a.” firm ‘ xufl my? J . .3. 3.1. A.“ A... ‘ 11.1.: I: ‘0 '1 I I}; .1 Lc'. NO! 3 . . a imam.» U 1. n1. $.35 é $25.4... V Ex. Lab“ V‘ .9, JIII- 5’ J .1. :b.‘nu... .715: r . ..7 . l; .: 9.2.3.3.. :5 - A {Nam . ma. ‘ y , EaJmpzwif “33%? ‘L. I' $1 9398 17/3 ('5‘? 7 LIBRARY Michigan State University This is to certify that the dissertation entitled ANALYSIS OF 17p11.2 DELETIONS AND CANDIDATE GENE CHARACTERIZATION IN SMITH-MAGENIS SYNDROME presented by CHRISTOPHER NICHOLAS VLANGOS has been accepted towards fulfillment of the requirements for the PhD. degree in Genetics A V Major Professor's Signature 5/1 1/2005 Date MSU is an Affinnative Action/Equal Opportunity Institution .- ._.— _—-—.—._.—.—--—---n-o—o—u—o-n-I-.-.-.-o-O-I-o-c-o-o-O-o-o-0-o-n-o-I-o-u-n-3-.-o—o-I—D-a-o-o--o-o-c-o-o--.-.-o-o-o-o-o- 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 2/05 cz/cFaCIDateouejnaa-pAs ANALYSIS OF 17p11.2 DELETIONS AND CANDIDATE GENE CHARACTERIZATION IN SMITH-MAGENIS SYNDROME By Christopher Nicholas Vlangos A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Genetics Program 2005 ABSTRACT ANALYSIS OF 17pll.2 DELETIONS AND CANDIDATE GENE CHARACTERIZATON IN SMITH-MAGENIS SYNDROME By Christopher Nicholas Vlangos Smith-Magenis syndrome (SMS) is a multiple congenital anomalies and mental retardation disorder usually associated with an interstitial deletion of chromosome 17p11.2. The SMS phenotype includes craniofacial/skeletal anomalies, otolaryngeal abnormalities, and neurological anomalies. The incidence of SMS is estimated at 1:25,000 births. Unfortunately, the syndrome is not widely known and under-diagnosis is likely. In order to identify the genes responsible for SMS, we constructed a physical and transcription map of the region. Using the small number of markers known to map to SMS critical region (SMCR), we identified large insert DNA clones mapping to 17p11.2. We established the location of the known markers within these clones by Southern analysis and created a genomic contig across the ~1.5 Mb SMCR. The order and orientation of 29 potential candidate genes was also determined within the SMCR. The candidate genes were initially characterized by bioinformatic analysis in order to determine any known or predicted protein motifs in these sequences. The developmentally regulated GTP-binding protein 2 (DRGZ) gene was chosen for further investigation by immunohistochemislry on sectioned mouse embryos where expression was found localized within the developing nervous system. DRG2 was localized to the endoplasmic reticulum and the Goligi apparatus with a GFP fusion product. Drg2 may play a role in embryonic neuronal development by regulating protein trafficking. Concurrent to construction of the physical map and gene characterization we used fluorescent in situ hybridization to characterize the deletion sizes carried by a cohort of 22 SMS patients in order to decrease the size of the SMS critical region. Through extensive FISH analysis of these SMS patient samples, we reduced the smallest region of overlap involving 17p11.2 deletion associated with SMS from 1.5 Mb to ~7OO kb. This refinement reduced the number of candidate genes from >40 to 11. Within our cohort of 22 patients there was a group of 7 patients in whom a 17p11.2 deletion could not be detected with FISH analysis. We hypothesized that these patients might carry mutations in gene(s) mapping to the SMCR. Using systematic direct sequencing of candidate genes, we identified 4 SMS patients with dominant frame shift mutations in the retinoic acid induced l (RAII) gene. Haploinsufficiency of the RAII gene is the cause of the physical, neurological, and behavioral characteristics of Smith-Magenis syndrome. ACKNOWLEDGEMENTS The work presented in this dissertation would not have been possible without the assistance and support from many people. I cannot begin to describe how lucky I was to perform this work uner the guidance of Dr. Sarah Elsea. Sarah provided an amazing atmosphere for learning. The dedication and support she provides to her students in unparralled. The knowledge and skills that I have learned from Sarah are priceless and will serve me well in the future. My guidance committee included Dr. Karen Friderici, Dr. Kathy Meek, Dr. Helga Torriello, Dr. VilmaYuzbasian-Gurkan, and Dr. Michael Grotewiel. Their assistance and time is greatly appreciated. My co-workers and friends in the Elsea Laboratory were also key in the success of this endeavor and include fellow graduate students Rebecca Slager and Santhosh Girirajan, research technicians Dennis Lettau and Barbara Szomju, and undergraduate students Tanisha Jain and Suzanne Shunn. A special thank you goes out to the Genetics Program at MSU, especially to Jeannine Lee. Without the help of Jeannie no Genetics student would be successful in their graduate career. Additionally, I would like to thank those in the Departments of Pediatrics and Human Genetics at Virginia Commonwealth University. In my brief stay at VCU as a visiting student they made me feel welcome and treated me as one of their own. Amazing support also came from my family and friends. Without constant encouragement fi'om my parents Peter Vlangos and Sue Smith, sister Georgette iv Vlangos, and grandparents Nick and Carol Smith I would not have come this far. I also have to acknowledge Jim and Sue Vogel for feeding me dinner for months during the more lean times in my graduate career. It is during the course of my graduate career that my friend Scott Asakevich introduced me to Jim and Sue’s daughter Michelle Vogel. Michelle is an amazing and loving woman who provided encouragement and love. As we begin our lives together as husband and wife I know I can always count on Michelle. TABLE OF CONTENTS LIST OF TABLES - . -- ix LIST OF FIGURES _ -- - -- - - - -_ -- - ...... .1 LIST OF ABBREVIATIONS - -- _ - ..... xii CHAPTER 1. INTRODUCTION - - - - - - - - - -- 1 The SMS phenotype ....................................................................................................... 2 l7p11.2 deletions associated with SMS ......................................................................... 9 Microdeletion syndromes: from critical region to gene identification ........................ 12 Animal modeling of Smith-Magenis syndrome ............. 16 Summary ...................................................................................................................... 18 CHAPTER 2. PHYSICAL MAPPING OF THE SMITH-MAGENIS SYNDROME CRITICAL REGION 19 Background .................................................................................................................. 19 The Smith-Magenis syndrome common and critical regions ............................. 20 Rationale ...................................................................................................................... 23 Results .......................................................................................................................... 23 Mapping of new genes and markers using a somatic cell hybrid mapping panel ........................................................................................................... 23 Construction of the physical map and contig of the SMS critical region ........... 26 Analysis and characterization of ESTs mapped to the SMS critical region ....... 29 Summary of known candidate genes mapping within the SMS critical region ......................................................................................................... 37 Summary ...................................................................................................................... 42 Materials and methods ................................................................................................. 43 CHAPTER 3. ANALYSIS OF l7p11.2 DELETIONS USING FLUORESCENT IN SI T U HYBRIDIZATION-_ ....... 48 Background .................................................................................................................. 48 Rationale ...................................................................................................................... 51 Results ......................................................................................................................... 52 Commercially available diagnostic FISH probes for SMS will not detect all cases. . ........................................................................................................ 56 The SMS common deletion occurs less frequently than reported ...................... 63 Summary ...................................................................................................................... 65 vi Materials and mehods .................................................................................................. 67 CHAPTER 4. ANALYSIS OF SMITH-MAGENIS SYNDROME PATIENTS WITHOUT A FISH DETECTABLE 17P11.2 DELETION ......... 72 Backround and rationale .............................................................................................. 72 Results .......................................................................................................................... 73 Mutation analysis in SMS patients without a FISH detectable deletion ............. 73 The retinoic acid induced l (RAII) gene ............................................................ 84 Summary ...................................................................................................................... 86 Materials and methods ................................................................................................. 87 CHAPTER 5. DEVELOPMENTALLY REGULATED GTP-BINDING PROTEIN(DRGZ) 94 Background .................................................................................................................. 94 Cloning and identification of mammalian DRG genes ....................................... 95 Rationale ...................................................................................................................... 96 Results .......................................................................................................................... 97 Identification of the STS WI-13499 as DRGZ and mapping to the SMS critical region. ...................................................................................................... 97 Bioinformatic analysis of DRGZ ......................................................................... 99 Expression levels of DRGZ/DrgZ using northern analysis ............................... 101 Spatial and temporal expression of Drg2 using immunohistochemistry. . .......102 Cellular localiztion of Drg2 .............................................................................. 1 10 Summary .................................................................................................................... 114 Materials and methods ............................................................................................... 115 CHAPTER 6. TARGET OF MYB-l (CHICKEN) LIKE 2 (TOM1L2) ............. 120 Background ................................................................................................................ 120 Rationale .................................................................................................................... 120 Results ........................................................................................................................ 120 Bioinformatic analysis fo TOMILZ .................................................................... 121 Northern analysis of TOMILZ ............................................................................ 122 T am] 12 gene trapped ES cell line ....................................................................... 124 XG909 genotyping and creation of XG909 mice ............................................... 125 Analysis of the F2 generation of Tom112 gene-trapped mice ............................ 126 Summary .................................................................................................................... 130 Materials and methods ............................................................................................... 135 CHAPTER 7. DISCUSSION 136 Future studies in the Elsea Laboratory ....................................................................... 137 TOMILZ/Toml 12 ....................................................................................................... 137 DRG2/Drg2 ................................................................................................................ 140 vii RAH/Rail ................................................................................................................... 141 Conclusion ................................................................................................................. 145 APENDIXA - - -- - - 147 APPENDIXB 156 APPENDIXC - ......... - - _ _ ..... 157 REFERENCES- - -_ - - 163 viii LIST OF TABLES Table l. ESTs mapped by Lucas et al., within the 2001 ~l .5 Mb critical interval identified as known genes ....................................................... 34 Table 2. Novel ESTs mapped by the Elsea Lab within the 2001 ~1.5 Mb critical interval ..................................................................................... 35 Table 3. ESTs recently mapped by other groups within the 2001 ~l .5 Mb critical interval ..................................................................................... 36 Table 4. SMS candidate gene disruption in mouse ..................................................... 39 Table 5. Phenotypic comparison of SMS patients with 17p deletions ....................... 61 Table 6. Frequency and type of SMS deletions .......................................................... 66 Table 7. Phenotypic comparison of SMS patients with deletions and RA]! mutations ..................................................................................... 82 ix LIST OF FIGURES Images in this dissertation are presented in color. Figure 1. Figure 2. Figure 3. Figure 4. Figure 5. Figure 6. Figure 7. Figure 8. Figure 9. Figure 10. Figure 11. Figure 12. Figure 13. Figure 14. Figure 15. Figure 16. Figure 17. Figure 18. Figure 19. Figure 20. Chromosome 17p somatic cell hybrid mapping panel ................................ 21 Somatic cell mapping of the RAII gene ...................................................... 25 Screening of the gridded RPll BAC library with D17S740 ....................... 27 Mapping of RA]! to BACs/PACs ................................................................ 28 Contig and transcription map of the SMS critical region ............................ 32 Human RAII northern analysis .................................................................... 33 Examples of deleted and not deleted FISH experiments ............................. 50 Summary of 17p11.2 deletions identified in this study ............................... 55 SMS patients with unusual deletions used to refine the SMCR .................. 58 Location of SMS FISH probes in relation to 17p11.2 deletions ............... 6O SMSlZ9 RAII mutation analysis ............................................................... 74 SMSlS3 RAII mutation analysis ............................................................... 77 SMS 1 56 RAII mutation analysis ............................................................... 79 SMS 1 59 RA]! mutation analysis ............................................................... 81 Somatic cell mapping of DRGZ ................................................................. 98 Assignment of DRG2 amino acid sequences .......................................... 100 Northern analysis of DRGZ/DrgZ ............................................................ 103 Transfection of Drg2 into COS-7 cells .................................................... 105 Temporal expression of Drg2 during mouse development ..................... 107 Cellular localiztion of Drg2 with GFP .................................................... 113 Figure 21. T 0M1L2 northern analysis ...................................................................... 123 Figure 22. Genetic analysis of the XG909 cell line ................................................... 128 Figure 23. Statistical analysis of XG909 mouse measurements ................................ 132 xi a.a.: AGS: ARF' ATPAFZ: aMT6: ANOVA: BAC: BLAST: CEPH: CLP: CL YN2: del: DRGZ: ELN: ES: EST: FISH: GFP: FLII: FZD3: HBSS: HGP: HTGS: IHC: kb: JA GI : LCR: LIMKZ: LLGLI : Mb: MR: NAHR: NCBL NHGRI: NIH: OMIM: PCR: PAC: PEMT 2: RA: RAII : RASDI : LIST OF ABBREVIATIONS amino acid Alagille syndrome ADP-ribosylation factor ATP synthase mitochondria F l comlex assembly factor 2 6-sulphatoxymelatonin analysis of variance bacterial artificial chromosome basic local aligment search tool Centre d'Etude du Polymorphisme Humain coactosin like protein cytoplasmic linker 2 deletion or deleted developmentally regulated GTP-binding protein elastin embryonic stem expressed sequence tag fluorescent in situ hybridization green fluorescent protein Drosophila flightless I homolog frizzled 3 Hank’s balanced salt solution Human Genome Project high-troughput genome sequencing immunohistochemistry kilobase jagged 1 low-copy repeats LIM domain kinase 2 lethal giant larvae homolog 1 megabase mental retardation nonallelic homologous recombination National Center for Biotechnology Information National Human Genome Research Institute National Institues of Health Online Mendelian Inheritance in Man polymerase chain reaction Pl-artificial chromosome phosphatidylethanolamine N-methyltransferase retinoic acid retinoic acid induced 1 Res, dexamethasome-induced l xii RF C2: REM sleep: RSTS: SCN: SIB: SMS: SMCR: SMS-REPD: SMS-REPM: SMS-REPP: SNP: SRP: SSCP: STX 1A: SVAS: TOMILZ: TRE: UTR: VHS: WS: YAC: replication factor c subunit 2 rapid eye movement sleep Rubinstein-Taybi syndrome suprachiasmatic nucleus self-injurious behavior Smith-Magenis syndrome Smith-Magenis critical region distal SMS repetitive element middle SMS repetitive element proximal SMS repetitive element singal nucleotide polymorphism signal recognition particle from canine pancreas signal strand conformation polymorphism syntaxin 1 supravalvular aortic stenosis Target of myb-l (chicken) like 2 human tumor specific antigen untranslated region Vps27p, Hrs and STAM Williams syndrome yeast artificial chromosome xiii Chapter I Introduction Smith-Magenis syndrome (SMS; OMIM#182290) is a multiple congenital anomalies and mental retardation syndrome usually associated with a de novo interstitial deletion of chromosome 17p1 1.2 (Smith et a1. 1986; Stratton et al. 1986; Lockwood et a1. 1988; de Almeida et a1. 1989). The 17p deletions associated with SMS are difficult to detect using standard cytogenetic Giemsa staining (G-banding) techniques and are thus termed microdeletions. In addition to SMS, there are many other microdeletion syndromes including Rubinstein-Taybi syndrome (del 16p13.3), Miller-Dieker syndrome (del l7p13.3), Alagille syndrome (del 20p11.23), DiGeorge/velocardiofacial syndrome (del 22q11.2), and Williams syndrome (del 7q11.2). As cytogenetic techniques improved, additional microdeletion disorders were described. The SMS-associated l7p11.2 deletions usually require high resolution banding to the 550 or 850 band level in order to be detected with G- banding. This technology did not become reliably available until the early 19805, which is likely why the SMS phenotype was not associated with 17p11.2 deletions until 1982 with the initial description by Ann Smith and Ellen Magenis. With the advent of fluorescent in situ hybridization (FISH), diagnostic testing for SMS has become more routine. In just 20 years, the number of known SMS cases now totals in the thousands worldwide. Unfortunately, the disease is not widely known and under- diagnosis is likely. Birth incidence of SMS was estimated in one study at 1:25,000 live births (Greenberg et a1. 1991). The association of SMS with a microdeletion of chromosome 17p11.2 makes the syndrome an excellent candidate as a contiguous gene disorder. Contiguous gene syndromes are a group of identifiable disorders caused by chromosomal abnormalities such as deletions or duplications affecting normal gene dosage (Schmickel 1986). The genes affected by the deletion or duplication are generally related only by their proximity to each other along the chromosome and not by their function. The dosage sensitive genes are thought to contribute independently to the characteristics of the contiguous gene syndrome; although, it is possible that a change in the gene dosage of some genes would have no biochemical effect and thus may not contribute to the overall phenotype. It is also possible that dosage changes in one gene causes the entire phenotype seen in the microdeletion syndrome, making the disease a single gene disorder. Additionally, some aspects of the contiguous gene disorder phenotype may segregate as Mendelian inherited traits (Schmickel 1986). The ultimate goal when studying contiguous gene disorders is to find the gene or genes responsible for the characteristics that make up the syndrome phenotype. The SMS Phenotype The SMS newborn is usually born at term of normal weight and length; APGAR scores are generally normal. The baby usually has a facial appearance that is described as cherubic, caused by mid-face hypoplasia, full checks, 3 tented upper lip, upward slanting eyes, an upturned nose, and micrognathia. The facial features can be subtle and are easily overlooked (Allanson et a1. 1999). Infantile hypotonia is usually present, and the infant may display poor sucking/swallowing and feeding. Failure to thrive has been noted in some cases (Smith et a1. 1986; Greenberg et al. 1996). Parents describe the SMS infant as the perfect baby who cries infrequently, has a smiling disposition, and sleeps well through the night. The infant is usually described as complacent, lethargic, and generally happy. These features overlap with other genetic disorders, and SMS children are often misdiagnosed based on phenotypic characteristics with Down syndrome or Prader-Willi syndrome (Allanson et al. 1999). Additional congenital anomalies including cleft lip/palate, renal/urinary tract anomalies, seizures, thyroid abnormalities, and heart defects have been noted in ~25% of patients (Chen et al. 1996; Greenberg et a1. 1996). During childhood, the SMS phenotype begins to become more apparent. Though overall length is normal at birth, growth is slow and children are generally short during childhood. Slower growth results in a shified growth curve, but the patients usually end up within the normal height range after puberty. Though height is within normal limits, SMS patients are usually shorter than their genetically predicted heights. Brachydactyly is almost always present. With age, the facial features begin to become more distinctive to SMS. The eyes remain up slanting. The lower jaw becomes more broad and protrusive causing a distinct square looking face especially during puberty. Mid-face hypoplasia becomes more noticeable, and mild brachycephaly is common. The brow is usually noted as heavy with the presence of synophrys adding to the effect (Allanson et al. 1999). Scoliosis can become problematic during childhood and is reported in over 50% of SMS children over age 4 (Greenberg et al. 1996). Further pediatric evaluations in SMS patients can reveal additional anomalies. Opthalmic abnormalities have been thoroughly studied in SMS patients with a variety of irregularities reported. These include iris abnormalities, microcornia, myopia, and strabismus occurring in ~50 to 60% of patients (Finucane et a1. 1993; Chen et al. 1996). Blood workup shows hypercholesterolemia in over 50% of patients (Smith et al. 2002), and decreased immunoglobulins, specifically IgA, in <25% of patients examined (Greenberg et al. 1996; Cassidy and Allanson 2001). By childhood, the presence of mental retardation (MR) is apparent and is usually in the mild to moderate range (IQ 40-54) (Dykens et al. 1997); however, cases of borderline MR to severe MR have been reported (Greenberg et a1. 1996). Parents of SMS patients can expect delay of motor and speech developmental milestones. Signs of peripheral neuropathy including pes planus, pes cavus, depressed deep tendon reflexes, and insensitivity to pain are seen in >75% of SMS patients (Greenberg et al. 1996). SMS patients also display a spectrum of otolaryngeal anomalies. Otitis media is common leading to placement of ventilation tubes in the ears to alleviate the frequent infections. Sensorineural or conductive hearing loss are frequent and occur in ~68% of patients (Greenberg et al. 1996). Almost all patients have a very characteristic hoarse deep voice. Laryngeal anomalies reported include polyps, nodules, edema and/or partial vocal cord paralysis (Cassidy and Allanson 2001). Additional otolaryngologic problems seen in SMS patients include, velopharyngeal insufficiency, weak bilabial seal, palatal anomalies, limited tongue motion, and fiequent drooling (Greenberg et al. 1996; Cassidy and Allanson 2001). SMS children also have an increased incidence of sinusitis, some requiring surgical intervention (Cassidy and Allanson 2001). Parents also complain that children are sometimes affected with bouts of constipation (Smith et al. 1998b). The neurobehavioral hallmarks of SMS discussed below are the most recognizable characteristic and give parents and professionals the most difficulty in dealing with SMS patients. Sleep disturbance is common in >75% of SMS patients (Smith et al. 1998b). Though sleep disturbance is generally not a problem at birth, alteration of sleep patterns begins around 9 months, and sleep progressively deteriorates. In an initial sleep study performed in 1996, 50% of SMS participants (12/24) had a reduced amount of REM sleep (Greenberg et al. 1996). Further study of sleep in SMS patients has revealed an increased number of awakenings during the night and early awakening time in the morning leading to an overall decrease in the amount of total sleep time, daytime sleepiness, and increased number of daytime naps (Smith et al. 1998b). One of the main hormonal components of sleep is melatonin. Melatonin secretion is controlled by the suprachiasmatic nucleus (SCN) of the brain. The SCN is sometimes referred to as the human biologic clock and is an endogenous keeper of circadian rhythm. The SCN controls secretion of melatonin by signaling through the neurotransmitter norepinephrine to the pineal gland. Light/dark cycles have an impact on the SCN through the retinohypothalarnic tract that connects the retina to the SCN. Thus, melatonin secretion can be affected by changes in exposure to light and darkness. Exposure of the photoreceptor cells to light causes their hyperpolarization, inhibiting both the release of norepinephrine and melatonin synthesis. When light is reduced, the cells become less polarized and norepinephrine is released to the pineal gland, which in turn produces and secretes melatonin to the blood stream (Brzezinski 1997). In humans, melatonin secretion peaks in the middle of the night somewhere between 2 and 4 am. and is at its lowest around 12 hours later. Melatonin is quickly metabolized in the liver by hydroxylation and conjugation with sulfuric or glucuronic acid to 6-sulphatoxymelatonin (aMT6s) which is excreted in the urine. Levels of aMT6s in mine mirror blood serum levels of melatonin (Lynch et al. 1975). Thus, measurement of aMT6s in urine is a reliable way to measure melatonin levels over a period of time without the need for repetitive blood draws. Two studies have reported the circadian rhythm of melatonin in SMS patients. These studies have shown an inverted rhythm in 37 of 38 SMS cases studied (Potocki et al. 2000b; De Leersnyder et al. 2001a). In the SMS individual, melatonin peaks midday and is at its lowest in the middle of the night. This inverted circadian rhythm of melatonin may contribute to the nighttime restlessness and early awakening experienced by SMS patients. In order to try to reduce the nighttime awakenings and early rising, parents of SMS patients have tried melatonin supplementation. Improvement in sleep has not been widely seen with this supplementation. This is likely due to the fact that increasing the amount of melatonin alone will only boost the total amount in the system resulting in two peaks rather than reversion of circadian rhythm back to normal. Two studies were conducted in France in an attempt to correct the sleep disturbance in SMS patients (De Leersnyder et al. 2001b; De Leersnyder et al. 2003). In the initial study, 10 SMS children were treated with the selective Bl-adrenergic antagonist acebutolol in the early morning. With this treatment the patient’s midday melatonin peak went away and sleep at night improved (De Leersnyder et al. 2001b). In the follow-up study children were treated with the 61 -adrenergic antagonist in the morning, and a melatonin dose in the evening (De Leersnyder et al. 2003). Following this scheme restored the normal circadian rhythm of melatonin. The sleep in the children involved in the study improved, nighttime awakenings diminished, and wake time was delayed thus allowing for a longer more restful sleep (De Leersnyder et al. 2001b; De Leersnyder et al. 2003). Currently, clinical trials using similar methodologies and further sleep studies to better understand the SMS sleep disturbance are underway at The Oregon Health and Science University and at The National Institutes of Health. Self-injurious (SIB) and maladaptive behaviors are another neurobehavioral hallmark of SMS patients. In fact, these behaviors are the most recognizable characteristic that usually causes the physician to suspect SMS. The behaviors associated with SMS can begin as early as age two and usually begin with head- banging and wrist biting (Smith et al. 1998a). These behaviors are often sparked when the child becomes upset or frusterated. Uncontrollable tantrums usually develop in combination with the head-banging and hand/wrist biting. As the children age and become stronger, they can become a danger to themselves or others during periods of tantrum. With age, additional self-injurious behaviors begin to manifest including onychotillomania (pulling/picking of finger and toenails), and polyembolokoilomania (insertion of objects into bodily orifices), usually beginning after age 5 (Greenberg et al. 1996). Skin and nail picking and biting can be persistent and quite severe causing permanent scaning. Additional stereotypical behaviors common in SMS patients include the spasmodic upper-body squeeze (known as “self- hugging”) and “licking and flipping.” The self-hug takes on two forms, one is an upper body hug, while the other includes clapping together the hands at chin level followed by the interlocking of the fingers and pushing of the hands together with the elbows pointed away from the body. These self-hugging behaviors occur usually when the child is very excited and appear involuntary (Dykens et al. 1997; Smith et al. 1998a). The licking and flipping behavior involves licking the finger before turning the pages of a book. The motion is quick and repetitive with little stopping between pages (Dykens et a1. 1997). Various drugs have been tried to combat the behaviors associated with SMS including antidepressants, antipsychotics, and anticonvulsants though none has proven extremely helpful over time and effectiveness seems to vary between individuals (Smith et al. 1998a). After puberty and throughout adulthood the self-injurious and maladaptive behaviors become less prominent. However, SIB can still be an issue during times of anger. The facial features continue to become more coarse, the chin can become very prominent, and the brow very heavy. Scoliosis can become more severe with age. The life expectancy of people with SMS seems to be normal. The oldest known living SMS patient is over 80 years old. There does not seem to be any increase in the incidence of cancer in SMS patients. Though the phenotype in infants and children is well known, less is known about what can be expected in the SMS adult. Studies of the natural history of SMS are being currently being conducted by Ann Smith at NHGRI/NII-I in order to refine the characteristics of the phenotype over the lifetime. 17p11.2 deletions associated with SMS Association between microduplication and microdeletion syndromes and flanking repetitive elements termed low-copy repeats (LCRs) is known (Ji et al. 2000). It is estimated that LCRs cover between 5%-10% of the human genome (Eichler 1998; Ji et a1. 2000). The LCRs can range in size from ~10-400 kb, averaging between 150-200kb, and having >97% homology (Eichler 1998; Ji et al. 2000; Stankiewicz et al. 2003). Because of the size similarity to BACs and PACs used in the human genome project and the repetitive nature of the LCRs (independently occurring in between 2-10 copies), they caused considerable difficulty in mapping and sequencing of the human genome (Bailey et a1. 2001). LCRs are different than the highly repetitive repeats, i.e. Alu, SINE/LINE, and satellite repeats, which are present in numerous copies and were identified based on reassociation kinetics. LCRs are made up of genes, gene fi'agments, pseudogenes, and retroviral sequences. Data have shown that nonallelic homologous recombination (NAHR) between the LCRs causes the segmental anomalies seen in the microdeletion/microduplication disorders (J i et al. 2000). NAI-IR can occur interchromosomally or intrachromosomally resulting in deletion, duplication, or segmental rearrangement depending on the individual characteristics of the LCRs involved (Stankiewicz and Lupski 2002). The first physical map of the SMS deletion region constructed in 1997 by Chen et. al. using yeast artificial chromosomes (Y ACs) identified the LCRs mapping to the SMS region (Chen et al. 1997). Using a combination of PCR, somatic cell hybrid mapping, and cosmid mapping, the authors were able to locate three LCRs mapping within l7p11 which were termed SMS-REPS. Each SMS-REP was represented by YACs that contained unique flanking sequence solidifying the hypothesis that there were three unique repetitive regions. The authors termed the regions SMS-REPP (proximal), SMS-REPM (middle), and SMS-REPD (distal). The SMS-REPS were determined to be repeated gene clusters of ~200 kb, though it was not clear whether the genes within the REPS are expressed or are pseudogenes. Three genes map to all three REP sequences including type-l keratin (KER), signal recognition particle from canine pancreas (SRP), and the human tumor specific antigen TRE (T RE). Another gene, coactosin like protein (CLP) only maps to the proximal and distal REPS, and not the middle REP. Based on Southern analysis, the authors estimated the size between the proximal and distal REP to be 5 Mb. The existence of the REP sequences led the authors to investigate whether they could detect the junction fragment that would remain after recombination events leading to 17p deletions associated with SMS. Using a cDNA representing the CLP gene, the authors were able to demonstrate an SMS specific junction fragment in 90% of patient samples studied. The authors hypothesized that NAHR between SMS-REPP and SMS-REPD facilitates the majority of the deletions seen in SMS, and that 90% of all SMS patients carry the same deletion that was estimated to cover ~5 Mb (Chen et al. 1997). The reciprocal of the SMS deletion at 17p11.2 would be a segmental duplication. The segmental duplication of 17p11.2 was identified in 7 patients with mild mental retardation and dental anomalies (Potocki et al. 2000a). Genetic proof of NAHR between the distal and proximal SMS-REPS was 10 provided via genotyping of 24 patient samples harboring 17p11.2 deletions/duplications. The analysis of the 24 cases showed 11 rearrangements resulting from interchromosomal recombination and 13 from intrachromosomal recombination indicating no preference for exchange between or within sister chromatids or homologs (Shaw et al. 2002). Recombination between the proximal and distal SMS-REPS causes the majority of SMS deletions, but these are not the only 17p11.2 deletions associated with SMS. Deletions overlapping the SMS common interval have been reported (Juyal et al. 1996; Trask et a1. 1996; Elsea et al. 1997). These unusual sized deletions can be larger or smaller than the common deletion, and are currently reported to occur in ~25% of patients, and are mediated by NAHR between the distal and middle SMS-REPS and/or other newly discovered low copy repeats in the region (Stankiewicz et al. 2003; Vlangos et al. 2003). Unusual deletions provide an invaluable resource for the study of microdeletion disorders. Overlapping deletions fi'om patients who retain all aspects of the syndrome phenotype delineate the critical region of the disorder. The gene(s) causing the disease phenotype are most likely to map within the disease critical region. The SMS critical region (SMCR) was first described in 1997 (Elsea et al. 1997). The SMCR was represented by an unusual deletion harbored by a single patient. This patient’s deletion was located between the distal and middle SMS-REPS and spanned ~1.5 Mb. Haploinsufficiency of a gene, or genes, mapping within this 1.5 Mb deletion was hypothesized to be the cause of the Smith-Magenis syndrome phenotype. 11 Microdeletion syndromes: from critical region to gene identification Determining the role that any of the positional candidate genes within the nricrodeletion syndrome critical region play in producing the characteristics of the disease phenotype is a complicated process. Multiple approaches are usually employed simultaneously in order to successfully determine the role the candidate genes may play in disease etiology. The techniques employed in this research have been successfully used to determine disease etiology in other microdeletion syndromes including Williams syndrome (WS; del 7q11.2; OMIM#194050) and Alagille syndrome (AGS; del 20p12; OMIM#118450). Williams syndrome is an autosomal dominant disorder associated with deletion of chromosome 7q11.23. Most deletions occur de novo, though incidences of parental transmission have been reported. Patients with Williams syndrome have a distinct facial appearance including a wide mouth, flat nasal bridge, small mandible, and prominent cheeks (Morris et al. 1988). Puberty is often precocious, and there can be a loss of subcutaneous tissue giving rise to a look of premature aging (Donnai and Karmiloff-Smith 2000). Cardiac anomalies in the fornr of supravalvular aortic stenosis (SVAS) and peripheral pulmonary artery stenosis are common to WS patients and occur in >80% of patients (Osborne 1999). Stenosis of other peripheral vessels including the subclavian, corornary, carotid, mesenteric, and renal have also been reported. The behavioral/cognitive profile of WS patients is a striking array of extremes. WS patients are usually affected with moderate to severe mental retardation; average IQ ranges between 51 and 70 (wain and Yule 1991). Though 12 MR is present WS patients do not have problems with language or face and emotional processing. This is in stark contrast to the extreme problems with visouspatial cognition (Donnai and Karmiloff-Smith 2000). An amazing propensity for music is displayed, but hypercusis is present and patients are often startled and display extreme anxiety in the presence of loud noise. Anxiety is also present when the WS patient is placed in a new situation. However, their personality is extremely friendly and social, and fear of strangers is not present (Osborne 1999). The deletion associated with WS is mediated by recombination between LCR sequences as is seen in SMS (Urban et al. 1996). The WS deletion interval was defined in 1998 by assessing 200 WS patients using FISH. The interval spans ~1.5 Mb of DNA. In order to facilitate mapping of candidate genes a complete BAC/PAC physical map was constructed across the region (Meng et al. 1998). When the physical map of the WS region was constructed the etiology of one aspect of the phenotype was already known. Isolated SVAS, an autosomal dominant trait, was found to map to the same chromosomal region by linkage analysis and by discovery of a translocation involving chromosome 7q co-segregating in a family with SVAS (Curran et a1. 1993; Ewart et al. 1993). It was determined that the translocation breakpoints in these patients disrupted the elastin gene (ELN). Investigation of Williams syndrome patients determined that >98% were hemizygous for ELN (Lowery et al. 1995). In evaluation of DNA samples from patients with isolated SVAS nonsense and splice site mutations were found within the elastin gene (Li et al. 1997a; Tassabehji et a1. 1997). Haploinsufficiency of ELN in humans causes the hardening of the vessels seen in SVAS and WS. 13 Elastin is produced in smooth muscles and eventually is formed into concentric rings of elastic lamellae alternating with rings of smooth muscle (Uin et al. 1991). Haploinsufficiency of ELN accounts for the SVAS seen in WS, but not the other characteristics of the syndrome. Thus, Williams syndrome is a true contiguous gene syndrome where other genes in the critical region are responsible for other aspects of the phenotype. Additional genes mapping to the region include LIM domain kinase-2 (LIMKZ) and cytoplasmic linker 2 (CL 1W2) thought to be responsible for the impaired visuospatial cognition and the neurologic alterations respectively. Replication factor c subunit 2 (RFCZ) is also a positional candidate for WS (Osborne 1999). Additional genes that were thought to be excellent candidate genes included syntaxin 1 (STXIA) and fiizzled (FZD3). Study of two classic WS patients with unusual deletions showed that these genes are not included in the WS critical region as these patients are not deleted for these genes (Botta et a1. 1999). To date, haploinsufficiency of the elastin gene is the only known cause for characteristics seen in Williams syndrome. The search for additional causative genes in WS continues through attempts to further narrow the critical region and characterization of the positional candidate genes mapping to the region. Alagille syndrome (AGS) is an autosomal dominant disorder with variable expression that can be associated with a deletion of chromosome 20p12. Phenotypic diagnosis of AGS is given upon confirmation of paucity of the interlobular bile ducts upon liver biopsy, and at least three of the five major clinical features described by Alagille in 1975 including chronic cholestasis, cardiac disease, skeletal anomalies, ocular anomalies, and characteristic facial features (Alagille et al. 1975; Krantz et al. 14 1999). Hepatic, cardiac, and opthalrnologic manifestations are the most common features seen in AGS. Most patients are of normal intelligence, though mild delay, delay of gross motor skills, and mild MR have been associated with the disease. Additional anomalies associated with the syndrome include delayed puberty, hearing loss, renal defect, and pancreatic insufficiency (Alagille et al. 1975; Li et al. 1997b; Oda et al. 1997b; Krantz et al. 1999). From the initial description of the disease it was clear that AGS was an autosomal dominant disorder with variable expressivity based on familial inheritance of the disease (Alagille et al. 1975). In fact, clinical manifestations in AGS are highly variable between individuals, ranging from subclirrical to life threatening. Birth incidence is reported between 1:70,000 to 1:100,000 births (Oda et al. 1997b; Krantz et al. 1999). Cytogenetic analysis of most AGS patients reveal a normal karyotype, though a small subset (~7%), have interstitial deletions or rearrangement of chromosome 20p12. Patients who harbor large cytogenetically visible deletions of chromosome 20p are usually more affected with the spectrum of phenotypic characteristics. Thus, AGS was an excellent candidate as a contiguous gene syndrome. The AGS critical region was delineated using overlapping 20p deletions, and was estimated to span between 8 and 11.3 cM based on CEPH mapping and linkage analysis in AGS patients without a 20p deletion (Hol et a1. 1995; Pollet et al. 1995). In the mid 19908 the number of mapped genes and available genetic markers were small. A physical map of a continuous path of YAC clones was constructed spanning 3.7 Mb of 20p12 including the AGS critical region in order to facilitate mapping of 15 new markers and identification of possible candidate genes via elucidation of CpG islands (Pollet et al. 1995). This first physical map containing 15 new anonymous markers was an excellent start toward identification of AGS candidate genes. Further analysis of AGS patients with 20p deletions narrowed the critical region to ~250 kb. A contig of the refined region was created using BAC clones which are less susceptible to recombination than the more unstable YAC clones. At the time only one gene mapped wholly within this refined AGS critical region, human Jagged] (JAGI), a gene orthologous to the rat Jagged gene which is involved in the notch signaling pathway (Oda et al. 1997b). The JA GI gene encodes a ligand for the Notch receptor. The notch receptor through association with its ligands creates a signaling pathway used in directing cell fate (Luo et a1. 1997; Oda et al. 1997a). Because of its possible function and map location JA GI was an excellent candidate gene for AGS. Analysis of DNA samples from patients without a deletion of chromosome 20p was performed via SSCP and direct sequencing of PCR amplicons of the JAGI gene. Two separate groups identified 11 mutations in the JAGI gene segregating in affected AGS patients (Li et al. 1997b; Oda et al. 1997b). Haploinsufficiency of the JAG] gene via hemizygous deletion or mutation is the cause of Alagille syndrome. In contrast to the theory of the contiguous gene syndrome hemizygous deletion of other genes flanking the JA GI gene does not seem to affect the syndrome phenotype. Animal modeling of Smith-Magenis syndrome The mouse is an excellent organism for use in animal modeling of human 16 disease due to the similarity of physiology, anatomy, and genetics. Mice are also relatively inexpensive to house and propagate. The techniques for manipulating the mouse genome have become readily available and are performed routinely. In addition, the completion of sequencing of the mouse and human genomes facilitates comparative analysis aiding experimental design. Genetic mutation of the mouse can include inactivation of an individual gene, or manipulation of entire segments of DNA via chromosomal engineering. In the mouse chromosome 11 is syntenic to human chromosome 17. Comparative analysis of the SMS critical region between mice and humans shows that order and orientation of 19 genes are conserved (Bi et al. 2002). Chromosomal engineering was undertaken in order to examine the phenotypic effect of deleting the SMS common region in mouse (Walz et al. 2003). Mice lacking a 3 Mb segment of chromosome 11 syntenic to the SMS common region were created and termed Df(11)17 animals. Heterozygous animals displayed craniofacial anomalies, EEG abnormalities, weight differences, and decreased fertility when compared to normal litterrnates in a mixed genetic background. Gross organ abnormalities in the heart, urinary tract, and eyes were not noted (Walz et a1. 2003). Behaviorally, the male Df(1 I )1 7 mice were reported to by hypoactive, while the female heterozygotes had normal locomotor activity. Both male and female heterozygotes showed shorter circadian periods when placed in constant darkness after light entraining (Walz et al. 2004). This response indicates a reduced precision in the circadian clock. Heterozygous mice had no self-injurious behaviors, decreased sensation to pain, or startle responses (Walz et al. 2004). These results indicate that a dosage sensitive l7 gene(s) maps within the chromosomal engineered region. Targeting of individual genes in the region will hopefirlly aid in understanding the genotype:phenotype correlation in humans. SUMMARY The goal of the research project presented here was to determine the gene(s) responsible for Smith-Magenis syndrome. The work explained in the following chapters follows the methods employed in elucidating the etiology of Alagille and Williams syndromes, as well as other rrricrodeletion disorders. Chapter 2 focuses on work performed in creation of a physical and transcriptional map of the SMS critical region. Chapter 3 describes narrowing of the SMS critical region using FISH, as well as determination of a more efficient diagnostic SMS probe. Chapter 4 illustrates direct sequencing efforts of candidate genes in SMS patients where deletion of chromosome l7p11.2 could not be detected. This research led to the identification of mutations in the RAII gene which is likely responsible for the majority of SMS characteristics. Chapters 5 and 6 focus on characterization of two positional candidate genes, DRG2 and T OMILZ respectively, which may play a role in the less penetrant SMS characteristics. Chapter 7 concludes the dissertation by discussing how the work presented could be incorporated into future research projects studying Smith-Magenis syndrome. 18 Chapter II Physical mapping of the Smith-Magenis syndrome critical region BACKGROUND The goal of the research projects in the Elsea Lab is to identify the dosage sensitive gene(s) mapping to chromosome 17p11.2 responsible for the characteristics making up the SMS phenotype. Prior to my joining the project, much work had been performed toward this goal. The SMS critical region (SMCR) had been defined, a somatic cell hybrid mapping panel had been created, and many markers had been mapped to the short arm of chromosome 17 by the lab. Using the markers previously mapped, we embarked on construction of a continuous path of large insert clones (a “contig”) across the SMS critical region. The creation of the contig also allowed for mapping of additional markers, genes, and ESTs to the SMS critical region. The final contig includes the order and orientation of the genes, ESTs, and markers mapped to the critical region. We initially characterized all the genes and ESTs mapping to the region using various bioinformatic methods and northern analysis. The genes were prioritized for further study based on initial characterization as candidate genes for SMS. Our lab published this work in the European Journal of Human Genetics (Lucas et al. 2001). 19 The Smith-Magenis syndrome common and critical regions As discussed in Chapter I, the deletions associated with SMS can range in size fi'om <2 Mb to >9 Mb (Trask et al. 1996), although it has been reported that >95% of SMS patients carry a common ~3.5 Mb deletion (Chen et al. 1997). Though there are a spectrum of deletions associated with SMS, the phenotype remains consistent in most patients. The dosage-sensitive genes responsible for the syndrome phenotype are theorized to map to the smallest shared region of deletion in patients displaying the full syndrome phenotype. This region is referred to as the critical region. The SMS common deletion and the critical region were delineated in 1996 by studying the deletions carried by 62 SMS patients (Juyal et al. 1996). At the start of this project, the SMS critical region was defined by the deletion carried by a single patient, HOU142-540 (Figure 1) (Elsea et al. 1997). This patient carries an unusual deletion spanning ~1.5 Mb of DNA roughly between the distal and middle SMS-REPS. Molecularly, the region is bounded by the genomic marker D17SZ9 proxirnally and an anonymous cosmid cCIl7-638 (Figure 1) (Juyal et al. 1996; Elsea et al. 1997). The deletion size was delineated by FISH using cosmid probes for the small number of known markers mapping to the region (Juyal et al. 1996; Elsea et al. 1997). At the beginning of this project, 7 genes, 13 different ESTs, and 7 markers had been mapped within the SMS critical region (Elsea et al. 1997). At the same time that the SMS critical region was delineated, the common region boundaries were also identified. The common interval was shown to map between genomic marker D17SS8 proxirnally and the anonymous cosmid cCIl7-498 20 egaééfi-a ififiEfiefls N 14 '2: II“. 13 S 12 | 11 01‘ all 10 :1 .. 9 a. 3 S {I 7 a. | 6 i E ' A. ET :3 ||‘-‘ 5 "l I 4 7,—— 111— s a I I I "‘ IIMII 2 '1. ‘1] W: 3 l l 1 E‘ v-' Bin Figure l. Chromsome 17p somatic cell hybrid mapping panel. Rodent-human somatic cell hybrids retaining a deleted chromosome 17 are illustrated at the right. The breakpoints of the naturally occurring deletions divide the region into bins. Genes and markers mapping to bins were used as anchors in the construction of the critical interval contig. The common deletion seen in SMS is indicated in orange and spans ~3.5 Mb and bins 4-10. The SMS critical interval indicated in red spans ~l.5 Mb and bins 6-8. The approximate location of the SMS- REPs are indicated in blue. 21 This region encompasses ~3.5 Mb of DNA roughly between the proximal and distal SMS-REPS (Figure 1) (Juyal et al. 1996; Elsea et al. 1997). In molecularly defining the SMS critical region, markers were also mapped to common SMS region along chromosome 17p. The authors mapped 85 additional markers to chromosome 17p surrounding the SMS critical region (Elsea et a1. 1997). Mapping of the markers was facilitated by the creation of a somatic cell hybrid mapping panel of overlapping 17p deletions (Figure 1) (Guzzetta et al. 1992; Patel et al. 1992; Elsea et a1. 1997). The rodentzhuman hybrid cells were grown and maintained to harbor the human chromosome 17 of interest (Guzzetta et al. 1992). The boundaries of the overlapping deletions were used to divide the p-arm of chromosome 17 into 14 bins. The SMS critical interval contained bins six, seven, and eight as delineated by the unusual deletions canied by HOU142-540, and patients with deletions overlapping the SMS critical region carried by HOU261-765 and HOU92-357 (Figure 1) (Elsea et al. 1997). The SMS common deletion was reported to span bins 3 and 10/11 as delineated by numerous patients with overlapping 17p deletions (Figure 1) (Elsea et al. 1997). A preliminary physical map of the region created using YAC clones was reported in 1997 along with the data showing the presence of the SMS-REPS (Chen et al. 1997). The map and contig reported contained many gaps, and only some of the markers were in the correct order. The orientation of some of the map was inverted based on data reported in the delineation of the critical region. The problems with this initial map likely stem from the lack of markers in the region, and the fact that YAC clones are very prone to recombination during propagation. 22 RATIONALE We undertook construction of a physical map and contig of the 1.5 Mb critical interval using BAC, PAC, and cosmid clones. These clones are more stable than the YAC clones used in previous physical maps (Chen et al. 1997). In construction of the physical map, new markers, ESTs, and genes were mapped to the SMS region. Initial characterization of the genes using bioinforrnatics and northern expression analysis was performed in order to prioritize the candidate genes for further study to determine the role, if any, they play in the SMS phenotype. RESULTS Mapping of new genes and markers using a somatic cell hybrid mapping panel DNA isolated fi'om the rodentzhuman hybrid cells making up the mapping panel was used in PCR reactions and Southern analysis to fine map newly obtained markers. The panel includes hybrids retaining a deleted human chromosome 17 from SMS patients and non-SMS patients harboring different sized 17p deletions. The following control cell lines were used for mapping along human chromosome 17; MHZZ-6 retains one entire normal human chromosome 17; LS-l carries an iso-q 17 chromosome, and 88H5 carries a fused chromosome of l7p:Xq. In order to exclude positive products resulting fiom the endogenous rodent DNA in the cell lines rodent parental lines a23 and Cl-lD were used. As the study of the human genome progressed, new markers and ESTs were reported, and their sequences were submitted to public databases at The National 23 Center for Biotechnology Information (NCBI; http://www.ncbi.n1m.nih.gov) including dbEST, dbSTS, and GenBank, as well as to The GDB Human Genome Database (http://www.gdb.org). Members of the laboratory regularly searched the databases to identify newly deposited information. The reagents necessary for fine mapping the markers to the SMS critical interval were obtained or created as described below. To map newly identified genomic markers to the region, specific oligonucleotide primer pairs were obtained for use in PCR using template DNA isolated from the rodentzhuman somatic cell hybrids. PCR reactions were performed, and products were analyzed for presence or absence of the expected banding pattern based on a positive human control sample. Once markers mapped within the SMS region, they could be used for screening of gridded clone libraries as described below. Newly identified EST sequences were obtained as clones through The Integrated Molecular Analysis of Genomes and their Expression (I.M.A.G.E.) consortirun (http://image.llnl.gov) or other international EST consortiurns located in Germany and Japan. Plasmid DNA was isolated and sequenced to ensure the correct clone was sent. The clone DNA was used as probe in Southern analysis to filters created through EcoRl digestion of the hybrid DNAs and controls. Banding patterns were analyzed to determine map location of the ESTs (Figure 2). Cloned EST DNA was also used in initial characterization of the ESTs as described below. 24 . .éeéee .. NairLé-tbli 03 gaazs§a§~§= =>.>.>~ mhg¢° Emmiiiii =2 >20kb—> pm- 1: .1 Figure 2. Somatic cell mapping of the RA]! gene. Fine mapping of RA]! within the SMS critical interval was performed by hybridizing the ~2.0 kb insert fiom EST DKFZp434A139Q2 to a panel of EcoRI digested somatic cell hybrids. The hybrids carry deleted chromosome 17 from SMS patients (Hy14720-D, Hy540-1D, and Hy484—2D), and non SMS patients with overlapping deletions along 17p (Hy765-1D and Hy357-2D). Proper positive and negative controls are described in the text (M822-6, Hy88H5, LS-l, a23, and Cl-lD). A >20 kb band representing the RA]! gene is evident in total human DNA, MI-122-6, and Hy88H5 (present but faint because of limited exposure time), demonstrating that RA]! is deleted in all SMS patients and maps to the central portion of the SMS critical region. 25 Construction of the physical map and contig of the SMS critical region Construction of a continuous path of large insert clones across the critical region was started while the Human Genome Project (HGP) was in its infancy. Sequence data was slow to appear within the SMS critical interval, and sequence data that did appear contained large gaps. Additionally, the markers identified by the HGP that matched those that we had previously fine-mapped to the region usually were in a different order than our data showed. These problems were likely caused by incorrect construction of the sequence fiagments produced by shotgun sequencing due to the highly repetitive nature of the DNA in the region. In order to find large insert clones mapping to the region, we repeatedly screened the RPll BAC library using the markers and ESTs mapping to the SMS critical region. PCR amplified marker, isolated EST DNA, or isolated cosmid DNA was used as probe and hybridized to the gridded arrays using standard Southern protocol. Positive clones are identified by specific spotting patterns displayed on the grid and decoded using a key provided by the library creators (Figure 3). Positive BACs were obtained and DNA was isolated using a modified Qiagen protocol designed in the lab. An initial contig was created using a combination of Ala-PCR and analysis of the EcoRI restriction patterns of the BACs. In order to determine the marker order on the individual BACs, Southern analysis was performed using the collection of markers mapped to the region as probes on EcoRI digested BAC DNA (Figure 4). Using these methods we were able to construct three contigs 26 Figure 3. Screening of the gridded RPll BAC library with D17S740. The gridded RPCI-ll BAC library was screened using ESTs and genomic markers with lcnown map positions inside the SMS critical region. Clones are spotted in duplicate within a 4 x 4 grid on the array. An example of the gridding pattern of the library is indicated in the upper left. Standard Southern technique with radio-labeled DNA probes was used in the screening. In the example above, the library was screened with the genomic marker D17S740. Positive clones are noted with black arrows. Positive clones were ordered and verified for map position by EcoRI Southern analysis. 27 .2 E 2 u 0 NO '- A) '_ °" N—nfag Noe: B) E o I: Guts—mhlnuam r: g u mam—mmvnvu o n" N "‘ a 2 ages: m. - .. a—e “an 5 u 903 £32 0 r: "' I~ 2 = Susan. 39. f 6I 353:3:32 a E oceanic—welt)? = 000%0%0 - E 1 can._a_r:.o 'j, g bc189D22 ‘. pc178F10 Figure 4. Mapping of RAII to BACs/PACs. A) Eight micrograms of DNA from BACs and PACs mapping within the SMS critical region and human genomic DNA (10 pg) were digested and then electrophoresed overnight in circulating buffer. The gel was photographed and DNA was transferred to nylon membranes using standard Southern technique. B) An autoradiograph of the Southern blot is shown. The insert from plasmid DKFZp434A139Q2 represents the 3’ end of the RAII gene and was used as a probe. The blue arrows indicate pc253P07 and a faint corresponding band in the digested human DNA lane. 28 >1) lb within the critical region, but were unable to fill the gaps with the BAC resources available at the time. As our project continued, the speed of data conring from the HGP began to increase. We were able to fill gaps in our contig using sequence data from the high-throughput genomic sequences (HTGS) being deposited into the public databases. We searched the HTGS databases using BLAST analysis with known marker and EST sequences which we had already mapped. Using this approach we were able to electronically identify numerous RPCI BACs/PACs and CIT BACs mapping to the SMS critical region. We obtained the positive BACs and PACs and confirmed the results obtained in silica using Southen analysis on EcaRI digested DNA and PCR analysis. Additional alignments and conti g organization were performed using the UCSC Genome Browser (http://genome.ucsc.edu) and Ensembl (http://www.ensebl.org). Using these methods, we were able to construct a fluid path of clones with a minimum tiling path of 16 BACs and 2 PACs (Figure 5) (Lucas et al. 2001). In this map several overlapping clones were included to provide greater coverage (Figure 5). Transcriptional orientation of the genes in the region was determined using the data from the HGP, sequencing of cosmids, and known overlap of genes, such as the 3' ends of the FLII and LLGLI genes (Campbell et al. 1997). Cosmid genomic clone DNA was isolated fi‘om previously mapped cosrnids (J uyal et al. 1996; Elsea et al. 1997) as well as from screening of gridded filters of the Los Alarnos flow sorted chromosome 17-specific cosmid library (Kallioniemi et al. 1994). At the time of publication, we were able to determine transcriptional orientation of 18 of the 29 expressed sequences mapping to the critical region (Lucas et al. 2001). The contig as published in 2001 (Lucas et al. 2001) contained 17 known genes, 12 ESTs, and 6 genomic markers (Figure 5). Analysis and characterization ESTs mapped to the SMS critical region Through the creation of the physical map of the SMS critical region we were able to identify numerous expressed sequences mapping to the region. These sequences were not only important in creation of the map, but because of their map location likely represented positional candidate genes for SMS. Clones for 13 of the ESTs were commercially obtained and plasmid DNA was isolated. The plasmid DNA was sequenced and database analysis of the DNA sequence was performed against online databases. Further, the DNA sequences of the ESTs were translated in all 6 flames and the amino acid sequences were examined to look for homologous genes and/or sequence motifs. Tissue expression patterns were then explored by probing commercially available multiple tissue northem (MTN) blots of adult and fetal tissues with the ESTs obtained. Northern analysis of EST DKFZp434A139Q2 is shown in Figure 6. The clone represents the 3' end of the human retinoic acid induced l (RAII) gene. The analysis shows an ~8.0 kb transcript in all tissues examined. Examination of all ESTs was performed as discussed above. Summary of the genes mapping within the SMS critical region as defined by the deletion carried by HOUl42-540 are described in Tables 2-4. Table 1 shows seven ESTs reported to map to the SMCR by our lab in 2001 (Lucas et al. 2001); those indicated in gray no 30 Figure 5. Transcription map of the ~1.5 MB SMS critical interval (adapted from Lucas at al., 2001). The SMS critical interval as of 2001 (Juyal et al., and Elsea et al., 1997) spanned between the two vertical hatch marks. This distance is roughly between the proximal and middle SMS-REPS. The contig is comprised of overlapping BAC and PAC clones which span the distance from the genomic marker D17S40 to the proximal end of the distal SMS-REP. Markers, ESTs, and known genes were mapped to the contig though a combination of PCR mapping, Southern hybridization to digested BAC and PAC DNA, and database analysis of draft human genome sequence. Overlap of some BAC/PAC sequence confirmed the position of clones in the absence of available markers. Refinement of the SMS critical region has been performed and will be discussed firrther in Chapter 3. Genes shown in blue no longer map within the SMS critical region. 31 .1 32 g 98-x“: o M 3 lo manage 2 C m o J n is. m. m 1 M / / _IV h l . B can?» ”Q su no Emmy .vnw 1w3nsro M 7025213 02 3 O 4 J 10_V23 5 14F 8 7 83MPLF1 962A2321R3 3:3 Y2P1PR7 S 7 8 WWI 8 PMlBl osm1 2TD61MSC1 6211I4SR 166 on” RST BWTKMEH 67 FWJMSOMSPaCDOJRJDmO7FPTPP Um 7 ADOR MlME LEMO TOLGHOL.L OlNLmSR H l _w»» mmlhumm lmslwwww»...mmmnmmmwmmmmfmmime «h 4 _ 111... 1111111 11...1:.WHH1...H.111111111111121 11.1 111 I 285ng 0.0:. Unmmwzfi 1|. UnwAOHo.I.I.I° coquoil U0www§wD OD} OqumuH e $835.»; 63523» i 035% a 3.33qu e333; OOHHHEO 1+: eowmszuo Toto-Or confine}; enmmmpirflrbl enumHHSrOrOrO uowwommwlllbl 538:3; .0359} O - won—Sena 33me I -EHOm UnsoHanOIUITnTIII - 1. no: I - 1 Name: 0 e w 5838.99 D -29 cue—vane I -Eamroa monsoon." UnwanMWM-Ou U0 3...: O - wow . Unwummfig aano canon no" 8:98am an» mzm N; 1 523311. . . . nu - Canaan 32.on ”Hgmonuncefi Unwmmqul I -5595 mono noose: entwmeITsaq _ 608.25 - E I 2 g m ‘3’ 3 ci 3 “a” 5 E s a .3 :I .2 J - a kb 9.. A A a: :2 a. 9.5—1.1‘F 2.4 I 1.35—— B-Actin B) Brain Lung Liver Kidney l.35-—* B-Actin Figure 6. Human RAII northern analysis. The ~2.0 kb insert from EST DKFZp434A139Q2 represents the 3' end of the RA]! gene. The insert was hybridized to A) adult and B) fetal multiple tissue northern blots from Clontech. An ~8.0 kb transcript is evident in all adult and fetal tissues. B-actin is included as an mRNA loading control for both blots. 33 Table l. ESTs mapped by Lucas et al. within the 2001 ~1.5 Mb critical interval which have been identified as known genes. EST amplimer(s) Unigene Tissue expression Protein motifs Known gene (GenBank accession) WI~13499 Hs.78582 2.2 kb transcript in all GTP-binding DRGZ (GenBank R41366) adult and fetal protein motif (GenBank tissues' X80754) D1782021 Hs.13434 1.8 kb transcript in all Mitochondrial ATPAFZ (GenBank AA281720) adult and fetal leader (GenBank tissues; highest in sequence AF052185) heart A003A44 Hs.8125 6.5 kb transcript in all VHS and GAT TOM1L2 (GenBank AA236905) Hs.12537 adult tissues, highest domains (GenBank stSG9692 in heart, brain, sk. NM_144678) (GenBank AF038192) muscle DKFZp434A139Q2 Hs.278684 8.0 kb transcript in all CAG repeat, RAII (GenBank AL133649) adult tissues nuclear (GenBank localization AY172 136) signal, PHD zinc finger domain 'NlB1041 Hs. 106359 5 lib transcript in all Member of RASDI (GenBank T16275) adult tissuesc Ras subfamily (GenBank , NM 016084) : WI-l 1472 Hs. 16614 1.6 kb transcript in Hydrolase NT5M (GenBank R72633) adult heart, brain, and domain (GenBank skeletal muscle” NM 020201) - stSGZ6124 Hs.84883 6.0 kb transcript in all KOG4807: M—RIP (interim '1 (GenBank AI570799) adult and fetal tissues domain from gene symbol) F -actin (GenBank binding NM_OlS 134) protein Note: Genes have been ordered proximal to distal and shading indicates gene is located outside of the current SMS critical interval (V langos et al., 2003). Protein motifs identified from NCBI conserved domain database and NCBI protein-protein BLAST alignments. “Results reported in Vlangos et al., 2000; orthem results similar to those reported in Rarnpazzo et al., 2000; cNorthern results similar to those reported in Tu and Wu, 1999. 34 Table 2. Novel ESTs mapped by the Elsea lab within the 2001 ~1.5 Mb critical interval. EST Amplimer Unigene Tissue expression Protein motifs (GenBank Accession) _ T78887 Hs. 187422 Clontech MTE blot showed 378 bp LINE] repeat (GenBank AA730163) expression in heart, GI tract, germ cell, lung, kidney, lacenta stSG8339 Clontech MTE blot 93 bp MIR repeat (GenBank H57290) showed faint expression in heart, GI tract, brain, lung, placenta, germ cell FL20308 Hs.356770 3.5 and 4.0 kb transcript in KOG4176: (GenBank BCO62339) all adult and fetal tissues uncharacterized examined conserved domain MGC3048 Hs.115437 4.4 kb and 4.0 transcript in KOG4635: domain (GenBank BC000636) adult heart, sk. muscle, found in vacuolar import kidney, pancreas and fetal and degradation proteins liver and lung; 4.0 kb transcript in adult and fetal brain, lung and placenta DKFZp586M1120 Hs.159068 Unigene reports 5.8 kb 43% similarity (140 aa (GenBank AL136926) transcript and ESTs from alignment) to sd322 several adult tissues protein homolog (GenProt $68209) FLJ23022 Hs.287717 Unigene reports SAGE tags none (GenBank from several adult tissues NM 025051) A006Y21 Hs.31652 Unigene reports ESTs from 322 bp Alu repeat; (GenBank H50830) several adult tissues 85.7% alignment to phospholipase D active site motif FLJ22382 Hs.46783 ESTs from several adult none ; (GenBank AK026035) tissues FLJ11532 Hs.296656 ESTs from brain and 78 bp Alu repeat ' (GenBank AK021594) embryo A004R11 Hs. 16899 2.4 kb transcript in adult and none . (GenBank T91728) fetal liver ‘ A006R41/FLJ10193 Hs.235195 6.0 kb transcript in adult Moderately similar to (GenBank A1500641) skeletal muscle and fetal Drosophila protein brain RH42446p (GenProt AAN71576) MGC13008 Hs.326732 ESTs from none (GenBank rhabdomyosarcoma NM 032686) Note: Genes have been ordered proximal to distal and shading indicates that this gene is localized outside of the current SMS critical interval (V langos et al. 2003). It remains a possibility that some of these unique ESTs can contains genomic contamination or may be cloning artifacts. Protein motifs were identified from NCBI conserved domain database and NCBI protein-protein BLAST alignments. 35 Table 3. ESTs recently mapped by other groups within the 2001 ~1.5 Mb critical interval. EST Unigene Tissue Expression Protein motifs Reference Amplimer (GenBank Accession) SMCR8 Hs.513986 2.8, 3.0, and 6.5 kb KOG3715: Bi et al. (Genbank AF467440) transcript in all adult domain found in (2002) and fetal tissues LST7 permease Golgi transport protein SMCR7 Hs.100448 2.4 and 3.4 kb KOG3963: motif Bi et a1. (GenBank AF467443) transcripts in all adult found in Mab—Zl- (2002) and fetal tissues, like cell fate predominantly in heart proteins and sk. muscle SMCR6 Hs.443639 RT—PCR demonstrated none Bi et al. (GenBank BF 51 1382) expression in all tissues (2002) SMCR5 Hs.352643 RT-PCR demonstrated none Bi et a1. (GenBank AF467442) expression in all tissues (2002) SMCR4 none RT-PCR demonstrated none Bi et al. (GenBank BG992883) expression in all adult (2002) tissues and fetus SMCR3 Hs.373 802 RT—PCR demonstrated none Bi et al. (GenBank AA609047) expression in all adult (2002) tissues and fetus SMCR2 none RT-PCR demonstrated none Bi et a1. expression in adult (2002) brain, and fetus SMCR9 - none. RT-PCR demonstrated wa PDZW. 'Bi et al.- 1. expression in all adult " "(20023 "g _ tissues and fetus ': i“ BED ' Flier-396533 3.8 kb transcript in all KOG3715: -' Nickersoniis (GenBank AFS 17523) adult tissues domain found in et al. ”2:. LST7 aa permease , (2002) - ii Golgi transport Followrng the publication of our physical and transcription map in 2001 (Lucas et al., 2001), several other genes were mapped to this region by other groups. Genes have been ordered proximal to distal and shading indicates that this gene is localized outside of the current SMS critical interval (Vlangos et al., 2003). identified from NCBI conserved domain database. 36 Domains were longer map within the critical region and will be discussed in Chapter 3. ESTs listed in Table 2 are additional novel ESTs we have since mapped to the SMCR. The current expression and bioinformatic information about these expressed sequences is included. Some of the sequences listed in Table 2 are novel and/or do not show banding patterns upon northern analysis. These sequences may be artifacts from EST cloning in the form of genomic sequence. It is also possible that the sequences are not expressed in tissues represented or are expressed at too low of a level to be detected on northern analysis. Further bioinformatic analysis may reveal more about these possible transcripts as the genomic sequence is polished. Additional groups have mapped EST sequences within the 1.5 Mb region represented by the deletion carried by HOU142-540. These sequences are shown in Table 3. Any of the EST sequences mapping within the SMS critical region may represent genes that play a role in modification or expression of the SMS phenotype. It is also possible that genes mapping outside the critical region may have an effect on the phenotype due to breakpoint location within the expressed sequence or disruption or deletion of yet to be discovered promoter or enhancer sequences. Though, the focus of this work is on genes mapping within the SMS critical region as they are most likely responsible for the characteristics making up the SMS phenotype. Summary of known candidate genes mapping within the SMS critical region Mapping of genes and ESTs to the SMS critical region provided us with numerous potential positional candidate genes for SMS. We were most interested in genes showing expression during fetal development or in brain tissues. Those genes 37 containing known protein motifs important in cell signaling, DNA binding, or protein-protein interaction domains were given priority. We believed genes with these characteristics were likely to be most susceptible to changes in dosage. Several genes mapping within the SMS critical region have been well characterized by others and are no longer candidates of high priority for SMS; these include TOP3A (Hanai et al. 1996; Fritz et al. 1997; Elsea et a1. 1998), FLII (Campbell et al. 1993; Chen et al. 1995; Campbell et a1. 1997; Campbell et al. 2002), MYOISA (Wang et al. 1998; Liang et al. 1999), SREBFI (Hua et al. 1995; Smith et al. 2002), PEMT 2 (Sesca et al. 1996; Vance 1996; Vance et al. 1997; Walkey et al. 1997), and COPS3 (Elsea et al. 1999; Potocki et al. 2000b; Yan et al. 2003). Further, gene targeting of mouse orthologs of the six genes listed above (Tap3a, F Iiih, Myal5, Srebfl, Pemt, and Caps3) show no phenotype in heterozygous mice (Table 4). This is in contrast to the report indicating an SMS like physical phenotype in mice with heterozygous deletion of the syntenic SMS region on mouse chromosome 11 (Walz et al. 2003). Combined, these data suggest that these six genes are not likely dosage- sensitive in the hemizygous state and are no longer high priority candidates for our study. The candidate genes of most interest to the lab during my tenure included LLGLI, DRGZ, ATPAF2, TOMILZ, and RAII . The contribution of these genes to the SMS phenotype was unknown at the beginning of this project. Studies of RAII, DRGZ, and T 0M1L2 will be discussed at length in chapters 4, 5, and 6, respectively. A summary of the two remaining candidate genes LLGLI and AT PAFZ is given below: 38 Table 4. SMS candidate ggne disruption in mouse'. Gene Publication Heterozygous Homozygous phenotype disrupted phenotype Pemt Walkey et al. No abnormal No abnormal phenotype on (1997) phenotype normal diet Srebfl Shimano et al. No abnormal No abnormal phenotype in live (1997) phenotype births, ~65% embryonic lethal My015 Probst et al. No abnormal Profound deafness and vestibular (1998) phenotype defects Fliih Campbell et al No abnormal Embryonic lethal, can be rescued (2002) phenotype with human FLII transgenically Tap3a Li and Wang No abnormal Embryonic lethal (1998) phenotype “Table is adapted from Vlangos et al., 2003. 39 Lethal giant larvae homolog 1 (LLGLI): LLGLI (NCBI Entrez Gene ID#3996; Unigene Hs.513983; OMIM 600966) is the human homolog of the Drosophila tumor suppressor gene, D-lethal (2) giant larvae (D-lgl). When disrupted, D-lgl can produce trunors in the irnaginal disks and abnormal transformation of the adult optic centers in larval brains. D-lgl was found to be associated with the cytoskeleton (Strand et al. 1995). In the initial description, human LLGLI was reportedly expressed in brain, kidney, and muscle, with little expression reported in heart and placenta (Strand et a1. 1995). Current electronic expression profiling of human ESTs in Unigene shows highest expression levels in stomach. Antibodies against LLGLI were able to co-irnmunoprecipitate LLGLI and non-muscle myosin heavy chain. This protein interaction is conserved in Drosophila (Strand et al. 1995). LLGLI and D-lgl are also associated with a serine kinase, which is able to specifically recognize and phosphorylate serine residues within the protein likely allowing for regulation (Strand et a1. 1995). LLGLI was first mapped to chromosome l7p11.2 with FISH and confirmed with Southern blotting (Strand et al. 1995). It is interesting that the 3' end of LLGLI overlaps with the 3' end of the FLII gene (Campbell et al. 1997) which helped in orientation of these genes during construction of our transcription map (Lucas et al. 2001). ATP synthase mitochondrial F1 complex assembly (A TPAFZ; formerly ATPIZ): Originally, AT PAF 2 (NCBI EntrezGene ID# 91647; Unigene Hs.13434; OMIM 608918) was investigated by our group as the novel EST arnplimer D17S2021 (Elsea et a1. 1997). The ATPAFZ gene is the human homolog of the yeast nuclear 40 gene Atp12p as determined via BLAST analysis (Lucas et al. 2001). The role of the AthZp gene is reportedly limited to ATP synthase assembly, and it is required to mediate formation of the Fla subunit of the yeast mitochondiral ATPase (Wang et al. 2000; Wang et al. 2001). We examined expression patterns of ATPAFZ via commercially available northern blots where a 1.8 kb transcript was noted in all tissues examined (Table 2). An equivalent function for the yeast and human homologs was proven by showing that transfection of the ATPAFZ cDNA into an inviable yeast atpaf? mutant rescues the phenotype (Wang et al. 2001). Partial deficiency of complex V of the oxidative phosphorylation system has been associated to recessive missense mutation of the ATPAFZ in one patient (De Meirleir et a1. 2004). Heterozygous carriers of the reported mutation appear normal (De Meirleir et al. 2004). It is unclear what percentage of gene function is lost in the reported patients with DNA changes. Complete abolishment of gene function is likely incompatible with life (De Meirleir et al. 2004); thus, the homozygous patient must have some gene function. It is interesting that the affected patient displays features consistent with SMS including infantile hypotonia and poor sucking, a prominent nasal bridge, and hypoplastic kidneys (De Meirleir et al. 2004). At this point it is unclear if haploinsufficiency of AT PAF 2 via hemizygous deletion has any role in the SMS phenotype. 41 SUMMARY Using genomic markers and ESTs mapping to chromosome 17p11.2, we were able to create a fluid genomic contig and transcription map across the 1.5 Mb SMS critical region. In constructing of the contig and transcription map we were able to identify many positional SMS candidate genes. Initial characterization of the candidate sequences led us to focus on five potential candidate genes. Additional in depth analysis of these genes will focus on identifying the cellular function of these genes, as well as any potential role they may play in generation of the SMS phenotype. 42 MATERIALS AND METHODS DNA isolation BAC and PAC DNA extrgztion: BAC and PAC clones were obtained from Research Genetics (now Invitrogen) and BAC/PAC Resources. Plasmid DNA was isolated from E. cali cultures using a modified Qiagen very low copy protocol created in our lab. Bacteria containing individual BACs or PACs were streaked on antibiotic selective LB agar plates. Single colonies were picked and grown with continuous shaking for 16 to 18 hours at 37°C in 2 ml of Luria-Bertani broth (LB) containing appropriate selective antibiotic (12.5 jig/ml chloramphenicol for BACs or 100 ug/ml kanamycin for PACs). Starter cultures were diluted 1/500 in 250 ml LB containing selective antibiotic and grown for 16 to 18 hours at 37°C while shaking. Bacteria were pelleted by spinning at 6000 x g for 15 minutes at 4°C. The supernatant was removed, followed by immediate resuspension of the pellet in 60 ml of buffer P1 (50 mM Tris-HCl, pH 8.0, 10 mM EDTA, pH 8.0, 0.1 mg/ml RNase A). Sixty milliliters of lysing buffer P2 (0.2 N NaOH, 1% sodium dodecyl sulfate{SDS}) were added to the cells and solutions were gently mixed via inversion to ensure complete cell lysis. Sixty milliliters of buffer P3 (3 M potassium acetate) were immediately added to the lysed solution and gently mixed. Tubes were placed on ice for thirty minutes to ensure complete precipitation of cellular debris. Following this incubation, the tubes were centrifuged at 4°C at 220,000 x g. Supernatant containing plasmid DNA was promptly filtered through filter paper (Whatrnan #4). The BAC/PAC DNA was precipitated by adding 0.7 volumes of room temperature isopropanol and centrifuged at 215,000 x g for 30 minutes at 4°C. The pelleted DNA was allowed to dry slightly 43 (inverted for 5 minutes) and then resuspended in 500 p1 sterile 1x TE buffer (10 mM Tris-HCl, pH 8.0, 1 mM EDTA). After the DNA was completely resuspended, 5 ml of buffer QBT (0.75 M NaCl and 50 mM MOPS buffer containing 15% isopropanol and 0.15% Triton X-100, pH 7.0) was added to the plasmid DNA. Resuspended DNA was applied to a buffer QBT equilibrated Qiagen Tip 500. The column was washed three times with 10 ml of buffer QC (1 M NaCl, 50 mM MOPS buffer, pH 7.0 containing 15% isopropanol). The BAC/PAC DNA was precipitated from the column by using 15 ml of buffer QF (1.25 M NaCl, 50 mM Tris, pH 8.5, containing 15% isopropanol) preheated to 65°C. The purified DNA was precipitated by adding 0.7 ml of room temperature isopropanol and centrifuged at 215,000 x g at 4°C for 30 minutes. The supernatant was decanted and the plasmid pellet was allowed to air dry for ~10 minutes. The pellet was resuspended in 200 pl sterile 1x TE. Purified BAC/PAC DNA was electrophoresed on a 1% TBE agarose gel and visualized under a UV transilluminator after staining with ethidium bromide. Finally, DNA quantity and quality was determined using a spectrophotometer. EST plasmid DNA isolation: EST clones representing markers mapping to chromosome 17p11.2 were obtained (Research Genetics, Incyte Genomics, the German Genome Project, or RIKEN) and grown on LB agar plates containing appropriate antibiotic for selection. Individual colonies were then picked and grown in 5 ml LB broth containing antibiotic. DNA was isolated from bacterial cultures using the Qiagen miniprep kit according to manufacturer’s instructions. After isolation plasmid DNA was electrophoresed on a 1% TBE agarose gel and visualized under a UV transilluminator after staining with ethidium bromide. DNA quantity and 44 quality was determined using a spectrophotometer. Southern analysis: Standard Southern analysis was used to map markers to BACs/PACs within the contig and to the hybrid mapping panel. BAC or PAC DNA (6 pg), human genomic DNA (9 pg), or rodent-human hybrid DNA (15 pg) was digested with 4 U/pg of EcoRI and 2.5 mM spermidine for ~16-18 hours at 37°C. Digested DNAs were electrophoresed on 1% TAE agarose gels (TAE; 0.04 M Tris- acetate, 1 mM EDTA) with continuous buffer recirculation at room temperature. Gels were depurinated in two gel volumes of 0.25 N HCl followed by denaturation in 2 gel volumes of 0.4 N NaOH. DNA was transferred to an Amersham Hybond-N+ nylon membrane via wicking with 10x SSC (SSC; 20x stock solution is 3 M NaCl, 0.3 M Na-citrate). After transfer, DNA was UV-crosslinked to the membrane using a Stratalinker (Stratagene). Membranes were prehybridized and hybridized in a solution of 1 M NaCl, 1% SDS, 10% dextran sulfate, and 0.1 mg/ml herring sperm DNA at 65°C with rotation in a hybridization oven. All DNA probes (purified PCR products or plasmid inserts) were labeled with 32P-dCTP using an Amersham Rediprime H kit. Unincorporated nucleotides were removed with a sperrnine and herring sperm DNA precipitation followed by radioactivity quantification in a Brinkrnan scintillation counter. During preassociation, ~106 cpm/ml of probe was annealed to 0.25 mg/ml placental DNA to mask repeat sequences and then hybridized to the membrane for ~l6-18 hours. Blots were washed for 20-30 minutes in 0.1x SSC, 0.1% SDS at room temperature, followed by a stringent wash in preheated 0.1x SSC, 0.1% SDS at 65°C. The blots were exposed to X-ray film with two intensifying 45 screens at -80°C for 1-5 days before developing. Database searches: Extensive information about the genes and ESTs in the region was obtained through the National Center for Biotechnology Information (NCBI) website (http:/www.ncbi.nlm.nih.gov), including Unigene, the EST database (dbEST), and the STS database (dbSTS). Draft assembly of the hmnan genome project was found at the UCSC genome bioinfonnatics site (http://www.genome.ucsc.edu) and through Project ENSEMBL (http://www.ensembl.org). Supplementary marker information was derived from the Genome Database (http://www.gdb.org). Sequence alignments and unfinished high- throughput genome sequence BLAST searches were conducted through the Baylor College of Medicine Search Launcher (http://searchlauncher.bcm.tmc.edu) or NCBI. Conserved sequence domains were identified through databases at NCBI (http://www.ncbi.nlm.nih.gov/Structure/cdd/wrpsb.cgi). PCR: The polymerase chain reaction (PCR) was performed in 25 p1 volumes. Amplification of 50—100 ng of template DNA was performed using 1 U of Taq polymerase in a cocktail consisting of 0.8 pM of each primer, 0.25 mM dNTPs, and 1x PCR buffer (10x buffer contains 100mM Tris-HCl pH 8.3, 15 mM MgC12, 500 mM KCl, 0.01% gelatin). Amplification was performed in an Applied Biosystems (ABI) or MJ Research therrnocycler at the following conditions, unless otherwise noted: initial denature at 94°C for 4 minutes, followed by 30 cycles of 94°C for 1 minute, 55°C for 1 minute, and 72°C for 3 minutes, followed by a final extension of 46 72°C for 10 minutes. PCR products were then electrophoresed on 1% or 2% TBE agarose gels, stained with ethidium bromide, and visualized on a UV transilluminator. Northern analysis: The expression of various ESTs was visualized using human multiple tissue, fetal, and brain 11 northern blots purchased from Clontech. Prehybridizations and hybridizations were performed in 5x SSPE (0.75 M Nacl, 0.05 M NaHzPO4, 5 mM EDTA), 10x Denhardt’s solution (0.2% BSA, 0.2% ficoll, 0.2% polyvinylpyrrolidone), 100 pg/ml herring sperm DNA, 2.0% SDS, and 50% deionized formarnide at 42°C with rotation in a hybridization oven. Radiolabeled probes were created as described for Southern analysis. Approximately 106 cpm/ml of probes was denatured and added to prehybridized filters. Filters were hybridized for ~18 to 20 hours at 42°C, followed by washing in 2x SSC, 0.05% SDS for ~30 minutes at room temperature with 1 to 2 changes of wash solution. Filters were then transferred to a wash of 0.1x SSC, 0.1% SDS for ~30-40 minutes at 50°C. The blots were exposed to X-ray film between two intensifying screens at -80°C for 2-5 days before developing. 47 Chapter 111 Analysis of 17p11.2 deletions using fluorescent in sitn hybridization BACKGROUND Until the early 19903 detection of chromosomal anomalies, including microdeletion disorders, depended on staining and analysis of the entire chromosome complement. This was usually accomplished by techniques such as Geimsa staining, also known as G-banding. The chromosomes were stained with a mix of the toxic dyes methylene azure and eosin, which bind to the phosphate groups of DNA. The regions of the chromosome where the DNA is more densely packaged (heterochromatin) stains darker than regions where the DNA is more loosely packaged (euchromatin). Thus, staining reveals a recognizable pattern of bands along the chromosomes. Using this standard technique, the entire genome usually shows ~350 distinct bands. Analysis of the number of total chromosomes and the banding patterns uncovers the chromosomal abnormalities. This standard karyotype analysis works very well for detection of aneusomy (i.e. Down syndrome, trisomy 21) and large segmental anomalies (i.e. translocations, inversions, deletions, or duplications) and is still routinely used today. Though the technique is very reliable, there are limitations. Increasing the ntunber of bands to the 550 or 850 level by preparing chromosomes of increased metaphase length helps intensify the sensitivity of the test. Even so, small anomalies involving less than ~1.5 Mb of DNA cannot be unambiguously detected. Technological advances using fluorescent dyes have aided greatly in the detection of these smaller anomalies. 48 The use of fluorescent in situ hybridization (FISH) for gene mapping dates back to the late 19708 when it was used to map the human globin genes to the long arms of chromosomes 4 and 5 (Cheung et al. 1977). It wasn’t until 1986 that FISH was reported as a use for detecting chromosomal anomalies using human genomic DNA as a probe (Pinkel et al. 1986). DNA probes made of genomic DNA representing known loci were labeled with nucleotides conjugated with haptens such as biotin or digoxigenin. The labeled nucleotides are incorporated, at a defined density, into the nucleic acid probes by DNA polymerases. The labeled probes were then hybridized to denatured metaphase chromosomes much in the way radiolabeled probes are hybridized to Southern blots. The labeled probes are detected with fluorescently labeled antibodies to the hapten, and viewed using a properly equipped fluorescent microscope. The number of fluorescent signals detected is a direct measure of DNA copy number (Figure 7). If a microdeletion syndrome is detected via karyotype, the deletion is usually confirmed via FISH. Additionally, FISH may be ordered directly without a prior karyotype if a specific chromosomal anomaly is suspected. The current guidelines for diagnosis of microdeletion syndromes, including SMS, recommend testing and/or confirmation via FISH (ACMG/ASHG positional statement. 2000). Technology has advanced to make FISH experiments faster and cheaper. Today, the flourochrome is attached directly to the nucleotide. This negates the need for secondary antibody detection. The probes are more robust and emit a greater amount of light fi'om the same excitation input. Initial FISH experiments in the Elsea Laboratory were performed with indirect labeling methods, but we have since 49 ‘— nol tIL‘I not do! l 1101 \It‘I Figure 7. Examples of not deleted and deleted FISH experiments. Metaphase chromosomes were hybridized with fluorescently labeled BAC probes. Green signals are indicative of clones mapping within the SMS critical region. The red signals represent a BAC mapping to l7q and is used as a control. A) Green and red signals of both chromosomes 17 indicate that this patient is not deleted for the test probe. B) In this patient sample the green test probe is only present on one chromosome 17, while the red control probe is present on both chromosomes 17. This indicates that this patient is deleted for this test probe. 50 switched to using directly labeled probes. All FISH experiments described herein have been performed with directly labeled FISH probes. FISH is not only a wonderful diagnostic tool but also a powerful tool for research of microdeletion syndromes. Using probes mapping in the region of the disorder, it is possible to determine which DNA sequences are absent or present. SMS patients displaying the full phenotype of the disorder may have different sized deletions (Trask et al. 1996). The smallest region of overlap of the deletions carried by these patients defines the syndrome critical region. The genes mapping within this region are the positional candidate genes for the disorder. This method has been used successfiilly in many microdeletion disorders including Smith-Magenis syndrome (Bettio et al. 1995; Gong et al. 1996; Juyal et al. 1996; Elsea et a1. 1997). RATIONALE In studying deletions of chromosome 17p11.2 associated with Smith-Magenis syndrome, we were most interested in patients with deletions overlapping the SMS critical region (described in Chapters 1 and 2). Using FISH analysis on SMS patient samples we identified deletions that overlapped the smallest SMS deletion known at the time. Detailed analysis of the unusual deletions carried by these patients allowed us to reduce the size of the SMS critical region and the number of candidate genes. The work presented in this chapter resulted in the following publications; (V langos et al. 2003; Vlangos et al. 2004). 51 RESULTS FISH detection of 17p11.2 deletions and refinement of the SMS critical region Using FISH, we characterized the 17p11.2 deletion size in 22 patient samples. Clinical analysis of the patients was obtained from patient medical records, and from a questionnaire developed in house (Appendix A). Because the common SMS deletion was reported to occur in >90% of SMS patients we expected that the majority of our patient cohort would harbor the common SMS deletion (Chen et a1. 1997; Lupski 1998; Potocki et al. 2000b; Bi et al. 2002; Shaw et al. 2002). Thorough FISH analysis using probes mapping along chromosome l7p11.2 revealed that 8 of our 22 patient samples carried the common SMS deletion (Figure 8). The number of patients with the SMS common deletion was significantly lower than we expected. Four of the 22 patient samples analyzed showed deletions that were uncommon, but that did not overlap the 1.5 Mb SMS critical region (Figure 8). These patients all showed the complete spectrum of the SMS phenotype. The unusual deletions these patients carry do give further insight into the mechanisms causing 17p11.2 deletions associated with SMS, as the deletions are not due to NAHR between the SMS-REP sequences. These data indicate that the true incidence of the SMS common deletion may be lower than < 90% previously reported (Chen et al. 1997; Lupski 1998; Potocki et al. 2000b; Bi et al. 2002; Shaw et al. 2002). Since 1997 the SMS critical region has been defined by a single l7p11.2 deletion spanning ~1.5 Mb of DNA (Elsea et al. 1997). In our analysis of l7p11.2 deletions associated with SMS, we were very interested in finding patients displaying the SMS phenotype whose deletions overlapped the 1.5 Mb carried by HOU142-540. 52 Through thorough FISH analysis on the patient samples received, we found three patients who carry deletions overlapping with the deletion carried by patient HOUl42-540 (Figure 8). SMS135 is the first patient referred to us whose deletion helped narrow the smallest region of overlapping deletion on chromosome 17p11.2 associated with SMS. SMSIBS is a 3 year, 8 month-old male of Filipino descent who displays the characteristic SMS phenotype (Figure 9A, Table 5). FISH with clones mapping along chromosome 17p11.2 allowed for identification of his deletion breakpoints. FISH data indicate that the proximal breakpoint of this deletion occurs at or near the proximal SMS-REP. The distal breakpoint, however, occurs within the current SMS critical interval (Figure 8). FISH analysis indicated that this patient is not deleted for genomic clones on l7p11.2 from the SMS-REPD through BAC RP11-335P17 containing the COPS3 gene (Figure 8). The patient is deleted for BAC CTD-40123, containing the PEMT2 gene (Figure 8). Finer mapping with cosmids was performed using clones from the chromosome 17 library (LA17NC01) (Kallioniemi et al. 1994) for RASDI (c107A2) and PEMT2 (c26ElO) (Figure 10). The patient is deleted for c26E10 and not deleted for c107A2 which places the distal breakpoint in the ~30 kb between these cosmids. Two additional patient samples, SMS182 and M2359 (Figure 9A and 9B, Table 5), were sent to us for analysis because high resolution karyotyping showed possible interstitial deletion of chromosome 17p11.2 though diagnostic testing for SMS with commercially available FISH probes was negative in both cases. We strongly suspected SMS in both patients, and performed FISH analysis using probes 53 Figure 8. Summary of 17p11.2 deletions identified in this study. Probes used in FISH analysis and their respective locus are indicated in the table to the left. Deletions found in the course of this study are indicated by the ideograms. Hatched boxes indicated DNA that is not deleted. Open boxes indicated DNA that is deleted. The critical deletion as reported in J uyal et al., 1996 and Elsea et al., 1997 is indicated for reference. The number of patients determined to carry each deletion is listed under each ideogram. 54 C .9 ad _d_) O 'O .5 'C O Locus Probe PMP22 bc76406 FOXO3B bc815l9 srs-we7727 bc529l10 REP-D bc198H15 TNFRSF13B pc48J14 cCI17-638 bc989815 D17S740 bc367GQ COPS3 bc335P17 PEMT2 bc40l23 RAI1 pc253P07 FLII bc347A12 FLII c62F2 SHMT1 bc219k23 017329 bc457L16 MFAP4 bc187M2 ULK2 b07807 D17S446 c105G12 01782201 bc746M1 Common deletion 55 Uncommon deletions V F lJl i 1 Lil mapping along l7p11.2. The proximal breakpoints for both SMS182 and M2359 map within the region between the FL]! and LLGLI genes (Figures 8 and 10). Distally, the deletion carried by SMS182 extends into 17p12 near the ADORA2B gene. The distal breakpoint for M2359 is located near the genomic marker STS-W67727 in 17p11.2 (Figures 8 and 10). Using the proximal deletion breakpoint for these two patients and the distal breakpoint for patient SMS135, we determined that the smallest shared region of deletion of studied SMS patients is ~700 kb (Figure 10) and currently contains 11 known genes (V langos et al. 2004). Commercially available diagnostic FISH probes for SMS will not detect all cases Both SMS182 and M2359 display characteristics typical of the SMS phenotype for their respective ages. Both patients tested negative for SMS using commercially available SMS FISH probes. Utilizing genomic probes mapping along 17p11.2 for FISH, we were able to determine that both SMSl82 and M2359 carry interstitial deletions of 17p11.2 and that both patients are deleted for the RA]! gene. As will be discussed further in Chapter 4, patients carrying mutations in the RA]! gene mapping within the SMS critical region display features consistent with the SMS phenotype (Slager et al. 2003; Bi et al. 2004; Girirajan et al. 2005). Characteristics that are not seen patients with mutation in the RAII gene include short stature, infantile hypotonia, and cardiac and/or renal anomalies. These features occur at a greater frequency in SMS patients who harbor a 17p11.2 deletion than in the general population and thus, are included in the SMS phenotype. It is possible that 56 Figure 9. SMS patients with unusual deletions used to refine the smallest region of overlapping deletion in SMS. The three patients who carry deletions allowing for refinement of the SRO in SMS all display typical SMS facial features; A) SMS135 at age 3 years, 8 months, B) SMS182 at age 19 months, and C) M2359 at age 10 years. D) FISH analysis of metaphase chromosomes from SMS135 indicate he is not deleted for bc3073J20 as green test signal is only detected on one chromosome 17. This probe is deleted original SMS critical interval described be Elsea et al., 1997. E) Metaphase chromosomes from SMSl35 indicate he is deleted for the pc253P07 probe containing the RA]! gene. F) Metaphase chromosomes from M2359 indicate he is also deleted for the pc253P07 probe. Both SMS182 and M2359 both tested negative for SMS using currently available commercially available FISH probes. 57 58 Figure 10. Location of SMS FISH probes in relation to l7p11.2 deletions. The genomic region of chromosome 17p11.2 is represented and indicates the location of the three SMS-REPS, as well as genes and markers mapping to this region. The red hatched bar indicates the smallest region of DNA that is deleted in all SMS patients carrying a 17p11.2 deletion, the open/closed bars indicate deletions of SMS patients in this study. The open portion of the bars represents deleted DNA, while the closed portion represents non-deleted DNA. The green bars represent the currently available FISH probes from Vysis and Cytocell. The red bar represents the PAC RP1-253P07 used in our FISH experiments. The purple bars represent the previously available FISH probes from Oncor. The blue bars indicate experimental test probes used in this study. This figure is not to scale. 59 7 2 WW. w n L we. 7mwm 0 mm Emma... H u so Dru...) M .5 ”3-3:.ch 0.52 U. emmmm I gait... SI '2: _-~ _ 20m. 033.. Income .1 9 w 8 .l 2 8 fi m S S 7 7 7 l ' all 1 s w 0 0 0 . .. ”11......3/ . l I <5? mzm Edda I U _ ammo 3030 I980: mgm Edda mZ—mBm 9323 I 38% ll IOCENGS l WAILSEIV. I also»: 8E8 it Have I 00389.93 wove I W333: b3 . I 988265 enodaieo Samoa “EB—ego 60 Table 5. Phenotypic comparison of SMS patients with 17p deletions'I SMS126 HOU142 SMS135 SMS182 M2359 Cormnon -540 Unusual Unusual Unusual SMS Smallest 17p1 1.2 17p1 1.2 17p1 1.2 deletion SMS deletion deletion deletion deletion to date Features common in >75% of SMS Pts. Craniofacial/Skeletal Brachycephaly - - + + + Midface hypoplasia + + + + + Progrthism (relative to age) + + N N + Tented upper lip + + + + + Broad, square face + - + + + Brachydactyly + - + + + Short stature + + - - Otol_a_rmgologic abnormalities Chronic ear infections/PE tubes + + N + + Hoarse, deep voice + + N N + Neurological Mental retardation + + + + + jpeech delay + + + + + Motor delay + - + + + Infantile hypotonia + - + + + Sleep disturbance + + + + + Behavioral Self-hugginghind wringing + + + N + Onychotillomania + + N N - Polyembolokoilorrrania + + N N + Head banging / face slapping + + N + + Hmd-bifigg + + N N + Attention seeking + + N N + Features common in 50 - 75% of SMS patients Hearing 1088 + + - + - Myopia + + - - - Strabismus - + + + - Iris abnormalities - + - - .- Synophrys - + - + + Scoliosis + - + N + Features common in < 50% of SMS patients Cardiovascular anomalies + + - - - Renal anomalies - + - - - Seizures - + - + - Cleft lip/palate - - - - - Gender/Age at evaluation Female Male Male Male Male 41y 10y, 14y, 3y 8m 14m, 2y 10y 15y ' Modified from the Gene-Reviews website (www.genereviews.com) and Slager et al., (2003) Note: +, feature present; -, feature not present; and N, feature not evaluated/ too young to evaluate 61 haploinsufficiency of RAII may also cause these characteristics; however, we have not yet identified patients with RA]! mutations who display these features. It is also possible that haploinsufficiency of another gene(s) mapping to l7p11.2 is responsible for these characteristics. Thus, studies of patients with unusual deletions will continue in order to narrow SMS region and the number of possible candidate genes to further understand genotype:phenotype correlation in SMS. Narrowing of critical region in microdeletion syndromes is common and can greatly aid in the search for causative genes as has been seen in Rubinstein-Taybi syndrome [del(16)(pl3.3)], due to haploinsufficiency of CREBBP (Petrij et al. 1995), and Alagille syndrome [del(20)(p12)] due to haploinsufficiency of JAG] (Li et al. 1997b; Oda et al. 1997b). As the critical regions are further reduced and the causative genes for the microdeletion syndromes are located, it is important that FISH probes are moved to represent these causative genes. Clinical FISH for Alagille syndrome associated deletions is now conducted with a probe containing the entire JAGI gene, while clinical FISH for Rubinstein-Taybi associated deletions is conducted with a contig of five cosrrrids covering the CREBBP gene (http://www.genereviews.org). It is important to note that so far, all SMS patients who carry a 17p11.2 deletion are deleted for the RAII gene (Figures 8 and 10). Currently available commercial FISH probes do not contain RAII . We have shown here that these currently available commercial FISH probes from Vysis Inc. (A subsidiary of Abbot Laboratories) and Cytocell Technologies will not detect all cases of SMS with a l7p11.2 deletion. We suggest that all future diagnostic FISH tests be performed with 62 probes containing the RAII gene. This will require that companies offering commercial FISH probes re-evaluate the location of their probes based on current data. Certain patients suspected to have SMS who previously tested negative with currently available FISH probes (i.e. those containing FLII or D17S29) may need to be re-tested with a probe containing RAJ], but we strongly recommend careful consideration of the SMS phenotype by thorough clinical re-evaluation before costly re-testing is pursued. It is also important to note that patients tested with the Oncor D178258 FISH probe, which is no longer commercially available, do not need to be re-tested by FISH. The D178258 probe is a cosmid, and the genetic marker D17S258 is located within an intron of the RAII gene (Figure 10). Some patients may display the typical SMS phenotype yet still test negative with FISH probes containing the RA]! gene. These patients may be candidates for further testing and sequencing of the RAII gene as will be discussed in Chapter 4. The SMS common deletion occurs less frequently than reported The deletion seen with greatest fiequency, known as the common deletion, is ~3.5 Mb and was been reported to occur in 90% - 95% of SMS patients (Chen et al. 1997; Lupski 1998; Potocki et al. 2000b; Bi et al. 2002; Shaw et al. 2002). The remaining ~5% are reported to be deletions of differing sizes, though size distribution, larger or smaller, has not been reported. During our FISH analysis we noticed that unusual deletions were occurring more frequently than expected. We reviewed the deletions of patients reported in the literature in addition to those reported here (Table 6). To date, including the 16 new SMS patients with l7p11.2 63 deletions reported here, there are a total of 85 reported SMS patient deletions fully characterized (Juyal et al. 1996; Chen et al. 1997; Yang et al. 1997; Potocki et al. 2000b; Bi et al. 2002; Park et al. 2002). The common deletion was reported as either the presence of a junction fragment on a PF GE Southern blot resulting from homologous recombination between SMS-REPP and SMS-REPD (Chen et al. 1997) or by FISH analysis. The common deletion has been detected in 60/85 (70.59%) SMS deletions, while unusual deletions have been found in 25/85 (29.41%) SMS deletions (Table 6). Approximately 30% of SMS deletions are likely mediated by other sequences, including the middle SMS-REP and/or other LCR elements, or the location of the SMS region within the pericentromeric region on the chromosome. An analysis of the August 2001 release of the human genome sequence discovered 169 sequences flanked by similar duplications of which only 24 are known to be associated with human disease (Bailey et al. 2001). It is thought that 5—10% of the human genome is duplicated (Ji et al. 2000) leaving many repetitive sequences uncharacterized. Newly described LCR elements have been characterized which flank the SMS-REP sequences, and these newly characterized LCRs are postulated to mediate recombination in unusual deletions (Park et al. 2002). The data presented here firrther suggest that LCRs yet to be characterized mediate recombination at a much higher rate than previously thought. Additionally, many microdeletion syndromes occur in the pericentromeric region of the chromosome (Williams syndrome del(7)(q11.2), DiGeorge syndrome del(22)(q11.2), Smith-Magenis syndrome del(17)(pll.2), NFl del(17)(q11.2), and Prader-Willi/Angelman syndromes del(15)(q11-q13)). The location of these syndromes in the pericentromeric region of the chromosome may also facilitate the homologous recombination leading to unusually sized deletions. SUMMARY Analysis of SMS patient samples has allowed us to narrow the smallest region of overlapping deletion of chromosome l7p11.2 associated with Smith-Magenis syndrome from 1.5 Mb to ~700 kb. This region contains the RA]! gene, which likely causes the majority of the SMS phenotype when in the hemizygous/haploinsufficient state. Currently available commercial FISH probes for SMS do no contain the RA]! gene, and not all SMS patients with 17p11.2 deletions are deleted for the currently available commercial FISH probes. Thus, not all SMS patients with deletions will be diagnosed with these probes. This will require that companies offering commercial FISH probes re-evaluate the location of their probes based on current data. Further, we have determined that the ~3.5Mb common SMS deletion occurs less frequently then the >90% reported. The common deletion occurs in ~70% of SMS patients. Other newly described, or undiscovered repetitive sequences likely contribute to the recombination of chromosome 17p11.2 leading to SMS. 65 Table 6. Frequency and type of SMS patient deletions‘. Publication Total # Common Deletion other completeb Deletion than common Juyal et al. 59 50 9 (1996) Yang et al. 1 0 l (1997) Chen et al., 2 2 0 (1997) Park et al., 1 0 1 (1998) Potocki et 4 0 4 al., (2000) Bi et al., 2 0 2 (2002) Vlangos et 11 7 4 al., (2003) This Report 5 l 4 Total 85 60/85 (70.6%) 25/85 (29.4%) ‘ Modified from Vlangos et al., 2003 b Total number complete represents fully characterized deletions. If patients have multiple reports, they are counted only once. 66 MATERIALS AND METHODS Patient and sample ascertainment: Contact between potential research participants was established between the patient’s guardian and/or medical care provider and the project Principal Investigator (Dr. Sarah H. Elsea, Ph.D.). Research samples and completed questionnaire (see appendix A) were collected in accordance with Institutional Review Board approved protocols at Michigan State University, Virginia Commonwealth University, and The National Institutes of Health. Peripheral blood samples were collected and shipped to the Elsea laboratory for preparation of transformed lymphoblastoid cell lines, metaphase chromosomes, and genomic DNA. Clinical descriptions afSMS135, SMS182, and M2359: Patient SMS135: SMS135 is a male of Filipino descent evaluated at age 3 years, 8 months by collaborator Dwight Yim, M.D. SMS135 displays the common SMS facial features including a distinct tented upper lip, brachycephaly, and midface hypoplasia (Figure 9A, Table 5). SMS135 also has speech and motor delay, short stature, brachydactyly, and was hyptonic as an infant. He is mentally retarded, has stereotypic self-hugging behavior, and experiences sleep disturbance common in SMS patients. SMS135 also has myopia and scoliosis, which are seen in < 50% of SMS patients. SMS135 has not yet developed self-injurious behaviors, though these may develop with further aging. Commercial FISH testing was positive for SMS. The patient was referred because of the family’s interest in SMS research. Patient SMSlSZ: SMSlS2 is healthy, non-verbal male evaluated at 14 months 67 (Figure 9B) and again at 2 years (Table 5). At age two years his height was 83.82 cm (15%ile), weight 12.93 kg (60%ile), head circumference 47.94 cm (25%ile), and BMI 18.41 (88%ile). He displays the common physical and neurological SMS characteristics expected for his age (Fig. 9B and Table 5). The parents of SMS182 also reported that he developed significant sleep disturbance at ~18 months of age, and head-banging and face slapping upon becoming frustrated at age 3. He displays some features seen less commonly in SMS, including hearing loss, strabismus, synophrys, and seizures (Table 5). High-resolution karyotypes at two different facilities showed an interstitial deletion of one chromosome 17p11.2. Commercial FISH testing, performed using the Vysis SMS probe, was negative (Fig. 13). Patient M2359: Evaluated by colleagues at NIH and in Australia, M2359 (Figure 9C, Table 5) is a male evaluated at 10 years of age with height 144.15 cm (90%ile), weight 43.4 kg (97%ile), head circumference 55.5 cm (90%ile), and BMI 20.89 (90- 95%ile). He has nearly all features most common to SMS, including typical craniofacial anomalies, speech and motor delay, mental retardation, and infantile hypotonia. In addition, he has synophrys, scoliosis, and bifid uvula. He displays a majority of the characteristic behaviors seen in SMS patients including self-hugging, polyembolokoilomarria, head banging, hand biting, and attention seeking, and he has evidence of nail damage, although he does not yet display nail yanking as is often seen in older SMS patients (Finucane et al. 2001). Significant sleep disturbance was reported by parents and validated by actigraphy. He does not have short stature or cardiac/renal anomalies. A high-resolution karyotype of M2359 showed a suspected 68 deletion of one chromosome 17p11.2 region. FISH was performed with SMS probes from Vysis and Cytocell, but both tests were negative. Isolation of metaphase chromosomes: Two hundred and fifty microliters of whole blood collected in a sodium heparin Vacutainer was inoculated in 5 ml of RPMI media (Gibco) supplemented with 10% fetal bovine serum and 1x antibiotic/antimycotic solution (Gibco). Fifty nricroliters of the mitogen phytohemaggultinin (PHA) were added to a final concentration of 1 x, tubes were mixed well and incubated at 37°C under 5% C02 for 72 hours with gentle mixing at least once per 24 hours. After 72 hours incubation, synchronization of the cell cycle was started by the addition of 10 pl of 50 pM methotrexate. After 16—18 hours incubation at 37°C under 5% C02, 50 pl of 1 mM thymidine was added. Cells were incubated another 4 hours at 37°C under 5% C02. Harvest of synchronized cells was started by the addition of 25 pl of 10 pg/ml colchicine. Cells were incubated 30 minutes at 37°C under 5% C02 before centrifugation at room temperature for 8 minutes at 1,200 rpm. Supernatant was discarded, followed immediately by the addition of 6 ml of 75 mM KCl. Cells were incubated at room temperature for 15 rrrinutes before the addition of 10—12 drops of ice cold fixative (3:1 methanolzacetic acid) with a Pasteur pipette. Tubes were immediately mixed and centrifiiged as above. All but 0.5 ml of the supernatant was removed, and cells were resuspended in 1 ml fixative and gently mixed. Five milliliters of additional fixative was immediately added and tubes were mixed and centrifuged as above. After centrifugation, the supernatant was completely removed and cells were resuspended 69 in 5 ml of fixative. Centrifugation and washing with fixative was repeated 2 additional times. After final centrifugation, the pellet was resuspended in ~4 ml fixative and incubated overnight at 4°C. After incubation metaphase chromosome spreads were prepared on glass slides using standard cytogentic methods. Fluorescent in situ hybridization: Probes for FISH were chosen based on mapping information from our recently completed SMS critical interval contig, which was constructed with large insert bacterial artificial chromosomes (BACs) and P1 artificial chromosomes (PACs). Additional probes (BACs, PACs, and cosnrids) were obtained using mapping data obtained during construction of our conti g, and from online databases, including the Genome Browser at UCSC (http://www.genome.ucsc.edu) and GenBank at NCBI (http://www.ncbi.nlm.nih.gov). FISH probes were created using BAC, PAC, or cosmid DNA isolated as described in Chapter 2 and labeled by using a commercially available nick translation kit to incorporate Spectrum Green or Spectrum Orange dUTP by following manufacturer instructions (Vysis Inc.). Metaphase chromosomes were prepared for hybridization by incubating at 37°C in 2x SSC for 30 rrrinutes. Slides were dehydrated by placing in an ethanol series (70%, 80%, 95%, and 100%, two minutes each) and allowed to air dry. Chromosomes were then denatured at 72°C for two minutes in a solution of 70% deionized fonnanride and 2 x SSC at pH 7.0 and immediately dehydrated in ethanol as above. Probe DNA (100 ng BAC/PAC or 180 ng cosmid) was precipitated in the presence of 1 pg COT-1 DNA and 2 pg human placental DNA by adding 0.1 volume of 3 M sodium acetate and 2.5 volumes 100% ethanol. The probes were placed at -80°C for 15 minutes then 70 immediately centrifuged at 4°C at > 12,000 x g for 30 minutes. The supernatant was discarded and the precipitated probe was dried to completion in a SpeedVac. The probe was then resuspended in 3 pl H20 and 7 pL hybrisol VII (Oncor Inc.). The probes were denatured for five minutes at 72°C and hybridized to chromosomes at 37°C for 14—16 hours. Slides were washed per manufacturer recommendations (Vysis Inc.) and counterstained using Vectashield antifade with DAPI (Vector Labs). Analysis of the FISH experiments was canied out on a Zeiss Axioplan2 microscope and photographed with a Harnarnatsu black and white camera using Zeiss AxioVision software version 2.0 (Carl Zeiss Inc). 71 Chapter IV Analysis of Smith-Magenis syndrome patients without a FISH detectable l7p11.2 deletion BACKGROUND AND RATIONALE Analysis of patient deletion sizes by FISH reduced the SMS critical region (SMCR) from 1.5 Mb to ~700 kb. This narrowing of the SMCR greatly reduced the number of candidate genes fi'om >30 to 11. However, of 22 patient samples analyzed using FISH, we were unable to detect a 17p11.2 deletion in six samples even though thorough clinical analysis of these patients indicated a strong suspicion of SMS. We believed that these patients indeed had SMS and hypothesized that there may be dominant mutations in a gene within the SMS critical region that could cause SMS. We therefore undertook direct sequencing of three SMS candidate genes on samples from patients without a detectable SMS deletion. Sequencing was performed on exonic DNA sequence from the RAS-dexamethasone induced 1 (RASDI ) gene, the developmentally regulated GTP-binding protein 2 (DRGZ) gene, and the retinoic acid induced l (RAII) gene. In our investigation of these 3 genes, we found dominant frameshift mutations in the RA]! gene in 4 of the 6 patients samples tested resulting in two publications from the lab (Slager et al. 2003; Girirajan et al. 2005). 72 RESULTS Mutation analysis in SMS patients without a FISH detectable deletion As described in Chapter 3, thorough FISH analysis was performed on metaphase chromosomes from suspected SMS patients. In six patient samples we were unable to detect a deletion of chromosome 17p11.2. Though FISH can detect smaller deletions than G-banding, it does have limitations. Standard FISH analysis will not detect deletions smaller than ~40 kb. Nor will FISH detect small chromosomal anomalies (i.e. inversions or translocations) occurring intrachromosomally within the same sub-band, though these would be extremely rare. The clinical description of many of our patient samples without FISH detectable deletions strongly suggested Smith-Magenis syndrome. The patients displayed craniofacial abnormalities, sleep disturbances and characteristics behaviors, including onychotillomania, polyembolokoilomania, spasmodic self-hugging, and explosive and aggressive episodes (Table 7). Because of these features, we decided that firrther analysis of individual genes within the SMS critical region using these patient samples was warranted. We theorized that mutations in a gene, or genes, mapping within the SMS critical region might cause the phenotype seen in these patients. We started this study by systematically sequencing three candidate genes, RASDI , DRGZ, and RAII on DNA isolated from patient SMS129 (Figure 11A). These genes were three of our best candidates based on the initial characterization methods described in Chapter 2. Using PCR amplification and sequencing of all exons from the RASDI, gene we detected several previously reported SNPs, but no deleterious DNA changes were 73 B)O SMSlZ8 SMSlZ7 C)GESVILLGPTVGTESK AGGGGAGTC TCATCCTGCTGGGCCCI‘ACAGTGGGCA GAGTCAAA I I“. {l . I I ll 1-. . ._.. .‘ .1 1.1-1.1411. “II “Milli/WM Normal allele G E S R V K G AGGGGAGTC+GAGTCAAAGG WW Deleted allele - Figure 11. SMS129 RAII mutation analysis. A) SMS129 pictured at age19 and 30. B) Pedigree analysis of the SMSlZ9, his parents (SMS127 and SMS128) and one sibling (SMS164). The RA13/ 14 PCR product is present in lanes 1, 3, 5, and 7; note doublet in affected individual SM8129. The PspOMI digestion of the RA13/l4 PCR product is shown in lanes 2, 4, 6, and 8; note undigested mutant allele in affected individual SMS129 which is not present in the parents or sibling. The 29 bp mutation was not detected in 102 Caucasian control samples which were analyzed by PCR amplification and PspOMI restriction digestion. C) The sequence tracing represents the the 29 bp deletion detected following direct sequencing of the RA13/14 amplimer of RA]! fi'om SMS129, as well as the wild type sequence detected on the other allele; this deletion eliminates a PspOMI restriction site, misincorporates 8 amino acids (4 of which are shown in the diagram) and produces a downstream stop codon. 74 found. Amplification and sequencing of the exons of the DRGZ I gene showed no changes, polymorphic or mutagenic, from the consensus sequence available from the human genome project. This was not a surprise as the DRGZ gene is highly conserved through evolution as will be discussed in Chapter 5. Interestingly, a deletion of 29 base pairs was found in exon three of the RAII gene on one allele in individual SMSlZ9 (Slager et al. 2003). This deletion was clearly demonstrated as two distinct bands representing the normal and deleted allele when the PCR product was run on a 2% agarose gel (Figure 11B). Both bands were extracted and purified from the agarose gel, cloned, and then sequenced. The chromatograms fiom the cloned PCR products revealed one normal allele, and one allele missing 29 base pairs (Figure 11C). This 29 base pair deletion produces a frameshift that introduces 8 incorrect amino acids, followed by a stop codon, truncating the protein produced by this allele (Slager et al. 2003). Bioinformatic sequence analysis using web based programming of the deleted allele revealed the abolishment of a PspOMI restriction site (Figure 11B). Using direct sequencing and restriction digest analysis of DNA isolated from the parents and sibling of SMS129 (SMS127, SMS128, and SMS164) we determined that the 29 base pair deletion occurred de novo and is not familial (Figure 11B). Though, low level and/or garnetic mosaicism cannot be ruled out using this methodology]. The PspOMI restriction analysis allowed for screening of 100 normal individuals (200 chromosomes) for this deletion (Slager et al. 2003). The deletion was not detected in any of the normal 1 DHPLC analysis of DNA isolated fi'om blood revealed that the mother of SMSlZ9 is ~20% mosaic for the same 29 base pair mutation. The mother is phenotypically normal. 75 chromosomes analyzed and thus, does not represent a normal polymorphism. Haploinsufficieny of the RAN protein due to the 29 base pair deletion likely causes the SMS phenotype seen in SM8129. With the discovery of a dominant frameshifi mutation in SM8129, we undertook sequencing of additional patients in whom a 17p11.2 deletion could not be detected by FISH analysis. Dominant fiameshift mutations were found in SMS153, SMSlS6, and SMS159. As reported by the lab in 2005, amplification of exon 3 of the RAII gene in SMS153 (Figure 12B) shows a 19 base pair deletion beginning at nucleotide 253 (Figure 12) (Girirajan et al. 2005). Similar to the mutation carried by SMS129, the deleted allele of SMS153 incorporates 60 incorrect amino acids into the RMI protein followed by a premature stop codon. The deletion was not seen in parental DNA samples, nor in >110 normal chromosomes analyzed (Girirajan et al. 2005). Analysis of RA]! exon 3 amplified from template DNA from SMS156 and SMSlS9 revealed single base pair deletions causing introduction of premature stop codons (Slager et al. 2003). In the case of SMS156 (Figure 14A, Table 7) a single cytosine within a run of 6 C’s ending at nucleotide position 5265 of the RA]! mRNA was deleted on one allele (Figure 14B). The 5265delC causes a frameshift on the coding DNA strand introducing 74 incorrect amino acids before the addition of a premature stop codon. Bioinformatic analysis of the DNA sequence indicated that this single nucleotide deletion abolishes a Bgll restriction enzyme recognition site. The 5265delC mutation was not found in either parent of SMS156 (SMS154 and SM8155) or in 100 normal control samples (equal to 200 chromosomes). 76 uo AGGCAGCAAGGCCTGCAGCGGAGGCCGGCT Deletedallele 253de| 19 Figure 12. SMS153 RAII mutation analysis A) SMS153 pictured at age 19. B) The sequence tracing represents the the 19 bp deletion detected following direct sequencing RAII exon 3 from SMS153, as well as the wild type sequence detected on the other allele. Figure 13. SMS156 RAII mutation analysis. A) SMS156 pictured at age 31. B) Pedigree analysis of the SMSlS6, her parents (SMS154 and SMS155). The RA25/26 PCR product is present in lanes 1, 3, and 5 and the Bgll digestion of the RA25/26 PCR product is shown in lanes 2, 4, 6, and 8 (note undigested mutant band present in SMS156 which is not present in parental samples). The 5265delC mutation was not detected in 110 Caucasian control DNA samples which were analyzed by PCR amplification and BglI restriction digestion. C) The sequence tracing fi'om the RA25/26 RAII amplimer reveals the 5265 delC in exon 3 on one allele; this mutation eliminates a BglI restriction site, misincorporates 74 amino acids, eliminates a BglI restriction site, and produces a downstream stop codon 78 3 E mama. mam—um was: _ N u a u a .22... IV unmet 3.5.. 0V Hulxl.l_.lllm|-mllwl > e _. > x Fe =e.._=e_s:o_o O 7. 1 O>OOOOHO>OOOOOG>Dnfi0>nnfifinOO>>OO>GODO GOO >>Ofififinn >Ofiflfido>finflnnfi>fifidfi>fififififififi>>06>fififififi 3:33:13; 1 O P .—. > O _. a. Z 1 m I > > > a mwammofln 79 Figure 14. SMSlS9 RAII mutation analysis. A) SMSlS9 pictured at age 11. B) SMSlS9 pictured at age 19. C) The 1449delC mutation in RA]! exon 3 on one allele is shown, which misincorporates 34 (12 of which are shown in the sequence tracing) amino acids and produces a premature stop codon. As no known restriction site was altered by the 1449delC mutation, amplified and sequenced the amplimer containing the l449delC mutation from >100 Caucasian samples (including the parents of SMSlS9), and this mutation was not detected in this population. 80 F -mI uoI ImI k > > a. > > 0 .000 00000fi>0 0>0000>000000 000 ~5—:d::.mu:m_¢ LLL LLL L L, L I 34390—0 81 Table 7. Phenotypic comparison of SMS patients with 17p deletions and RA]! mutations SMS HOU14 SMS SMS SMS SMS] 126 2-540 129 153 156 59 typical small deletion deletion Features common in >75% of SMS Patients Craniofacial/Skeletal Midface relative to Tented face Short stature abnormalities Middle anomalies voice ++ N Mental retardation Motor Infantile disturbance SMS behavior Self behavior Features common in 50 — 75% of SMS + + + + + + + + + + + + + + loss Ocular abnormalities Scoliosis Features common in < 50% of SMS Cardiovascular anomalies + - Renal anomalies - - - - - Seizures - - - - + Cleft - - - - - - Gender/Age at evaluation Female Male Male Female Female Male 41 10,14, 14 14 17, 21 13 and 16 82 Similar to the mutation in SMS156, DNA isolated and amplified from SMSlS9 (Figure 14 and Table 7) showed a deletion of a cytosine in a run of 4 C’s on one allele starting at base pair 1449 of the RA]! mRNA (Figure 14) (Slager et al. 2003). The deletion of the cytosine at base pair 1449 causes incorporation of 34 incorrect anrino acids before addition of a premature stop codon. Bioinformatic analysis of this mutation did not reveal abolishment or addition of a restriction site. Thus, template DNA from the family of SMSlS9 and normal control DNAs was amplified from exon 3 of the RA]! gene and sequenced. The l449delC deletion was not detected in the parental or control samples analyzed. Additional dominant mutations in the RA]! gene have now been reported from DNA isolated fi'om patients without a FISH detectable 17p11.2 (Bi et al. 2004; Girirajan et al. 2005). The total number of SMS patients carrying dominant mutations in the RAII gene totals 9 (Bi et al. 2004; Girirajan et al. 2005). Though the frame shift and nonsense mutations reported have been theorized to prematurely truncate the RAN protein (Slager et al. 2003; Bi et al. 2005; Girirajan et al. 2005). Because SMS is also associated With microdeletion of 17p1 1.2 including the RA]! gene, a dominant gain of function mutation in the RAII gene is unlikely. Research reported in the last 10 years indicates that premature stop codons caused by a frameshift or nonsense mutation may trigger decay of the mRNA before it is translated into a protein product (Cheng and Maquat 1993; Belgrader et al. 1994). Currently, research in the Elsea Laboratory is underway to determine whether premature stop codons in the RAII mRN A trigger the nonsense mediated decay pathway. It is likely that nonsense mediated decay of the mutant RAII mRNAs 83 causes haploinsufficiency of the RAN protein. The retinoic acid induced 1 (RAII) gene The mouse ortholog of RA]! was cloned prior to the discovery of the human gene. In 1995 a Japanese group identified Rail, then called Gt], as a transcript that was up-regulated in mouse embryonal carcinoma cells following treatment with retinoic acid (Imai et a1. 1995). Mouse embryonal carcinoma cells are known to differentiate into neuronal and glial cells when treated with retinoic acid. The original study aimed to understand the genetic factors underlying this differentiation (Irnai et al. 1995). RNA in situ hybridization and irnmunohistochemistry revealed that Rail gene expression is localized throughout the adult mouse brain, specifically in neurons (Irnai et al. 1995). The role, if any, the Rail gene plays in neuronal development has yet to be reported. As discussed in Chapter 2, our group mapped the human RAII gene to the middle of the SMS critical region as the EST clone DKFZp434A139 (Figures 3, 5, and 6) (Lucas et al. 2001). Further, using commercially available adult and fetal tissue northern blots we determined that the RAII gene is expressed as an ~8.0 kb transcript in all tissues examined (Figure 6). Upon publication of these data in 2001 (Lucas et al. 2001), we were unable to determine any possible cellular role for the RA]! gene due to lack of any homology of our EST to known proteins. Further complicating analysis of the gene was the number of different EST 8 deposited into the databases that were thought to represent the RAII/Rail genes. In 2003, the major human RAII transcript was cloned fiom SKN SH neuroblastoma cells 84 treated with retinoic acid, the sequence was deposited as GenBank accession AY172136 (Toulouse et a1. 2003). This group identified both the 5' and 3' UTR sequences, as well as the presence of an upstream CpG island and a putative retinoic acid response element immediately upstream of exon 1 (Toulouse et al. 2003). The mouse Rail gene is represented by Genbank accession AY5481752. Bioinformatic analysis indicates that both mouse and human sequences contain a bibartite nuclear localization signal, a polyglutarnine tract, a polyserine tract, and a conserved extended PHD domain at the carboxy end of the amino acid sequence (Girirajan et al. 2005). In depth bioinformatic analyses were performed in the lab by fellow graduate student Santhosh Girirajan on the human and mouse RAIl/Rail protein sequences. The human and mouse sequences are well conserved with 84% identity and 89% similarity. It was determined that the polyserine tracts seen in these proteins are similar to the human DRPLA and Drosophila hairless gene. Consistent with the original RAII /Rai1 cloning experiments, both DRPLA and hairless are reported to play a role in neuronal development (Maier et al. 1992; Onodera et al. 1995; Seranski et al. 2001; Girirajan et al. 2005). Additionally, polyserine and polygutarnine stretches are known to modulate transcriptional activation (Gerber et al. 1994). The extended PHD domain in the carboxy terminal of the protein is similar to that seen in the trithorax nuclear proteins. In Drosophila these proteins maintain stable expression of homebox-like genes (Paro 1993; Aasland et al. 1995). The PHD-finger domains likely bind zinc ions and act similarly to Zn-RING finger containing proteins (Aasland et al. 1995). Currently, no evidence has been reported on the binding of RAIl/Rail to DNA, RNA, or other proteins. The only other protein sharing 85 homology to RAIl/Rail is the human ARI gene also known as transcription factor 20, or TCF20 (Seranski et al. 2001; Girirajan et al. 2005). The protein homology of RAIl/Rail indicates that the gene is involved in transcription at some level, either as a bonefide transcription factor or a transcriptional co-activator. No reports of the in viva function of the RAH/Rail gene currently exist. Members of the lab have shown that a GFP-Rail fusion protein is localized within the nucleus. Further study of the RAH/Rail genes will help understand their role in mammalian development and in expression of the SMS phenotype. SUMMARY The discovery of 9 dominant RAII mutations together with data analyzed from heterozygous krrock-out of mouse Rail (Bi et al. 2005) indicate that haploinsufficiency of the RAII gene is the cause of the craniofacial, neurological, otolaryngological, and behavioral characteristics seen in SMS. Additional in depth studies are underway to better understand the cellular and biochemical role of the RAII gene. Currently, we postulate that other genes in the region may play a role in the more variable characteristics seen in SMS patients with l7p11.2 deletions, including cleft lip/palate, and visceral anomalies including the kidney and heart. Due to the fiequency of hearing loss in SMS patients we also believe that additional genes affecting hearing may map to the SMS critical region. We are continuing to study the role that additional genes mapping within the SMS critical region play toward expression of the SMS phenotype. 86 MATERIALS AND METHODS Patient ascertainment: SMS129, SMS156, and SMSlS9 were evaluated by Brenda Finucane, CGC at the Elwyn Inc. (formerly Elwyn Institute). Clinical data for SMS153 were collected by Dr. Sarah H. Elsea, Ph.D. via contact with the patient’s health care provider, and through our in house questionnaire (Appendix A). All clinical data and samples were collected in accordance with Institutional Review Boards at Elwyn Inc., Michigan State University (MSU), and Virginia Commonwealth University 0! CU). Clinical description ofSMS129, SMS153, SMS156, and SMS159: SMS129: SMS129 is a 30-year-old male who was admitted to residential placement at age 14 because of aggressive and disruptive behaviors which could no longer be managed at home. He was the product of an uncomplicated full term pregnancy, weighing 7 lbs.,1 oz. Motor milestones were normal, with no history of hypotonia, but speech development was significantly delayed. From an early age he exhibited aggressive and self-injurious behaviors, including onychotillomania and polyembolokoilarnarria, as well as sleep disturbance, and frequent self-hugging. He is obese and has spina bifida occulta. His behavior is currently stable, although he continues to require residential placement and psychotropic medications because of aggression. Results of fragile X and chromosome analyses at age 14 were normal. At ages 21 and 30, he was reevaluated in the genetics clinic and felt to have many behavioral and physical characteristics of SMS. A repeat cytogenetic study (650 band resolution) was normal, as were results of chromosome analysis on skin 87 fibroblasts and FISH for del(17)(p1 1.2). _S_l\_/1_$l_53: SMS153 is a l9-year-old obese, white female. Pregnancy was normal, though delivery was by emergency cesarean section because of failure to progress and a decreased fetal heart rate upon inducing contractions. SMS153 was diagnosed in the operating room with Down syndrome because of floppy muscle tone, upslanting palpebral fissures, midface hypoplasia and presence of a simian crease. Laboratory studies performed were negative for Down syndrome. Motor and speech were significantly delayed. When she did begin speaking her voice was noted as hoarse. Her facial features are consistent with the SMS phenotype and include brachycephaly, tented upper lip, and broad square face. SMS153 suffers hour the self-injurious behaviors commonly seen in SMS patients. These have been particularly difficult on the family. SMS153 also suffers from the characteristic SMS sleep disturbance. SMS156: SMSlS6 is a 31-year-old obese, white female. Pregnancy and delivery were normal; birth weight was 8 lbs. Motor milestones were achieved on time, and there was no history of hypotonia. Speech development was mildly delayed compared to that of her siblings. The patient has a history of mild mental retardation and emotional disturbance, including aggressive and defiant behaviors, which prompted residential placement during adolescence. She had self-injurious behaviors, including onychotillomania and polyembolokoilamania, as well as significant sleep disturbance. Her behavior improved in the residential setting, and she was able to return home to live with her parents at age 21. On reevaluation at age 31, her behaviors had stabilized, and she required no psychotropic medications. She 88 continues to exhibit skin and nail picking, and when excited, the self-hugging stereotypy typical of people with SMS. She also has a persistent habit of inserting string into her nose. Her facial features are subtly similar to those seen in patients with a l7p11.2 cytogenetic deletion. Chromosome and fragile X analyses at age 17 were normal, as were results of subsequent FISH studies for deletion 17p11.2. $148122: SMSlS9 is a 19-year-old male patient who has a history of mild mental retardation, self-injury, and aggressive behaviors which resulted in residential placement at age 13. Pregnancy and delivery were uncomplicated; birth weight was 8 lbs.1 oz. A functional heart murmur was detected in childhood and later resolved. Motor milestones were significantly delayed (walked at 21 months) due to infantile hypotonia. Speech developed within normal limits. Since early childhood, the patient has exhibited self-hugging behavior when excited. Macrocephaly was noted in infancy, and MRI studies of his head and spine at age 11 revealed mild hydrocephalus, Amold-Chiari malformation, and spina bifida occulta. On physical exam at age 13, he was obese and had facial features suggestive of SMS. The patient also had gynecomastia and hypogonadism. Results of fragile X and cytogenetic studies were normal, including FISH analysis for deletion l7p11.2. At age 19, the patient lived at home and received homebound school services. He suffers from recurrent, debilitating syncopal episodes likely related to his Amold-Chiari malformation. These episodes were temporarily alleviated by brainstem decompression surgery at age 15 but have gradually increased since then. The patient continues to present behavioral problems, including onychotillomania, which are treated with psychotropic medications. Polyembolokoilarnania has not occurred for 89 several years. Fluorescent in situ hybridization (FISH): FISH on patient chromosomes without detectable l7p11.2 deletions was performed as described in Chapter 3. Isolation of patient genomic DNA: In order to isolate template suitable for PCR amplification, DNA was isolated from peripheral blood or buccal cells fiom all putative SMS patients, parental and sibling controls, and Caucasian control samples. DNA isolation from whole blood: Approximately 15-50 ml of whole blood was collected and centrifuged at 2500 RPM plasma was removed, and the remaining cells were mixed with “red blood cell lysis solution A” (0.32 M sucrose, 10 mM Tris, pH 7.5, 5 mM MgCl2 and 1% Triton X-100) and placed on ice for 30 minutes. The mixture was then centrifuged at 2500 RPM, the supernatant was removed, and 50 ml of solution A were added to the pellet. This solution was placed on ice for 20 minutes, centrifuged as above, and the supernatant was removed. The pellet was resuspended in 5 ml of solution B (10 mM Tris, pH 7.5, 400 mM NaCl, and 2 mM EDTA, pH 8) and digested overnight at 37°C with the addition of 100 pl of 20% SDS and 50 pl of 20 mg/ml proteinase K solution. The following day, 3 ml of saturated phenol pH 8.0 were added to the solution while rocking, and then the samples were centrifuged at 2500 RPM for 15 minutes. The upper phase was removed with a Pasteur pipet and transferred to a new 15 ml polypropylene tube and 3 ml of chloroform:isoamy1 alcohol (24: 1) were added to the aqueous phase with rocking for 90 15 minutes. Following another centrifugation at 2500 rpm, the DNA upper phase was removed and the DNA was precipitated with 2 volumes of 95% ethanol. The DNA was removed with a Pasteur pipet and allowed to sit 70% ethanol for 5 minutes, then placed in 200 pl of 1x TE, pH 7.5. DNA isolation from buccal cells: Genomic DNA was isolated fi‘om buccal cells by boiling the cheek brushes in 400 pl of 50 mM NaOH at 95°C for 10 minutes. The brush was then discarded and the sample was placed on ice for 10 minutes. This solution was neutralized with 40 pl of l M Tris, pH 8.0. RAIl PCR amplification and sequencing reactions: Analysis of RAII in patient and control DNA was performed by PCR and subsequent sequencing and analysis of PCR products was performed by Rebecca Slager in the Elsea Lab. PCR primers covering the entire RAII coding sequence, 5’ and 3’ untranslated regions (UTR), and alternative splice variants were generously provided by Dr. Laura Schmidt of NCI- Frederick or were designed by this laboratory and synthesized at the Michigan State University Macromolecular Structure Sequencing and Synthesis Facility. PCR was performed in a 25 pL volume with 50-200 ng DNA template as described in Chapter 2. Informative primer sequences and conditions are indicated in Appendix B. In order to check each PCR amplification 5 pL of the reaction was electrophoresed in 2% agarose gels containing ethidium bromide. Successful reactions were then purified using the Qiagen Gel Extraction Kit according to manufacturer’s instructions, or treated enzymatically in the following manner: 2 pL of USB shrimp alkaline phosphatase (1 units/pL) and 1 pL USB exonuclease I (10 91 units/pL) were added to 5 pL of PCR amplification mixture, the solution was mixed and incubated at 37°C for 15 minutes in a thennocycler, and then inactivated at 80°C for 15 minutes. A sequencing reaction containing at least 10-40 ng of purified PCR product template in distilled water and 30 pmol of sequencing primer (the forward or reverse PCR primer or an internal primer) was then prepared and sequencing was conducted at the Michigan State University Genomics Technology Support Facility or the Virginia Commonwealth University Nucleic Acid Research Facility using an ABI PRISM® 3100 Genetic Analyzer or ABI PRISM® 3730xl DNA Analyzer. Sequence analysis: PCR amplified RAIII fragments were analyzed for mutation by pairwise BLAST analysis at NCBI (http:/lwww.ncbi.n1m.nih.gov/BLAST) against the published GenBank sequence for the RAII mRNA (GenBank Accession: AJ271790 and AY172136) and the genomic sequence data from the RAII locus (GenBank Accession: AJ271791 and NT_010718). SNP databases at NCBI (http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=snp) were also queried in order to identify polymorphic DNA changes. Sequencing chromatograms were also thoroughly visually inspected in order to identify sequence anomalies missed in the automated sequence analysis. Restriction enzyme analysis of RAIl mutations: The 29 base pair deletion detected in SMS129 abolishes a PspOMI restriction site. Parental and 100 normal control DNA samples were tested for this deletion via restriction analysis. PspOMI digestion of the amplimer from primer pair RA13/14 (Appendix B) normally cleaves the 493 base 92 pair product into 361 base pair and 132 base pair fragments. PspOMI does not cleave the allele with the 29 base pair deletion. PCR products were digested for 2 hours at 37°C with 1-2 U of PspOMI, then resolved on a 2% agarose gel in 1x TBE buffer containing ethidium bromide. Agarose gels were photographed on an AlphaImager using software version 5.5. The 5265delC mutation detected in SMSlS6 abolishes a Bgll restriction site. Parental and 100 normal control DNA samples were tested for this deletion via restriction analysis. Bgll digestion of the amplimer fiom primer pair RA25/26 (Appendix B) normally cleaves the 496 base pair product into 171 base pair and 325 base pair fragments. Bgll does not cleave the 5265delC allele. PCR products were digested for 2 hours at 37°C with 4-5 U of Bgll, then resolved on a 2% agarose gel in 1x TBE buffer containing ethidium bromide. Agarose gels were photographed on an AlphaImager using software version 5.5. Restriction analysis was performed by Rebecca Slager in the Elsea Lab. Analysis of control samples: Analysis normal control samples for mutations that could not be resolved with restriction analysis were directly sequenced and analyzed as described in the methods above. 93 Chapter V Developmentally regulated GTP-binding protein 2 (DRGZ) BACKGROUND Guanine triphosphate hydrolases (also known as GTPases or G-proteins) make up a superfarnily of proteins that act as regulatory molecules. The GTPases are known as molecular switches because they are activated while they are bound to GTP and switched off upon hydrolysis of the GTP to GDP. This group of proteins is expansive and is made up of three well-known sub-families; the monomeric GTP- binding protein Ras and its homologs, bacterial translation elongation factors, and the heterotrimeric G proteins (Bourne et al. 1991). All G-proteins work in the same mechanistic manner even though the primary structures are extremely divergent. The proteins maintain their fimction in the absence of long stretches of homology through conservation of four distinct domains that allow for conserved folding of the GTP binding core. The differences in primary structure allow for the extreme diversity seen in overall protein firnction, while conservation of the short GTP binding domains create the very well conserved secondary structure necessary for GTP binding and hydrolysis. As additional expressed sequences became available additional sub-fanrilies of GTPases were described. One such family includes the developmentally regulated GTP-binding proteins (DRG) proteins. The DRG proteins retain the G1-G4 binding motifs necessary for GTP-binding, though they do not share similarity to any of the 94 well-known GTPase sub-families. One member of the DRG family maps within the SMS critical region on chromosome 17 (Schenker and Trueb 1997; Vlangos et al. 2000). The map location of the developmentally regulated GTP-binding protein 2 (DRGZ) gene and limited information about its possible role in development, made it one of our best SMS candidate genes. In addition to fine mapping of the gene, we undertook characterization of DRGZ through expression patterning at the RNA and protein levels. Cloning and identification of mammalian DRG genes In 1992 a subtractive cloning approach was used to identify genes that are expressed at high levels in developing and differentiating mouse brain when compared to adult brain. This approach identified many potential transcripts including the Nedd3 gene (neural precursor cell expressed, developmentally downregulated-3) (Kumar et al. 1992). Identification and characterization of a longer cloned sequence for Nedd3 showed that it contained short regions of homology to Drosophila and Halabacterium sequences deposited in public databases. Early bioinformatic analysis of the three related sequences revealed that they all shared domains of GTP-binding. In vitro analysis of the Nedd3 protein showed its ability to bind GTP. Based on the results fi‘om the in vitra GTP-binding assay and the subtractive cloning method enriching for developmental genes used in its original identification the Nedd3 gene was assigned the name developmentally regulated GTP-binding protein (Sazuka et al. 1992b). The putative human homolog of Drg was also identified by subtractive cDNA 95 cloning where it was shown to be expressed in human fibroblasts but repressed in virally transformed cells (Schenker et al. 1994). Using FISH techniques, the human gene was mapped to chromosome l7p12 -)13 (Schenker and Trueb 1997). Searches of DNA databases revealed that there were at least 2 DRG genes in humans, DRGI and DRGZ. In 2000 we reported that the human gene identified by Schenker et al is actually DRGZ, while original murine Drg gene is actually homologous to human DRGI (V langos et al. 2000). RATION ALE Our lab mapped the DRG2 gene to chromosome 17p11.2 within the SMS critical region. In order to begin to understand the function of DRGZ and any role it might play in SMS, we looked at RNA expression patterns of the gene in adult and developing human and developing mouse. We employed irnmunohistochemistry techniques to determine spatial and temporal expression through mouse development. At the cellular level, localization of the protein was performed using green fluorescent protein technique. The data presented in this chapter are currently being drafted into a manuscript for publication. 96 RESULTS Identification of the STS WI-l3499 as DRGZ and mapping to the SMS critical region In order to clarify the map location and identification of DRGZ to chromosome 17p11.2, we used the sequence tagged site (STS) WI-l3499. We and others previously mapped STS WI-13499 to chromosome 17p11.2 (Elsea et al. 1997; Liang et al. 1999). Subsequent BLAST searched and Unigene analysis of the published WI-13499 sequence indicated 94% identity to the DRGZ gene. The STS WI-13499 was created from the I.M.A.G.E. cDNA clone 29328. Analysis of the DNA sequence of clone 29328 revealed that it bore 99% identity to DRGZ. Lower identity of the DRGZ sequence to WI-l3499 was likely due to one-pass sequencing errors. To ensure that we were truly looking at a single-copy gene and that clone 29328 (WI-13499) and the reported DRGZ sequences were representative of the same gene, we fully sequenced clone 29328 and the PCR product WI—13499 and aligned them to the reported DRGZ sequences from GenBank. The results indicate that the sequences are truly identical and representative of the same locus. DRG2 was originally mapped to chromosome 17p13 -) p12 by FISH (Schenker and Trueb 1997). Experiments using Southern analysis (Figure 15) and PCR mapping with somatic cell hybrids retaining portions of chromosome 17 indicate that DRGZ maps more proximally on chromosome 17, to band 11.2 and the SMS critical region. In order to confirm our mapping data, we obtained a 1,300 base pair DRGZ clone from Dr. B. Trueb (originally reported in Schenker ant Trueb, 1997) and 97 17p 13.3 13.2 13.1 I 12 11.2 MH22-6 — 255-110 — — ‘ 485-3D _ 540-1D — — 357-2D _ — — c b a: “9 E m ‘é’é’i— : a “O :: oo :23 Kb Q0. a» E! ‘t 12 3:3“? ' _/11 -10 ,. —9 ##“II‘ -8 ' —7 “L It» -6 O -5 Figure 15. Somatic cell mapping of DRGZ. EcoRI digests of humanm rodent, and hybrid genomic DNAs is shown. The entire ~l.9 kb insert from IMAGE clone 29328 was PCR amplified and used as a probe. Bars above the blot indicate relative regions of chromosome 17 deleted in human and hybrid DNAs. Arrows to the left indicate chromosome 17 specific genomic bands. 98 obtained the same mapping results using the somatic cell hybrid mapping panel. These data confirmed that DRGZ maps within the SMS critical region (V langos et al. 2000). Bioinformatic analysis of DRGZ By direct sequencing we determined that clone 29328 contained an EST insert of 1,917 base pairs. Our southern analysis showed that the DRGZ gene likely spans a minimum genomic region of 32 kb (V langos et al. 2000). Alignment of the cloned EST sequence to the latest version of the human genome sequence shows that DRGZ is composed of 13 coding exons spanning 20 kb of DNA. A potential ATG start site begins at base number 76 and fits the Kozak consensus sequence of ACCATGG (Kozak 1986). Six-frame translation of the DNA sequence showed an open reading frame resulting in a product of 364 amino acids. This protein has an estimated molecular weight of 40.5 kDa. Four conserved GTP binding domains remain intact in the DRGZ protein (Figure 16). The G1 domain is unique to this subfamily of G- proteins and has been termed OBG/GTPI based on the orthologous B.subtilis and S. pombe genes. Alignment of DRG2 amino acid sequences reveals striking homology through evolution (Figure 16). The human sequence is 98.6% identical to the mouse sequence, 93.9% identical to the Xenopus, 80.5% identical to Drosophila sequence, and 65.9% identical to the fission yeast sequence. Though the DRGZ sequences are well conserved through evolution, DRG] and DRGZ only share 57% identity at the amino acid level. It is interesting that human DRG2 is more similar to the fission 99 HS DRGZ .4 .. ._ . ._3 ..-.. 1 ...I‘:'...‘" 515:." '.: .-, :‘ Mm Dr92 1 '.LE1-3111‘1'1‘T £1.11; 1::II1 11E1'111. (3.:11.1 911:1 £r1~21011hp X1 Dr92 1 1L1:.1:ISE1E.1‘:1..-:1"1')111Ti‘311'.3'J1..1_.1‘”:11':13n1:LPQQLLEPof “‘1‘1KJEKJ1‘D 1111 Dm DRG2 1 -E1IgEILRLI bTHrI1AT1IMLG131~1L11YR§QL1LF {Kr-E1b01FDVLx Sp GTPl 1 .. -«:. III'“I' 13G; ' H8 DRGZ 'i'..EI7.33.1.1.31‘1'3Yi3KL}1EI'I..L71.1-'.1:117..‘7':‘:.»';~.i7'-'13 FITLlCIPK‘: .711'3'1'1'1313311C21313D1.1I"~3i- MN Drgz 31 F‘HLIDFP)‘UF3TELQLHI TH)HM%SLEFIILrCIPD IE"KGANIQLLDLPC1L X1 Drgz 5UMRV :£LIGFPS I‘C:1.3TFISLIIT TnSEsnLIEFTTIICIPGVIEI1GANIQLLDLPG-. Dm DRGZ '6 - ' g. : MAIIQLLDLF.1 sp GTPl .. - - .-._ :. I~.‘ - .. ' ‘ :1 . . .v‘..‘,. .. ....-..... .-...,, .. . ..... ,... .....-...., 8 . .. i~', ,w- .n f .. .. . .. . ‘. . ~- . . ~~ .. r—g J . ...._7-.. ..- . -.'*‘- ... .... ‘ ........... 3 . .. . . .. . 1.. ..., ..-.-...... Mm DrgZ inAQGE-GRGRQVIAL'AP: I‘AUVV Evil-21.3.3.1 Egg. (2 11.2131. E E1. 1381131 R1 IIKflK’ r [I 1 . ;' x1 Drgz vNAflQGKGRGRQVIA”ARTiDVV MMI.DAIKG ’ORSLI.EI LL3\LIVLN1EAPUI.r Dm DRGZ " 3.:‘1AQGKG1'JL31‘30‘JIA r1I1'TAD . :1 (811 .E 151.135" (JIPIJ‘JKéI‘ [)1 J11 Sp GTPl v 3:7. ,‘C‘ -. ‘, - " ‘ 1 .3 "7 .1 713111.11" QPE‘II H8 DRG2 71.13? ’,".“IEI.'~'.I,‘.':;I.'. . .Ei'r '-.":~'. IT. " Mm Drgz z'I 313GIS1II: - ' ‘“ I Z .‘-.'QI311311'E;'£K1113AE I31 rLDCfiI‘DEFID "1. 1311111. X1 DrgZ ‘ .1113 3 ’3 1 31.. 1.'I‘OC;31~31‘CI.‘~.'Q.. 11.11171 'KaE-‘II “131' 1. F REDCTPDEFI D‘.’ "JGIJR" 1. Dm DRG2 :/:"1‘ _. . ITuaKIFI; A13 LWHJFTEUEFHN. Sp GTPl "I? , fi~ " . r . .-. ~ ... ~ H8 DRG2 ., . -... , .-.U- .. . . Mm Drgz -L.V.NKIDQ1.d‘LVDkIAPK1U3.‘lbLUHRIULDILLEH.‘1 LALICIITKKI x1 Drgz Dm DRGZ Sp GTPl Ha DRGZ .e 3g:1::v: I“ - . H ._- . “v:~ . . - Mm Drgz AF DflrrLIIGa..1a (METHRILfiuQFYYAL“‘b rr.er . x1 Drgz Icy DAIIIPI x mm (IFIHRTLaquP..LI QISIRLSPOR Dm DRG2 . :ng 11131 by . C1n111 RTL.‘ A311 1 .11.? .\31L 11314.11: Sp GTPl Si"TV SIR "”' i"‘ 12.-I’u7rc ” ‘3"7' Ha DRG2 Mm Drgz X1 Drgz Dm DRG2 Sp GTPl Figure 16. Alignment of DRGZ amino acid sequences. Amino acid sequences from human DRG2 (Hs DRGZ), mouse DRGZ (Mm DRG2), Xenopus (X1 Drg2), Drosophila DRG2 (Dm DRG2), and fission yeast gtpl (Sp gtpl) were obtained and aligned using the CLUSTALW 1.8 program at the Baylor College of Medicine Search Launcher web site (http://searchlauncher.bcm.tmc.edu). The black shading indicates identical amino acids, the grey shading indicates similar amino acids, the white shading indicates differing amino acids. The star indicates stop codon, and the - indicates a gap in alignment. Afier alignment, the figure was created using the BOXSHADE program (http://http://www.ch.embnet.org/sofiware/BOX_form.html). 100 yeast DRG2 than to the human DRGl sequence. Previously published phylogenetic analysis of the DRG sequences available in 2000 revealed that 2 genes seen in higher organisms likely evolved fi'om a gene duplication event (Li and Trueb 2000). Only a single DRG gene is present in bacteria. These data reveal that the DRGZ proteins are more similar to the DRG gene seen in bacteria and that the DRG] proteins are likely the result of gene duplication. Expression levels of DRGZ/DrgZ using northern analysis Since DRGZ is thought to be developmentally regulated, we characterized both human and mouse DRGZ/DrgZ using northern analysis on adult and fetal tissues. Northern analysis of human tissues on a commercially available blot showed an ~2.2 kb transcript in all adult and fetal tissues represented (Figure 17A and 17B). Expression was not significantly elevated in any human tissue analyzed. Rather, the gene seems to be expressed at fairly equal levels in all tissues represented on the northern blots. Electronic expression profiling of human samples using the Unigene database at NCBI currently shows highest levels of Drg2 expression in cervix and small intestine. In order to characterize the expression pattern of Drg2 in the developing mouse, we utilized a northern blot representing embryonic day 5.5 to day 18.5. The blot does not contain RNA fiom maternal or placental tissues. Using a full-length EST clone for Drg2 as a probe a ~2.0 kb transcript representing Drg2 expression was seen in all developmental days at a consistent level (Figure 17C). The blot did not reveal any drastic peaks or depression in gene expression during development. This 101 is in contrast to reports of mouse Drg] where there was a significant decline in gene expression starting afier embryonic day 10 (Sazuka et al. 1992a). Electronic expression profiling of mouse Drg2 indicate expression in all tissues examined and at all developmental stages. Our studies, thus far, do not indicate that DRG2/Drg2 is developmentally regulated at the RNA level. Spatial and temporal expression of Drg2 using immunohistochemistry In order to understand the expression pattern of the Drg2 protein in the developing mouse embryo we undertook irnmunohistochemistry (II-1C) experiments on sectioned C57Bl/6J embryos. The IHC experiments were performed using a polyclonal antibody designed to an 11 amino acid sequence near the carboxy end of the DRGZ protein (Figure 16). The sequence of DRGZ was extensively analyzed in the design of the peptide used to create the antibody. The sequence was run through computer programs generating a hydropathy plot showing regions of hydrophilic amino acids most likely to reside on the outside of the folded protein. BLAST analysis of all deposited amino acid sequences revealed no known homology to any other proteins (known or predicted) except DRG2. The 11 amino acid sequence does not occur in the DRG] protein. The antibody produced by this peptide is recognized in mouse Drg2, Xenopus drg2, and Drosophila DRGZ allowing for experimental use in multiple organisms. The peptide was injected into two rabbits for polyclonal antibody production. 102 A) 2 B) U ‘2 33$ .. a a ‘- OD “win Eggs mm 5.415QO an .4 .452 T" 3 , “E 2.4— ' C H 1.35 1.35_ : DRGZ DRGZ 2.4a8 C." I.‘ 2.4 1.35— " 1.35 . B—Actin B—Actm C) In try try us II} In no In In W ‘n ‘n. V1“ 6 -« Nviv vi \0' [‘06 W \O l\ 00 ON .— v-I r—I v—l '- —a —c —I —t LL] Lil-l [L] m LL} LL] m LL} LL] LU LU LL] LL] LL] Figure 17. Northern analysis of DRGZ/DrgZ. A) Human DRGZexpression analysis is seen throughout all tissues in this Clontech adult multiple tissue northern blot. B) Human DRGZ is seen in all tissues present in this fetal Clontech multple tissue blot. C) A consistent expression of an ~1.7 kb transcript throughout mouse development is visualized. Northern analysis was performed on a mouse embryonic blot purchased from SeeGene USA (Rockville, MD). IMAGE clone 4910172 representing mouse Drg2 was used as a probe. B-actin was used as a loading control. 103 ELISA analysis of the polyclonal antisera showed that the antibody could detect the DRG2 peptide at 1:100,000 dilution. The polyclonal antiserum from rabbit 8139 was further tested in order to determine whether or not it had the ability to recognize the mouse Drg2 protein. The mouse Drg2 gene was cloned into a vector containing a cytomegalovirus (CMV) promoter. The plasmid was transfected into COS-7 cells allowing for Drg2 expression from the CMV promoter. Lysate from cells transfected with varying amounts of DNA was used to create a western blot. The DRGZ polyclonal antiserum from rabbit 8139 was used to detect the Drg2 protein expressed in the transfected cells. The western analysis revealed an ~40 kDa band of varying intensity which correlated directly with the amount of EST DNA transfected into the COS—7 cells (Figure 18). The Drg2 banding pattern was not detected in untransfected cells. These data show that the antiserum has the ability to detect the proper epitope. Sectioned mouse embryos representing embryonic day 8 though birth were examined via IHC to determine the spatial expression pattern of the Drg2 gene through development. Gestation in the mouse is a fairly rapid process lasting ~21 days. Dissection of embryos prior to day 8 was not attempted due to the small size of the embryo. At embryonic day 8 Drg2 is highly expressed within the neural tube, migrating neural crest, and somites (Figure 19A), while low level staining is present throughout the rest of the embryonic section. 104 bowel) cocoon 60211100111 iooo 100:: Eomwxoomwxo kDaE, , { 1.. 191- 97 f 64 -~ 51 —' S i _ (fl 2-..... 40.7 kDa 39" M l 28f Figure 18. Transfection of Drg2 into COS-7 cells. COS-7 cells were transfected with the full length Drg2 sequence under control of the CMV promoter. Cell lysate from tranfected cells was run on a western blot and analyzed with a polyclonal antibody created to the Drg2 protein. A 40.7 kDa band is seen in transfected cells. The band intensity correlates directly with the amount of DNA added to the cells. The amount of DNA input into the cells is indicated at the top of the figure. 105 Figure 19. Protein expression of Drg2 during mouse development. A) An 8.5 day mouse embryo transverse sectioned is stained with anti-Drg2. Expression is seen throughout the embryo, and is highest in the neural tube and neural crest. C) A 10 day sagital section shows Drg2 expression throughout the embryo with highest expression in the developing brain and neural tube. E) and G) A day 12 and day 14 embryo respectively stained with Drg2 antibody showing expression in the developing nervous system. I) and K) show a 16 day and 19 day mouse embryo respectively and show expression in the neural tissue and spinal ganglia. B D, F, H, J, and L) Show corresponding pre-immune serum controls for respective embryos. 106 107 108 By embryonic day 10, the most impressive development has occurred in the embryonic brain. The optic vesicle is beginning its transformation into the optic cup. Expression at this time is seen throughout the embryo with higher expression in the optic stalk, forebrain, hindbrain, and neural tube (Figure 19C). Between developmental day 10 and 12 significant maturing has occurred in the mouse embryo. The heart, vascular system, digestive system, and external features are more clearly defined. Drg2 expression at embryonic day 12 is clearly seen in the developing spinal cord, optic recess, and the developing brain (Figure 19E). At developmental days 14 and 15 the most drastic changes are in the skeletal system where the pre- cartilage has developed into mature cartilage. Additionally, the heart has continued to develop, and the components have reached their post-natal configuration. Drg2 expression at day 14 is very clear in the developing neural tissues of the entire brain and spinal cord. Additionally, expression is seen in the cervico-thoracic ganglia (Figure 196). The expression in these ganglia mirrors the reported expression of Drg] in day 14 embryos as measured by RNA in situ hybridization (Sazuka et al. 1992a) Expression of Drg2 at day 15 is seen diffusely throughout the developing mouse, with higher expression still being seen in the developing brain and neural tissues (Figure 191). At embryonic day 20, Drg2 expression is seen diffusely throughout the entire developing animal. In Figure 19K slightly higher expression can still be noted in the brain and spinal ganglia. Analysis of the RNA expression patterns of Drg2 in the developing mouse did not reveal developmental regulation as reported with Drg]. As with the RNA expression, analysis of the spatial and temporal expression of the gene using IHC also 109 reveals expression at all developmental stages evaluated. Greater expression was noted in the developing neural tissue throughout development. These results are similar to those reported for Drg] expression as reported using RNA in situ experiments. Both human DRG homologs may play an interesting role in neural development. Cellular localization of Drg2 The spatial and temporal expression patterns of Drg2 revealed in the developing mouse embryo show that the gene is expressed throughout development, especially in developing neural tissue. In order to further understand the biochemical role of the Drg2 gene, we used green fluorescent protein (GFP) techniques to evaluate the spatial expression of the gene at the cellular level. The Drg2 gene was cloned into a vector containing the sequence of the GFP gene. Cloning was performed such that the Drg2 gene was cloned directionally and in frame with the GFP gene. Expression of the GFP-Drg2 fusion protein was performed by transfection of COS-7 cells. Examination of cells transfected with the GFP-Drg2 fusion protein revealed localization to the endoplasmic reticulum (ER) and/or Golgi apparatus (Figure 20). Transfection of COS-7 cells was repeated in order to verify the cellular localization with ER and Golgi control stains. After transfection but before visualization, cells were treated with a dye that localizes to. the ER and fluoresces a blue-white color under a long-pass DAPI filter. Ceramide stain localizing to the Golgi apparatus fluoresces red under a rhodamine or Texas red filter. The two 110 control stains co-localize with the GFP-Drg2 fusion protein indicating that Drg2 likely functions in the ER and Golgi apparatus. The best studied GTPases associated with the Golgi apparatus and the ER are the ADP-ribosylation factor (ARF) genes and the (Rab) genes. ARF activity has been shown to regulate intracellular vesicular membrane trafficking and stimulate a phospholipase D isoform. Phospholipase D may be involved in cell morphology alterations through intracellular protein trafficking (Hammond et al. 1997). Homozygous mutations in a homologous ARF like gene have been shown to be one possible cause of Bardet-Biedl syndrome type 3 (Fan et al. 2004). The Rab proteins regulate vesicular transport in endocytosis and exocytosis. They are small GTPases that are distantly related to the Ras proteins. The role of the Rab proteins seems to be as a regulator of the sequential events that must occur for proper vesicular transport (Rodman and Wadinger-Ness 2000). Rab genes have been associated with Griscelli syndrome which manifests with immunological anomalies and partial albinism. Recently, it has been suggested that Rab genes may be one of the many causes of Batten disease (Luiro et al. 2004). These genes are also currently being investigated as the cause of many cancers (Cheng et al. 2005). Many additional GTPases including Rabs, ARFs, and DRGs (and their associated proteins) are being investigated for roles in human genetic diseases. 111 Figure 20. Cellular localization of Drg2. COS-7 monkey fibroblast cells were transfected with a Drg2-pcDNADEST47 construct, allowing expression of a Drg2-GFP fusion protein. Cells were treated with Bodipy TR Ceramide and ER-Tracker Blue-White to stain the golgi apparatus and endoplasmic reticulum, respectively. A) Drg2-GP P fluorescence is visualized under a FITC filter, B) ER fluorescence under a DAPI filter and C) Golgi fluorescence under a rhodamine filter, D) A merged image of the DAPI and rhodamine filter showing colocalization of the ER and Golgi, and E) A merged image of the DAPI, FIT C, and rhodamie filters showing co-loclaization of Drg2-GFP to the ER and Golgi. 112 113 SUMMARY Developmentally regulated GTP-binding protein-2 was named because of its homology to Drgl, a possible developmentally regulated gene. The Drg] gene was originally reported as being developmentally regulated because of the subtractive cloning method used in its original isolation (Sazuka et al. 1992b). Further, at the RNA level Drg] is reported to be highly expressed at embryonic day 10 with decreasing expression seen through birth and into adulthood (Sazuka et al. 1992a). Unfortunately, the authors of this paper failed to include positive controls in their analysis of gene expression. Expression of Drg2 during development is highest in the developing neural tissues. The Drg2 gene may function in the developing neural tissue by regulating protein trafficking in the Golgi apparatus and the ER. It may directly interact with proteins being moved through the cell, or similar to its relatives Rab and Alf it may regulate the formation of the vesicles used in the trafficking system. Further biochemical analysis of the protein will help reveal its fimction in the developing neural tissue and its role at the cellular level. Any role that the gene may play in the SMS phenotype will need to be further evaluated upon resolution of the protein biochemistry, and/or creation of a Drg2 targeted mouse. 114 MATERIALS AND METHODS Alignment of orthologous DRGZ proteins: DRG2 protein sequence fi'om Homo sapien (I.M.A.G.E. clone 29328 and Genbank accession NP_001379), Mus musculus (I.M.A.G.E. clone 4910172 and Genbank accession NP_067329), Drosophila melanogaster (Genbank accession NP_53673 3), and Schizosaccharomyces pombe (Genbank accession JT0741) were aligned using ClustalW 1.8 at the Baylor College of Medicine Search Launcher (http:/lsearchlauncher.bcm.tmc.edu). Aligned sequence was graphically transformed using Boxhade 3.21 (http://www.ch.embnet.org/software/BOX_form.html) Northern Analysis: A mouse embryonic northern blot was obtained commercially (SeeGene USA, Rockville, MD). Northern analysis was performed as described in Chapter 2. Human B-actin DNA (Clontech Inc.) cross reacts with all mammalian B- actins and was used as a control probe on the mouse blot. Northern analysis of B- Actin was also canied out as described in Chapter 2, except that 5x105 CPM/ml was used, and the hybridization temperature was reduced to 42°C. Cloning of Drg2 into a GFP vector: The Gateway cloning system from Invitrogen was employed to clone the Drg2 EST sequence into a vector containing the GFP sequence. The Gateway system uses site-specific recombination properties of bacteriaphage lamba to shuttle cloned sequences between various vectors. The entire 115 coding sequence for mouse Drg2 was amplified from I.M.A'.G.E clone 4910172 using primer pair SHE242/243. Primer SHE242 anneals to the 5’ end of the Drg2 coding sequence and contains a Kozak consensus sequence upstream of the m start site for the Drg2 gene allowing for proper gene expression upon transfection (SHE242: 5’-CACC;£I‘_GGGGATCTTGGACA-3’). SHE243 anneals to the 5’ end of the Drg2 coding sequence and was designed without the Drg2 stop codon to allow for proper in-frame read-trough transcription to the GFP gene (SHE243: 5’- CTTCACAATCTGCATGA). PCR product fiom SHE242/SHE343 amplification was cloned into the pENTR vector from Invitrogen per manufacturer instructions. Using the Gateway recombination technology, the Drg2 sequence was moved fiom the ent1y vector (pENTR) to a vector containing C-terminus GFP expression (pDEST47) per manufacturer instructions (http://www.invitrogen.com). All cloning reactions were confirmed via direct sequencing of the product. Plasmid DNA was isolated using the protocols described in Chapter 2. Calcium-phosphate transfection of COS-7 cells: COS-7 cells were grown to confluence in T-25 flasks for Drg2 expression or on sterile cover glass in 6 well plates for GFP-Drg2 fusion protein expression. Cells were grown to ~60% confluence in commercially available RPMI media (Invitrogen) supplemented with 10% fetal bovine serum, and 1x antibiotic/antimycotic solution (Invitrogen). Media on cells was changed 2 hours prior to transfection. DNA to be transfected was isolated and diluted to a total volume of 500 pl in ddHZO. To the DNA 50 pl of 2.5 M CaClz was added. DNA solution was briefly vortexed. Five hundred microliters of 116 2x HeBS (2x HeBS is 50 mM HEPES [pH 7.1], 280 mM NaCl, 1.5 mM Na21-1P04) was placed in a 15 ml conical tube. Air was bubbled through the HeBS solution with a sterile 5 ml pipette connected to a battery operated pipette aid. While bubbling HeBS, the DNA solution was added dropwise. The 15 ml tubes were vortexed briefly, and incubated at room temperature for 15 minutes. After 20 minutes the DNA solution was added to the cells. Cells were incubated for 7 hours at 37°C under 5% C02. After incubation, media was replaced and cells were allowed to grow for 2 days. After 2 days, the coverglass with cells transfected with GFP-Drg2 plasmid DNA was removed and rinsed with sterile lx Hank’s balanced salt solution (HBSS; Invitrogen). Cells not stained with Golgi or ER control stains were mounted onto glass slides with Vectashield with DAPI (Vector Labs) and viewed on a fluorescent AxioPlan2 microscope (Zeiss). Fluorescing cells were photographed using an AxioCam and AxioVision software version 4 (Zeiss). Cells grown to be stained prior to viewing were rinsed with HBSS and a mixture of 1 nM ER tracker blue white- solution (Invitrogen), 5 1.1M BODIPY TR ceramide (Invitrogen) in RPMI media or HBSS was added. Staining mixture was allowed to incubate for 30 minutes before cells were rinsed with HBSS and mounted to glass slides with Vecatshield without DAPI (Vector Labs). Cells were viewed and photographed as above. Cells transfected for Drg2 expression were removed from the T-25 flasks using 2 ml 1x Trypsin-EDTA (Invitrogen). Cells were collected in 15 ml tubes via centrifugation, rinsed in 1x HBSS, and pelleted via centrifirgation. Cell pellets were lysed in 200 pl 1x Laemmli buffer (2x Laemmli buffer is 4% SDS, 120 mM Tris pH 117 6.8), 1x protease inhibitor (Boehringer) for lysis. Lysate was vortexed briefly and run through a 20-gauge needle in order to shear the DNA. Lysate was stored at -20°C until use. Western blotting: Protein samples were prepared for western blotting in 1x loading dye (Invitrogen) and 1x reducing agent (Invitrogen) to a total volume of 25 pl. Samples were heated to 65°C for 10 minutes prior to separation on polyacrylamide gels. Invitrogen’s western blotting system includes precast 5-12% Bis-Tris acrylamide gel (Invitrogen) run and transferred in an XCell western blotting apparatus. Protein gels were run and transferred to PVDF membrane per manufacturer instructions. Western analysis was performed using antibodies in a 1:1000 dilution. A chromogenic WestemBreeze kit from Invitrogen was used for western analysis following manufacturer instructions (http://www.invitrogen.com). Immunohistochemistry: IHC was performed using a polyclonal antibody designed and created by Alpha Diagnostics. Timed matings were performed using C5 7Bl/6J (The Jackson Laboratory). Embryos were dissected and fixed in 4% paraformaldehyde overnight. Sectioning in paraffin blocks was performed at the Michigan State University Histology Lab. Sections were de-waxed in xylene, and re- hydrated through an ethanol series before use. The sections were exposed to antigen unmasking by heating to boiling in a microwave, boiling continued for 5 minutes in antigen unmasking solution (Vector Labs). Blocking was performed overnight at 118 room temperature in a humid chamber. Blocking reagent was 1x BM blocking reagent (Roche), 5% goat serum, 0.2% Tween 20, 1x PBS. Sera were diluted 1:1000 in blocking solution and incubated to sections overnight at room temperature. Sections were washed and stained using the Vectastain Elite ABC Kit and DAB/Ni detection (Vector Labs) per manufacturer instructions. 119 Chapter VI Target of myb-l (chicken) like 2 (TOMILZ) BACKGROUND In addition to studies involving the DRGZ gene, I also embarked on characterization of the novel EST stSG9692. The stSG9692 EST was previously mapped to the SMS critical interval (Elsea et al. 1997), though no additional information was available upon beginning the study. Through sequence analysis we determined that stSG9692 represents the 3' UTR of a gene known as target of myb-l (chicken) like 2 (T 0M1L2). Initial characterization using northern analysis revealed low—level expression in all adult human tissues studied, including the brain. At this point in the study, we had identified dominant fi'ame shift mutations in the RAII gene in persons with SMS. Study of the TOM112 gene was given less priority in favor of studies of the RA]! gene. RATIONALE Bioinformatic and northern analysis of TOMILZ was performed in our initial charaterization of all the known genes and ESTs mapping to the SMS critical region. Upon finding mutations in SMS patients in the RAII gene research of TOM 1L2 was given less priority. Study of TOM1L2/Tom112 resumed upon discovery of a mouse embryonic stem cell line fi'eely available to academic researchers carrying a gene- trapping vector inserted within the T om] 12 gene. The gene trapping method disrupts genes by random insertion in the genome. The ES cells were obtained and mice 120 carrying the targeted Tom112 were generated. In addition to the initial characterization of the TOM1L2 gene, we also analyzed the Tom112 mice in order to determine if any of the SMS characteristics could be recapitulated in the gene-trapped \ mice. RESULTS Bioinformatic analysis of TOMILZ Initial database searching of the small amount of sequence available for EST stSG9692 revealed only that it was represented by IMAGE clone 23808. This clone was commercially obtained and DNA was isolated and sequenced. When compared to nucleotide databases at NCBI, the sequence only aligned with the small amount of sequence available for itself and the now completely sequenced PAC RP1-253P07. Six frame translation of the clone sequence did not reveal an open reading frame. As newer gene prediction software became available we were able to match the cloned EST sequences to predicted genes reported online (http://www.genome.ucsc.edu). Searching the Gene Scan program at the Golden Path website (http://genome.ucsc.edu), we found that clone 23808 was predicted to lie within the 3' untranslated region of a predicted gene with accession ctg17005.29l. This predicted gene spans 53.1kb and is made up of 15 exons. BLAST analysis at NCBI showed sequence homology to the gene target of mybl (chicken) homolog (T 0M1). T 0M1 was originally identified in chicken cells as a gene target of the proto-oncogene v-myb when chicken myelomonicytic cells were transformed with avian myeloblastosis virus or avian leukemia virus E26 (Burk et al. 1997). The 121 human T 0M1 gene was mapped to chromosome 22ql3. l , expression analysis showed a 2.3 kb transcript in all adult tissues. In situ hybridization was performed on mouse embryo and demonstrated ubiquitous expression (Seroussi et al. 1999). We wanted to confirm the predicted Tom] 12 sequence in vivo in preparation for further experiments on mouse Tom] 12. RT-PCR using RNA isolated fiom mouse tail was used to amplify the Tom112 cDNA. The resulting cDNA was isolated and sequenced. The T om! 12 gene in mouse contains 15 exons and spans 122 kb (Figure 22). Six frame translation of the cDNA sequence revealed an open reading frame resulting in a 507 amino acid product. Sequence analysis of the TOMILZ/ Tom] 12 sequences using BLAST programs at NCBI show two conserved domains within the sequence. There is a VHS domain present, which is seen in proteins involved in vesicular trafficking and endocytosis (Lohi and Lehto 1998). Also present within the predicted sequence is a GAT domain thought to be responsible for stabilizing membrane-bound ARF 1 in the GTP state (Takatsu et al. 2001). Northern Analysis of TOM1L2 Expression analysis was carried out using commercially available northern blots from Clontech (Figure 21). Hybridization was carried out on the multiple tissue and brain 2 northern blots. The expression pattern on both northern blots revealed that the transcript of this EST is ~6.5 kb. Expression pattern in the adult tissues (Figure 19A) shows approximately equal expression levels in heart, brain, skeletal muscle and kidney. Lower levels of expression are seen in placenta, lung, liver, and pancreas. 122 X .8 a *3 E 333 3 '3 5 2 8 '5 .3 '3 a S W 8 = 8 '9‘ ._ -— H 0 c.55ws§§§ 8933£fi§§§ 3885.23'25 bau'ngeoa kb EmE—l—lmxm kb OUEmOmr— 9.5 9.5: i 7'5 if ' 7.5 f- 1!! u- I- 4 1 44 %. 4 ’ 4.4— 1a? a“ \ TOM 1 L2 TOM 1 L2 2.4—‘ .. ..‘ 2.4— -... 1.35— 1.35— B-actin B-actin Figure 21. T 0M1L2 northern analysis. The ~ 2 kb insert from clone 23808, representing the 3’ end of human T 0M1L2, was hybridized to Clontech northern blots. Panel A) shows hybridization signal of ~65 kb to the Clontech multiple tissue northern blot. Panel B) shows hybridization of ~6.5 kb to the Clontech Brain Two northern blot. B-actin blots are shown as controls for both blots. 123 Expression in the brain (Figure 213) is fairly equal in the cerebellum, cerebral cortex, medulla, occipital pole, frontal lobe, and temporal lobe. There is slightly lower expression in the putamen and a very low level signal is seen in the spinal cord. TOMILZ gene trapped ES cell lines . Gene trapping is a method of disrupting gene function that is rapid and fairly inexpensive when compared to traditional gene lmockout methods. The gene-trap relies on a vector that randomly inserts itself into the genome. The vector is equipped with a reporter gene flanked by a splice acceptor and polyadenylation sites. If the vector lands within a gene it disrupts the mRNA by creating a fusion RNA of the endogenous gene and sequence from the vector which codes for reporter and selector genes. An antibiotic resistance gene within the gene vector allows for positive selection of cells in which the vector has properly inserted. The insertion site of the gene-trap vector is determined using 5’ RACE experiments. Amplification fiom vector sequence in the fusion mRNA into the endogenous RNA allows for determination of the insertion site. The gene-trap method has been verified and used successfully to knock-out gene expression in a multitude of genes (Skarnes et al. 1992; Mitchell et al. 2001). Access to databases of gene-trapped ES cell lines are available for searching on the Internet. The cells are freely available to academic researchers from gene- trapping centers around the world. Using BLAST analysis for SMS candidate genes we determined that a cell line was available with a gene-trap in Tom112 from the BayGenomics consortium (http:l/www.baygenomics.ucs£edu). Bioinformatic 124 analysis of the 5’ RACE performed from the XG909 cell line indicated that the gene- trapping vector inserted between exons 10 and 11 in the mouse T om112 gene (Figure 22A and 22B). The gene product resulting for this gene-trapped allele in the Tom112 gene would be missing 147 amino acids from the carboxy end. XG909 genotyping and creation of XG909 mice Genotyping of the XG909 cell line (and mice subsequently developed fi‘om the cell line) was performed by Southern analysis. Digestion of mouse genomic DNA was performed with Bgll . When Southern blots are probed with a PCR product fiom the intronic sequence between Tom112 exons the wild type allele is indicated by a band of 2812 base pair (Figures 22B and 22C). The insertion of the gene-trap vector between exons 10 and 11 shifts the native 2812 base pair band up to an ~3800 base pair band (Figures 22B and 22C). Wildtype animals only have the ~2800 base pair band. Heterozygous animals display both the ~2800 base pair and ~3800 base pair bands. Homozygous targeted animals show only the 3800 base pair band. The XG909 cell line was obtained and sent to the Transgenic Animal Model Core (TAMC) at the University of Michigan for creation of gene-trapped founder mice. The XG909 cell line was expanded and then injected into C57Bl/6J for creation of a chimeric (founder) line of mice. The XG909 cell line was created from 129/OlaI-Isd embryonic stem cells. These mice have white coat color, while the C57Bl/6J mice have black coats. Chimeric animals are a mix of both cell lines and display a mottled coat with patches of black or white fur (Figure 22D). Male chimera animals were mated with C57Bl/6J females to produce an F1 line. Chimeras whose 125 germ cells were of the 129/OlaHsd lineage produced agouti colored offspring when mated with C57B1/6] animals. Agouti color results fi'om crossing of white and black coated animals. One-half of the agouti animals were expected to carry the gene- trapped Tom112 allele. The F l agouti animals were genotyped, and brother and sister carriers were mated in order to obtain an F2 generation for phenotypic analysis. Analysis of the F2 generation of Tom112 gene-trapped mice Brother sister mating of carrier F 1 gene-targeted Tom112 mice resulted in 15 F2 offspring. The gene-target occurs at an autosomal locus and should follow simple Mendilian inheritance; v. should be XG909”, 1/2 should be XG909“; and v. should be XG909"’. or the 15 F2 nrice 3 XG909”, 3 XG909”; and 9 XG909"' were obtained, which is not in a Mendelian fashion. Increasing the number of offspring may bring the inheritance back to agreement with Mendelian inheritance. The F2 offspring were physically assessed using the guide in Appendix B. Mice were thoroughly evaluated for physical, neurological, and behavioral anomalies at age 5 and 13 weeks. All physical measurements were within normal range. Total body length, body weight, and cage hang time measurements were statistically analyzed as described below. 126 Figure 22. Genetic analysis of the XG909 cell line. A) A schematic representation of the Tom112 gene with location of the pGTlle gene-targeting vector. The Tom112 gene is composed of 15 exons. The gene- trapping vector inserted into the intron between exons 10 and 11. B) A schematic of the wild type and targeted alleles in the XG909 cell line. The insert causes a Bgll frament to increase fi'om 2812 bp to ~3 800 bp. The Bgll sites are indicated in red, the probe used in Southern analysis is indicated in blue, the pGTlle vector is indicated in green. C) Southern analysis of F1 mice generated from the XG909 cell line. Mice heterozygous for the trapped allele are seen in the first and last lanes as indicated by the presence of both the 2812 bp and ~3800 bp bands. D) Photograph of the chimeric animals generated from the XG909 cell line. 127 A) , lllllll , 5'i Illllll 3 B) 59 Wild type allele = 2812 kb Q 5, Gene-trapped allele = ~3800 kb Trapped allele Wild type allele 128 The data obtained from the mice studies were grouped based on the age at evaluation and their genotype. The means and standard deviations of the groups were compared using the student t-test. Also, comparisons within the groups were performed by the analysis of variance (ANOVA). Comparisons of all the three groups at 5 weeks and 13 weeks indicated statistically significant difference between homozygous trapped and wild-type littermates only in length at 13 weeks. Total body length was measured fiom the tip of the nose to the tip of the tail while the mice were under light anesthetic. At 5 weeks, there is no statistically significant difference in body length between groups (variance of 0.78 and mean comparisons showed a student t test value of 2.7 (p=0.08)(Figure 21A). A significant difference was seen while comparing the lengths of 13 week-old mice. A significant p-value of 0.02, at Df=3 was obtained when comparing the means and standard deviation (student t-test value of 2.3). A significant difference was seen between the means from the homozygous trapped and wild type (p=0.03) and between normal and the heterozygous mice (p=0.01), though no difference can be deduced between the transgenic and the heterozygous mice (Figure 23B). This cannot be a conclusive evidence of the difference as power calculations were not performed for adequacy of the sample size for evaluation. Weight assessments performed at 5 weeks and 13 weeks showed no significant differences between the genotypes (p=0.48 and 0.315 and variance of 0.28 and 0.24 respectively)(Figures 23C and 23D). Comparison of mean values of each group by student t-test, gave a value of 2.36. Analysis of the hang time of the mice to the cage tops did not show statistical 129 significance (p=0.55 at 5 weeks and 0.0505 at 13 weeks). The ANOVA showed an R2 value of 0.24 and 0.56. (Figures 23B and 23F) SUMMARY The T 0M1L2 gene was first mapped as an EST during definition of the SMS critical region (Elsea et al. 1997). Using bioinformatic analysis and RT-PCR we were able to identify the full length sequence of the human and mouse TOMILZ/TomlLZ genes. Northern analysis revealed expression in all human adult tissues studies. Work on the TOM1L2 gene was placed on hold upon discovery of mutations in the RAII gene in some SMS patients. Mouse analysis of the Tom112 gene began when we discovered that embryonic stem cells with a gene-trapping vector in the Tom112 gene were available at no cost. We created mice from these embryonic stem cells in order to try to identify any phenotype due to dosage changes in Tom112 expression. Initial evaluation of the F2 generation of gene-trapped only showed statistical differences in total body length at 13 weeks of age between wild-type and homozygous trapped mice. Though, due to the small number of animals analyzed the statistical analysis lacks the power needed to be reliable. In order to determine whether the gene trap is having an effect on phenotype additional F2 mice are currently being generated. 130 Figure 23. Statistical analysis of XG909 mouse measurements. Mice were measured at 5 and 13 weeks for length and weight. Hang time from the cage top was also assessed. Data was analyzed using the student t-test and ANOVA with JMP software. Student t-test results are indicated by overlapping circles. AN OVA analysis is represented by the diamonds. Measurements for heterozygous mice are colored green, wild type measurements are colored yellow, and homozygous targeted are colored red. A) Comparison of length at 5 weeks. B) Comparison of length at 13 weeks. C) Comparison of weight at 5 weeks. D) Comparison of length at 13 weeks. E) Comparison of cage top hang time at 5 weeks. F) Comparison of cage top hang time at 13 weeks. 131 A) B) 155 ”E 150- I e. A l g) 145- _ 140- l 135- 130 I i r . +/- +/+ -/- BOW student's Genotype 175 170 . A E 165 M 'E) 160 _— __ c: ’3 155 150 145 140 ' .r I +/. +/+ ./- Each pair student's t-test Genotype Group 1 Group Difference Lower CL Upper CL p-Value Difference 2 +/ +/- | 1.80000 3.6426 19.95736 0.0102961 +/ -/- 7.66250 0.5980 14.72698 0.0368724 -V- +/- 4.13750 -1 . 1281 9.40305 0.1075639 Figure 23 132 C) E Weight (g) i 17.5 - 15 12.5 r++| {CD \> J Each Pair Stuthit's t-test D) E Weight (g) i 17.5 -1 15 12.5 n—q ) / <0! Each Pair Student's t-test Figure 23 continued 133 E) F) 7" 6o- l—i- ‘ \ E 50- \ E __ _ ‘5' 40‘ i” :1 30'1 20' 10' ‘ G l I ' ' r ‘ j v' ‘ +4 +/+ 4. W Studen Genotype 7G \ 60- A 50?“ I ‘ m 1 E 40' m _ g 30 20 10- l u l +/ ' +/+' _/_ Each pair Studenttesn Genotype Figure 23 continued 134 MATERIALS AND METHODS Norhtern analysis: Northern analysis was performed as described in Chatper 2. Mouse DNA isolation from tail biopsies: Tail biopsies were obtained fi'om 21 day old mice after weaning. Biopsies were digested/lysed over night in 600 pl lx TNES (10 mM Tris, pH 7.5, 400 mM NaCl, 10 mM EDTA, and 0.6% SDS), and 35 pl of 10 mg/ml overnight at 55°C. After digestion/lysis DNA was isolated using P:C:I as described in Chapter 2. Southern analysis: Eight micrograms of mouse DNA was digested with 34 U Bgll overnight at 37°C. Southern blots were created fi'om digested DNA as described in Chapter 2. Probe for southern analysis was generated by PCR amplification of the intron between Tom112 exons 10 and 11 was performed as described in chapter 2. Southern hybridization, washes, and detection were performed exactly as described in Chapter 2. Mouse assessment: Assessments of the F2 mouse generation was performed using the guide attached as Appendix C. All protocols for work with mice were approved by Animal Use Committees at MSU and VCU. Statistical analysis: Analyses were performed using the JMP software, version 5.1 with the significance value set at 0.05. 135 Chapter VII Discussion Through the research presented here significant progress has been made in the understanding of the genes that play a role in Smith-Magenis syndrome. Our efforts have resulted in construction of a contig of large insert clones across the 1.5 Mb SMS critical interval that included a transcription map of the region (Lucas et al. 2001). Analysis of SMS patients with 17p11.2 deletions allowed for two refinements of the smallest deletion region of overlap in SMS (V langos et al. 2003; Vlangos et al. 2004). The SMS critical region has been reduced fi'om 1.5 Mb to ~700 kb. Correspondingly, the number of genes mapping within this interval has also been reduced from ~40 to 11 (V langos et al. 2004). Research on samples from patients who display the SMS phenotype but in whom a deletion could not be detected with extensive FISH analysis yielded the most insight into the genetic basis of SMS. Systematic sequencing of genes mapping within the SMS critical region led to discovery of dominant fiameshifi mutations in the RA]! gene. Nine total SMS patients with RAII mutations have been reported since our initial findings in three patients (Slager et al. 2003; Bi et al. 2004; Girirajan et al. 2005). Research on the expression pattern of the DR GZ gene revealed dynamic expression in the developing mouse nervous system. Though DRGZ is no longer thought to be developmentally regulated, it likely does play an important role in the proper development of the nervous system. Though its role in SMS is still 136 inconclusive, the results of the IHC experiments presented here make it tempting to hypothesize that haploinsufficieny of DRGz may play some role in infantile hypotonia seen in SMS patients with a deletion but not seen in patients with mutations in RA]! . Future studies in the Elsea Laboratory My work in the Elsea Laboratory has helped develop an environment full of opportunity with regard to research on SMS and the genes mapping to chromosome l7p11.2. Additional research on TOMILZ, DRGZ, and RA]! will yield additional insight into human genetic disease. In order to fitrther understand the function of these genes, and any possible role they may play in human disease, the following research is proposed. TOM1L2/Tom112 Mice with gene trapped Tom112 alleles did not show an overt phenotype in either the heterozygous or homozygous state in the C5 7Bl/6J background. Many questions remain with respect to the gene-targeted Tom112 allele. Although we were able to determine the genotype of the Tom112 mice, we did not yet study whether expression levels of the Tom112 gene were affected by the gene-trapping. Because the ES cells cannot survive the positive antibiotic selection in propagation of the cell line it is unlikely, but not impossible, that the gene-trapping vector inserted incorrectly. PCR analysis and direct sequencing of the vector insertion location could be used to measure the accuracy and location of the insert site. PCR primers have 137 been designed throughout the vector and flanking genomic sequence to amplify across the insertion breakpoint. The PCR reactions to accomplish this task need to be optimized. Unlike traditional gene targeting via “knocking-out,” gene-trapping does not guarantee a null allele. Genes that have been gene-targeted can be leaky and result in a hypomorphic allele instead of a true null allele. Though no publications are reported regarding the efficiency of null alleles in gene-trapping, the groups promoting gene-trapping report that ~10% of gene-trap lines are leaky (http://genetrap.gsf.de). One report characterizing two gene trap lines that were leaky report that the actual percentage of leaky lines may be upwards of 50% (V oss et al. 1998). To determine whether a null or hypomorphic allele has been generated measurement of gene expression in Tom112 gene-trapped mice should be investigated. Tom112 expression could be analyzed fiom the normal and trapped alleles at both the RNA and protein levels. Northern analysis of RNA isolated from the Tom112 mouse line probed with 5’ sequence from the Tom112 gene should reveal the normal ~65 kb message but should also show a shifted band of RNA containing the Tom112-vector fusion RNA. Some studies using this technique have found that the normal RNA is preferentially expressed over the trapping exons resulting in normal gene expression in the homozygous trapped mice (McClive et al. 1998; Voss et al. 1998). Further complicating the analysis is that the varying expression can be tissue specific (McClive et al. 1998). Additional analysis could be performed at the protein level. We have created 138 a polyclonal antibody near the amino terminal of TOM1L2/Tom112. Protein expression of the trapped allele could be analyzed via western analysis in order to determine whether protein levels are reduced in the gene-trapped mice. The recent reports of leaky gene trapping in mice and previous reports of gene trapping in Drosophila suggest that the method may not reduce gene expression from trapped alleles as much as predicted. Even if the Tom112 gene-trapped mice are hypomorphic they may be very useful in studying Tom112 gene expression in mice. In addition to positive selection by antibiotic resistance, the gene-trapping vector allows for expression analysis by expression of the lacZ reporter gene. Gene expression can be studied by staining mouse sections or whole mount mice for LacZ expression. Expression analysis could be performed both in developing mouse embryos and adult tissues. LacZ expression could be verified by RNA ISH and/or IHC to the Tom112 gene. Conserved domains within the Tom112 gene predict that it plays a role in trafficking of proteins fi'om the trans-Golgi network to the lysozome. I have cloned the full length Tom112 sequence into the Gateway system entry vector in proper orientation for use in GFP studies. GFP studies would provide insight into the cellular localization of the Tom112 protein. Human genetic disease related to trafficking is not rare, and includes mannosidosis, Neiman-Pick disease, and Zellweger syndrome. Though the role T 0M1L2 plays in human genetic disease remains unknown, it is a good candidate gene that should be explored further. 139 DRGZ/Drgz From the data presented we hypothesize that the DRGZ/DrgZ genes likely play an important role in mammalian development. The highly conserved evolutionary sequence and the expression patterns seen in the developing mouse make DR GZ/DrgZ an interesting gene for further research as the role that DRGZ may play in SMS or other genetic disorders is still unknown. With the discovery of novel genes comes the job of characterizing function, regulation, and possible interactions with other proteins. In order to determine the biochemical fiinction of DRG2 the lab plans to use techniques to pursue any protein- protein interactions involving DRG2. Proposed experiments involve the use of yeast two hybrid assays to study possible protein-protein interactions. The yeast two hybrid assay was first reported in 1989 as a screen for binary protein interactions (Fields and Song 1989). The assay involves cloning your protein of interest into a plasmid with a DNA binding domain such as GAL4. The GAIA fusion protein is the “bait” that allows for screening for interactions with “prey" proteins. Prey proteins are created from cDNA libraries where the clones are created fitsed to a transcriptional activating domain. Fusion of the bait and prey allow for expression of a reporter gene such as B«galactosidase, or for genes allowing for growth in selective media. Positive yeast colonies are screened to determine which cDNAs interact with the bait protein. The yeast two hybrid assay is a powerful tool for determination of protein-protein interactions. The technique has been very successful in identification of proteins in cell signaling pathways (F ashena et al. 2000). The yeast two hybrid is a good assay for finding protein-protein interactions in 140 novel genes, but it is not without faults. The false positive rate can be very high, though newer techniques are being developed to reduce the number of false hits. Further, many interactions that naturally occur in vivo may not occur in this system. The yeast two hybrid system operates in the nucleus in order to activate the reporter gene. Transmembrane proteins, an extremely important class of signaling molecules, cannot be studied effectively with this assay. Interactions with proteins that require post-translational modification such as glycosylation before becoming active rrright also be missed in the traditional yeast two hybrid assay (Stagljar 2003). With these limitations in mind, the lab also plans to use coirnmunoprecipitation techniques to identify interactions that may be missed with the yeast two hybrid assay and to verify any positive interactions identified. We hypothesize that if we are able to determine the protein partners of DRGZ, we may further understand its role in mammalian development. The expression patterning in the developing mouse shows that Drg2 likely plays a role in the developing nervous system. Generation of a Drg2 knock-out mouse may reveal any phenotype associated with dosage changes in the Drg2 gene. RAH/Rail Our finding of SMS patients with dominant fiameshift mutations in the RA]! gene unraveled some of the mystery behind the etiology of SMS; however, it also created many more questions that need to be addressed. Though research will continue on other candidate genes in the SMS region, the thrust of the work in the lab will focus on research of the RAII gene. 141 Mouse modeling to study the effect of dosage changes in Rail has begun. With the hypothesis that Rail is a dosage sensitive gene, the lab has generated a line of transgenic Rail mice with multiple copies of a BAC containing the Rail gene. Current work in the lab involves assessing whether increased dosage of the Rail gene results in a phenotypic effect. Mice in the line generated carry between 2 and 5 copies of a BAC containing the Rail gene. Phenotypic measurements taken thus far have not revealed a difference between normal littermates and those with increased Rail copy number (Rebecca Slager, MSU Ph.D. Dissertation, 2004). Though increased copy number at the DNA level has been confirmed, it is unknown if the level of Rail expression is also increased. Experiments to determine expression levels at the RNA level will likely begin soon. The transgenic mice were created in conjunction with an Rail targeted knockout project. At this time, Rail has been successfully knocked-out in embryonic stem cells. The targeted cells are in queue for injection into blastocysts for creation of chimeric animals. Thorough physical and behavioral analysis of the Rail knockout animals is planned. As discussed in Chapter 1, mice deficient for the portion of mouse chromosome 11 syntenic to human chromosome 17p11.2 show a distinct phenotype overlapping with some of the features seen in the SMS phenotype (Walz et al. 2003; Walz et al. 2004). Recently, it has been shown that inactivation of the Rail gene recapitulates the physical features seen in the deletion mice (Bi et al. 2005). Behavioral studies of the Rail targeted mice were not reported. Both mouse studies reported were performed in a mixed C57BI/6J x 129Sva background (Walz et al. 142 2003; Walz et al. 2004; Bi et al. 2005). The Rail knockout mouse that the Elsea lab is creating will be in a full C57BL/6J background. There are many documented reports showing that phenotypic differences in knockout animals can vary greatly in different background strains (Dobkin et al. 2000; Fleming et al. 2001; Humphries et al. 2001). It is preferential to analyze behavior and circadian rhythm of mutant mice in a firll C57B1/6J background. Differences in phenotype may be found between the knockout mice produced in different labs. Not much is known about the biochemistry of RAIl/Rail. By sequence analysis it is postulated to act in transcriptional regulation (Seranski et al. 2001; Girirajan et al. 2005). Studies utilizing GFP show that Rail is localized to the nucleus (Elsea lab unpublished data and Bi et al. 2005). Initial studies of Rail in yeast show that there are two functional transcriptional transactivation domains present near the N-terminal end of the protein (Bi et al. 2005). It is unknown if the protein interacts directly with DNA or acts through association with other transcription factors. Vitamin A (retinol) and its derivatives are extremely important in embryonic development (Ross et al. 2000). Retinoic acid (RA) is the product of oxidation of vitamin A. Rail was originally cloned because of increased expression in response to retinoic acid (Irnai et al. 1995). However, the correlation between RA, RAll, and development is unknown. It has been shown that RA plays a direct role in the proper regulation of the Hox genes (Langston and Gudas 1994). Correct expression of the ~38 known Hox genes is crucial for proper development (Langston and Gudas 1994; Ross et al. 2000). The lab may choose to investigate whether RAIl acts in the 143 signaling pathway in response to RA resulting in Hox gene regulation. This could be performed by testing to see if RAIl binds to the promoters of any of the large number of Hox genes. The spatial and temporal expression of most of the Hox genes during development is known (Langston and Gudas 1994; Ross et al. 2000). In addition to identification of the potential DNA binding function of RAIl, determination of any protein-protein interactions would help determine potential partners in transcriptional activation and help determine the transcriptional targets of RAH/Rail. The yeast two hybrid assay or coimmunoprecipitation are two of the best techniques to study protein interactions. These are techniques that would also be of use in studying other novel genes in the lab. Determining which proteins act with RAIl/Rail rrright lead to investigation of a potential second locus for SMS. The lab has a cohort of samples from patients without detectable l7p11.2 deletions in whom mutations in RA]! have not been located. It is possible that dosage changes in genes that interact with RAIl could phenocopy the characteristics seen with haploinsufficiency of RAIl. Heterogeneity in Rubinstein-Taybi syndrome (RSTS) has recently been reported (Roelfsema et al. 2005). RSTS is characterized by mental retardation, craniofacial and skeletal anomalies, EEG abnormalities and seizures, and an increased incidence of tumor formation (Petrij et a1. 1995). Rubinstein-Taybi syndrome was originally reported associated with a hemizygous deletion of chromosome 16p 1 3.3. Research then revealed that heterozygous mutations in the CREBBP gene mapping to 16p13.3 also caused Rubinstein-Taybi (Petrij et al. 1995). It is interesting to note that the CREBBP and RAII genes are likely transcriptional co-activators that both contain a PHD domain (Kalkhoven et al. 144 2003). The PHD domain in CREBBP has been shown to be extremely important in acetyltransferase activity towards histones and CBP. This reduction in acyltransferase activity led directly to reduction in coactivator function for the transcription factor CREB (Kalkhoven et al. 2003). The authors of this study postulate that disruption of the PI-ID domain in CREBBP alone may be enough to cause RSTS (Kalkhoven et al. 2003). Research of the CREBBP gene found that it sometimes interacts with the EP300 gene as a transcriptional coactivator in the regulation of gene expression through various signal transduction pathways (Weaver et al. 1998; Roelfsema et a1. 2005). Some patients who were clinically diagnosed with RSTS do not have detectable deletions of chromosome l6p13.3 or mutations in CREBBP. Mutations in the EP300 gene were found in samples fi'om this cohort of patients by direct DNA sequencing (Roelfsema et al. 2005). It is possible that SMS may display heterogeneity similar to that seen in RSTS. Research on our cohort of SMS patients without 17p11.2 deletion or RAll mutation may reveal a second locus for SMS. Conclusion The research presented here sets the groundwork for continuing study of the genes mapping to chromosome l7p11.2. We now know that the RAII gene is dosage sensitive and that haploinsufficiency of M11 leads to most of the characteristics in the SMS phenotype. Currently, we can only speculate on the many developmental pathways that RAll could potentially play a role. Further research using in vivo and in vitro techniques, as well as physical and behavioral analysis of Rail mouse models, will help determine the true biochemical role of M11 . 145 Though M11 is the cause of the majority of the characteristics seen in the SMS phenotype we cannot yet say that it is the only gene involved. Patients with mutations in RA]! do not display short stature, infantile hypotonia, or visceral anomalies seen in some SMS patients with a l7p11.2 deletion. Further research into other genes including DRGZ and TOM 1 L2 may show that these genes are responsible for these characteristics. Clinically, we have improved the diagnostic testing for SMS by identification of a more efficient FISH probe and by helping implement commercially available RAII mutation screening. With improved testing and knowledge of SMS, patients can begin to receive intervention through proper therapies at an earlier age. Early intervention greatly increases the quality of life for SMS patients and their families. Further work using the research presented here will no doubt answer even more questions with regard to SMS and proper mammalian development. 146 Appendix A Features of Smith-Magenis Syndrome A Questionnaire for Parents and Guardians This questionnaire is designed for parents or guardians of children diagnosed with Smith-Magenis syndrome (SMS). The information provided in this survey will be used to better understand the correlation of the syndrome phenotype (the “syrnptoms”) and the genotype (the DNA content). We will use the information collected to try to determine the gene or genes that play a role in SMS. Participation in this survey is completely voluntary. All answers will be kept strictly confidential. Survey participants will not gain any direct benefit for participating in the survey, however, this information will be very important for understanding the genes involved in SMS. If at anytime you have additional questions during the course of this study about the research or your rights as a research subject, you may address them to The Michigan State University Committee on Research Involving Human Subjects at (517) 355- 2180. In the event that any problem or question arises you may contact Dr. Sarah Elsea at (517) 355-5597, or email at elsea@msu.edu. I agree to voluntarily participate in this survey titled “Features of Smith-Magenis syndrome: a questionnaire for parents and guardians.” Participants may refirse to participate or discontinue involvement AT ANY TIME without penalty. Signature Date Please Print Name This Consent form is not valid without The Institutional Review Board stamp of certification. For Office Use Only: S # F ' ID SMS ID M 9 R/I Sent? Date S Sent Date S Rec’d 147 Personal Information: Your name Your relationship to person with SMS Patient’s name Patient’s sex Patient’s date of birth Mother’s Name Father’s Name Mother's date of birth Mother's Ethnic Background Father's date of birth Father's Ethnic Background Address information is optional (see below): Street address City State Zip/Postal Code Country Telephone Number (with area code) e-mail address Pregnancy/Birth/Diagnosis: 148 Was the pregnancy considered normal? (If not, please explain any complications in the space below) Was your child carried to term? If not, how premature was your child? What was your child’s birth weight? What was your child’s length at birth? At what age was your child diagnosed with SMS? How was the diagnosis confirmed (e. g. FISH, karyotype)? In the space below please explain any other features that pertain to the pregnancy/birth of your child: 149 Physical Features: Ifyour child displays or has been diagnosed with any of the following features, please circle yes or no. If you can recall when the feature first became apparent or was diagnosed, please indicate in the space to the right. Additional comments may be made in space to the right or at the end of this section. F eature/Question Cirlce Age of onset/Additional comments Short stature Yes No Smaller than average midface Yes (hypoplasia) No General shortness of the head Yes (brachycephaly) No Large forehead (frontal bossing) Yes No Broad nasal bridge Yes N o Abnormal ear shape Yes No Down-turned upper lip Yes N o Projecting of the jaw Yes (prognathism) No Cleft lip Yes If yes, has it been corrected? and No when? Cleft palate Yes If yes, has it been corrected? and No when? Dental abnormalities Yes If yes please explain in space No below Unison of the eyebrow into one Yes large brow No (synophryS) Upslanting eyes Yes No Iris anomalies Yes No Presence of a wandering eye Yes (strabismus) N 0 Does the eye wander toward the Yes nose (esotropia) No or is wandering away from the Yes nose (exotropia) No Micro cornea Yes 150 No Hoarse Voice Yes No Short, broad hands Yes No Decreased use of hands Yes No Fusion of the fingers or toes Yes syndactyly) No Cold hands/feet Yes No Difficulty finding shoes that fit Yes No Hammer toes Yes No High arched feet (pes cavus) Yes No Flat feet (pes planus) Yes No Curvature of the spine (scoliosis) Yes Has surgery been performed to No correct/prevent? If yes, at what age? Yes No Skeletal/vertebral changes Yes If yes, please explain below No Elbow limitations (decreased Yes abaility to extend arm fully) No Abnormal gait Yes No Dry skin or psoriasis Yes N 0 Muscle weakness Yes No Decreased tolerance to exercise Yes N o In the space below please explain any other features that may pertain to the physical appearance of your child. 151 Development: Please indicate the age at which your child performed the following features. When did your child first... Ag high Smile Roll over Sit up without aid Crawl Pull themselves up to standig Stand without aid Take first steps Begin to walk Speak first word Speak in complete sentences If your child displays or has been diagnosed with any of the following features with regard to development, please circle yes or no. If you can recall when the feature first became apparent or was diagnosed, please indicate in the space to the right. Additional comments may be made in space to the right or at the end of this section. F eature/Question Circle Age of onset or diagnosis/additional comments Was your child hypotonic Yes N o (“florapr”) Do you know the developmental Yes Age? No age of your child Has your child had their IQ Yes Measurement: N 0 measured Age of measurement: Does your child attend school Yes No Is it a special school/program Yes No Does your child have an aide at Yes No 152 school Does (did)your child use sign Yes Age used? No language Did you notice a change in Yes No frustration level when your child began to communicate more effectively Medical History: If your child displays or has been diagnosed with any of the following features with regard to their medical history, please circle yes or no. If you can recall when the feature first became apparent or was diagnosed, please indicate in the space to the right. Additional comments may be made in space to the right or at the end of this section. F eature/Question Circle Age of onset or diagnosis/additional comments Vision problems due to myopia Yes No (nearsightedness) Wear glasses Yes No Retinal detachment Yes No Macular degeneration Yes N 0 Hearing impairment Yes Specify: No 153 If yes, has the impairment been Yes How? No corrects Frequent ear infections (Otitis Yes # per year No media) Have ear tubes been inserted Yes No Sinus infection Yes No Peripheral neuropathy Yes No Decreased sensitivity to pain Yes If yes, please explain which parts of No body below Muscle cramps Seizures Yes Types: No Is medication used to control? Yes Explain: No Tremors Yes No Congenital heart defects Yes No Breathing problems . Yes No Feeding problems during infancy Yes No Swallowing problems Yes No Excessive choking Yes No Excessive drooling Yes No Difficulty digesting foods Yes No Regular constipation problems Yes No Bowel obstruction Yes No Regular loose bowel movements Yes No 154 Frequent urinary tract infections Yes No Kidney abnormality Yes No Diabetes Yes N o Obesity Yes No High cholesterol Yes No Please list any medications your child receives in the space below: Please list other items you believe would be helpful below: 155 Appendix B RAII gene amplification primers RAIl Forward Primer Reverse Primer Annealing Exon Temp 1 RA1:CCI‘TCCCTCCCTCCCTCCC RA2:CACCCCTGCAGGTAGTGG 65°C 'ITCC CT G 1+2 RA3:CGCI‘ATGCTGGTGAGGAG RA4:CCGACTGGTAGGCATGAA 64 °C AGCC GATTC 2 RAS :CCATGACAGGCCGCTGAC RA6:CAGGGAGCTT GTCCTT CT G 62 °C TGC AAG I'24-3 RA7:CT GACCACAGCCAC'I'T CA RA8:CACGGACTCGGGCITGGC 63 °C TGCC CI‘T CG 3 RA9:CAGC'IT CCT CT ACT GCAAC RA 1 0:GCGAAGGCCACGGAAGG 60 °C CAG GTCTT C 3 M1 1 :GCCCGACTCCTTGCAGCT RA 1 2:CCGGTCAGCC'I'1’GGCCAC 65 °C GGAC CT CGG 3 RAl3zGGACTTCAAGCAGGAGG RA14:CAGAGAGGCGTCCGAGG 64 °C AGGTGG TGGTG *3 RA 1 5 :CACATGAAGCAGGTGAA RA 1 6:CTGGAGGCAGCCTTGGG 65 °C GAGG TGAG *3 RA] 7:CG'ITCT CT CACGGCCCT G RA18:GCCACTGGCGTTGCTGCT 68 °C AGTG GCTGC *3 RA19:GCGCT CAAAAGGAAGTC RA20:CCACAT'I'I‘ACCAGGCCI‘T 64 °C GGCC ‘ C'I'T CC ‘3 RA21:CCCI'TTCCGACAAAGACC RA22:GTGTGGCCTGGCTGTTTC 64 °C GTGG TGTG ‘3 RA23:GGAGGAGCTGGGCCTGG RA24 :CAAGTGCATCGTGGAGG 64 °C CCT C AGAGG 4 RA25:CCTGGCCACACTCCCTGG RA26:CTGCCGGAGCCTCCTTGC 68 °C AGG TGCAC 5 SHEl :TGTGCAGCTGCCGCCACT SHE2 :ACT CT GCAGATTGTCCCG 57 °C AGA 6 SHES :GCACACACCACCAACCC SHE6:AATGCCTCATITCCATGT 62 °C TCACT CC 7 SHE7:GGTGCACAGGTTGCCCIT SHE8:GCTGTCCTTGCTGTGGGT 59 °C AAT TCT 8 RA45 :GGACTGTGAAGGAGGTG RA46:GGAGTGGAGTGGAGTGT 66 °C CGAGG GGAGG 9 RA35zGAGGCTCCTGTGCTAC'IT RA36zG'I‘TGACACAGCCCAACC 64 °C TGCC ATGTGC 9 RA37:GCACATGGTTGGGCTGTG RA38bzGTCAATAAAGATACAAC 62 °C TCAAC GATTG 9 RA39:CAGCTCGATACACACAA RA40:CCGTTGTGCACCACCAGG 64 °C TCT'T C GACC 9 RA41:GGTCCCTGGTGGTGCACA RA42 :GTGGGAGACGGCTTTGTC 64 °C ACGG CT GG 9 RA43 :CCAGGACAAAGCCGTCT RA44:GACTGTGAAGTCCGAGG 57 °C CCCAC TCGTC Primer pairs noted with "' require addition of Q solution of 5 M betaine 156 APPENDIX C This mouse assessment was developed by the project PI, Dr. Sarah H. Elsea. The form usually has entried for mice up to one year old. Elsea lab physical and behavioral assessment for transgenidgene-targeted mice Name of investigator: Mouse number: Sex: DOB: Coat color: Strain: Genotype: ‘Please note any physical or behavioral abnormalies, even if not specifically indicated on this form"I Directions for performing most of the physical and behavioral tests are given at the end of this form Please perform the assays requiring anesthesia last! {“131 l“""1".’1’"".“ {{T‘ Y;:5 HP“); Vt" “writ“ Fag“ f {.4 i he. , ,- v; 3* “rt t 5‘51 i: Q. "6* t .312: ‘5' ‘ Whisker appearance and length (in mm) Skin or fur abnormalities? yes/no yes/no yes/no If so, where? Bald patches? yes/no yes/no yes/no If so, where? Condition of genital/rectal areas Condition of nails Condition of teeth Cage movement? yes/no yes/no yes/no Briefly describe Righting? Did mouse Did mouse Did mouse right itself? right itself? right itself? yes/no time in secs: time in secs: time in secs: Sound orienting? Right: Right: Right: yes/no yes/no yes/no Left: Left: Left: yes/no yes/no yes/no Pupil constriction/dilation? Right eye: Right eye: Right eye: Constriction? Constriction? Constriction? yes/no yes/no yes/no Dilation? Dilation? Dilation? yes/no yes/no yes/no Left eye: Left eye: Left eye: Constriction? Constriction? Constriction? yes/no yes/no yes/no Dilation? Dilation? Dilation? yes/no yes/no yes/no Distance from outer ear to outer ear (rmn) Whisker response? Normal/ Normal/ Normal! Normal/abnormal If abnormal, please comment abnormal abnormal abnormal Eye blink? Right eye: Right eye: Right eye: Ifno, please comment yes/no yes/no yes/no Left eye: Lefl eye: Left eye: yes/no yes/no yes/no Ear twitch? Right ear: Right ear: Right car: If no, please comment yes/no yes/no yes/no Left ear: Left ear: Left ear: Distance from outer eye to outer eye (mm) Distance from top of head to tip of nose (mm) Distance from tip of nose to tip of warm) Length of trunk from mandible (mm) If so, please comment Length of limb -front left (m) From toe to From toe to From toe to knee: knee: knee: From knee to From knee to From knee to hip: hip: hip: Length of limb-front right (m) From toe to From toe to From toe to knee: knee: knee: From knee to From knee to From knee to hip: hip: hip: Length of limb-back left (m) From toe to From toe to From toe to knee: knee: knee: From knee to From knee to From knee to hip: hip: hip: Length of limb-back right (rmn) From toe to From toe to From toe to knee: knee: knee: From knee to From knee to From knee to thigh: thigh: thigh: Wild running? yes/no yes/no yes/no 158 Ifunusual gaitz please comment 159 Freezing? yes/no yes/no yes/no If so, please comment Sniffing? yes/no yes/no yes/no Licking? yes/no yes/no yes/no Rearing? yes/no yes/no yes/no Defecation? yes/no yes/no yes/no Urination? yes/no yes/no yes/no Movement around entire cage? yes/no yes/no yes/no Postural reflex Cage shake Cage shake Was animal able to stay upright for 10s test? from side to from side to If no, please comment side: side: yes/no yes/no Cage shake up Cage shake up and down: and down: yes/no Yes/no Response to being picked up by tail Normal] Normal/ Normal/abnormal abnormal abnormal If abnormal, please comment Cage top hang test (seconds) Test 1: Test 1: Test three times (max 60 seconds) Test 2: Test 2: Test 3: Test 3: Gaiting test Normal Normal/ Normal/abnormal abnormal Hot plate test (seconds) Time: Note time in seconds and appropriate assessment (using letters) of mouse‘s response: a. jump b raise paw c. paw lick Response: d. paw shake e other (please comment) Response: Max trial time is 30 seconds Has this animal displayed normal breeding behavior? # of litters? Size of litters? Comments: Cause of death? Euthanasia or other Date of death: Age at death: 160 Description of physical tests Righting: turn the mouse onto its back and measure time in seconds for mouse to right itself; comment if mouse is unable to right itself or unusual response is noted Sound orienting: make brief, sharp sound to the right and left of mouse; note if mouse turns head in appropriate direction Pupil constriction/dilation: with a pen-light, shine a beam of light in the direction of the mouse’s eye; constriction should occur when light is shone and dilation should occur when light is removed Whisker response: lightly brush the whiskers of freely moving animal with a small paint brush and note response (normal mice will stop moving their whiskers and may turn head) Eye blink: approach the eye with the tip of a clean cotton swab and note if eye blinks (test both eyes) Ear twitch: touch with ear with the tip of a clean cotton swab and note if ear twitches (test both ears) Bodily measurements Anesthesize the mouse with isofluorane prior to conducting measurements. A cotton ball at the bottom of the jar is soaked with 300 pL of isofluorane and the mice are placed in the chamber. A cover is placed on the jar but oxygen flow is retained by not sealing the jar completely. At this isofluorane concentration, the mice take approximately 3 minutes to lose consciousness. Once the mice are asleep, we carry out the measurements, which takes <5 rrrinutes. If the mice wake up during the procedure, they are placed back in the chamber. After every third mouse, an additional 300 pL of isofluorane is added. The mice tolerate this dosage of anesthesic well and wake up and move about within 5 minutes of performing the measurements. Sensorimotor and reflexes Postural reflex: place mouse in empty cage shake from side to side and up and down for 10 seconds each; note mouse’s ability to maintain upright position Response to being picked up by tail: note response when mouse is picked up by tail for 10 8; normal response is to raise head, extend extremities and reach for ground when lowered Cage top hang test: hang mouse in empty cage with a cage top; measure time in seconds for mouse to remain hanging and note crawling movement along cage top; repeat test three times (max 60 seconds) Gaiting test: gaiting is measured by coloring each mouse’s foot with one color (using non-toxic materials) and walking mouse through a small tunnel 161 on white paper Hot-plate test: place mouse on analgesic hot plate set at 60°C and measure time in seconds for mouse to display a common response (jump, raise paw, paw lick or paw shake) or an unusual response (note and comment); max trial time is 30 secs. 162 REFERENCES --------- (2000) Technical and clinical assessment of fluorescence in situ hybridization: an ACMG/ASHG position statement. 1. Technical considerations. Test and Technology Transfer Committee. Genet Med 2:356- 361 Aasland R, Gibson TJ, Stewart AF (1995) The PHI) finger: implications for chromatin-mediated transcriptional regulation. Trends Biochem Sci 20:56-59 Alagille D, Odievre M, Gautier M, Dommergues JP (1975) Hepatic ductular hypoplasia associated with characteristic facies, vertebral malformations, retarded physical, mental, and sexual development, and cardiac murmur. J Pediatr 86:63-71 Allanson J E, Greenberg F, Smith AC (1999) The face of Smith-Magenis syndrome: a subjective and objective study. J Med Genet 36:394-397 Bailey J A, Yavor AM, Massa HF, Trask BJ, Eichler EE (2001) Segmental duplications: organization and impact within the current human genome project assembly. Genome Res 11:1005-1017 Belgrader P, Cheng J, Zhou X, Stephenson LS, Maquat LE (1994) Mammalian nonsense codons can be cis effectors of nuclear mRNA half-life. Mol Cell Biol 14:8219-8228 Bettio D, Rizzi N, Giardino D, Grugrri G, Briscioli V, Selicomi A, Carnevale F, Larizza L (1995) FISH analysis in Prader-Willi and Angelman syndrome patients. Am J Med Genet 56:224—228 Bi W, Ohyarna T, Nakarnura H, Yan J, Visvanathan J, Justice MJ, Lupski JR (2005) Inactivation of Rail in mice recapitulates phenotypes observed in chromosome engineered mouse models for Smith-Magenis syndrome. Hum Mol Genet Bi W, Saifi GM, Shaw CJ, Walz K, Fonseca P, Wilson M, Potocki L, Lupski JR (2004) Mutations of RAM , a PHD-containing protein, in nondeletion patients with Smith-Magenis syndrome. Hum Genet 115:515-524 Bi W, Yan J, Stankiewicz P, Park SS, Walz K, Boerkoel CF, Potocki L, Shaffer LG, Devriendt K, Nowaczyk MJ, Inoue K, Lupski JR (2002) Genes in a refined Smith-Magenis syndrome critical deletion interval on chromosome l7p11.2 and the syntenic region of the mouse. Genome Res 12:713-728 Botta A, Novelli G, Mari A, Novelli A, Sabani M, Korenberg J, Osborne LR, Digilio MC, Giannotti A, Dallapiccola B (1999) Detection of an atypical 7q11.23 163 deletion in Williams syndrome patients which does not include the STXlA and FZD3 genes. J Med Genet 36:478—480 Boume HR, Sanders DA, McCormick F (1991) The GTPase superfamily: conserved structure and molecular mechanism. Nature 349:117-127 Brzezinski A (1997) Melatonin in humans. N Engl J Med 336:186-195 Burk O, Worpenberg S, Haenig B, Klempnauer KH (1997) tom-1, a novel v-Myb target gene expressed in AMV- and E26-transformed myelomonocytic cells. Embo J 16:1371-1380 Campbell HD, Fountain S, McLennan IS, Berven LA, Crouch MF, Davy DA, Hooper JA, Waterford K, Chen KS, Lupski JR, Ledermann B, Young IG, Matthaei KI (2002) Fliih, a gelsolin-related cytoskeletal regulator essential for early mammalian embryonic development. Mol Cell Biol 22:3518-3526 Campbell I—ID, Fountain S, Young IG, Claudianos C, Hoheisel JD, Chen KS, Lupski JR (1997) Genomic structure, evolution, and expression of human FLII, a gelsolin and leucine-rich-repeat family member: overlap with LLGL. Genomics 42:46-54 Campbell HD, Schimansky T, Claudianos C, Ozsarac N, Kasprzak AB, Cotsell JN, Young IG, de Couet HG, Miklos GL (1993) The Drosophila melanogaster flightless-I gene involved in gastrulation and muscle degeneration encodes gelsolin-like and leucine-rich repeat domains and is conserved in Caenorhabditis elegans and humans. Proc Natl Acad Sci U S A 90:11386- l 1390 Cassidy SB, Allanson JE (2001) Management of genetic syndromes / edited by Suzanne B. Cassidy, Judith E. Allanson. Wiley-Liss, New York Chen KS, Gunaratne PH, Hoheisel JD, Young IG, Miklos GL, Greenberg F, Shaffer LG, Campbell I-ID, Lupski JR (1995) The human homologue of the Drosophila melanogaster flightless-I gene (flil) maps within the Smith- Magenis microdeletion critical region in 17p11.2. Am J Hum Genet 56:175- 1 82 Chen KS, Manian P, Koeuth T, Potocki L, Zhao Q, Chinault AC, Lee CC, Lupski JR (1997) Homologous recombination of a flanking repeat gene cluster is a mechanism for a common contiguous gene deletion syndrome. Nat Genet 1 7: 1 54-163 Chen KS, Potocki L, Lupski JR (1996) The Smith-Magenis syndrome [del(17)p11.2]: clinical review and molecular advances. Ment Retard Dev Disabil Res Rev 2: 122-129 Cheng J, Maquat LE (1993) Nonsense codons can reduce the abundance of nuclear mRNA without affecting the abundance of pre-mRNA or the half-life of 164 cytoplasmic mRNA. Mol Cell Biol 13:1892-1902 Cheng KW, Lahad JP, Gray J W, Mills GB (2005) Emerging role of RAB GTPases in cancer and human disease. Cancer Res 65:2516-2519 Cheung SW, Tishler PV, Atkins L, Sengupta SK, Modest EJ, Forget BG (1977) Gene mapping by fluorescent in situ hybridization. Cell Biol Int Rep 1:255-262 Curran ME, Atkinson DL, Ewart AK, Morris CA, Leppert MF, Keating MT (1993) The elastin gene is disrupted by a translocation associated with supravalvular aortic stenosis. Cell 73:159-168 de Almeida J C, Reis DF, Martins RR (1989) Interstitial deletion of (l7)(pl 1.2). A rrricrodeletion syndrome. Another example. Ann Genet 32: 184-186 De Leersnyder H, Bresson JL, de Blois MC, Souberbielle J C, Mogenet A, Delhotal- Landes B, Salefianque F, Munnich A (2003) Beta l-adrenergic antagonists and melatonin reset the clock and restore sleep in a circadian disorder, Smith- Magenis syndrome. J Med Genet 40:74-78 De Leersnyder H, De Blois MC, Claustrat B, Romana S, Albrecht U, Von Kleist- Retzow J C, Delobel B, Viot G, Lyonnet S, Vekemans M, Munnich A (2001a) Inversion of the circadian rhythm of melatonin in the Srrrith-Magenis syndrome. J Pediatr 139:111-116 De Leersnyder H, de Blois MC, Vekemans M, Sidi D, Villain E, Kindermans C, Munnich A (2001b) beta(1)-adrenergic antagonists improve sleep and behavioural disturbances in a circadian disorder, Snritlr-Magenis syndrome. J Med Genet 38:586-590 De Meirleir L, Seneca S, Lissens W, De Clercq I, Eyskens F, Gerlo E, Smet J, Van Coster R (2004) Respiratory chain complex V deficiency due to a mutation in the assembly gene ATP12. J Med Genet 41 : 120-124 Dobkin C, Rabe A, Dumas R, El Idrissi A, Haubenstock H, Brown WT (2000) Fmrl knockout mouse has a distinctive strain-specific learning impairment. Neuroscience 100:423-429 Donnai D, Karmiloff-Smith A (2000) Williams syndrome: from genotype through to the cognitive phenotype. Am J Med Genet 97:164-171 Dykens EM, F inucane BM, Gayley C (1997) Brief report: cognitive and behavioral profiles in persons with Snrith-Magenis syndrome. J Autism Dev Disord 27:203-21 1 Eichler EE (1998) Masquerading repeats: paralogous pitfalls of the human genome. Genome Res 8:758-762 Elsea SH, Fritz E, Schoener-Scott R, Meyn MS, Patel PI (1998) Gene for topoisomerase III maps within the Smith-Magenis syndrome critical region: 165 analysis of cell-cycle distribution and radiation sensitivity. Am J Med Genet 75: 104-108 Elsea SH, Mykytyn K, Ferrell K, Coulter KL, Das P, Dubiel W, Patel PI, Metherall JE (1999) Henrizygosity for the COP9 signalosome subunit gene, SGN3, in the Smith-Magenis syndrome. Am J Med Genet 87:342-348 Elsea SH, Purandare SM, Adell RA, Juyal RC, Davis JG, Finucane B, Magerris RE, Patel PI (1997) Definition of the critical interval for Smith-Magenis syndrome. Cytogenet Cell Genet 79:276-281 Ewart AK, Morris CA, Ensing GJ, Loker J, Moore C, Leppert M, Keating M (1993) A human vascular disorder, supravalvular aortic stenosis, maps to chromosome 7. Proc Natl Acad Sci U S A 90:3226-3230 Fashena SJ, Serebriiskii I, Golerrris EA (2000) The continued evolution of two-hybrid screening approaches in yeast: how to outwit different preys with different baits. Gene 250:1-14 Fields S, Song 0 (1989) A novel genetic system to detect protein-protein interactions. Nature 340:245-246 F inucane B, Dinigl KH, Simon EW (2001) Characterization of self-injurious behaviors in children and adults with Srrrith-Magenis syndrome. Am J Ment Retard 106:52-58 Finucane BM, J aeger ER, Kurtz MB, Weinstein M, Scott CI, Jr. (1993) Eye abnormalities in the Srrrith-Magenis contiguous gene deletion syndrome. Am J Med Genet 45:443-446 Fleming RE, Holden CC, Tomatsu S, Waheed A, Brunt EM, Britton RS, Bacon BR, Roopenian DC, Sly WS (2001) Mouse strain differences determine severity of iron accumulation in er knockout model of hereditary hemochromatosis. Proc Natl Acad Sci U S A 98:2707-2711 Fritz E, Elsea SH, Patel PI, Meyn MS (1997) Overexpression of a truncated human topoisomerase III partially corrects multiple aspects of the ataxia- telangiectasia phenotype. Proc Natl Acad Sci U S A 94:4538-4542 Gerber HP, Seipel K, Georgiev O, Hofferer M, Hug M, Rusconi S, Schaffner W (1994) Transcriptional activation modulated by homopolymeric glutamine and proline stretches. Science 263:808-811 Girirajan S, Elsas Ii LJ, Devriendt KH, Elsea SH (2005) RAIl variations in Smith- Magerris syndrome patients without l7p11.2 deletions. J Med Genet Gong W, Emanuel BS, Collins J, Kim DH, Wang Z, Chen F, Zhang G, Roe B, Budarf ML (1996) A transcription map of the DiGeorge and velo-cardio-facial syndrome minimal critical region on 22q11. Hum Mol Genet 5:789-800 166 Greenberg F, Guzzetta V, Montes de Oca-Luna R, Magenis RE, Smith AC, Richter SF, Kondo I, Dobyns WB, Patel PI, Lupski JR (1991) Molecular analysis of the Smith-Magenis syndrome: a possible contiguous-gene syndrome associated with del(17)(p11.2). Am J Hum Genet 49: 1207-1218 Greenberg F, Lewis RA, Potocki L, Glaze D, Parke J, Killian J, Murphy MA, Williamson D, Brown F, Dutton R, McCluggage C, Friedman E, Sulek M, Lupski JR (1996) Multi-disciplinary clinical study of Smith-Magenis syndrome (deletion 17p11.2). Am J Med Genet 62:247-254 Guzzetta V, Franco B, Trask BJ, Zhang H, Saucedo-Cardenas O, Montes de Oca- Luna R, Greenberg F, Chinault AC, Lupski JR, Patel PI (1992) Somatic cell hybrids, sequence-tagged sites, simple repeat polymorphisms, and yeast artificial chromosomes for physical and genetic mapping of proximal 17p. Genomics 13:551-559 Hanai R, Caron PR, Wang JC (1996) Human TOP3: a single-copy gene encoding DNA topoisomerase HI. Proc Natl Acad Sci U S A 93:3653-3657 Hol FA, Hamel BC, Geurds MP, Hansmann I, Nabben FA, Daniels 0, Mariman EC (1995) Localization of Alagille syndrome to 20p11.2-p12 by linkage analysis of a three-generation family. Hum Genet 95:687-690 Hua X, Wu J, Goldstein JL, Brown MS, Hobbs I-IH (1995) Structure of the human gene encoding sterol regulatory element binding protein-1 (SREBFl) and localization of SREBF 1 and SREBF2 to chromosomes 17p11.2 and 22q13. Genomics 25:667-673 Humphries MM, Kiang S, McNally N, Donovan MA, Sieving PA, Bush RA, Machida S, Cotter T, Hobson A, Farrar J, Humphries P, Kenna P (2001) Comparative structural and functional analysis of photoreceptor neurons of Rho-/- mice reveal increased survival on C57BL/6J in comparison to 129Sv genetic background. Vis Neurosci 18:43 7-443 Imai Y, Suzuki Y, Matsui T, Tohyarna M, Wanaka A, Takagi T (1995) Cloning of a retinoic acid-induced gene, GT1, in the embryonal carcinoma cell line P19: neuron-specific expression in the mouse brain. Brain Res Mol Brain Res 31 :1- 9 J i Y, Eichler EE, Schwartz S, Nicholls RD (2000) Structure of chromosomal duplicons and their role in mediating human genorrric disorders. Genome Res 10:597-610 J uyal RC, Figuera LE, Hauge X, Elsea SH, Lupski JR, Greenberg F, Baldini A, Patel PI (1996) Molecular analyses of 17p11.2 deletions in 62 Smith-Magenis syndrome patients. Am J Hum Genet 58:998-1007 Kalkhoven E, Roelfsema JH, Teunissen H, den Boer A, Ariyurek Y, Zantema A, Breuning MH, Hennekam RC, Peters DJ (2003) Loss of CBP 167 acetyltransferase activity by PHD finger mutations in Rubinstein-Taybi syndrome. Hum Mol Genet 12:441-450 Kallionienri OP, Kallioniemi A, Mascio L, Sudar D, Pinkel D, Deaven L, Gray J (1994) Physical mapping of chromosome 17 cosmids by fluorescence in situ hybridization and digital image analysis. Genomics 20: 125-128 Kozak M (1986) Point mutations define a sequence flanking the AUG initiator codon that modulates translation by eukaryotic ribosomes. Cell 44:283-292 Krantz ID, Piccoli DA, Spinner NB (1999) Clinical and molecular genetics of Alagille syndrome. Curr Opin Pediatr 11:558-564 Kumar S, Tomooka Y, Noda M (1992) Identification of a set of genes with developmentally down-regulated expression in the mouse brain. Biochem Biophys Res Commun 185:1155-1161 Langston AW, Gudas LJ (1994) Retinoic acid and homeobox gene regulation. Curr Opin Genet Dev 4:550-555 Li B, Trueb B (2000) DRG represents a family of two closely related GTP-binding proteins. Biochirn Biophys Acta 1491 : 196-204 Li DY, Toland AE, Boak BB, Atkinson DL, Ensing GJ, Morris CA, Keating MT (19973) Elastin point mutations cause an obstructive vascular disease, supravalvular aortic stenosis. Hum Mol Genet 6: 1021-1028 Li L, Krantz ID, Deng Y, Genin A, Banta AB, Collins CC, Qi M, Trask BJ, Kuo WL, Cochran J, Costa T, Pierpont ME, Rand EB, Piccoli DA, Hood L, Spinner NB (1997b) Alagille syndrome is caused by mutations in human J aggedl, which encodes a ligand for Notchl. Nat Genet 16:243-251 Liang Y, Wang A, Belyantseva IA, Anderson DW, Probst F J , Barber TD, Miller W, Touchman JW, J in L, Sullivan SL, Sellers JR, Camper SA, Lloyd RV, Kachar B, Friedman TB, Fridell RA (1999) Characterization of the human and mouse unconventional myosin XV genes responsible for hereditary deafness DFNB3 and shaker 2. Genorrrics 61 :243-258 Lockwood D, Hecht F, Dowman C, Hecht BK, Rizkallah TH, Goodwin TM, Allanson J (1988) Chromosome subband 17p11.2 deletion: a minute deletion syndrome. J Med Genet 25:732-737 Lohi O, Lehto VP (1998) VHS domain marks a group of proteins involved in endocytosis and vesicular trafficking. F EBS Lett 440:255-257 Lowery MC, Morris CA, Ewart A, Brothman LJ, Zhu XL, Leonard CO, Carey J C, Keating M, Brothman AR (1995) Strong correlation of elastin deletions, detected by FISH, with Williams syndrome: evaluation of 235 patients. Am J Hum Genet 57:49-53 168 Lucas RE, Vlangos CN, Das P, Patel P1, Elsea SH (2001) Genomic organisation of the approximately 1.5 Mb Srrrith-Magenis syndrome critical interval: transcription map, genomic contig, and candidate gene analysis. Eur J Hum Genet 9:892-902 Luiro K, Yliannala K, Ahtiainen L, Maunu H, J arvela I, Kyttala A, J alanko A (2004) Interconnections of CLN3, Hookl and Rab proteins link Batten disease to defects in the endoch pathway. Hum Mol Genet 13:3017-3027 Luo B, Aster JC, Hasserjian RP, Kuo F, Sklar J (1997) Isolation and fimctional analysis of a cDNA for human J agged2, a gene encoding a ligand for the Notchl receptor. Mol Cell Biol 17:6057-6067 Lupski JR (1998) Genomic disorders: structural features of the genome can lead to DNA rearrangements and human disease traits. Trends Genet 14:417-422 Lynch HJ, Wurtrnan RJ, Moskowitz MA, Archer MC, Ho MH (1975) Daily rhythm in human urinary melatonin. Science 187 :169-1 71 Maier D, Stumm G, Kuhn K, Preiss A (1992) Hairless, a Drosophila gene involved in neural development, encodes a novel, serine rich protein. Mech Dev 38: 143- 156 McClive P, Pall G, Newton K, Lee M, Mullins J, Forrester L (1998) Gene trap integrations expressed in the developing heart: insertion site affects splicing of the PTl-ATG vector. Dev Dyn 212:267-276 Meng X, Lu X, Li Z, Green ED, Massa H, Trask BJ, Morris CA, Keating MT (1998) Complete physical map of the common deletion region in Williams syndrome and identification and characterization of three novel genes. Hum Genet 1 03 :590-599 Mitchell KJ, Pinson KI, Kelly OG, Brennan J, Zupicich J, Scherz P, Leighton PA, Goodrich LV, Lu X, Avery BJ, Tate P, Dill K, Pangilinan E, Wakenight P, Tessier-Lavigrre M, Skarnes WC (2001) Functional analysis of secreted and transmembrane proteins critical to mouse development. Nat Genet 28:241-249 Morris CA, Demsey SA, Leonard CO, Dilts C, Blackburn BL (1988) Natural history of Williams syndrome: physical characteristics. J Pediatr 113:318-326 Oda T, Elkahloun AG, Meltzer PS, Chandrasekharappa SC (1997a) Identification and cloning of the human homolog (JAGl) of the rat J aggedl gene from the Alagille syndrome critical region at 20p12. Genomics 43:376-379 Oda T, Elkalrloun AG, Pike BL, Okajima K, Krantz 1D, Genin A, Piccoli DA, Meltzer PS, Spinner NB, Collins F S, Chandrasekharappa SC (1997b) Mutations in the human J aggedl gene are responsible for Alagille syndrome. Nat Genet 16:235-242 Onodera O, Oyake M, T akano H, Ikeuchi T, Igarashi S, Tsuji S (1995) Molecular 169 cloning of a firll-length cDNA for dentatorubral-pallidoluysian atrophy and regional expressions of the expanded alleles in the CNS. Am J Hum Genet 57:1050-1060 Osborne LR (1999) Williams-Beuren syndrome: unraveling the mysteries of a microdeletion disorder. Mol Genet Metab 67: 1-10 Park SS, Stankiewicz P, Bi W, Shaw C, Lehoczky J, Dewar K, Birren B, Lupski JR (2002) Structure and evolution of the Smith-Magenis syndrome repeat gene clusters, SMS-REPS. Genome Res 12:729-738 Paro R (1993) Mechanisms of heritable gene repression during development of Drosophila. Curr Opin Cell Biol 5:999-1005 Patel PI, Roa BB, Welcher AA, Schoener-Scott R, Trask BJ, Pentao L, Snipes GJ, Garcia CA, Francke U, Shooter EM, et al. (1992) The gene for the peripheral myelin protein PMP-22 is a candidate for Charcot-Marie-Tooth disease type 1A. Nat Genet 1:159-165 Petrij F, Giles RH, Dauwerse HG, Saris JJ, Hennekam RC, Masuno M, Tommerup N, van Ommen GJ, Goodman RH, Peters DJ, et al. (1995) Rubinstein-Taybi syndrome caused by mutations in the transcriptional co-activator CBP. Nature 376:348-351 Pinkel D, Straume T, Gray JW (1986) Cytogenetic analysis using quantitative, high- sensitivity, fluorescence hybridization. Proc Natl Acad Sci U S A 83:2934- 2938 Pollet N, Dhome-Pollet S, Deleuze JF, Boccaccio C, Driancourt C, Raynaud N, Le Paslier D, Hadchouel M, Meunier-Rotival M (1995) Construction of a 3.7-Mb physical map within human chromosome 20p12 ordering 18 markers in the Alagille syndrome locus. Genorrrics 27 :467-474 Potocki L, Chen KS, Park SS, Osterholm DE, Withers MA, Kimonis V, Summers AM, Meschino WS, Anyane-Yeboa K, Kashork CD, Shaffer LG, Lupski JR (2000a) Molecular mechanism for duplication 17p11.2- the homologous recombination reciprocal of the Smith-Magenis microdeletion. Nat Genet 24:84-87 Potocki L, Glaze D, Tan DX, Park SS, Kashork CD, Shaffer LG, Reiter RJ, Lupski JR (2000b) Circadian rhythm abnormalities of melatonin in Srrrith-Magenis syndrome. J Med Genet 37:428-433 Rodman S, Wadinger-Ness A (2000) Rab GTPases coordinate endocytosis. J Cell Sci 1 1 3: 1 83-192 Roelfsema JH, White SJ, Ariyurek Y, Bartholdi D, Niedrist D, Papadia F, Bacino CA, den Dunnen J T, van Ommen GJ, Breuning MI-I, Hennekam RC, Peters DJ (2005) Genetic heterogeneity in Rubinstein-Taybi syndrome: mutations in 170 both the CBP and EP300 genes cause disease. Am J Hum Genet 76:572-580 Ross SA, McCaffery PJ, Drager UC, De Luca LM (2000) Retinoids in embryonal development. Physiol Rev 80: 1021-1054 Sazuka T, Kinoshita M, Tomooka Y, Ikawa Y, Noda M, Kumar S (1992a) Expression of DRG during murine embryonic development. Biochem Biophys Res Commun 189:371-377 Sazuka T, Tomooka Y, Ikawa Y, Noda M, Kumar S (1992b) DRG: a novel developmentally regulated GTP-binding protein. Biochem Biophys Res Commun 189:363-370 Schenker T, Lach C, Kessler B, Calderara S, Trueb B (1994) A novel GTP-binding protein which is selectively repressed in SV4O transformed fibroblasts. J Biol Chem 269:25447-25453 Schenker T, Trueb B (1997) Assignment of the gene for a developmentally regulated GTP-binding protein (DRG2) to human chromosome bands 17pl3-->p12 by in situ hybridization. Cytogenet Cell Genet 79:274-275 Schmickel RD (1986) Contiguous gene syndromes: a component of recognizable syndromes. J Pediatr 109:231-241 Seranski P, Hoff C, Radelof U, Henrrig S, Reinhardt R, Schwartz CE, Heiss NS, Poustka A (2001) RM] is a novel polyglutarrrine encoding gene that is deleted in Smith-Magenis syndrome patients. Gene 270:69-76 Seroussi E, Kedra D, Kost-Alimova M, Sandberg-Nordqvist AC, Fransson 1, Jacobs JF, Fu Y, Pan HQ, Roe BA, Irnreh S, Dumanski JP (1999) TOM] genes map to human chromosome 22q13.1 and mouse chromosome 8C1 and encode proteins similar to the endosomal proteins HGS and STAM. Genonrics 57:380-388 Sesca E, Perletti GP, Binasco V, Chiara M, Tessitore L (1996) Phosphatidylethanolarnine N-methyltransferase 2 and CTP-phosphocholine cytidylyltransferase expressions are related with protein kinase C isozymes in developmental liver growth. Biochem Biophys Res Commun 229:158-162 Shaw CJ, Bi W, Lupski JR (2002) Genetic proof of unequal meiotic crossovers in reciprocal deletion and duplication of 17p11.2. Am J Hum Genet 71 :1072- 108 1 Skarnes WC, Auerbach BA, Joyner AL (1992) A gene trap approach in mouse embryonic stem cells: the lacZ reported is activated by splicing, reflects endogenous gene expression, and is mutagenic in mice. Genes Dev 6:903-918 Slager RB, Newton TL, Vlangos CN, Finucane B, Elsea SH (2003) Mutations in RAIl associated with Smith-Magenis syndrome. Nat Genet 33:466-468 Smith AC, Dykens E, Greenberg F (1998a) Behavioral phenotype of Srrrith-Magenis l 71 syndrome (del 17p11.2). Am J Med Genet 81:179-185 Smith AC, Dykens E, Greenberg F (1998b) Sleep disturbance in Smith-Magenis syndrome (del 17 p11.2). Am J Med Genet 81:186-191 Smith AC, Gropman AL, Bailey-Wilson J E, Goker-Alpan O, Elsea SH, Blancato J, Lupski JR, Potocki L (2002) Hypercholesterolemia in children with Smith- Magenis syndrome: del (l 7) (p11.2p11.2). Genet Med 4:118-125 Smith AC, McGavran L, Robinson J, Waldstein G, Macfarlane J, Zonona J, Reiss J, Lahr M, Allen L, Magenis E (1986) Interstitial deletion of (17)(p1 1.2p1 1.2) in nine patients. Am J Med Genet 24:393-414 Stagljar I (2003) Finding partners: emerging protein interaction technologies applied to signaling networks. Sci STKE 2003:pe56 Stankiewicz P, Lupski JR (2002) Genome architecture, rearrangements and genomic disorders. Trends Genet 18:74-82 Stankiewicz P, Shaw CJ, Dapper JD, Wakui K, Shaffer LG, Withers M, Elizondo L, Park SS, Lupski JR (2003) Genome architecture catalyzes nonrecurrent chromosomal rearrangements. Am J Hum Genet 72:1101-1116 Strand D, Unger S, Corvi R, Hartenstein K, Schenkel H, Kalrnes A, Merdes G, Neumann B, Krieg-Schneider F, Coy JF, et a1. (1995) A human homologue of the Drosophila tumour suppressor gene l(2)gl maps to l7p11.2-12 and codes for a cytoskeletal protein that associates with nonmuscle myosin H heavy chain. Oncogene 11:291-301 Stratton RF, Dobyns WB, Greenberg F, DeSana JB, Moore C, F idone G, Runge GH, Feldman P, Sekhon GS, Pauli RM, et al. (1986) Interstitial deletion of (17)(p11.2p11.2): report of six additional patients with a new chromosome deletion syndrome. Am J Med Genet 24:421-432 Takatsu H, Katoh Y, Shiba Y, Nakayarna K (2001) Golgi-localizing, gamma-adaptin ear homology domain, ADP-ribosylation factor-binding (GGA) proteins interact with acidic dileucine sequences within the cytoplasmic domains of sorting receptors through their VpsZ7p/Hrs/STAM (VHS) domains. J Biol Chem 276:28541-28545 Tassabehji M, Metcalfe K, Donnai D, Hurst J, Reardon W, Burch M, Read AP (1997) Elastin: genomic structure and point mutations in patients with supravalvular aortic stenosis. Hum Mol Genet 6: 1029-1036 Toulouse A, Rochefort D, Roussel J, J oober R, Rouleau GA (2003) Molecular cloning and characterization of human RAIl, a gene associated with schizophrenia. Genomics 82:162-171 Trask BJ, Mefford H, van den Engh G, Massa HF, Juyal RC, Potocki L, Finucane B, Abuelo DN, Witt DR, Magenis E, Baldini A, Greenberg F, Lupski JR, Patel 172 PI (1996) Quantification by flow cytometry of chromosome-17 deletions in Smith-Magenis syndrome patients. Hum Genet 98:710-718 wain O, Yule W (1991) A cognitive and behavioural phenotype in Williams syndrome. J Clin Exp Neuropsychol 13:232-244 Uitto J, Christiano AM, Kahari VM, Bashir MM, Rosenbloom J (1991) Molecular biology and pathology of human elastin. Biochem Soc Trans 19:824-829 Urban Z, Helms C, F ekete G, Csiszar K, Bonnet D, Munnich A, Donis-Keller H, Boyd CD (1996) 7ql 1.23 deletions in Williams syndrome arise as a consequence of unequal meiotic crossover. Am J Hum Genet 59:958-962 Vance DE (1996) Phosphatidylethanolarnine N-methyltransferase: unexpected findings from curiosity-driven research. Eur J Med Res 1:182-188 Vance DE, Walkey CJ, Cui Z (1997) Phosphatidylethanolarnine N-methyltransferase from liver. Biochim Biophys Acta 1348: 142-150 Vlangos CN, Das P, Patel PI, Elsea SH (2000) Assignment of developmentally regulated GTP-binding protein (DRG2) to human chromosome band 17p11.2 with somatic cell hybrids and localization to the Srrrith-Magenis syndrome critical interval. Cytogenet Cell Genet 88:283-285 Vlangos CN, Wilson M, Blancato J, Smith AC, Elsea SH (2004) Diagnostic FISH probes for del(l7)(pl l.2p11.2) associated with Smith-Magenis syndrome should contain the RAIl gene. Am J Med Genet A 132Az278-282 Vlangos CN, Yirn DK, Elsea SH (2003) Refinement of the Srrrith-Magenis syndrome critical region to approximately 950kb and assessment of l7p11.2 deletions. Are all deletions created equally? Mol Genet Metab 79:134-141 Voss AK, Thomas T, Gruss P (1998) Compensation for a gene trap mutation in the murine microtubule-associated protein 4 locus by alternative polyadenylation and alternative splicing. Dev Dyn 212:258-266 Walkey CJ, Donohue LR, Bronson R, Agellon LB, Vance DE (1997) Disruption of the murine gene encoding phosphatidylethanolarnine N-methyltransferase. Proc Natl Acad Sci U S A 94:12880—12885 Walz K, Caratini-Rivera S, Bi W, Fonseca P, Mansouri DL, Lynch J, Vogel H, Noebels J L, Bradley A, Lupski JR (2003) Modeling del(17)(p11.2p11.2) and dup(17)(p11.2p11.2) contiguous gene syndromes by chromosome engineering in mice: phenotypic consequences of gene dosage imbalance. Mol Cell Biol 23:3646-3655 Walz K, Spencer C, Kaasik K, Lee CC, Lupski JR, Paylor R (2004) Behavioral characterization of mouse models for Smith-Magenis syndrome and dup(l7)(pl 1.2p1 1.2). Hum Mol Genet 13:367-378 173 Wang A, Liang Y, Fridell RA, Probst FJ, Wilcox ER, Touchman JW, Morton CC, Morell RJ, Noben-Trauth K, Camper SA, Friedman TB (1998) Association of unconventional myosin W01 5 mutations with human nonsyndrorrric deafness DFNB3. Science 280:1447-1451 Wang ZG, Sheluho D, Gatti DL, Ackerman SH (2000) The alpha-subunit of the mitochondrial F(l) ATPase interacts directly with the assembly factor Atp12p. Embo J 19:1486-1493 Wang ZG, White PS, Ackerman SH (2001) Atpl 1p and Atp12p are assembly factors for the F(l)-ATPase in human mitochondria. J Biol Chem 276:30773-30778 Weaver BK, Kumar KP, Reich NC (1998) Interferon regulatory factor 3 and CREB- binding protein/p300 are subunits of double-stranded RNA-activated transcription factor DRAF 1. Mol Cell Biol 18: 1359-1368 Yan J, Walz K, Nakarnura H, Carattini-Rivera S, Zhao Q, Vogel H, Wei N, Justice MJ, Bradley A, Lupski JR (2003) COP9 signalosome subunit 3 is essential for maintenance of cell proliferation in the mouse embryonic epiblast. Mol Cell Biol 23:6798-6808 Yang SP, Bidichandani SI, F iguera LE, Juyal RC, Saxon PJ, Baldini A, Patel PI (1997) Molecular analysis of deletion (17)(pll.2p11.2) in a family segregating a 17p paracentric inversion: implications for carriers of paracentric inversions. Am J Hum Genet 60:1184-1193 174 K-