146 results on '"Lawrence C. Layman"'
Search Results
2. Identification of Rare Genetic Variants in the PCDH Genetic Family in a Cohort of Transgender Women
- Author
-
John Theisen, Lynn P. Chorich, Hongyan Xu, James Knight, Hyung-Goo Kim, and Lawrence C. Layman
- Published
- 2023
3. Unexplained Female Infertility Associated with Genetic Disease Variants
- Author
-
Michael P. Dougherty, Alexandra M. Poch, Lynn P. Chorich, Zoe A. Hawkins, Hongyan Xu, Robert A. Roman, Haitao Liu, Soumia Brakta, Hugh S. Taylor, James Knight, Hyung-Goo Kim, Michael P. Diamond, and Lawrence C. Layman
- Subjects
General Medicine - Published
- 2023
4. A rigorous in silico genomic interrogation at 1p13.3 reveals 16 autosomal dominant candidate genes in syndromic neurodevelopmental disorders
- Author
-
Afif Ben-Mahmoud, Kyung Ran Jun, Vijay Gupta, Pinang Shastri, Alberto de la Fuente, Yongsoo Park, Kyung Chul Shin, Chong Ae Kim, Aparecido Divino da Cruz, Irene Plaza Pinto, Lysa Bernardes Minasi, Alex Silva da Cruz, Laurence Faivre, Patrick Callier, Caroline Racine, Lawrence C. Layman, Il-Keun Kong, Cheol-Hee Kim, Woo-Yang Kim, and Hyung-Goo Kim
- Subjects
Cellular and Molecular Neuroscience ,Molecular Biology - Abstract
Genome-wide chromosomal microarray is extensively used to detect copy number variations (CNVs), which can diagnose microdeletion and microduplication syndromes. These small unbalanced chromosomal structural rearrangements ranging from 1 kb to 10 Mb comprise up to 15% of human mutations leading to monogenic or contiguous genomic disorders. Albeit rare, CNVs at 1p13.3 cause a variety of neurodevelopmental disorders (NDDs) including development delay (DD), intellectual disability (ID), autism, epilepsy, and craniofacial anomalies (CFA). Most of the 1p13.3 CNV cases reported in the pre-microarray era encompassed a large number of genes and lacked the demarcating genomic coordinates, hampering the discovery of positional candidate genes within the boundaries. In this study, we present four subjects with 1p13.3 microdeletions displaying DD, ID, autism, epilepsy, and CFA. In silico comparative genomic mapping with three previously reported subjects with CNVs and 22 unreported DECIPHER CNV cases has resulted in the identification of four different sub-genomic loci harboring five positional candidate genes for DD, ID, and CFA at 1p13.3. Most of these genes have pathogenic variants reported, and their interacting genes are involved in NDDs. RT-qPCR in various human tissues revealed a high expression pattern in the brain and fetal brain, supporting their functional roles in NDDs. Interrogation of variant databases and interacting protein partners led to the identification of another set of 11 potential candidate genes, which might have been dysregulated by the position effect of these CNVs at 1p13.3. Our studies define 1p13.3 as a genomic region harboring 16 NDD candidate genes and underscore the critical roles of small CNVs in in silico comparative genomic mapping for disease gene discovery. Our candidate genes will help accelerate the isolation of pathogenic heterozygous variants from exome/genome sequencing (ES/GS) databases.
- Published
- 2022
5. A rigorous
- Author
-
Afif, Ben-Mahmoud, Kyung Ran, Jun, Vijay, Gupta, Pinang, Shastri, Alberto, de la Fuente, Yongsoo, Park, Kyung Chul, Shin, Chong Ae, Kim, Aparecido Divino, da Cruz, Irene Plaza, Pinto, Lysa Bernardes, Minasi, Alex, Silva da Cruz, Laurence, Faivre, Patrick, Callier, Caroline, Racine, Lawrence C, Layman, Il-Keun, Kong, Cheol-Hee, Kim, Woo-Yang, Kim, and Hyung-Goo, Kim
- Abstract
Genome-wide chromosomal microarray is extensively used to detect copy number variations (CNVs), which can diagnose microdeletion and microduplication syndromes. These small unbalanced chromosomal structural rearrangements ranging from 1 kb to 10 Mb comprise up to 15% of human mutations leading to monogenic or contiguous genomic disorders. Albeit rare, CNVs at 1p13.3 cause a variety of neurodevelopmental disorders (NDDs) including development delay (DD), intellectual disability (ID), autism, epilepsy, and craniofacial anomalies (CFA). Most of the 1p13.3 CNV cases reported in the pre-microarray era encompassed a large number of genes and lacked the demarcating genomic coordinates, hampering the discovery of positional candidate genes within the boundaries. In this study, we present four subjects with 1p13.3 microdeletions displaying DD, ID, autism, epilepsy, and CFA.
- Published
- 2022
6. OR06-3 ESR1 Pathogenic Variant With Incomplete Estrogen Insensitivity
- Author
-
Lynn P Chorich, Michael P Diamond, Janet E Hall, Kenneth S Korach, Lawrence C Layman, Yin Li, Haitao Liu, and Robert A Roman
- Subjects
Endocrinology, Diabetes and Metabolism - Abstract
Introduction Estrogen is vital to human reproduction and acts primarily through two receptors: estrogen receptor alpha (ERα, encoded by ESR1) and estrogen receptor beta (ERβ, encoded by ESR2). Surprisingly, very few human ESR1 pathogenic variants have been recognized, despite the existence of >1000 different androgen receptor pathogenic variants causing androgen insensitivity syndrome. Functional analysis of an ESR1 homozygous missense variant (p.Glu375His) in the ERα ligand-binding domain (LBD) showed a 240-fold decrease in estrogen response in a patient with complete estrogen insensitivity and abnormal pubertal development. We predict that less severe ESR1 variants will present with unexplained infertility. Previously, we performed whole exome sequencing on 200 females with unexplained infertility and identified 4 likely pathogenic ESR1 heterozygous missense variants confirmed by Sanger sequencing, including one variant (p.Thr313Met) in the LBD. In this study, we performed in vitro functional analysis to evaluate if the p.Thr313Met likely pathogenic variant found in a patient with unexplained infertility causes incomplete estrogen insensitivity. Hypothesis We hypothesize that the p.Thr313Met likely pathogenic variant in the ERa LBD will result in incomplete estrogen insensitivity. Methods The p.Thr313Met variant plasmid was constructed using site-directed mutagenesis (SDM), and confirmed by Sanger sequencing. An in vitro cell model was used to evaluate estrogenic activity. HepG2 cells were transiently transfected with WT ESR1, p.Thr313Met, or p.Glu375His plasmid, along with the Firefly estrogen response element (ERE) luciferase reporter (3X ERE TATA luc) and Renilla luciferase (pRL-TK luc) plasmids using the Effectene transfection protocol. Cells were treated with 17β-estradiol (0.01, 0.1, 1, 10, and 100 nM). A dual luciferase reporter assay was used to compare relative Firefly and Renilla luciferase activity between WT ESR1, p.Thr313Met, and p.Glu375His plasmids using a luminometer. Estrogen dose response curves were generated, and a two-way ANOVA with Tukey's test was performed to compare the relative luciferase activity across WT and variant plasmids using GraphPad Prism v.9. Differences were considered significant at p Results Estrogen response curves were generated for WT, p.Thr313Met, and p.Glu375His plasmids. The p.Thr313Met variant showed lower overall activity with a right shifted dose response curve with a statistically significant decrease in relative luciferase activity in comparison to WT (p Conclusion We have identified a human ESR1 variant that impairs estrogen signaling in vitro, suggesting incomplete estrogen insensitivity as a putative mechanism for unexplained infertility. The identification of ESR1 pathogenic variants and the evaluation of their functionality could provide improved diagnosis and genetic counseling for patients with unexplained infertility. Presentation: Saturday, June 11, 2022 12:00 p.m. - 12:15 p.m.
- Published
- 2022
7. Evidence of Sex Differences in the Endothelin Pathway in Diabetes
- Author
-
Cassandra C. Derella, Jeffrey Thomas, Lawrence C. Layman, and Ryan A. Harris
- Subjects
Genetics ,Molecular Biology ,Biochemistry ,Biotechnology - Published
- 2022
8. Heterogeneous Inheritance in Autism Genes Shared Across Neurodevelopmental and Neuromuscular Disorders in Consanguineous Singlets
- Author
-
Mohammad-Reza Ghasemi, Hossein Sadeghi, Farzad Hashemi-Gorji, Reza Mirfakhraie, Vijay Gupta, Afif Ben-Mahmoud, Katayoon Razjouyan, Shadab Salehpour, Seyed Hassan Tonekaboni, Mehdi Dianatpour, Davood Omrani, Lawrence C. Layman, Hyung-Goo Kim, and Mohammad Miryounesi
- Abstract
Background Autosomal recessive genetic disorders are leading health concerns in countries with consanguineous marriage. However, previously there was no strong association of consanguinity with autosomal dominant or X-linked inheritance patterns. Whether this would be the case in singlet consanguineous families remained unexplored. Methods The current study aimed to identify the disease genes and their inheritance patterns in 12 independent Iranian consanguineous singlet families with syndromic autism. Results Exome sequencing revealed two missense and two nonsense variants (one in each gene: IQSEC2, FOXG1, DMD, and CHKB) in four out of 12 families, thereby offering an aetiologic diagnosis. Autism, intellectual disability, learning disability, developmental delay, and behavioral problems were common features in these four families, and three individuals presented with impaired motor skills such as abnormal gait. Muscular dystrophy coupled with hypotonia were noted in two individuals, whereas in the remaining two families ADHD combined with language/speech delay were present. Along with the two X-linked (IQSEC2 and DMD), one autosomal dominant (FOXG1) and one autosomal recessive genes (CHKB) were found in these four consanguineous families. Two genes-DMD and CHKB- are known to be mainly involved in muscular dystrophy. Limitation: our study significance is limited by the small sample size, 12 individuals, and lack of functional experiment on a VUS on the DMD variant. Conclusion Our findings illustrate the mixed inheritance pattern in consanguineous singlet families and highlight the association of autism spectrum disorder with muscular dystrophy, both of which are seemingly not related.
- Published
- 2022
9. ESR1 HINGE REGION VARIANT IN A PATIENT WITH UNEXPLAINED INFERTILITY
- Author
-
Robert A. Roman, Haitao Liu, Lynn Chorich, Yin Li, Janet E. Hall, Michael P. Diamond, Kenneth S. Korach, and Lawrence C. Layman
- Subjects
Reproductive Medicine ,Obstetrics and Gynecology - Published
- 2022
10. PREVALENCE OF DIGENIC DISEASE IN PATIENTS DIAGNOSED WITH IDIOPATHIC HYPOGONADOTROPIC HYPOGONADISM/KALLMANN SYNDROME
- Author
-
Alexandra Poch, Michael P. Dougherty, Robert A. Roman, Lynn Chorich, Zoe Hawkins, and Lawrence C. Layman
- Subjects
Reproductive Medicine ,Obstetrics and Gynecology - Published
- 2022
11. Loss of Kallmann syndrome-associated gene WDR11 disrupts primordial germ cell development by affecting canonical and non-canonical Hedgehog signalling
- Author
-
Hyung-Goo Kim, Yeonjoo Kim, Paris Ataliotis, Jiyoung Lee, Soo-Hyun Kim, Dae-Won Kim, Nigel A. Brown, Dorothy C. Bennett, and Lawrence C. Layman
- Subjects
endocrine system ,biology ,Somatic cell ,Kallmann syndrome ,medicine.disease ,CREB ,Cell biology ,PTCH2 ,medicine.anatomical_structure ,Knockout mouse ,medicine ,biology.protein ,Hedgehog ,Germ cell ,Hypogonadotrophic hypogonadism - Abstract
Mutations ofWDR11are associated with Kallmann syndrome (KS) and congenital hypogonadotrophic hypogonadism (CHH), typically caused by defective functions of gonadotrophin-releasing hormone (GnRH) neurones in the brain. We previously reported that Wdr11 knockout mice show profound infertility with significantly fewer germ cells present in the gonads. To understand the underlying mechanisms mediated by WDR11 in these processes, we investigated the effects ofWdr11deletion on primordial germ cell (PGC) development. Using live-tracking of PGCs and primary co-cultures of genital ridges (GR), we demonstrated thatWdr11-deficient embryos contained reduced numbers of PGCs which had delayed migration due to significantly decreased proliferation and motility. We found primary cilia-dependent canonical Hedgehog (Hh) signalling was required for proliferation of the somatic mesenchymal cells of GR, while primary cilia-independent non-canonical Hh signalling mediated by Ptch2/Gas1 and downstream effectors Src and Creb was required for PGC proliferation and migration, which was disrupted by the loss of function mutations of WDR11. Therefore, canonical and non-canonical Hh signalling are differentially involved in the development of somatic and germ cell components of the gonads, and WDR11 is required for both of these pathways operating in parallel in GR and PGCs, respectively, during normal PGC development. Our study provides a mechanistic link between the development of GnRH neurones and germ cells mediated by WDR11, which may underlie some cases of KS/CHH and ciliopathies.
- Published
- 2020
12. Genetics of agenesis/hypoplasia of the uterus and vagina: narrowing down the number of candidate genes for Mayer-Rokitansky-Küster-Hauser Syndrome
- Author
-
John A. Capra, Hugh S. Taylor, Sasha Mikhael, Madison Morton, Michael J. Friez, Amy C. Lossie, Lynn P. Chorich, Kerlene Berwick Tam, Souhrid Mukherjee, John A. Phillips, Lawrence C. Layman, Hyung-Goo Kim, James R. Knight, and Sonal Dugar
- Subjects
Male ,Candidate gene ,46, XX Disorders of Sex Development ,Translocation Breakpoint ,Chromosomal translocation ,Biology ,Translocation, Genetic ,Article ,Congenital Abnormalities ,03 medical and health sciences ,symbols.namesake ,Mice ,Exome Sequencing ,Genetics ,Animals ,Humans ,Gene ,Mullerian Ducts ,Genetics (clinical) ,Exome sequencing ,030304 developmental biology ,Sanger sequencing ,0303 health sciences ,030305 genetics & heredity ,Breakpoint ,Uterus ,Genetic Variation ,Human genetics ,Vagina ,symbols ,Female - Abstract
PURPOSE: Mayer–Rokitansky–Küster–Hauser (MRKH) syndrome consists of congenital absence of the uterus and vagina and is often associated with renal, skeletal, cardiac, and auditory defects. The genetic basis is largely unknown except for rare variants in several genes. Many candidate genes have been suggested by mouse models and human studies. The purpose of this study was to narrow down the number of candidate genes. METHODS: Whole exome sequencing was performed on 111 unrelated individuals with MRKH; variant analysis focused on 72 genes suggested by mouse models, human studies of physiological candidates, or located near translocation breakpoints in t(3;16). Candidate variants (CV) predicted to be deleterious were confirmed by Sanger sequencing. RESULTS: Sanger sequencing verified 54 heterozygous CV from genes identified through mouse (13 CV in 6 genes), human (22 CV in seven genes), and translocation breakpoint (19 CV in 11 genes) studies. Twelve patients had ≥ 2 CVs, including four patients with two variants in the same gene. One likely digenic combination of LAMC1 and MMP14 was identified. CONCLUSION: We narrowed 72 candidate genes to 10 genes that appear more likely implicated. These candidate genes will require further investigation to elucidate their role in the development of MRKH.
- Published
- 2020
13. SUN-738 Establishing the Link Between Genetic Variations of Estrogen Receptor 2 and Unexplained Infertility
- Author
-
Janet E. Hall, Lynn P. Chorich, Adam B. Burkholder, Michael P. Diamond, Kenneth S. Korach, Sophia Halassy, Kerlene Bertwick Tam, Lawrence C. Layman, and Sasha Mikhael
- Subjects
Sanger sequencing ,Infertility ,medicine.drug_class ,business.industry ,Endocrinology, Diabetes and Metabolism ,media_common.quotation_subject ,medicine.medical_treatment ,Steroid Hormones and Receptors ,medicine.disease ,Bioinformatics ,symbols.namesake ,Hypergonadotropic hypogonadism ,Estrogen ,medicine ,symbols ,Missense mutation ,Ovulation induction ,Steroid and Nuclear Receptors ,business ,Ovulation ,AcademicSubjects/MED00250 ,media_common ,Unexplained infertility - Abstract
Background: Unexplained or idiopathic infertility comprises approximately 30% of couples who present with infertility. This has led to investigations seeking to determine the cause(s) of this important diagnosis of exclusion. Estrogen’s role in reproduction has been well- established. Estrogens bind to two hormone receptors (namely estrogen receptor-alpha and estrogen receptor-beta), which are distributed differentially throughout the body. Specifically, the estrogen receptor-beta, coded by the Estrogen Receptor 2 (ESR2) gene, is highly expressed in granulosa cells and growing follicles. The one female patient reported with an ESR2 mutation presented with hypergonadotropic hypogonadism. However, subfertility with inefficient ovulation and resistance to exogenous ovulatory stimulation is seen in an ESR2 knockout mouse model. We therefore hypothesized that less severe ESR2 variants could lead to a normal female phenotype and pubertal development but could be a cause subfertility. Methods: DNA samples from 200 women with unexplained infertility were obtained from the Assessment of Multiple Intrauterine Gestations from Ovarian Stimulation (AMIGOS) clinical trial, which investigated optimal ovulation induction medications for unexplained infertility. These samples were subjected to targeted next-generation sequencing (NGS) for the ESR2 gene. Likely pathogenic variants that occurred with a minor allele frequency of < 0.01 in the gnomAD database and a Combined Annotation Dependent Depletion (CADD) score of > 20 were selected for confirmation by Sanger sequencing. Results: From the 200 patient samples, five heterozygous missense variants and one heterozygous in-frame deletion identified by targeted NGS were confirmed by Sanger sequencing. Further studies will need to be performed in vitro to confirm the likely pathogenicity of these variants. Conclusion: These studies raise the possibility that If these variants in ESR2 that impair estrogen signaling, they could be a potential newly recognized etiology of unexplained infertility in women with unexplained infertility. Conclusion: These studies raise the possibility that variants in ESR2 that impair estrogen signaling could be a potential newly recognized etiology of unexplained infertility in women.
- Published
- 2020
14. Long-Term Follow-Up and Treatment of a Female With Complete Estrogen Insensitivity
- Author
-
Laurel A. Coons, Hyung-Goo Kim, John A. Katzenellenbogen, Kenneth S. Korach, Lynn P. Chorich, Janet E. Hall, Lawrence C. Layman, and Soumia Brakta
- Subjects
0301 basic medicine ,Delayed puberty ,Adult ,medicine.medical_specialty ,Adolescent ,medicine.drug_class ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Diethylstilbestrol ,Drug Resistance ,Biochemistry ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Endocrinology ,Internal medicine ,Chlorocebus aethiops ,medicine ,Vaginal smear ,Animals ,Humans ,Amenorrhea ,Clinical Research Articles ,Puberty, Delayed ,business.industry ,Biochemistry (medical) ,Estrogen Receptor alpha ,Hep G2 Cells ,Polycystic ovarian disease ,Ovarian Cysts ,030104 developmental biology ,030220 oncology & carcinogenesis ,COS Cells ,Female ,medicine.symptom ,Gonadotropin ,business ,Luteinizing hormone ,Estrogen receptor alpha ,medicine.drug ,Hormone ,Follow-Up Studies - Abstract
ContextWe previously reported the first female with a causative ESR1 gene variant, who exhibited absent puberty and high estrogens. At age 15 years, she presented with lower abdominal pain, absent breast development, primary amenorrhea, and multicystic ovaries. The natural history of complete estrogen insensitivity (CEI) in women is unknown.ObjectiveThe purpose of this report is to present the neuroendocrine phenotype of CEI, identify potential ligands, and determine the effect of targeted treatment.DesignWe have characterized gonadotropin pulsatility and followed this patient’s endocrine profile and bone density over 8 years. Seventy-five different compounds were tested for transactivation of the variant receptor. A personalized medicine approach was tailored to our patient.SettingAcademic medical center.Patient or Other ParticipantsA 24-year-old adopted white female with CEI.Intervention(s)The patient was treated with diethylstilbestrol (DES) for approximately 2.5 years.Main Outcome Measure(s)Induction of secondary sexual characteristics.ResultsLuteinizing hormone (LH) pulse studies demonstrated normal pulsatile LH secretion, elevated mean LH, and mildly elevated mean follicle-stimulating hormone (FSH) in the presence of markedly increased estrogens. DES transactivated the variant ESR1 in vitro. However, DES treatment did not induce secondary sexual characteristics in our patient.ConclusionsTreatment with DES was not successful in our patient. She remains hypoestrogenic despite the presence of ovarian cysts with a hypoestrogenic vaginal smear, absent breast development, and low bone mineral mass. Findings suggest additional receptor mechanistic actions are required to elicit clinical hormone responses.
- Published
- 2020
15. Comparative Genomic Mapping Implicates LRRK2 for Intellectual Disability and Autism at 12q12, and HDHD1, as Well as PNPLA4, for X-Linked Intellectual Disability at Xp22.31
- Author
-
Kim, Jonathan D. J. Labonne, Terri M. Driessen, Marvin E. Harris, Il-Keun Kong, Soumia Brakta, John Theisen, Modibo Sangare, Lawrence C. Layman, Cheol-Hee Kim, Janghoo Lim, and Hyung-Goo
- Subjects
microdeletion ,LRRK2 ,autism ,HDHD1 ,PNPLA4 ,intellectual disability ,parkinsonism ,XLID ,12q12 ,Xp22.31 ,nervous system diseases - Abstract
We report a genomic and phenotypic delineation for two chromosome regions with candidate genes for syndromic intellectual disability at 12q12 and Xp22.31, segregating independently in one family with four affected members. Fine mapping of three affected members, along with six unreported small informative CNVs, narrowed down the candidate chromosomal interval to one gene LRRK2 at 12q12. Expression studies revealed high levels of LRRK2 transcripts in the whole human brain, cerebral cortex and hippocampus. RT-qPCR assays revealed that LRRK2 transcripts were dramatically reduced in our microdeletion patient DGDP289A compared to his healthy grandfather with no deletion. The decreased expression of LRRK2 may affect protein–protein interactions between LRRK2 and its binding partners, of which eight have previously been linked to intellectual disability. These findings corroborate with a role for LRRK2 in cognitive development, and, thus, we propose that intellectual disability and autism, displayed in the 12q12 microdeletions, are likely caused by LRRK2. Using another affected member, DGDP289B, with a microdeletion at Xp22.31, in this family, we performed the genomic and clinical delineation with six published and nine unreported cases. We propose HDHD1 and PNPLA4 for X-linked intellectual disability in this region, since their high transcript levels in the human brain substantiate their role in intellectual functioning.
- Published
- 2020
- Full Text
- View/download PDF
16. PGT-M for Couples with a Single-Gene Disorder
- Author
-
Lawrence C. Layman
- Subjects
Pediatrics ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Genetic counseling ,Duchenne muscular dystrophy ,Grandparent ,Preimplantation genetic diagnosis ,medicine.disease ,Medicine ,Medical history ,Muscular dystrophy ,Family history ,business ,Genetic testing - Abstract
For all patients considering fertility, a detailed family history should be ascertained from the patient and partner (if there is a partner). Ideally, this would be a full pedigree of family members up to three generations from the individual. This entails getting the medical history of parents and sibs (first degree); aunts, uncles, and grandparents (second degree); and first cousins (third degree). This may be best done by a genetic counselor, geneticist, a nurse, or extended provider trained in performing pedigrees. It is important to ascertain the symptoms, specific diagnosis, and age of diagnosis of the disorder. Only knowing there is a history of muscular dystrophy, for example, is not sufficient to perform genetic testing. There are many different types of muscular dystrophy, so it cannot be assumed that Duchenne muscular dystrophy is present—it must be confirmed. Medical records with precise genetic testing results are required if there is to be any genetic testing of embryos. If there is a history of cancer, the age at diagnosis is more important than the age of death with regard to familial cancer, which is more likely under the age of 50 at diagnosis.
- Published
- 2020
17. Genetic Spectrum of Syndromic and Non-Syndromic Hearing Loss in Pakistani Families
- Author
-
Sher Alam Khan, Marcus Dittrich, Tobias Müller, Hamed Nawaz, Julia Doll, Imran Khan, Lawrence C. Layman, Laura Kühlewein, Naseer Ahmad, Susanne M. Kolb, Noor Muhammad, Hyung-Goo Kim, Il-Keun Kong, Jonathan D J Labonne, Ajmal Khan, Franz Rüschendorf, Tabea Röder, Saadullah Khan, Indrajit Nanda, Barbara Vona, Linda Schnapp, Thomas Haaf, Michaela A.H. Hofrichter, and Tyler D. Graves
- Subjects
Adult ,Male ,0301 basic medicine ,MYO15A ,lcsh:QH426-470 ,Adolescent ,Population ,Genes, Recessive ,Consanguinity ,Deafness ,030105 genetics & heredity ,Biology ,Compound heterozygosity ,Article ,Frameshift mutation ,Genetic Heterogeneity ,genetic diagnosis ,03 medical and health sciences ,Gene mapping ,Exome Sequencing ,Ethnicity ,Genetics ,otorhinolaryngologic diseases ,Humans ,Family ,Genetic Predisposition to Disease ,Pakistan ,ddc:610 ,Child ,Hearing Loss ,education ,Genetics (clinical) ,Exome sequencing ,Aged ,education.field_of_study ,Genetic heterogeneity ,Homozygote ,Middle Aged ,Pedigree ,lcsh:Genetics ,030104 developmental biology ,Cardiovascular and Metabolic Diseases ,Mutation ,Female ,genome-wide linkage analysis ,Genome-Wide Association Study - Abstract
The current molecular genetic diagnostic rates for hereditary hearing loss (HL) vary considerably according to the population background. Pakistan and other countries with high rates of consanguineous marriages have served as a unique resource for studying rare and novel forms of recessive HL. A combined exome sequencing, bioinformatics analysis, and gene mapping approach for 21 consanguineous Pakistani families revealed 13 pathogenic or likely pathogenic variants in the genes GJB2, MYO7A, FGF3, CDC14A, SLITRK6, CDH23, and MYO15A, with an overall resolve rate of 61.9%. GJB2 and MYO7A were the most frequently involved genes in this cohort. All the identified variants were either homozygous or compound heterozygous, with two of them not previously described in the literature (15.4%). Overall, seven missense variants (53.8%), three nonsense variants (23.1%), two frameshift variants (15.4%), and one splice-site variant (7.7%) were observed. Syndromic HL was identified in five (23.8%) of the 21 families studied. This study reflects the extreme genetic heterogeneity observed in HL and expands the spectrum of variants in deafness-associated genes.
- Published
- 2020
18. The Use of Whole Exome Sequencing in a Cohort of Transgender Individuals to Identify Rare Genetic Variants
- Author
-
Viji Sundaram, Megan E. Sullivan, Hyung-Goo Kim, J. Graham Theisen, Mary S. Filchak, Lawrence C. Layman, Lynn P. Chorich, and James R. Knight
- Subjects
Male ,0301 basic medicine ,Gender dysphoria ,Candidate gene ,Mutation, Missense ,lcsh:Medicine ,Genomics ,Development ,Biology ,Transgender Persons ,Article ,Frameshift mutation ,03 medical and health sciences ,symbols.namesake ,0302 clinical medicine ,Exome Sequencing ,Genetic variation ,Transgender ,medicine ,Humans ,lcsh:Science ,Frameshift Mutation ,10. No inequality ,Exome sequencing ,Sanger sequencing ,Genetics ,Multidisciplinary ,lcsh:R ,Chromosome Mapping ,Genetic Variation ,Sequence Analysis, DNA ,Sex Determination Processes ,medicine.disease ,Alternative Splicing ,030104 developmental biology ,symbols ,lcsh:Q ,Female ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
Approximately 0.5–1.4% of natal males and 0.2–0.3% of natal females meet DSM-5 criteria for gender dysphoria, with many of these individuals self-describing as transgender men or women. Despite recent improvements both in social acceptance of transgender individuals as well as access to gender affirming therapy, progress in both areas has been hampered by poor understanding of the etiology of gender dysphoria. Prior studies have suggested a genetic contribution to gender dysphoria, but previously proposed candidate genes have not yet been verified in follow-up investigation. In this study, we expand on the topic of gender identity genomics by identifying rare variants in genes associated with sexually dimorphic brain development and exploring how they could contribute to gender dysphoria. To accomplish this, we performed whole exome sequencing on the genomic DNA of 13 transgender males and 17 transgender females. Whole exome sequencing revealed 120,582 genetic variants. After filtering, 441 variants in 421 genes remained for further consideration, including 21 nonsense, 28 frameshift, 13 splice-region, and 225 missense variants. Of these, 21 variants in 19 genes were found to have associations with previously described estrogen receptor activated pathways of sexually dimorphic brain development. These variants were confirmed by Sanger Sequencing. Our findings suggest a new avenue for investigation of genes involved in estrogen signaling pathways related to sexually dimorphic brain development and their relationship to gender dysphoria.
- Published
- 2019
19. Evaluation of Mayer-Rokitansky-Kuster-Hauser (MRKH) Patient Families by Whole Genome Sequencing
- Author
-
Lynn P. Chorich, Michael P. Dougherty, and Lawrence C. Layman
- Subjects
Whole genome sequencing ,Genetics ,Mayer rokitansky kuster hauser ,Endocrinology, Diabetes and Metabolism ,From Bench to Bedside: Genetics, Development and Cell Signaling in Endocrinology ,Biology ,AcademicSubjects/MED00250 ,Genetics and Development (including Gene Regulation) - Abstract
Introduction: MRKH is a characterized by the congenital absence of the uterus and vagina in 46,XX individuals. A subset of these patients also has associated renal, skeletal, cardiac and/or auditory defects. Familial cases suggest a genetic component, but to date only pathogenic variants in WNT4 and HNF1B have been confirmed. We hypothesize that de novo heterozygous variants in candidate genes will be present in some patients with MRKH. Methods: DNAs from 30 quads (an MRKH proband and three relatives) were subjected to whole genome sequencing (WGS), and heterozygous variants in coding regions with < 0.02 frequency were filtered by two different methods. In the first approach, variants were filtered by 1) top consequence variant (splice site, stop-gain, frameshift, and missense, respectively); 2) impact score; 3) mapping quality; 4) cytobands; 5) intolerance; 6) de novo variants; and 7) plausibility based on familial genotype. The second approach considered only heterozygous variants found in the proband and absent in all other family members, which were then filtered by top consequence (splice donor and acceptor sites, stop-gain, frameshift). Results: Five pedigrees were excluded for inadequate sequence in one or more individuals. 55,033 variants in coding regions with < 2% frequency were identified in the 25 remaining quads for analysis. Using the first approach, 42 candidate gene variants in 32 genes were identified - 12 splice variants, 10 stop-gains, 15 frameshift variants and 5 missense variants. Of these, MUC22 contained 2 missense variants from different families. Additionally, DICER1 had multiple splice variants and is essential for mouse urogenital tract development. In the second approach, 39 candidate genes were identified—6 splice variants in 6 genes, 18 stop-gains in 17 genes, and 17 frameshift variants in 16 genes. Zinc finger genes (ZNF418, ZNF646, ZNF135, and ZNF772) comprised the most frequent class of the 39 genes. Two genes (MIR4436A and ZNF418) contained attractive variants in two different families. Conclusion: WGS has been shown to improve detection of gene variants in coding regions, more so than whole exome sequencing (WES). We previously performed WES on 111 MRKH probands without family members and analyzed variants in candidate genes suggested by mouse and preliminary human studies. Interestingly, in this study, only three genes overlapped with previously suspected candidate genes. Here, we identified new candidates based upon potential deleteriousness. These candidate genes will be studied further in our families to determine their role in Mullerian development.
- Published
- 2021
20. Would combined glucocorticoid and gonadotropin therapy improve pregnancy rates in women with primary ovarian insufficiency?
- Author
-
Lawrence C. Layman
- Subjects
Pregnancy ,business.industry ,medicine.drug_class ,Primary ovarian insufficiency ,Physiology ,Reflection ,medicine.disease ,lcsh:Diseases of the genitourinary system. Urology ,lcsh:RC870-923 ,lcsh:Gynecology and obstetrics ,Text mining ,medicine ,Gonadotropin ,business ,Glucocorticoid ,lcsh:RG1-991 ,medicine.drug - Published
- 2020
21. Genetics of hypogonadotropic Hypogonadism—Human and mouse genes, inheritance, oligogenicity, and genetic counseling
- Author
-
Hyung-Goo Kim, Soo-Hyun Kim, Lawrence C. Layman, Afif Ben-Mahmoud, Alexandra Poch, and Erica Louden
- Subjects
Male ,0301 basic medicine ,Candidate gene ,Kallmann syndrome ,Genetic counseling ,Inheritance Patterns ,Genetic Counseling ,Nerve Tissue Proteins ,030209 endocrinology & metabolism ,Biology ,Biochemistry ,Mice ,03 medical and health sciences ,symbols.namesake ,0302 clinical medicine ,Endocrinology ,Hypogonadotropic hypogonadism ,medicine ,Animals ,Humans ,Receptor, Fibroblast Growth Factor, Type 1 ,Molecular Biology ,Gene ,Sanger sequencing ,Genetics ,Extracellular Matrix Proteins ,Genetic Variation ,Oligogenic Inheritance ,Kallmann Syndrome ,medicine.disease ,Phenotype ,Disease Models, Animal ,030104 developmental biology ,symbols ,Receptors, LHRH - Abstract
Hypogonadotropic hypogonadism, which may be normosmic (nHH) or anosmic/hyposmic, known as Kallmann syndrome (KS), is due to gonadotropin-releasing hormone deficiency, which results in absent puberty and infertility. Investigation of the genetic basis of nHH/KS over the past 35 years has yielded a substantial increase in our understanding, as variants in 44 genes in OMIM account for ~50% of cases. The first genes for KS (ANOS1) and nHH (GNRHR) were followed by the discovery that FGFR1 variants may cause either nHH or KS. Associated anomalies include midline facial defects, neurologic deficits, cardiac anomalies, and renal agenesis, among others. Mouse models for all but one gene (ANOS1) generally support findings in humans. About half of the known genes implicated in nHH/KS are inherited as autosomal dominant and half are autosomal recessive, whereas only 7% are X-linked recessive. Digenic and oligogenic inheritance has been reported in 2-20% of patients, most commonly with variants in genes that may result in either nHH or KS inherited in an autosomal dominant fashion. In vitro analyses have only been conducted for both gene variants in eight cases and for one gene variant in 20 cases. Rigorous confirmation that two gene variants in the same individual cause the nHH/KS phenotype is lacking for most. Clinical diagnosis is probably best accomplished by targeted next generation sequencing of the known candidate genes with confirmation by Sanger sequencing. Elucidation of the genetic basis of nHH/KS has resulted in an enhanced understanding of this disorder, as well as normal puberty, which makes genetic diagnosis clinically relevant.
- Published
- 2021
22. Berberine Inhibits Uterine Leiomyoma Cell Proliferation via Downregulation of Cyclooxygenase 2 and Pituitary Tumor-Transforming Gene 1
- Author
-
Lawrence C. Layman, Ayman Al-Hendy, Cristina McCrary, Jie Min, Yen Hao Chen, Ricardo Azziz, Hsiao Li Wu, Tung Yueh Chuang, and Michael P. Diamond
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Berberine ,Cell ,Down-Regulation ,Mice, Nude ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Nude mouse ,Downregulation and upregulation ,In vivo ,Internal medicine ,medicine ,Animals ,Cell Proliferation ,030219 obstetrics & reproductive medicine ,Leiomyoma ,biology ,Cell growth ,business.industry ,Obstetrics and Gynecology ,biology.organism_classification ,Securin ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Cyclooxygenase 2 ,Apoptosis ,Cell culture ,Uterine Neoplasms ,Cancer research ,Heterografts ,Female ,business ,Immortalised cell line - Abstract
We previously demonstrated that berberine (BBR) inhibits cell proliferation and induces apoptosis in a human uterine leiomyoma (UtLM) cell line but does not demonstrate a significant cytotoxic effect in a normal human uterine smooth muscle (UtSM) cell line. However, the mechanisms of this inhibition are unclear. Of note, cyclooxygenase 2 (COX2) and pituitary tumor-transforming gene 1 (PTTG1) are overexpressed in human uterine leiomyomata and are involved in the pathogenesis of uterine fibroids (UFs). We found that COX2 and PTTG1 were overexpressed in UtLM and that BBR decreased COX2 and PTTG1 expression in UtLM cells. Our data support that UtLM and UtSM are immortalized cell lines without phenotypic alterations from parental cell types and suggest that COX2 and PTTG1 are molecular targets for BBR. However, studies in these cell lines may not reveal all activities of BBR in vivo, and we therefore proceeded to test in this report the antitumor effects of BBR in an UF nude mouse xenograft model. When UF nude mice were killed at 7 weeks, tumor weight in controls was 45 ± 7 mg versus 20 ± 3 mg ( P < .05) in the low-dose (5 mg/kg) and 7 ± 3 mg ( P < .01) in the high-dose (10 mg/kg) BBR groups, respectively. Expression of proliferation markers, cell cycle-related genes, and UF-related genes was downregulated in tumors. No unusual behavioral changes and no signs of kidney or liver damage were observed in the animals with BBR treatment. In conclusion, our data suggests that (a) COX2 and PTTG1 are molecular targets for BBR and (b) BBR is potentially an effective and safe anti-UF agent.
- Published
- 2017
23. Disruption of PHF21A causes syndromic intellectual disability with craniofacial anomalies, epilepsy, hypotonia, and neurobehavioral problems including autism
- Author
-
Marianne McGuire, Daryl A. Scott, Jason Brown, Sonal Mahida, Hyung-Goo Kim, Jonathan D J Labonne, Eric Muller, Arthur Sorlin, Vincent des Portes, Duchwan Ryu, Gerard Bénédicte, Fan Xia, Zeyaul Islam, Suneeta Madan-Khetarpal, Sakkubai Naidu, Jill A. Rosenfeld, Il-Keun Kong, Dianalee McKnight, Erin Torti, Lawrence C. Layman, Jacqueline Gutierrez, Franz Rüschendorf, Gaetan Lesca, Yline Capri, Oliver Hummel, Cheol-Hee Kim, Ange Line Bruel, and Prasanna R. Kolatkar
- Subjects
Male ,lcsh:RC346-429 ,Craniofacial Abnormalities ,Intellectual disability (ID) ,Missense mutation ,Potocki-Shaffer syndrome (PSS) ,Child ,Exome sequencing ,Genetics ,0303 health sciences ,030305 genetics & heredity ,Neurodevelopmental disorders ,Brain ,Syndrome ,Autism spectrum disorder (ASD) ,Hypotonia ,Psychiatry and Mental health ,Autism spectrum disorder ,Child, Preschool ,Muscle Hypotonia ,Female ,medicine.symptom ,Haploinsufficiency ,BHC80 ,Adolescent ,Nonsense mutation ,Histone Deacetylases ,Frameshift mutation ,Intrinsically disordered region (IDR) ,03 medical and health sciences ,Developmental Neuroscience ,Protein Domains ,Intellectual Disability ,medicine ,Humans ,AT Hook domain ,Amino Acid Sequence ,RNA, Messenger ,Autistic Disorder ,Molecular Biology ,lcsh:Neurology. Diseases of the nervous system ,030304 developmental biology ,Behavior ,Epilepsy ,business.industry ,Research ,Infant, Newborn ,Infant ,medicine.disease ,PHF21A ,Cardiovascular and Metabolic Diseases ,KDM1A ,Mutation ,Autism ,business ,Developmental Biology - Abstract
Background PHF21A has been associated with intellectual disability and craniofacial anomalies based on its deletion in the Potocki-Shaffer syndrome region at 11p11.2 and its disruption in three patients with balanced translocations. In addition, three patients with de novo truncating mutations in PHF21A were reported recently. Here, we analyze genomic data from seven unrelated individuals with mutations in PHF21A and provide detailed clinical descriptions, further expanding the phenotype associated with PHF21A haploinsufficiency. Methods Diagnostic trio whole exome sequencing, Sanger sequencing, use of GeneMatcher, targeted gene panel sequencing, and MiSeq sequencing techniques were used to identify and confirm variants. RT-qPCR was used to measure the normal expression pattern of PHF21A in multiple human tissues including 13 different brain tissues. Protein-DNA modeling was performed to substantiate the pathogenicity of the missense mutation. Results We have identified seven heterozygous coding mutations, among which six are de novo (not maternal in one). Mutations include four frameshifts, one nonsense mutation in two patients, and one heterozygous missense mutation in the AT Hook domain, predicted to be deleterious and likely to cause loss of PHF21A function. We also found a new C-terminal domain composed of an intrinsically disordered region. This domain is truncated in six patients and thus likely to play an important role in the function of PHF21A, suggesting that haploinsufficiency is the likely underlying mechanism in the phenotype of seven patients. Our results extend the phenotypic spectrum of PHF21A mutations by adding autism spectrum disorder, epilepsy, hypotonia, and neurobehavioral problems. Furthermore, PHF21A is highly expressed in the human fetal brain, which is consistent with the neurodevelopmental phenotype. Conclusion Deleterious nonsense, frameshift, and missense mutations disrupting the AT Hook domain and/or an intrinsically disordered region in PHF21A were found to be associated with autism spectrum disorder, epilepsy, hypotonia, neurobehavioral problems, tapering fingers, clinodactyly, and syndactyly, in addition to intellectual disability and craniofacial anomalies. This suggests that PHF21A is involved in autism spectrum disorder and intellectual disability, and its haploinsufficiency causes a diverse neurological phenotype.
- Published
- 2019
24. OR15-4 Long-Term Follow-Up of a Female with a Mutation in the Estrogen Receptor Alpha (ESR1) Gene
- Author
-
Janet E. Hall, Lynn P. Chorich, Soumia Brakta, Kenneth Korach, Lawrence C. Layman, Laurel Coons, and Hyung Goo Kim
- Subjects
Text mining ,business.industry ,Long term follow up ,Genetics and Development of the Endocrine System in Health and Disease ,Endocrinology, Diabetes and Metabolism ,Mutation (genetic algorithm) ,Cancer research ,Biology ,business ,Estrogen receptor alpha ,Gene ,Genetics and Development (including Gene Regulation) - Abstract
We previously reported the first female with a mutation in the ESR1 gene encoding estrogen receptor-α (ER-α), which dramatically reduced estrogen signaling in vitro. She presented at 179/12 years with absent breast development, markedly elevated serum estradiol and estrone levels, modestly high gonadotropins, and multicystic ovaries by ultrasound. The natural history of estrogen insensitivity in women is unknown. The purpose of this report is to present: 1) the neuroendocrine phenotype; 2) studies to identify a ligand to stimulate the mutant receptor; and 3) the effect of targeted treatment. We now provide long-term clinical follow up and a detailed hormonal analysis of the patient at age 234/12 years. On no treatment, serum FSH and LH were drawn every 10 minutes for 8 hours, and estradiol and estrone levels were measured every hour during the study. Various compounds that are potential for treatment were used in transient transfection assays to determine if estrogen signaling could be induced. A personalized medicine approach was tailored to our patient, in which she received increasing doses of diethylstilbestrol (DES) with the primary endpoints being increased vaginal discharge and breast development. Follow up exams, labs, pelvic ultrasounds, DXA, and bone age X-rays were completed at various intervals up to age 234/12 years. The Bioethics Board approved this treatment. During the frequent sampling study, E2 was 604 pg/mL, E1 was 596 pg/mL, FSH was 10.0+/-0.1 IU/L (mean+/-SE), and LH was 6.7+/-0.2 IU/L. There were 3 pulses in 8 hr with an LH pulse amplitude of 2.3+/-0.5 IU/L. Although DES was shown to successfully transactivate the mutant ESR1in vitro, both in HEK293 cells and in her lymphoblastoid cells, the patient had no clinical response. After 2.5 years of treatment with increasing doses of DES, there was no vaginal discharge and breasts remained at Tanner 1. At 234/12 years, LH, FSH and E2 were unchanged. A vaginal maturation index showed a predominance of parabasal cells consistent with a lack of estrogen effect. Ultrasound showed bilateral ovarian cysts. At chronologic age 234/12 years, her bone age was 17 years and the epiphysis remained open in the ulnar bone. DXA revealed a Z-score of -2.9 at the femoral neck and -4.5 at the lumbar spine. Our findings in this severely estrogen insensitive female indicate elevated gonadotropins with normal amplitude and low frequency LH pulse dynamics in the presence of markedly increased estrogens. Treatment with DES, which stimulated estrogen signaling in vitro, did not induce secondary sexual characteristics in our patient. She remains hypoestrogenic despite the presence of ovarian cysts with a hypoestrogenic vaginal smear, absent breast development, and low bone mineral mass. Future studies will be directed towards further identification of other potential signaling mechanisms and activators of this mutant receptor.
- Published
- 2019
25. SAT-404 Effect of Nsmf Knockout upon Hypothalamic and Pituitary Gene Expression in the Nsmf KO Mouse
- Author
-
Lawrence C. Layman, Erica Louden, Eunkyung Ko, and Lynn P. Chorich
- Subjects
endocrine system ,Neuroendocrinology and Pituitary ,Text mining ,business.industry ,Endocrinology, Diabetes and Metabolism ,Gene expression ,Neuroendocrinology ,Biology ,business ,Cell biology - Abstract
Genetic approaches in humans with gonadotropin releasing hormone (GnRH) deficiency causing hypogonadotropic hypogonadism (HH) have been important to understand normal reproduction. NSMF (NMDA receptor synaptonuclear signaling & neuronal migration factor), formerly known as NELF (nasal embryonic LHRH factor), gene mutations have been identified in humans with normosmic HH (nHH) and Kallmann syndrome (KS). The Nsmf knockout mouse (KO) females have normal puberty, but also manifest subfertility of uncertain etiology. Although many causative nHH/KS genes are known, a critical barrier is our lack of understanding the mechanisms by which gene mutations impair reproduction. Previously we have shown that Nsmf knockdown impairs GnRH neuronal cell migration and is associated with reduced JAK/STAT expression in migratory GnRH neurons. We also showed that Nsmf mRNA expression in LβT2 cells increased 3-fold after GnRH agonist administration, suggesting a pituitary role. Furthermore, Nsmf is expressed in the ovary, but the role there is unknown. Our central hypothesis is that subfertility in the female Nsmf KO mouse is due to a combination of hypothalamic, pituitary and/or gonadal defects. We first wanted to determine baseline expression of key reproductive genes in the hypothalamus (Gnrh1 and Kiss1), pituitary (Cga, Fshb, Lhb, and Gnrhr), and ovary (Fshr, Lhr, Prkaca, Prkar1a, and Gdf9) in Nsmf KO females vs wild type (WT) controls. RT-qPCR was performed on RNA extracted from individual hypothalami, pituitaries, and ovaries from WT or KO 8 week old adult female mice. Comparison of WT vs. KO was performed using ΔΔ CT method with Gapdh as the internal control. Our preliminary findings suggest a modest decrease in Gnrh1 expression and a doubling of Kiss1 expression in the hypothalamus. However, all pituitary genes studied (Cga, Fshb, Lhb, and Gnrhr) had markedly reduced expression in the KO vs WT. Ovarian gene expression was unaltered with the exception of a moderate decrease in Gdf9. These preliminary findings suggest a hypothalamic and/or pituitary etiology for subfertility in the Nsmf KO mouse. Studies with Gnrh agonist are ongoing to differentiate hypothalamic from pituitary dysfunction. Future experiments will include treatment with PMSG/hCG to evaluate the ovarian role in the subfertility. Our results reveal that NSMF may play a role in the central regulation of reproductive function via effects at the hypothalamus and/or pituitary levels. Elucidation of the reproductive phenotype of the Nsmf KO will increase our understanding of the pathogenesis of the Nsmf KO mouse as well as normal reproduction.
- Published
- 2019
26. The genetics of Mullerian aplasia
- Author
-
Lawrence C. Layman
- Subjects
Genetics ,medicine.medical_specialty ,Microarray ,Positional cloning ,Endocrinology, Diabetes and Metabolism ,Molecular genetics ,medicine ,Mayer-Rokitansky-Kuster-Hauser Syndrome ,Copy-number variation ,Biology ,HNF1B ,Gene ,Exome sequencing - Abstract
Mayer-Rokitansky-Kuster-Hauser (MRKH) syndrome consists of Mullerian aplasia with or without other anomalies, most commonly renal and skeletal. The genetic etiology of MRKH syndrome is unknown for most patients, but supportive evidence exists for heterozygous mutations in WNT4, LHX1, and HNF1B. Chromosomal microarray analyses have demonstrated chromosomal regions with copy number variants in multiple patients - deletions in17q12 and 16p11.2, and either deletions or duplications in 22q11.2. Genomic analyses of expression and methylation have also suggested potential molecular pathways. Positional cloning in MRKH patients with chromosomal rearrangements and exome sequencing are likely to result in new genes. Although some single gene defects and copy number variant regions have been identified, the molecular basis for the vast majority of MRKH remains unknown.
- Published
- 2019
27. Concomitant partial exon skipping by a unique missense mutation of RPS6KA3 causes Coffin–Lowry syndrome
- Author
-
Julie R. Jones, Lawrence C. Layman, Priya Anand, Jonathan D J Labonne, Hyung Goo Kim, Wolfgang Wenzel, Daniela Iacoboni, and Min Ji Chung
- Subjects
Male ,0301 basic medicine ,Technology ,RNA Splicing ,Mutant ,Mutation, Missense ,Biology ,Ribosomal Protein S6 Kinases, 90-kDa ,03 medical and health sciences ,Exon ,Coffin-Lowry Syndrome ,Genetics ,medicine ,Humans ,Missense mutation ,Coffin–Lowry syndrome ,Exons ,General Medicine ,medicine.disease ,Molecular biology ,Exon skipping ,RPS6KA3 ,030104 developmental biology ,Mutation (genetic algorithm) ,RNA splicing ,Female ,RNA Splice Sites ,ddc:600 - Abstract
Coffin-Lowry syndrome (CLS) is an X-linked semi-dominant disorder characterized by diverse phenotypes including intellectual disability, facial and digital anomalies. Loss-of-function mutations in the Ribosomal Protein S6 Kinase Polypeptide 3 (RPS6KA3) gene have been shown to be responsible for CLS. Among the large number of mutations, however, no exonic mutation causing exon skipping has been described. Here, we report a male patient with CLS having a novel mutation at the 3' end of an exon at a splice donor junction. Interestingly, this nucleotide change causes both a novel missense mutation and partial exon skipping leading to a truncated transcript. These two transcripts were identified by cDNA sequencing of RT-PCR products. In the carrier mother, we found only wildtype transcripts suggesting skewed X-inactivation. Methylation studies confirmed X-inactivation was skewed moderately, but not completely, which is consistent with her mild phenotype. Western blot showed that the mutant RSK2 protein in the patient is expressed at similar levels relative to his mother. Protein modeling demonstrated that the missense mutation is damaging and may alter binding to ATP molecules. This is the first report of exon skipping from an exonic mutation of RPS6KA3, demonstrating that a missense mutation and concomitant disruption of normal splicing contribute to the manifestation of CLS.
- Published
- 2016
28. Academic pursuits in board-certified reproductive endocrinologists
- Author
-
Scott J. Morin, Lawrence C. Layman, Thomas M Price, Yolanda R. Smith, Jamie L Morris, Bradley S. Hurst, Eve C. Feinberg, and Margareta D. Pisarska
- Subjects
medicine.medical_specialty ,Biomedical Research ,Certification ,Reproductive endocrinology and infertility ,Efficiency ,03 medical and health sciences ,Endocrinology ,0302 clinical medicine ,Specialty Boards ,Surveys and Questionnaires ,medicine ,Humans ,030212 general & internal medicine ,Publishing ,Academic Success ,030219 obstetrics & reproductive medicine ,Publications ,Outcome measures ,Obstetrics and Gynecology ,United States ,Important research ,Endocrinologists ,Reproductive Medicine ,Family medicine ,Psychology ,Construct (philosophy) - Abstract
Objective To determine research interests of reproductive endocrinology and infertility (REI) physicians and assess their academic productivity. Design A questionnaire composed by the Society for REI (SREI) board members was e-mailed to members. PubMed was queried to quantify peer-reviewed publications. Setting An internal SREI questionnaire to members and online publication search. Patient(s) Not applicable. Intervention(s) Questions involving research being performed, funding, relevance to fellow thesis, and important areas of future research. Publications were ascertained in the past 3 years, past 10 years, and total publications for SREI members. Main Outcome Measure(s) Question responses and number of peer-reviewed publications. Result(s) Most respondents currently conduct research, which was predominantly clinical. One-third have current research funding and two-thirds were ever funded. One-third had a National Institutes of Health grant and about half were principal investigators. Two-thirds had a basic science fellow thesis and 44% of respondents perform research related to their fellowship thesis. Important research areas included infertility outcomes, implantation, preimplantation genetic testing, and genetics. In the past 3 years, SREI members published 3,408 peer-reviewed articles (mean ± standard deviation [SD], 4.4 ± 9.0). In the past 10 years, SREI members had 10,162 peer-reviewed publications (mean±SD, 13.0 ± 24.3). When all publications were considered, SREI members published 24,088 peer-reviewed articles (mean±SD, 30.9 ± 53.0). Conclusion(s) The REI fellows have learned to construct scientific articles, which will help them to better interpret the literature in the care of patients. The SREI members continue to pursue scientific investigation, commonly related to their fellowship thesis. Respondents support SREI funding research; the success of which should be judged by publications. Overall, SREI members have demonstrated significant academic productivity and published about 1,000 articles/year for the past 10 years, affirming the importance of research training.
- Published
- 2020
29. WDR11-mediated Hedgehog signalling defects underlie a new ciliopathy related to Kallmann syndrome
- Author
-
Francesc Miralles, Juan Pedro Martinez-Barbera, Nina Brandstack, Jiyoung Lee, Soo-Hyun Kim, Hyun Taek Kim, Hyung-Goo Kim, Yeonjoo Kim, Nigel A. Brown, Masatake Araki, Lawrence C. Layman, Johanna Känsäkoski, Paris Ataliotis, Kimi Araki, Taneli Raivio, Cheol-Hee Kim, Daniel P. S. Osborn, Timothy J. Mohun, Raivio Group, Clinicum, Department of Diagnostics and Therapeutics, Medicum, Department of Physiology, HUS Children and Adolescents, and HUS Medical Imaging Center
- Subjects
0301 basic medicine ,Purmorphamine ,WDR11 ,Kallmann syndrome ,Biopsy ,Gene Expression ,PROTEIN ,MOUSE ,Biochemistry ,DISEASE ,PATHWAY ,Gene Knockout Techniques ,Mice ,0302 clinical medicine ,Molecular Biology of Disease ,Promoter Regions, Genetic ,Zebrafish ,Mice, Knockout ,ELECTRON-MICROSCOPY ,Genetics ,0303 health sciences ,Cilium ,Articles ,Magnetic Resonance Imaging ,Hedgehog signaling pathway ,Cell biology ,Patched-1 Receptor ,Protein Transport ,Phenotype ,Organ Specificity ,hedgehog signal pathway ,HORMONE NEURONS ,Haploinsufficiency ,Signal Transduction ,Protein Binding ,Genotype ,PRIMARY CILIA ,kallmann syndrome ,Biology ,Article ,03 medical and health sciences ,CONGENITAL HYPOGONADOTROPIC HYPOGONADISM ,Hypogonadotropic hypogonadism ,Ciliogenesis ,GLI3 ,medicine ,Animals ,Humans ,Hedgehog Proteins ,MUTANT MICE ,Molecular Biology ,Hedgehog ,Genetic Association Studies ,030304 developmental biology ,Gene Expression Profiling ,Membrane Proteins ,hypogonadotropic hypogonadism ,medicine.disease ,GENE ,Ciliopathies ,Ciliopathy ,ciliopathy ,030104 developmental biology ,Mutation ,3111 Biomedicine ,Cell Adhesion, Polarity & Cytoskeleton ,Transcriptome ,030217 neurology & neurosurgery - Abstract
WDR11 has been implicated in congenital hypogonadotropic hypogonadism (CHH) and Kallmann syndrome (KS), human developmental genetic disorders defined by delayed puberty and infertility. However, WDR11’s role in development is poorly understood. Here we report that WDR11 modulates the Hedgehog (Hh) signalling pathway and is essential for ciliogenesis. Disruption of WDR11 expression in mouse and zebrafish results in phenotypic characteristics associated with defective Hh signalling, accompanied by dysgenesis of ciliated tissues.Wdr11null mice also exhibit early onset obesity. We found that WDR11 shuttles from the cilium to the nucleus in response to Hh signalling. WDR11 was also observed to regulate the proteolytic processing of GLI3 and cooperate with EMX1 transcription factor to induce the expression of downstream Hh pathway genes and gonadotrophin releasing hormone production. The CHH/KS-associated human mutations result in loss-of-function of WDR11. Treatment with the Hh agonist purmorphamine partially rescued the WDR11-haploinsufficiency phenotypes. Our study reveals a novel class of ciliopathy caused by WDR11 mutations and suggests that CHH/KS may be a part of the human ciliopathy spectrum.
- Published
- 2017
30. Copy number variation and regions of homozygosity analysis in patients with MÜLLERIAN aplasia
- Author
-
Özgül M. Alper, Yiping Shen, Hyung Goo Kim, Lawrence C. Layman, Meric Bilekdemir, Guven Luleci, Megan E. Sullivan, Lynn P. Chorich, Elanur Yilmaz, Durkadin Demir Eksi, Munire Erman, ALKÜ, and 0-belirlenecek
- Subjects
0301 basic medicine ,Candidate gene ,Turkish population ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,lcsh:QH426-470 ,Regions of homozygosity ,CNV ,Biology ,Copy number variant ,Biochemistry ,Müllerian aplasia ,03 medical and health sciences ,0302 clinical medicine ,Mayer-Rokitansky-Küster-Hauser syndrome ,Mullerian aplasia ,TP63 ,Genetics ,medicine ,Copy-number variation ,Allele ,Molecular Biology ,Genetics (clinical) ,030219 obstetrics & reproductive medicine ,Research ,Biochemistry (medical) ,ROH ,Cytogenetics ,HNF1B ,Human genetics ,lcsh:Genetics ,030104 developmental biology ,Mayer-Rokitansky-Kuster-Hauser syndrome ,Congenital absence of the uterus and vagina ,Molecular Medicine ,MRKH - Abstract
Yilmaz, Elanur/0000-0001-7045-5068; Alper, Ozgul/0000-0003-1536-2111 WOS: 000424131300001 PubMed: 29434669 Background: Little is known about the genetic contribution to Mullerian aplasia, better known to patients as Mayer-Rokitansky-Kuster-Hauser (MRKH) syndrome. Mutations in two genes (WNT4 and HNF1B) account for a small number of patients, but heterozygous copy number variants (CNVs) have been described. However, the significance of these CNVs in the pathogenesis of MRKH is unknown, but suggests possible autosomal dominant inheritance. We are not aware of CNV studies in consanguineous patients, which could pinpoint genes important in autosomal recessive MRKH. We therefore utilized SNP/CGH microarrays to identify CNVs and define regions of homozygosity (ROH) in Anatolian Turkish MRKH patients. Result(s): Five different CNVs were detected in 4/19 patients (21%), one of which is a previously reported 16p11.2 deletion containing 32 genes, while four involved smaller regions each containing only one gene. Fourteen of 19 (74%) of patients had parents that were third degree relatives or closer. There were 42 regions of homozygosity shared by at least two MRKH patients which was spread throughout most chromosomes. Of interest, eight candidate genes suggested by human or animal studies (RBM8A, CMTM7, CCR4, TRIM71, CNOT10, TP63, EMX2, and CFTR) reside within these ROH. Conclusion(s): CNVs were found in about 20% of Turkish MRKH patients, and as in other studies, proof of causation is lacking. The 16p11.2 deletion seen in mixed populations is also identified in Turkish MRKH patients. Turkish MRKH patients have a higher likelihood of being consanguineous than the general Anatolian Turkish population. Although identified single gene mutations and heterozygous CNVs suggest autosomal dominant inheritance for MRKH in much of the western world, regions of homozygosity, which could contain shared mutant alleles, make it more likely that autosomal recessively inherited causes will be manifested in Turkish women with MRKH. NIHUnited States Department of Health & Human ServicesNational Institutes of Health (NIH) - USA [HD33004]; Department of Ob/Gyn at Augusta University; Research Funds Office of Akdeniz University, Antalya, Turkey [2012. 03.0122.001] LCL was funded by NIH HD33004 and the Department of Ob/Gyn at Augusta University and OMA was funded by the Research Funds Office of Akdeniz University, Antalya, Turkey (Grant #2012. 03.0122.001).
- Published
- 2017
31. Abstracts of papers presented at the 27th Genetics Society's Mammalian Genetics and Development Workshop held at the UCL Great Ormond Street Institute of Child Health, University College London on 18th November 2016
- Author
-
Paris Ataliotis, H-G Kim, Masatake Araki, Juan Pedro Martinez-Barbera, Lawrence C. Layman, Kimi Araki, Yr. Kim, S-H Kim, Nigel A. Brown, Daniel P. S. Osborn, Timothy J. Mohun, and J-Y Lee
- Subjects
medicine.medical_specialty ,business.industry ,Kallmann syndrome ,General Medicine ,medicine.disease ,Hedgehog signaling pathway ,Ciliopathy ,Endocrinology ,Internal medicine ,Genetics ,medicine ,Congenital Hypogonadotropic Hypogonadism ,business ,Abstract - Published
- 2017
32. Genetic basis of eugonadal and hypogonadal female reproductive disorders
- Author
-
Lawrence C. Layman, Ricardo Azziz, Daria Lizneva, Tatiana Trofimova, Yen Hao Chen, Larisa Suturina, and Walidah Walker
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Kallmann syndrome ,Endometriosis ,Ovarian hyperstimulation syndrome ,Bioinformatics ,03 medical and health sciences ,Ovarian Hyperstimulation Syndrome ,0302 clinical medicine ,Hypergonadotropic hypogonadism ,Turner syndrome ,medicine ,Humans ,Mullerian Ducts ,Gynecology ,030219 obstetrics & reproductive medicine ,Leiomyoma ,business.industry ,Hypogonadism ,Obstetrics and Gynecology ,General Medicine ,medicine.disease ,Premature ovarian failure ,030104 developmental biology ,Phenotype ,Etiology ,Female ,business ,Hyperandrogenism ,Hypogonadotrophic hypogonadism ,Genome-Wide Association Study ,Polycystic Ovary Syndrome - Abstract
This review discusses the current state of our understanding regarding the genetic basis of the most important reproductive disorders in women. For clarity, these disorders have been divided into eugonadal and hypogonadal types. Hypogonadal disorders have been further subdivided according to serum gonadotropin levels. Our review focuses on historical and recent data regarding the genetics of the hypothalamic–pituitary–gonadal axis dysfunction, as well as the development and etiology of eugonadal disorders including leiomyomata, endometriosis, spontaneous ovarian hyperstimulation syndrome, polycystic ovarian syndrome, mullerian aplasia, and steroid hormone resistance syndromes. We discuss the known genes most commonly involved in hypergonadotropic hypogonadism (Turner syndrome and premature ovarian failure) and hypogonadotrophic hypogonadism (Kallmann syndrome and normosmic types). In addition, we summarize the current clinical testing approaches and their utility in practical application.
- Published
- 2017
33. Genetic analysis of Mayer-Rokitansky-Kuster-Hauser syndrome in a large cohort of families
- Author
-
Yiping Shen, Lawrence C. Layman, Özgül M. Alper, Hyung Goo Kim, Megan E. Sullivan, Lynn P. Chorich, John A. Phillips, Durkadin Demir Eksi, Lacey S. Williams, Amy C. Lossie, and Munire Erman
- Subjects
0301 basic medicine ,Proband ,Adult ,Genetic Markers ,Candidate gene ,Internationality ,46, XX Disorders of Sex Development ,LIM-Homeodomain Proteins ,Gene mutation ,Biology ,Genetic analysis ,Polymorphism, Single Nucleotide ,Article ,Congenital Abnormalities ,Cohort Studies ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Risk Factors ,Wnt4 Protein ,Prevalence ,Humans ,Family ,Genetic Predisposition to Disease ,Copy-number variation ,Mullerian Ducts ,Hepatocyte Nuclear Factor 1-beta ,Genetics ,030219 obstetrics & reproductive medicine ,Obstetrics and Gynecology ,HNF1B ,030104 developmental biology ,Reproductive Medicine ,Etiology ,Sample collection ,Transcription Factors - Abstract
Objective To study the genetic cause of Mayer-Rokitansky-Kuster-Hauser syndrome (MRKH). Although a few candidate genes and genomic domains for have been reported for MRKH, the genetic underpinnings remain largely unknown. Some of the top candidate genes are WNT4 , HNF1B , and LHX1 . The goals of this study were to: 1) determine the prevalence of WNT4 , HNF1B , and LHX1 point mutations, as well as new copy number variants (CNVs) in people with MRKH; and 2) identify and characterize MRKH cohorts. Design Laboratory- and community-based study. Setting Academic medical centers. Patient(s) A total of 147 MRKH probands and available family members. Interventions(s) DNA sequencing of WNT4 , HNF1B , and LHX1 in 100 MRKH patients, chromosomal microarray analysis in 31 North American MRKH patients, and characterization and sample collection of 147 North American and Turkish MRKH probands and their families. Main Outcome Measure(s) DNA sequence variants and CNVs; pedigree structural analysis. Result(s) We report finding CNVs in 6/31 people (∼19%) with MRKH, but no point mutations or small indels in WNT4 , HNF1B , or LHX1 in 100 MRKH patients. Our MRKH families included 43 quads, 26 trios, and 30 duos. Of our MRKH probands, 87/147 (59%) had MRKH type 1 and 60/147 (41%) had type 2 with additional anomalies. Conclusion(s) Although the prevalence of WNT4 , HNF1B , and LHX1 point mutations is low in people with MRKH, the prevalence of CNVs was ∼19%. Further analysis of our large familial cohort of patients will facilitate gene discovery to better understand the complex etiology of MRKH.
- Published
- 2017
34. Spectrum of genes involved in a unique case of Potocki Schaffer syndrome with a large chromosome 11 deletion
- Author
-
Bernd F. M. Romeike, Hiromi Koso Nishimoto, Lawrence C. Layman, Cynthia C. Morton, Yiping Shen, and Hyung Goo Kim
- Subjects
Genetics ,Letter ,Extramural ,Chromosome ,General Medicine ,Neuropathology ,Biology ,Pathology and Forensic Medicine ,Transcriptome ,Neurology ,Potocki Schaffer syndrome ,Neurology (clinical) ,Gene - Abstract
Letter to the Editor.
- Published
- 2014
35. Targeted next generation sequencing approach identifies eighteen new candidate genes in normosmic hypogonadotropic hypogonadism and Kallmann Syndrome
- Author
-
Samuel D. Quaynor, Lynn P. Chorich, Maggie E. Bosley, Lawrence C. Layman, Kelsey R. Porter, Megan E. Sullivan, Richard S. Cameron, Jeong Hyeon Choi, Hyung Goo Kim, Soo-Hyun Kim, and Christina G. Duckworth
- Subjects
Male ,0301 basic medicine ,Nonsynonymous substitution ,Candidate gene ,Kallmann syndrome ,Biology ,Biochemistry ,DNA sequencing ,Frameshift mutation ,03 medical and health sciences ,symbols.namesake ,0302 clinical medicine ,Endocrinology ,Hypogonadotropic hypogonadism ,medicine ,Humans ,Missense mutation ,Molecular Biology ,Genetic Association Studies ,Genetics ,Sanger sequencing ,Hypogonadism ,High-Throughput Nucleotide Sequencing ,Kallmann Syndrome ,medicine.disease ,Pedigree ,Phenotype ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation ,symbols ,Female - Abstract
The genetic basis is unknown for ∼60% of normosmic hypogonadotropic hypogonadism (nHH)/Kallmann syndrome (KS). DNAs from (17 male and 31 female) nHH/KS patients were analyzed by targeted next generation sequencing (NGS) of 261 genes involved in hypothalamic, pituitary, and/or olfactory pathways, or suggested by chromosome rearrangements. Selected variants were subjected to Sanger DNA sequencing, the gold standard. The frequency of Sanger-confirmed variants was determined using the ExAC database. Variants were classified as likely pathogenic (frameshift, nonsense, and splice site) or predicted pathogenic (nonsynonymous missense). Two novel FGFR1 mutations were identified, as were 18 new candidate genes including: AMN1, CCKBR, CRY1, CXCR4, FGF13, GAP43, GLI3, JAG1, NOS1, MASTL, NOTCH1, NRP2, PALM2, PDE3A, PLEKHA5, RD3, and TRAPPC9, and TSPAN11. Digenic and trigenic variants were found in 8/48 (16.7%) and 1/48 (2.1%) patients, respectively. NGS with confirmation by Sanger sequencing resulted in the identification of new causative FGFR1 gene mutations and suggested 18 new candidate genes in nHH/KS.
- Published
- 2016
36. An ovarian component involved in subfertility of the Nsmf KO mouse
- Author
-
Erica Louden, Lawrence C. Layman, and Lynn P. Chorich
- Subjects
Reproductive Medicine ,Component (UML) ,Obstetrics and Gynecology ,Biology ,Cell biology - Published
- 2019
37. Delayed Puberty and Estrogen Resistance in a Woman with Estrogen Receptor α Variant
- Author
-
Earl W. Stradtman, Hyung Goo Kim, Yiping Shen, Samuel D. Quaynor, Lawrence C. Layman, Lynn P. Chorich, and Derek A. Schreihofer
- Subjects
medicine.medical_specialty ,Breast development ,medicine.drug_class ,business.industry ,Estrogen receptor ,General Medicine ,Androgen ,Polycystic ovarian disease ,body regions ,Estrogen-related receptor alpha ,Endocrinology ,Estrogen ,Internal medicine ,medicine ,business ,Estrogen receptor alpha ,Estrogen receptor beta - Abstract
Although androgen resistance has been characterized in men with a normal chromosome complement and mutations in the androgen-receptor gene, a mutation in the gene encoding estrogen receptor α (ESR1) was previously described only in one man and not, to our knowledge, in a woman. We now describe an 18-year-old woman without breast development and with markedly elevated serum levels of estrogens and bilateral multicystic ovaries. She was found to have a homozygous loss-of-function ESR1 mutation in a completely conserved residue that interferes with estrogen signaling. Her clinical presentation was similar to that in the mouse orthologue knockout. This case shows that disruption of ESR1 causes profound estrogen resistance in women. (Funded by the National Institutes of Health.)
- Published
- 2013
38. The genetic basis of female reproductive disorders: Etiology and clinical testing
- Author
-
Lawrence C. Layman
- Subjects
Male ,medicine.medical_specialty ,46, XX Disorders of Sex Development ,Kallmann syndrome ,Disorders of Sex Development ,Endometriosis ,Ovarian hyperstimulation syndrome ,Biology ,Gene mutation ,Bioinformatics ,Biochemistry ,Article ,Congenital Abnormalities ,Fragile X Mental Retardation Protein ,Ovarian Hyperstimulation Syndrome ,Endocrinology ,Hypergonadotropic hypogonadism ,Pituitary Hormones, Anterior ,Wnt4 Protein ,Hypogonadotropic hypogonadism ,Internal medicine ,medicine ,Humans ,Receptor, Fibroblast Growth Factor, Type 1 ,Mullerian Ducts ,Molecular Biology ,Genetic testing ,Homeodomain Proteins ,Leiomyoma ,medicine.diagnostic_test ,Hypogonadism ,DNA Helicases ,medicine.disease ,FMR1 ,Polycystic ovary ,DNA-Binding Proteins ,Female ,Genital Diseases, Female ,Polycystic Ovary Syndrome - Abstract
With the advent of improved molecular biology techniques, the genetic basis of an increasing number of reproductive disorders has been elucidated. Mutations in at least 20 genes cause hypogonadotropic hypogonadism including Kallmann syndrome in about 35–40% of patients. The two most commonly involved genes are FGFR1 and CHD7. When combined pituitary hormone deficiency includes hypogonadotropic hypogonadism as a feature, PROP1 mutations are the most common of the six genes involved. For hypergonadotropic hypogonadism, mutations in 14 genes cause gonadal failure in 15% of affected females, most commonly in FMR1. In eugonadal disorders, activating FSHR mutations have been identified for spontaneous ovarian hyperstimulation syndrome; and WNT4 mutations have been described in mullerian aplasia. For other eugonadal disorders, such as endometriosis, polycystic ovary syndrome, and leiomyomata, specific germline gene mutations have not been identified, but some chromosomal regions are associated with the corresponding phenotype. Practical genetic testing is possible to perform in both hypogonadotropic and hypergonadotropic hypogonadism and spontaneous ovarian hyperstimulation syndrome. However, clinical testing for endometriosis, polycystic ovary syndrome, and leiomyomata is not currently practical for the clinician.
- Published
- 2013
39. The Molecular Basis of Impaired Follicle-Stimulating Hormone Action: Evidence From Human Mutations and Mouse Models
- Author
-
Eric T. Siegel, Hyung Goo Kim, Lawrence C. Layman, and Hiromi Koso Nishimoto
- Subjects
Male ,endocrine system ,medicine.medical_specialty ,Reviews ,Biology ,FSHB ,Mice ,Follicle-stimulating hormone ,Internal medicine ,Follicular phase ,medicine ,Animals ,Humans ,Receptor ,Azoospermia ,Gonadal Disorders ,Obstetrics and Gynecology ,Sertoli cell ,medicine.disease ,Disease Models, Animal ,Endocrinology ,medicine.anatomical_structure ,Mutation ,Knockout mouse ,Receptors, FSH ,Female ,Follicle Stimulating Hormone ,Follicle-stimulating hormone receptor ,hormones, hormone substitutes, and hormone antagonists - Abstract
The pituitary gonadotropin follicle-stimulating hormone (FSH) interacts with its membrane-bound receptor to produce biologic effects. Traditional functions of FSH include follicular development and estradiol production in females, and the regulation of Sertoli cell action and spermatogenesis in males. Knockout mice for both the ligand (Fshb) and the receptor (Fshr) serve as models for FSH deficiency, while Fshb and Fshr transgenic mice manifest FSH excess. In addition, inactivating mutations of both human orthologs (FSHB and FSHR) have been characterized in a small number of patients, with phenotypic effects of the ligand disruption being more profound than those of its receptor. Activating human FSHR mutants have also been described in both sexes, leading to a phenotype of normal testis function (male) or spontaneous ovarian hyperstimulation syndrome (females). As determined from human and mouse models, FSH is essential for normal puberty and fertility in females, particularly for ovarian follicular development beyond the antral stage. In males, FSH is necessary for normal spermatogenesis, but there are differences in human and mouse models. The FSHB mutations in humans result in azoospermia; while FSHR mutations in humans and knockouts of both the ligand and the receptor in mice affect testicular function but do not result in absolute infertility. Available evidence also indicates that FSH may also be necessary for normal androgen synthesis in males and females.
- Published
- 2013
40. Liquid Chromatography–Tandem Mass Spectrometry Analysis of Human Adrenal Vein 19-Carbon Steroids Before and After ACTH Stimulation
- Author
-
Michael R. Kennedy, Seijiro Honma, Lawrence C. Layman, Ryo Morimoto, Hironobu Sasano, William E. Rainey, Juilee Rege, Fumitoshi Satoh, and Yasuhiro Nakamura
- Subjects
medicine.medical_specialty ,Estrone ,medicine.drug_class ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Dehydroepiandrosterone ,Context (language use) ,Adrenocorticotropic hormone ,Biochemistry ,Veins ,chemistry.chemical_compound ,Endocrinology ,Dehydroepiandrosterone sulfate ,Adrenocorticotropic Hormone ,Tandem Mass Spectrometry ,Internal medicine ,medicine ,Endocrine Research ,Humans ,Testosterone ,Cells, Cultured ,Retrospective Studies ,Androstenols ,Estradiol ,Dehydroepiandrosterone Sulfate ,Adrenal cortex ,Chemistry ,Biochemistry (medical) ,Androstenedione ,Middle Aged ,Androgen ,Hormones ,medicine.anatomical_structure ,Receptors, Androgen ,Adrenal Cortex ,Androgens ,Metabolome ,Female ,hormones, hormone substitutes, and hormone antagonists ,Chromatography, Liquid - Abstract
A broad analysis of adrenal gland-derived 19-carbon (C19) steroids has not been reported. This is the first study that uses liquid chromatography-tandem mass spectrometry to quantify 9 C19 steroids (androgens and their precursors), estrone, and estradiol in the adrenal vein (AV) of women, before and after ACTH stimulation.The objective of this study was to define the adrenal androgen metabolome in women before and after ACTH infusion.This was a retrospective study.Seven women, aged 50.4 ± 5.4 years, with suspected diagnosis of an adrenal aldosterone-producing adenoma were included in the study.AV and iliac serum samples were collected before and after administration of ACTH (15 minutes). AV samples were analyzed using for concentrations of 9 unconjugated C19 steroids, estrone, and estradiol. Dehydroepiandrosterone sulfate (DHEA-S) was quantified by radioimmunoassay.AV levels of DHEA-S were the highest among the steroids measured. The most abundant unconjugated C19 steroids in AV were 11β-hydroxyandrostenedione (11OHA), dehydroepiandrosterone (DHEA), and androstenedione (A4). ACTH significantly increased the adrenal output of 9 of the 12 steroids that were measured. ACTH increased the mean AV concentration of DHEA-S by 5-fold, DHEA by 21-fold, A4 by 7-fold, and 11OHA by 5-fold. 11β-Hydroxytestosterone and testosterone were found to be potent androgen receptor agonists when tested with an androgen-responsive cell reporter model.The current study indicates that the adrenal gland secretes primarily 3 weak androgens, namely DHEA, 11OHA, and A4. Active androgens, including testosterone and 11β-hydroxytestosterone, are also produced but to a lesser degree.
- Published
- 2013
41. Contribution of copy number variants involving nonsense-mediated mRNA decay pathway genes to neuro-developmental disorders
- Author
-
Yiping Shen, Jozef Gecz, Lam Son Nguyen, Lawrence C. Layman, James F. Gusella, Lisa G. Shaffer, Yves Lacassie, Hyung Goo Kim, and Jill A. Rosenfeld
- Subjects
Male ,Adolescent ,DNA Copy Number Variations ,Developmental Disabilities ,Nonsense-mediated decay ,Gene Dosage ,RNA-binding protein ,Biology ,medicine.disease_cause ,Gene dosage ,DEAD-box RNA Helicases ,Transcriptome ,Intellectual Disability ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,RNA, Messenger ,Copy-number variation ,Child ,Telomerase ,Molecular Biology ,Gene ,Genetics (clinical) ,Neurons ,Mutation ,Sequence Analysis, RNA ,Computational Biology ,RNA-Binding Proteins ,Heterozygote advantage ,General Medicine ,Nonsense Mediated mRNA Decay ,Ribonucleoproteins ,Codon, Nonsense ,Case-Control Studies ,Eukaryotic Initiation Factor-4A ,Female ,Gene Deletion ,Transcription Factors - Abstract
The nonsense-mediated mRNA decay (NMD) pathway functions not only to degrade transcripts containing premature termination codons (PTC), but also to regulate the transcriptome. UPF3B and RBM8A, important components of NMD, have been implicated in various forms of intellectual disability (ID) and Thrombocytopenia with Absent Radius (TAR) syndrome, which is also associated with ID. To gauge the contribution of other NMD factors to ID, we performed a comprehensive search for copy number variants (CNVs) of 18 NMD genes among individuals with ID and/or congenital anomalies. We identified 11 cases with heterozygous deletions of the genomic region encompassing UPF2, which encodes for a direct interacting protein of UPF3B. Using RNA-Seq, we showed that the genome-wide consequence of reduced expression of UPF2 is similar to that seen in patients with UPF3B mutations. Out of the 1009 genes found deregulated in patients with UPF2 deletions by at least 2-fold, majority (95%) were deregulated similarly in patients with UPF3B mutations. This supports the major role of deletion of UPF2 in ID. Furthermore, we found that four other NMD genes, UPF3A, SMG6, EIF4A3 and RNPS1 are frequently deleted and/or duplicated in the patients. We postulate that dosage imbalances of these NMD genes are likely to be the causes or act as predisposing factors for neuro-developmental disorders. Our findings further emphasize the importance of NMD pathway(s) in learning and memory.
- Published
- 2013
42. Steric Clash in the SET Domain of Histone Methyltransferase NSD1 as a Cause of Sotos Syndrome and Its Genetic Heterogeneity in a Brazilian Cohort
- Author
-
Julie R. Jones, Lawrence C. Layman, Chong Ae Kim, Kyungsoo Ha, Jennifer A. Lee, Priya Anand, Hyung Goo Kim, Débora Romeo Bertola, Jonathan D J Labonne, and Wolfgang Wenzel
- Subjects
0301 basic medicine ,Technology ,lcsh:QH426-470 ,030105 genetics & heredity ,Biology ,medicine.disease_cause ,NSD1 ,Article ,03 medical and health sciences ,Genetics ,medicine ,Missense mutation ,Sotos syndrome ,SET domain ,histone methyltransferase ,intellectual disability ,Gene ,Genetics (clinical) ,Mutation ,Genetic heterogeneity ,medicine.disease ,3. Good health ,lcsh:Genetics ,030104 developmental biology ,Histone methyltransferase ,Overgrowth syndrome ,Haploinsufficiency ,ddc:600 - Abstract
Most histone methyltransferases (HMTase) harbor a predicted Su(var)3–9, Enhancer-of-zeste, Trithorax (SET) domain, which transfers a methyl group to a lysine residue in their substrates. Mutations of the SET domains were reported to cause intellectual disability syndromes such as Sotos, Weaver, or Kabuki syndromes. Sotos syndrome is an overgrowth syndrome with intellectual disability caused by haploinsufficiency of the nuclear receptor binding SET domain protein 1 (NSD1) gene, an HMTase at 5q35.2–35.3. Here, we analyzed NSD1 in 34 Brazilian Sotos patients and identified three novel and eight known mutations. Using protein modeling and bioinformatic approaches, we evaluated the effects of one novel (I2007F) and 21 previously reported missense mutations in the SET domain. For the I2007F mutation, we observed conformational change and loss of structural stability in Molecular Dynamics (MD) simulations which may lead to loss-of-function of the SET domain. For six mutations near the ligand-binding site we observed in simulations steric clashes with neighboring side chains near the substrate S-Adenosyl methionine (SAM) binding site, which may disrupt the enzymatic activity of NSD1. These results point to a structural mechanism underlying the pathology of the NSD1 missense mutations in the SET domain in Sotos syndrome. NSD1 mutations were identified in only 32% of the Brazilian Sotos patients in our study cohort suggesting other genes (including unknown disease genes) underlie the molecular etiology for the majority of these patients. Our studies also found NSD1 expression to be profound in human fetal brain and cerebellum, accounting for prenatal onset and hypoplasia of cerebellar vermis seen in Sotos syndrome.
- Published
- 2016
- Full Text
- View/download PDF
43. Translocations Disrupting PHF21A in the Potocki-Shaffer-Syndrome Region Are Associated with Intellectual Disability and Craniofacial Anomalies
- Author
-
Lisa G. Shaffer, Douglas M. Ruderfer, Fei Lan, Kyungsoo Ha, Bernd F. M. Romeike, Cynthia C. Morton, Tjitske Kleefstra, Lawrence C. Layman, Ihn Sik Seong, James F. Gusella, Bradley J. Quade, Anja Nowka, Hans-Hilger Ropers, Hyun Taek Kim, Sarah H. Elsea, Vera M. Kalscheuer, Oliver Bartsch, Ji Hyun Lee, Asli Silahtaroglu, David J. Harris, Yang Shi, Marcy E. MacDonald, Susanne Kjaergaard, Ingo Kurth, Cheol-Hee Kim, Niels Tommerup, Caron D. Glotzbach, Kerstin Kutsche, Reinhard Ullmann, Michael E. Talkowski, Ethylin Wang Jabs, Natalia T. Leach, Hyung Goo Kim, Alex V. Levin, and Yiping Shen
- Subjects
Adult ,Male ,Adolescent ,Genotype ,Potocki–Shaffer syndrome ,Chromosome Disorders ,Haploinsufficiency ,Biology ,Histone Deacetylases ,Sodium Channels ,Translocation, Genetic ,Article ,Chromatin remodeling ,Craniofacial Abnormalities ,03 medical and health sciences ,SCN3A ,0302 clinical medicine ,Intellectual Disability ,NAV1.3 Voltage-Gated Sodium Channel ,medicine ,Transcriptional regulation ,Genetics ,Animals ,Humans ,Deletion mapping ,Genetics(clinical) ,Craniofacial ,Zebrafish ,Genetics (clinical) ,030304 developmental biology ,0303 health sciences ,Chromosomes, Human, Pair 11 ,Infant, Newborn ,medicine.disease ,Genetics and epigenetic pathways of disease DCN MP - Plasticity and memory [NCMLS 6] ,Child, Preschool ,Homeobox ,Female ,Chromosome Deletion ,Exostoses, Multiple Hereditary ,030217 neurology & neurosurgery - Abstract
Contains fulltext : 110038.pdf (Publisher’s version ) (Closed access) Potocki-Shaffer syndrome (PSS) is a contiguous gene disorder due to the interstitial deletion of band p11.2 of chromosome 11 and is characterized by multiple exostoses, parietal foramina, intellectual disability (ID), and craniofacial anomalies (CFAs). Despite the identification of individual genes responsible for multiple exostoses and parietal foramina in PSS, the identity of the gene(s) associated with the ID and CFA phenotypes has remained elusive. Through characterization of independent subjects with balanced translocations and supportive comparative deletion mapping of PSS subjects, we have uncovered evidence that the ID and CFA phenotypes are both caused by haploinsufficiency of a single gene, PHF21A, at 11p11.2. PHF21A encodes a plant homeodomain finger protein whose murine and zebrafish orthologs are both expressed in a manner consistent with a function in neurofacial and craniofacial development, and suppression of the latter led to both craniofacial abnormalities and neuronal apoptosis. Along with lysine-specific demethylase 1 (LSD1), PHF21A, also known as BHC80, is a component of the BRAF-histone deacetylase complex that represses target-gene transcription. In lymphoblastoid cell lines from two translocation subjects in whom PHF21A was directly disrupted by the respective breakpoints, we observed derepression of the neuronal gene SCN3A and reduced LSD1 occupancy at the SCN3A promoter, supporting a direct functional consequence of PHF21A haploinsufficiency on transcriptional regulation. Our finding that disruption of PHF21A by translocations in the PSS region is associated with ID adds to the growing list of ID-associated genes that emphasize the critical role of transcriptional regulation and chromatin remodeling in normal brain development and cognitive function.
- Published
- 2012
- Full Text
- View/download PDF
44. Role of ART in Imprinting Disorders
- Author
-
Lawrence C. Layman and Ali Eroglu
- Subjects
Male ,Infertility ,medicine.medical_specialty ,RNA, Untranslated ,Reproductive Techniques, Assisted ,Endocrinology, Diabetes and Metabolism ,Reproductive technology ,Biology ,Article ,Histones ,Genomic Imprinting ,Mice ,Endocrinology ,Pregnancy ,Physiology (medical) ,Angelman syndrome ,Prevalence ,medicine ,Animals ,Humans ,Abnormalities, Multiple ,Imprinting (psychology) ,Psychiatry ,Genetics ,Incidence ,Gene Expression Regulation, Developmental ,Obstetrics and Gynecology ,DNA Methylation ,medicine.disease ,Pregnancy Complications ,Reproductive Medicine ,Transient neonatal diabetes mellitus ,Female ,Genomic imprinting ,Developed country - Abstract
Assisted reproductive technologies (ART) offer revolutionary infertility treatments for millions of childless couples around the world. Currently, ART accounts for 1 to 3% of annual births in industrialized countries and continues to expand rapidly. Except for an increased incidence of premature births, these technologies are considered safe. However, new evidence published during the past decade has suggested an increased incidence of imprinting disorders in children conceived by ART. Specifically, an increased risk was reported for Beckwith-Wiedemann syndrome (BWS), Angelman syndrome (AS), Silver-Russell syndrome, and retinoblastoma. In contrast, some studies have found no association between ART and BWS, AS, Prader-Willi syndrome, transient neonatal diabetes mellitus, and retinoblastoma. The variability in ART protocols and the rarity of imprinting disorders complicate determining the causative relationship between ART and an increased incidence of imprinting disorders. Nevertheless, compelling experimental data from animal studies also suggest a link between increased imprinting disorders and ART. Further comprehensive, appropriately powered studies are needed to better address the magnitude of the risk for ART-associated imprinting disorders. Large longitudinal studies are particularly critical to evaluate long-term effects of ART not only during the perinatal period but also into adulthood. An important consideration is to determine if the implicated association between ART and imprinting disorders is actually related to the procedures or to infertility itself.
- Published
- 2012
45. The role of CHD7 and the newly identified WDR11 gene in patients with idiopathic hypogonadotropic hypogonadism and Kallmann syndrome
- Author
-
Hyung Goo Kim and Lawrence C. Layman
- Subjects
Genetics ,GnRH Neuron ,medicine.medical_specialty ,Coloboma ,Positional cloning ,Heart malformation ,Kallmann syndrome ,Biology ,medicine.disease ,Biochemistry ,Chromodomain ,body regions ,CHARGE syndrome ,Endocrinology ,Hypogonadotropic hypogonadism ,Internal medicine ,medicine ,Molecular Biology - Abstract
Mutations in the chromodomain helicase DNA binding protein-7 (CHD7) cause CHARGE syndrome, which includes eye coloboma, heart malformations, atresia of the choanae, retardation of growth/development, genital anomalies, and ear abnormalities. CHARGE syndrome is usually sporadic, but is also autosomal dominant. CHD7 encodes a large protein that participates in chromatin remodeling and transcription. Findings from studies of mouse models employing ENU-mutagenesis or gene-trap methods recapitulate human CHARGE syndrome. CHARGE patients may manifest anosmia and/or hypogonadism, features that overlap with idiopathic hypogonadotropic hypogonadism (IHH) and Kallmann syndrome (KS). Similarly, IHH/KS patients may also display partial CHARGE features. Therefore, it has been hypothesized that IHH/KS represents a milder allelic variant of CHARGE syndrome, which has been supported by the identification of heterozygous CHD7 mutations in both normosmic IHH and KS. Developmental expression within the hypothalamus and the presence of human mutations indicate that CHD7 has an important role in puberty and reproduction. In addition, WDR11 was recently identified by positional cloning; and mutations in were identified in IHH/KS patients, suggesting a role for this gene in normal puberty.
- Published
- 2011
46. Retrieval of rhesus monkey (Macaca mulatta) oocytes by ultrasound-guided needle aspiration: Problems and solutions
- Author
-
Wei Si, Lawrence C. Layman, Ali Eroglu, Nancy A. Rodriguez, and Adelina M. Emmi
- Subjects
Infection risk ,biology ,medicine.diagnostic_test ,medicine.medical_treatment ,Cell Biology ,Anatomy ,Reproductive technology ,Oocyte ,Bioinformatics ,Ultrasound guided ,Recovery period ,medicine.anatomical_structure ,biology.animal ,Laparotomy ,Biopsy ,Genetics ,medicine ,Primate ,Developmental Biology - Abstract
Oocytes of nonhuman primates such as rhesus monkeys are excellent models for diverse studies on developmental biology, epigenetics, human reproduction, and assisted reproductive technologies, as well as on transgenics. Such studies require numerous oocytes that can be retrieved after controlled ovarian stimulation. Currently, most primate centers use laparoscopic aspiration or laparotomy followed by aspiration to collect rhesus oocytes, although the ultrasound-guided needle aspiration is more advantageous due to reduced infection risk, less injury, and a shorter recovery period. Yet, some initial difficulties associated with the ultrasound-guided needle aspiration limit its broader application. The objective of the present study was to address these obstacles. By presenting practical solutions to the initial difficulties, results from our study show that it is possible to collect a mean number of 38 +/- 10 rhesus oocytes per hormonally stimulated female. These results compare favorably to the average number of rhesus oocytes collected using the laparoscopic approach and suggest that when initial obstacles are overcome, the ultrasound-guided oocyte retrieval represents a good alternative to more invasive approaches.
- Published
- 2009
47. Phenotypic spectrum of 45,X/46,XY males with a ring Y chromosome and bilaterally descended testes
- Author
-
Andrew D. Clark, Anita S. Kulharya, Lawrence C. Layman, Sandra P.T. Tho, and Paul G. McDonough
- Subjects
Adult ,Male ,medicine.medical_specialty ,Genotype ,Ring chromosome ,Gonadal dysgenesis ,Biology ,Y chromosome ,Severity of Illness Index ,Short stature ,Article ,Internal medicine ,Testis ,medicine ,Body Size ,Humans ,Ring Chromosomes ,Testosterone ,Child ,Spermatogenesis ,Azoospermia ,Retrospective Studies ,Gonadal Dysgenesis, 46,XY ,Chromosomes, Human, Y ,Puberty ,Obstetrics and Gynecology ,Karyotype ,Oligospermia ,medicine.disease ,Phenotype ,Endocrinology ,Reproductive Medicine ,Karyotyping ,Gonadal Dysgenesis, Mixed ,medicine.symptom ,Gonadotropins - Abstract
Objective To characterize the phenotypic spectrum of males with bilaterally descended testes and a 45,X/46,X,(r)Y karyotype. Design Retrospective review of patient records; cytogenetic and molecular analysis. Setting Tertiary medical center setting. Participant(s) Five males, two prepubertal and three postpubertal, with a 45,X/46,X(r)Y karyotype and bilaterally descended testes. Intervention(s) Linear growth evaluation, testicular endocrine and exocrine studies, cytogenetic and molecular analysis on each patient. Main Outcome Measure(s) Clinical phenotype versus genotype. Result(s) Both prepubertal males had short stature and low testosterone. All three adults had normal puberty and normal testosterone levels. Two of the adults (one with short stature and one with normal stature) had elevated gonadotropins and azoospermia. The third adult had normal stature, severe oligospermia, normal gonadotropins, and normal serum testosterone. Conclusion(s) The phenotypic spectrum of males with a 45,X/46,X(r)Y karyotype and bilaterally descended testes varies greatly from males with short stature and spermatogenic failure to males without short stature and less severely affected spermatogenesis. This broad spectrum of phenotypic findings needs to be taken into account when the clinical geneticist encounters a prenatal diagnosis of a 45,X/46,X(r)Y karyotype. This information will also be helpful for pediatric and reproductive endocrinologists in counseling males with bilaterally descended testes and a 45,X/46,X(r)Y karyotype.
- Published
- 2009
48. Effects of follicle-stimulating hormone and human chorionic gonadotropin on gonadal steroidogenesis in two siblings with a follicle-stimulating hormone β subunit mutation
- Author
-
Lawrence C. Layman, Lucilia Domingues Casulari da Motta, Luciana Ansaneli Naves, Luiz Augusto Casulari, Leonardo Abdala Giacomini, Paula P. Nascimento, and Adriana Lofrano-Porto
- Subjects
Adult ,Male ,endocrine system ,medicine.medical_specialty ,medicine.drug_class ,Ovary ,Testicle ,Biology ,Chorionic Gonadotropin ,FSHB ,Human chorionic gonadotropin ,Follicle-stimulating hormone ,Internal medicine ,medicine ,Humans ,Endocrine system ,Androstenedione ,Gonadal Steroid Hormones ,Gonads ,Siblings ,Obstetrics and Gynecology ,Endocrinology ,medicine.anatomical_structure ,Reproductive Medicine ,Follicle Stimulating Hormone, beta Subunit ,Mutation ,Drug Therapy, Combination ,Female ,Follicle Stimulating Hormone ,Gonadotropin ,hormones, hormone substitutes, and hormone antagonists - Abstract
Objective To quantify gonadal steroid responses to different gonadotropin regimens. Design Transversal clinical study. Setting Academic medical center. Patient(s) A 41-year-old woman and her 37-year-old brother with isolated FSH deficiency due to a homozygous Tyr76X FSH β subunit gene ( FSHB ) mutation. Intervention(s) Initially, serial LH samples were drawn overnight. After 2-day dexamethasone suppression, steroids were measured at baseline and after hCG, recombinant FSH, or hCG + recombinant FSH administration. Main Outcome Measure(s) Pulse number, peak amplitude, and mean overnight LH levels, as well as basal and stimulated FSH, LH, T, E 2 , DHEAS, 17α-hydroxyprogesterone (17-OHP), and androstenedione (A). Result(s) The mean ±SD overnight LH was 49.2 ± 5.7 mIU/mL and 9.1 ± 2.9 mIU/mL; there were 8 pulses/8 hours and 9 pulses/9 hours, with mean amplitudes of 53.4 ± 6.5 mIU/mL and 11.7 ± 1.9 mIU/mL, for the woman and man, respectively. There was no steroid response to recombinant FSH, hCG, or hCG + recombinant FSH in the woman. In the man, T increased after hCG, recombinant FSH, and hCG + recombinant FSH, whereas E 2 , A, and 17-OHP increased only after hCG + recombinant FSH. Conclusion(s) This report constitutes the first detailed endocrine study of a man with isolated FSH deficiency due to an FSHB mutation and suggests that FSH may have a positive regulatory effect on healthy LH-stimulated Leydig cells, probably mediated by its primary action on Sertoli cells, in a paracrine mechanism.
- Published
- 2008
49. Mutations in CHD7, Encoding a Chromatin-Remodeling Protein, Cause Idiopathic Hypogonadotropic Hypogonadism and Kallmann Syndrome
- Author
-
Ingo Kurth, Gil Bu Kang, Metin Ozata, Irene Meliciani, Mustafa Tekin, Richard J. Sherins, Yang Shi, Wolfgang Wenzel, Georg Rosenberger, Fei Lan, Soo Hyun Eom, Lawrence C. Layman, James F. Gusella, David P. Bick, Hyung Goo Kim, and Steven L. Walker
- Subjects
Male ,medicine.medical_specialty ,Kallmann syndrome ,Molecular Sequence Data ,Molecular Conformation ,030209 endocrinology & metabolism ,Biology ,medicine.disease_cause ,Human puberty ,03 medical and health sciences ,CHARGE syndrome ,0302 clinical medicine ,Hypogonadotropic hypogonadism ,Report ,Internal medicine ,Genetics ,medicine ,Humans ,Missense mutation ,Genetics(clinical) ,Amino Acid Sequence ,Genetics (clinical) ,030304 developmental biology ,0303 health sciences ,Mutation ,Base Sequence ,Sequence Homology, Amino Acid ,Genetic heterogeneity ,Hypogonadism ,DNA Helicases ,Exons ,Kallmann Syndrome ,medicine.disease ,Chromatin ,Protein Structure, Tertiary ,3. Good health ,DNA-Binding Proteins ,body regions ,Endocrinology ,Female ,Congenital Hypogonadotropic Hypogonadism - Abstract
CHARGE syndrome and Kallmann syndrome (KS) are two distinct developmental disorders sharing overlapping features of impaired olfaction and hypogonadism. KS is a genetically heterogeneous disorder consisting of idiopathic hypogonadotropic hypogonadism (IHH) and anosmia, and is most commonly due to KAL1 or FGFR1 mutations. CHARGE syndrome, a multisystem autosomal-dominant disorder, is caused by CHD7 mutations. We hypothesized that CHD7 would be involved in the pathogenesis of IHH and KS (IHH/KS) without the CHARGE phenotype and that IHH/KS represents a milder allelic variant of CHARGE syndrome. Mutation screening of the 37 protein-coding exons of CHD7 was performed in 101 IHH/KS patients without a CHARGE phenotype. In an additional 96 IHH/KS patients, exons 6-10, encoding the conserved chromodomains, were sequenced. RT-PCR, SIFT, protein-structure analysis, and in situ hybridization were performed for additional supportive evidence. Seven heterozygous mutations, two splice and five missense, which were absent inor = 180 controls, were identified in three sporadic KS and four sporadic normosmic IHH patients. Three mutations affect chromodomains critical for proper CHD7 function in chromatin remodeling and transcriptional regulation, whereas the other four affect conserved residues, suggesting that they are deleterious. CHD7's role is further corroborated by specific expression in IHH/KS-relevant tissues and appropriate developmental expression. Sporadic CHD7 mutations occur in 6% of IHH/KS patients. CHD7 represents the first identified chromatin-remodeling protein with a role in human puberty and the second gene to cause both normosmic IHH and KS in humans. Our findings indicate that both normosmic IHH and KS are mild allelic variants of CHARGE syndrome and are caused by CHD7 mutations.
- Published
- 2008
50. Dicavitary Uteri with Twin Gestation: A Case Following Clomiphene Citrate Therapy and Review of Obstetric Outcomes
- Author
-
David T. Vandermolen, Lawrence D. Devoe, and Lawrence C. Layman
- Subjects
Adult ,medicine.medical_specialty ,Dizygotic twin ,Twins ,Uterus bicornis ,Clomiphene ,Anovulation ,Pregnancy ,medicine ,Humans ,Twin Pregnancy ,Gynecology ,Obstetrics ,business.industry ,Uterus ,Pregnancy Outcome ,Obstetrics and Gynecology ,medicine.disease ,Pregnancy Complications ,Twin gestation ,Pediatrics, Perinatology and Child Health ,Female ,Pregnancy, Multiple ,Congenital disease ,business ,Infertility, Female - Abstract
We report the first case of dicavitary twin pregnancy, following clomiphene citrate therapy, in a patient with uterus bicornis bicollis and anovulation. A review of the literature is presented, and obstetric outcomes and management of these rare pregnancies are discussed.
- Published
- 2008
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.