Deletion of amino acids 430 through 486 of SPRK to yield SPRKAzip was accomplished by digestion of the expression vector pRKS-sprk with BssH H, followed by ligation with T4 DNA ligase. DNA modifying enzymes were purchased fi'om New England Biolabs or Life Technologies, Inc.. 3.2 Expression and purification of recombinant SPRK An Nca I-Hind III fragment, containing the full length SPRK cDNA, was subcloned fi'om pRKS-sprk into the pFastBac HTb baculovirus expression vector (Life Technologies, Inc.) which contains a hexahistidine tag. Recombinant histidine- tagged SPRK was expressed in Sf9 cells and purified by nickel affinity chromatography according to the manufacturer's protocol. Fractions containing histidine-tagged SPRK, as determined by SDS-PAGE followed by Coomassie Blue staining, were pooled and concentrated using a Centriprep concentrator (Amicon). 51 3.3 Cell lines and transfections Human fetal kidney 293 cells were maintained in Ham’s F12:low glucose Dulbecco’s modified Eagle’s media (1:1) (Life Teclmologies, Inc.) supplemented with 8% fetal bovine serum (Life Technologies, Inc.), 2 mM glutamine, and penicillin/streptomycin (Life Technologies, Inc.). Plasmids (5 pg each for 60 mm dishes; 10 pg each for 100 mm dishes) were used to transfect 293 cells using the calcium phosphate technique (Gorman, 1990). Cell monolayers were incubated with the DNA precipitate for 4 h, then washed once with PBS (phosphate-bufi‘ered saline), and cultured in the mdium described above. After 18 h cells were harvested. 3.4 Cell lysis and immunoprecipitation Cells were washed with ice cold PBS and lysed for 5 min on ice by the addition of 1 ml of lysis buffer (50 mM HEPES (pH 7.5), 150 mM NaCl, 1.5 mM MgCh, 2 mM EGTA, 1% Triton X—100, 10% glycerol, 10 mM NaF, 1 mM NaIP., 100 uM B- glycerophosphate, 1 mM Na3VO4, 2 mM PMSF, and 0.15 U/ml aprotinin). The lysate was clarified by centrifugation for 20 min at 14,000 rpm in an Eppendorf centrifuge at 4 °C. Rabbit polyclonal antiserum was raised against a peptide corresponding to the COOH-terminal eight amino acids of SPRK and was purified by Protein A-Sepharose chromatography. Antibodies against the proteins of interest were prebound to protein A—agarose beads [SPRK antiserum (0.25 rig/u] slurry), M2 monoclonal antibody (Kodak IBI) directed against the Flag epitope (0.45 rig/u] slurry) and INK C-17 52 antibody (Santa Cruz Biotechnology) (50 ng/ul slurry) as previously described (Gallo er al., 1994). Clarified lysate (300 pl) was incubated with 20 u] of antibody-bound Protein A-agarose for 90 min at 4 °C. Immunoprecipitates were washed with HNTG buffer (20 mM HEPES (pH 7.5), 150 mM NaCl, 0.1% Triton-X-100, 10% glycerol). Immunoprecipitates used for kinase assays were washed three times with HNTG buffer containing 1 M LiCl, three times with [MTG buffer, and twice with kinase assay bufi‘er (50 mM Tris-HCl (pH 7.5), 100 mM NaCl, 1 mM MnClz, 10 mM MgC12, 0.1 mM Na3VO4). 3.5 Gel electrophoresis and Western blot analysis Lysates and immunoprecipitates of SPRK and Cdc42 were resolved by SDS- polyacrylamide gel electrophoresis (SDS-PAGE) according to Laemmli (Laemmli, 1970). Proteins were transferred to nitrocellulose and irnmunoblotted using either SPRK antiserum (1 ug/ml) or M2 Flag monoclorml antibody (9 rig/ml), followed by the appropriate horseradish peroxidase-conjugated secondary antibody (Life Technologies, Inc.). Western blots were developed by chemiluminescence. Multiple exposures of the Western blots were developed, and densitometry (NIH Image) of unsaturated films was used to determine relative expression levels. Statistics were compiled using an unpaired Student's t-test. A p—value smaller than 0.05 was considered statistically significant. 53 3.6 In vitro kinase assays Kimse assays were performed in 20 pl of kinase assay buffer containing 50 pM ATP and 5 pCi [y-3ZP]-ATP (3000 Ci/mmol) (NEN Life Science Products). For the SPRK kinase assay 10 pg of mixed histones (Boehringer Mannheim) was used as a substrate and the reaction was carried out for 30 min at room temperature. Independent experiments showed that the reaction was linear within this time range. The reactions were terminated by the addition of an equal volume of 2x SDS sample buffer (100 mM Tris (pH 6.8), 4% SDS, 20% glycerol, 0.2% bromphenol blue, 100 mM DTT, 1% B—mercaptoethanol) containing 50 mM EDTA (pH 8.0). For the kinase assays involving purified kinases, recombinant SPRK or PAK-2 (1 pg and 2 pg, respectively) was incubated in 50 pl of kinase assay buffer containing 4 pg of GST (glutathione S-transferase)-Cdc42 that had been preloaded with GTPyS or GDP and assays were performed as above. GST-Cdc42 (19 pM) was preloaded with GTPyS or GDP (4 mM) in buffer containing SOmM Tris (pH 7.5), 5 mM EDTA, and 1 mM DTT. The mixture was incubated for 15 min at 30 °C. The nucleotide loading reaction was quenched by the addition of 10 mM MgC12. Purified GST-Cdc42 was obtained from an E. cali overexpression system; and purified PAK-2, obtained using the baculovirus expression system, was kindly provided by Dr. Arie Abo (Onyx Pharmaceuticals) (Martin et al., 1995). For the INK assays, 8 pg of GST-c-jun was used as the substrate, and the reaction was carried out for 15 min at room temperature. The pGEX-c-jun (1-115) 54 vector was kindly provided by Dr. Ajay Rana (Massachusetts General Hospital, Harvard Medical School, Boston, MA). GST-c-jun was expressed in XL-l Blue E. cali and purified by glutathione Sepharose chromatography. Following the kinase assay, proteins were separated by SDS-PAGE. Gels were rinsed in PBS, dried, and incorporation of radioactivity into kinase, or substrates was determined by Phosphorlmaging (Molecular Dynamics). To detect INK expression, proteins were transferred fi'om an SDS polyacrylamide gel to a polyvinylidene difluoride (PVDF) membrane and irnmunoblotted using the INK C-17 antibody (0.5 pg/ml). 3.7 In viva phosphopeptide mapping After a 24 h transfection with pRKS-sprk in the presence or absence of pRKS- Nflag.cdc42m, 293 cells were washed five times with phosphate-flee medium (Dulbecco’s modified Eagle’s medium supplemented with 10% dialyzed fetal bovine serum (Summit Biotechnology», and incubated at 37 °C for 2 h. The cells were then incubated in phosphate-free medium containing 3 mCi/ml [”P] orthophosphate (carrier free; NEN Life Science Products) for 4 h at 37 °C. Cells were washed five times with ice cold PBS and then lysed in 1 ml of lysis buffer. Lysates were clarified by centrifugation for 15 min at 14,000 rpm in an Eppendorf centrifuge at 4 °C. SPRK was immunoprecipitated with SPRK antiserum as described above. Immunoprecipitated proteins were resolved by SDS-PAGE and transferred to a PVDF membrane. Radiolabeled bands that co-migrated with SPRK, 55 as judged by Western blotting, were excised fi'orn the PVDF membrane. After washing three times with methanol and three times with water, the radioactive piece of membrane was blocked with 1 ml of 0.5% polyvinylpyrrolidine-36O (Sigma) containing 100 mM acetic acid for 30 min at 37 °C, and then washed five times with water. Tryptic digestion was performed with 10 pg of sequencing grade trypsin (Boehringer Mannheim) for 2 h in 200 pl of 50 mM NH4HC03, pH 8.3, at 37 °C. An additional 10 pg of trypsin was added and the digestion mixture was incubated for an additional 2 h at 37 °C. The membrane was then sonicated for 3 min in 300 pl of water to remove additional tryptic peptides. The solution containing the released tryptic peptides was concentrated in a SpeedVac (Savant Instruments). The peptides were separated on cellulose thin layer chromatography (TLC) plates (Kodak, 20 x 20 cm) by thin layer electrophoresis (TLE) in the first dimension in pH 1.9 bufier (formic acid (88% w/v)/glacial acetic acid/water, 25:78:897, v/v/v) at 0°Cand1000Vfor30min,andseparatedintheseconddimensionbyTLCin phosphochromatography buffer (n-butanol/pyridine/ glacial acetic acid/water, 15:10:3zl2, v/v/v/v). The radiolabeled phosphopeptides were visualized and quantitated using a phosphorimager. 56 4. Results 4.1 Association of activated Cdc42 with SPRK does not require SPRK kinase activity Recently Hall and coworkers have shown that several proteins, including SPRK, associate with GTP-bound Cdc42 and Rac in filter binding assays (Burbelo et al., 1995). All of these proteins contain a 14-16 amino acid sequence that includes eight consensus residues, which has been coined the CRIB motif. The CRIB motif of SPRK contains six of the eight consensus amino acids (Fig. 8). In this study we examined the structural requirements and mechanism of Cdc42 binding and activation of SPRK. SPRK and Flag epitope-tagged Cdc42 expression vectors were transiently transfected in 293 cells. To mimic the GTP-bound state of Cdc42 we used a constitutively active mutant of the GTPase, i.e.Cdc42V'2. Cellular lysates from 293 cells were immunoprecipitated with the Flag antibody and the presence of associated SPRK was assessed by Western blot analysis with a SPRK antibody. In these co- immunoprecipitation experiments (Fig. 9), the constitutively active form of Cdc42, but not wild type Cdc42, associates with SPRK (first panel). We have not observed association of SPRK with the dominant negative variant Cdc42N”, but, in our hands. expression of this Cdc42 variant has been low. Expression levels of SPRK and Cdc42 in celluLar lysates were determined by Western blot analysis (Fig. 9, lower pane18). 57 These data indicate that SPRK preferentially associates with the GTP-bound form of Cdc42. SPRK can autophosphorylate in vitra (Gallo et al., 1994). Although SPRK’s postulated site of interaction with Cdc42 is not within the catalytic domain, it is plausible that either SPRK autophosphorylation or SPRK phosphorylation of another interacting molecule might modulate the Cdc42-SPRK interaction. To examine whether SPRK’s catalytic activity effects its ability to bind to Cdc42, the kinase- defective SPRK variant (SPRKK'W‘) was tested for its ability to associate with Cdc42m. The SPRKKIMA variant shows no autophosphorylation in an in vitra kinase assay (Gallo et al., 1994). Cotransfection and co-irmnunoprecipitation experiments with active and inactive SPRK, and Cdc42m, demonstrate that constitutively active Cdc42 associates equally well with wild type SPRK and the kinase—defective variant SPRKW" (Fig. 10, upper panel). This indicates that SPRK’s catalytic activity is not required for its association with Cdc42. The retarded electrophoretic mobility of wild type SPRK, as compared with that of inactive SPRK, suggests a more highly phosphorylated form of SPRK that is likely due, at least in part, to autophosphorylation (Fig. 10, upper and middle panels). To determine whether association with Cdc42 effects SPRK’s catalytic activity, an in vitra kinase assay for SPRK was developed. While its kinase domain shares overall sequence similarity with both serine/threonine and tyrosine kinases, 58 SPRK shows high amino acid sequence identity with the serine/threonine kinase B- Raf in a small region of subdomain VIb and VIII that is important for substrate specificity and catalytic activity (Hanks et al., 1988). In addition, both Rafand SPRK appear to function as MKKKs leading prirmrily to the activation of ERK and INK, respectively. Since histones are commonly employed as exogenous substrates for Raf in in vitra kinase assays (Kanakura et al., 1991; Tarnaki et al., 1992), a mixture of histones was tested as an exogenous substrate for SPRK. SPRK, immunOprecipitated from cellular lysates of transiently transfected 293 cells, exhibits basal autophosphorylation as well as phosphorylation of the histones H3 and H4 (Fig. 11). In contrast, the kinase-defective SPRK shows no autophosphorylation or histone phosphorylation, indicating that the phosphorylation events observed in these assays are attributable to SPRK, rather than to contaminating kinases. W2 2 The expression of Cdc4 in transiently transfected 293 cells increases the autophosphorylation activity as well as the substrate phosphorylation activity of SPRK about 3- to 5-fold (Fig. 11). No background phosphorylation is observed with the kinase-defective SPRK variant in the presence or absence of Cdc42v'2. Interestingly, under these in vitra kinase assay conditions, we do not detect Cdc42 associated with SPRK by Western blotting. 59 4.2 SPRK’s CRIB motif is necessary for association with Cdc42 and for Cdc42 induced activation of SPRK To test whether SPRK’s potential CRIB motif actually functions in the binding of Cdc42 we took a site-directed mutagenesis approach. Three difierent SPRK variants were generated by mutating conserved amino acids in the CRIB motif to alanine residues: SPRKF‘98‘, SPRKHW, and Sl>RK"”"'S493A (Fig. 8). While we cannot absolutely rule out the possibility that introduction of these mutations might alter SPRK’s conformation, the expression levels of the SPRK CRIB mutants in transient transfections of 293 cells are at least as high as that of wild type SPRK (Fig. 12), suggesting that these variants are stable. The CRIB variants were coexpressed with Cdc42V12 in 293 cells and the cellular lysates were subjected to co- immunoprecipitation experiments. While wild type SPRK associates with Cdc42m, none of the SPRK CRIB mutants detectably associates with the activated GTPase (Fig. 12). If Cdc42-induced activation of SPRK is mediated through its interaction with SPRK’s CRIB motif, one would expect that the SPRK CRIB mutants should exhibit a defect in Cdc42-induced activation. Accordingly, cells were transiently transfected with cDNAs encoding wild type SPRK or SPRKWZA‘8493A, in the presence or absence of Cdc42m. The activity of the immunoprecipitated SPRK or SPRK CRIB mutant was measured in an in vitra kinase assay. In the absence of Cdc42m, both wild type SPRK and the SPRK CRIB variant show similar levels of autophosphorylation and 60 substrate phosphorylation (Fig. 13, 14a and b). In the absence of Cdc42V”, the differences in the activities of SPRK and the SPRK CRIB variants were not statistically significant. This further supports the idea that the mutations in the CRIB motif do not grossly perturb SPRK’s structure or inherent catalytic activity. However, both autophosphorylation and substrate phosphorylation of the CRIB variant is markedly lower (3-fold) than that of wild type SPRK, when each is co- expressed with the activated GTPase (Fig. 13, 14a and b). The small increase in the catalytic activity of the SPRK CRIB mutant when activated Cdc42 is coexpressed my be due to Cdc42-activated endogenous SPRK. Alternatively there may be residual binding of the CRIB variant to activated Cdc42 in viva, which we do not detect in our in vitro co-immunoprecipitation assay. Taken together these results demonstrate that SPRK does contain a functional CRIB motif, and that Cdc42- induced activation of SPRK is mediated via association with this CRIB motif. 4.3 Effects of deleting the COOH-terminal portion of SPRK’s zipper domain In WASP (Rudolph et al., 1998), PAK (Thompson et al., 1998; Zhao et al., 1998) and ACK (Manser et al., 1993) the CRIB motif is necessary but not suflicient for GTPase binding. Outside of the CRIB motif SPRK shares no sequence similarity with the minimal GTPase binding domains that have been defined for these proteins. Instead, SPRK contains two closely spaced leucine/isoleucine zipper motifs spanning amino acids 400-462, NHz-terminal to the CRIB motif (Fig. 7). Considering the close 61 vicinity in linear sequence of the zippers and the CRIB motif we asked whether the zipper motif might contribute to the binding of SPRK to Cdc42m. Accordingly, a variant of SPRK, SPRKm’, which lacks amino acids 430-486, as shown in Fig. 7, was constructed. This deletion removes the second half of the zipper region as well as 22 COOH-terminal amino acids, which include a short basic region, but leaves the entire CRIB motif intact. SPRK“ip is expressed in transiently transfected 293 cells at levels comparable to that of wild type SPRK (Fig. 15). The ability of Cdc42v'2 to associate with SPRKM" was tested in co-imrnunoprecipitation experiments with cellular lysates harvested from transiently transfected 293 cells. The deletion of the second zipper/basic stretch greatly diminishes the ability of SPRK to bind to Cdc42v'2, despite the presence of the complete CRIB motif (Fig. 15). Based on these and our previous results, both the CRIB motif, the second half of the zipper region and a stretch of basic amino acids may contribute to Cdc42 binding. Alternatively, the COOH-terminal zipper/basic stretch may not directly interact with Cdc42, but may be required for the proper presentation and binding of the CRIB motif to the GTPase. SPRK“? has approximately 70% ofthe basal autophosphorylation activity of wild type SPRK (Fig. 16, 17a). However, in contrast to wild type SPRK, there is no Cdc42-induced increase in autophosphorylation of SPRK”, consistent with the finding that SPRKM" binds Cdc42VIZ only very weakly. In an in vitra kinase assay, SPRK” expressed with or without mm"12 (Fig. 16, 17b) kicks the ability to 62 phosphorylate histones. To address whether the lack of histone phosphorylation might be due to some unique feature of histones we performed the same experiment with myelin basic protein as a substrate and obtained analogous results (data not shown). These data suggest that the zipper domain/basic stretch may be fimdamentally required for substrate pho sphorylation. 4.4 SPRKA" fails to activate JNK SPRK has been identified as an upstream activator of the INK/SAPK pathway (Rana et al., 1996; Teramoto et al., 1996; Tibbles et al., 1996). INK activity was measured after immunoprecipitation of endogenous INK from cellular lysates of transiently transfected 293 cells in an immune complex in vitra kinase assay using GST-c—jun as the substrate. Overexpression of SPRK in 293 cells leads to a 5-fold increase in INK activity over the basal activity in vector control-transfected cells (Fig. 18, 19). Transient expression of Cdc42V12 increases INK activity some 2- to 3-fold. Although coexpression of SPRK and Cdc42VIZ increases SPRK catalytic activity, as measured in an in vitra kinase assay, the observed SPRK-induced INK activation with co-expressed Cdc42V12 over that of SPRK alone does not reach statistical significance. A likely explanation for this finding is that the activity of overexpressed SPRK alone is sufficient to maximally activate the endogenous INK in 293 cells. SPRKAZ" is unable to phosphorylate an exogenous substrate in an in vitra kinase assay, and, indeed, we have found tint it completely lacks the ability to activate the 63 INK pathway (Fig. 18, 19). Thus, it appears that the zipper motif is critical for substrate phosphorylation by SPRK both in vitra and in viva. 4.5 Activated Cdc42 fails to activate SPRK in vilra The small GTPases Rae and Cdc42 can stimulate the autophosphorylation activity of the CRIB-containing serine/threonine kinase PAK2 in vitra (Martin et al., 1995). To determine whether Cdc42-induced activation of SPRK can be recapitulated in a completely in vitra system', hexahistidine NHz-terminal tagged SPRK was expressed using the baculovirus system, and purified by metal-chelate chronntography’. GST- Cdc42 was expressed in and purified fiom E. coli’. The purified SPRK is catalytically active as judged by its basal autophosphorylation activity. GTPyS- or GDP-loaded GST-Cdc42 was incubated with purified SPRK or PAK2 in an in vitra kinase assay (Fig. 20b). While GTP-yS-loaded Cdc42 activates purified PAK2, it Eils to activate purifed SPRK. Likewise, SPRK immunoprecipitated fi'om transfected 293 cells carmot be activated in vitra by the addition of GTPyS-loaded Cdc42 (Fig. 20a). These data support the requirement of a cellular context or coactivator for SPRK activation by Cdc42. 4.6 Cdc42 alters the in viva phosphorylation pattern of SPRK As described above, co-immunoprecipitation experiments and in vitra kinase assays show that Cdc42“2 when coexpressed with SPRK can associate with SPRK and modulate its catalytic activity. However, purified, activated Cdc42 cannot stimulate the autophosphorylation of SPRK in vitra. In order to determine if the presence of activated Cdc42 alters SPRK phosphorylation in viva, two-dimensional phosphopeptide analysis of SPRK labeled in viva, in the absence and in the presence of Cdc42m, was performed“. The net incorporation of radiolabel into SPRK increased approximately three- fold when SPRK was coexpressed with Cdc42m. Two-dimensional TLE/TLC revealed that while the basic pattern of phosphopeptides from the two samples is similar (Fig. 21a), there are notable differences. The major changes are observed in the triangular cluster of phosphopeptides b, c, and d. Phosphopeptide a predominates in both samples. Phosphopeptides b and c are detected in the triangular cluster of the SPRK map, but are low in abundance relative to peptide 0. In the corresponding map of SPRK that had been expressed in the presence of Cdc42m, however, phosphopeptide b is not detected. Instead, phosphopeptide c is the prominent phosphopeptide in the triangular cluster, with 70% of the radioactivity of phosphopeptide a. (Fig. 21b). Furthermore, phosphopeptide (I, nearly undetectable in the map of SPRK, appears at high levels in the map of SPRK expressed with Cdc42m. For comparison, the level of another peptide (1:) relative to peptide 0 is essentially constant in both maps. The labeling and mapping fiom three independent experiments yielded the same results. These data indicate that the presence of 65 activated Cdc42 changes the in viva phosphorylation state of SPRK, which correlates with an increase in SPRK catalytic activity. 66 Fig. 7. Schematic of SPRK. The numbers in the above diagram represent amino acid number. The glycine-rich region (amino acids 1-42) is denoted by Gly. The region containing the zipper motif and the polybasic stretch of amino acids includes amino acids 400-486. The amino acid sequence corresponding to this region is shown below with the basic stretch of amino acids printed in bold letters and the non- aromatic hydrophobic residues predicted to occupy the d position in the zipper motifs underlined. The sequence deleted in SPRKAzip (amino acids 430-486) is boxed. 67 5% r "trill 10444.waEJWSOSJImmggmmagammmmmi—mxEJBEJGO—mz; 0:-.. no 0.. . 23m > a ham «8 8m «aims So an .3 m2 9 F 68 Consensus stpxxxxrxnxxava MLK2 m-rsxr. .xxr'xrixxrivca-487 WASP 233-IG'XP. .xxri'xnxxavcr-z51 ACK 510-:rsxraxxxxrl'xuxxuea-527 SPRK 492-stpxxxxrxnxx1vQ—sos SPRKWW ALI l SPRKWA a SPRKHESOOA A Fig. 8. Alignment of CRIB motifs and SPRK variants. The consensus sequence for the CRIB motif as defined by Burbelo et a1. is shown aligned with the CRIB motif of MLK2, WASP, ACK, and SPRK. Amino acid numbers are indicated to the left and the right of each sequence. Amino acids occupying the consensus residues are indicated by bold, with those that differ from the consensus italicized. Absent amino acids are indicated by period The mutations in the engineered CRIB variants of SPRK are shown with the conserved residue to alanine changes indicated by bold letters. 69 SPRK: - - .. + + + Cdc42: - V12 wr - v12 wr lmmunoblot Flag IP . SPRK I .5 .‘ SPRK Flag-Cdc42 Fig. 9. Co-immunoprecipitation of SPRK with wild type (WT) Cdc42 or Cdc42‘m. 293 cells were transfected with expression vectors containing the cDNA indicated above. A minus sign indicates that a control empty vector was transfected. Cdc42 was immunoprecipitated (IP) using the M2 antibody directed against the Flag epitope appended to the NHz-terminus of Cdc42 and Cdc42m. The presence of SPRK was determined by irnmunoblotting with a SPRK antibody (upper panel). The middle and lower panels show SPRK and Cdc42 expression, respectively. 70 SPRK: WT K144A WTK144A Flag IP - - SPRK . ~~ SPRK w Flag-Cdc42 Fig. 10. Co—immunoprecipitation of SPRKKI‘“ with activated Cdc42v". 293 cells were transfected with expression vectors containing the cDNA indicated above. The upper panel shows the immunoprecipitation of wild type SPRK or SPRKK‘W‘ with activated Cdc42m. The presence of SPRK was determined using a SPRK antibody. The middle and lower panel show SPRK and Cdc42 expression, respectively. 71 SPRK: _ WT K144A WT K144A Cdc42m= - .. - + + autoradiogram I <— SPRK " ‘— histonee immunoblot - ~~ SPRK v Flag-Cdc42 Fig. 1]. In vitra kinase assay of SPRK and SPRK'm“ coexpressed with Cdc42m. SPRK was immunoprecipitated from cellular lysates and subjected to an in vitra kinase assay using histories as a substrate. The top panel shows an autoradiogram with bands corresponding to SPRK autophophorylation and histone phophorylation, indicated by arrows. The middle and lower panels show SPRK and Cdc42 expression, respectively. 72 SPRK= -wrF H IWTF Cdc42‘m:-. .++ Flag IF it ‘ ' ' ”a i I‘“. SPRK H I + immunoblot SPRK Flag-Cdc42 Fig. 12. Effects of mutations in the CRIB motif an association of SPRK with Cdc42v". Co-irnmunoprecipitation experiments of SPRK or SPRK CRIB variants with Cdc42m. The presence of bound SPRK or SPRK CRIB variant was assessed by immunoblotting with a SPRK antibody as described previously (upper panel). The middle and lower panels show SPRK and Cdc42 expression, respectively. WT indicates wild type SPRK, F indicates SPRKFW, H indicates SPRKHSW, and I indicates SPRKMQA'S493A. 73 SPRK: - wr |492A- - wr mm- S493A S493A Cdc42m: . _ _ + + + autoradiogram 4— SPRK ‘ ' ‘_ histories Q ... - lmmunoblot it. than... In. spar ** w Flag-Cdc42 Fig. 13. Effects of mutations in the CRIB motif on SPRK in vitra kinase activity. In vitro kinase assay of SPRK and SPRKWA'W“. The autoradiogram (upper panel) shows SPRK autophosphorylation and histone phosphorylation indicated by the arrows. The middle and lower panels show SPRK and Cdc42 expression, respectively. 74 auophoephoryletlen C d N 0) ‘ U! a N r a a r a l 4 Fold lncreaee In h I F 0 -e N U 1‘ (1| a N zu_ A L_ # 4 4L A 1 Fold lncreeee In hletone phoephoryleflon Fig. 14. Quantitation of in vitro kinase assays of SPRK or SPRKmu‘w“. a, SPRK autophosphorylation and b, histone phosphorylation which were determined by in vitra kinase assays were quantified by PhosphorImaging and normalized to relative SPRK expression levels. The mean :t SE. of three independent experiments are shown. 75 SPRK: _ WT Azlp WT Azlp Cdc42‘m: + 4- Flag lP . eaSPRK — - - .9er t - Flag-Cdc42 immunoblot Fig. 15. Effects of partial deletion of the zipper/basic stretch an association with Cdc42v". Co-immunoprecipitation experiment of SPRK and SPRKM" with Cdc42v'2. The presence of bound SPRK or SPRKM" was assessed by irnmunoblotting with a SPRK antibody as described previously (top panel). The middle and lower panels show SPRK expression, respectively. 76 SPRK: _ wr Azlp _ wr Azip Cdc42”: - - - + + "’ autoradiogram WP ‘. . “ELK ‘ <— SPRK . . * hietonee '4'" u ‘ immunoblot ~ ~ SPRK m ' Flag-Cdc42 Fig. 16. Effects of partial deletion of the zipper/basic stretch on SPRK in vitro kinase activity. In vitra kinase assay of SPRK and SPRKM" using histones as a substrate. The top panel shows an autoradiogram with bands corresponding to autophosphorylated SPRK and phosphorylated histories indicated by arrows. The lower and middle panels show SPRK and Cdc42 expression, respectively. 77 ‘ J (.0 hleeene phoephoryleuon Fold Inereaee In Fig. 17. Quantitation of in vitro kinase assays of SPRK and SPRKMP. a, SPRK autophosphorylation and b, histone phosphorylation which were determined by in vitra kinase assays were quantified by PhosphorImaging and normalized relative to SPRK expression levels. The mean i 8.13. of three independent experiments are shown. 78 SPRK: _ wr Azip .. wr Azip m. 66°42 - - - - + + + 'utondiognm JNKlP V I ”(I' ’ A”: m is”; _4—GST-e-jun immunoblot ' “"91”". r.“‘!*"" ‘ JNKIP ”A“ + ”j. m...» 7w. pi JNK ”- ”Mm Fig. 18. Effects of SPRKMp on JNK activity. Endogenous INK was immunoprecipitated fi'om cellular lysates that had been transiently transfected with cDNAs encoding the specified SPRK variants and/or Cdc42m. An in vitra kinase assay for INK was performed using GST-c-jun as a substrate. The top panel shows an autoradiogram with bands corresponding to phosphorylated GST-c-jun indicated by an arrow. The second panel shows a INK irnmunoblot of the same immunoprecipitated samples from the in vitra kinase assay. The third and bottom panels show SPRK and Cdc42 expression, respectively. 79 Fold lnereaee In GST-e-jun Fig. 19. Quantitation of JNK in vitra kinase assays. 293 cells were transiently transfected with cDNAs encoding the specified SPRK variant and/or Cdc42. INK activity was determined by GST-c-jun phosphorylation, which was quantified by Phosphorlrnaging and normalized relative to SPRK expression levels. The mean t SE. of three independent experiments are shown. 80 Cdc42: GDP GTPYS autoradiogram ‘ " a“ d—SPRK immunoblot ”~— SPRK Cdc42: GDP GTP-18 GDP GTPyS . r, autoradlogram W m d—SPRK PAK 2 —> 'I‘ Fig. 20. In vitro kinase assay using purified Cdc42. a, SPRK was immunoprecipitated fiom cellular lysates that had been transfected with cDNA encoding SPRK and was incubated in an in vitra kinase assay in the presence of 4 pg of GST-Cdc42 preloaded with either GDP or GTPyS. The top panel shows an autoradiogram with bands corresponding to SPRK autophosphorylation indicated by an arrow. The lower panel shows SPRK expression. b, purified SPRK and PAK2 (1 and 2 pg, respectively) were incubated in an in vitra kinase assay in the presence of 4 pg of GST-Cdc42 preloaded with either GDP or GTPyS. Shown is an autoradiogram with bands corresponding to SPRK or PAK2 autophosphorylation. 81 Fig. 21. Phosphopeptide mapping of tryptic peptides derived from in viva phosphorylated SPRK. a, two-dimensional phosphopeptide mapping of 32P-labeled SPRK from 293 cells transfected with expression vectors for SPRK (top) or SPRK and Cdc42V12 (bottom). SPRK was immunopurified fi'om cellular lysates, blotted onto polyvinylidene difluoride membrane, and subjected to partial tryptic digestion. Equal amounts of radioactivity, as determined by Cerenkov counting of the resultant tryptic peptides , were analyzed by TLE in the first dimension and TLC in the second dimension. The direction of electrophoresis and chromatography are indicated by long arrows. Phosphopeptides were visualized by PhosphorImaging. The phosphopeptides of interest are alphabetically labeled. Shown is a map representative of three independent experiments. b, the percent radioactivity of the indicated phosphopeptides compared with phosphopeptide a, calculated as: [(volume- background)phosphopeptide]/[(volume-background)phosphopeptide a] x 100, using Image Quant software (Molecular Dynamics). 82 ' ' SPRK x a C b C + d w SPRK + T ctle42V12 a .5; b ehromato- . i- d graph)! G A 4 electrophoresis _—P b % Radioactivity of Phosphopeptide a * Peptides b c d x SPRK 14 13 7 56 3:32:12 0 71 5o 44 83 5. Discussion Small GTPases of the Ras superfamily have been shown to regulate protein kinases. PAK has emerged as the paradigm CRIB-containing serine/threonine kinase that is activated by GTP-bound Cdc42 and/or Rac. The PAKs play roles in diverse processes, including apoptosis, modulation of actin cytoskeleton, gene transcription, and cell cycle (Sells and Chernoff, 1997). SPRK is a member of the so-called mixed lineage kinases. Except for the presence of a loosely conserved CRIB motif, SPRK differs dramatically fiom the PAKs, both structurally and functionally. While the mammalian PAK], 2, and 3 share 95% sequence similarity in their catalytic domains, SPRK’s catalytic domain is just 20% similar to those of the mammalian PAKs. The CRIB motif of the PAKs is found NHz-terrninal to the catalytic domain, whereas SPRK’s CRIB motif is COOH-terminal to the catalytic domain. Flanking SPRK’s catalytic domain is an NHz-terminal SH3 domain and a COOH-terminal leucine zipper motif, both lacking in the PAKs. The only well-established function thus Er ascribed to SPRK is as an MKKK in the activation of the INK pathway. Because the MLK3 are so different fi'om the PAK3, it is important to determine whether the structural features of their binding to and the mechanisms of activation by Cdc42 and/or Rac also differ fi'om that of the PAKs. Whereas the three marmnalian PAK isoforms contain perfect consensus CRIB motifs, as defined by Burbelo et al. (Burbelo et al., 1995), SPRK’s CRIB motif contains only six of the eight consensus residues (Fig. 8). We show that mutations in 84 conserved residues of SPRK’s CRIB motif disrupt the ability of the Cdc42 to bind to and activate SPRK, indicating that SPRK does indeed contain a functional CRIB motif. WASP and ACK, two other proteins whose CRIB-dependent binding to Cdc42 has been well-established, also contain less than perfect CRIB motifs (Fig. 8), with WASP (Aspenstrom et al., 1996; Kolluri et al., 1996), and ACK (Manser et al., 1993) containing 7 of the 8 consensus residues, and 6 of the 8 consensus residues, respectively. It may well be that the CRIB consensus motif is biased towards PAK, due to the large number of PAK isoforms that have been identified. SPRK and the closely-related MLK2 lack the second of the two conserved histidine residues of the consensus CRIB motif (Fig. 8). We show here that mutation of the first conserved histidine in SPRK to an alanine residue (1-1500A) disrupts binding to Cdc42. The Ect that both SPRK (Burbelo et al., 1995) and MLK2 (Burbelo et al., 1995; Nagata et al., 1998) have been demonstrated to bind Cdc42 may indicate that the second of the two conserved histidines in the consensus CRIB motif in other CRIB-containing proteins is not required for binding to Cdc42. Further support for this notion is provided by the finding that the conserved Asp 38 in Cdc42 interacts primarily with the first of the two conserved histidine residues (11520) in ACK (Matt at al., 1999). In addition, mutation of the first of the two conserved histidine residues in N-WASP to aspartate (1-1208D) decreases the in vitra binding aflinity for Cdc42 and Rac, as well as the activity of N-WASP in viva and in vitro (Mild et al., 1998). The regions outside of the CRIB motifs of ACK and WASP 85 exhibit low sequence similarity, and, perhaps not surprisingly, low structural similarity when bound to Cdc42 (Abdul-Manan et al., 1999; Mott et al., 1999). It is likely that the GTPase binding domain of SPRK, with the exception of the CRIB motif, will difl‘er structurally from those of both WASP and ACK. Because of the proximity of SPRK’s zipper and CRIB motifs in linear sequence, and because sequences flanking the CRIB motif in other proteins contribute to Cdc42/Rae binding, we tested whether deletion of the COOH—terminal half of the zipper motif effects Cdc42 binding. The binding of SPRK‘W (Fig. 7), which contains an intact CRIB motif, to activated Cdc42 is reduced more than lO-fold, suggesting that, in addition to SPRK’s CRIB motif, the zipper domain or the following short basic region of SPRK may contribute to Cdc42 binding. Interestingly, the basal autophosphorylation activity of SPRKM" is about 70% that ofwild type SPRK. This may indicate some intrarnolecular autophosphorylation activity. Alternatively, SPRK”? may homooligomerize and undergo intermolecular autophosphorylation. Leung et al. (Leung and Lassam, 1998) recently reported a very large reduction in GST-SPRK/MLK3 autophosphorylation activity in vitra upon deleting the entire zipper region, but leaving the basic stretch intact. Recent site-directed mutagenesis studies indicate that the serine/threonine kinase PAK contains a basic stretch consisting of three contiguous lysine residues upstream of the CRIB motif, whose charge is important for binding to Rae] and Rac2, and whose presence is required for efficient PAK-1 activation by Rae], Rac2, 86 and Cdc42 (Knaus et al., 1998). SPRK contains four contiguous arginine residues between the zipper domain and the CRIB motif. These basic amino acids are deleted in the SPRK“ip variant, which exhibits greatly reduced binding to activated Cdc42. Thus, it is plausible that the arginine tract in SPRK may contribute to Cdc42 binding or activation of SPRK. We show that SPRK and activated Cdc42 can be coirnrnunoprecipitated fiom cellular lysates. However, under the conditions of our in vitra kinase assay, which clearly show a Cdc42-induced increase in SPRK autophosphorylation and histone phosphorylation, Cdc42 is not detected. Possibly once SPRK is activated by Cdc42, SPRK lms a reduced affinity for the GTPase as has been demonstrated with PAK2 (Manser et al., 1994). Furthermore Panayiotis Vacratsis demonstrated that the sites of in vitra autophosphorylation of SPRK expressed with and without Cdc42, and isolated from nmmmalian cells, are essentially identical as judged by mapping of tryptic phosphopeptides. The mechanism by which the highly conserved PAK Emily members (PAKl, 2, and 3) are activated by Rac and Cdc42 has been well studied. Increased autophosphorylation activity is observed upon incubation of purified activated Cdc42 with purified PAK In contrast to PAK, purified, catalytically active SPRK cannot be further activated in vitro by the addition of GTP-bound Cdc42. These data are consistent with a catalytic model in which Cdc42 activates SPRK in viva, but is not required to maintain SPRK in its activated state. We therefore decided to assess whether Cdc42 induces differential phosphorylation of SPRK in viva. 87 Two-dimensional tryptic phosphopeptide mapping studies of in viva labeled SPRK, expressed with or without constitutively active Cdc42, showed similar phosphopeptide maps, with major differences observed in a triangular cluster of phosphopeptides b, c, and d (Fig. 21a). In the map of SPRK alone, phosphopeptides b and c are present, but at low levels. When activated Cdc42 is coexpressed with SPRK, pho sphopeptide b is not detected and phosphopeptides c and d appear at high levels. Since the change in in viva phosphorylation of SPRK with Cdc42 correlates with increased SPRK activity, it is likely that phosphopeptides c and d contain activating phosphorylation sites. Phosphopeptides c and d may be distinct phosphopeptides. However, since their chromatographic mobilities are essentially identical, it is also possible that phosphopeptides c and d result from differential trypsin digestion. Phosphopeptides b and c lie on a diagonal which slopes toward the anode, characteristic of peptides that are phosphoisomers. Upon phosphorylation, the negative charge and polar nature of the added phosphate group reduces a peptide‘s mobility in both the electrophoretic and chromatographic dimensions (Boyle et al., 1991). Therefore, phosphopeptide c may differ from phosphopeptide b by the addition of a phosphate group(s). This is consistent with the observation that when activated Cdc42 is coexpressed with SPRK, phosphopeptide c emerges while phosphopeptide b disappears. Cdc42 may induce differential SPRK autophosphorylation in viva or, alternatively, another SPRK-activating kinase may be responsible for the in viva 88 change in SPRK phosphorylation. Because Cdc42 is geranylgeranylated (Maltese and Sheridan, 1990) and has been localized to cellular membranes (Backlund, 1993; Erickson et al., 1996; Hart et al., 1990) as well as to cytoskeletal elements (Dash et al., 1995; Hotta et al., 1996), it is possible that Cdc42 recruits SPRK to the vicinity of an activating kinase. SPRK and the serine/threonine kinase Raf both appear to function as MKKKs, activating the INK and ERK pathways, respectively. The idea that Cdc42 may recruit SPRK to an activating kinase is reminiscent of the proposed mechanism by which the small GTPase Ras activates Raf. Analogous to our findings with Cdc42 and SPRK, the addition of purified, activated Ras to Raf in vitra is not suficient for El] activation of Raf. However, appending a membrane targeting motif to the COOH-terminus of Raf causes Raf translocation and activation in the absence of activated Ras (Stokoe et al., 1994). It has been recently shown that PAK3 phosphorylates Raf in viva and in vitra leading to an increase in Raf activity (King et al., 1998), although it has yet to be determined whether this event requires Raf translocation In yeast, the PAK homolog STE20 functions as an MKKK in the activation of a yeast MAPK pathway. By analogy, potential SPRK-activating kinases may include PAK-related kinases (Sells and Chernoff, 1997) such as HPK-l. In coexpression studies HPK-l binds to SPRK's SH3 domain and phosphorylates SPRK, but the effect on SPRK activity is not reported (Kiefer et al., 1996). Interestingly, unlike PAK-1, -2, and -3, HPK-l lacks a CRIB motif. Our finding that catalytically active SPRK cannot be further activated in 89 vitra by GTP-bound Cdc42 suggests that the GTPase activates SPRK differently than the PAKs. Whereas the PAKs can be activated in vitra by interaction with unprenylated GTP-bound Cdc42, the activation of SPRK by Cdc42 appears to require prenylation of Cdc42, the cellular environment, or an as yet unidentified cellular component. Our data support a model in which the in viva CRIB—dependent interaction of SPRK and Cdc42 either allows SPRK to adopt a conformation that leads to autophosphorylation or recruits SPRK to the vicinity of a serine/threonine kinase that phosphorylates and activates SPRK Determination of the precise sites of Cdc42-induced phosphorylation of SPRK, coupled with subcellular localization studies, should shed finther light on the detailed mechanism of SPRK activation by Cdc42. ' The in vitro kinase assay was performed by Panayiotis Vacratsis 2 Expression and purification of SPRK was done by Brian Qamirani 3 Expression and purification of GST-Cdc42 was performed by Hua Zhang ‘ Two-dimensional in viva phosphopeptide mapping was performed by Panayiotis Vacratsis 90 IV. Cdc42 Changes the Subcellular Distribution of the Mixed-Lineage Kinase SPRK 1. Abstract SPRK/MLK3 is a serine/threonine kinase that upon overexpression activates the INK pathway. The small GTPase Cdc42 has been shown to associate with SPRK and to increase its catalytic activity. Coexpression of SPRK with activated Cdc42 changes the in viva phosphorylation of the protein kinase. However, the Cdc42- induced SPRK activation cannot be recapitulated in viira using recombinant, purified proteins. Taken together these results suggest that SPRK activation by Cdc42 requires a cellular context. In this study the effect of Cdc42 on the subcellular distribution of SPRK was investigated. Upon overexpression in COS-7 cells, SPRK is predominantly found at the Golgi apparatus. Coexpression of SPRK and Cdc42 in 293 cells induces a shift of SPRK protein to a plasma membrane-enriched fraction, suggesting tlmt Cdc42 may indeed change SPRK's subcellular distribution. The Cdc42-induced change in SPRK's localization requires prenylation, since a Cdc42 variant, which Eils to undergo prenylation cannot induce the observed shift. Preliminary comparative two- dirnensional phosphopeptide mapping suggests that subcellular targeting may be required for Cdc42-induced activation of SPRK. 91 2. Introduction Small GTPases act as molecular switches by cycling between two interconvertible conformational states, a GTP-bound active form and a GDP-bound inactive form. The activity of small GTPases is regulated by the ratio of GDP/GTP- bound states. Guanine nucleotide exchange factors (GEFs) promote the exchange of GDP and GTP, converting the GTPase to its activated state. GTPase activating proteins (GAPS) enhance GTP hydrolysis, thus inactivating the GTPase (Boguski and McCormick, 1993). GTPases regulate the activity of a variety of effector molecules, including protein kinases. A well-characterized target of the small Rho GTPases Rae and Cdc42 is the protein kinase PAK. PAK was originally identified in rat brain as a serine/threonine kinase that is activated by the small GTPases Rae and Cdc42 in a GTP-dependent manner (Manser et al., 1994). The structural requirements and the mechanism of PAK activation by these small GTPases have been vigorously investigated (Gatti et al., 1999; Knaus et al., 1998; Manser et al., 1997; Manser et al., 1994; Zenke et al., 1999; Zhao et al., 1998). The association of multiple PAK isoforms with Rae and Cdc42 requires the CRIB motif of the protein kinase (Burbelo et al., 1995). The CRIB motif is a 14-16 amino acid sequence containing eight conserved residues. The direct interaction between PAK and Cdc42/Rae increases the autophosphorylation activity of PAK, both in vitra and in viva. Small GTPases can also change the subcellular distribution of protein kinases. For example, the Ras-induced activation of the serine/threonine kinase Raf depends on membrane targeting by activated, Ernesylated Ras. In contrast to PAK activation 92 by Cdc42, Raf activation by Ras cannot be recapitulated in vitra (Stokoe et al., 1994). However, appending a membrane-targeting signal to Raf, which promotes membrane association independent of Ras, induces Raf activation, suggesting that not Ras binding per se, but rather membrane targeting, is the key-activating event (Leevers et al., 1994; Stokoe et al., 1994). By virtue of prenylation, small GTPases are able to associate with cellular membranes and cytoskeletal elements (Dash et al., 1995; Erickson et al., 1996; Hancock et al., 1991b; Hart et al., 1990; Hotta et al., 1996; Thissen et al., 1997). Thus they have the potential to change the subcellular distribution of target proteins. Prenylation is a posttranslational lipid modification, whereby a Ernesyl or geranylgeranyl isoprenoid is covalently attached to the COOH-terminus of proteins. Prenylation requires a so-called CAAX (cysteine-aliphatic-aliphatic-X) motif at the extreme COOH-terminus of the protein. The prenyl group is attached by a thioether linkage to the cysteine within the CAAX motif (Clarke, 1992). Prenylation is a three-step process, which takes place in different compartments of the cell. First the prenyl group is attached to the cysteine by soluble prenyltransferases. Depending on the amino acid at the X position within the CAAX motif, Ernesyltransferases or geranylgeranyltransferases catalyze this process. The second step is the removal of the AAX residues by a prenyl-CAAX protease. Finally the new COOH-terminus is methylated by a carboxyl methyltransferases (Casey and Seabra, 1996). Whereas the prenyltransferases are soluble (Casey and Seabra, 1996), the activities of the prenyl-CAAX proteases (Hancock et al., 1991a) and the prenylcysteine carboxyl methyltransferase (Pillinger 93 et al., 1994) are associated with endomembranes. Recent evidence from studies examining prenylation of various Ras isoforms supports the idea that the prenylated CAAX motif serves as a target signal for association with the endOplasmic reticulum, where the subsequent processing by proteolysis and methylation takes place (Choy et al., 1999). The authors propose that after prenylation, the small GTPases use cellular transport pathways to target to the plasma membrane. The small GTPase Cdc42 is posttranslationally modified by geranylgeranylation (Maltese and Sheridan, 1990), promoting membrane association. Indeed, Cdc42 has been localized to cellular membranes, including the Golgi apparatus (Erickson et al., 1996) and the plasma membrane (Hart et al., 1990), as well as to cytoskeletal elements (Dash et al., 1995; Hotta et al., 1996). SPRK contains a CRIB motif. The work described in this thesis and by Bock et al., demonstrates that this CRIB motif is required for association of SPRK with Cdc42 and for Cdc42-induced activation. Furthermore, the increase in activity is accompanied by a change in the in viva phosphorylation of SPRK as determined by two-dimensional phosphopeptide mapping, suggesting an activating phosphorylation event. However, the Cdc42-induced activation of SPRK cannot be recapitulated in vitra indicating the necessity for a cellular component in order to promote Cdc42- induced SPRK activation. One intriguing possibility is that Cdc42 may induce a change in localization of SPRK, for example by bringing SPRK into the vicinity of an activating kinase. To investigate this possibility, studies were undertaken to determine the effects of Cdc42 on SPRK subcellular localization and SPRK activity. 94 3. Materials and Methods 3.1 Construction of mammalian expression vectors and site-directed mutagenesis Construction of the cytomegalovirus-based vectors carrying the cDNAs for wild type SPRK (pRK5 -sprk), for the kinase-defective variant (pRKS-sprkxm") and the zipper mutant (pRKS-sprkw") has been described elsewhere (Beck et al., 2000; Gallo et al., 1994). Expression plasmids encoding NHz-terminal Flag epitope-tagged constitutively activated Cdc42 (pRK5-N-Flag.cdc42v'2) were kindly provided by Avi Ashkermzi (Genentech, Inc.). The Cdc42VIZ variant bearing an amino acid change of cysteine to serine in the CAAX motif, Cdc42v'2'C'8ss was generated by site-directed mutagenesis using the Quick Change Mutagenesis Kit (Stratagene). The mutagenesis primers used were S’-GATGT‘TCATAGCAGCACAGATCTGCGGCTCTTCTTCG-3’ and 5’-CGAA- GAAGAGCCGCAGATCTGTGCTGCTATGAACATC-3’. The expression vector pRK5-Nflag.cdc42V12 was used as a template for the mutagenesis reaction. The Cdc42 variant with a substitution of the polybasic amino acids in the COOH- terminus to glutamines was generated by using the Quick Change Mutagenesis Kit (Stratagene). To ensure that all four amino acids are changed to glutamine the mutation was performed in two steps. For the first step and second step the following mutagenesis primers were used, respectively: 5'-CCAGAACCGAAGAAGAGCC- AGAGGTCTGTGCTGCTATGAAC-3' and 5'-GTTCATAGCAGCACAGACCT- CTGGCTCTTCTTCGGTTCTGG-3'; 5'-GCCTCCAGAACCGCAGCAGAGCCA- GCAGTCTGTGCTATGAACGCT-3' and 5 '-GTTCATAGCAGCACAGACTGC- 95 TGGCTCTGCTGCGGT'I‘CTGGAGGC-3'. The expression vector pRK5- Nflag.ca’c42‘”2'c'388 was used as a template for the mutagenesis reaction. 3.2 Cell lines and transfections Human fetal kidney 293 cells were maintained in Ham’s F 12:10w glucose Dulbecco’s modified Eagle’s media (1:1) (Life Technologies, Inc.) supplemented with 8% fetal bovine serum (Life Technologies, Inc.), 2 mM glutamine, and penicillin/streptomycin (Life Technologies, Inc.). Plasmids (5 pg each for 60 mm dishes; 10 pg each for 100 mm dishes) were used to transfect 293 cells using the calcium phosphate technique [Goreman, 1990 #191]. Cell mono layers were incubated with the DNA precipitate for 4 h, then rinsed once with PBS (phosphate- buffered saline), cultured in the medium described above, and lmrvested for lysis after 18 h. COS-7 cells were maintained in high glucose Dulbecco’s modified Eagle’s media (Life Technologies, Inc.) supplemented with 10% fetal bovine serum (Life Technologies, Inc.), 2 mM glutamine, and penicillin/streptomycin (Life Technologies, Inc.). For transfection 7500 cells were plated on glass cover slips (1.5, 22x22 mm) and transfected as described above. Cells were fixed for microscopy 8 h post transfection. 3.3 Cell lysis, membrane fractionation, and immunoprecipitation The crude membrane fi'actionation into 8100 and P100 fiactions was performed as described previously (Stokoe et al., 1994). Briefly, transiently transfected 293 cells were rinsed twice with ice-cold PBS and lysed in 1 ml Buffer A (10 mM Tris 96 (pH 7.5), 35 mM NaF, 5 mM MgC12, 1 mM EGTA, 1 mM Na3VOa, 1 mM NaaPPi, 100 pM B—glycerophosphate, 2 mM PMSF and 0.15 U/ml aprotinin). After 10 min on ice, the cells were homogenized with 30 strokes in a Dounce homogenizer. Nuclei were pelleted by two centrifugation steps at 500 x g for 5 min at 4 °C. The supernatant was ultracentrifuged at 100,000 x g for 1 h at 4 °C. The sedimented P100 fi'action was rinsed twice with Buffer A and resuspended in Buffer A containing 1% Nonidet P-40 (NP-40), resulting in the appearance of a particulate solution. To obtain a plasma membrane-enriched fraction the experiment was carried out as described previously with some modifications (Thissen and Casey, 1993). The cells were lysed in Buffer A, Dounce homogenized, and the nuclei pelleted as described above. The supernatant was subjected to a centrifugation at 16,900 x g for 1 h at 4 °C to obtain a sedimented plasma membrane-emiched fraction (Pl6.9). The P169 fi'action was rinsed twice with Buffer A and resuspended in Buffer B (50 mM HEPES (pH 7.5), 150 mM NaCl, 1.5 mM MgC12, 2 mM EGTA, 1% Triton X-100, 10% glycerol, 10 mM NaF, 1 mM NaaPPi, 100 pM B- glycerophosphate, 1 mM Na3VOa, 2 mM PMSF, and 0.15 U/ml aprotinin). The supernatant ($16.9) was removed and subjected to an ultracentrifugation at 100,000 x g for 1 h at 4 °C. The sedimented fi‘action (PIOO) was rinsed twice with Buffer A and the pellet was resuspended in Buffer B. Rabbit polyclonal antiserum was raised against a peptide corresponding to the C-terminal eight amino acids of SPRK and was purified by Protein A-Sepharose chromatography. SPRK antiserum was prebound to protein A—agarose beads (0.25 pg/ p1 slurry) as previously described (Gallo et al., 1994). For immunoprecipitation 97 of SPRK 200-400 pl from a P16.9 fraction the resuspended P16.9 fiaction was incubated with 20 pl of antibody-bound Protein A-agarose (Life Technologies, Inc.) for 90 min at 4 °C. Immunoprecipitates were rinsed 3x with HNTG buffer (20 mM HEPES (pH 7.5), 150 mM NaCl, 0.1% Triton-X-100, 10% glycerol) containing 1 M LiCl, 3x with HNTG buffer and twice with kinases buffer (50 mM Tris-HCl (pH 7.5), 100 mM NaCl, 1 mM MnClz, 10 mM MgC12, 0.1 mM Na3V04) and then subjected to an in vitra immune complex kinase assay. Irnmunoprecipitation of SPRK fiom total lysates, subjected to in viira kinase assays is described elsewhere (Beck et al., 2000). For co-irnmunoprecipitation experiments and in vitra kinase assays of SPRK from total lysates, cells were lysed in 1 ml of Buffer B for 5 min on ice. The lysate was clarified by centrifugation for 20 min at 14,000 rpm in an Eppendorf centrifuge at 4 °C. The Flag epitope-tagged Cdc42 variants were immunoprecipitated fi'om the clarified lysate (300 pl) with the M2 monoclonal antibody (Kodak IBI) directed against the Flag epitope (0.45 pg/pl slurry). 3.4 Gel electrophoresis and Western blot analysis Lysates and irnmunoprecipitates of SPRK and Cdc42 were resolved by SDS- PAGE according to Laemmli (Laemmli, 1970). Proteins were transferred to nitrocellulose and irnmunoblotted using either SPRK antiserum (1 pg/ml) or the M2 Flag monoclonal antibody (9 pg/ml), followed by the appropriate horseradish peroxidase-conjugated secondary antibody (Life Technologies, Inc.). To determine the distribution of SPRK and Cdc42 in the different membrane fiactions equal 98 amounts of protein were resolved by SDS-PAGE as described above. The protein concentration of the various fractions was determined by Bradford analysis (BioRad). Western blots were developed by chemiluminescence, and densitometry (NIH Image) of unsaturated film was used to determine SPRK distribution in the non-nuclear fiactions. 3.5 In vitra kinase assays and in viva phosphopeptide mapping Kinase assays were performed as described previously (Bock et al., 2000), with the exception that 10pg of purified GST-MKK4 was used as substrate. For non- radioactive in vitra kinase assays the reaction was carried out in 20 pl of kinase buffer containing 50 pM ATP and 10 pg GST-MKK4 for 30 min at room temperature. The reaction was terminated by the addition of 2x SDS sample buffer containing 50 mM EDTA (pH 8.0). To determine SPRK kinase activity 10 pl of the reaction were separated by SDS-PAGE and transferred to nitrocellulose. The Western blots were probed with an anti-phospho-MKK4 (New England Biolabs) according to the manchturer’s protocol. To determine SPRK expression the same blot was reprobed with SPRK polyclonal antiserum. GST-MKK4 was expressed fiom the pGEX-vector in BL-21 E. cali and purified by glutathione Sepharose aflinity chromatography as described previously (V acratsis and Gallo, 2000). Two- dimensional in viva phosphopeptide mapping was performed as described previously (Beck et al., 2000) 99 3.6 Immunofluorescence and microscopy Transiently transfected COS-7 cells were fixed in 2% formaldehyde/PBS for 30 min at room temperature, rinsed 5x with PBS, and then permeabilized in PBS/0.2% Triton-X-100 for 10 min at 37 °C. After three rinses with PBS, the samples were blocked in PBS/5% BSA for 10 min at 37 °C. The cells were incubated with SPRK antiserum (1:500) in PBS/0.5%BSA for l h at 37 °C. After three rinses cells were incubated with SPRK antiserum (1:500) or the M2 monoclonal antibody (1:20) in PBS/0.5%BSA for 1 h at 37 °C. Prior to incubation with the secondary antibody cells were rinsed 3x with PBS. For single protein staining, the cells were incubated with Alexa 488 goat anti-rabbit IgG (1:250) (Molecular Probes) or with the FluoroLinltTM Cy3TM-labelled goat anti-mouse IgG (1:1000) (Amersham); for double staining a dilution of 1:500 was used of each antibody. All incubations at 37 °C were carried out in a humidified atmosphere. For Golgi apparatus staining, cells were fixed as described above and incubated with 5 pM BODIPY TR ceramide (Molecular Probes) in high glucose DMEM supplemented with 0.34 mg/ml BSA for 30 min at 4 °C. Cells were rinsed 5x with PBS containing 3.4 mg/ml BSA. For double staining of SPRK and the Golgi apparatus cells were incubated in 1:750 Alexa 488 goat anti-rabbit IgG. After incubation with the secondary antibody and 5 rinses with PBS cells were incubated with 5 pM BODIPY TR ceramide in high glucose DMEM supplemented with 0.34 mg/ml BSA for 30 min at 4 °C. Following the incubation with ceramide the cells were rinsed 5x with PBS containing 3.4 mg/ml BSA. To examine cells stained with one fluorophore a Zeiss 210 confocal laser scanning microscope was used. For the analysis of F luoroLinkTM Cy3m- 100 labelled and ceramide stained cells a helium neon laser with an excitation at 543 nm was used. Staining with Alexa 488 was examined with an argon laser at 488 nm. Doubly stained cells were examined with a Meridian INSIGHT microscope (Genomic Solution, Lansing Division) at 488 nm and 514 nm. Images included in this thesis are presented in color. 3.7 Protein markers To analyze the subcellular fi'actions various cellular marker proteins were employed. To determine the cytosolic fiaction a polyclonal rabbit antiserum against lactate dehydrogenase (LDH) from pig muscle (~ 37 kDa) was used at a dilution of 1:500. The LDH antiserum (anti-LDH) was kindly provided by Dr. John Wilson (Department of Biochemistry, Michigan State University). The presence of endoplasmic reticulum proteins was determined using a rabbit anti-ERp72 polyclonal antibody (StressGen Biotechnologies Corp.) in a dilution 1:1000 according to the manufacturer’s protocol. The presence of Golgi proteins was determined using the monoclonal antibody M3A5 (Sigma-Aldrich), which recognizes the coatamer protein B—COP, at a dilution of 1:50 according to the manufacturer’s protocol. To verify the plasma membrane-enriched fi'action (Pl6.9) biotinylation of membrane proteins using the EZ-Linkm Sulfa-NHS-LC-Biotin cross-linker (Pierce) was performed (Shetty et al., 1993). The cell monolayer, grown on a 100 mm dish, was rinsed twice with ice cold PBS and incubated in 1 ml of biotinylation buffer (120 mM NaCl, 30 mM NaHCO,, 5 mM KCl, (pH 8.5)), containing 0.1 mg/ml cross-linker for 30 min at 4 °C. Following biotinylation, cells were lysed in Buffer A and the membrane 101 fractionation was carried out as described above. Equal amounts of protein of the S169 and P16.9 fiaction were separated by SDS-PAGE and tested for biotinylation by Western blotting using horseradish peroxidase-conjugated strepavidin (Pierce) at a dilution 1:500. The percentage of the protein amount in each fraction compared to the total non-nuclear cellular protein in the single fractions was calculated as: [pg protein fiaction y x 100]/(pg P16.9 protein + pg P100 protein + pg S100 protein). y represents either the P16.9, P100, or either 8100. 4. Results 4.1 Analysis of cellular fractions Initial studies of SPRK's subcellular distribution used a one-step crude membrane fractionation method resulting in a soluble and membrane fraction. To analyze the localization of SPRK in more detail, an additional centrifugation step was introduced, resulting in a soluble 816.9 and a pelleted P16.9 fiaction. The 816.9 fi‘action was firrther separated, yielding 8100 and P100 fiactions. Fractions obtained using the two-step method were analyzed by Western blot analysis employing different marker proteins. Lactose dehydrogenase (LDH) has been demonstrated to be present in the cytosol (Vyakarnarn et al., 1998). As shown in Fig. 23a the soluble 8100 fiaction contains LDH, whereas both pellet fiactions lack this enzyme, indicating that they are not contaminated with cytosolic proteins. To determine which fi'action contains plasma membrane proteins, a biotinylation method was employed. Membrane proteins are translated at the rough 102 endoplasmic reticulum and posttranslationally modified in the endoplasmic reticulum and the Golgi system before they are inserted into the plasma membrane. Thus membrane proteins may have limited usefulness as marker proteins. Therefore intact cells were treated with a biotin-conjugated membrane impermeable crosslinking agent. Then a membrane fiactionation was performed to separate 816.9 and P16.9 fi'actions. After SDS-PAGE, the proteins were transferred to nitrocellulose and the biotinylated proteins were visualized by chemiluminescence using horseradish peroxidase-conjugated strepavidin. Over 95% of biotinylated proteins are found in the P16.9 fi'action (Fig. 23b). Additionally the transmembrane protein sodium potassium ATPase can be detected in the P16.9 fraction, but not in the $16.9 fraction (personal communication, Dr. Julia V. Busik), validating that the P16.9 fraction contains the majority of integral plasma membrane proteins. Additionally the P16.9 fi'action contains endoplasmic reticulum as judged by the presence of the endoplasmic reticulum protein (ERp)72,(Fig. 23a). ERp72 is a member of the protein disulfide isomerase family and serves as a molecular chaperone for newly translocated and glycosylated proteins. Currently antibodies against human proteins, which reside within the Golgi are not commercially available. Analysis of the obtained fi'actions with an antibody directed against murine or-mannosidase II, a glycosylation enzyme within the Golgi apparatus, was not successful since this antibody does not crossreact with the human counterpart. Therefore a monoclonal antibody against B-Cop was used. B-COP is a coatamer protein, which is found in the periphery of the Golgi system and on vesicles scattered throughout the cytoplasm. As shown in Fig. 23 a, B-COP is 103 predominantly found in the 8100 fraction. The P100 fi'action is reported to contain Golgi membranes (Thissen and Casey, 1993). It is likely that smaller Golgi- associated vesicles, which are distributed within the cytosol, are found in the S100 fiaction whereas large Golgi stacks may pool in the P100 fi'action. A more reliable method to determine the Golgi fraction may be provided by a galactosyl transferase activity assay. Galactosyl transferase is an internal Golgi protein making it a better marker for the Golgi complex. In addition to B-COP, ERp72 is detected in the $100 fiaction, indicating that the 8100 fiaction also contains endoplasmic reticulum. A summary of the analysis of the fractions by marker proteins is given in Table 1. 4.2 Activated Cdc42 targets SPRK to cellular membranes Transiently transfected 293 cells were used to test the effect of Cdc42 on SPRK's subcellular distribution. A crude biochemical membrane fi'actionation, employing one centrifirgation step at 100,000 x g was performed. Cellular lysates were separated into a soluble and a membrane fi'action. As shown in Fig. 24, overexpressed SPRK, in the absence of Cdc42, is distributed in both the soluble and the membrane fiaction but, upon coexpression with activated Cdc42, nearly all SPRK is found in the membrane fraction. From this biochemical data we conclude that activated Cdc42 indeed affects SPRK's subcellular distribution, causing an increased association of SPRK with cellular membranes. 104 4.3 Analysis of the subcellular distribution of SPRK by confocal microscopy The biochemical fractionation shows that SPRK, overexpressed in 293 cells, is distributed approximately equally between a soluble and a pelleted fraction. To visually determine SPRK's distribution, COS-7 cells overexpressing SPRK were analyzed by confocal microscopy. Recently it has been shown by confocal and electron microscopy that the endogenous dual leucine zipper-bearing kinase (DLK) associates with the cytoplasmic face of the Golgi apparatus in NIH 3T3 cells (Douziech et al., 1999). Since DLK is related to SPRK and shares structural similarity, SPRK may also localize to the Golgi apparatus. Because the Golgi system has a diflerent appearance depending on the cell type, a crude visualization of the Golgi apparatus, in COS-7 cells was performed using a ceramide analogue coupled to the fluorophore Texas Red (Pagano et al., 1991). Cerarnides are precursors for glycosphingolipids and sphingomyelin, which are synthesized in the Golgi apparatus and hence can be employed to visualize the Golgi complex (Pagano et al., 1991). The appearance of the Golgi apparatus in COS-7 cells is shown in Fig. 25a. Confocal microscopy using a polyclonal SPRK antiserum results in a perinuclear staining and a Golgi-like staining (Fig. 25b). Additionally, SPRK shows a difitlse distribution in the cytosol. Fig. 25c shows an overlay (yellow, right image) of a cell stained for the Golgi apparatus (red, left image) and overexpressed SPRK protein (green, middle image). The overlay suggests that SPRK indeed colocalizes with the Golgi apparatus. The perinuclear staining may be indicative of some SPRK association with the endoplasmic reticulum. 105 To analyze the effect of Cdc42V12 on SPRK distribution, COS-7 cells were transfected with SPRK and Cdc42V12 and analyzed by irnmunofluorescence using confocal microscopy. The Cdc42 distribution was determined using the M2 monoclonal antibody, which recognizes the Flag epitope, which is appended to the NHz-terminus of Cdc42v’2. When using the same transfection conditions as in the previous experiments, large vacuoles within the cells were observed. The cells appeared to lose structural integrity and thus were impossible to analyze by confocal microscopy. SPRK and Cdc42 have been shown to activate the INK pathway (Coso et al., 1995; Minden et al., 1995; Tibbles et al., 1996), which has been implicated in the induction of cellular death (Verheij et al., 1996; Xia et al., 1995; Zanke et al., 1996). Therefore it is possible that overexpression of the two proteins may be toxic to the cells and may promote the induction of cell death. This idea is also supported by the morphological changes observed in 293 cells cotransfected with SPRK and Cdc42V‘2. The 293 cells started rounding up 18 h post transfection and lifted fi'om the culture dish approximately 48 h post transfection. This phenomena was not seen when SPRK or Cdc42V12 were expressed alone. To determine the earliest time point post transfection at which SPRK and Cdc42V12 can be detected by Western blot analysis, a time course of the expression of both proteins was analyzed. SPRK expression was already apparent 6 h post transfection whereas Cdc42 expression was detected after 8 h (Fig. 26). Based on this result the 8 h time point was chosen for further experiments to hopefirlly avoid the gross morphological changes, which were observed when using longer transfection times. 106 SPRK, expressed alone, is predominantly found in the Golgi apparatus, with a diffuse distribution within the cytosol and some perinuclear localization (Fig. 25b). Upon coexpression with constitutively active Cdc42, the majority of SPRK appears at a Golgi-like structure and within the cytosol (Fig. 27a and b, H). The cytosolic distribution appears more punctuate compared to SPRK alone. This may be indicative of SPRK association with microtubules or other components of the cytoskeleton. However, a possible association of SPRK with cytoskeletal elements was not further investigated. A small amount of SPRK is also detected at the plasma membrane, when activated Cdc42 is coexpressed. Analysis of Cdc42Vlz distribution is very similar to that of SPRK with Cdc42 being found in wlmt appears to be the Golgi apparatus, the cytosol, and maybe the plasma membrane (27 and b, I). The overlay of SPRK and Cdc42V12 staining supports the idea that SPRK and activated Cdc42 colocalize within COS-7 cells upon overexpression (Fig. 27a and b, 111). Surprisingly, the small GTPase was also detected in the nucleus, independent of the presence of SPRK (Fig. 27a and b, I and Fig. 283 and b). To exclude the possibility that the nuclear staining results from non-specific binding of the M2 antibody or the secondary antibody, the subcellular distribution of NHz-terminal F lag-tagged SPRK was determined using the M2 monoclonal antibody and the SPRK antibody in parallel. No nuclear staining was detected when examining SPRK distribution. To rule out that nuclear staining results fi'om the used concentrations of antibody, lower concentrations of primary and secondary antibody were tested, again resulting in nuclear staining. These data my suggest that under these experimental conditions Cdc42 may be nuclear or may be associated with the nuclear envelope. 107 Due to geranylgeranylation, Cdc42 is able to associate with cellular membranes (Erickson et al., 1996; Hart et al., 1990). Since the association of activated Cdc42 with the plasma membrane depends upon its prenylation, the translocation of SPRK by activated Cdc42 might also depend on this posttranslational modification. To investigate whether the prenylation of Cdc42 is required for Cdc42-induced membrane targeting of SPRK a prenylation-defective variant of Cdc42 was constructed. To prevent prenylation the cysteine residue within the CAAX motif of Cdc42V12 was changed to a serine, resulting in the CAAX mutant Cdc42v12cms (Fig. 22). The analogous mutation in yeast prevents prenylation and rescues the lethal phenotype induced by constitutively active Cdc42V12 (Ziman et al., 1991). To compare the subcellular distribution of Cdc42v'2 and Cdc42Vlzmss when overexpressed in COS-7 cells, confocal microscopy was performed. The observed plasma membrane staining with overexpressed Cdc42V12 (Fig. 28a and b) is largely blunted with the Cdc42 CAAX variant (Fig. 28c and d), supporting the idea that prenylation of Cdc42 contributes to membrane association of this small GTPase. However, despite the lack of the prenyl group residual, Cdc42vncrsss is detected at the plasma membrane. SPRK alone is found predominately at the Golgi system and difiuse in the cytosol (Fig. 25b). In contrast, overexpression of SPRK with activated Cdc42 results mainly in a Golgi-like staining and a punctuate scattered distribution in the cytosol, as well as some plasma membrane staining (27a and b, 11). When analyzing cells, which coexpress SPRK and the Cdc42vrzcms mutant, SPRK distribution appears very similar to the one seen with SPRK alone (Fig 29a and b; 11), with localization of 108 SPRK to a Golgi-like structure. Interestingly, Cdc42v'2'C1888 is also found at a Golgi-like structure (Fig. 29a and b, I) where it colocalizes with SPRK (Fig. 29 a and b, 11). Taken together the data obtained by confocal microscopy suggest that activated and prenylated Cdc42‘“2 changes the subcellular distribution of SPRK from the Golgi system to a more punctuate distribution within the cytosol. This change in distribution requires activated prenylated Cdc42. However, since the morphology of cells coexpressing SPRK and Cdc42VIZ differs substantially from the phenotype observed with SPRK alone, or SPRK coexpressed with the Cdc42 CAAX variant. it is difficult to draw any hard conclusions. To further examine the effect on Cdc42 on SPRK subcellular distribution, an additional modified biochemical fiactionation was undertaken. 4.4 Analysis of SPRK distribution by biochemical fractionation To complement confocal microscopy, the distribution of SPRK was further examined by biochemical fractionation experiments. To obtain a plasma membrane- enriched fiaction the one-step fractionation method was modified by introducing an initial centrifirgation step at 16,900 x g, according to Thissen and Casey (Thissen and Casey, 1993). This additional centrifugation yields a plasma membrane-enriched fraction (Pl6.9). The soluble 816.9 fiaction was further separated into a soluble (S100) and a pelleted (PlOO) fiaction by a second centrifirgation step at 100,000 x g. It is estimated, based on four different fiactionation experiments, that approximately 109 25% of total non-nuclear cellular protein is found within the P16.9 fi'action, 8% within the P100 fraction, and 67% within the $100 fraction (Table 2). Analysis of the subcellular distribution of SPRK, when equal amounts of protein are subjected to SDS-PAGE, showed that SPRK alone is distributed nearly equally between the 816.9 and the P16.9 fiactions. Upon coexpression with Cdc42“2 nearly all SPRK protein is found in the pelleted (Pl6.9) fiaction (Fig. 303, top left panel), supporting the idea that activated Cdc42 may target SPRK to the plasma membrane. SPRK coexpressed with Cdc42v'2c188$ shows a similar distribution to that of overexpressed SPRK alone, supporting the idea that Cdc42-induced SPRK membrane targeting requires prenylation of Cdc42. Interestingly, when examining the subcellular distribution of Cdc42“20888 it becomes clear that this mutant associates to some extent with the plasma membrane-enriched fi'action, despite the lack of the prenyl group (Fig. 30a, lower panel). Therefore, this mutant does not yield a clean phenotype and thus, may target some SPRK protein to the plasma membrane. Hancock and coworkers showed tint, in addition to the CAAX motif, a so- called polylysine stretch upstream of the CAAX-motif contributes to membrane association of K-Ras (Hancock et al., 1990). Cdc42 also contains forn' basic amino acids (two lysines and two arginines) near its CAAX motif (Fig.22), which may contribute to membrane targeting. In yeast Cdc42“2 changing the basic residues near the CAAX motif to glutamines results in a decrease in membrane association (Davis et al., 1998). To test whether these four positively charged residues are 110 important for membrane association of human Cdc42 all four basic residues were mutated to neutral ones in the Cdc42 CAAX mutant, resulting in a CAAX variant, termed Cdc42v'2'cms'wR40 (Fig. 22). When tested in membrane fractionation experiments, this Cdc42 variant shows no detectable association with the plasma membrane-enriched P16.9 fraction (Fig. 30b, lower left panel), indicating that the polybasic stretch in Cdc42, along with the CAAX-motif, participates in membrane association of this small GTPase. It appears that both Cdc42 CAAX variants do not influence the subcellular distribution of SPRK, when compared to SPRK expressed alone, and are devoid of inducing the shift of SPRK to the P16.9 fiaction. This result suggests that activated, prenylated Cdc42 is required for the observed change in SPRK's subcellular distribution. The distribution of overexpressed SPRK between the S169 and P16.9 fiaction is nearly equal when equivalent amounts of protein are analyzed (Fig. 30a and b, top left panel). However, estimation of the fractional distribution of non- nuclear SPRK reveals that approximately 27% of SPRK protein is found in the P16.9 fraction and 63% in the $16.9 fraction (Table 3). This distribution is dramatically changed to 70% of SPRK protein in the P16.9 fiaetion when SPRK is coexpressed with activated Cdc42. Coexpression of constitutively active Cdc42 does not influence SPRK distribution in the P100 fraction, but in the 8100 fraction SPRK protein is reduced fiom 62% to 18% as when compared to SPRK expressed alone (Table 3). We conclude therefore that the Cdc42-induced redistribution of SPRK is due to a shift of SPRK fi'om the 8100 to the P16.9 fraction, supporting the idea of the Cdc42-mediated translocation of SPRK from the Golgi apparatus to the plasma 1]] membrane. The percentage of SPRK protein found in the P100 fraction is not influenced by Cdc42 (Table 3). The small GTPase Rac also contains six contiguous basic amino acids NH;- terminal to the CAAX motif. Changing these residues to neutral glutamine residues results in the inability of Rac to bind PAK (Knaus et al., 1998). To rule out the possibility that loss of membrane localization of SPRK is due to its inability to bind to the Cdc42 CAAX variants, a co-irnmunoprecipitation experiment was performed. The result of this experiment clearly shows that all Cdc42V12 variants associate with SPRK to the same extent (Fig. 31, upper panel). A portion of SPRK, upon overexpression, is found in the plasma membrane- enriched fraction (Fig. 30a and b, top left panel). Overexpressed SPRK has high autophosphorylation activity (Gallo et al., 1994). Therefore, it is conceivable that this autophosphorylation activity is responsible for the presence of SPRK in the plasma-membrane fraction. However, the subcellular distribution of SPRKKI’M, a SPRK variant which lacks catalytic activity (Gallo et al., 1994), in presence or absence of Cdc42v'2 revealed similar results as observed with wild type SPRK (Fig. 323) suggesting that membrane association is not dependent on SPRK's catalytic activity. SPRK contains a polyarginine tract of four contiguous arginines following the zipper region. To test whether this polybasic stretch may mediate Cdc42- independent plasma membrane association of SPRK the subcellular distribution of the SPRK” variant was examined. This spinezip variant lacks the polybasic tract as well as the second half of the zipper domain. SPRKAZ'I’ shows the same 112 distribution as wild type SPRK (Fig. 32b), suggesting that the polybasic stretch is not required for Cdc42-independent membrane association of SPRK. Because all of these experiments were performed with overexpressed SPRK the observed subcellular distribution of SPRK may not reflect the distribution of endogenous SPRK. To exclude the possibility that these results are influenced by overexpression, the subcellular distribution of endogenous SPRK in the presence and absence of activated Cdc42 was analyzed in 293 cells. A preliminary experiment suggests that endogenous SPRK in the presence and absence of activated Cdc42 shows no striking difference in subcellular distribution compared to overexpressed SPRK (Fig. 33). 4.5 Effect of membrane-targeting on SPRK's kinase activity Activated Cdc42 increases the catalytic activity of SPRK and changes its in viva phosphorylation. The Cdc42-induced activation cannot be recapitulated in vitra (Bock et al., 2000), suggesting the requirement of a cellular component. Since the results presented above show that Cdc42V12 is capable of localizing SPRK to a plasma membrane-enriched fiaction, we wished to determine whether this membrane targeting contributes to SPRK's activation. SPRK, when coexpressed with Cdc42vn, has a slower electrophoretic mobility compared to SPRK alone or coexpressed with Cdc42v'2‘C1838 (Fig. 34). Increased phosphorylation is often correlated with a decrease in electrophoretic mobility. Thus, this mobility shift may suggest a change in phosphorylation of SPRK that depends on prenylated activated Cdc42. 113 To test the hypothesis that membrane targeting by Cdc42 may be accompanied by a change in in viva phosphorylation of SPRK two-dimensional comparative in viva phosphopeptide mapping was performed'. Fig. 35 shows the in viva phosphopeptide maps of SPRK expressed alone, or coexpressed with Cdc42V12 or Cdc42V'2C'888. The basic pattern of the three phophopeptide maps is very similar. The major changes are detected in the triangular cluster 0, b, and c. When SPRK is expressed alone phosphopeptide a and b are detected in the triangular cluster. In the 2"”, phosphopeptide b is corresponding map of SPRK, when coexpressed with Cdc4 not detected, whereas phosphopetides a and c are present at high levels. Analysis of the preliminary phosphopeptide map of SPRK upon coexpression with Cdc42V'2' Cms shows that phosphopeptide b is detected, but also a and c are highly abundant, perhaps suggesting an intermediate phosphorylation phenotype. This may suggest that membrane targeting is not required for the Cdc42-induced change in SPRK phosphorylation. Alternatively, it is possible that this intermediate phenotype is observed due to the fact that Cdc42v'2'c'8ss is detected within the plasma membrane- enriched biochemical fiaction, and thus may target residual SPRK to the plasma membrane, where in viva phosphorylation may take place. To more rigorously test if membrane targeting of SPRK changes the in viva phosphorylation pattern of SPRK this experiment should be performed employing the Cdc42‘mc'889"”1W2 variant, which is devoid of membrane association. To determine whether Cdc42-induced SPRK activation is dependent on membrane targeting, total lysates of 293 cells were examined for their SPRK kinase activity upon coexpression with Cdc42V12 or Cdc42v'zc'sss. Surprisingly, no 114 difference in kinase activity could be observed (Fig. 36) suggesting that Cdc42- induced activation of SPRK under these experimental conditions may be independent of membrane targeting. As shown in Fig. 30a the Cdc42 CAAX mutant retains the ability to associate to some extent with the plasma membrane. This partial membrane association is abolished in the c:dc42V"’-w”s"‘IR4Q variant (Fig. 30b). Therefore the kinase activity of SPRK when coexpressed with Cdc42V12'C'885'm’Q was measured. Upon coexpression of the Cdc42w2cms'wmQ variant with SPRK no detectable difference in SPRK activity compared with Cdc42V12 in total cellular lysates was found (Fig. 37). To determine the kinase activity of membrane-associated SPRK, in vitra kinase assays of SPRK in the P16.9 fiaction were performed. SPRK and either Cdc42"12 or Cdc42V'2'Cm‘S'K’R40 was coexpressed in 293 cells. Following a membrane fi'actionation, SPRK was immunoprecipitated from the P16.9 fiaction using SPRK antiserum and the immunoprecipitates were subjected to an in vitra kinase assay. The kinase assay employed in previous experiments using radiolabeled [y-32P]ATP and histones as substrates were accompanied by high background radioactivity. Therefore a non-radioactive kinase assay was developed using ATP and GST-MKK4 as substrates. Activation of MKK4 requires phosphorylation on serine 254 and threonine 258 (Yan et al., 1994; Zheng and Guan, 1994). The activating phosphorylation of MKK4 by SPRK was detected using an antibody, which recognizes the phosphorylated form of threonine 258 of MKK4. Using this method the background was dramatically reduced and SPRK substrate phosphorylation activity could be determined (Fig. 38). In membrane fiactions SPRK coexpressed 115 with Cdc42V12 has a 3-fold higher kinase activity compared to SPRK alone, or SPRK coexpressed with Cchvrzcrsssx/mo. The data shown in Fig. 38 is a representative of three independent experiments. A preliminary experiment examining endogenous SPRK in the P16.9 fiaetion for kinase activity in presence or absence or Cdc42V12 also shows an increase in SPRK kinase activity when activated, prenylated Cdc42 is expressed (Fig. 39). As shown earlier, even without activated Cdc42, SPRK can be detected in the immunoprecipitates of the P16.9, but as determined by in vitra kinase assays this portion of SPRK is not active. 116 P16.9 P100 8100 LDH - - + ERp72 + - + B-COP - - + Biotinylation + - - 117 Table 1. Analysis of subcellular fractions by marker proteins and biotinylation Cdc42": PPEKKSRRCVLL Cdc42v‘2‘c’m PPEKKSRRSVLL ctlt:42‘""“""’°""m PPEQQSQQSVLL Fig. 22. Alignment of various Cdc42 variants. The last 12 amino acid residues of the COOH-terminus of the Cdc42 variants, which were used in this study, are shown. The CAAX motifofczdc42V12 is in bold letters; amino acids, which were changed by site-directed mutagenesis, are shown in bold and italicized letters. 118 816.9 P16.9 100 P16.9 S P . LDH can B-COP Fig. 23. Analysis of subcellular fractions. a, the P16.9, $100, and P100 fiactions were analysed using lactose dehydrogenase, ERp72, and B-COP as rmrker proteins. Each lane contains equal amounts of protein. b, to determine the plasma membrane- containing fi'action, the plasma membrane-bound proteins were cross-linked using a biotinylated plasma membrane-impermeable crosslinker as described in "Materials and Methods". After a membrane fi'actionation the proteins were separated by SDS- PAGE and the biotinylated proteins were detected using horseradish peroxidase- coupled strepavidin. Both lanes contain 0.3 pg of total protein. 119 SPRK: - + «l- Cde42‘m: - - + S a S A U y S M Immunoblot . SPRK “A Fug4ana2 Fig. 24. Subcellular distribution of SPRK. Cells were transfected with expression vectors containing the cDNA indicated above the figure. The minus sign indicates the transfection of a control vector. S indicates the soluble fiaction and M the membrane fiaction after a centrifugation at 100,000 x g. Transient transfections of 293 cells, membrane fractionation, and SDS-PAGE were performed as described in "Materials and Methods". The upper panel shows SPRK distribution, the lower one shows distribution of Cdc42v'2. Each lane contains equal amounts of protein. The membrane fractionation shown is representative of five independent experiments. 120 Golgi SPRK Golgi SPRK Overlay Fig. 25. Subcellular localization of SPRK. COS-7 cells were plated onto glass coverslips and transfected with an expression vector containing the SPRK cDNA. SPRK subcellular distribution was determined by confocal irnmunofluorescence. a, staining of the Golgi apparatus in COS-7 using a BODIPY-ceramide coupled to Texas Red. b, localization of SPRK using an anti-SPRK antiserum as primary and Alexa 488 goat anti-rabbit IgG as secondary antibody. c, cells were treated as described under b and stained for both the Golgi apparatus (red, left panel) and SPRK (green, middle panel). The right panel shows costaining of SPRK and the Golgi apparatus (yellow). 12] hours: 5 6 s 10 12 imam”, . ‘7‘} f ,e . FIag-Cde42 Fig. 26. Time course of SPRK and Cdc42Vlz expression. COS-7 cells were transfected with expression vectors containing the cDNAs for SPRK and Cdc42V”. Cells were harvested post transfection at the time points indicated above. The top panel shows SPRK expression, the lower one expression of Cdc42V'2. 122 Flag-Cdc42": SPRK Overlay Fig. 27. Subcellular distribution of coexpressed SPRK and Cdc42vu. COS-7 cells were plated onto glass coverslips, and transfected with expression vectors encoding for SPRK or Cdc42V”. SPRK and Cdc42V12 subcellular distribution was determined by confocal irnmunofluorescence. Both the a and b panels show cells which coexpress SPRK and Cdc42V'2. (I), staining of Cdc42 (red) was performed using the M2 monoclonal antibody as primary and the FluoroLinkTM Cy3TM-labelled goat anti-mouse IgG as secondary antibody. (11) staining of SPRK protein (green) was performed using an anti-SPRK antiserum as primary and the Alexa 488 goat anti-rabbit IgG as secondary antibody. Arrowheads indicate areas where plasma membrane staining is observed. (111), colocalization of SPRK and Cdc42V12 (yellow). 123 Fig. 28. Subcellular distribution of Cdc42Vlz and Cdc42V‘m“. cos-7 cells were plated onto glass coverslips, and transfected with expression vectors encoding for Cdc42V12 (a and b) or Cdc42v'2'cms (c and d). The subcellular distribution of the two Cdc42 variants was determined by immunofluorescence using the M2 monoclonal antibody as primary and the FluoroLinkTM Cy3TM-labelled goat anti- mouse IgG as secondary antibody. Arrowheads indicate areas of plasma membrane staining. 124 Flag-Cdc42"“"“ SPRK Overlay Fig. 29. Subcellular localization of coexpressed SPRK and Cdc42v'2‘cms. COS- 7 cells were plated onto glass coverslips, and transfected with expression vectors encoding for SPRK and Cdc42v'2‘c‘m. SPRK and Cde42‘”w“s subcellular distribution was determined by confocal immunofluorescence as described above. The a and b panels show cells which coexpress SPRK and Cdc42V'2‘ms. (I), staining of Cdc42v'2'c‘m (red); (11) staining of SPRK (green); (111), costaining of SPRK and Cdc42V'2‘3ms (yellow). 125 - Ib‘“ - . Flag-Cdc42 16,900 x 9 100,000 x g b SPRK: + + + + + + CIEIZ v11 - + 01W - + crass-mo 8 P 8 P 8 P 8 P 8 P 8 P Inlnunoblot ‘ SPRK . e- r... M -Q a...“ ' ._ it ~ Plug-06042 16.900 x a 100,000 rt 9 Fig. 30. SPRK's subcellular distribution upon coexpression with Cdc42v" variants. 293 cells were transfected with expression vectors containing the cDNA indicated above each figure. The membrane fiactionation was carried out as described in "Materials and Methods". S indicates the soluble fiaction and P, the pellet fiaction. The left hand panels contain soluble and pellet fiactions obtained flour a centrifugation at 16,900 x g, the right hand panels contain soluble and pellet fractions of a centrifugation at 100,000 x g. a, subcellular distribution of SPRK when expressed alone or with either Cdc42V‘2, or Cdc42v'2'c'883. b, subcellular distribution of SPRK when coexpressed with either Cdc42VIZ or Cdc42V‘M'835’WQ. The reduced electrophoretic mobility of Cdc42V‘ZC'883'WQ may be due to the substitution of positively charged residues to neutral residues. Equal amounts of protein were loaded in corresponding experiments. 126 Fraction P16.9 P100 S100 % Protein 25 8 67 Table 2. Estimated distribution of total cellular non-nuclear protein among biochemical fractions. The percentage of total cellular non-nuclear proteins found in a given fiaction y was calculated as: [pg protein in fiaction y x 100]/(pg protein in P16.9 fiaction + pg of protein in P100 fiaction + pg of protein in $100 fiaetion). y represents either P16.9, P100, or 8100. 127 SPRK SPRK (- Cdc42‘m) (+ Cdc42v”) P16.9 27% 70% 816.9 (%P100 + %S]00) 63% 30% P100 1 1% 12% $100 62% 18% Table 3. Estimated distribution of SPRK protein among biochemical fractions in presence and absence of Cdc42v". The distribution of SPRK among the non- nuclear biochemical fiactions was determined by densitometry (NIH Image) as described in “Materials and Methods”, and corrected for the fiactional distribution of total non-nuclear protein. 128 SPRK: - + + + + m crass Cdc42 = - - + crass KIR4Q - . ‘ ‘ immunoblot ”'9", . - . SPRK _-._..._.._._ . ’54; one W .O. Fla-scam Fig. 31. Co-immunoprecipitation of SPRK and Cdc42 variants. Cdc42v‘2, Cdc42V'2‘C'sss, or Cdc42 V‘“'”‘~‘*“"“Q was immunoprecipitated (IP) with the M2 antibody directed against the Flag epitope. The co-irnmunoprecipitation of SPRK by the Cdc42Vlz variants was determined by immunoblotting with a SPRK antibody (top panel). The middle panel and lower panel show Cdc42 expression, respectively. 129 SPRK'm“: + + Cdc42m: _ + S P madam realm .~ . . .‘ .’c,_', i _ i‘ FSPRKW q Fig 32. Subcelluhr distribution of sprud‘mA and SPRKA". 293 cells were transiently transfected with expression plasmids encoding a, SPRKK'W‘ and Cdc42"12 or b, SPRK“. Cellular lysates were subjected to membrane fractionation. The distribution of the SPRK variants was determined by Western blot analysis. P indicates the pelleted 16.9 plasma membrane-emiched fraction and S the soluble $16.9 fiaction. Each lane contains equal amounts of protein. 130 “7'. .. ,1 . It“ '_ -""M~"-~..fi‘ SIP 100,000 x 9 Fig. 33. Subcellular distribution of endogenous SPRK. 293 cells were transfected with a control vector or an expression plasmid containing the Cdc42“2 cDNA. The cellular lysates were subjected to membrane fractionation as described in "Materials and Methods" and the SPRK distribution was determined by Western blot analysis. S indicates the soluble fiaction, P the pellet fiaction of the indicated centrifugation speed. Each lane contains equal amounts of protein. 131 SPRK: _ + + + + 01888 V12. - - Cdc42 . __ .. . lmmunoblot SPRK Fig. 34. Electrophoretic mobility shift of SPRK. 293 cells were transiently transfected with expression plasmids encoding the cDNAs indicated above. Cells were lysed and the proteins were analyzed by Western blot analysis. 132 SPRK SPRK + SPRK + Cduzv12ec1m Cdc42V12 . g ”h, . are. a , W, ‘ E b I b b ehromato- .‘ ‘M . graphy a c i ‘, C a“ c electrophoresis —> Fig. 35. Phosphopeptide mapping of in viva phosphorylated SPRK. Two- dirnensional phosphopeptide mapping of 32P-labeled SPRK alone or coexpressed with either Cdc42Vl2 or Cdc42V'2‘mS. cellular lysates, proteins were separated by SDS-PAGE and blotted on polyvinylidene difluoridemembrane, and subjected to a partial tryptic digest. The resultant tryptic peptides were analyzed by TLE in the first and by TLC in the second dimension. The elecreophoretic and the chromatographic directions are indicated by SPRK was immunoprecipitated fi-om arrows. The peptides of interest are indicated by italicized letters. 133 SPRK: - + + + Cdc42”: - - + 01888 autoradiogram *.., épg,.g;‘-giiig;i.jiig_,