SDG | UPG | Main Page

 

clinical genomics

 

Owen M. Rennert, MD, Head, Section on Developmental Genomics

Wai-yee Chan, PhD, Adjunct Investigator

Shao-Ming Wu, PhD, Staff Scientist

Margarita Raygada, PhD, Staff Genetic Counselor

Cigdem F. Dogulu, MD, PhD, Clinical Fellow

Sergei Kvasha, PhD, Postdoctoral Fellow

Michael Y.K. Leung, PhD, Postdoctoral Fellow

Alan L.Y. Pang, PhD, Postdoctoral Fellow

Queen P. Vong, PhD, Postdoctoral Fellow

Vanessa Baxendale, MS, Research Associate

Jeffrey Benson, MS, Research Associate

Diana Alba, BS, Postbaccalaureate Fellow

Deborah Bellan, BS, Postbaccalaureate Fellow

Warren Johnson, BS, Postbaccalaureate Fellow

Lisa Ruszczyk, BS, Postbaccalaureate Fellow

Evelyn Law, BS, Summer Medical Student

Angele Nalbandian, BS, Graduate Student

Deondra Simons, BS, Howard Hughes Student

 

Given that it involves mitosis, meiosis, and post-meiotic differentiation, spermatogenesis provides an ideal system for studying the intricate regulatory mechanism of cellular proliferation and differentiation. We have obtained the profile of the genes expressed in mitotic, meiotic, and post-meiotic germ cells and thus can take advantage of a rich resource for studying development-specific mechanisms of gene expression. To understand the change in gene expression from primordial germ cells to differentiated spermatids, we mapped the transcriptome of embryonic gonads at different stages of development; that work will provide information about genetic regulation during early gonad development. An alternative approach is to study the consequences of perturbation of normal development, such as in the case of genetic mutations. Thus, we investigated the effects of a mutated luteinizing hormone receptor in cultured cells as well as in intact animals. In addition, our clinical protocols study pediatric patients with genetic and metabolic disorders, giving us access to various genetic disorders as well as providing our fellows with clinical genetics training. One such clinical protocol concerns the identification of the role of susceptibility to thrombosis in the pseudotumor cerebri of nephropathic cystinosis, which has led to the development of a screen for mutational combinations in several venous thrombosis-related molecules.

Studies of differentially expressed genes in spermatogenesis

Pang, Johnson, Rennert, Chan; in collaboration with Dym

Using Serial Analysis of Gene Expression (SAGE), we profiled the transcriptome of male germ cells at different stages of development, i.e., mitosis, meiosis, and post-meiosis, as represented by type A spermatogonia, pachytene spermatocytes, and round spermatids, respectively. We deposited the SAGE data in our publicly accessible Website (http://www.nichddirsage.nichd.nih.gov/publicsage/).  Using 15,000-cDNA microarrays from the National Institute on Aging, we also profiled expressed genes in pachytene spermatocyte and round spermatid. Based on the similarity in changes in profile of germ cells demonstrated by both techniques, we selected a number of genes for further study, hypothesizing that the distinct expression pattern of a gene reflects its specific role in different stages of spermatogenesis. An X-linked gene, Testis expressed gene 13 (Tex13), was expressed predominantly in type A spermatogonia. Further studies showed that a potential antisense transcript of Tex13, complementary to the 3´ end of the sense transcript, is present and that the expression pattern of the sense and antisense transcripts is similar but that the relative expression level of the transcripts differs at different stages of germ cells, suggesting that the expression of these transcripts is potentially subject to post-transcriptional regulation by a mechanism such as gene silencing. The regulation of expression, the biological activities, and the relationship between the sense and antisense transcripts of Tex13 are currently under study.

In a similar study, we found that, similar to Tex13, the mouse Lin-28 homolog is preferentially expressed during the mitotic stage. In contrast, SAGE analysis of the embryonic male gonad indicated that the expression patterns of the two transcripts differ during early gonadal development. Previous investigations in worms demonstrated that Lin-28 is responsible for developmental timing regulation and that its expression is subject to the action of microRNA. We speculate that Tex13 and Lin-28 exert their effects early in spermatogenesis. To elucidate the functional roles of the genes and the mechanisms of gene expression regulation, we plan to generate gene knockout mouse models.

Based on microarray analysis, we have cloned a novel isoform of the mouse Ard-1 gene, which encodes a putative co-subunit of murine N-terminal acetyltransferase 1 complex. We found this novel Ard-1 transcript expressed predominantly in meiotic male germ cells but not in nontesticular tissues, suggesting that the encoded product may exert a meiotic stage–specific effect. From the mouse genome sequence database, we identified the X-linked Ard-1 gene and another homologous transcript present on the autosomes. Surprisingly, the latter two transcripts are universally expressed but, during spermatogenesis, display expression patterns that differ from the novel Ard-1 transcript. The presence of a long unique 3´ untranslated region may subject the novel Ard-1 transcript to a germ cell–specific mode of regulation of expression. The open reading frames of the three translated products, although highly homologous, differ slightly from each other. Further characterization of the novel Ard-1 protein is under way to elucidate its role during spermatogenesis.

Pang ALY, Taylor HC, Johnson W, Alexander S, Chen Y, Su YA, Li X, Ravindranath N, Dym M, Rennert OM, Chan WY. Identification of differentially expressed genes in mouse spermatogenesis. J Androl 2003;24:59-71.

Antisense transcription in differentiating germ cells

Wu, Ruszczyk, Law, Baxendale, Pang, Rennert, Chan; in collaboration with Stitely

Antisense transcripts have been shown to be involved in transcriptional and post-transcriptional gene regulation, including genomic imprinting, X-inactivation, RNAi, RNA editing, and mRNA processing, splicing, stability, transport, and translation. A recent computational analysis identified 2,500 pairs of putative sense-antisense transcripts out of 60,770 full-length mouse cDNA (about 8 percent). An earlier study reported the identification of about 1,600 sense-antisense transcriptional units (SATs) in the human genome. With the exception of the antisense transcript of the SPEER2 gene, no other example of sense-antisense transcript pairs in mammalian germ cell has been reported. Given that SAGE libraries are derived from transcripts, the SAGE database of mouse type A spermatogonia, pachytene spermatocytes, and round spermatids, as previously generated by our laboratory, offers the opportunity to examine the presence of antisense transcripts in these cells. Examination of 62 differentially expressed genes identified in the three types of germ cell showed the presence of antisense transcripts of 41 genes (66 percent). Various types of antisense transcript can be identified, as shown in Figure 9.1. Of these 41 genes, 35 have overlapping SATs. Nine of the genes have more than one SAT pair; 12 have nonoverlapping antisense bidirectional transcripts (NABTs); and seven have both SATs and NABTs. Using orientation-specific RT-PCR, we confirmed 29 genes with an appreciable number of SAGE tags among the 41 genes. We confirmed the presence of antisense transcripts experimentally for 17 genes. We compared relative levels of expression of the SATs by quantitative real-time RT-PCR. We examined tissue distribution of the SATs of the nine genes Uba52, Calm2, Ppp1cc, Ppic, Tsg1, Tcte3, Pdcl2, Prm 1, and Prm2 and observed a wide spectrum of tissue-specific expression of SATs. The antisense transcripts of four genes, namely Uba52, Tcte3, Prm1, and Prm2, were cloned and characterized. Alignment of the nucleotide sequence of the antisense transcripts with the genomic sequence of the genes encoding the sense transcripts allowed localization of the antisense transcripts to exons and introns of the sense gene and to pseudogene, intronic, and intergenic sequences. A number of antisense transcripts contain appreciable open reading frames that could be encoding novel proteins. The data show that antisense transcription occurs more frequently in differentiating germ cells than in somatic cells. We are identifying in vitro and in vivo systems suitable for testing functional activities of the cloned antisense transcripts.

Wu SM, Baxendale V, Chen Y, Li X, Pang ALY, Stitely T, Munson PJ, Leung MYK, Ravindranath N, Dym M, Rennert OM, Chan WY. Analysis of mouse germ cell transcriptome at different stages of spermatogenesis: biological significance. Genomics 2004;84:971-981.

Novel activity of cytochrome c oxidase in germ cell development

Wu, Ruszczyk, Johnson, Baxendale, Law, Rennert, Chan

Extensive apoptosis occurs during spermatogenesis, particularly in spermatogonia and spermatocytes, but the mechanism has not been fully defined. Recent studies suggested that cytochrome c plays a critical role in germ cell apoptosis. Cytochrome c exists in loosely and tightly bound pools attached to the inner mitochrondrial membrane by association with cardiolipin, a complex that must first be disrupted to generate a soluble pool of this protein. Once cytochrome c is soluble, permeabilization of the outer mitochondrial membrane with calcium or Bax is sufficient to allow extrusion of the protein into the cytosol, resulting in the onset of apoptosis. Analysis of our germ cell SAGE database showed the presence of the testis-specific cytochrome c with the same differential expression pattern as the testis-specific isoform cytochrome c oxidase VIb-2. Cloning of the transcript corresponding to the most abundant novel tag in the three SAGE libraries showed that the transcript corresponds to mouse cytochrome c oxidase subunit-3 (Cox 3). We cloned a further abundant novel tag, which corresponded to another subunit of cytochrome c oxidase (subunit 1, Cox 1). A search of the germ cell SAGE libraries revealed that all 13 subunits of cytochrome c oxidase, complex IV of the respiratory chain, are expressed at appreciable levels and demonstrate comparable differential expression patterns in these cells. On the other hand, some of the subunits of complexes II, III, and V of the respiratory chain are absent, and only 12 of the 43 subunits of complex I have the same differential expression pattern. The results imply that cytochrome c oxidase may not function as a component of the respiratory chain in germ cells. We hypothesize that cytochrome c oxidase oxidizes cytochrome c, causing its release from the inner mitochrondrial membrane. Transfer of the soluble cytochrome c into the cytosol results in amplification of calcium-dependent apoptosis. We are in the process of testing this hypothesis by using in vitro cell models.

Functional genomic studies of gonad development and sexual dimorphism of the brain

Baxendale, Vong, Bellan, Alba, Rennert, Chan; in collaboration with Lau, Su

To understand the mechanisms that regulate the transition of primordial germ cells to gonocytes and the initiation of sexual dimorphism, we are profiling the genes expressed in embryonic gonads of the mouse. We are using SAGE to examine male and female embryonic gonads at embryonic day 10.5 (E10.5), E11.5, E12.5, E13.5, E15.5, and E17.5 and the mesonephros at E13.5, E15.5, and E17.5. We have completed an analysis of about 152,000 SAGE tags for each of the male E10.5, E11.5, and E12.5 gonads. The tags identify 24,460, 214,762, and 26,378 genes in the E10.5, E11.5, and E12.5 gonads, respectively. The 10 most abundant tags represent cytochrome b-245 beta polypeptide (Cybb), Cyp2e1 cytochrome P450 (COX5b), Translationally controlled tumor protein (Tctp1), hemoglobin Y beta-like embryonic chain (Hbb-y), tubulin alpha 2 (Tuba2), four ribosomal proteins (X-linked S4, L26, 29, and L10A), and one uncharacterized cDNA. The gene encoding embryonic hemoglobins beta and that encoding the X-linked ribosomal protein S4 are either absent from or expressed at a very low level in germ cells, indicating that both are specific for activities of the embryonic gonads. Among the 10 most abundant tags present in embryonic gonads but absent from germ cells, four represent genes encoding hemoglobin chains, namely, Hba-X, Hbb-b1, Hbb-Y, and Hba-a1. The role of the hemoglobin genes in early embryonic gonad development is not presently understood. However, a recent paper that examined gene expression in embryonic lens also demonstrated the expression of hemoglobin isoforms (Hba-a1, Hba-X, Hbb-b1, Hbb-b2, and Hbb-Y). Even though fewer tags were sequenced for the germ cell SAGE libraries (about 111,000 tags for each germ cell type versus about 152,000 tags for each embryonic gonad stage), we found (Wu SM et al., 2004) more specific genes in germ cells than in embryonic gonads (4,946 germ cell specific-gene tags versus 4,755 embryonic gonad specific-gene tags). These preliminary observations have important implications for the regulation of gonad development and germ cell differentiation. We are continuing our analysis of expressed genes in male gonads at later embryonic ages and in female gonads and mesonephros.

We are interested in exploring the role of sex chromosome–linked genes in sexual dimorphism of the brain. We have generated human cDNA sex-linked gene microarrays of 724 X-linked and 28 Y-linked human genes on glass slides. We will use the microarrays to profile expressed genes in the brain and gonad of male and female mice at E10.5, E13.5, E15.5, E17.5, newborn mice, and adult mice. We will compare the expression profile of the male and the female brain and gonad at different time points and then examine the relationship between gonad development and onset of sexual dimorphism of the brain.

Chan WY, Rennert OM. Molecular aspects of sexual differentiation. Curr Mol Med 2002;2:25-37.

Wu SM, Baxendale V, Chen Y, Li X, Pang ALY, Stitely T, Munson PJ, Leung MYK, Ravindranath N, Dym M, Rennert OM, Chan WY. Analysis of mouse germ cell transcriptome at different stages of spermatogenesis: biological significance. Genomics 2004;84:971-981.

The role of Ddx3y in mouse gonad and germ cell development

Vong, Wu, Rennert, Chan; in collaboration with Lau, Dym

Among Y-encoded genes, Ddx3y (formerly known as Dby) has been considered a strong candidate as a mediator of Y chromosome function in spermatogonial proliferation. The tag representing Ddx3y is present in the spermatogonial SAGE library. We subsequently cloned full-length Ddx3y cDNA from mouse type A spermatogonia in the form of two transcripts that differ only in the length of the 3´untranslated region due to the presence of a different polyadenylation signal. Both the long form (Dby-L) and the short form (Dby-S) of Ddx3y are ubiquitously expressed in nontesticular tissues except the ovary, with Dby-L expressed especially in brain and heart. We observed a higher level of expression of Dby-L and Dby-S in type A spermatogonia than in spermatocytes and spermatids. In addition to the two variants, we found two other types of alternatively spliced transcripts containing nonoverlapping sequences of 48 and 120 nucleotides, respectively.

We compared the expression of Ddx3y and its X and autosomal homologs Ddx3 and D1Pas1 (formerly known as PL10) in gonads at E10.5 with 18, 24, and 30 days after birth. Expression of Ddx3y rises from E10.5 to E17.5 and then declines slowly after birth. In embryonic gonads, expression of Ddx3 is five- to 10-fold higher than that of Ddx3y but drops after birth. It has been reported that D1Pas1 is expressed only in pachytene spermatocytes and round spermatids, and we noted its first expression at postnatal day 18, reaching its highest level from day 24 to 30, when spermatocytes and spermatids first appear. Alignment of Ddx3y, Ddx3, and D1Pas1 reveals significant homology; Ddx3y and Ddx3 show 90 and 84 percent identity at the amino acid and nucleic acid level, respectively. The differential expression of these three genes despite their high degree of sequence homology suggests that their differential action may be dictated by their less homologous 5´ and 3´ untranslated sequences. The differential regulation of expression of the three genes is currently under investigation.

Genetic, physiological, and biochemical effects of disease-causing mutations of the luteinizing hormone receptor

Leung, Bellan, Baxendale, Wu, Rennert, Chan; in collaboration with Fechner, Steinbach, Su

Constitutive activating mutations of the human luteinizing hormone/chorionic gonadotropin receptor (hLHR) cause familial male-limited precocious puberty (FMPP), a noncentral form of gonadotropin-independent precocious puberty. Even though constitutive production of testosterone, which occurs in FMPP patients, is not known to be tumorigenic, we identified two FMPP patients who developed testicular neoplasia. Another study of testicular tumor by Dr. Shenker’s group at Northwestern University also identified an activating mutation of the hLHR in a subgroup of testicular tumor patients. We hypothesize that an activating mutation of hLHR turns on genes that ultimately may lead to development of testicular tumors. To study the potential tumorigenic effect of a constitutively activated LHR, we generated an in vitro cell model. MA-10 cells, a mouse Leydig cell line, were transfected with hLHR carrying activating mutations. Using cDNA microarrays of the NIH mouse 23K genes, we compared the profile of expressed genes in cells expressing the mutated hLHR with that of control cells. Of 22,684 genes, 10,353 display high-quality informative expression data across all microarray images, including MA10 (control) and the experimental samples, in both forward and reverse labeling. Asp578Gly, the most common activating mutation, induces 42 differentially expressed genes, including Bscl2, Tcfe2a, Ercc3, Crsp9, Gabarapl1, Il10rb, and Hsh2. The somatic activating mutation Asp578His, found in some patients with testicular tumors, induces 74 differentially expressed genes. Several genes, including Tcfe2a, Crsp9, and Rpo1-2, are shared by cells expressing hLHR-Asp578Gly and those expressing hLHR-Asp578His. All three genes are involved in transcription. We plan to investigate further their relationship with mutated hLHR.

The impact of an activating mutation of the hLHR has always been considered limited to sexual development of the patient. The abnormal social behavior of patients was thought to be secondary to precocious sexual maturation. It is known that LHR is expressed in the brain. We speculate that the abnormal behavior of FMPP patients is due to expression of the mutated LHR in the brain. To test this hypothesis and study the impact of constitutively activated LHR on spermatogenesis as well as on sexual and neurological development, we generated a transgenic mouse strain that expresses a fusion protein of enhanced green fluorescent protein (EGFP) to an hLHR mutant with substitution Asp578Gly or Asp578His. Interestingly, all the transgenic mice expressing the mutated hLHR are females. The reason for this phenomenon is unclear. We are currently investigating the impact of the mutated receptor on embryonic growth and development.

The antithesis of FMPP is Leydig cell hypoplasia (LCH). In LCH patients, a mutation inactivates the LHR, resulting in reduced production of testosterone, which causes hypergonadotrophic hypogonadism or male pseudohermaphroditism. A novel missense mutation A340T identified in a patient with LCH results in substitution of Ile-114 by Phe, which affects one of the leucine-rich repeats (LRRs) in the extracellular domain of the hLHR. The mutant receptor fails to trigger cAMP production upon hCG stimulation in transient expression studies. A fluorescence microscopy study of the fusion protein of receptor with green fluorescent protein revealed that the mutation does not affect trafficking of the mutated receptor but rather affects binding of the hormone by the receptor. To study the effect of the mutation on the conformation of the receptor, we generated a computer model of the LRR, which clearly demonstrates the conformational effect of the mutation and may explain the impact of mutations on the biological activity of other proteins with LRRs.

Leung MLY, Al-Muslim O, Wu SM, Azizs A, Inam S, Awadh M, Rennert OM, Chan WY. A novel missense homozygous inactivating mutation in the fourth transmembrane helix of the luteinizing hormone receptor in leydig cell hypoplasia. Amer J Med Genet 2004;130A:146-153.

Salameh W, Shoucair M, Guo TB, Zahed L, Wu SM, Rennert OM, Chan WY. Leydig cell hypoplasia due to inactivation of luteinizing hormone receptor by a novel homozygous nonsense truncation mutation in the seventh transmembrane domain. Mol Cell Endocrinol 2004, in press.

Role of susceptibility to thrombosis in pseudotumor cerebri of nephropathic cystinosis

Dogulu, Raygada, Chan, Rennert; in collaboration with Gahl, Kaiser

Given our recent findings regarding genetic susceptibility to thrombosis in pseudotumor cerebri (PTC), we instituted a clinical protocol to study the role of thrombosis susceptibility in the development of PTC in nephropathic cystinosis patients. We are screening nephropathic cystinosis patients who develop PTC and control nephropathic cystinosis patients without PTC by using a thrombosis susceptibility screening panel: thrombin time (TT), activated partial thromboplastin (APT), activated protein C resistance, serum levels of protein C, protein S, antithrombin III, fibrinogen, factor VIII, factor IX, factor XI, total homocysteine, antiphospholipid antibodies (ACA and Lupus AC), Factor V Leiden mutation, Factor V G1628A polymorphism, Factor V R2 allele, Prothrombin 20210 mutation, and 5,10-methylenetetrahydrofolate reductase (MTHFR) gene C677T polymorphism.

To date, we have recruited two patients with pseudotumor cerebri with pre-existing nephropathic cystinosis. The thrombosis screening panel revealed shortened TT in both patients. Thrombin time measures the rate of fibrin monomer polymerization and is the most sensitive screening test for decreases or abnormalities in fibrinogen. The shortened TT demonstrates an acceleration of fibrin monomer polymerization leading to a thrombotic tendency.

Dogulu CF, Kansu T, Leung MYK, Baxendale V, Wu SM, Ozguc M, Chan WY, Rennert OM. Evidence for genetic susceptibility to thrombosis in idiopathic intracranial hypertension. Thromb Res 2003;111:389-395.

Dogulu CF, Tsilou E, Rubin B, FitzGibbon EJ, Kaiser MI, Rennert OM, Gahl WA. Idiopathic intracranial hypertension in cystinosis. J Pediat 2004;145:673-678.

Method evolved for recognition of thrombophilia (MERT)

Dogulu, Chan, Rennert

Venous thrombosis affects one in every 1,000 individuals annually and is one of the leading causes of mortality and morbidity, resulting in approximately 300,000 hospitalizations and 50,000 fatalities a year in the United States alone. It is, however, an avoidable disease if currently available prophylactic treatment is instituted. To avoid the development of venous thromboembolism, it would be beneficial to estimate the thrombotic risk of individuals in order to develop stratification protocols for risk-adapted prophylaxis. We have devised an approach (patent pending) that will allow rapid screening of an array of mutations and polymorphisms reported to be risk factors for the development of venous thrombosis, thus permitting highly accurate assessment in several ethnic populations.

Clinical protocol on the studies of pediatric patients with genetic and metabolic disorders

Raygada, Dogulu, Rennert; in collaboration with Kaler, Stratakis

We have instituted a clinical protocol that provides care for patients with a variety of rare genetic disorders, offers an opportunity for training in clinical genetics, dysmorphology, and metabolic genetics, and serves to spearhead the development of new research protocols on particular aspects of diagnosis and treatment of specific genetic diseases. We evaluate patients with a broad spectrum of metabolic and genetic conditions, offer genetic counseling services to patients and their families to assess risk, and provide information on preventive measures and testing options. The disorders we study include chromosomal and Mendelian disorders of childhood and/or adults, congenital anomalies and/or birth defects, dysmorphic syndromes, familial cancer syndromes, multifactorial disorders, and metabolic abnormalities. Patients and/or family members with genetic disorders may provide DNA for storage and/or testing. The overall purpose of the protocol is to support NICHD’s training and research missions by expanding the spectrum of diseases that can be seen in our clinics and wards and to recruit a diverse population of patients and/or biological samples to provide NICHD investigators and trainees with hands-on experience related to diagnosis, management, follow-up, treatment, and genetic counseling. The protocol also provides an opportunity to evaluate patients with unusual or challenging genetic and metabolic disorders who may not be eligible for an existing research protocol; often it is not possible to determine protocol eligibility without prospective evaluation conducted at the NIH. Such patients may be of exceptional educational value for clinical staff at all levels, and their evaluation may catalyze the recognition of new disease processes and need for research initiatives. Furthermore, the evaluation of such challenging patients is necessary to sustain the analytic and innovative faculties of clinical research staff at all levels, from student to clinical fellow to senior staff member.

Ifon ET, Pang ALY, Johnson W, Cashman K, Zimmerman S, Muralidhar S, Chan WY, Casey J, Rosenthal LJ. U94 alters FN1 and ANGPTL4 gene expression and inhibits tumorigenesis of prostate cancer cell line PC3. Cancer Cell Internat 2004, in press.

Ohta S, Lai EW, Pang ALY, Brouwers FM, Chan WY, Eisenhofer G, de Krijger R, Ksinantova L, Blazicek P, Breza J, Kvetnansky R, Wesley RA, Pacak K. Down-regulation of metastasis suppressor gene in malignant pheochromocytoma. Internat J Cancer 2004, Nov 2; [Epub ahead of print].

Raygada M, Rennert OM. Congenital generalized lipodystrophy: profile of the disease and gender differences in two siblings. Clin Genet 2004, in press.

Roth J, Raygada M, Devaskar S, Montrose-Rafizadeh C, LeRoith D. Insulin and the brain. In: Adelman G, Smith BH, eds. Encyclopedia of Neuroscience, 3rd edition, [on CD Rom], Oxford, UK: Elsevier; 2003.

Stratakis C, Rennert OM. Turner syndrome: an update. The Endocrinologist 2004, in press.

COLLABORATORS

Martin Dym, PhD, Georgetown University Medical Center, Washington, DC

Patricia Fechner, MD, Stanford University, Palo Alto, CA

William Gahl, MD, PhD, Clinical Director, NHGRI, Bethesda, MD

Muriel I. Kaiser, MD, Ophthalmic Genetics and Visual Function Branch, NEI, Bethesda, MD

Stephen Kaler, MD, MPH, Clinical Director, NICHD, Bethesda, MD

Chris Y.F. Lau, PhD, University of California, San Francisco, CA

Malcolm M. Martin, MD, Georgetown University, Washington, DC

Neelakanta Ravindranath, PhD, Center for Scientific Review, NIH, Bethesda, MD

Peter Steinbach, PhD, Center for Information Technology, NIH, Bethesda, MD

Timothy Stitely, MS, Unit on Computer Support Services, NICHD, Bethesda, MD

Constantine Stratakis, MD, DSc, Heritable Disorders Branch, NICHD, Bethesda, MD

Yan Su, MD, PhD, Loyola University, Chicago, IL

For further information, contact chanwy@mail.nih.gov