28 results on '"Denise M. Kay"'
Search Results
2. Exome sequencing of child–parent trios with bladder exstrophy: Findings in 26 children
- Author
-
Lawrence C. Brody, James L. Mills, Faith Pangilinan, Jennita Reefhuis, Gary M. Shaw, Richard H. Finnell, Kristin M Conway, Marcia L. Feldkamp, Charlotte A. Hobbs, Georgia Pitsava, Andrew F. Olshan, Lynn M Almli, John Lane, Michael J. Bamshad, Deborah A. Nickerson, Mary M. Jenkins, Robert J. Sicko, Nathan Pankratz, Denise M. Kay, Paul A. Romitti, James C. Mullikin, and Daniel McGoldrick
- Subjects
Adult ,Male ,Tetraspanins ,media_common.quotation_subject ,Nonsense ,Biology ,Compound heterozygosity ,Article ,Cell Movement ,Pregnancy ,Tubulin ,Exome Sequencing ,Genetic variation ,Cell Adhesion ,Genetics ,medicine ,Humans ,Missense mutation ,Exome ,Genetic Predisposition to Disease ,Allele frequency ,Gene ,Genetics (clinical) ,Exome sequencing ,media_common ,Bladder Exstrophy ,Infant, Newborn ,medicine.disease ,Bladder exstrophy ,Mutation ,Female - Abstract
Bladder exstrophy (BE) is a rare, lower ventral midline defect with the bladder and part of the urethra exposed. The etiology of BE is unknown but thought to be influenced by genetic variation with more recent studies suggesting a role for rare variants. As such, we conducted paired-end exome sequencing in 26 child/mother/father trios. Three children had rare (allele frequency ≤ 0.0001 in several public databases) inherited variants in TSPAN4, one with a loss-of-function variant and two with missense variants. Two children had loss-of-function variants in TUBE1. Four children had rare missense or nonsense variants (one per child) in WNT3, CRKL, MYH9, or LZTR1, genes previously associated with BE. We detected 17 de novo missense variants in 13 children and three de novo loss-of-function variants (AKR1C2, PRRX1, PPM1D) in three children (one per child). We also detected rare compound heterozygous loss-of-function variants in PLCH2 and CLEC4M and rare inherited missense or loss-of-function variants in additional genes applying autosomal recessive (three genes) and X-linked recessive inheritance models (13 genes). Variants in two genes identified may implicate disruption in cell migration (TUBE1) and adhesion (TSPAN4) processes, mechanisms proposed for BE, and provide additional evidence for rare variants in the development of this defect.
- Published
- 2021
3. Genetic drivers of Cushing's disease: Frequency and associated phenotypes
- Author
-
Laura C. Hernández-Ramírez, Nathan Pankratz, John Lane, Fabio R. Faucz, Prashant Chittiboina, Denise M. Kay, Zachary Beethem, James L. Mills, and Constantine A. Stratakis
- Subjects
Phenotype ,Neoplasms ,Exome Sequencing ,Humans ,Female ,Genetic Predisposition to Disease ,Pituitary ACTH Hypersecretion ,Genetics (clinical) ,Germ-Line Mutation - Abstract
Cushing's disease (CD) is often explained by a single somatic sequence change. Germline defects, however, often go unrecognized. We aimed to determine the frequency and associated phenotypes of genetic drivers of CD in a large cohort.We studied 245 unrelated patients with CD (139 female, 56.7%), including 230 (93.9%) pediatric and 15 (6.1%) adult patients. Germline exome sequencing was performed in 184 patients; tumor exome sequencing was also done in 27 of them. A total of 43 germline samples and 92 tumor samples underwent Sanger sequencing of specific genes. Rare variants of uncertain significance, likely pathogenic (LP), or pathogenic variants in CD-associated genes, were identified.Germline variants (13 variants of uncertain significance, 8 LP, and 11 pathogenic) were found in 8 of 19 patients (42.1%) with positive family history and in 23 of 226 sporadic patients (10.2%). Somatic variants (1 LP and 7 pathogenic) were found in 20 of 119 tested individuals (16.8%); one of them had a coexistent germline defect. Altogether, variants of interest were identified at the germline level in 12.2% of patients, at the somatic level in 7.8%, and coexisting germline and somatic variants in 0.4%, accounting for one-fifth of the cohort.We report an estimate of the contribution of multiple germline and somatic genetic defects underlying CD in a single cohort.
- Published
- 2022
4. Implementation of population-based newborn screening reveals low incidence of spinal muscular atrophy
- Author
-
Kristin Engelstad, Colleen F. Stevens, Emma Laureta, Claudia A. Chiriboga, April Parker, Simona Treidler, Emma Ciafaloni, Leslie Delfiner, Michele Caggana, Denise M. Kay, Yaacov Anziska, Carlos A. Saavedra-Matiz, Osman Farooq, Ai Sakonju, Bo Hoon Lee, Sohail Malek, Virginia Sack, and Wendy K. Chung
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Genetic counseling ,New York ,Prenatal diagnosis ,Reproductive technology ,SMN1 ,030105 genetics & heredity ,Muscular Atrophy, Spinal ,03 medical and health sciences ,Neonatal Screening ,Pregnancy ,Internal medicine ,Medicine ,Humans ,Genetics (clinical) ,Newborn screening ,business.industry ,Incidence ,Homozygote ,Infant, Newborn ,Obstetrics and Gynecology ,Infant ,General Medicine ,Spinal muscular atrophy ,SMA ,medicine.disease ,Survival of Motor Neuron 1 Protein ,030104 developmental biology ,Real-time polymerase chain reaction ,Female ,business - Abstract
Spinal muscular atrophy (SMA) was added to the Recommended Uniform Screening Panel (RUSP) in July 2018, following FDA approval of the first effective SMA treatment, and demonstration of feasibility of high-throughput newborn screening using a primary molecular assay. SMA newborn screening was implemented in New York State (NYS) on 1 October 2018. Screening was conducted using DNA extracted from dried blood spots with a multiplex real-time quantitative polymerase chain reaction (qPCR) assay targeting the recurrent SMN1 exon 7 gene deletion. During the first year, 225,093 infants were tested. Eight screened positive, were referred for follow-up, and confirmed to be homozygous for the deletion. Infants with two or three copies of the SMN2 gene, predicting more severe, earlier-onset SMA, were treated with antisense oligonucleotide and/or gene therapy. One infant with ≥4 copies SMN2 also received gene therapy. Newborn screening permits presymptomatic SMA diagnosis, when treatment initiation is most beneficial. At 1 in 28,137 (95% confidence interval [CI]: 1 in 14,259 to 55,525), the NYS SMA incidence is 2.6- to 4.7-fold lower than expected. The low SMA incidence is likely attributable to imprecise and biased estimates, coupled with increased awareness, access to and uptake of carrier screening, genetic counseling, cascade testing, prenatal diagnosis, and advanced reproductive technologies.
- Published
- 2020
5. A genome-wide association study implicates the BMP7 locus as a risk factor for nonsyndromic metopic craniosynostosis
- Author
-
Julie M. Phipps, Jenny Morton, Elizabeth Sweeney, Araceli Cuellar, Jeremy A. Sabourin, Assen Bussarsky, Val C. Sheffield, James L. Mills, Michael L. Cunningham, David W. Johnson, Karen Crawford, Krithi Bala, Mary M. Jenkins, Marta Barba, Louise C. Wilson, Cristina M. Justice, Nadezhda Yaneva, Lawrence C. Brody, Simeon A. Boyadjiev, Paul A. Romitti, Katie E. M. Rees, Andrew O.M. Wilkie, Wanda Lattanzi, Peter H. Langlois, Peter Noons, Yan Zhou, Rachel K. Tittle, Steven A. Wall, Tony Roscioli, Marike Zwienenberg, Denise M. Kay, Deirdre Cilliers, Kiril Georgiev, Jo C. Byren, Robert J. Sicko, Craig W. Senders, Lorenzo D. Botto, Alexander F. Wilson, Radka Kaneva, E Simeonov, Astrid Weber, Gianpiero Tamburrini, Kristin M Conway, James E. Boggan, and Janine M. LaSalle
- Subjects
Proband ,Linkage disequilibrium ,Genotype ,Bone Morphogenetic Protein 7 ,Genome-wide association study ,Single-nucleotide polymorphism ,Biology ,genome‑wide association study ,Polymorphism, Single Nucleotide ,Linkage Disequilibrium ,Article ,Craniosynostosis ,Paediatrics and Reproductive Medicine ,03 medical and health sciences ,single nucleotide polymorphisms ,Craniosynostoses ,Complementary and Alternative Medicine ,non syndromic craniosynostosis ,Genes, Reporter ,Risk Factors ,Genetics ,medicine ,GWAS ,Settore BIO/13 - BIOLOGIA APPLICATA ,Humans ,Genetic Predisposition to Disease ,Promoter Regions, Genetic ,Genetics (clinical) ,Alleles ,030304 developmental biology ,PCDH11X ,Genetics & Heredity ,0303 health sciences ,030305 genetics & heredity ,Genetic Variation ,Odds ratio ,DNA Methylation ,medicine.disease ,Introns ,craniosynostosis ,Imputation (genetics) ,Genome-Wide Association Study - Abstract
Our previous genome-wide association study (GWAS) for sagittal nonsyndromic craniosynostosis (sNCS) provided important insights into the genetics of midline CS. In this study, we performed a GWAS for a second midline NCS, metopic NCS (mNCS), using 215 non-Hispanic white case-parent triads. We identified six variants with genome-wide significance (P ≤ 5 × 10(−8)): rs781716 (P = 4.71 × 10(−9); odds ratio [OR] = 2.44) intronic to SPRY3; rs6127972 (P = 4.41 × 10(−8); OR = 2.17) intronic to BMP7; rs62590971 (P = 6.22 × 10(−9); OR = 0.34), located ~155 kb upstream from TGIF2LX; and rs2522623, rs2573826, and rs2754857, all intronic to PCDH11X (P = 1.76 × 10(−8), OR = 0.45; P = 3.31 × 10(−8), OR = 0.45; P = 1.09 × 10(−8), OR=0.44, respectively). We performed a replication study of these variants using an independent non-Hispanic white sample of 194 unrelated mNCS cases and 333 unaffected controls; only the association for rs6127972 (P = 0.004, OR = 1.45; meta-analysis P = 1.27 × 10(−8), OR = 1.74) was replicated. Our meta-analysis examining single nucleotide polymorphisms common to both our mNCS and sNCS studies showed the strongest association for rs6127972 (P = 1.16 × 10(−6)). Our imputation analysis identified a linkage disequilibrium block encompassing rs6127972, which contained an enhancer overlapping a CTCF transcription factor binding site (chr20:55,798,821–55,798,917) that was significantly hypomethylated in mesenchymal stem cells derived from fused metopic compared to open sutures from the same probands. This study provides additional insights into genetic factors in midline CS.
- Published
- 2020
6. Clinical outcomes of children with abnormal newborn screening results for Krabbe disease in New York State
- Author
-
Lissette Estrella, Patricia K. Duffner, Alejandro Iglesias, David A. Wenger, James M. Provenzale, Joseph J. Orsini, Jennifer M. Kwon, Denise M. Kay, Patricia Galvin-Parton, Georgianne L. Arnold, Joan E. Pellegrino, Maria L. Escolar, David Kronn, Michele Caggana, Joanne Kurtzberg, Richard W. Erbe, Alan M. Aron, Paul A. Levy, Mary R. Andriola, Thomas P. Naidich, Melissa P. Wasserstein, and Thomas J. Langan
- Subjects
0301 basic medicine ,Pediatrics ,medicine.medical_specialty ,medicine.medical_treatment ,New York ,Disease ,Hematopoietic stem cell transplantation ,Asymptomatic ,03 medical and health sciences ,Neonatal Screening ,0302 clinical medicine ,Risk Factors ,medicine ,Humans ,Mass Screening ,Genetics (clinical) ,Mass screening ,Newborn screening ,business.industry ,Leukodystrophy ,Hematopoietic Stem Cell Transplantation ,Infant, Newborn ,Infant ,medicine.disease ,Leukodystrophy, Globoid Cell ,Medicolegal issues ,030104 developmental biology ,Krabbe disease ,Female ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Early infantile Krabbe disease is rapidly fatal, but hematopoietic stem cell transplantation (HSCT) may improve outcomes if performed soon after birth. New York State began screening all newborns for Krabbe disease in 2006. Infants with abnormal newborn screen results for Krabbe disease were referred to specialty-care centers. Newborns found to be at high risk for Krabbe disease underwent a neurodiagnostic battery to determine the need for emergent HSCT. Almost 2 million infants were screened. Five infants were diagnosed with early infantile Krabbe disease. Three died, two from HSCT-related complications and one from untreated disease. Two children who received HSCT have moderate to severe developmental delays. Forty-six currently asymptomatic children are considered to be at moderate or high risk for development of later-onset Krabbe disease. These results show significant HSCT-associated morbidity and mortality in early infantile Krabbe disease and raise questions about its efficacy when performed in newborns diagnosed through newborn screening. The unanticipated identification of “at risk” children introduces unique ethical and medicolegal issues. New York’s experience raises questions about the risks, benefits, and practicality of screening newborns for Krabbe disease. It is imperative that objective assessments be made on an ongoing basis as additional states begin screening for this disorder. Genet Med 18 12, 1235–1243.
- Published
- 2016
7. Copy number variants in a population-based investigation of Klippel-Trenaunay syndrome
- Author
-
Charlotte M. Druschel, Ruzong Fan, Marilyn L. Browne, James L. Mills, Aggeliki Dimopoulos, Robert J. Sicko, Denise M. Kay, Lawrence C. Brody, Shannon L. Rigler, Paul A. Romitti, and Michele Caggana
- Subjects
0301 basic medicine ,Klippel-Trenaunay-Weber Syndrome ,Methyltransferase ,DNA Copy Number Variations ,Genotype ,Single-nucleotide polymorphism ,Polymorphism, Single Nucleotide ,Histone Deacetylases ,Article ,03 medical and health sciences ,0302 clinical medicine ,Gene duplication ,Prevalence ,Genetics ,Humans ,Genetic Testing ,Registries ,Copy-number variation ,Genetic Association Studies ,Genetics (clinical) ,Comparative Genomic Hybridization ,biology ,HDAC9 ,Chromosome Mapping ,Repressor Proteins ,030104 developmental biology ,Histone ,Case-Control Studies ,Population Surveillance ,030220 oncology & carcinogenesis ,biology.protein ,Histone deacetylase ,DNA microarray ,Maternal Age - Abstract
Klippel-Trenaunay syndrome (KTS) is a rare congenital vascular disorder that is thought to occur sporadically; however, reports of familial occurrence suggest a genetic component. We examined KTS cases to identify novel, potentially causal copy number variants (CNVs). We identified 17 KTS cases from all live-births occurring in New York (1998-2010). Extracted DNA was genotyped using Illumina microarrays and CNVs were called using PennCNV software. CNVs selected for follow-up had ≥10 single nucleotide polymorphisms (SNPs) and minimal overlap with in-house controls or controls from the Database of Genomic Variants. We identified 15 candidate CNVs in seven cases; among them a deletion in two cases within transcripts of HDAC9, a histone deacetylase essential for angiogenic sprouting of endothelial cells. One of them also had a duplication upstream of SALL3, a transcription factor essential for embryonic development that inhibits DNMT3A, a DNA methyltransferase responsible for embryonic de novo DNA methylation. Another case had a duplication spanning ING5, a histone acetylation regulator active during embryogenesis. We identified rare genetic variants related to chromatin modification which may have a key role in regulating vascular development during embryogenesis. Further investigation of their implications in the pathogenesis of KTS is warranted. © 2016 Wiley Periodicals, Inc.
- Published
- 2016
8. Copy-number variant analysis of classic heterotaxy highlights the importance of body patterning pathways
- Author
-
Robert J. Sicko, Lawrence C. Brody, Margaret H. Doleman, Erin M. Hagen, Shannon L. Rigler, Shabbir Ahmad, Michele Caggana, Denise M. Kay, Marilyn L. Browne, Ruzong Fan, Paul A. Romitti, James L. Mills, Gary M. Shaw, Aggeliki Dimopoulos, and Laura L. Jelliffe-Pawlowski
- Subjects
Heart Defects, Congenital ,Male ,0301 basic medicine ,DNA Copy Number Variations ,Genotype ,Microarray ,RBFOX1 ,Single-nucleotide polymorphism ,Heterotaxy Syndrome ,030204 cardiovascular system & hematology ,Biology ,Polymorphism, Single Nucleotide ,Article ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,Humans ,Copy-number variation ,Genetics (clinical) ,Body Patterning ,Wnt signaling pathway ,Infant ,NIPBL ,Human genetics ,Fibroblast Growth Factors ,MicroRNAs ,030104 developmental biology ,Female ,RNA Splicing Factors ,Heterotaxy ,Signal Transduction - Abstract
Classic heterotaxy consists of congenital heart defects with abnormally positioned thoracic and abdominal organs. We aimed to uncover novel, genomic copy-number variants (CNVs) in classic heterotaxy cases. A microarray containing 2.5 million single-nucleotide polymorphisms (SNPs) was used to genotype 69 infants (cases) with classic heterotaxy identified from California live births from 1998 to 2009. CNVs were identified using the PennCNV software. We identified 56 rare CNVs encompassing genes in the NODAL (NIPBL, TBX6), BMP (PPP4C), and WNT (FZD3) signaling pathways, not previously linked to classic heterotaxy. We also identified a CNV involving FGF12, a gene previously noted in a classic heterotaxy case. CNVs involving RBFOX1 and near MIR302F were detected in multiple cases. Our findings illustrate the importance of body patterning pathways for cardiac development and left/right axes determination. FGF12, RBFOX1, and MIR302F could be important in human heterotaxy, because they were noted in multiple cases. Further investigation into genes involved in the NODAL, BMP, and WNT body patterning pathways and into the dosage effects of FGF12, RBFOX1, and MIR302F is warranted.
- Published
- 2016
9. Rare copy number variants implicated in posterior urethral valves
- Author
-
Benjamin R. Cole, Ruzong Fan, Marilyn L. Browne, Robert J. Sicko, Denise M. Kay, Nansi S. Boghossian, James L. Mills, Michele Caggana, Paul A. Romitti, Edwina Yeung, Michael Y. Tsai, Charlotte M. Druschel, Aiyi Liu, Nathan Pankratz, Lawrence C. Brody, and Shannon L. Rigler
- Subjects
Male ,0301 basic medicine ,Posterior urethral valve ,DNA Copy Number Variations ,Genotype ,Tetraspanins ,Bone Morphogenetic Protein 7 ,Molecular Sequence Data ,Population ,New York ,030232 urology & nephrology ,Gene Expression ,Biology ,Polymorphism, Single Nucleotide ,Article ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Urethra ,mental disorders ,Gene duplication ,Genetics ,medicine ,Humans ,Copy-number variation ,education ,Gene ,Genetics (clinical) ,Oligonucleotide Array Sequence Analysis ,Sequence Deletion ,Urethral Stricture ,Comparative Genomic Hybridization ,education.field_of_study ,Base Sequence ,Infant ,Chromosome ,Cadherins ,medicine.disease ,Fibroblast Growth Factors ,Phenotype ,030104 developmental biology ,Case-Control Studies ,Child, Preschool ,Chromosomes, Human, Pair 6 ,Chromosomes, Human, Pair 3 ,Chromosomes, Human, Pair 17 ,Comparative genomic hybridization - Abstract
The cause of posterior urethral valves (PUV) is unknown, but genetic factors are suspected given their familial occurrence. We examined cases of isolated PUV to identify novel copy number variants (CNVs). We identified 56 cases of isolated PUV from all live-births in New York State (1998-2005). Samples were genotyped using Illumina HumanOmni2.5 microarrays. Autosomal and sex-linked CNVs were identified using PennCNV and cnvPartition software. CNVs were prioritized for follow-up if they were absent from in-house controls, contained ≥ 10 consecutive probes, were ≥ 20 Kb in size, had ≤ 20% overlap with variants detected in other birth defect phenotypes screened in our lab, and were rare in population reference controls. We identified 47 rare candidate PUV-associated CNVs in 32 cases; one case had a 3.9 Mb deletion encompassing BMP7. Mutations in BMP7 have been associated with severe anomalies in the mouse urethra. Other interesting CNVs, each detected in a single PUV case included: a deletion of PIK3R3 and TSPAN1, duplication/triplication in FGF12, duplication of FAT1--a gene essential for normal growth and development, a large deletion (>2 Mb) on chromosome 17q that involves TBX2 and TBX4, and large duplications (>1 Mb) on chromosomes 3q and 6q. Our finding of previously unreported novel CNVs in PUV suggests that genetic factors may play a larger role than previously understood. Our data show a potential role of CNVs in up to 57% of cases examined. Investigation of genes in these CNVs may provide further insights into genetic variants that contribute to PUV.
- Published
- 2015
10. Clinical Sensitivity of Cystic Fibrosis Mutation Panels in a Diverse Population
- Author
-
Patrick Van Roey, Ran D. Anbar, Karen Z. Voter, Catherine Kier, Zhen Zhang, Andrew Ting, Denise M. Kay, Allen J. Dozor, Norma P. Tavakoli, Maria Berdella, Danielle Goetz, Lea M. Krein, Colleen F. Stevens, Louis Guida, Michele Caggana, Breanne Maloney, Erin E. Hughes, Beth Vogel, Meyer Kattan, Paul G. Comber, Joan DeCelie-Germana, April Parker, and Carlos A. Saavedra-Matiz
- Subjects
0301 basic medicine ,Mutation ,education.field_of_study ,medicine.medical_specialty ,Newborn screening ,Population ,Biology ,medicine.disease_cause ,medicine.disease ,Bioinformatics ,Cystic fibrosis ,03 medical and health sciences ,030104 developmental biology ,Diverse population ,Internal medicine ,Genetics ,medicine ,Clinical validity ,education ,Dried blood ,Genotyping ,Genetics (clinical) - Abstract
Infants are screened for cystic fibrosis (CF) in New York State (NYS) using an IRT-DNA algorithm. The purpose of this study was to validate and assess clinical validity of the US FDA-cleared Illumina MiSeqDx CF 139-Variant Assay (139-VA) in the diverse NYS CF population. The study included 439 infants with CF identified via newborn screening (NBS) from 2002 to 2012. All had been screened using the Abbott Molecular CF Genotyping Assay or the Hologic InPlex CF Molecular Test. All with CF and zero or one mutation were tested using the 139-VA. DNA extracted from dried blood spots was reliably and accurately genotyped using the 139-VA. Sixty-three additional mutations were identified. Clinical sensitivity of three panels ranged from 76.2% (23 mutations recommended for screening by ACMG/ACOG) to 79.7% (current NYS 39-mutation InPlex panel), up to 86.0% for the 139-VA. For all, sensitivity was highest in Whites and lowest in the Black population. Although the sample size was small, there was a nearly 20% increase in sensitivity for the Black CF population using the 139-VA (68.2%) over the ACMG/ACOG and InPlex panels (both 50.0%). Overall, the 139-VA is more sensitive than other commercially available panels, and could be considered for NBS, clinical, or research laboratories conducting CF screening.
- Published
- 2015
11. Genome-wide meta-analysis identifies BARX1 and EML4-MTA3 as new loci associated with infantile hypertrophic pyloric stenosis
- Author
-
Frank Geller, James L. Mills, Bjarke Feenstra, Line Skotte, Michele Caggana, Sanne Gørtz, Denise M. Kay, Heather A. Boyd, David M. Hougaard, João Fadista, Agneta Nordenskjöld, Mads Melbye, Jonas Bybjerg-Grauholm, Paul A. Romitti, and Hans Matsson
- Subjects
medicine.medical_specialty ,Linkage disequilibrium ,Population ,Transcription Factors/genetics ,Serine Endopeptidases/genetics ,Single-nucleotide polymorphism ,Cell Cycle Proteins ,Pyloric Stenosis, Hypertrophic ,Biology ,Polymorphism, Single Nucleotide ,Cohort Studies ,03 medical and health sciences ,Neoplasm Proteins/genetics ,Genetics ,medicine ,SNP ,Humans ,Genetic Predisposition to Disease ,education ,Association Studies Article ,Pyloric Stenosis, Hypertrophic/genetics ,Molecular Biology ,Genetics (clinical) ,Hypertrophic Pyloric Stenosis ,Cell Cycle Proteins/genetics ,Homeodomain Proteins ,0303 health sciences ,education.field_of_study ,030305 genetics & heredity ,Serine Endopeptidases ,Infant, Newborn ,Infant ,General Medicine ,Odds ratio ,Microtubule-Associated Proteins/genetics ,Pylorus ,3. Good health ,Neoplasm Proteins ,medicine.anatomical_structure ,Case-Control Studies ,Medical genetics ,Homeodomain Proteins/genetics ,Microtubule-Associated Proteins ,Genome-Wide Association Study ,Transcription Factors - Abstract
Infantile hypertrophic pyloric stenosis (IHPS) is a disorder of young infants with a population incidence of ∼2/1000 live births, caused by hypertrophy of the pyloric sphincter smooth muscle. Reported genetic loci associated with IHPS explain only a minor proportion of IHPS risk. To identify new risk loci, we carried out a genome-wide meta-analysis on 1395 surgery-confirmed cases and 4438 controls, with replication in a set of 2427 cases and 2524 controls. We identified and replicated six independent genomic loci associated with IHPS risk at genome wide significance (P < 5 × 10-8), including novel associations with two single nucleotide polymorphisms (SNPs). One of these SNPs, rs6736913 [odds ratio (OR) = 2.32; P = 3.0 × 10-15], is a low frequency missense variant in EML4 at 2p21. The second SNP, rs1933683 (OR = 1.34; P = 3.1 × 10-9) is 1 kb downstream of BARX1 at 9q22.32, an essential gene for stomach formation in embryogenesis. Using the genome-wide complex trait analysis method, we estimated the IHPS SNP heritability to be 30%, and using the linkage disequilibrium score regression method, we found support for a previously reported genetic correlation of IHPS with lipid metabolism. By combining the largest collection of IHPS cases to date (3822 cases), with results generalized across populations of different ancestry, we elucidate novel mechanistic avenues of IHPS disease architecture.
- Published
- 2018
12. Copy number variants in hypoplastic right heart syndrome
- Author
-
Laura L. Jelliffe-Pawlowski, Michele Caggana, James L. Mills, Gary M. Shaw, Andreas Giannakou, Robert J. Sicko, Denise M. Kay, Wei Zhang, and Paul A. Romitti
- Subjects
0301 basic medicine ,Adult ,Heart Defects, Congenital ,Male ,DNA Copy Number Variations ,Heart Ventricles ,030204 cardiovascular system & hematology ,Biology ,California ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Chromosome 16 ,Pregnancy ,Risk Factors ,mental disorders ,Gene duplication ,Genetics ,medicine ,SALL1 ,Humans ,Genetic Predisposition to Disease ,Copy-number variation ,Heart Atria ,Genetics (clinical) ,ERBB4 ,Genetic Association Studies ,Chromosome Aberrations ,Heart development ,Pregnancy Outcome ,Middle Aged ,medicine.disease ,030104 developmental biology ,Phenotype ,RPGRIP1L ,Population Surveillance ,Female ,Hypoplastic right heart syndrome - Abstract
Hypoplastic right heart syndrome (HRHS) is a rare congenital defect characterized by underdeveloped and malformed structures of the right heart. Familial recurrence of HRHS indicates genetic factors contribute to its etiology. Our study investigates the presence of copy number variants (CNVs) in HRHS cases. We genotyped 42 HRHS cases identified from live births throughout California (2003-2010) using the Illumina HumanOmni2.5-8 array. We identified 14 candidate CNVs in 14 HRHS cases (33%) based on the genes included in the CNVs and their functions. Duplications overlapping part of ERBB4 were identified in two unrelated cases. ERBB4 is a neuregulin receptor with a pivotal role in cardiomyocyte differentiation and heart development. We also described a 7.5 Mb duplication at 16q11-12. Multiple genes in the duplicated region have previously been linked to heart defects and cardiac development, including RPGRIP1L, RBL2, SALL1, and MYLK3. Of the 14 validated CNVs, we identified four CNVs in close proximity to genes linked to the Wnt signaling pathway. This study expands on our previous work supporting the role of genetics in HRHS. We identified CNVs affecting crucial genes and signaling pathways involved in right heart development. ERBB4 and duplication of the 16q11-12 region are important areas for future investigation.
- Published
- 2018
13. Rare copy number variants identified in prune belly syndrome
- Author
-
Paul A. Romitti, Denise M. Kay, Ruzong Fan, Edwina Yeung, Michael Y. Tsai, Aggeliki Dimopoulos, Nansi S. Boghossian, James L. Mills, Andreas Giannakou, Aiyi Liu, Robert J. Sicko, Nathan Pankratz, Marilyn L. Browne, Michele Caggana, and Benjamin R. Cole
- Subjects
0301 basic medicine ,Adult ,Male ,DNA Copy Number Variations ,Genotype ,Population ,030232 urology & nephrology ,Biology ,Article ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Prune belly syndrome ,Gene duplication ,mental disorders ,Genetics ,medicine ,Humans ,Prune Belly Syndrome ,Copy-number variation ,education ,Gene ,Genetics (clinical) ,education.field_of_study ,Infant, Newborn ,General Medicine ,Sequence Analysis, DNA ,medicine.disease ,Phenotype ,3. Good health ,030104 developmental biology ,Female ,DNA microarray ,Congenital disorder - Abstract
Prune belly syndrome (PBS), also known as Eagle-Barrett syndrome, is a rare congenital disorder characterized by absence or hypoplasia of the abdominal wall musculature, urinary tract anomalies, and cryptorchidism in males. The etiology of PBS is largely unresolved, but genetic factors are implicated given its recurrence in families. We examined cases of PBS to identify novel pathogenic copy number variants (CNVs). A total of 34 cases (30 males and 4 females) with PBS identified from all live births in New York State (1998–2005) were genotyped using Illumina HumanOmni2.5 microarrays. CNVs were prioritized if they were absent from in-house controls, encompassed ≥10 consecutive probes, were ≥20 Kb in size, had ≤20% overlap with common variants in population reference controls, and had ≤20% overlap with any variant previously detected in other birth defect phenotypes screened in our laboratory. We identified 17 candidate autosomal CNVs; 10 cases each had one CNV and four cases each had two CNVs. The CNVs included a 158 Kb duplication at 4q22 that overlaps the BMPR1B gene; duplications of different sizes carried by two cases in the intron of STIM1 gene; a 67 Kb duplication 202 Kb downstream of the NOG gene, and a 1.34 Mb deletion including the MYOCD gene. The identified rare CNVs spanned genes involved in mesodermal, muscle, and urinary tract development and differentiation, which might help in elucidating the genetic contribution to PBS. We did not have parental DNA and cannot identify whether these CNVs were de novo or inherited. Further research on these CNVs, particularly BMP signaling is warranted to elucidate the pathogenesis of PBS.
- Published
- 2017
14. Pilot study of population-based newborn screening for spinal muscular atrophy in New York state
- Author
-
Ritu Jain, Jennifer N Kraszewski, Bianca Haser, Michele Caggana, Carrie Koval, Nicole M. LaMarca, Sally Dunaway Young, Wendy K. Chung, Anthony Albertorio, Colleen F. Stevens, Sarah P Andrew, Darryl C. De Vivo, Denise M. Kay, Lilian L. Cohen, and Veronica Ortiz
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Pediatrics ,Population ,Gene Dosage ,New York ,Pilot Projects ,Population based ,SMN1 ,030105 genetics & heredity ,Muscular Atrophy, Spinal ,03 medical and health sciences ,0302 clinical medicine ,Neonatal Screening ,medicine ,Humans ,education ,Genetics (clinical) ,education.field_of_study ,Newborn screening ,business.industry ,Infant, Newborn ,Infant ,Spinal muscular atrophy ,Exons ,medicine.disease ,SMA ,Survival of Motor Neuron 1 Protein ,Surgery ,Clinical trial ,Nusinersen ,Female ,business ,030217 neurology & neurosurgery ,Gene Deletion - Abstract
PurposeTo determine feasibility and utility of newborn screening for spinal muscular atrophy (SMA) in New York State.MethodsWe validated a multiplex TaqMan real-time quantitative polymerase chain reaction assay using dried blood spots for SMA. From January 2016 to January 2017, we offered, consented, and screened 3,826 newborns at three hospitals in New York City and tested newborns for the deletion in exon 7 of SMN1.ResultsNinety-three percent of parents opted in for SMA screening. Overall the SMA carrier frequency was 1.5%. We identified one newborn with a homozygous SMN1 deletion and two copies of SMN2, which strongly suggests the severe type 1 SMA phenotype. The infant was enrolled in the NURTURE clinical trial and was first treated with Spinraza at age 15 days. She is now age 12 months, meeting all developmental milestones, and free of any respiratory issues.ConclusionOur pilot study demonstrates the feasibility of population-based screening, the acceptance by families, and the benefit of newborn screening for SMA. We suggest that SMA be considered for addition to the national recommended uniform screening panel.
- Published
- 2017
15. Copy-number variants and candidate gene mutations in isolated split hand/foot malformation
- Author
-
Michele Caggana, James L. Mills, Robert J. Sicko, Ruzong Fan, Aiyi Liu, Zoё L Edmunds, Charlotte M. Druschel, Paul A. Romitti, Denise M. Kay, Marilyn L. Browne, Lawrence C. Brody, and Tonia C. Carter
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Candidate gene ,DNA Copy Number Variations ,Population ,Limb Deformities, Congenital ,Biology ,Regulatory Sequences, Nucleic Acid ,Real-Time Polymerase Chain Reaction ,Polymorphism, Single Nucleotide ,Article ,03 medical and health sciences ,Molecular genetics ,TP63 ,Genetics ,medicine ,Humans ,Copy-number variation ,education ,Genetics (clinical) ,Alleles ,Genetic Association Studies ,Chromosome Aberrations ,education.field_of_study ,congenital abnormalities ,TP63 gene ,Breakpoint ,copy number variation ,Reproducibility of Results ,Sequence Analysis, DNA ,split hand foot malformation ,3. Good health ,030104 developmental biology ,Phenotype ,Statistical genetics ,ectrodactyly ,Mutation ,Female ,congenital limb deformities ,Haploinsufficiency - Abstract
Split hand/foot malformation (SHFM) is a congenital limb deficiency with missing or shortened central digits. Some SHFM genes have been identified but the cause of many SHFM cases is unknown. We used single-nucleotide polymorphism (SNP) microarray analysis to detect copy-number variants (CNVs) in 25 SHFM cases without other birth defects from New York State (NYS), prioritized CNVs absent from population CNV databases, and validated these CNVs using quantitative real-time polymerase chain reaction (qPCR). We tested for the validated CNVs in seven cases from Iowa using qPCR, and also sequenced 36 SHFM candidate genes in all the subjects. Seven NYS cases had a potentially deleterious variant: two had a p.R225H or p.R225L mutation in TP63, one had a 17q25 microdeletion, one had a 10q24 microduplication and three had a 17p13.3 microduplication. In addition, one Iowa case had a de novo 10q24 microduplication. The 17q25 microdeletion has not been reported previously in SHFM and included two SHFM candidate genes (SUMO2 and GRB2), while the 10q24 and 17p13.3 CNVs had breakpoints within genomic regions that contained putative regulatory elements and a limb development gene. In SHFM pathogenesis, the microdeletion may cause haploinsufficiency of SHFM genes and/or deletion of their regulatory regions, and the microduplications could disrupt regulatory elements that control transcription of limb development genes.
- Published
- 2016
16. Anorectal atresia and Variants at Predicted Regulatory Sites in Candidate Genes
- Author
-
James L. Mills, Paul A. Romitti, Marilyn L. Browne, Mary Conley, Charlotte M. Druschel, Aiyi Liu, Tonia C. Carter, Denise M. Kay, Michele Caggana, Devon Kuehn, and Lawrence C. Brody
- Subjects
Genetics ,Candidate gene ,animal structures ,TCF4 ,Biology ,medicine.disease ,MKKS ,DNA binding site ,GLI2 ,DNA methylation ,medicine ,Imperforate anus ,Gene ,Genetics (clinical) - Abstract
Anorectal atresia is a serious birth defect of largely unknown etiology but candidate genes have been identified in animal studies and human syndromes. Because alterations in the activity of these genes might lead to anorectal atresia, we selected 71 common variants predicted to be in transcription factor binding sites, CpG windows, splice sites, and miRNA target sites of 25 candidate genes, and tested for their association with anorectal atresia. The study population comprised 150 anorectal atresia cases and 623 control infants without major malformations. Variants predicted to affect transcription factor binding, splicing, and DNA methylation in WNT3A, PCSK5, TCF4, MKKS, GLI2, HOXD12, and BMP4 were associated with anorectal atresia based on a nominal P value < 0.05. The GLI2 and BMP4 variants are reported to be moderately associated with gene expression changes (Spearman's rank correlation coefficients between -0.260 and 0.226). We did not find evidence for interaction between maternal pre-pregnancy obesity and variants in MKKS, a gene previously associated with obesity, on the risk of anorectal atresia. Our results for MKKS support previously suggested associations with anorectal malformations. Our findings suggest that more research is needed to determine whether altered GLI2 and BMP4 expression is important in anorectal atresia in humans.
- Published
- 2012
17. Evaluation of genes involved in limb development, angiogenesis, and coagulation as risk factors for congenital limb deficiencies
- Author
-
Charlotte M. Druschel, Paul A. Romitti, Tonia C. Carter, Lawrence C. Brody, Marilyn L. Browne, James L. Mills, Michele Caggana, Aiyi Liu, Denise M. Kay, and Devon Kuehn
- Subjects
Adult ,Male ,Apical ectodermal ridge ,Oncology ,medicine.medical_specialty ,Genotype ,Population ,Limb Deformities, Congenital ,Neovascularization, Physiologic ,Single-nucleotide polymorphism ,Polymorphism, Single Nucleotide ,Article ,Young Adult ,Internal medicine ,Morphogenesis ,Genetics ,medicine ,Humans ,Limb development ,SNP ,education ,Blood Coagulation ,Genetics (clinical) ,education.field_of_study ,biology ,Infant, Newborn ,Proteins ,Extremities ,Odds ratio ,Case-Control Studies ,Methylenetetrahydrofolate reductase ,biology.protein ,Female ,Fibroblast Growth Factor 10 - Abstract
We conducted a population-based case-control study of single nucleotide polymorphisms (SNPs) in selected genes to find common variants that play a role in the etiology of limb deficiencies (LDs). Included in the study were 389 infants with LDs of unknown cause and 980 unaffected controls selected from all births in New York State (NYS) for the years 1998-2005. We used cases identified from the NYS Department of Health (DOH) Congenital Malformations Registry. Genotypes were obtained for 132 SNPs in genes involved in limb development (SHH, WNT7A, FGF4, FGF8, FGF10, TBX3, TBX5, SALL4, GREM1, GDF5, CTNNB1, EN1, CYP26A1, CYP26B1), angiogenesis (VEGFA, HIF1A, NOS3), and coagulation (F2, F5, MTHFR). Genotype call rates were97% and SNPs were tested for departure from Hardy-Weinberg expectations by race/ethnic subgroups. For each SNP, odds ratios (OR)s and confidence intervals (CI)s were estimated and corrected for multiple comparisons for all LDs combined and for LD subtypes. Among non-Hispanic white infants, associations between FGF10 SNPs rs10805683 and rs13170645 and all LDs combined were statistically significant following correction for multiple testing (OR = 1.99; 95% CI = 1.43-2.77; uncorrected P = 0.000043 for rs10805683 heterozygous genotype, and OR = 2.37; 95% CI = 1.48-3.78; uncorrected P = 0.00032 for rs13170645 homozygous minor genotype). We also observed suggestive evidence for associations with SNPs in other genes including CYP26B1 and WNT7A. Animal studies have shown that FGF10 induces formation of the apical ectodermal ridge and is necessary for limb development. Our data suggest that common variants in FGF10 increase the risk for a wide range of non-syndromic limb deficiencies.
- Published
- 2012
18. Folate and vitamin B12-related genes and risk for omphalocele
- Author
-
Denise M. Kay, Paul A. Romitti, Charlotte M. Druschel, Tonia C. Carter, James L. Mills, Aiyi Liu, Michele Caggana, Lawrence C. Brody, and Marilyn L. Browne
- Subjects
Risk ,medicine.medical_specialty ,Genotype ,Homocysteine ,Pilot Projects ,Receptors, Cell Surface ,Single-nucleotide polymorphism ,Polymorphism, Single Nucleotide ,Article ,chemistry.chemical_compound ,Folic Acid ,Transcobalamin ,Internal medicine ,Genetics ,medicine ,Humans ,SNP ,Genetic Predisposition to Disease ,Vitamin B12 ,Methylenetetrahydrofolate Reductase (NADPH2) ,Genetics (clinical) ,Omphalocele ,biology ,Infant, Newborn ,medicine.disease ,Vitamin B 12 ,Endocrinology ,chemistry ,Methylenetetrahydrofolate reductase ,biology.protein ,Population study ,Hernia, Umbilical - Abstract
Both taking folic acid-containing vitamins around conception and consuming food fortified with folic acid have been reported to reduce omphalocele rates. Genetic factors are etiologically important in omphalocele as well; our pilot study showed a relationship with the folate metabolic enzyme gene methylenetetrahydrofolate reductase (MTHFR). We studied 169 non-aneuploid omphalocele cases and 761 unaffected, matched controls from all New York State births occurring between 1998 and 2005 to look for associations with single nucleotide polymorphisms (SNPs) known to be important in folate, vitamin B12, or choline metabolism. In the total study population, variants in the transcobalamin receptor gene (TCblR), rs2232775 (p.Q8R), and the MTHFR gene, rs1801131 (c.1298A>C), were significantly associated with omphalocele. In African-Americans, significant associations were found with SNPs in genes for the vitamin B12 transporter (TCN2) and the vitamin B12 receptor (TCblR). A SNP in the homocysteine-related gene, betaine-homocysteine S-methyltransferase (BHMT), rs3733890 (p.R239Q), was significantly associated with omphalocele in both African-Americans and Asians. Only the TCblR association in the total population remained statistically significant if Bonferroni correction was applied. The finding that transcobalamin receptor (TCblR) and transporter (TCN2) SNPs and a BHMT SNP were associated with omphalocele suggests that disruption of methylation reactions, in which folate, vitamin B12, and homocysteine play critical parts, may be a risk factor for omphalocele. Our data, if confirmed, suggest that supplements containing both folic acid and vitamin B12 may be beneficial in preventing omphaloceles.
- Published
- 2011
19. Genetic association between α-synuclein and idiopathic parkinson's disease
- Author
-
Denise M. Kay, Donald S. Higgins, Robert G. Keefe, Berta C. Leis, John W. Roberts, April J. Atkins, Dora Yearout, Ali Samii, Haydeh Payami, Alida Griffith, Jennifer S. Montimurro, Cyrus P. Zabetian, Stewart A. Factor, and John G. Nutt
- Subjects
Adult ,medicine.medical_specialty ,Neurogenetics ,Biology ,Cellular and Molecular Neuroscience ,Gene Frequency ,Polymorphism (computer science) ,Internal medicine ,medicine ,Humans ,Genetic Predisposition to Disease ,Age of Onset ,Risk factor ,Allele ,Promoter Regions, Genetic ,Allele frequency ,Alleles ,Genetics (clinical) ,Aged ,Genetic association ,Family Health ,Genetics ,Polymorphism, Genetic ,Parkinson Disease ,Odds ratio ,Middle Aged ,Psychiatry and Mental health ,Endocrinology ,Case-Control Studies ,alpha-Synuclein ,Age of onset - Abstract
Point mutations and copy number variations in SNCA, the gene encoding alpha-synuclein, cause familial Parkinson's disease (PD). A dinucleotide polymorphism (REP1) in the SNCA promoter may be a risk factor for common forms of PD. We studied 1,802 PD patients and 2,129 controls from the NeuroGenetics Research Consortium, using uniform, standardized protocols for diagnosis, subject recruitment, data collection, genotyping, and data analysis. Three common REP1 alleles (257, 259, and 261 bp, with control frequencies of 0.28, 0.65, and 0.06) and several rare alleles (combined frequency
- Published
- 2008
20. Exploring gene-environment interactions in Parkinson’s disease
- Author
-
Berta C. Leis, Denise M. Kay, Alida Griffith, Colin Craig McCulloch, John W. Roberts, John G. Nutt, Donald S. Higgins, Haydeh Payami, Jennifer S. Montimurro, Stewart A. Factor, Ali Samii, and Cyrus P. Zabetian
- Subjects
Adult ,Male ,Apolipoprotein E ,Oncology ,medicine.medical_specialty ,Genotype ,Neurogenetics ,tau Proteins ,Disease ,Environment ,Biology ,Logistic regression ,Apolipoproteins E ,Gene interaction ,Risk Factors ,Surveys and Questionnaires ,Internal medicine ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Genotyping ,Genetics (clinical) ,Aged ,Aged, 80 and over ,Polymorphism, Genetic ,Haplotype ,Genetic Variation ,Parkinson Disease ,Odds ratio ,Middle Aged ,Haplotypes ,Case-Control Studies ,alpha-Synuclein ,Ubiquitin Thiolesterase - Abstract
The objective of this study was to explore combined effects of four candidate susceptibility genes and two exposures on Parkinson's disease (PD) risk; namely, alpha-synuclein (SNCA) promoter polymorphism REP1, microtubule-associated protein tau (MAPT) H1/H2 haplotypes, apolipoprotein E (APOE) epsilon2/epsilon3/epsilon4 polymorphism, ubiquitin carboxy-terminal esterase L1 (UCHL1) S18Y variant, cigarette smoking and caffeinated coffee consumption. 932 PD patients and 664 control subjects from the NeuroGenetics Research Consortium, with complete data on all six factors, were studied. Uniform protocols were used for diagnosis, recruitment, data collection and genotyping. A logistic regression model which included gene-exposure interactions was applied. Likelihood ratio tests (LRTs) were used for significance testing and Bayesian inference was used to estimate odds ratios (ORs). MAPT (P = 0.007), SNCA REP1 (P = 0.012), smoking (P = 0.001), and coffee (P = 0.011) were associated with PD risk. Two novel interactions were detected: APOE with coffee (P = 0.005), and REP1 with smoking (P = 0.021). While the individual main effects were modest, each yielding OR1.6, the effects were cumulative, with some combinations reaching OR = 12.6 (95% CI: 5.9-26.8). This study provides evidence for the long-held notion that PD risk is modulated by cumulative and interactive effects of genes and exposures. Furthermore, the study demonstrates that while interaction studies are useful for exploring risk relationships that might otherwise go undetected, results should be interpreted with caution because of the inherent loss of power due to multiple testing. The novel findings of this study that warrant replication are the evidence for interaction of coffee with APOE, and of smoking with REP1 on PD risk.
- Published
- 2008
21. Newborn screening for Krabbe disease in New York State: the first eight years' experience
- Author
-
Patricia Galvin-Parton, David A. Wenger, Melissa P. Wasserstein, Denise M. Kay, Joan E. Pellegrino, Lea M. Krein, David Kronn, Carlos A. Saavedra-Matiz, Michele Caggana, Richard W. Erbe, Jennifer M. Kwon, Maria L. Escolar, Alejandro D. Iglesias, Chad K. Biski, Natasha Shur, Monica Martin, Georgianne L. Arnold, Joseph J. Orsini, Paul A. Levy, Matthew Nichols, Joanne Kurtzberg, Darius J. Adams, and Patricia K. Duffner
- Subjects
0301 basic medicine ,Pediatrics ,medicine.medical_specialty ,medicine.medical_treatment ,New York ,Neurological examination ,Hematopoietic stem cell transplantation ,Asymptomatic ,Polymorphism, Single Nucleotide ,Mass Spectrometry ,03 medical and health sciences ,0302 clinical medicine ,Neonatal Screening ,Predictive Value of Tests ,Medicine ,Humans ,Genetics (clinical) ,Newborn screening ,medicine.diagnostic_test ,business.industry ,Leukodystrophy ,Hematopoietic Stem Cell Transplantation ,Infant, Newborn ,medicine.disease ,Leukodystrophy, Globoid Cell ,Transplantation ,030104 developmental biology ,Treatment Outcome ,Predictive value of tests ,Krabbe disease ,Dried Blood Spot Testing ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Algorithms ,Galactosylceramidase - Abstract
Krabbe disease (KD) results from galactocerebrosidase (GALC) deficiency. Infantile KD symptoms include irritability, progressive stiffness, developmental delay, and death. The only potential treatment is hematopoietic stem cell transplantation. New York State (NYS) implemented newborn screening for KD in 2006. Dried blood spots from newborns were assayed for GALC enzyme activity using mass spectrometry, followed by molecular analysis for those with low activity (≤12% of the daily mean). Infants with low enzyme activity and one or more mutations were referred for follow-up diagnostic testing and neurological examination. Of >1.9 million screened, 620 infants were subjected to molecular analysis and 348 were referred for diagnostic testing. Five had enzyme activities and mutations consistent with infantile KD and manifested clinical/neurodiagnostic abnormalities. Four underwent transplantation, two are surviving with moderate to severe handicaps, and two died from transplant-related complications. The significance of many sequence variants identified is unknown. Forty-six asymptomatic infants were found to be at moderate to high risk for disease. The positive predictive value of KD screening in NYS is 1.4% (5/346) considering confirmed infantile cases. The incidence of infantile KD in NYS is approximately 1 in 394,000, but it may be higher for later-onset forms. Genet Med 18 3, 239–248.
- Published
- 2015
22. Novel copy-number variants in a population-based investigation of classic heterotaxy
- Author
-
Michele Caggana, Robert J. Sicko, Marilyn L. Browne, Paul A. Romitti, Aiyi Liu, Ruzong Fan, James L. Mills, Denise M. Kay, Charlotte M. Druschel, Lawrence C. Brody, and Shannon L. Rigler
- Subjects
Male ,Candidate gene ,Microarray ,DNA Copy Number Variations ,Genotype ,New York ,Biology ,Polymorphism, Single Nucleotide ,Article ,Congenital Abnormalities ,Risk Factors ,Humans ,Copy-number variation ,Genotyping ,Genetics (clinical) ,Genetic Association Studies ,Sequence Deletion ,Genetics ,Comparative Genomic Hybridization ,Genetic heterogeneity ,Infant, Newborn ,Infant ,Phenotype ,Case-Control Studies ,Population Surveillance ,Female ,DNA microarray ,Heterotaxy ,Comparative genomic hybridization - Abstract
Heterotaxy is a clinically and genetically heterogeneous disorder. We investigated whether screening cases restricted to a classic phenotype would result in the discovery of novel, potentially causal copy-number variants. We identified 77 cases of classic heterotaxy from all live births in New York State during 1998–2005. DNA extracted from each infant’s newborn dried blood spot was genotyped with a microarray containing 2.5 million single-nucleotide polymorphisms. Copy-number variants were identified with PennCNV and cnvPartition software. Candidates were selected for follow-up if they were absent in unaffected controls, contained 10 or more consecutive probes, and had minimal overlap with variants published in the Database of Genomic Variants. We identified 20 rare copy-number variants including a deletion of BMP2, which has been linked to laterality disorders in mice but not previously reported in humans. We also identified a large, terminal deletion of 10q and a microdeletion at 1q23.1 involving the MNDA gene; both are rare variants suspected to be associated with heterotaxy. Our findings implicate rare copy-number variants in classic heterotaxy and highlight several candidate gene regions for further investigation. We also demonstrate the efficacy of copy-number variant genotyping in blood spots using microarrays. Genet Med 17 5, 348–357.
- Published
- 2014
23. Replication and exploratory analysis of 24 candidate risk polymorphisms for neural tube defects
- Author
-
Barry Shane, Marilyn L. Browne, Anne Parle-McDermott, Hatice Ozel Abaan, James Troendle, John M. Scott, Michele Caggana, Lawrence C. Brody, Marie Sutton, Emily C. McGrath, Denise M. Kay, Peadar N. Kirke, James L. Mills, Faith Pangilinan, and Anne M. Molloy
- Subjects
Folate ,Folic acid ,Adenosine Deaminase ,5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase ,Genotype ,2.1 Biological and endogenous factors ,Genetics(clinical) ,Neural Tube Defects ,Aetiology ,Genetics (clinical) ,Pediatric ,African Americans ,Genetics & Heredity ,Genetics ,education.field_of_study ,Nuclear Proteins ,Single Nucleotide ,One-carbon metabolism ,Asians ,DNA-Binding Proteins ,Cohort ,Research Article ,Asian Continental Ancestry Group ,congenital, hereditary, and neonatal diseases and abnormalities ,European Continental Ancestry Group ,Clinical Sciences ,Population ,New York ,Replication ,Single-nucleotide polymorphism ,Biology ,Polymorphism, Single Nucleotide ,Aldehyde Dehydrogenase 1 Family ,White People ,Rare Diseases ,Asian People ,Clinical Research ,SNP ,Humans ,Genetic Predisposition to Disease ,Polymorphism ,Allele ,education ,Spina bifida ,Genetic Association Studies ,Genetic association ,Whites ,Prevention ,Neurosciences ,Retinal Dehydrogenase ,United Kingdom ,Black or African American ,Good Health and Well Being ,Multiple comparisons problem ,Congenital Structural Anomalies ,Aldehyde Dehydrogenase 1 ,Transcription Factors - Abstract
Background Neural tube defects (NTDs), which are among the most common congenital malformations, are influenced by environmental and genetic factors. Low maternal folate is the strongest known contributing factor, making variants in genes in the folate metabolic pathway attractive candidates for NTD risk. Multiple studies have identified nominally significant allelic associations with NTDs. We tested whether associations detected in a large Irish cohort could be replicated in an independent population. Methods Replication tests of 24 nominally significant NTD associations were performed in racially/ethnically matched populations. Family-based tests of fifteen nominally significant single nucleotide polymorphisms (SNPs) were repeated in a cohort of NTD trios (530 cases and their parents) from the United Kingdom, and case–control tests of nine nominally significant SNPs were repeated in a cohort (190 cases, 941 controls) from New York State (NYS). Secondary hypotheses involved evaluating the latter set of nine SNPs for NTD association using alternate case–control models and NTD groupings in white, African American and Hispanic cohorts from NYS. Results Of the 24 SNPs tested for replication, ADA rs452159 and MTR rs10925260 were significantly associated with isolated NTDs. Of the secondary tests performed, ARID1A rs11247593 was associated with NTDs in whites, and ALDH1A2 rs7169289 was associated with isolated NTDs in African Americans. Conclusions We report a number of associations between SNP genotypes and neural tube defects. These associations were nominally significant before correction for multiple hypothesis testing. These corrections are highly conservative for association studies of untested hypotheses, and may be too conservative for replication studies. We therefore believe the true effect of these four nominally significant SNPs on NTD risk will be more definitively determined by further study in other populations, and eventual meta-analysis. Electronic supplementary material The online version of this article (doi:10.1186/s12881-014-0102-9) contains supplementary material, which is available to authorized users.
- Published
- 2014
24. Hirschsprung’s disease and variants in genes that regulate enteric neural crest cell proliferation, migration and differentiation
- Author
-
Devon Kuehn, Marilyn L. Browne, Aiyi Liu, Paul A. Romitti, Tonia C. Carter, James L. Mills, Mary Conley, Michele Caggana, Lawrence C. Brody, Denise M. Kay, and Charlotte M. Druschel
- Subjects
Adult ,Male ,Candidate gene ,L1 ,endocrine system diseases ,Adolescent ,Cellular differentiation ,Population ,Biology ,Polymorphism, Single Nucleotide ,Proto-Oncogene Mas ,Article ,Enteric Nervous System ,Cell Movement ,Genetics ,medicine ,Ethnicity ,Humans ,Hirschsprung Disease ,education ,Child ,Hirschsprung's disease ,Genetics (clinical) ,Genetic Association Studies ,Cell Proliferation ,Homeodomain Proteins ,education.field_of_study ,Proto-Oncogene Proteins c-ret ,Neural crest ,Cell Differentiation ,medicine.disease ,Neural Crest ,Child, Preschool ,Homeobox ,Female ,RNA Splice Sites ,Transcription Factors - Abstract
Hirschsprung's disease (HSCR) results from failed colonization of the embryonic gut by enteric neural crest cells (ENCCs); colonization requires RET proto-oncogene (RET) signaling. We sequenced RET to identify coding and splice-site variants in a population-based case group and we tested for associations between HSCR and common variants in RET and candidate genes (ASCL1, homeobox B5 (HOXB5), L1 cell adhesion molecule (L1CAM), paired-like homeobox 2b (PHOX2B), PROK1 and PROKR1) chosen because they are involved in ENCC proliferation, migration and differentiation in animal models. We conducted a nested case-control study of 304 HSCR cases and 1215 controls. Among 38 (12.5%) cases with 34 RET coding and splice-site variants, 18 variants were previously unreported. We confirmed associations with common variants in HOXB5 and PHOX2B but the associations with variants in ASCL1, L1CAM and PROK1 were not significant after multiple comparisons adjustment. RET variants were strongly associated with HSCR (P-values between 10(-3) and 10(-31)) but this differed by race/ethnicity: associations were absent in African-Americans. Our population-based study not only identified novel RET variants in HSCR cases, it showed that common RET variants may not contribute to HSCR in all race/ethnic groups. The findings for HOXB5 and PHOX2B provide supportive evidence that genes regulating ENCC proliferation, migration and differentiation could be risk factors for HSCR.
- Published
- 2012
25. Genome-Wide Gene-Environment Study Identifies Glutamate Receptor Gene GRIN2A as a Parkinson's Disease Modifier Gene via Interaction with Coffee
- Author
-
Jianjun Gao, William K. Scott, Albert Tenesa, Denise M. Kay, Yvette Bordelon, Pinky Agarwal, Silviu Alin Bacanu, Patricia Sheehan, Jeffery M. Vance, Beate Ritz, John G. Nutt, Cyrus P. Zabetian, Erin M. Hill-Burns, Ali Samii, Shannon L. Rhodes, Liyong Wang, Stewart A. Factor, Muthukrishnan Eaaswarkhanth, Haydeh Payami, Jennifer S. Montimurro, Taye H. Hamza, John W. Roberts, Yikyung Park, Victoria I. Kusel, Kenneth S. Kendler, Dora Yearout, and Honglei Chen
- Subjects
Male ,Cancer Research ,Genotype ,Neurogenetics ,Single-nucleotide polymorphism ,Genome-wide association study ,QH426-470 ,Bioinformatics ,Coffee ,Polymorphism, Single Nucleotide ,Receptors, N-Methyl-D-Aspartate ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Genetics ,SNP ,Humans ,Genetic Predisposition to Disease ,Genetics(clinical) ,Gene–environment interaction ,10. No inequality ,Biology ,Molecular Biology ,Genetics (clinical) ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,0303 health sciences ,biology ,Genome, Human ,Parkinson Disease ,3. Good health ,Case-Control Studies ,biology.protein ,Medicine ,GRIN2A ,Female ,Gene-Environment Interaction ,030217 neurology & neurosurgery ,Pharmacogenetics ,Research Article ,Genome-Wide Association Study - Abstract
Our aim was to identify genes that influence the inverse association of coffee with the risk of developing Parkinson's disease (PD). We used genome-wide genotype data and lifetime caffeinated-coffee-consumption data on 1,458 persons with PD and 931 without PD from the NeuroGenetics Research Consortium (NGRC), and we performed a genome-wide association and interaction study (GWAIS), testing each SNP's main-effect plus its interaction with coffee, adjusting for sex, age, and two principal components. We then stratified subjects as heavy or light coffee-drinkers and performed genome-wide association study (GWAS) in each group. We replicated the most significant SNP. Finally, we imputed the NGRC dataset, increasing genomic coverage to examine the region of interest in detail. The primary analyses (GWAIS, GWAS, Replication) were performed using genotyped data. In GWAIS, the most significant signal came from rs4998386 and the neighboring SNPs in GRIN2A. GRIN2A encodes an NMDA-glutamate-receptor subunit and regulates excitatory neurotransmission in the brain. Achieving P2df = 10−6, GRIN2A surpassed all known PD susceptibility genes in significance in the GWAIS. In stratified GWAS, the GRIN2A signal was present in heavy coffee-drinkers (OR = 0.43; P = 6×10−7) but not in light coffee-drinkers. The a priori Replication hypothesis that “Among heavy coffee-drinkers, rs4998386_T carriers have lower PD risk than rs4998386_CC carriers” was confirmed: ORReplication = 0.59, PReplication = 10−3; ORPooled = 0.51, PPooled = 7×10−8. Compared to light coffee-drinkers with rs4998386_CC genotype, heavy coffee-drinkers with rs4998386_CC genotype had 18% lower risk (P = 3×10−3), whereas heavy coffee-drinkers with rs4998386_TC genotype had 59% lower risk (P = 6×10−13). Imputation revealed a block of SNPs that achieved P2df, Author Summary Parkinson's disease (PD), like most common disorders, involves interactions between genetic make-up and environmental exposures that are unique to each individual. Caffeinated-coffee consumption may protect some people from developing PD, although not all benefit equally. In a genome-wide search, we discovered that variations in the glutamate-receptor gene GRIN2A modulate the risk of developing PD in heavy coffee drinkers. The study was hypothesis-free, that is, we cast a net across the entire genome allowing statistical significance to point us to a genetic variant, regardless of whether it fell in a genomic desert or an important gene. Fortuitously, the most significant finding was in a well-known gene, GRIN2A, which regulates brain signals that control movement and behavior. Our finding is important for three reasons: First, it is a proof of concept that studying genes and environment on the whole-genome scale is feasible, and this approach can identify important genes that are missed when environmental exposures are ignored. Second, the knowledge of interaction between GRIN2A, which is involved in neurotransmission in the brain, and caffeine, which is an adenosine-A2A-receptor antagonist, will stimulate new research towards understanding the cause and progression of PD. Third, the results may lead to personalized prevention of and treatment for PD.
- Published
- 2011
26. Visualizing disease associations: graphic analysis of frequency distributions as a function of age using moving average plots (MAP) with application to Alzheimer's and Parkinson's disease
- Author
-
Gerard D. Schellenberg, Denise M. Kay, Stewart A. Factor, Cyrus P. Zabetian, Haydeh Payami, and Colin Craig McCulloch
- Subjects
Adult ,Genetic Markers ,Epidemiology ,Genome-wide association study ,Quantitative trait locus ,Biology ,Article ,Apolipoproteins E ,Gene Frequency ,Alzheimer Disease ,Risk Factors ,Humans ,Age of Onset ,Spurious relationship ,Allele frequency ,Alleles ,Genetics (clinical) ,Aged ,Genetic association ,Aged, 80 and over ,Genetics ,Models, Statistical ,Models, Genetic ,Confounding ,Age Factors ,Parkinson Disease ,Middle Aged ,Case-Control Studies ,Age of onset ,Frequency distribution ,Cartography ,Genome-Wide Association Study - Abstract
Age-related variation in marker frequency can be a confounder in association studies, leading to both false positive and false negative findings and subsequently to inconsistent reproducibility. We have developed a simple method, based on a novel extension of moving average plots (MAP), which allows investigators to inspect the frequency data for hidden age-related variations. MAP uses the standard case-control association data and generates a birds-eye view of the frequency distributions across the age spectrum; a picture in which one can see if, how, and when the marker frequencies in cases differ from that in controls. The marker can be specified as an allele, genotype, haplotype, or environmental factor; and age can be age at onset, age when subject was last known to be unaffected, or duration of exposure. Signature patterns that emerge can help distinguish true disease associations from spurious associations due to age effects, age-varying associations from associations that are uniform across all ages, and associations with risk from associations with age-at-onset. Utility of MAP is illustrated by application to genetic and epidemiological association data for Alzheimer's and Parkinson's disease. MAP is intended as a descriptive method, to complement standard statistical techniques. Although originally developed for age patterns, MAP is equally useful for visualizing any quantitative trait.
- Published
- 2009
27. Validity and utility of a LRRK2 G2019S mutation test for the diagnosis of Parkinson's disease
- Author
-
Berta C. Leis, Donald S. Higgins, Sean Philpott, Cyrus P. Zabetian, Haydeh Payami, Jennifer S. Montimurro, Alida Griffith, Thomas D. Bird, Ali Samii, Stewart A. Factor, John W. Roberts, Denise M. Kay, and John G. Nutt
- Subjects
Adult ,Male ,medicine.medical_specialty ,Parkinson's disease ,Adolescent ,Disease ,Test validity ,Protein Serine-Threonine Kinases ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Likelihood ratios in diagnostic testing ,Sensitivity and Specificity ,Internal medicine ,medicine ,TaqMan ,Humans ,Genetic Testing ,Family history ,Genetics (clinical) ,Genetic testing ,Aged ,Aged, 80 and over ,medicine.diagnostic_test ,business.industry ,Parkinson Disease ,Middle Aged ,medicine.disease ,LRRK2 ,Amino Acid Substitution ,Physical therapy ,Female ,business - Abstract
The G2019S mutation in the LRRK2 gene, the most common known cause of Parkinson's disease (PD), will soon be widely available as a molecular clinical test for PD. The objective of this study was to assess performance characteristics of G2019S as a clinical test for PD in the setting of typical movement disorder clinics in the United States. Subjects included 1,518 sequentially recruited PD patients from seven movement disorder clinics in the United States, and 1,733 unaffected subjects. All 3,251 subjects were genotyped for the G2019S mutation using a TaqMan assay, and mutations were verified by direct sequencing. Test validity estimates were calculated using standard methods. A total of 20/1518 patients and 1/1733 controls carried the G2019S mutation. Specificity was 99.9% (95% CI, 99.6-100%), sensitivity was 1.3% (0.8-2.1%), and the positive likelihood ratio was 22.8. A positive family history of PD increased the positive likelihood ratio to 82.5. Information on gender, age at disease onset, or age at testing did not improve test performance. The gene test was highly accurate in classifying mutation carriers as PD, but it performed poorly in predicting the phenotype of non-mutation carriers. A G2019S molecular test for PD would be highly specific, technically simple, and inexpensive. Test interpretation is straightforward when used for diagnosis of symptomatic individuals, but is more complex for risk assessment and predictive testing in asymptomatic individuals. Test results can have psychological, social, and economical ramifications; thus, proper counseling is essential.
- Published
- 2006
28. LRRK2 G2019S in families with Parkinson disease who originated from Europe and the Middle East: evidence of two distinct founding events beginning two millennia ago
- Author
-
Thomas D. Bird, Carolyn M. Hutter, Alida Griffith, Alexis N. Lopez, Karen L. Edwards, Denise M. Kay, Dora Yearout, Berta C. Leis, Donald S. Higgins, John W. Roberts, Haydeh Payami, Ali Samii, Stewart A. Factor, Cyrus P. Zabetian, and John G. Nutt
- Subjects
Male ,Population genetics ,Protein Serine-Threonine Kinases ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Polymorphism, Single Nucleotide ,White People ,03 medical and health sciences ,Middle East ,0302 clinical medicine ,Africa, Northern ,Report ,Genetics ,Humans ,Genetics(clinical) ,Family ,Genotyping ,Genetics (clinical) ,030304 developmental biology ,Ancestor ,0303 health sciences ,Haplotype ,Parkinson Disease ,LRRK2 ,Ashkenazi jews ,Genealogy ,3. Good health ,Geography ,Amino Acid Substitution ,Haplotypes ,Case-Control Studies ,Jews ,Microsatellite ,Female ,030217 neurology & neurosurgery ,Software ,Founder effect - Abstract
The leucine-rich repeat kinase 2 (LRRK2) G2019S mutation is the most common genetic determinant of Parkinson disease (PD) identified to date. It accounts for 1%–7% of PD in patients of European origin and 20%–40% in Ashkenazi Jews and North African Arabs with PD. Previous studies concluded that patients from these populations all shared a common Middle Eastern founder who lived in the 13th century. We tested this hypothesis by genotyping 25 microsatellite and single-nucleotide–polymorphism markers in 22 families with G2019S and observed two distinct haplotypes. Haplotype 1 was present in 19 families of Ashkenazi Jewish and European ancestry, whereas haplotype 2 occurred in three European American families. Using a maximum-likelihood method, we estimated that the families with haplotype 1 shared a common ancestor 2,250 (95% confidence interval 1,650–3,120) years ago, whereas those with haplotype 2 appeared to share a more recent founder. Our data suggest two separate founding events for G2019S in these populations, beginning at a time that coincides with the Jewish Diasporas.
- Published
- 2006
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.