66 results on '"Jon G. Seidman"'
Search Results
2. LAMP2 Cardiomyopathy: Consequences of Impaired Autophagy in the Heart
- Author
-
Ronny Alcalai, Michael Arad, Hiroko Wakimoto, Dor Yadin, Joshua Gorham, Libin Wang, Elia Burns, Barry J. Maron, William C. Roberts, Tetsuo Konno, David A. Conner, Antonio R. Perez‐Atayde, Jon G. Seidman, and Christine E. Seidman
- Subjects
autophagy ,calcium transients ,cardiomyopathy ,Danon disease ,mouse model ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Background Human mutations in the X‐linked lysosome‐associated membrane protein‐2 (LAMP2) gene can cause a multisystem Danon disease or a primary cardiomyopathy characterized by massive hypertrophy, conduction system abnormalities, and malignant ventricular arrhythmias. We introduced an in‐frame LAMP2 gene exon 6 deletion mutation (denoted L2Δ6) causing human cardiomyopathy, into mouse LAMP2 gene, to elucidate its consequences on cardiomyocyte biology. This mutation results in in‐frame deletion of 41 amino acids, compatible with presence of some defective LAMP2 protein. Methods and Results Left ventricular tissues from L2Δ6 and wild‐type mice had equivalent amounts of LAMP2 RNA, but a significantly lower level of LAMP2 protein. By 20 weeks of age male mutant mice developed left ventricular hypertrophy which was followed by left ventricular dilatation and reduced systolic function. Cardiac electrophysiology and isolated cardiomyocyte studies demonstrated ventricular arrhythmia, conduction disturbances, abnormal calcium transients and increased sensitivity to catecholamines. Myocardial fibrosis was strikingly increased in 40‐week‐old L2Δ6 mice, recapitulating findings of human LAMP2 cardiomyopathy. Immunofluorescence and transmission electron microscopy identified mislocalization of lysosomes and accumulation of autophagosomes between sarcomeres, causing profound morphological changes disrupting the cellular ultrastructure. Transcription profile and protein expression analyses of L2Δ6 hearts showed significantly increased expression of genes encoding activators and protein components of autophagy, hypertrophy, and apoptosis. Conclusions We suggest that impaired autophagy results in cardiac hypertrophy and profound transcriptional reactions that impacted metabolism, calcium homeostasis, and cell survival. These responses define the molecular pathways that underlie the pathology and aberrant electrophysiology in cardiomyopathy of Danon disease.
- Published
- 2021
- Full Text
- View/download PDF
3. Joint analysis of left ventricular expression and circulating plasma levels of Omentin after myocardial ischemia
- Author
-
Louis A. Saddic, Sarah M. Nicoloro, Olga T. Gupta, Michael P. Czech, Joshua Gorham, Stanton K. Shernan, Christine E. Seidman, Jon G. Seidman, Sary F. Aranki, Simon C. Body, Timothy P. Fitzgibbons, and Jochen D. Muehlschlegel
- Subjects
Ischemia ,Adipokine ,Omentin ,RNA-seq ,Cardiovascular ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Abstract Background Omentin-1, also known as Intelectin-1 (ITLN1), is an adipokine with plasma levels associated with diabetes, obesity, and coronary artery disease. Recent studies suggest that ITLN1 can mitigate myocardial ischemic injury but the expression of ITLN1 in the heart itself has not been well characterized. The purpose of this study is to discern the relationship between the expression pattern of ITLN1 RNA in the human heart and the level of circulating ITLN1 protein in plasma from the same patients following myocardial ischemia. Methods A large cohort of patients (n = 140) undergoing elective cardiac surgery for aortic valve replacement were enrolled in this study. Plasma and left ventricular biopsy samples were taken at the beginning of cardiopulmonary bypass and after an average of 82 min of ischemic cross clamp time. The localization of ITLN1 in epicardial adipose tissue (EAT) was also further characterized with immunoassays and cell fate transition studies. Results mRNA expression of ITLN1 decreases in left ventricular tissue after acute ischemia in human patients (mean difference 280.48, p = 0.001) whereas plasma protein levels of ITLN1 increase (mean difference 5.24, p
- Published
- 2017
- Full Text
- View/download PDF
4. Contribution of Noncanonical Splice Variants toTTNTruncating Variant Cardiomyopathy
- Author
-
Barbara McDonough, Alireza Haghighi, Joshua M. Gorham, Christine E. Seidman, Min Young Jang, Diane Fatkin, Neal K. Lakdawala, Parth N Patel, Amy E. Roberts, Kaoru Ito, Lien Lam, Steven R. DePalma, Jon G. Seidman, Renee Johnson, Stuart A. Cook, Jon A. L. Willcox, Paul J.R. Barton, Guys & St Thomas NHS Foundation Trust, Imperial College Healthcare NHS Trust- BRC Funding, and British Heart Foundation
- Subjects
Genetics ,intron ,Cardiomyopathy ,Intron ,heart failure ,General Medicine ,Biology ,medicine.disease ,Exon ,Heart failure ,RNA splicing ,Idiopathic dilated cardiomyopathy ,myocardium ,medicine ,splice ,exon ,Invariant (mathematics) ,cardiomyopathy - Abstract
Background:HeterozygousTTNtruncating variants cause 10% to 20% of idiopathic dilated cardiomyopathy (DCM). Although variants which disrupt canonical splice signals (ie, invariant dinucleotide of the splice donor site, invariant dinucleotide of the splice acceptor site) at exon-intron junctions are readily recognized asTTNtruncating variants, the effects of other nearby sequence variations on splicing and their contribution to disease is uncertain.Methods:Rare variants of unknown significance located in the splice regions of highly expressedTTNexons from 203 DCM cases, 3329 normal subjects, and clinical variant databases were identified. The effects of these variants on splicing were assessed using an in vitro splice assay.Results:Splice-altering variants of unknown significance were enriched in DCM cases over controls and present in 2% of DCM patients (P=0.002). Application of this method to clinical variant databases demonstrated 20% of similar variants of unknown significance inTTNsplice regions affect splicing. Noncanonical splice-altering variants were most frequently located at position +5 of the donor site (P=4.4×107) and position -3 of the acceptor site (P=0.002). SpliceAI, an emerging in silico prediction tool, had a high positive predictive value (86%–95%) but poor sensitivity (15%–50%) for the detection of splice-altering variants. Alternate exons spliced out of mostTTNtranscripts frequently lacked the consensus base at +5 donor and −3 acceptor positions.Conclusions:Noncanonical splice-altering variants inTTNexplain 1-2% of DCM and offer a 10-20% increase in the diagnostic power ofTTNsequencing in this disease. These data suggest rules that may improve efforts to detect splice-altering variants in other genes and may explain the low percent splicing observed for many alternateTTNexons.
- Published
- 2021
- Full Text
- View/download PDF
5. Direct Reprogramming of Non-limb Fibroblasts to Cells with Properties of Limb Progenitors
- Author
-
Joshua M. Gorham, Christine E. Seidman, Vannier J, Charlotte Colle, Patrick Tschopp, Olivier Pourquié, Chao-Zong Lee, Yuji Atsuta, Clifford J. Tabin, Jon G. Seidman, Alan R. Rodrigues, Reiko R. Tomizawa, and Lujan Eg
- Subjects
body regions ,Cell type ,Limb bud ,Lateral plate mesoderm ,Mesenchymal stem cell ,Context (language use) ,Biology ,Progenitor cell ,LIN28 ,Reprogramming ,Cell biology - Abstract
SUMMARYThe early limb bud consists of mesenchymal progenitors (limb progenitors) derived from the lateral plate mesoderm (LPM) that produce most of the tissues of the mature limb bud. The LPM also gives rise to the mesodermal components of the trunk, flank and neck. However, the mesenchymal cells generated at these other axial levels cannot produce the variety of cell types found in the limb bud, nor can they be directed to form a patterned appendage-like structure, even when placed in the context of the signals responsible for organizing the limb bud. Here, by taking advantage of a direct reprogramming approach, we find a set of factors (Prdm16, Zbtb16, and Lin28) normally expressed in the early limb bud, that are capable of imparting limb progenitor-like properties to non-limb fibroblasts. Cells reprogrammed by these factors show similar gene expression profiles, and can differentiate into similar cell types, as endogenous limb progenitors. The further addition of Lin41 potentiates proliferation of the reprogrammed cells while suppressing differentiation. These results suggest that these same four key factors may play pivotal roles in the specification of endogenous limb progenitors.
- Published
- 2021
- Full Text
- View/download PDF
6. In Memorium, Philip Leder (1934–2020)
- Author
-
Shirley M. Tilghman and Jon G. Seidman
- Subjects
Art history ,Cell Biology ,Biology ,Molecular Biology ,Developmental Biology - Published
- 2020
- Full Text
- View/download PDF
7. Cells and gene expression programs in the adult human heart
- Author
-
Joshua M. Gorham, Christine E. Seidman, Krishnaa T. Mahbubani, Jon G. Seidman, Krzysztof Polanski, Eric L. Lindberg, Daniel M. DeLaughter, Kenny Roberts, Hongbo Zhang, Eirini S. Fasouli, Emily R. Nadelmann, Catherine L. Worth, Michela Noseda, Daniel Reichart, Norbert Hubner, Hiroko Wakimoto, Barbara McDonough, Matthias Heinig, Giannino Patone, Gavin Y. Oudit, Liz Tuck, Sarah A. Teichmann, Omer Ali Bayraktar, Carlos Talavera-López, Monika Litviňuková, Anissa Viveiros, Kourosh Saeb-Parsy, Hao Zhang, Henrike Maatz, Sara Samari, Masatoshi Kanda, and Joseph J. Boyle
- Subjects
education.field_of_study ,government.form_of_government ,Cell ,Population ,Skeletal muscle ,Biology ,Phenotype ,Transcriptome ,Lymphatic Endothelium ,medicine.anatomical_structure ,Cardiovascular and Metabolic Diseases ,medicine ,government ,cardiovascular system ,Interventricular septum ,education ,Neuroscience ,Homeostasis - Abstract
SummaryCardiovascular disease is the leading cause of death worldwide. Advanced insights into disease mechanisms and strategies to improve therapeutic opportunities require deeper understanding of the molecular processes of the normal heart. Knowledge of the full repertoire of cardiac cells and their gene expression profiles is a fundamental first step in this endeavor. Here, using large-scale single cell and nuclei transcriptomic profiling together with state-of-the-art analytical techniques, we characterise the adult human heart cellular landscape covering six anatomical cardiac regions (left and right atria and ventricles, apex and interventricular septum). Our results highlight the cellular heterogeneity of cardiomyocytes, pericytes and fibroblasts, revealing distinct subsets in the atria and ventricles indicative of diverse developmental origins and specialized properties. Further we define the complexity of the cardiac vascular network which includes clusters of arterial, capillary, venous, lymphatic endothelial cells and an atrial-enriched population. By comparing cardiac cells to skeletal muscle and kidney, we identify cardiac tissue resident macrophage subsets with transcriptional signatures indicative of both inflammatory and reparative phenotypes. Further, inference of cell-cell interactions highlight a macrophage-fibroblast-cardiomyocyte network that differs between atria and ventricles, and compared to skeletal muscle. We expect this reference human cardiac cell atlas to advance mechanistic studies of heart homeostasis and disease.
- Published
- 2020
8. Whole Genome De Novo Variant Identification with FreeBayes and Neural Network Approaches
- Author
-
Felix Richter, Jason Homsy, Hongjian Qi, Jiayao Wang, Christine E. Seidman, Steve Depalma, Yufeng Shen, Kitaygorodsky A, Sarah U. Morton, Nihir Patel, Jon G. Seidman, and Bruce D. Gelb
- Subjects
Sanger sequencing ,Source code ,Artificial neural network ,Computer science ,media_common.quotation_subject ,Computational biology ,Integrative genomics ,Genome ,Pipeline (software) ,Identification (information) ,symbols.namesake ,symbols ,Indel ,media_common - Abstract
MotivationDe novo variant (DNV) calling typically relies on heuristic filters intrinsic to specific platforms and variant calling algorithms. FreeBayes and neural network approaches have overcome this limitation for variant calling, and we implemented a similar approach for DNV identification.ResultsWe developed a DNV calling framework that uses Genome Analysis Toolkit (GATK), FreeBayes and a neural network trained on Integrative Genomics Viewer pile-up plots (IGV-bot). We identified DNVs in 2,390 WGS trios and benchmarked results against heuristics based on GATK parameters. Results were validated in silico and with Sanger sequencing, with the latter showing true positive rates of 98.4% and 97.3% for SNVs and indels, respectively. Taken together we describe a scalable framework for DNV identification based on both FreeBayes and neural network methods.AvailabilitySource code and documentation are available at https://github.com/ShenLab/igv-classifier and https://github.com/frichter/dnv_pipeline under the MIT license.Contactys2411@cumc.columbia.edu
- Published
- 2020
- Full Text
- View/download PDF
9. Early post-zygotic mutations contribute to congenital heart disease
- Author
-
Joshua M. Gorham, Christine E. Seidman, Steve Depalma, Elizabeth Goldmuntz, Bruce D. Gelb, Kathryn B. Manheimer, Alexander Hsieh, Richard P. Lifton, Yufeng Shen, Jane W. Newburger, Sarah U. Morton, Wendy K. Chung, Deepak Srivastava, Emily Leann Griffin, George A. Porter, Richard W. Kim, Hongjian Qi, Daniel Bernstein, Martina Brueckner, Jon G. Seidman, Angela Tai, Martin Tristani-Firouzi, David M. McKean, and Jon A. L. Willcox
- Subjects
Genetics ,Proband ,0303 health sciences ,Zygote ,Heart disease ,Somatic cell ,Biology ,medicine.disease ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Tissue mosaicism ,medicine ,Allele ,Exome ,Gene ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
BackgroundThe contribution of somatic mosaicism, or genetic mutations arising after oocyte fertilization, to congenital heart disease (CHD) is not well understood. Further, the relationship between mosaicism in blood and cardiovascular tissue has not been determined.ResultsWe developed a computational method, Expectation-Maximization-based detection of Mosaicism (EM-mosaic), to analyze mosaicism in exome sequences of 2530 CHD proband-parent trios. EM-mosaic detected 326 mosaic mutations in blood and/or cardiac tissue DNA. Of the 309 detected in blood DNA, 85/97 (88%) tested were independently confirmed, while 7/17 (41%) candidates of 17 detected in cardiac tissue were confirmed. MosaicHunter detected an additional 64 mosaics, of which 23/46 (50%) among 58 candidates from blood and 4/6 (67%) of 6 candidates from cardiac tissue confirmed. Twenty-five mosaic variants altered CHD-risk genes, affecting 1% of our cohort. Of these 25, 22/22 candidates tested were confirmed. Variants predicted as damaging had higher variant allele fraction than benign variants, suggesting a role in CHD. The frequency of mosaic variants above 10% mosaicism was 0.13/person in blood and 0.14/person in cardiac tissue. Analysis of 66 individuals with matched cardiac tissue available revealed both tissue-specific and shared mosaicism, with shared mosaics generally having higher allele fraction.ConclusionsWe estimate that ~1% of CHD probands have a mosaic variant detectable in blood that could contribute to cardiac malformations, particularly those damaging variants expressed at higher allele fraction compared to benign variants. Although blood is a readily-available DNA source, cardiac tissues analyzed contributed ~5% of somatic mosaic variants identified, indicating the value of tissue mosaicism analyses.
- Published
- 2019
- Full Text
- View/download PDF
10. Abstract 467: Identification of Novel Pathogenic Mutations in Non-Canonical RNA Splice Sites in Congenital Heart Disease
- Author
-
Kaoru Ito, Angela C. Tai, Min Young Jang, Alexandre C. Pereira, Jon G. Seidman, David M. McKean, Joshua M. Gorham, Christine E. Seidman, and Parth N Patel
- Subjects
Genetics ,Heart disease ,Physiology ,media_common.quotation_subject ,Nonsense ,Gene mutation ,Biology ,medicine.disease ,Frameshift mutation ,RNA Splice Sites ,Non canonical ,medicine ,Identification (biology) ,Cardiology and Cardiovascular Medicine ,media_common - Abstract
Rationale: Congenital heart disease (CHD) occurs in about 1 in 100 live births, yet known genetic causes explain less than 20% of CHD cases. While variants that cause frameshift, nonsense, start/stop site gain or loss, and canonical splice site alterations are readily categorized as being pathogenic or loss-of-function (LOF), interpreting the clinical significance of variants without obvious functional consequences remains challenging. Here, we aim to improve classification of variants of unknown significance (VUS) in non-canonical RNA splice sites that may be pathogenic for CHD. Methods: We tested candidate LOF de novo (DNV) and rare (p < 2E-5) inherited variants from whole exome sequencing of 4474 CHD probands and their parents in the NHLBI Pediatric Cardiac Genetics Consortium. Briefly, variants underwent computational selection to prioritize VUS in splice regions that are likely to alter splicing (“high-likelihood VUS”). These variants then underwent in vitro analysis including Minigene construction, transfection, RNA isolation, and sequencing to confirm splicing outcomes. Results: Preliminary results limited to DNV variants showed that 163 of 2678 (6.08%) were high-likelihood VUS. Subsequent analysis in vitro assay of high-likelihood VUS yielded 53 as splice-altering (p < 0.05) and thus LOF. Combined with previously identified 366 DNV LOF variants, the addition of these splice-altering variants represents a 15.3% increase in total LOF DNV variant calls. This includes new pathogenic mutations in known CHD genes such as KMT2D . In one case, a CHD proband with features of Kabuki Syndrome without a definitive diagnosis was found to have a splice-altering variant in KMT2D . We have extended this assay to 34518 rare, inherited variants in the same cohort, of which 868 (2.54%) are in genes previously associated with CHD. Conclusion: Consideration of non-canonical RNA splice sites in this assay increased the yield of LOF mutations from traditional sequencing methods by 15.3% in the CHD cohort. Further analysis of splice-altering variants in both known and unknown pathogenic genes will improve diagnostic classification of VUS and gene-based diagnosis of CHD.
- Published
- 2019
- Full Text
- View/download PDF
11. Abstract 785: Modeling Congenital Heart Disease-Associated Variants in GATA6 Using CRISPR/Cas9 and Human Induced Pluripotent Stem Cells
- Author
-
Seongwon Kim, Joshua M. Gorham, Christopher N. Toepfer, David A. Conner, Jon A. L. Willcox, Lauren K. Wasson, Christine E. Seidman, Megan Jang, Daniel M. DeLaughter, Tarsha Ward, Steven R. DePalma, Alexandre C. Pereira, Angela Tai, Manuel Schmid, Sarah U. Morton, Meraj Neyazi, Arun Sharma, Yuri Kim, Benoit G. Bruneau, Jon G. Seidman, and Radhika Agarwal
- Subjects
Genetics ,endocrine system ,GATA6 ,Heart disease ,Physiology ,Biology ,Gene mutation ,medicine.disease ,medicine ,CRISPR ,Identification (biology) ,Human Induced Pluripotent Stem Cells ,Cardiology and Cardiovascular Medicine ,Stem cell biology ,Exome sequencing - Abstract
The discovery of damaging gene mutations in congenital heart disease (CHD) patients enables identification of regulators of cardiac development. Exome sequencing identified de novo heterozygous loss-of-function (LoF) and missense variants in GATA6 among CHD probands, most with outflow tract malformations. Other subjects with GATA6 LoF mutations developed pancreatic agenesis. To elucidate the molecular basis for the predominance of this heart defect, we modeled GATA6 mutations in cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs). GATA6 variants were introduced into isogenic hiPSCs using CRISPR/Cas9 genome editing. Genome-wide molecular profiles including chromatin accessibility (ATAC-Seq) and gene expression (single cell and bulk RNA-Seq) were evaluated during hiPSC-CM differentiation. Analyses of GATA6 mutant hiPSC-CMs showed deficits in hiPSC-CM differentiation, chromatin accessibility and transcriptional profiles. Heterozygous GATA6 LoF hiPSCs made hiPSC-CMs but exhibited reduced expression of second heart field genes. Homozygous GATA6 LoF hiPSCs failed to differentiate and adopted fibroblast expression profiles. hiPSCs carrying a homozygous GATA6 missense variant, R456G, which altered a DNA-binding domain residue, showed enhanced capacity to differentiate into neuroepithelial-like cells. Chromatin-accessibility studies confirmed that GATA6 normally binds to genes in the promoter region and other genes at distal enhancers. Human GATA6 haploinsufficiency disrupts developmental transcriptional responses driving cardiac morphogenesis. The HAND2 -dependent genetic program, operant during outflow tract development, is particularly sensitive to GATA6 dosage. The mixed differentiation patterns observed in mutation-carrying hiPSCs likely contributes to vascular phenotypes observed in CHD patients. GATA6 haploinsufficiency preferentially alters binding of distal enhancers to promoters in genes where GATA6 normally binds the enhancer rather than the promoter. We speculate that pathogenicity of GATA6 haploinsufficiency is mediated by weaker binding of GATA6 to distal enhancers than to promoter elements, altering expression of these genes in GATA6 haploinsufficient patients.
- Published
- 2019
- Full Text
- View/download PDF
12. Abstract 210: Transcriptomic Changes During Induced Pluripotent Stem Cell-derived Neural Crest Cell Differentiation Highlight Genes Involved in Endocardial Cushion and Cardiac Outflow Tract Development
- Author
-
Joshua M. Gorham, Christine E. Seidman, Tarsha Ward, Arun Sharma, Min Young Jang, Alexandre C. Pereira, Jon G. Seidman, Radhika Agarwal, David M. McKean, Daniel Reichart, and Daniel M. DeLaughter
- Subjects
Transcriptome ,Neural crest cell differentiation ,Heart disease ,Physiology ,medicine ,Neural crest ,Biology ,Cardiology and Cardiovascular Medicine ,medicine.disease ,Induced pluripotent stem cell ,Atrioventricular cushions ,Gene ,Cell biology - Abstract
Rationale: Neural crest cells (NCCs) play a critical role in normal cardiac development, and defects in NCCs likely cause congenital heart disease (CHD). NCCs are transient and multipotent migratory stem cells that give rise to diverse tissues, including cardiac structures such as the smooth muscles of the great arteries and semilunar valves. Induced pluripotent stem cells (iPSC) can be differentiated into NCCs, as demonstrated by expression of several marker proteins including NGFR and HNK1. To better define iPSC-NCCs and to better understand the progression of iPSCs to NCCs, we have compared the transcriptomes of iPSC and iPSC-NCCs. We have also begun to investigate the consequences of loss-of-function mutations in genes implicated in CHD on the differentiation of iPSC-NCCs. Methods and Results: PGP1 iPSCs were differentiated to NCCs and RNA was collected at 0, 5, 10, and 15 days of differentiation. RNAseq analysis showed that by day 15, 6483 genes were upregulated in NCC vs iPSCs, 6406 downregulated, and 6715 unchanged (FDR 5%). Enrichment analysis for the top 500 upregulated genes showed 4 cardiac gene ontology (GO) terms in the top 10, including ‘endocardial cushion morphogenesis’ (fold enrichment 11.85, p = 0.012). Notably, of 45 genes under GO term ‘endocardial cushion development’, 38 (84%) differentially expressed in NCCs by day 15 (FDR 5%). Next, we compared this RNAseq data with that of iPSC-derived cardiomyocyte (CM) differentiation in a subset of 253 genes previously implicated in CHD. Of these, 143 genes were differentially expressed in both iPSC-CMs and iPSC-NCCs. However, 27 genes (10.6%) including MYH6, PITX2, and TBX5 were uniquely upregulated during CM differentiation, while 65 genes (25.7%) including CHD4 and NOTCH1 were uniquely upregulated during NCC differentiation. Conclusion: Transcriptomic changes during iPSC-NCC differentiation, assessed by both bulk and single cell RNAseq, reveal an upregulation of genes involved in cardiac development, particularly endocardial cushions and the outflow tract. Importantly, a subset of genes implicated in CHD are altered during iPSC-NCC differentiation but not in iPSC-CM differentiation. Thus, iPSC-NCCs offer a new model with which to investigate the pathogenesis and mechanisms of CHD.
- Published
- 2019
- Full Text
- View/download PDF
13. Author Correction: poly(UG)-tailed RNAs in genome protection and epigenetic inheritance
- Author
-
Scott Kennedy, Daniel J. Pagano, Marvin Wickens, Jon G. Seidman, Yuhan Fei, Anne E. Dodson, Jenny Jiacheng Yan, Aditi Shukla, and Josh Gorham
- Subjects
Small interfering RNA ,Multidisciplinary ,RNA interference ,Posttranslational modification ,Gene silencing ,Epigenetics ,Computational biology ,Biology ,Genome - Published
- 2021
- Full Text
- View/download PDF
14. P5090Sortilin is a key driver of fibrocalcific aortic valve disease
- Author
-
Farouc A. Jaffer, Jon G. Seidman, Elena Aikawa, Joshua D. Hutcheson, Florian Schlotter, Shinya Goto, W D Merryman, Masanori Aikawa, Sasha A Singh, Lang Lee, Mark C. Blaser, Maximillian A. Rogers, Daniel M. DeLaughter, Simon C. Body, and Claudia Goettsch
- Subjects
Aortic valve disease ,medicine.medical_specialty ,business.industry ,Internal medicine ,Key (cryptography) ,Cardiology ,Medicine ,Cardiology and Cardiovascular Medicine ,business - Published
- 2018
- Full Text
- View/download PDF
15. Molecular Genetics of Lidocaine-containing Cardioplegia in the Human Heart during Cardiac Surgery
- Author
-
Sary F. Aranki, Jon G. Seidman, Mahyar Heydarpour, Maroun Yammine, Josh Gorham, Michael S. Gilfeather, Simon C. Body, Gregory Stone, Julius I. Ejiofor, Christine E. Seidman, and Jochen D. Muehlschlegel
- Subjects
Pulmonary and Respiratory Medicine ,Male ,medicine.medical_specialty ,Lidocaine ,030204 cardiovascular system & hematology ,Pharmacology ,Risk Assessment ,Article ,law.invention ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,law ,Reference Values ,Gene expression ,Cardiopulmonary bypass ,Medicine ,Humans ,Viability assay ,Cardiac Surgical Procedures ,Neutrophil aggregation ,Cardioplegic Solutions ,Molecular Biology ,Aged ,Retrospective Studies ,chemistry.chemical_classification ,Aged, 80 and over ,Heart Valve Prosthesis Implantation ,Academic Medical Centers ,Cardiopulmonary Bypass ,business.industry ,Oxygen transport ,Age Factors ,Middle Aged ,Cardiac surgery ,Treatment Outcome ,030228 respiratory system ,chemistry ,Gene Expression Regulation ,Transferrin ,Aortic Valve ,Heart Arrest, Induced ,Surgery ,Cardiology and Cardiovascular Medicine ,business ,medicine.drug - Abstract
Background During cardiac surgery with cardiopulmonary bypass, delivery of cardioplegia solution to achieve electromechanical cardiac quiescence is obligatory. The addition of lidocaine to cardioplegia has advantages, although its consequences at a molecular level remain unclear. We performed whole-genome RNA sequencing of the human left ventricular (LV) myocardium to elucidate the differences between whole-blood (WB) cardioplegia with and without addition of lidocaine (LC) on gene expression. Methods We prospectively enrolled 130 patients undergoing aortic valve replacement surgery. Patients received high-potassium blood cardioplegia either with (n = 37) or without (n = 93) lidocaine. The LV apex was biopsied at baseline, and after an average of 74 minutes of cold cardioplegic arrest. We performed differential gene expression analysis for 18,258 genes between these 2 groups. Clinical and demographic variables were adjusted in the model. Gene ontology (GO) and network enrichment analysis of the retained genes were performed using g:Profiler and Cytoscape. Results A total of 1,298 genes were differentially expressed between cardioplegic treatments. Compared with the WB group, genes upregulated in the LC group were identified by network enrichment to play a protective role in ischemic injury by inhibiting apoptosis, increasing transferrin endocytosis, and increasing cell viability. Downregulated genes in the LC group were identified to play a role in inflammatory diseases, oxygen transport, and neutrophil aggregation. Conclusions The addition of lidocaine to cardioplegia had pronounced effects on a molecular level with genes responsible for decreased inflammation, reduced intracellular calcium binding, enhanced antiapoptotic protection, augmented oxygen accessibility through transferrins, and increased cell viability showing measurable differences.
- Published
- 2018
16. Sex differences in gene expression in response to ischemia in the human myocardium
- Author
-
Joshua M. Gorham, Gregory Stone, Christine E. Seidman, Ashley Choi, Vincent J. Carey, Mahyar Heydarpour, Barbara E. Stranger, Sary F. Aranki, Jochen D. Muehlschlegel, Benjamin A. Raby, Meritxell Oliva, Simon C. Body, and Jon G. Seidman
- Subjects
0303 health sciences ,business.industry ,Ischemia ,Physiology ,Infarction ,030204 cardiovascular system & hematology ,medicine.disease ,Sexual dimorphism ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Ventricle ,Heart failure ,Expression quantitative trait loci ,medicine ,Myocardial infarction ,Risk factor ,business ,030304 developmental biology - Abstract
and KeywordsBackgroundSex differences exist in the prevalence, presentation, and outcomes of ischemic heart disease. Females have higher risk of heart failure post myocardial infarction relative to males and the female sex is an independent risk factor for hospital and operative mortality after cardiac surgery. However, the mechanisms underlying this sexual dimorphism remain unclear. We examined sex differences in human myocardial gene expression in response to ischemia.MethodsLeft ventricular biopsies from 68 male and 46 female patients undergoing aortic valve replacement surgery were obtained at baseline and after a median 74 minutes of cold cardioplegic arrest/ischemia and respective transcriptomes were quantified by RNA-Seq. Sex-specific responses to ischemia were quantified by differential gene expression, expression quantitative trait loci (eQTL) and pathway and functional analysis. Cell-type enrichment analysis. was used to obtain an estimate of the identity and relative proportions of different cell types present in each sample.ResultsA sex-specific response to ischemia was observed for 271 genes. Functional annotation analysis revealed sex-specific modulation of the oxytocin signaling pathway and common pathway of fibrin clot formation. The eQTL analysis identified variant-by-sex interaction eQTLs at baseline and post-ischemia, indicative of sex differences in the genotypic effects on gene expression, and cell-type enrichment analysis showed sex-bias in proportion of specific cell types.ConclusionIn response to myocardial ischemia, the human left ventricle demonstrates changes in gene expression that differ between the sexes. These differences provide insight into the sexual dimorphism of ischemic heart disease and may aid in the development of sex-specific therapies that reduce myocardial injury.
- Published
- 2018
- Full Text
- View/download PDF
17. Using Next-generation RNA Sequencing to Examine Ischemic Changes Induced by Cold Blood Cardioplegia on the Human Left Ventricular Myocardium Transcriptome
- Author
-
David M. McKean, Steven R. DePalma, Joshua M. Gorham, Christine E. Seidman, Mahyar Heydarpour, Erica Mazaika, Stanton K. Shernan, Danos C. Christodoulou, Sary F. Aranki, Simon C. Body, Jon G. Seidman, Tjorvi E. Perry, Amanda A. Fox, Grace M. Lee, and Jochen D. Muehlschlegel
- Subjects
Male ,medicine.medical_specialty ,Myocardial ischemia ,Heart Ventricles ,Myocardial Ischemia ,Ischemia ,Article ,law.invention ,Transcriptome ,Downregulation and upregulation ,law ,Internal medicine ,Cardiopulmonary bypass ,Humans ,Medicine ,Blood cardioplegia ,Aged ,Aged, 80 and over ,Sequence Analysis, RNA ,business.industry ,Myocardium ,RNA ,medicine.disease ,Cold Temperature ,Anesthesiology and Pain Medicine ,Heart Arrest, Induced ,Left ventricular myocardium ,Cardiology ,Female ,business - Abstract
Background: The exact mechanisms that underlie the pathological processes of myocardial ischemia in humans are unclear. Cardiopulmonary bypass with cardioplegic arrest allows the authors to examine the whole transcriptional profile of human left ventricular myocardium at baseline and after exposure to cold cardioplegia-induced ischemia as a human ischemia model. Methods: The authors obtained biopsies from 45 patients undergoing aortic valve replacement surgery at baseline and after an average of 79 min of cold cardioplegic arrest. Samples were RNA sequenced and analyzed with the Partek® Genomics Suite (Partek Inc., St. Louis, MO) for differential expression. Ingenuity Pathway Analysis (Ingenuity Systems, Redwood City, CA) and Biobase ExPlain (Biobase GmbH, Wolfenbuettel, Germany) systems were used for functional and pathway analyses. Results: Of the 4,098 genes with a mean expression value greater than 5, 90% were down-regulated and 9.1% were up-regulated. Of those, 1,241 were significantly differentially expressed. Gene ontology analysis revealed significant down-regulation in immune inflammatory response and complement activation categories and highly consistent was the down-regulation of intelectin 1, proteoglycan, and secretory leukocyte peptidase inhibitor. Up-regulated genes of interest were FBJ murine osteosarcoma viral oncogene homolog and the hemoglobin genes hemoglobin α1 (HBA1) and hemoglobin β. In addition, analysis of transcription factor–binding sites revealed interesting targets in factors regulating reactive oxygen species production, apoptosis, immunity, cytokine production, and inflammatory response. Conclusions: The authors have shown that the human left ventricle exhibits significant changes in gene expression in response to cold cardioplegia-induced ischemia during cardiopulmonary bypass, which provides great insight into the pathophysiology of ventricular ischemia, and thus, may help guide efforts to reduce myocardial damage during surgery.
- Published
- 2015
- Full Text
- View/download PDF
18. Pitx2 modulates a Tbx5 -dependent gene regulatory network to maintain atrial rhythm
- Author
-
Xinan Yang, Yun Qiao, Jenna Bekeny, Elizabeth M. McNally, Rangarajan D. Nadadur, Ivan P. Moskowitz, Igor R. Efimov, Tarsha Ward, Jon G. Seidman, Michael Broman, Stefan R Mazurek, Christopher R. Weber, Christine E. Seidman, Min Zhang, Margaret Gadek, Bastiaan J. Boukens, Vincent M. Christoffels, James F. Martin, Malou van den Boogaard, and Medical Biology
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Gene regulatory network ,Haploinsufficiency ,Biology ,Ryanodine receptor 2 ,Article ,Translational Research, Biomedical ,Mice ,03 medical and health sciences ,Heart Rate ,Internal medicine ,Atrial Fibrillation ,Gene expression ,medicine ,Transcriptional regulation ,Animals ,Humans ,Gene Regulatory Networks ,Genetic Predisposition to Disease ,Calcium Signaling ,Homeodomain Proteins ,Mice, Knockout ,PITX2 ,Effector ,General Medicine ,Atrial Function ,Myocardial Contraction ,Cell biology ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,T-Box Domain Proteins ,Transcription Factor Gene ,Genome-Wide Association Study ,Transcription Factors - Abstract
Cardiac rhythm is extremely robust, generating 2 billion contraction cycles during the average human life span. Transcriptional control of cardiac rhythm is poorly understood. We found that removal of the transcription factor gene Tbx5 from the adult mouse caused primary spontaneous and sustained atrial fibrillation (AF). Atrial cardiomyocytes from the Tbx5 -mutant mice exhibited action potential abnormalities, including spontaneous depolarizations, which were rescued by chelating free calcium. We identified a multitiered transcriptional network that linked seven previously defined AF risk loci: TBX5 directly activated PITX2, and TBX5 and PITX2 antagonistically regulated membrane effector genes Scn5a , Gja1 , Ryr2 , Dsp , and Atp2a2 . In addition, reduced Tbx5 dose by adult-specific haploinsufficiency caused decreased target gene expression, myocardial automaticity, and AF inducibility, which were all rescued by Pitx2 haploinsufficiency in mice. These results defined a transcriptional architecture for atrial rhythm control organized as an incoherent feed-forward loop, driven by TBX5 and modulated by PITX2. TBX5/PITX2 interplay provides tight control of atrial rhythm effector gene expression, and perturbation of the co-regulated network caused AF susceptibility. This work provides a model for the molecular mechanisms underpinning the genetic implication of multiple AF genome-wide association studies loci and will contribute to future efforts to stratify patients for AF risk by genotype.
- Published
- 2016
- Full Text
- View/download PDF
19. Allele-specific expression in the human heart and its application to postoperative atrial fibrillation and myocardial ischemia
- Author
-
Gregory S. Couper, Tzuu-Wang Chang, Martin I. Sigurdsson, Stanton K. Shernan, Jon G. Seidman, Simon C. Body, Sary F. Aranki, Mahyar Heydarpour, Prem Shekar, Jochen D. Muehlschlegel, and Louis A. Saddic
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,animal structures ,Genotype ,Genotyping Techniques ,Heart Ventricles ,Ischemia ,Myocardial Ischemia ,Single-nucleotide polymorphism ,030204 cardiovascular system & hematology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Internal medicine ,Atrial Fibrillation ,medicine ,Genetics ,SNP ,Animals ,Humans ,Genetics(clinical) ,Cardiac Surgical Procedures ,Molecular Biology ,Genetics (clinical) ,Alleles ,Whole blood ,Aged ,business.industry ,Research ,High-Throughput Nucleotide Sequencing ,Atrial fibrillation ,Middle Aged ,medicine.disease ,Pathophysiology ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Ventricle ,Ubiquitin ligase complex ,Cardiology ,Molecular Medicine ,Female ,business - Abstract
Background Allele-specific expression (ASE) is differential expression of each of the two chromosomal alleles of an autosomal gene. We assessed ASE patterns in the human left atrium (LA, n = 62) and paired samples from the left ventricle (LV, n = 76) before and after ischemia, and tested the utility of differential ASE to identify genes associated with postoperative atrial fibrillation (poAF) and myocardial ischemia. Methods Following genotyping from whole blood and whole-genome sequencing of LA and LV samples, we called ASE using sequences overlapping heterozygous SNPs using rigorous quality control to minimize false ASE calling. ASE patterns were compared between cardiac chambers and with a validation cohort from cadaveric tissue. ASE patterns in the LA were compared between patients who had poAF and those who did not. Changes in ASE in the LV were compared between paired baseline and post-ischemia samples. Results ASE was found for 3404 (5.1%) and 8642 (4.0%) of SNPs analyzed in the LA and LV, respectively. Out of 6157 SNPs with ASE analyzed in both chambers, 2078 had evidence of ASE in both LA and LV (p
- Published
- 2016
20. Genome-wide identification of mouse congenital heart disease loci
- Author
-
Kathryn E. Hentges, Karen L. Svenson, Bryan C. Bjork, Christine E. Seidman, Ivan P. Moskowitz, Jennifer L. Moran, Tharinda W. Rajapaksha, Michael A. Peterson, Anna Kamp, David R. Beier, Monica J. Justice, and Jon G. Seidman
- Subjects
Heart Defects, Congenital ,Male ,Candidate gene ,Heart disease ,Locus (genetics) ,Genome-wide association study ,Biology ,Mice ,Genetics ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,cardiovascular diseases ,Molecular Biology ,Gene ,Crosses, Genetic ,Genetics (clinical) ,Genetic testing ,medicine.diagnostic_test ,Myocardium ,Chromosome Mapping ,Articles ,General Medicine ,medicine.disease ,Phenotype ,Mice, Mutant Strains ,Mice, Inbred C57BL ,Disease Models, Animal ,Animals, Newborn ,Ethylnitrosourea ,Female ,Genome-Wide Association Study ,Genetic screen - Abstract
Empirical evidence supporting a genetic basis for the etiology of congenital heart disease (CHD) is limited and few disease-causing mutations have been identified. To identify novel CHD genes, we performed a forward genetic screen to identify mutant mouse lines with heritable CHD. Lines with recessive N-ethyl-N-nitrsourea-induced CHD-causing mutations were identified using a three-generation backcross. A hierarchical screening protocol was used to test the hypothesis that the fetal-to-neonatal circulatory transition unmasks the specific structural heart defects observed in CHD. Mice with heart defects were efficiently ascertained by selecting for pups exhibiting perinatal lethality and characterizing their cardiac pathology. A marked increase of perinatal lethality was observed in the mutagen-treated cohort compared with an untreated backcross population. Cardiac pathology on perinatal lethals revealed cardiovascular defects in 79 pups from 47 of 321 mutagenized lines. All identified structural abnormalities were analogous to previously described forms of human CHD. Furthermore, the phenotypic recurrence and variance patterns across all lines were similar to human CHD prevalence and recurrence patterns. We mapped the locus responsible for heritable atrioventricular septal defects in six lines (avc1-6). Our screen demonstrated that 'sporadic' CHD may have major genetic component and established a practical, efficient approach for identifying CHD candidate genes.
- Published
- 2010
- Full Text
- View/download PDF
21. Ca 2+ dysregulation in Ryr1 I4895T/wt mice causes congenital myopathy with progressive formation of minicores, cores, and nemaline rods
- Author
-
Eric Bombardier, Jon G. Seidman, David H. MacLennan, Frederic Depreux, Robert A. McCloy, Christine E. Seidman, Alexander Kraev, Douglas Holmyard, Elena Zvaritch, Natasha Kraeva, and A. Russell Tupling
- Subjects
Mutant ,Biology ,Myopathies, Nemaline ,Pathogenesis ,Mice ,Microscopy, Electron, Transmission ,medicine ,Animals ,RNA, Messenger ,Muscle, Skeletal ,RYR1 ,Multidisciplinary ,Reverse Transcriptase Polymerase Chain Reaction ,Ryanodine receptor ,Skeletal muscle ,Ryanodine Receptor Calcium Release Channel ,Anatomy ,Biological Sciences ,medicine.disease ,Congenital myopathy ,Cell biology ,Phenotype ,medicine.anatomical_structure ,Calcium ,Myofibril ,Central core disease - Abstract
Ryr1 I4895T/wt (IT/+) mice express a knockin mutation corresponding to the human I4898T EC-uncoupling mutation in the type 1 ryanodine receptor/Ca 2+ release channel (RyR1), which causes a severe form of central core disease (CCD). IT/+ mice exhibit a slowly progressive congenital myopathy, with neonatal respiratory stress, skeletal muscle weakness, impaired mobility, dorsal kyphosis, and hind limb paralysis. Lesions observed in myofibers from diseased mice undergo age-dependent transformation from minicores to cores and nemaline rods. Early ultrastructural abnormalities include sarcomeric misalignment, Z-line streaming, focal loss of cross-striations, and myofibrillar splitting and intermingling that may arise from defective myofibrillogenesis. However, manifestation of the disease phenotype is highly variable on a Sv129 genomic background. Quantitative RT-PCR shows an equimolar ratio of WT and mutant Ryr1 transcripts within IT/+ myofibers and total RyR1 protein expression levels are normal. We propose a unifying theory in which the cause of core formation lies in functional heterogeneity among RyR1 tetramers. Random combinations of normal and either leaky or EC-uncoupled RyR subunits would lead to spatial differences in Ca 2+ transients; the resulting heterogeneity of contraction among myofibrils would lead to focal, irreversible tearing and shearing, which would, over time, enlarge to form minicores, cores, and nemaline rods. The IT/+ mouse line is proposed to be a valid model of RyR1-related congenital myopathy, offering high potential for elucidation of the pathogenesis of skeletal muscle disorders arising from impaired EC coupling.
- Published
- 2009
- Full Text
- View/download PDF
22. Variation in the 4q25 Chromosomal Locus Predicts Atrial Fibrillation After Coronary Artery Bypass Graft Surgery
- Author
-
Nancy J. Brown, Peter Lichtner, Tjorvi E. Perry, Dan M. Roden, Patrick T. Ellinor, Christopher Newton-Cheh, Charles D. Collard, Mias Pretorius, Thomas Meitinger, Amanda A. Fox, Jochen D. Muehlschlegel, Sary F. Aranki, Marylyn D. Ritchie, Arne Pfeufer, Stefan Kääb, Jon G. Seidman, Kuang Yu Liu, Simon C. Body, Christine E. Seidman, Dawood Darbar, Stanton K. Shernan, Brian S. Donahue, Juan Carlos Estrada, and Daniel S. Herman
- Subjects
Adult ,Male ,medicine.medical_specialty ,Polymorphism, Single Nucleotide ,Article ,Young Adult ,Postoperative Complications ,Predictive Value of Tests ,Internal medicine ,Atrial Fibrillation ,Genetics ,Humans ,Medicine ,Prospective Studies ,Coronary Artery Bypass ,Prospective cohort study ,Genetics (clinical) ,Aged ,Aged, 80 and over ,business.industry ,Genetic Variation ,Postoperative complication ,Atrial fibrillation ,Odds ratio ,Middle Aged ,medicine.disease ,Surgery ,Cardiac surgery ,Predictive value of tests ,Cohort ,Chromosomal region ,Cardiology ,Female ,Chromosomes, Human, Pair 4 ,Cardiology and Cardiovascular Medicine ,business - Abstract
Background— Atrial fibrillation (AF) is the most common adverse event following coronary artery bypass graft surgery. A recent study identified chromosome 4q25 variants associated with AF in ambulatory populations. However, their role in postoperative AF is unknown. We hypothesized that genetic variants in the 4q25 chromosomal region are independently associated with postoperative AF after coronary artery bypass graft surgery. Methods and Results— Two prospectively collected cohorts of patients undergoing coronary artery bypass graft surgery, with or without concurrent valve surgery, at 3 US centers. From a discovery cohort of 959 patients, clinical and genomic multivariate predictors of postoperative AF were identified by genotyping 45 single-nucleotide polymorphisms (SNPs) encompassing the 4q25 locus. Three SNPs were then assessed in a separately collected validation cohort of 494 patients. After adjustment for clinical predictors of postoperative AF and multiple comparisons, rs2200733, rs13143308, and 5 other linked SNPs independently predicted postoperative AF in the discovery cohort. Additive odds ratios for the 7 associated 4q25 SNPs ranged between 1.57 and 2.17 ( P =8.0�10 −4 to 3.4�10 −6 ). Association with postoperative AF were measured and replicated for rs2200733 and rs13143308 in the validation cohort. Conclusions— In 2 independently collected cardiac surgery cohorts, noncoding SNPs within the chromosome 4q25 region are independently associated with postoperative AF after coronary artery bypass graft surgery after adjusting for clinical covariates and multiple comparisons.
- Published
- 2009
- Full Text
- View/download PDF
23. Comprehensive genomic characterization defines human glioblastoma genes and core pathways
- Author
-
Sandy Aronson, Leslie Cope, Michael L. Bittner, Daniel C. Koboldt, Alex E. Lash, W. K. Alfred Yung, Margaret Morgan, Devin Absher, Carl F. Schaefer, Roger E. McLendon, Michael D. Prados, Josh Gould, Ju Han, Stacey Gabriel, Scott R. VandenBerg, Ilana Perna, Troy Shelton, Junyuan Wu, Sacha Scott, Steve Scherer, Michael J. T. O’Kelly, Li Ding, Erin Hickey, Elizabeth J. Thomson, Bahram Parvin, Kim D. Delehaunty, Gi Choi Yoon, Mark D. Robinson, Oliver Bogler, Darrell D. Bigner, Michael R. Reich, Jianhua Zhang, Robert S. Fulton, Allan H. Friedman, Tammi L. Vickery, Amita Aggarwal, Subhashree Madhavan, Liuda Ziaugra, Yuan Qi, Vandita Joshi, Eric Van Name, Jane Wilkinson, W. Ruprecht Wiedemeyer, Xiaoqi Shi, Richard A. Gibbs, Lynda Chin, Jessica Chen, Stefano Monti, Erwin G. Van Meir, John Ngai, Amy Hawkins, Elizabeth Lenkiewicz, Brad Ozenberger, Shannon Dorton, Georgia Ren, John N. Weinstein, Gena M. Mastrogianakis, Asif T. Chinwalla, Scott L. Carter, Nicholas D. Socci, Rachel Abbott, Gavin Sherlock, Lucinda Fulton, Hyun Soo Kim, Fei Pan, Magali Cavatore, Gabriele Alexe, Francis S. Collins, Narayanan Sathiamoorthy, Lakshmi Jakkula, Brian H. Dunford-Shore, Jireh Santibanez, Tom Mikkelsen, Huy V. Nguyen, Levi A. Garraway, Christopher A. Miller, Jinghui Zhang, Ken Chen, Timothy Fennell, Robert Sfeir, James A. Robinson, Alexey Stukalov, Richard K. Wilson, Matthew Meyerson, Daniel J. Weisenberger, Mi Yi Joo, Yevgeniy Antipin, Anna Lapuk, Gerald V. Fontenay, Nicolas Stransky, Adam B. Olshen, Elizabeth Purdom, Josh Korn, Huyen Dinh, Sai Balu, Victoria Wang, James G. Herman, Christie Kovar, Kristian Cibulskis, Tisha Chung, Agnes Viale, Paul T. Spellman, Supriya Gupta, Melissa Parkin, Peter J. Park, Maddy Wiechert, John W. Wallis, Peter W. Laird, Nikolaus Schultz, James D. Brooks, David Nassau, Jun Li, John R. Osborne, Anna D. Barker, Peter Fielding, Boris Reva, Karen Vranizan, D. Neil Hayes, Aleksandar Milosavljevic, Lawrence A. Donehower, Won Kong Sek, Daniela S. Gerhard, Otis Hall, Rameen Beroukhim, Audrey Southwick, George M. Weinstock, Chris Markovic, Roel G.W. Verhaak, David Van Den Berg, Joe W. Gray, Yanru Ren, Ethan Cerami, Yiming Zhu, Amrita Ray, Yonghong Xiao, Kristin G. Ardlie, William L. Gerald, Michael S. Lawrence, Gerald R. Fowler, Mark S. Guyer, Isaac S. Kohane, Kornel E. Schuebel, Mitchel S. Berger, Jeffrey J. Olson, Gary W. Swift, Lora Lewis, Sheri Sanders, Norman L. Lehman, Eric S. Lander, Robert Penny, Liliana Villafania, John G. Conboy, Ari B. Kahn, Henry Marr, Heidi S. Feiler, Lynn Nazareth, David J. Dooling, Katherine A. Hoadley, Alicia Hawes, Marc Ladanyi, Aniko Sabo, Wendy Winckler, Vivian Peng, Barbara A. Weir, Daniel J. Brat, Scott Morris, Carolyn C. Compton, Todd R. Golub, Scott Abbott, Michael D. McLellan, Jiqiang Yao, Shalini N. Jhangiani, Michael D. Topal, Michael C. Wendl, Gad Getz, Jun Yao, Derek Y. Chiang, Larry Feng, Steffen Durinck, David A. Wheeler, Yuzhu Tang, Benjamin Gross, Barry S. Taylor, Kenneth Aldape, Craig Pohl, Rick Meyer, Peter J. Good, Ling Lin, Elaine R. Mardis, Robert C. Onofrio, Jane Peterson, Stephen B. Baylin, Li-Xuan Qin, Andrew Cree, Cameron Brennan, Charles M. Perou, William Courtney, Omar Alvi, Donna M. Muzny, Joseph G. Vockley, Jill P. Mesirov, Yan Shi, Alexei Protopopov, Jim Vaught, Craig H. Mermel, Scott Mahan, Laetitia Borsu, Heather Schmidt, Jennifer Baldwin, Tracie L. Miner, Toby Bloom, David E. Larson, Leander Van Neste, Nicholas J. Wang, Kenneth H. Buetow, Raju Kucherlapati, Anthony San Lucas, Martin L. Ferguson, Terence P. Speed, Venkatraman E. Seshan, Debbie Beasley, Carrie Sougnez, Carrie A. Haipek, Richard M. Myers, Chris Sander, Qing Wang Wei, Jon G. Seidman, Rob Nicol, Manuel L. Gonzalez-Garay, Shin Leong, Shannon T. Brady, and University of Groningen
- Subjects
Male ,Models, Molecular ,DNA Repair ,Gene Dosage ,NEUROFIBROMATOSIS TYPE-1 ,MISMATCH REPAIR ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Genes, Tumor Suppressor ,DNA Modification Methylases ,Proneural Glioblastoma ,Aged, 80 and over ,Genetics ,0303 health sciences ,Neurofibromin 1 ,Multidisciplinary ,Brain Neoplasms ,NF1 GENE ,Genomics ,Middle Aged ,TUMORS ,ALKYLATING-AGENTS ,3. Good health ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,DNA methylation ,Female ,DNA mismatch repair ,Functional genomics ,Signal Transduction ,Adult ,Adolescent ,CELL-LINES ,Oncogenomics ,Biology ,Article ,03 medical and health sciences ,PIK3CA GENE ,Humans ,Epigenetics ,Gene ,Aged ,Retrospective Studies ,030304 developmental biology ,HIGH-FREQUENCY ,Genome, Human ,Tumor Suppressor Proteins ,SOMATIC MUTATIONS ,Genes, erbB-1 ,DNA Methylation ,Protein Structure, Tertiary ,MALIGNANT GLIOMAS ,DNA Repair Enzymes ,Mutation ,Glioblastoma - Abstract
Human cancer cells typically harbour multiple chromosomal aberrations, nucleotide substitutions and epigenetic modifications that drive malignant transformation. The Cancer Genome Atlas ( TCGA) pilot project aims to assess the value of large- scale multi- dimensional analysis of these molecular characteristics in human cancer and to provide the data rapidly to the research community. Here we report the interim integrative analysis of DNA copy number, gene expression and DNA methylation aberrations in 206 glioblastomas - the most common type of primary adult brain cancer - and nucleotide sequence aberrations in 91 of the 206 glioblastomas. This analysis provides new insights into the roles of ERBB2, NF1 and TP53, uncovers frequent mutations of the phosphatidylinositol- 3- OH kinase regulatory subunit gene PIK3R1, and provides a network view of the pathways altered in the development of glioblastoma. Furthermore, integration of mutation, DNA methylation and clinical treatment data reveals a link between MGMT promoter methylation and a hypermutator phenotype consequent to mismatch repair deficiency in treated glioblastomas, an observation with potential clinical implications. Together, these findings establish the feasibility and power of TCGA, demonstrating that it can rapidly expand knowledge of the molecular basis of cancer.
- Published
- 2008
- Full Text
- View/download PDF
24. Control of In Vivo Contraction/Relaxation Kinetics by Myosin Binding Protein C
- Author
-
Jeffrey Robbins, Sakthivel Sadayappan, Jon G. Seidman, David A. Kass, James O. Mudd, Takahiro Nagayama, and Eiki Takimoto
- Subjects
Cardiac function curve ,medicine.medical_specialty ,Myosin light-chain kinase ,Contraction (grammar) ,Binding protein ,Biology ,Molecular biology ,Sarcomere ,Endocrinology ,Physiology (medical) ,Internal medicine ,medicine ,Phosphorylation ,Cardiology and Cardiovascular Medicine ,Protein kinase A ,Protein kinase C - Abstract
Background— Cardiac myosin binding protein-C (cMyBP-C) is a thick-filament protein whose presence and phosphorylation by protein kinase A (PKA) regulates cross-bridge formation and kinetics in isolated myocardium. We tested the influence of cMyBP-C and its PKA-phosphorylation on contraction/relaxation kinetics in intact hearts and revealed its essential role in several classic properties of cardiac function. Methods and Results— Comprehensive in situ cardiac pressure–volume analysis was performed in mice harboring a truncation mutation of cMyBP-C (cMyBP-C (t/t) ) that resulted in nondetectable protein versus hearts re-expressing solely wild-type (cMyBP-C WT:(t/t) ) or mutated protein in which known PKA-phosphorylation sites were constitutively suppressed (cMyBP-C AllP-:(t/t) ). Hearts lacking cMyBP-C had faster early systolic activation, which then terminated prematurely, limiting ejection. Systole remained short at faster heart rates; thus, cMyBP-C (t/t) hearts displayed minimal rate-dependent decline in diastolic time and cardiac preload. Furthermore, prolongation of pressure relaxation by afterload was markedly blunted in cMyBP-C (t/t) hearts. All 3 properties were similarly restored to normal in cMyBP-C WT:(t/t) and cMyBP-C AllP-:(t/t) hearts, which supports independence of PKA-phosphorylation. However, the dependence of peak rate of pressure rise on preload was specifically depressed in cMyBP-C AllP-:(t/t) hearts, whereas cMyBP-C (t/t) and cMyBP-C AllP-:(t/t) hearts had similar blunted adrenergic and rate-dependent contractile reserve, which supports linkage of these behaviors to PKA-cMyBP-C modification. Conclusions— cMyBP-C is essential for major properties of cardiac function, including sustaining systole during ejection, the heart-rate dependence of the diastolic time period, and relaxation delay from increased arterial afterload. These are independent of its phosphorylation by PKA, which more specifically modulates early pressure rise rate and adrenergic/heart rate reserve.
- Published
- 2007
- Full Text
- View/download PDF
25. Abstract 46: Identification of Novel Alternate Splicing Events in Humans With RBFOX2 Mutations and Hypoplastic Left Heart Syndrome
- Author
-
Richard B. Kim, Jason Homsy, Christine E. Seidman, Jon G. Seidman, Josh Gorham, David M. McKean, and Craig C. Benson
- Subjects
Gene isoform ,Genetics ,Physiology ,Alternative splicing ,medicine ,RNA-Seq ,RNA-binding protein ,Biology ,Cardiology and Cardiovascular Medicine ,medicine.disease ,Hypoplastic left heart syndrome - Abstract
Alternative splicing (AS) of protein isoforms is an integral mechanism for cardiac development. RNA Binding Protein, Fox-1 Homolog (C. Elegans) 2 (RBFOX2) is an RNA binding protein preferentially expressed in muscle and neuronal cells and regulates tissue-specific alternate exon splicing in ~2,100 target genes by binding the conserved RNA sequence motif (U)GCAUG. RBFOX2 was recently implicated in the pathogenesis of abnormal cardiac and cerebral development via loss-of-function studies in zebrafish and mouse. However, convincing evidence remains incomplete, as the full complement of RBFOX2 target genes and differential exon usage (DEU) in human cardiovascular cell lines are incompletely defined. We identified de novo mutations in RBFOX2 from four human cases of congenital heart disease (CHD) with hypoplastic left heart syndrome (HLHS) via whole exome sequencing. To test the hypothesis that RBFOX2 mutations alter DEU in known target genes, we performed RNA-seq on ductus arteriosus tissue from human CHD cases with and without RBFOX2 mutation. Analysis of RNA-seq for DEU was performed with DEXSeq. To limit the effect of differential gene expression, we restricted analysis to subjects with high global gene expression correlation (r2 > 0.9, case=1 vs. control=5). DEU in known RBFOX2 target genes were highly enriched compared to all known genes (115/2,100 vs. 589/26,310, p=5.78e-15). A high percentage of the DEU genes (60.0%, 69/115) have high heart expression (HHE) in the developing mouse (mean expression of four cardiac chambers at e14.5). DEU genes with HHE include VCL, TPM1, FN1, ACTN1, and CALD1. Functional annotation clustering reveals enrichment for several actin, cytoskeletal, and contractile Gene Ontology terms, suggesting a possible role in the epithelial-mesenchymal transition (EMT) developmental processes active during early cardiac formation. We also identified enrichment from genes implicated in CHD by allelic specific expression or de novo mutations (44/1,263 vs. 589/26,310, p=0.007). These results are the first in humans to identify differentially expressed exons associated with RBFOX2 mutations in CHD and suggests RBFOX2-mediated alternate splicing may influence EMT pathways implicated in the pathogenesis of HLHS.
- Published
- 2015
- Full Text
- View/download PDF
26. N488I Mutation of the γ2-Subunit Results in Bidirectional Changes in AMP-Activated Protein Kinase Activity
- Author
-
Joanne S. Ingwall, Mei Shen, Huamei He, Liqun Zou, Christine E. Seidman, Michael Arad, Rong Tian, Jon G. Seidman, and Bo Løfgren
- Subjects
medicine.medical_specialty ,Physiology ,Protein subunit ,Mutant ,Adenylate kinase ,Mice, Transgenic ,AMP-Activated Protein Kinases ,Protein Serine-Threonine Kinases ,Mice ,Adenosine Triphosphate ,AMP-activated protein kinase ,Multienzyme Complexes ,Internal medicine ,medicine ,Animals ,Humans ,Phosphorylation ,Protein kinase A ,biology ,Myocardium ,AMPK ,AMP-activated protein kinase activity ,Adenosine Monophosphate ,Cell biology ,Protein Subunits ,Endocrinology ,Mutation ,biology.protein ,Cardiology and Cardiovascular Medicine ,Glycogen - Abstract
Mutations in the human gene encoding the nucleotide-binding region in the gamma-subunit of AMP-activated protein kinase (AMPK) cause cardiomyopathy with preexcitation syndrome. Mutant AMPK showed reduced binding affinity to nucleotides in vitro raising the possibility that altered regulation of AMPK activity by AMP/ATP could contribute to the disease phenotype. In this study, we determined the sensitivity of AMPK activity to AMP/ATP in the beating hearts using transgenic mice expressing a mutant (N488I, gamma2-mutant) or wild-type gamma2-subunit (gamma2-TG). The [ATP] and [AMP] were unaltered in all hearts but the AMPK activity was increased by 2.5-fold in gamma2-mutant hearts freeze-clamped at normal AMP/ATP compared with nontransgenic (WT) or gamma2-TG. The increased basal AMPK activity was caused by increased Thr-172 phosphorylation of the alpha-subunit (p-AMPK, by 4-fold) at normal [ATP] and was not changed by reducing glycogen content by 60% in the gamma2-mutant hearts. A reversal of AMP/ATP, caused by ATP degradation, increased p-AMPK by 7-fold in WT but caused no change in gamma2-mutant hearts. These results demonstrate that the mutation renders AMPK insensitive to the inhibitory and stimulatory effects of the regulatory nucleotides ATP and AMP, respectively, suggesting that the pathogenesis of the human disease may not be attributable to a simple loss- or gain-of-function.
- Published
- 2005
- Full Text
- View/download PDF
27. Characterization of HPV and host genome interactions in primary head and neck cancers
- Author
-
J. Jack Lee, Carter Van Waes, Michael B. Prystowsky, Akinyemi I. Ojesina, Joshua M. Stuart, Mitchell J. Frederick, Angela By Hui, Saianand Balu, Scot Waring, Inanc Birol, Bayardo Perez-Ordonez, Aaron D. Black, Payal Sipahimalani, Wiktoria Maria Suchorska, Michele C. Hayward, Jeffrey Roach, Adrian Ally, Ranabir Guin, Samuel S. Freeman, J. Todd Auman, Hollie A. Harper, Mark E. Prince, David Van Den Berg, Nipun Kakkar, James G. Herman, Adam M. Zanation, Nilsa C. Ramirez, Daniel DiCara, Carol R. Bradford, Paul C. Boutros, Peter W. Laird, Evan G. Taylor, Yan Shi, Jonathan M. Irish, Kristian Cibulskis, Junyuan Wu, Darlene Lee, Katherine A. Hoadley, Jeff Bruce, Yingchun Liu, Heather M. Walline, Carol G. Shores, Raju Kucherlapati, Scott M. Lippman, Colleen Mitchell, Steven E. Schumacher, Wendell G. Yarbrough, Tanguy Y. Seiwert, Sophie C. Egea, Elena Ivanova, Troy Shelton, Jiexin Zhang, Patrick K. Kimes, Nikolaus Schultz, Noreen Dhalla, Maciej Wiznerowicz, Daniel J. Weisenberger, Martin L. Ferguson, Tamara R. Jones, Haiyan I. Li, Nishant Agrawal, Yiling Lu, Lixing Yang, Brandee T. Brown, Julie M. Gastier-Foster, Nils Weinhold, Harshad S. Mahadeshwar, Angela Tam, Lihua Zou, Tom Bodenheimer, Gordon B. Mills, W. Kimryn Rathmell, David Mallery, Semin Lee, Scott J. Morris, Lynn M. Herbert, Hailei Zhang, Thomas C. Motter, Walker Hale, Tina Wong, Yichao Sun, Heidi J. Sofia, Rileen Sinha, Dennis T. Maglinte, Robert I. Haddad, Matthew D. Wilkerson, Jinze Liu, Julien Baboud, Brenda Diergaarde, Michelle Q. Pham, Tanja M. Davidsen, Maureen A. Sartor, Robert A. Holt, Angela Hadjipanayis, Pei Lin, Anders Jacobsen, Stephen C. Benz, Dean Cheng, Matthew Meyerson, Andy Chu, Ezra E.W. Cohen, Joonil Jung, Ari B. Kahn, Scott Frazer, Leigh B. Thorne, Andrew J. Mungall, David A. Wheeler, Nina Thiessen, Jay Bowen, Hui Cheng, D. Neil Hayes, Jennifer Drummond, Patrick Kwok Shing Ng, Harshavardhan Doddapaneni, S. Onur Sumer, Katherine Tarvin, John W.F. Waldron, Nils Gehlenborg, Douglas B. Chepeha, Janae V. Simons, Robert L. Ferris, Paweł Golusiński, Christina Beard, Donna Morton, Boris Reva, Psalm Haseley, Andrew Wei Xu, Giovanni Ciriello, Gregory T. Wolf, Ken Burnett, Lisa A. Peterson, Sylvia Wrenn, Mark A. Jensen, Travis I. Zack, Deepak Srinivasan, Thomas E. Carey, Lisle E. Mose, Mark E. Sherman, Leslie Cope, Adel K. El-Naggar, Marjorie Romkes, D Wheeler, Christopher A. Bristow, Andrea Haddad, Mayya Malakh, Ludmila Danilova, Jean C. Zenklusen, Jiabin Tang, Laura S. Rozek, Jon G. Seidman, Steven J.M. Jones, Hsu Chao, Candance Shelton, Carmen Gomez-Fernandez, Carrie Sougnez, Piotr A. Mieczkowski, Lee Lichtenstein, Erik Zmuda, Johanna Gardner, Jianjiong Gao, Marco A. Marra, Juok Cho, Alexei Protopopov, Roy Tarnuzzer, Tod D. Casasent, Timothy M. Chan, Michael Parfenov, Jing Wang, Christine M. Komarck, Lauren Averett Byers, Rehan Akbani, Todd Pihl, Curtis R. Pickering, Aaron D. Tward, Chandra Sekhar Pedamallu, Han Si, Jan F. Prins, Vonn Walter, Simion I. Chiosea, Jacqueline E. Schein, James Stephen Marron, Ariane Nguyen, William W. Shockley, Jennifer R. Grandis, Zhining Wang, Jeffrey N. Myers, Konstanty Korski, Bradley A. Ozenberger, Margaret L. Gulley, Barry S. Taylor, B. Arman Aksoy, Ni Zhao, Ashley H. Salazar, Petar Stojanov, Tara M. Lichtenberg, Chu Chen, Donghui Tan, Bhishamjit S. Chera, Andrzej Mackiewicz, A. Gordon Robertson, Ronglai Shen, Lisa Wise, Charles M. Perou, Michael Mayo, Charles Saller, Timothy J. Triche, Dong Zeng, Yan Guo, Lixia Diao, Jonathan B. McHugh, Zhixiang Zuo, Gad Getz, John A. Demchok, Raja R. Seethala, Gordon Saksena, Liu Xi, Liming Yang, Justin A. Bishop, Jessica Walton, Greg Eley, Jessica Frick, John N. Weinstein, Kevin Lau, Jianhua Zhang, Kenna R. Mills Shaw, Miruna Balasundaram, Zubair Khan, Umadevi Veluvolu, Stuart R. Jefferys, Christie Kovar, Moiz S. Bootwalla, Rebecca Carlsen, Daniel Crain, Fei-Fei Liu, Matthiew Ibbs, Michael S. Noble, William K. Funkhouser, Shaowu Meng, Mei Huang, Ann Marie Egloff, George E. Sandusky, Yi Han, Eric S. Lander, Sam Ng, William Lee, Christina Yau, Reanne Bowlby, Erin Curley, Donna M. Muzny, Lynda Chin, Joel S. Parker, Stephen B. Baylin, Peter S. Hammerman, Julie Ahn, Christopher C. Benz, Benjamin Gross, Paul M. Weinberger, Gideon Dresdner, Luc G. T. Morris, Zhong Chen, You Hong Fan, Angeliki Pantazi, David I. Heiman, Bartosz Szybiak, Sahil Seth, Richard A. Gibbs, Trevor Hackman, Peggy Yena, Richard A. Moore, Darshan Singh, Yaron S.N. Butterfield, Andrew D. Cherniack, Jeffrey S. Moyer, Robert A. Burton, Peter J. Park, Rameen Beroukhim, Ricardo Ramirez, Natalia Issaeva, Michael S. Lawrence, Wen-Bin Liu, Xiaojia Ren, Chris Sander, Yunhu Wan, Daryl Waggott, Joseph A. Califano, David A. Pot, Mathew G. Soloway, Roni J. Bollag, Stacey Gabriel, Jeffrey S. Reid, Netty Santoso, Elizabeth Buda, Doug Voet, Xingzhi Song, Kyle Chang, Joseph Paulauskis, Thomas M. Harris, Jaegil Kim, Alan P. Hoyle, Marc Ladanyi, Anthony Saleh, Mark C. Weissler, Scott L. Carter, Kai Wang, Jonathan G. Seidman, Corbin D. Jones, Wojciech Golusiński, Robert Penny, Margi Sheth, and Lori Boice
- Subjects
Cervical Cancer ,Genome ,head and neck ,Cancer Genome Atlas Network ,2.1 Biological and endogenous factors ,2.2 Factors relating to the physical environment ,Papillomaviridae ,Aetiology ,Genetics ,Multidisciplinary ,biology ,HPV infection ,virus diseases ,Biological Sciences ,integration sites ,female genital diseases and pregnancy complications ,Gene Expression Regulation, Neoplastic ,Infectious Diseases ,Head and Neck Neoplasms ,DNA methylation ,Host-Pathogen Interactions ,HIV/AIDS ,Infection ,Human ,Virus Integration ,Molecular Sequence Data ,DNA sequencing ,Rare Diseases ,medicine ,Humans ,cancer ,Dental/Oral and Craniofacial Disease ,Gene ,genome rearrangement ,Neoplastic ,Base Sequence ,Genome, Human ,Human Genome ,DNA Methylation ,biology.organism_classification ,medicine.disease ,Human genetics ,Gene Expression Regulation ,Genes ,Neoplasm ,Sexually Transmitted Infections ,Human genome ,Genes, Neoplasm ,papilloma virus - Abstract
Previous studies have established that a subset of head and neck tumors contains human papillomavirus (HPV) sequences and that HPV-driven head and neck cancers display distinct biological and clinical features. HPV is known to drive cancer by the actions of the E6 and E7 oncoproteins, but the molecular architecture of HPV infection and its interaction with the host genome in head and neck cancers have not been comprehensively described. We profiled a cohort of 279 head and neck cancers with next generation RNA and DNA sequencing and show that 35 (12.5%) tumors displayed evidence of high-risk HPV types 16, 33, or 35. Twenty-five cases had integration of the viral genome into one or more locations in the human genome with statistical enrichment for genic regions. Integrations had a marked impact on the human genome and were associated with alterations in DNA copy number, mRNA transcript abundance and splicing, and both inter- and intrachromosomal rearrangements. Many of these events involved genes with documented roles in cancer. Cancers with integrated vs. nonintegrated HPV displayed different patterns of DNA methylation and both human and viral gene expressions. Together, these data provide insight into the mechanisms by which HPV interacts with the human genome beyond expression of viral oncoproteins and suggest that specific integration events are an integral component of viral oncogenesis.
- Published
- 2014
28. β–Myosin Heavy Chain Variant Met606Val Causes Very Mild Hypertrophic Cardiomyopathy in Mice, but Exacerbates HCM Phenotypes in Mice Carrying Other HCM Mutations
- Author
-
Christine E. Seidman, Katarzyna Hackert, Robert Blankenburg, Jon G. Seidman, Sebastian Wurster, René Deenen, Joachim P. Schmitt, and Martin J. Lohse
- Subjects
Models, Molecular ,medicine.medical_specialty ,Genotype ,Transcription, Genetic ,Protein Conformation ,Physiology ,Mutation, Missense ,macromolecular substances ,Biology ,medicine.disease_cause ,Article ,Muscle hypertrophy ,Ventricular Myosins ,Mice ,Internal medicine ,Myosin ,medicine ,Cardiomyopathy, Hypertrophic, Familial ,Missense mutation ,Animals ,Humans ,Point Mutation ,Gene Knock-In Techniques ,Ventricular remodeling ,Ultrasonography ,Genetics ,Mutation ,Ventricular Remodeling ,Myosin Heavy Chains ,Point mutation ,Hypertrophic cardiomyopathy ,medicine.disease ,Myocardial Contraction ,Disease Models, Animal ,Phenotype ,Endocrinology ,Amino Acid Substitution ,Cyclosporine ,Hypertrophy, Left Ventricular ,Cardiology and Cardiovascular Medicine ,Cardiac Myosins - Abstract
Rationale : Approximately 40% of hypertrophic cardiomyopathy (HCM) is caused by heterozygous missense mutations in β-cardiac myosin heavy chain (β-MHC). Associating disease phenotype with mutation is confounded by extensive background genetic and lifestyle/environmental differences between subjects even from the same family. Objective : To characterize disease caused by β-cardiac myosin heavy chain Val606Met substitution (VM) that has been identified in several HCM families with wide variation of clinical outcomes, in mice. Methods and Results : Unlike 2 mouse lines bearing the malignant myosin mutations Arg453Cys (RC/+) or Arg719Trp (RW/+), VM/+ mice with an identical inbred genetic background lacked hallmarks of HCM such as left ventricular hypertrophy, disarray of myofibers, and interstitial fibrosis. Even homozygous VM/VM mice were indistinguishable from wild-type animals, whereas RC/RC- and RW/RW-mutant mice died within 9 days after birth. However, hypertrophic effects of the VM mutation were observed both in mice treated with cyclosporine, a known stimulator of the HCM response, and compound VM/RC heterozygous mice, which developed a severe HCM phenotype. In contrast to all heterozygous mutants, both systolic and diastolic function of VM/RC hearts was severely impaired already before the onset of cardiac remodeling. Conclusions : The VM mutation per se causes mild HCM-related phenotypes; however, in combination with other HCM activators it exacerbates the HCM phenotype. Double-mutant mice are suitable for assessing the severity of benign mutations.
- Published
- 2014
29. Familial Dilated Cardiomyopathy Locus Maps to Chromosome 2q31
- Author
-
Hideshi Niimura, John A. Osborne, Jon G. Seidman, Benjamin L. Siu, Diane Fatkin, Calum A. MacRae, Christine E. Seidman, Scott D. Solomon, and D. Woodrow Benson
- Subjects
Adult ,Cardiomyopathy, Dilated ,Male ,Candidate gene ,Adolescent ,Heart disease ,Genetic Linkage ,Cardiomyopathy ,Locus (genetics) ,Genetic linkage ,Physiology (medical) ,medicine ,Humans ,Child ,Aged ,Genetics ,biology ,Chromosome Mapping ,Dilated cardiomyopathy ,Middle Aged ,medicine.disease ,Pedigree ,Chromosomes, Human, Pair 2 ,Heart failure ,biology.protein ,Female ,Titin ,Cardiology and Cardiovascular Medicine - Abstract
Background —Inherited gene defects are an important cause of dilated cardiomyopathy. Although the chromosome locations of some defects and 1 disease gene (actin) have been identified, the genetic etiologies of most cases of familial dilated cardiomyopathy remain unknown. Methods and Results —We clinically evaluated 3 generations of a kindred with autosomal dominant transmission of dilated cardiomyopathy. Nine surviving and affected individuals had early-onset disease (ventricular chamber dilation during the teenage years and congestive heart failure during the third decade of life). The disease was nonpenetrant in 2 obligate carriers. To identify the causal gene defect, linkage studies were performed. A new dilated cardiomyopathy locus was identified on chromosome 2 between loci GCG and D2S72 (maximum logarithm of odds [LOD] score=4.86 at θ=0). Because the massive gene encoding titin, a cytoskeletal muscle protein, resides in this disease interval, sequences encoding 900 amino acid residues of the cardiac-specific (N2-B) domain were analyzed. Five sequence variants were identified, but none segregated with disease in this family. Conclusions —A dilated cardiomyopathy locus (designated CMD1G ) is located on chromosome 2q31 and causes early-onset congestive heart failure. Although titin remains an intriguing candidate gene for this disorder, a disease-causing mutation is not present in its cardiac-specific N2-B domain.
- Published
- 1999
- Full Text
- View/download PDF
30. Neonatal cardiomyopathy in mice homozygous for the Arg403Gln mutation in the α cardiac myosin heavy chain gene
- Author
-
Shardha Srinivasan, Michael E. Christe, Orlando Aristizabal, Frederick J. Schoen, Jon G. Seidman, Diane Fatkin, Christine E. Seidman, Daniel H. Turnbull, and Bradley K. McConnell
- Subjects
medicine.medical_specialty ,Cell Survival ,Heart Ventricles ,Cardiomyopathy ,Mice, Transgenic ,Biology ,Sarcomere ,Article ,Mice ,Dystrophic calcification ,Internal medicine ,Myosin ,medicine ,Animals ,Humans ,Missense mutation ,Myocyte ,Heart Atria ,Myosin Heavy Chains ,Histocytochemistry ,Myocardium ,Homozygote ,Dilated cardiomyopathy ,General Medicine ,medicine.disease ,Disease Models, Animal ,Microscopy, Electron ,Endocrinology ,Echocardiography ,Mutation ,Cardiology ,MYH6 ,Cardiomyopathies - Abstract
Heterozygous mice bearing an Arg403Gln missense mutation in the alpha cardiac myosin heavy chain gene (alpha-MHC403/+) exhibit the histopathologic features of human familial hypertrophic cardiomyopathy. Surprisingly, homozygous alpha-MHC403/403 mice die by postnatal day 8. Here we report that neonatal lethality is caused by a fulminant dilated cardiomyopathy characterized by myocyte dysfunction and loss. Heart tissues from neonatal wild-type and alpha-MHC403/403 mice demonstrate equivalent switching of MHC isoforms; alpha isoforms in each increase from 30% at birth to 70% by day 6. Cardiac dimensions and function, studied for the first time in neonatal mice by high frequency (45 MHz) echocardiography, were normal at birth. Between days 4 and 6, alpha-MHC403/403 mice developed a rapidly progressive cardiomyopathy with left ventricular dilation, wall thinning, and reduced systolic contraction. Histopathology revealed myocardial necrosis with dystrophic calcification. Electron microscopy showed normal architecture intermixed with focal myofibrillar disarray. We conclude that 45-MHz echocardiography is an excellent tool for assessing cardiac physiology in neonatal mice and that the concentration of Gln403 alpha cardiac MHC in myocytes influences both cell function and cell viability. We speculate that variable incorporation of mutant and normal MHC into sarcomeres of heterozygotes may account for focal myocyte death in familial hypertrophic cardiomyopathy.
- Published
- 1999
- Full Text
- View/download PDF
31. Targeted Ablation of the Murine α-Tropomyosin Gene
- Author
-
David W. Maughan, David A. Conner, Edward M. Blanchard, Michael E. Christe, Anja Geisterfer-Lowrance, Jon G. Seidman, Frederick J. Schoen, Christine E. Seidman, and Kenji Iizuka
- Subjects
Male ,Mice, Knockout ,Physiology ,Myocardium ,Mutant ,Gene targeting ,Tropomyosin ,macromolecular substances ,Biology ,musculoskeletal system ,Molecular biology ,Loss of heterozygosity ,Transgenesis ,Mice ,Gene Targeting ,Knockout mouse ,Animals ,Cardiology and Cardiovascular Medicine ,Haploinsufficiency ,tissues ,Gene - Abstract
Abstract We created a mouse that lacks a functional α-tropomyosin gene using gene targeting in embryonic stem cells and blastocyst-mediated transgenesis. Homozygous α-tropomyosin “knockout” mice die between embryonic day 9.5 and 13.5 and lack α-tropomyosin mRNA. Heterozygous α-tropomyosin knockout mice have ≈50% as much cardiac α-tropomyosin mRNA as wild-type littermates but similar α-tropomyosin protein levels. Cardiac gross morphology, histology, and function (assessed by working heart preparations) of heterozygous α-tropomyosin knockout and wild-type mice were indistinguishable. Mechanical performance of skinned papillary muscle strips derived from mutant and wild-type hearts also revealed no differences. We conclude that haploinsufficiency of the α-tropomyosin gene produces little or no change in cardiac function or structure, whereas total α-tropomyosin deficiency is incompatible with life. These findings imply that in heterozygotes there is a regulatory mechanism that maintains the level of myofibrillar tropomyosin despite the reduction in α-tropomyosin mRNA.
- Published
- 1997
- Full Text
- View/download PDF
32. Transcriptional Profiling of Cultured, Embryonic Epicardial Cells Identifies Novel Genes and Signaling Pathways Regulated by TGFβR3 In Vitro
- Author
-
H. Scott Baldwin, Danos C. Christodoulou, Christine E. Seidman, Jon G. Seidman, Daniel M. DeLaughter, Cynthia R. Clark, and Joey V. Barnett
- Subjects
0301 basic medicine ,Cell signaling ,Cell ,lcsh:Medicine ,Gene Expression ,Signal transduction ,Ligands ,Transcriptome ,Mice ,Gene expression ,Medicine and Health Sciences ,lcsh:Science ,Musculoskeletal System ,Regulation of gene expression ,Smooth Muscles ,Multidisciplinary ,Gene Ontologies ,Muscles ,NF-kappa B ,Signaling cascades ,Heart ,Genomics ,Epicardium ,Cell biology ,medicine.anatomical_structure ,Proteoglycans ,Anatomy ,Pericardium ,Research Article ,Cell Physiology ,BMP signaling ,Biology ,Cell Line ,03 medical and health sciences ,Genetics ,medicine ,Animals ,Gene Regulation ,Biology and life sciences ,030102 biochemistry & molecular biology ,Sequence Analysis, RNA ,lcsh:R ,Computational Biology ,Genome Analysis ,Embryonic stem cell ,Mice, Inbred C57BL ,030104 developmental biology ,TGF-beta signaling cascade ,Cell culture ,Cardiovascular Anatomy ,lcsh:Q ,Cell Immortalization ,Receptors, Transforming Growth Factor beta - Abstract
The epicardium plays an important role in coronary vessel formation and Tgfbr3-/- mice exhibit failed coronary vessel development associated with decreased epicardial cell invasion. Immortalized Tgfbr3-/- epicardial cells display the same defects. Tgfbr3+/+ and Tgfbr3-/- cells incubated for 72 hours with VEH or ligands known to promote invasion via TGFβR3 (TGFβ1, TGFβ2, BMP2), for 72 hours were harvested for RNA-seq analysis. We selected for genes >2-fold differentially expressed between Tgfbr3+/+ and Tgfbr3-/- cells when incubated with VEH (604), TGFβ1 (515), TGFβ2 (553), or BMP2 (632). Gene Ontology (GO) analysis of these genes identified dysregulated biological processes consistent with the defects observed in Tgfbr3-/- cells, including those associated with extracellular matrix interaction. GO and Gene Regulatory Network (GRN) analysis identified distinct expression profiles between TGFβ1-TGFβ2 and VEH-BMP2 incubated cells, consistent with the differential response of epicardial cells to these ligands in vitro. Despite the differences observed between Tgfbr3+/+ and Tgfbr3-/- cells after TGFβ and BMP ligand addition, GRNs constructed from these gene lists identified NF-ĸB as a key nodal point for all ligands examined. Tgfbr3-/- cells exhibited decreased expression of genes known to be activated by NF-ĸB signaling. NF-ĸB activity was stimulated in Tgfbr3+/+ epicardial cells after TGFβ2 or BMP2 incubation, while Tgfbr3-/- cells failed to activate NF-ĸB in response to these ligands. Tgfbr3+/+ epicardial cells incubated with an inhibitor of NF-ĸB signaling no longer invaded into a collagen gel in response to TGFβ2 or BMP2. These data suggest that NF-ĸB signaling is dysregulated in Tgfbr3-/- epicardial cells and that NF-ĸB signaling is required for epicardial cell invasion in vitro. Our approach successfully identified a signaling pathway important in epicardial cell behavior downstream of TGFβR3. Overall, the genes and signaling pathways identified through our analysis yield the first comprehensive list of candidate genes whose expression is dependent on TGFβR3 signaling.
- Published
- 2016
- Full Text
- View/download PDF
33. Genome-wide association study identifies a susceptibility locus for thoracic aortic aneurysms and aortic dissections spanning FBN1 at 15q21.1
- Author
-
Ralph J. Johnson, Mary Nguyen, Scott A. LeMaire, Ludivine Russell, Luis M. Franco, Christine E. Seidman, Reed E. Pyeritz, Richard B. Devereux, Merry-Lynn McDonald, Suzanne M. Leal, Dongchuan Guo, Molly S. Bray, Joseph E. Bavaria, Kim A. Eagle, Jon G. Seidman, John W. Belmont, Dianna M. Milewicz, Mir Reza Bekheirnia, Kathryn W. Holmes, Cheryl L. Maslen, Charles C. Miller, Anthony L. Estrera, Simon C. Body, Joseph S. Coselli, Hazim J. Safi, and Eric M. Isselbacher
- Subjects
Marfan syndrome ,musculoskeletal diseases ,Linkage disequilibrium ,congenital, hereditary, and neonatal diseases and abnormalities ,Genotyping Techniques ,Fibrillin-1 ,Single-nucleotide polymorphism ,Genome-wide association study ,Biology ,Bioinformatics ,Fibrillins ,Polymorphism, Single Nucleotide ,Linkage Disequilibrium ,Article ,Marfan Syndrome ,Aortic aneurysm ,Genetics ,medicine ,Genetic predisposition ,Humans ,Genetic Predisposition to Disease ,Chromosomes, Human, Pair 15 ,Aortic Aneurysm, Thoracic ,Microfilament Proteins ,Case-control study ,Odds ratio ,DNA ,Sequence Analysis, DNA ,medicine.disease ,Genetic Loci ,Case-Control Studies ,Mutation ,Genome-Wide Association Study - Abstract
Although thoracic aortic aneurysms and dissections (TAAD) can be inherited as a single-gene disorder, the genetic predisposition in the majority of affected people is poorly understood. In a multistage genome-wide association study (GWAS), we compared 765 individuals who had sporadic TAAD (STAAD) with 874 controls and identified common SNPs at a 15q21.1 locus that were associated with STAAD, with odds ratios of 1.6-1.8 that achieved genome-wide significance. We followed up 107 SNPs associated with STAAD with P < 1 × 10(-5) in the region, in two separate STAAD cohorts. The associated SNPs fall into a large region of linkage disequilibrium encompassing FBN1, which encodes fibrillin-1. FBN1 mutations cause Marfan syndrome, whose major cardiovascular complication is TAAD. This study shows that common genetic variants at 15q21.1 that probably act via FBN1 are associated with STAAD, suggesting a common pathogenesis of aortic disease in Marfan syndrome and STAAD.
- Published
- 2011
34. Loss-of-function mutations in **PTPN11** cause metachondromatosis, but not Ollier disease or Maffucci syndrome
- Author
-
Matthew L. Warman, Luisa Bonafé, Eric D. Boyden, Belinda Campos-Xavier, Kyle C. Kurek, Elena Pedrini, Judith V.M.G. Bovée, Ravi Savarirayan, Livia Garavelli, Harry P.W. Kozakewich, Elena Andreucci, Jon G. Seidman, Wim Wuyts, Ingrid A. Holm, Bianca M. Regazzoni, Valérie Cormier-Daire, James R. Kasser, Andrea Superti-Furga, Sérgio B. Sousa, Margot E. Bowen, Miikka Vikkula, Mei Zhu, Caroline Pottinger, Shiro Ikegawa, Twinkal C. Pansuriya, Toshihiko Ogino, Luca Sangiorgi, Akinori Sakai, and UCL - SSS/DDUV - Institut de Duve
- Subjects
Cancer Research ,Genetic Linkage ,Gene Identification and Analysis ,Loss of Heterozygosity ,Protein Tyrosine Phosphatase, Non-Receptor Type 11 ,Pediatrics ,0302 clinical medicine ,Chromosomes, Human ,Ollier disease ,Genetics (clinical) ,Sanger sequencing ,Genetics ,0303 health sciences ,Massive parallel sequencing ,High-Throughput Nucleotide Sequencing ,Enchondromatosis ,Exons ,3. Good health ,Pedigree ,Maffucci syndrome ,Autosomal Dominant ,030220 oncology & carcinogenesis ,symbols ,Medicine ,Metachondromatosis ,Exostoses, Multiple Hereditary ,Research Article ,Pediatric Orthopedics ,musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,lcsh:QH426-470 ,DNA Copy Number Variations ,Nonsense mutation ,Biology ,Polymorphism, Single Nucleotide ,Frameshift mutation ,Molecular Genetics ,03 medical and health sciences ,symbols.namesake ,Cancer Genetics ,medicine ,Humans ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,Clinical Genetics ,Human Genetics ,Sequence Analysis, DNA ,medicine.disease ,Molecular biology ,lcsh:Genetics ,Pediatric Oncology ,Mutation ,Human medicine ,Gene Deletion - Abstract
Metachondromatosis (MC) is a rare, autosomal dominant, incompletely penetrant combined exostosis and enchondromatosis tumor syndrome. MC is clinically distinct from other multiple exostosis or multiple enchondromatosis syndromes and is unlinked to EXT1 and EXT2, the genes responsible for autosomal dominant multiple osteochondromas (MO). To identify a gene for MC, we performed linkage analysis with high-density SNP arrays in a single family, used a targeted array to capture exons and promoter sequences from the linked interval in 16 participants from 11 MC families, and sequenced the captured DNA using high-throughput parallel sequencing technologies. DNA capture and parallel sequencing identified heterozygous putative loss-of-function mutations in PTPN11 in 4 of the 11 families. Sanger sequence analysis of PTPN11 coding regions in a total of 17 MC families identified mutations in 10 of them (5 frameshift, 2 nonsense, and 3 splice-site mutations). Copy number analysis of sequencing reads from a second targeted capture that included the entire PTPN11 gene identified an additional family with a 15 kb deletion spanning exon 7 of PTPN11. Microdissected MC lesions from two patients with PTPN11 mutations demonstrated loss-of-heterozygosity for the wild-type allele. We next sequenced PTPN11 in DNA samples from 54 patients with the multiple enchondromatosis disorders Ollier disease or Maffucci syndrome, but found no coding sequence PTPN11 mutations. We conclude that heterozygous loss-of-function mutations in PTPN11 are a frequent cause of MC, that lesions in patients with MC appear to arise following a “second hit,” that MC may be locus heterogeneous since 1 familial and 5 sporadically occurring cases lacked obvious disease-causing PTPN11 mutations, and that PTPN11 mutations are not a common cause of Ollier disease or Maffucci syndrome., Author Summary Children with cartilage tumor syndromes form multiple tumors of cartilage next to joints. These tumors can occur inside the bones, as with Ollier disease and Maffuci syndrome, or on the surface of bones, as in the Multiple Osteochondroma syndrome (MO). In a hybrid syndrome, called metachondromatosis (MC), patients develop tumors both on and within bones. Only the genes causing MO are known. Since MC is inherited, we studied genetic markers in an affected family and found a region of the genome, encompassing 100 genes, always passed on to affected members. Using a recently developed method, we captured and sequenced all 100 genes in multiple families and found mutations in one gene, PTPN11, in 11 of 17 families. Patients with MC have one mutant copy of PTPN11 from their affected parent and one normal copy from their unaffected parent in all cells. We found that the normal copy is additionally lost in cartilage cells that form tumors, giving rise to cells without PTPN11. Mutations in PTPN11 were not found in other cartilage tumor syndromes, including Ollier disease and Maffucci syndrome. We are currently working to understand how loss of PTPN11 in cartilage cells causes tumors to form.
- Published
- 2011
35. Familial Dilated Cardiomyopathy caused by an Alpha-Tropomyosin Mutation: The Distinctive Natural History of Sarcomeric DCM
- Author
-
Elizabeth Sparks, Jon G. Seidman, Birgit H. Funke, Steven D. Colan, Allison L. Cirino, Carolyn Y. Ho, Lisa Dellefave, Elizabeth M. McNally, Paul Robinson, Neal K. Lakdawala, C Redwood, Christine E. Seidman, Steve Depalma, and Hugh Watkins
- Subjects
Adult ,Cardiomyopathy, Dilated ,Male ,Sarcomeres ,medicine.medical_specialty ,Adolescent ,genetic structures ,Familial dilated cardiomyopathy ,Tropomyosin ,Myosins ,Alpha-Tropomyosin ,complex mixtures ,Sarcomere ,Article ,Internal medicine ,Humans ,Medicine ,cardiovascular diseases ,Child ,Aged ,Genetics ,Ventricular Remodeling ,business.industry ,Age Factors ,Infant ,Middle Aged ,musculoskeletal system ,Phenotype ,Troponin ,Natural history ,Child, Preschool ,Mutation ,Mutation (genetic algorithm) ,cardiovascular system ,Cardiology ,Calcium ,Female ,Cardiology and Cardiovascular Medicine ,business - Abstract
We sought to further define the role of sarcomere mutations in dilated cardiomyopathy (DCM) and associated clinical phenotypes.Mutations in several contractile proteins contribute to DCM, but definitive evidence for the roles of most sarcomere genes remains limited by the lack of robust genetic support.Direct sequencing of 6 sarcomere genes was performed on 334 probands with DCM. A novel D230N missense mutation in the gene encoding alpha-tropomyosin (TPM1) was identified. Functional assessment was performed by the use of an in vitro reconstituted sarcomere complex to evaluate ATPase regulation and Ca(2+) affinity as correlates of contractility.TPM1 D230N segregated with DCM in 2 large unrelated families. This mutation altered an evolutionarily conserved residue and was absent in1,000 control chromosomes. In vitro studies demonstrated major inhibitory effects on sarcomere function with reduced Ca(2+) sensitivity, maximum activation, and Ca(2+) affinity compared with wild-type TPM1. Clinical manifestations ranged from decompensated heart failure or sudden death in those presenting early in life to asymptomatic left ventricular dysfunction in those diagnosed during adulthood. Notably, several affected infants had remarkable improvement.Genetic segregation in 2 unrelated families and functional analyses conclusively establish a pathogenic role for TPM1 mutations in DCM. In vitro results demonstrate contrasting effects of DCM and hypertrophic cardiomyopathy mutations in TPM1, suggesting that specific functional consequences shape cardiac remodeling. Along with previous reports, our data support a distinctive, age-dependent phenotype with sarcomere-associated DCM where presentation early in life is associated with severe, sometimes lethal, disease. These observations have implications for the management of familial DCM.
- Published
- 2010
36. Regional localization of the human G protein αi2 (GNAI2) gene: Assignment to 3p21 and a related sequence (GNAI2L) to 12p12–p13
- Author
-
Jon G. Seidman, Cynthia C. Morton, Ivana Magovcevic, Cynthia Tolman, Siew-Lan Ang, and Eva J. Neer
- Subjects
Molecular Sequence Data ,Hybrid Cells ,Biology ,Polymerase Chain Reaction ,Mice ,Gene mapping ,GTP-Binding Proteins ,Chromosome regions ,Genetics ,Animals ,Humans ,Chromosome 12 ,Chromosome 7 (human) ,Chromosomes, Human, Pair 12 ,Base Sequence ,Chromosome Mapping ,Nucleic Acid Hybridization ,DNA ,Molecular biology ,Chromosome Banding ,Chromosome 17 (human) ,Chromosome 3 ,Chromosomes, Human, Pair 3 ,Peptides ,Chromosome 21 ,Chromosome 22 - Abstract
Gi alpha proteins, members of the G protein signal transduction family, include a small number of polypeptides: Gi alpha 1 (GNAI1), Gi alpha 2 (GNAI2), and Gi alpha 3 (GNAI3). A cDNA for the human GNAI2 gene has been isolated from a human T-cell library and is mapped by chromosomal in situ hybridization to the short arm of chromosome 3 at 3p21. A related sequence, GNAI2L, is mapped by in situ hybridization to the short arm of chromosome 12 at p12-p13. These mapping results are further supported by amplification of GNAI2-specific sequences in a monochromosomal human/rodent somatic cell hybrid containing only human chromosome 3. Of note, these assignments are to chromosome regions in which other G proteins reside. Localization of GNAI2 to 3p21 is of great interest as this region of the short arm of chromosome 3 is frequently involved in rearrangements in various human tumors.
- Published
- 1992
- Full Text
- View/download PDF
37. Sequencing of TGF-β pathway genes in familial cases of intracranial aneurysm
- Author
-
Sumy Mathew-Joseph, Hariyadarshi Pannu, Christine E. Seidman, Dianna M. Milewicz, Jon G. Seidman, Dong H. Kim, and Teresa Santiago-Sim
- Subjects
Male ,Receptor, Transforming Growth Factor-beta Type I ,Single-nucleotide polymorphism ,Receptors, Cell Surface ,Protein Serine-Threonine Kinases ,Polymorphism, Single Nucleotide ,Article ,Pathogenesis ,Antigens, CD ,Transforming Growth Factor beta ,Medicine ,Humans ,Allele ,Allele frequency ,Gene ,Alleles ,Aged ,DNA Primers ,Advanced and Specialized Nursing ,Genetics ,biology ,business.industry ,Endoglin ,Receptor, Transforming Growth Factor-beta Type II ,Family aggregation ,Intracranial Aneurysm ,Transforming growth factor beta ,DNA ,Middle Aged ,Pedigree ,Stroke ,Immunology ,biology.protein ,Female ,Proteoglycans ,Neurology (clinical) ,Cardiology and Cardiovascular Medicine ,business ,Activin Receptors, Type I ,Receptors, Transforming Growth Factor beta ,Signal Transduction - Abstract
Background and Purpose— Familial aggregation of intracranial aneurysms (IA) strongly suggests a genetic contribution to pathogenesis. However, genetic risk factors have yet to be defined. For families affected by aortic aneurysms, specific gene variants have been identified, many affecting the receptors to transforming growth factor-beta (TGF-β). In recent work, we found that aortic and intracranial aneurysms may share a common genetic basis in some families. We hypothesized, therefore, that mutations in TGF-β receptors might also play a role in IA pathogenesis. Methods— To identify genetic variants in TGF-β and its receptors, TGFB1 , TGFBR1 , TGFBR2 , ACVR1 , TGFBR3 , and ENG were directly sequenced in 44 unrelated patients with familial IA. Novel variants were confirmed by restriction digestion analyses, and allele frequencies were analyzed in cases versus individuals without known intracranial disease. Similarly, allele frequencies of a subset of known SNPs in each gene were also analyzed for association with IA. Results— No mutations were found in TGFB1 , TGFBR1 , TGFBR2 , or ACVR1 . Novel variants identified in ENG (p.A60E) and TGFBR3 (p.W112R) were not detected in at least 892 reference chromosomes. ENG p.A60E showed significant association with familial IA in case-control studies ( P =0.0080). No association with IA could be found for any of the known polymorphisms tested. Conclusions— Mutations in TGF-β receptor genes are not a major cause of IA. However, we identified rare variants in ENG and TGFBR3 that may be important for IA pathogenesis in a subset of families.
- Published
- 2009
38. Familial hypertrophic cardiomyopathy is a genetically heterogeneous disease
- Author
-
Anja Geisterfer-Lowrance, R Germain, Jon G. Seidman, John A. Jarcho, R Salerni, Christine E. Seidman, Scott D. Solomon, and William J. McKenna
- Subjects
medicine.medical_specialty ,Genetic Linkage ,Locus (genetics) ,Disease ,Genetic linkage ,Humans ,Medicine ,cardiovascular diseases ,Gene ,Genes, Dominant ,Chromosomes, Human, Pair 14 ,Genetics ,Genetic heterogeneity ,business.industry ,Cytogenetics ,Hypertrophic cardiomyopathy ,Autosomal dominant trait ,General Medicine ,Cardiomyopathy, Hypertrophic ,medicine.disease ,Pedigree ,Echocardiography ,Lod Score ,business ,Polymorphism, Restriction Fragment Length ,Research Article - Abstract
We demonstrate that familial hypertrophic cardiomyopathy (FHC), an autosomal dominant disorder of heart muscle, is a genetically heterogeneous disease. The locus responsible for FHC in members of one large kindred was recently mapped to chromosome 14q11-12 (FHC-1). We have characterized three additional unrelated families in which the gene for FHC segregates as an autosomal dominant trait to determine if these disease loci also map to FHC-1. All family members were clinically studied by physical examination, electrocardiogram, and two-dimensional echocardiography. Genetic studies were performed using DNA probes which are derived from loci that are closely linked to FHC-1. In one family the genetic defect maps to the previously identified FHC-1 locus. However, the loci responsible for FHC in two other families were not linked to FHC-1. We conclude that FHC can be caused by defects in at least two loci and is a genetically heterogeneous disorder.
- Published
- 1990
- Full Text
- View/download PDF
39. Corrigendum to 'Tbx5-dependent rheostatic control of cardiac gene expression and morphogenesis' [Dev. Biol. 297 (2006) 566–586]
- Author
-
Allesandro D. Mori, Benoit G. Bruneau, Jon G. Seidman, Yonghong Zhu, Ilyas Vahora, Lorinda Davidson, Christine E. Seidman, R. Mark Henkelman, Brian J. Nieman, Kazuko Koshiba-Takeuchi, X. Josette Chen, and Anne Pizard
- Subjects
Genetics ,TheoryofComputation_COMPUTATIONBYABSTRACTDEVICES ,GeneralLiterature_INTRODUCTORYANDSURVEY ,Gene expression ,Morphogenesis ,ComputingMilieux_LEGALASPECTSOFCOMPUTING ,Data_CODINGANDINFORMATIONTHEORY ,Cell Biology ,Biology ,ComputingMethodologies_ARTIFICIALINTELLIGENCE ,Molecular Biology ,Developmental Biology - Published
- 2007
- Full Text
- View/download PDF
40. Functional role of AMP-activated protein kinase in the heart during exercise
- Author
-
Nicolas Musi, Nobuharu L. Fujii, Rong Tian, Jason M. Pomerleau, Ferhaan Ahmad, Jon G. Seidman, Michael Arad, Charles I. Berul, Yanqiu Xing, Laurie J. Goodyear, and Michael F. Hirshman
- Subjects
Cardiac function curve ,medicine.medical_specialty ,Biophysics ,p38 ,Mice, Transgenic ,AMP-Activated Protein Kinases ,Protein Serine-Threonine Kinases ,Biochemistry ,p38 Mitogen-Activated Protein Kinases ,Energy homeostasis ,Mice ,AMP-activated protein kinase ,Structural Biology ,Multienzyme Complexes ,Internal medicine ,Physical Conditioning, Animal ,Genetics ,medicine ,Stress Echocardiography ,Animals ,Exercise physiology ,Phosphorylation ,Protein kinase A ,Exercise ,Molecular Biology ,biology ,Acetyl CoA carboxylase ,Myocardium ,AMPK ,Heart ,Cell Biology ,Cardiovascular physiology ,Endocrinology ,Echocardiography ,biology.protein - Abstract
AMP-activated protein kinase (AMPK) plays a critical role in maintaining energy homeostasis and cardiac function during ischemia in the heart. However, the functional role of AMPK in the heart during exercise is unknown. We examined whether acute exercise increases AMPK activity in mouse hearts and determined the significance of these increases by studying transgenic (TG) mice expressing a cardiac-specific dominant-negative (inactivating) AMPKα2 subunit. Exercise increased cardiac AMPKα2 activity in the wild type mice but not in TG. We found that inactivation of AMPK did not result in abnormal ATP and glycogen consumption during exercise, cardiac function assessed by heart rhythm telemetry and stress echocardiography, or in maximal exercise capacity.
- Published
- 2004
41. Electrophysiologic characterization and postnatal development of ventricular pre-excitation in a mouse model of cardiac hypertrophy and Wolff-Parkinson-White syndrome
- Author
-
Charles I. Berul, Dorothy Branco, Ivan P. Moskowitz, Vickas V. Patel, Michael Arad, Jon G. Seidman, Christine E. Seidman, and Colin T. Maguire
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Adenosine ,Pre-Excitation Syndromes ,Heart disease ,Genotype ,Biopsy ,Mutation, Missense ,Cardiomegaly ,Mice, Transgenic ,Accessory pathway ,AMP-Activated Protein Kinases ,Procainamide ,Protein Serine-Threonine Kinases ,Muscle hypertrophy ,Electrocardiography ,Mice ,Animal model ,Heart Conduction System ,Multienzyme Complexes ,Internal medicine ,Ventricular Dysfunction ,Medicine ,Animals ,Single-Blind Method ,cardiovascular diseases ,medicine.diagnostic_test ,business.industry ,Age Factors ,medicine.disease ,Multienzyme complexes ,Electrophysiology ,Disease Models, Animal ,Endocrinology ,Phenotype ,Cardiology ,Disease Progression ,Wolff-Parkinson-White Syndrome ,Electrical conduction system of the heart ,Cardiology and Cardiovascular Medicine ,business ,Electrophysiologic Techniques, Cardiac ,Anti-Arrhythmia Agents ,Pre-excitation syndrome - Abstract
ObjectivesWe sought to characterize an animal model of the Wolff-Parkinson-White (WPW) syndrome to help elucidate the mechanisms of accessory pathway formation.BackgroundPatients with mutations in PRKAG2manifest cardiac hypertrophy and ventricular pre-excitation; however, the mechanisms underlying the development and conduction of accessory pathways remain unknown.MethodsWe created transgenic mice overexpressing either the Asn488Ile mutant (TGN488I) or wild-type (TGWT) human PRKAG2complementary deoxyribonucleic acid under a cardiac-specific promoter. Both groups of transgenic mice underwent intracardiac electrophysiologic, electrocardiographic (ECG), and histologic analyses.ResultsOn the ECG, ∼50% of TGN488Imice displayed sinus bradycardia and features suggestive of pre-excitation, not seen in TGWTmice. The electrophysiologic studies revealed a distinct atrioventricular (AV) connection apart from the AV node, using programmed stimulation. In TGN488Imice with pre-excitation, procainamide blocked bypass tract conduction, whereas adenosine infusion caused AV block in TGWTmice but not TGN488Imice with pre-excitation. Serial ECGs in 16 mice pups revealed no differences at birth. After one week, two of eight TGN488Ipups had ECG features of pre-excitation, increasing to seven of eight pups by week 4. By nine weeks, one TGN488Imouse with WPW syndrome lost this phenotype, whereas TGWTpups never developed pre-excitation. Histologic investigation revealed postnatal development of myocardial connections through the annulus fibrosum of the AV valves in young TGN488Ibut not TGWTmice.ConclusionsTransgenic mice overexpressing the Asn488Ile PRKAG2mutation recapitulate an electrophysiologic phenotype similar to humans with this mutation. This includes procainamide-sensitive, adenosine-resistant accessory pathways induced in postnatal life that may rarely disappear later in life.
- Published
- 2003
42. Assessment of diastolic function with Doppler tissue imaging to predict genotype in preclinical hypertrophic cardiomyopathy
- Author
-
Nancy K. Sweitzer, Christine E. Seidman, Barry J. Maron, Scott D. Solomon, Jon G. Seidman, Barbara McDonough, Susan A. Casey, and Carolyn Y. Ho
- Subjects
Adult ,Male ,medicine.medical_specialty ,Pathology ,Heart disease ,Genotype ,Population ,Diastole ,Cardiomyopathy ,Left ventricular hypertrophy ,Electrocardiography ,Physiology (medical) ,Internal medicine ,Cardiomyopathy, Hypertrophic, Familial ,Medicine ,Humans ,cardiovascular diseases ,education ,education.field_of_study ,Ejection fraction ,Myosin Heavy Chains ,business.industry ,Hypertrophic cardiomyopathy ,medicine.disease ,Echocardiography, Doppler ,Mutation ,Cardiology ,Female ,Cardiology and Cardiovascular Medicine ,business - Abstract
Background — Unexplained left ventricular hypertrophy (LVH) is considered diagnostic of hypertrophic cardiomyopathy (HCM) but fails to identify all genetically affected individuals. Altered diastolic function has been hypothesized to represent an earlier manifestation of HCM before the development of LVH; however, data regarding the clinical utility of imaging techniques that assess this parameter are limited. Methods and Results — Echocardiographic studies including Doppler tissue imaging (DTI) were performed in a genotyped HCM population with β-myosin heavy chain (β -MHC) mutations. Genotype (+) individuals with LVH (G+/LVH+; n=18) and genotype (+) individuals without LVH (G+/LVH−; n=18) were compared with normal control subjects (n=36). Left ventricular ejection fraction (EF) was significantly higher in both genotype (+) groups (75±5% and 71±6%, respectively, versus 64± 5% in control subjects; P P Conclusions — Abnormalities of diastolic function assessed by Doppler tissue imaging precede the development of LVH in individuals with HCM caused by β -MHC mutations. Although Ea velocity alone was not sufficiently sensitive as a sole diagnostic criterion, the combination of Ea velocity and EF was highly predictive of affected genotype in individuals without overt manifestations of HCM.
- Published
- 2002
43. A novel lamin A/C missense mutation in a family with autosomal dominant dilated cardiomyopathy with conduction abnormalities
- Author
-
Ryuichiro Anan, Takeshi Sasaki, Christine E. Seidman, Chuwa Tei, Jon G. Seidman, Hideshi Niimura, and Shinichi Minagoe
- Subjects
Pathology ,medicine.medical_specialty ,Conduction abnormalities ,business.industry ,medicine ,cardiovascular system ,Missense mutation ,Dilated cardiomyopathy ,medicine.disease ,business ,Cardiology and Cardiovascular Medicine ,Lamin - Published
- 2002
- Full Text
- View/download PDF
44. An abnormal Ca(2+) response in mutant sarcomere protein-mediated familial hypertrophic cardiomyopathy
- Author
-
Christopher Semsarian, Christine E. Seidman, Frederick J. Schoen, Ivan G.P. Moskowitz, James O. Mudd, Bradley K. McConnell, Jon G. Seidman, Michael Giewat, and Diane Fatkin
- Subjects
Sarcomeres ,medicine.medical_specialty ,Heart Ventricles ,Calcineurin Inhibitors ,Gene mutation ,Biology ,medicine.disease_cause ,Sarcomere ,Sudden death ,Article ,Tacrolimus ,Muscle hypertrophy ,Mice ,Internal medicine ,Myosin ,medicine ,Myocyte ,Humans ,Animals ,cardiovascular diseases ,Calcium Signaling ,Enzyme Inhibitors ,Mice, Knockout ,Mutation ,Myosin Heavy Chains ,Calcineurin ,Molecular Motor Proteins ,Muscles ,General Medicine ,Cardiomyopathy, Hypertrophic ,Survival Analysis ,Disease Models, Animal ,Endocrinology ,Echocardiography ,Cyclosporine ,Minoxidil ,Commentary ,Calcium - Abstract
Dominant-negative sarcomere protein gene mutations cause familial hypertrophic cardiomyopathy (FHC), a disease characterized by left-ventricular hypertrophy, angina, and dyspnea that can result in sudden death. We report here that a murine model of FHC bearing a cardiac myosin heavy-chain gene missense mutation (alphaMHC(403/+)), when treated with calcineurin inhibitors or a K(+)-channel agonist, developed accentuated hypertrophy, worsened histopathology, and was at risk for early death. Despite distinct pharmacologic targets, each agent augmented diastolic Ca(2+) concentrations in wild-type cardiac myocytes; alphaMHC(403/+) myocytes failed to respond. Pretreatment with a Ca(2+)-channel antagonist abrogated diastolic Ca(2+) changes in wild-type myocytes and prevented the exaggerated hypertrophic response of treated alphaMHC(403/+) mice. We conclude that FHC-causing sarcomere protein gene mutations cause abnormal Ca(2+) responses that initiate a hypertrophic response. These data define an important Ca(2+)-dependent step in the pathway by which mutant sarcomere proteins trigger myocyte growth and remodel the heart, provide definitive evidence that environment influences progression of FHC, and suggest a rational therapeutic approach to this prevalent human disease.
- Published
- 2000
45. Dilated cardiomyopathy in homozygous myosin-binding protein-C mutant mice
- Author
-
L. H. Arroyo, Hideshi Niimura, David A. Kass, Karen A. Jones, Diane Fatkin, David A. Conner, Orlando Aristizabal, Bradley K. McConnell, Daniel H. Turnbull, C. R. Seidman, Frederick J. Schoen, Richard T. Lee, Dimitrios Georgakopoulos, Jon G. Seidman, Meredith Bond, and D. A. Fischman
- Subjects
medicine.medical_specialty ,Myosin-binding protein C ,Internal medicine ,Clinical investigation ,Mutant ,medicine ,Cardiology ,Dilated cardiomyopathy ,General Medicine ,Biology ,medicine.disease ,Corrigendum ,Molecular biology - Published
- 1999
46. Altered crossbridge kinetics in the alphaMHC403/+ mouse model of familial hypertrophic cardiomyopathy
- Author
-
Martin M. LeWinter, Christine E. Seidman, David W. Maughan, Jon G. Seidman, and E. M. Blanchard
- Subjects
Male ,medicine.medical_specialty ,Physiology ,Cardiomyopathy ,chemistry.chemical_element ,Isometric exercise ,Calcium ,Myosins ,Mice ,CrossBridge ,Internal medicine ,Myosin ,medicine ,Animals ,Papillary muscle ,Myosin Heavy Chains ,Chemistry ,Viscosity ,Myocardium ,Hypertrophic cardiomyopathy ,Cardiomyopathy, Hypertrophic ,Papillary Muscles ,medicine.disease ,Myocardial Contraction ,Elasticity ,Mice, Mutant Strains ,Kinetics ,Endocrinology ,medicine.anatomical_structure ,Circulatory system ,Mutation ,Cardiology and Cardiovascular Medicine - Abstract
Abstract —A mutation in the cardiac β-myosin heavy chain, Arg403Gln (R403Q), causes a severe form of familial hypertrophic cardiomyopathy (FHC) in humans. We used small-amplitude (0.25%) length-perturbation analysis to examine the mechanical properties of skinned left ventricular papillary muscle strips from mouse hearts bearing the R403Q mutation in the α-myosin heavy chain (αMHC 403/+ ). Myofibrillar disarray with variable penetrance occurred in the left ventricular free wall of the αMHC 403/+ hearts. In resting strips (pCa 8), dynamic stiffness was ≈40% greater than in wild-type strips, consistent with elevated diastolic stiffness reported for murine hearts with FHC. At pCa 6 (submaximal activation), strip isometric tension was ≈3 times higher than for wild-type strips, whereas at pCa 5 (maximal activation), tension was marginally lower. At submaximal calcium activation the characteristic frequencies of the work-producing ( b ) and work-absorbing ( c ) steps of the crossbridge were less in αMHC 403/+ strips than in wild-type strips ( b =11±1 versus 15±1 Hz; c = 58±3 versus 66±3 Hz; 27°C). At maximal calcium activation, strip oscillatory power was reduced (0.53±0.25 versus 1.03±0.18 mW/mm 3 ; 27°C), which is partly attributable to the reduced frequency b , at which crossbridge work is maximum. The results are consistent with the hypothesis that the R403Q mutation reduces the strong binding affinity of myosin for actin. Myosin heads may accumulate in a preforce state that promotes cooperative activation of the thin filament at submaximal calcium but blunts maximal tension and oscillatory power output at maximal calcium. The calcium-dependent effect of the mutation (whether facilitating or debilitating), together with a variable degree of fibrosis and myofibrillar disorder, may contribute to the diversity of clinical symptoms observed in murine FHC.
- Published
- 1999
47. beta-Arrestin1 knockout mice appear normal but demonstrate altered cardiac responses to beta-adrenergic stimulation
- Author
-
Jon G. Seidman, Michael A. Mathier, Michael E. Christe, Christine E. Seidman, Stephen F. Vatner, Richard M. Mortensen, and David A. Conner
- Subjects
Male ,medicine.medical_specialty ,Physiology ,G protein ,Arrestins ,medicine.medical_treatment ,Adrenergic ,Blood Pressure ,Biology ,Mice ,Cell surface receptor ,GTP-Binding Proteins ,Heart Rate ,Internal medicine ,Receptors, Adrenergic, beta ,medicine ,Animals ,Receptor ,beta-Arrestins ,Desensitization (medicine) ,G protein-coupled receptor ,Mice, Knockout ,Beta-Arrestins ,Chromosome Mapping ,Endocrinology ,Knockout mouse ,Gene Targeting ,Cardiology and Cardiovascular Medicine - Abstract
Abstract β-Arrestin1 knockout mice were studied to define the physiological role of β-arrestin1 in the regulation of G protein–coupled receptors. β-Arrestin1 is thought to be involved in the desensitization of many G protein–associated cell surface receptors, particularly β-adrenergic receptors. Homozygous knockout mice are overtly normal. Resting cardiovascular parameters modulated by β-adrenergic receptors such as heart rate, blood pressure, and left ventricular ejection fraction are not changed. However, homozygous mutants are more sensitive to β-receptor agonist–stimulated increases in ejection fraction, consistent with a role of β-arrestin1 in β-adrenergic receptor desensitization. We conclude that β-arrestin1 is important for in vivo G protein–coupled receptor desensitization and that this aspect of desensitization represents a mechanism for fine-tuning responses. However, β-arrestin1 does not appear to be required for development or for other essential biological functions.
- Published
- 1997
48. The cloning of extracellular Ca(2+)-sensing receptors from parathyroid and kidney: molecular mechanisms of extracellular Ca(2+)-sensing
- Author
-
Christine E. Seidman, Edward M. Brown, Jon G. Seidman, Martin R. Pollak, Steven C. Hebert, and Ya-Huei Wu Chou
- Subjects
Medicine (miscellaneous) ,Receptors, Cell Surface ,Disorders of calcium metabolism ,Biology ,Kidney ,Parathyroid Glands ,medicine ,Extracellular ,Animals ,Homeostasis ,Cloning, Molecular ,Receptor ,Calcium metabolism ,Hyperparathyroidism ,Nutrition and Dietetics ,Familial hypocalciuric hypercalcemia ,Parathyroid chief cell ,medicine.disease ,Molecular biology ,Biochemistry ,Mutation ,Oocytes ,Calcium ,Calcium-sensing receptor ,Metabolism, Inborn Errors ,Signal Transduction - Abstract
The parathyroid cell detects changes in the extracellular ionized calcium concentration (Ca o 2+ ) with exquisite sensitivity, but the mechanisms through which it senses Ca o 2+ have remained obscure. Recently, we isolated a cDNA encoding a Ca o 2+ -sensing receptor from bovine parathyroid using expression cloning in Xenopus laevis oocytes. The expressed receptor stimulates phospholipase C and has a pharmacological profile almost identical to that of the native receptor. Furthermore, its deduced amino acid sequence confirms that it belongs to the superfamily of G-protein-coupled receptors. Receptor transcripts are present in parathyroid and other tissues sensing Ca o 2+ (e.g., kidney and thyroidal C-cells) as well as those not known to be involved in Ca o 2+ homeostasis (viz., in the brain). We have also shown that mutations in the receptor cause three inherited disorders of calcium metabolism : Familial hypocalciuric hypercalcemia (FHH) and neonatal severe hyperparathyroidism (NSHPT) result from inactivating mutations, when present in the heterozygous and homozygous states, respectively, whereas an autosomal dominant form of hypocalcemia is due to an activating mutation. Thus this Ca o 2+ -sensing receptor permits Ca o 2+ to act as an extracellular, first messenger in addition to its better known role as an intracellular second messenger
- Published
- 1995
49. A de novo mutation in alpha-tropomyosin that causes hypertrophic cardiomyopathy
- Author
-
Ryuichiro Anan, Christine E. Seidman, Hugh Watkins, Domenico A. Coviello, Paolo Spirito, and Jon G. Seidman
- Subjects
Proband ,Genetics ,Adult ,Male ,Adolescent ,Offspring ,Haplotype ,Cardiomyopathy ,Hypertrophic cardiomyopathy ,Tropomyosin ,Biology ,Cardiomyopathy, Hypertrophic ,Middle Aged ,medicine.disease ,Pedigree ,Centimorgan ,Haplotypes ,Physiology (medical) ,Mutation (genetic algorithm) ,Mutation ,medicine ,Missense mutation ,Humans ,Female ,Cardiology and Cardiovascular Medicine - Abstract
Background Two missense mutations in the gene for α-tropomyosin have been described that segregate with hypertrophic cardiomyopathy in single families. To confirm that these mutations are the cause of the disease, we have investigated the origins of one of these mutations, Asp175Asn, in a third and unrelated family. Methods and Results The presence or absence of an α-tropomyosin mutation and the haplotypes of the flanking chromosomal regions were determined for members of a family with hypertrophic cardiomyopathy. Haplotypes were constructed by use of an intragenic polymorphism and 10 flanking polymorphisms spanning a region of 35 centimorgans. The Asp175Asn missense mutation was present in the proband and his two affected offspring but not in any of the proband’s three siblings. Although both parents were deceased, the haplotypes of the four parental chromosomes could be reconstructed. One parental chromosome was transmitted to two offspring: one bearing the Asp175Asn mutation (the affected proband) and one clinically unaffected sibling who lacked the α-tropomyosin mutation. Thus, the Asp175Asn mutation must have arisen de novo. Conclusions De novo mutations in the α-tropomyosin gene can result in hypertrophic cardiomyopathy that may appear to be sporadic but in subsequent generations gives rise to familial disease. Individuals with sporadic hypertrophic cardiomyopathy should be advised of the risk of transmission to offspring. In addition, these findings provide the strongest genetic evidence that mutations in the α-tropomyosin gene are directly responsible for hypertrophic cardiomyopathy.
- Published
- 1995
50. Cloning, expression, and chromosomal location of SHH and IHH: two human homologues of the Drosophila segment polarity gene hedgehog
- Author
-
Clifford J. Tabin, Olga Tsukurov, Debra J. Gilbert, Christine E. Seidman, Douglas J. Epstein, Drucilla J. Roberts, Neal G. Copeland, Julie M. Gastier, Jon G. Seidman, Tatjana Levi, Valeria Marigo, Scott M. K. Lee, Nancy A. Jenkins, and Andrew P. McMahon
- Subjects
DNA, Complementary ,Molecular Sequence Data ,Gene Expression ,Locus (genetics) ,Biology ,Sequence-tagged site ,espressione in situ ,Mice ,Fetus ,Gene mapping ,Complementary DNA ,Embryonic morphogenesis ,Genetics ,Animals ,Drosophila Proteins ,Humans ,Hedgehog Proteins ,Amino Acid Sequence ,Cloning, Molecular ,Gene ,Hedgehog ,In Situ Hybridization ,Base Sequence ,Chromosome Mapping ,Proteins ,Sonic hedgehog ,Molecular biology ,mappatura cromosomica ,Segment polarity gene ,Organ Specificity ,Chromosomes, Human, Pair 2 ,Protein Biosynthesis ,Indian hedgehog ,embryonic structures ,Drosophila ,Sequence Alignment ,Chromosomes, Human, Pair 7 - Abstract
The hedgehog genes encode signaling molecules that play a role in regulating embryonic morphogenesis. We have cloned and sequenced human cDNA copies of two of these genes, SHH and IHH. The SHH clone includes the full coding sequence and encodes a protein 92.4% identical to its murine homologue. The IHH clone is 89% complete and encodes a protein 94.6% identical to its murine homologue. IHH is expressed in adult kidney and liver. SHH expression was not detected in adult tissues examined; however, it is expressed in fetal intestine, liver, lung, and kidney. SHH mapped to chromosome 7q and IHH to chromosome 2 by PCR with DNA from a panel of rodent-human somatic cell hybrids. To identify the chromosomal location of SHH more precisely, a P1 genomic clone of SHH was isolated. This phage contained a CA repeat sequence tagged site that was used to map SHH relative to a polysyndactyly disease locus, using DNA prepared from affected and unaffected members of a large pedigree. SHH is closely linked, but distinct from the polysyndactyly disease locus at 7q36 (maximum lod score = 4.82, theta = 0.05) tightly linked to the EN2 locus. The murine homologues Shh, Ihh, and Dhh were mapped using (C57BL/6J x Mus spretus)F1 x C57BL/6J interspecific backcross. Shh mapped to a position 0.6 cM distal to En2 and 1.9 cM proximal to Il6 on mouse chromosome 5. This location is closely linked but distinct from the murine limb mutation Hx and syntenic to human chromosome 7q36.
- Published
- 1995
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.