Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
bioRxiv ; 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38915555

ABSTRACT

LMNA -Related Dilated Cardiomyopathy (DCM) is an autosomal-dominant genetic condition with cardiomyocyte and conduction system dysfunction often resulting in heart failure or sudden death. The condition is caused by mutation in the Lamin A/C ( LMNA ) gene encoding Type-A nuclear lamin proteins involved in nuclear integrity, epigenetic regulation of gene expression, and differentiation. Molecular mechanisms of disease are not completely understood, and there are no definitive treatments to reverse progression or prevent mortality. We investigated possible mechanisms of LMNA -Related DCM using induced pluripotent stem cells derived from a family with a heterozygous LMNA c.357-2A>G splice-site mutation. We differentiated one LMNA mutant iPSC line derived from an affected female (Patient) and two non-mutant iPSC lines derived from her unaffected sister (Control) and conducted single-cell RNA sequencing for 12 samples (4 Patient and 8 Control) across seven time points: Day 0, 2, 4, 9, 16, 19, and 30. Our bioinformatics workflow identified 125,554 cells in raw data and 110,521 (88%) high-quality cells in sequentially processed data. Unsupervised clustering, cell annotation, and trajectory inference found complex heterogeneity: ten main cell types; many possible subtypes; and lineage bifurcation for Cardiac Progenitors to Cardiomyocytes (CM) and Epicardium-Derived Cells (EPDC). Data integration and comparative analyses of Patient and Control cells found cell type and lineage differentially expressed genes (DEG) with enrichment to support pathway dysregulation. Top DEG and enriched pathways included: 10 ZNF genes and RNA polymerase II transcription in Pluripotent cells (PP); BMP4 and TGF Beta/BMP signaling, sarcomere gene subsets and cardiogenesis, CDH2 and EMT in CM; LMNA and epigenetic regulation and DDIT4 and mTORC1 signaling in EPDC. Top DEG also included: XIST and other X-linked genes, six imprinted genes: SNRPN , PWAR6 , NDN , PEG10 , MEG3 , MEG8 , and enriched gene sets in metabolism, proliferation, and homeostasis. We confirmed Lamin A/C haploinsufficiency by allelic expression and Western blot. Our complex Patient-derived iPSC model for Lamin A/C haploinsufficiency in PP, CM, and EPDC provided support for dysregulation of genes and pathways, many previously associated with Lamin A/C defects, such as epigenetic gene expression, signaling, and differentiation. Our findings support disruption of epigenomic developmental programs as proposed in other LMNA disease models. We recognized other factors influencing epigenetics and differentiation; thus, our approach needs improvement to further investigate this mechanism in an iPSC-derived model.

2.
Urology ; 185: 44-48, 2024 03.
Article in English | MEDLINE | ID: mdl-38101581

ABSTRACT

OBJECTIVE: To examine the efficacy, safety, and long-term durability of the autologous pubovaginal sling for stress incontinence over a 29-year period. METHODS: A total of 192 consecutive female patients with stress urinary incontinence who underwent autologous pubovaginal sling from 1993 through 1999 were analyzed over a 29-year period. Intermediate and ultra long-term follow-up were obtained at a mean of 4 and 23 years, respectively. A total of 51 patients had sufficient data at both time intervals and were evaluated using a standardized questionnaire for resolution of stress incontinence, the primary endpoint, as well as resolution of urge incontinence, overall dryness, and voiding dysfunction. RESULTS: At intermediate-term follow-up (mean 45.3 months), 96% of 51 patients reported no stress incontinence. In addition, 76% of patients experienced resolution of their preoperative urge incontinence. Overall dryness occurred in 84%. At ultra long-term follow-up (mean 22.9 years), 84% reported no stress incontinence and an overall dryness rate of 53%. Postoperative voiding dysfunction included de novo urge incontinence (3 patients) and persistent urinary obstruction requiring urethrolysis (1 patient). CONCLUSION: The autologous pubovaginal sling is effective, safe, and durable at a mean of 23 years, the longest known follow-up in the literature. Given the U.S. Food and Drug Administration (FDA) warnings regarding transvaginal mesh and growing concerns with the synthetic midurethral sling, the autologous pubovaginal sling should be offered as an option to those women seeking treatment for stress urinary incontinence.


Subject(s)
Urethral Diseases , Urinary Incontinence, Stress , Female , Humans , Follow-Up Studies , Urinary Incontinence, Stress/surgery , Urinary Incontinence, Urge , Prostheses and Implants
3.
Ann Biomed Eng ; 49(12): 3524-3539, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34585335

ABSTRACT

Genetic mutations to the Lamin A/C gene (LMNA) can cause heart disease, but the mechanisms making cardiac tissues uniquely vulnerable to the mutations remain largely unknown. Further, patients with LMNA mutations have highly variable presentation of heart disease progression and type. In vitro patient-specific experiments could provide a powerful platform for studying this phenomenon, but the use of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) introduces heterogeneity in maturity and function thus complicating the interpretation of the results of any single experiment. We hypothesized that integrating single cell RNA sequencing (scRNA-seq) with analysis of the tissue architecture and contractile function would elucidate some of the probable mechanisms. To test this, we investigated five iPSC-CM lines, three controls and two patients with a (c.357-2A>G) mutation. The patient iPSC-CM tissues had significantly weaker stress generation potential than control iPSC-CM tissues demonstrating the viability of our in vitro approach. Through scRNA-seq, differentially expressed genes between control and patient lines were identified. Some of these genes, linked to quantitative structural and functional changes, were cardiac specific, explaining the targeted nature of the disease progression seen in patients. The results of this work demonstrate the utility of combining in vitro tools in exploring heart disease mechanics.


Subject(s)
Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/physiopathology , Gene Expression , Induced Pluripotent Stem Cells/cytology , Lamin Type A/genetics , Myocardial Contraction , Myocytes, Cardiac/physiology , Adult , Aged , Cell Line , Humans , Middle Aged
4.
Clin Epigenetics ; 13(1): 139, 2021 07 10.
Article in English | MEDLINE | ID: mdl-34246298

ABSTRACT

BACKGROUND: Mutations in LMNA, encoding lamin A/C, lead to a variety of diseases known as laminopathies including dilated cardiomyopathy (DCM) and skeletal abnormalities. Though previous studies have investigated the dysregulation of gene expression in cells from patients with DCM, the role of epigenetic (gene regulatory) mechanisms, such as DNA methylation, has not been thoroughly investigated. Furthermore, the impact of family-specific LMNA mutations on DNA methylation is unknown. Here, we performed reduced representation bisulfite sequencing on ten pairs of fibroblasts and their induced pluripotent stem cell (iPSC) derivatives from two families with DCM due to distinct LMNA mutations, one of which also induces brachydactyly. RESULTS: Family-specific differentially methylated regions (DMRs) were identified by comparing the DNA methylation landscape of patient and control samples. Fibroblast DMRs were found to enrich for distal regulatory features and transcriptionally repressed chromatin and to associate with genes related to phenotypes found in tissues affected by laminopathies. These DMRs, in combination with transcriptome-wide expression data and lamina-associated domain (LAD) organization, revealed the presence of inter-family epimutation hotspots near differentially expressed genes, most of which were located outside LADs redistributed in LMNA-related DCM. Comparison of DMRs found in fibroblasts and iPSCs identified regions where epimutations were persistent across both cell types. Finally, a network of aberrantly methylated disease-associated genes revealed a potential molecular link between pathways involved in bone and heart development. CONCLUSIONS: Our results identified both shared and mutation-specific laminopathy epimutation landscapes that were consistent with lamin A/C mutation-mediated epigenetic aberrancies that arose in somatic and early developmental cell stages.


Subject(s)
Cardiomyopathy, Dilated/complications , Lamin Type A/analysis , Laminopathies/etiology , Cardiomyopathy, Dilated/genetics , DNA Methylation/genetics , DNA Methylation/physiology , Humans , Lamin Type A/genetics , Laminopathies/genetics
5.
BMC Med Genet ; 21(1): 152, 2020 07 22.
Article in English | MEDLINE | ID: mdl-32698886

ABSTRACT

BACKGROUND: Intermediate filament proteins that construct the nuclear lamina of a cell include the Lamin A/C proteins encoded by the LMNA gene, and are implicated in fundamental processes such as nuclear structure, gene expression, and signal transduction. LMNA mutations predominantly affect mesoderm-derived cell lineages in diseases collectively termed as laminopathies that include dilated cardiomyopathy with conduction defects, different forms of muscular dystrophies, and premature aging syndromes as Hutchinson-Gilford Progeria Syndrome. At present, our understanding of the molecular mechanisms regulating tissue-specific manifestations of laminopathies are still limited. METHODS: To gain deeper insight into the molecular mechanism of a novel LMNA splice-site mutation (c.357-2A > G) in an affected family with cardiac disease, we conducted deep RNA sequencing and pathway analysis for nine fibroblast samples obtained from three patients with cardiomyopathy, three unaffected family members, and three unrelated, unaffected individuals. We validated our findings by quantitative PCR and protein studies. RESULTS: We identified eight significantly differentially expressed genes between the mutant and non-mutant fibroblasts, that included downregulated insulin growth factor binding factor protein 5 (IGFBP5) in patient samples. Pathway analysis showed involvement of the ERK/MAPK signaling pathway consistent with previous studies. We found no significant differences in gene expression for Lamin A/C and B-type lamins between the groups. In mutant fibroblasts, RNA-seq confirmed that only the LMNA wild type allele predominately was expressed, and Western Blot showed normal Lamin A/C protein levels. CONCLUSIONS: IGFBP5 may contribute in maintaining signaling pathway homeostasis, which may lead to the absence of notable molecular and structural abnormalities in unaffected tissues such as fibroblasts. Compensatory mechanisms from other nuclear membrane proteins were not found. Our results also demonstrate that only one copy of the wild type allele is sufficient for normal levels of Lamin A/C protein to maintain physiological function in an unaffected cell type. This suggests that affected cell types such as cardiac tissues may be more sensitive to haploinsufficiency of Lamin A/C. These results provide insight into the molecular mechanism of disease with a possible explanation for the tissue specificity of LMNA-related dilated cardiomyopathy.


Subject(s)
Cardiomyopathies/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Profiling , Lamin Type A/genetics , Signal Transduction/genetics , Base Sequence , Family , Gene Expression Regulation , Humans , MAP Kinase Signaling System/genetics , Nuclear Lamina/metabolism
6.
J Biomech Eng ; 142(6)2020 06 01.
Article in English | MEDLINE | ID: mdl-31233093

ABSTRACT

Although mutations in the Lamin A/C gene (LMNA) cause a variety of devastating diseases, the pathological mechanism is often unknown. Lamin A/C proteins play a crucial role in forming a meshwork under the nuclear membrane, providing the nucleus with mechanical integrity and interacting with other proteins for gene regulation. Most LMNA mutations result in heart diseases, including some types that primarily have heart disease as the main pathology. In this study, we used cells from patients with different LMNA mutations that primarily lead to heart disease. Indeed, it is a mystery why a mutation to the protein in every nucleus of the body manifests as a disease of primarily the heart in these patients. Here, we aimed to investigate if strains mimicking those within the myocardial environment are sufficient to cause differences in cells with and without the LMNA mutation. To test this, a stretcher device was used to induce cyclic strain upon cells, and viability/proliferation, cytoskeleton and extracellular matrix organization, and nuclear morphology were quantified. The properties of cells with Hutchinson-Gilford progeria syndrome (HGPS) were found to be significantly different from all other cell lines and were mostly in line with previous findings. However, the properties of cells from patients who primarily had heart diseases were not drastically different when compared to individuals without the LMNA mutation. Our results indicated that cyclic strain alone was insufficient to cause any significant differences that could explain the mechanisms that lead to heart diseases in these patients with LMNA mutations.


Subject(s)
Lamin Type A , Progeria , Cell Nucleus , Fibroblasts , Gene Expression Regulation , Humans , Mutation
7.
J Vis Exp ; (153)2019 11 22.
Article in English | MEDLINE | ID: mdl-31814616

ABSTRACT

Science relies on increasingly complex data sets for progress, but common data management methods such as spreadsheet programs are inadequate for the growing scale and complexity of this information. While database management systems have the potential to rectify these issues, they are not commonly utilized outside of business and informatics fields. Yet, many research labs already generate "medium sized", low velocity, multi-dimensional data that could greatly benefit from implementing similar systems. In this article, we provide a conceptual overview explaining how databases function and the advantages they provide in tissue engineering applications. Structural fibroblast data from individuals with a lamin A/C mutation was used to illustrate examples within a specific experimental context. Examples include visualizing multidimensional data, linking tables in a relational database structure, mapping a semi-automated data pipeline to convert raw data into structured formats, and explaining the underlying syntax of a query. Outcomes from analyzing the data were used to create plots of various arrangements and significance was demonstrated in cell organization in aligned environments between the positive control of Hutchinson-Gilford progeria, a well-known laminopathy, and all other experimental groups. In comparison to spreadsheets, database methods were enormously time efficient, simple to use once set up, allowed for immediate access of original file locations, and increased data rigor. In response to the National Institutes of Health (NIH) emphasis on experimental rigor, it is likely that many scientific fields will eventually adopt databases as common practice due to their strong capability to effectively organize complex data.


Subject(s)
Database Management Systems , Databases, Factual , Tissue Engineering , Cell Line , Humans , Lamin Type A/genetics , United States
8.
Cells ; 6(4)2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29104234

ABSTRACT

Dupuytren's disease (palmar fibromatosis) involves nodules in fascia of the hand that leads to flexion contractures. Ledderhose disease (plantar fibromatosis) is similar with nodules of the foot. While clinical aspects are well-described, genetic mechanisms are unknown. We report a family with cardiac disease due to a heterozygous LMNA mutation (c.736C>T, p.Gln246Stop) with palmar/plantar fibromatosis and investigate the hypothesis that a second rare DNA variant increases the risk for fibrotic disease in LMNA mutation carriers. The proband and six family members were evaluated for the cardiac and hand/feet phenotypes and tested for the LMNA mutation. Fibroblast RNA studies revealed monoallelic expression of the normal LMNA allele and reduced lamin A/C mRNAs consistent with LMNA haploinsufficiency. A novel, heterozygous missense variant (c.230T>C, p.Val77Ala) in the Asteroid Homolog 1 (ASTE1) gene was identified as a potential risk factor in fibrotic disease using exome sequencing and family studies of five family members: four LMNA mutation carriers with fibromatosis and one individual without the LMNA mutation and no fibromatosis. With a possible role in epidermal growth factor receptor signaling, ASTE1 may contribute to the increased risk for palmar/plantar fibromatosis in patients with Lamin A/C haploinsufficiency.

9.
PLoS One ; 12(11): e0188256, 2017.
Article in English | MEDLINE | ID: mdl-29149195

ABSTRACT

Nuclear shape defects are a distinguishing characteristic in laminopathies, cancers, and other pathologies. Correlating these defects to the symptoms, mechanisms, and progression of disease requires unbiased, quantitative, and high-throughput means of quantifying nuclear morphology. To accomplish this, we developed a method of automatically segmenting fluorescently stained nuclei in 2D microscopy images and then classifying them as normal or dysmorphic based on three geometric features of the nucleus using a package of Matlab codes. As a test case, cultured skin-fibroblast nuclei of individuals possessing LMNA splice-site mutation (c.357-2A>G), LMNA nonsense mutation (c.736 C>T, pQ246X) in exon 4, LMNA missense mutation (c.1003C>T, pR335W) in exon 6, Hutchinson-Gilford Progeria Syndrome, and no LMNA mutations were analyzed. For each cell type, the percentage of dysmorphic nuclei, and other morphological features such as average nuclear area and average eccentricity were obtained. Compared to blind observers, our procedure implemented in Matlab codes possessed similar accuracy to manual counting of dysmorphic nuclei while being significantly more consistent. The automatic quantification of nuclear defects revealed a correlation between in vitro results and age of patients for initial symptom onset. Our results demonstrate the method's utility in experimental studies of diseases affecting nuclear shape through automated, unbiased, and accurate identification of dysmorphic nuclei.


Subject(s)
Cell Nucleus/genetics , Fibroblasts/metabolism , Heart Diseases/diagnosis , Lamin Type A/genetics , Mutation , Progeria/diagnosis , Adult , Age Factors , Age of Onset , Aged , Case-Control Studies , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Exons , Female , Fibroblasts/ultrastructure , Gene Expression , Heart Diseases/genetics , Heart Diseases/pathology , Humans , Image Processing, Computer-Assisted , Lamin Type A/metabolism , Male , Microscopy , Middle Aged , Observer Variation , Organelle Shape , Primary Cell Culture , Progeria/genetics , Progeria/pathology
10.
PLoS One ; 11(5): e0155421, 2016.
Article in English | MEDLINE | ID: mdl-27182706

ABSTRACT

The goals are to understand the primary genetic mechanisms that cause Sick Sinus Syndrome and to identify potential modifiers that may result in intrafamilial variability within a multigenerational family. The proband is a 63-year-old male with a family history of individuals (>10) with sinus node dysfunction, ventricular arrhythmia, cardiomyopathy, heart failure, and sudden death. We used exome sequencing of a single individual to identify a novel LMNA mutation and demonstrated the importance of Sanger validation and family studies when evaluating candidates. After initial single-gene studies were negative, we conducted exome sequencing for the proband which produced 9 gigabases of sequencing data. Bioinformatics analysis showed 94% of the reads mapped to the reference and identified 128,563 unique variants with 108,795 (85%) located in 16,319 genes of 19,056 target genes. We discovered multiple variants in known arrhythmia, cardiomyopathy, or ion channel associated genes that may serve as potential modifiers in disease expression. To identify candidate mutations, we focused on ~2,000 variants located in 237 genes of 283 known arrhythmia, cardiomyopathy, or ion channel associated genes. We filtered the candidates to 41 variants in 33 genes using zygosity, protein impact, database searches, and clinical association. Only 21 of 41 (51%) variants were validated by Sanger sequencing. We selected nine confirmed variants with minor allele frequencies <1% for family studies. The results identified LMNA c.357-2A>G, a novel heterozygous splice-site mutation as the primary mutation with rare or novel variants in HCN4, MYBPC3, PKP4, TMPO, TTN, DMPK and KCNJ10 as potential modifiers and a mechanism consistent with haploinsufficiency.


Subject(s)
Cardiomyopathy, Dilated/genetics , Death, Sudden, Cardiac/etiology , Genetic Heterogeneity , Lamin Type A/genetics , Mutation , RNA Splice Sites , Sick Sinus Syndrome/genetics , Adult , Alleles , Biomarkers , Cardiomyopathy, Dilated/diagnosis , DNA Mutational Analysis , Exome , Female , Gene Expression Profiling , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Pedigree , Phenotype , Reproducibility of Results , Sequence Analysis, DNA , Sick Sinus Syndrome/diagnosis
11.
BMC Med Genet ; 14: 49, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23634718

ABSTRACT

BACKGROUND: Global developmental delay and mental retardation are associated with X-linked disorders including Hunter syndrome (mucopolysaccharidosis type II) and Fragile X syndrome (FXS). Single nucleotide mutations in the iduronate 2-sulfatase (IDS) gene at Xq28 most commonly cause Hunter syndrome while a CGG expansion in the FMR1 gene at Xq27.3 is associated with Fragile X syndrome. Gene deletions of the Xq27-28 region are less frequently found in either condition with rare reports in females. Additionally, an association between Xq27-28 deletions and skewed X-inactivation of the normal X chromosome observed in previous studies suggested a primary role of the Xq27-28 region in X-inactivation. CASE PRESENTATION: We describe the clinical, molecular and biochemical evaluations of a four year-old female patient with global developmental delay and a hemizygous deletion of Xq27.3q28 (144,270,614-154,845,961 bp), a 10.6 Mb region that contains >100 genes including IDS and FMR1. A literature review revealed rare cases with similar deletions that included IDS and FMR1 in females with developmental delay, variable features of Hunter syndrome, and skewed X-inactivation of the normal X chromosome. In contrast, our patient exhibited skewed X-inactivation of the deleted X chromosome and tested negative for Hunter syndrome. CONCLUSIONS: This is a report of a female with a 10.6 Mb Xq27-28 deletion with skewed inactivation of the deleted X chromosome. Contrary to previous reports, our observations do not support a primary role of the Xq27-28 region in X-inactivation. A review of the genes in the deletion region revealed several potential genes that may contribute to the patient's developmental delays, and sequencing of the active X chromosome may provide insight into the etiology of this clinical presentation.


Subject(s)
Chromosome Deletion , Chromosomes, Human, X , Developmental Disabilities/genetics , Fragile X Syndrome/genetics , X Chromosome Inactivation , California , Child, Preschool , Comparative Genomic Hybridization , Female , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/diagnosis , Humans , Phenotype
12.
Proc Natl Acad Sci U S A ; 110(9): 3453-8, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23401503

ABSTRACT

Mutations of both nuclear and mitochondrial DNA (mtDNA)-encoded mitochondrial proteins can cause cardiomyopathy associated with mitochondrial dysfunction. Hence, the cardiac phenotype of nuclear DNA mitochondrial mutations might be modulated by mtDNA variation. We studied a 13-generation Mennonite pedigree with autosomal recessive myopathy and cardiomyopathy due to an SLC25A4 frameshift null mutation (c.523delC, p.Q175RfsX38), which codes for the heart-muscle isoform of the adenine nucleotide translocator-1. Ten homozygous null (adenine nucleotide translocator-1(-/-)) patients monitored over a median of 6 years had a phenotype of progressive myocardial thickening, hyperalaninemia, lactic acidosis, exercise intolerance, and persistent adrenergic activation. Electrocardiography and echocardiography with velocity vector imaging revealed abnormal contractile mechanics, myocardial repolarization abnormalities, and impaired left ventricular relaxation. End-stage heart disease was characterized by massive, symmetric, concentric cardiac hypertrophy; widespread cardiomyocyte degeneration; overabundant and structurally abnormal mitochondria; extensive subendocardial interstitial fibrosis; and marked hypertrophy of arteriolar smooth muscle. Substantial variability in the progression and severity of heart disease segregated with maternal lineage, and sequencing of mtDNA from five maternal lineages revealed two major European haplogroups, U and H. Patients with the haplogroup U mtDNAs had more rapid and severe cardiomyopathy than those with haplogroup H.


Subject(s)
Adenine Nucleotide Translocator 1/deficiency , Adenine Nucleotide Translocator 1/genetics , Cardiomyopathies/genetics , Cardiomyopathies/pathology , DNA, Mitochondrial/genetics , Haplotypes/genetics , Adolescent , Cardiomyopathies/physiopathology , Disease Progression , Female , Homozygote , Humans , Male , Mutation , Myocardium/pathology , Myocardium/ultrastructure , Pedigree
13.
Mol Genet Metab ; 107(3): 428-32, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23031367

ABSTRACT

BACKGROUND: The tafazzin gene (TAZ) is located at Xq28 and encodes a protein involved in the transacylation of cardiolipin, an essential mitochondrial phospholipid. Mutations in TAZ are associated with Barth syndrome (BTHS), the X-linked recessive condition with dilated cardiomyopathy, skeletal myopathy, growth retardation, neutropenia and organic aciduria. TAZ mutations also contribute to left ventricular noncompaction (LVNC), a cardiomyopathy characterized by loose, trabeculated myocardium. CASE REPORT: We report a family with a novel TAZ mutation and the clinical spectrum from severe BTHS in an infant to skeletal myopathy with LVNC in an adult, the oldest individual with BTHS reported. The proband is a 51-year-old male with muscle weakness since early childhood. He remained stable until the age of 43. His initial evaluations found LVNC and borderline neutropenia with no elevation of urine 3-methylglutaconic acid. The proband's great nephew is a 3-year-old who presented at birth with poor feeding, hypotonia, lactic acidosis and hypoglycemia. At three months he was admitted with failure to thrive, lethargy and respiratory distress due to heart failure. Cardiac studies revealed dilated cardiomyopathy with a spongiform trabeculated pattern of the left ventricle. Laboratory studies showed cyclic neutropenia and elevated urine 3-methylglutaconic and 3-methylglutaric acids. At age 11months the patient had a heart transplant. We conducted sequence analysis of the TAZ gene for two affected individuals, the proband first and then his great-nephew. A novel, hemizygous nonsense mutation in TAZ exon 7 (c.583G>T, p.Gly195X) was detected. CONCLUSION: At his current age of 51years-old, the proband is the oldest surviving individual reported with a confirmed molecular diagnosis and features of Barth syndrome. Further studies will be conducted to identify the genetic modifying factor(s) associated with the wide phenotypic range seen in this family.


Subject(s)
Barth Syndrome/genetics , Cardiomyopathy, Dilated/genetics , Heart Defects, Congenital/genetics , Heart Failure/genetics , Transcription Factors/genetics , Acyltransferases , Barth Syndrome/pathology , Cardiolipins/metabolism , Cardiomyopathy, Dilated/pathology , Codon, Nonsense , Exons , Genetic Heterogeneity , Glutarates/urine , Heart Defects, Congenital/pathology , Heart Failure/pathology , Humans , Infant , Male , Middle Aged , Myocardium/pathology , Neutropenia/genetics , Neutropenia/pathology , Pedigree
14.
JACC Cardiovasc Imaging ; 4(1): 1-10, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21232697

ABSTRACT

OBJECTIVES: the aim of this study was to test the hypothesis that chronic mitochondrial energy deficiency causes dilated cardiomyopathy, we characterized the hearts of age-matched young and old adenine nucleotide translocator (ANT)1 mutant and control mice. BACKGROUND: ANTs export mitochondrial adenosine triphosphate into the cytosol and have a role in the regulation of the intrinsic apoptosis pathway. Mitochondrial energy deficiency has been hypothesized, on the basis of indirect evidence, to be a factor in the pathophysiology of dilated cardiomyopathies. Ant1 inactivation should limit adenosine triphosphate for contraction and calcium transport, thereby resulting in early cardiac dysfunction with later dilation and heart failure. METHODS: we conducted a multiyear study of 73 mutant (Ant1-/-) and 57 control (Ant1+/+) mice, between the ages of 2 and 21 months. Hearts were characterized by cardiac anatomy, echocardiographic imaging with velocity vector analysis, histopathology, and apoptosis assays. RESULTS: the Ant1-/- mice developed a distinctive concentric dilated cardiomyopathy, characterized by substantial myocardial hypertrophy and ventricular dilation, with cardiac function declining earlier in age as compared to control mice. Left ventricular circumferential, radial, and rotational mechanics were reduced even in the younger mutants with preserved systolic function. Histopathologic analysis demonstrated increased myocyte hypertrophy, fibrosis, and calcification in the mutant mice as compared with control mice. Furthermore, increased cytoplasmic cytochrome c levels and caspase 3 activation were observed in the mutant mice. CONCLUSIONS: our results demonstrate that mitochondrial energy deficiency is sufficient to cause dilated cardiomyopathy, confirming that energy defects are a factor in this disease. Energy deficiency initially leads to early mechanical dysfunction before a decline in left ventricular systolic function. Chronic energy deficiency with age then leads to heart failure. Our results now allow us to use the Ant1-/- mouse model for testing new therapies for ANT1 mutant patients.


Subject(s)
Apoptosis , Cardiomyopathy, Dilated/enzymology , Disease Models, Animal , Mitochondrial ADP, ATP Translocases/deficiency , Myocardium/pathology , Animals , Blotting, Western , Cardiomegaly/enzymology , Cardiomegaly/physiopathology , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/physiopathology , Echocardiography , Female , Histocytochemistry , Male , Mice , Mice, Mutant Strains , Mitochondria, Heart/metabolism , Mitochondrial ADP, ATP Translocases/genetics , Mutation , Myocardial Contraction , Stroke Volume
15.
Eur J Hum Genet ; 19(2): 200-7, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20978534

ABSTRACT

Pathogenic mitochondrial DNA (mtDNA) mutations leading to mitochondrial dysfunction can cause cardiomyopathy and heart failure. Owing to a high mutation rate, mtDNA defects may occur at any nucleotide in its 16 569 bp sequence. Complete mtDNA sequencing may detect pathogenic mutations, which can be difficult to interpret because of normal ethnic/geographic-associated haplogroup variation. Our goal is to show how to identify candidate mtDNA mutations by sorting out polymorphisms using readily available online tools. The purpose of this approach is to help investigators in prioritizing mtDNA variants for functional analysis to establish pathogenicity. We analyzed complete mtDNA sequences from 29 Italian patients with mitochondrial cardiomyopathy or suspected disease. Using MITOMASTER and PhyloTree, we characterized 593 substitution variants by haplogroup and allele frequencies to identify all novel, non-haplogroup-associated variants. MITOMASTER permitted determination of each variant's location, amino acid change and evolutionary conservation. We found that 98% of variants were common or rare, haplogroup-associated variants, and thus unlikely to be primary cause in 80% of cases. Six variants were novel, non-haplogroup variants and thus possible contributors to disease etiology. Two with the greatest pathogenic potential were heteroplasmic, nonsynonymous variants: m.15132T>C in MT-CYB for a patient with hypertrophic dilated cardiomyopathy and m.6570G>T in MT-CO1 for a patient with myopathy. In summary, we have used our automated information system, MITOMASTER, to make a preliminary distinction between normal mtDNA variation and pathogenic mutations in patient samples; this fast and easy approach allowed us to select the variants for traditional analysis to establish pathogenicity.


Subject(s)
Cardiomyopathies/genetics , DNA, Mitochondrial/genetics , Databases, Genetic , Genetic Variation , Mitochondrial Diseases/genetics , Phylogeny , Sequence Analysis, DNA/methods , Adolescent , Adult , Cardiomyopathies/pathology , Child , Echocardiography , Female , Genes, Mitochondrial , Humans , Infant , Italy , Male , Middle Aged , Mitochondria/genetics , Mitochondrial Diseases/pathology , Mutation , Pedigree
16.
PLoS One ; 5(8): e12295, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20808834

ABSTRACT

Mutations in mitochondrial DNA (mtDNA) may cause maternally-inherited cardiomyopathy and heart failure. In homoplasmy all mtDNA copies contain the mutation. In heteroplasmy there is a mixture of normal and mutant copies of mtDNA. The clinical phenotype of an affected individual depends on the type of genetic defect and the ratios of mutant and normal mtDNA in affected tissues. We aimed at determining the sensitivity of next-generation sequencing compared to Sanger sequencing for mutation detection in patients with mitochondrial cardiomyopathy. We studied 18 patients with mitochondrial cardiomyopathy and two with suspected mitochondrial disease. We "shotgun" sequenced PCR-amplified mtDNA and multiplexed using a single run on Roche's 454 Genome Sequencer. By mapping to the reference sequence, we obtained 1,300x average coverage per case and identified high-confidence variants. By comparing these to >400 mtDNA substitution variants detected by Sanger, we found 98% concordance in variant detection. Simulation studies showed that >95% of the homoplasmic variants were detected at a minimum sequence coverage of 20x while heteroplasmic variants required >200x coverage. Several Sanger "misses" were detected by 454 sequencing. These included the novel heteroplasmic 7501T>C in tRNA serine 1 in a patient with sudden cardiac death. These results support a potential role of next-generation sequencing in the discovery of novel mtDNA variants with heteroplasmy below the level reliably detected with Sanger sequencing. We hope that this will assist in the identification of mtDNA mutations and key genetic determinants for cardiomyopathy and mitochondrial disease.


Subject(s)
Cardiomyopathies/genetics , DNA, Mitochondrial/genetics , Genetic Variation , Sequence Analysis, DNA/methods , Base Sequence , Computational Biology , Female , Humans , Male , Mitochondrial Diseases/genetics , Mutation
17.
J Am Acad Dermatol ; 58(2): 303-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18222328

ABSTRACT

Infantile systemic hyalinosis (ISH) is a rare, progressive autosomal recessive disease, which is usually fatal by the age of 2 years. Clinical onset typically occurs within the first few weeks of life. The disease is characterized by joint contractures, osteopenia, failure to thrive, gingival hypertrophy, diarrhea, protein-losing enteropathy, and frequent infections. Dermatologic manifestations include thickened skin, hyperpigmentation, perianal nodules, and facial papules. Histopathology shows hyaline deposits in the dermis and visceral organs. We describe a patient with ISH confirmed by clinical and histopathologic findings, as well as DNA sequence analysis, which revealed a novel homozygous T118K mutation in the CMG2 gene.


Subject(s)
Contracture/pathology , Joint Diseases/pathology , Muscular Diseases/pathology , Skin Diseases/pathology , Amino Acid Substitution , Contracture/genetics , Diarrhea/pathology , Fatal Outcome , Female , Humans , Infant , Joint Diseases/genetics , Membrane Proteins/genetics , Muscular Diseases/genetics , Receptors, Peptide , Skin Diseases/genetics
18.
Curr Opin Pediatr ; 19(6): 619-27, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18025927

ABSTRACT

PURPOSE OF REVIEW: Noncompaction of the left ventricle is a descriptive anatomical term and recently recognized primary cardiomyopathy. Cardiac imaging now allows for prompt detection. The specific etiology remains poorly understood, however, and the major genetic determinants are unknown. This review describes recent data showing the genetic heterogeneity and overlap with other cardiomyopathies. Understanding the genetics may depend on clarifying the distinctive diagnostic features and investigating the contribution of all known cardiomyopathy-causing genes with overlapping morphology. RECENT FINDINGS: Adding to the known genes (TAZ, DTNA, LDB3 and LMNA), recent work has identified SCN5A, MYH7 and MYBPC3 as associated loci. LDB3 may also be a genetic modifier. Case reports and linkage studies suggest additional loci at 1p36, 1q43 and 11p15. Aside from Barth syndrome, other genetic and metabolic syndromes with noncompaction have been described. Despite this, large studies have failed to identify the etiology in the majority of patients. SUMMARY: Despite advances in detection, comprehensive clinical, pathological, genetic, and family studies are necessary to define the phenotypic overlap with other cardiomyopathies. Without a more precise understanding of its etiology, the answers to the questions regarding the clinical relevance and management of patients with noncompaction of the left ventricle will remain elusive.


Subject(s)
Cardiomyopathies/genetics , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Cardiomyopathies/pathology , Child , Genetic Heterogeneity , Heart Ventricles/pathology , Humans , Hypertrophy, Left Ventricular/genetics , LIM Domain Proteins , Neuromuscular Diseases/genetics , Phenotype , Proteins/genetics , Ryanodine Receptor Calcium Release Channel/genetics , Transcription Factors/genetics
19.
Hum Genet ; 117(1): 43-53, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15806394

ABSTRACT

Campomelic dysplasia (CD; MIM 114290), an autosomal dominant skeletal malformation syndrome with XY sex reversal, is caused by heterozygous de novo mutations in and around the SOX9 gene on 17q. We report a patient with typical signs of CD, including sex reversal, who was, surprisingly, homozygous for the nonsense mutation Y440X. Since neither parent carried the Y440X mutation, possible mechanisms explaining the homozygous situation were a de novo mutation followed by uniparental isodisomy, somatic crossing over, or gene conversion. As the patient was heterozygous for six microsatellite markers flanking SOX9, uniparental isodisomy and somatic crossing over were excluded. Analysis of intragenic single-nucleotide polymorphisms suggested that the homozygous mutation arose by a mitotic gene conversion event involving exchange of at least 440 nucleotides and at most 2,208 nucleotides between a de novo mutant maternal allele and a wild-type paternal allele. Analysis of cloned alleles showed that homozygous mutant cells constituted about 80% of the leukocyte cell population of the patient, whereas about 20% were heterozygous mutant cells. Heterozygous Y440X mutations, previously described in three CD cases, have been identified in seven additional cases, thus constituting the most frequent recurrent mutations in SOX9. These patients frequently have a milder phenotype with longer survival, possibly because of the retention of some transactivation activity of the mutant protein on SOX9 target genes, as shown by cell transfection experiments. The fact that the patient survived for 3 months may thus be explained by homozygosity for a hypomorphic rather than a complete loss-of-function allele, in combination with somatic mosaicism. This is, to our knowledge, the first report of mitotic gene conversion of a wild-type allele by a de novo mutant allele in humans.


Subject(s)
Bone Diseases, Developmental/genetics , Gene Conversion , High Mobility Group Proteins/genetics , Sex Chromosome Aberrations , Transcription Factors/genetics , Abnormalities, Multiple , Chromosomes, Human, Pair 17 , Codon, Nonsense , Disorders of Sex Development , Fatal Outcome , Female , Humans , Infant, Newborn , Inheritance Patterns , Karyotyping , Microsatellite Repeats , Mitosis , Mosaicism , Polymorphism, Single Nucleotide , SOX9 Transcription Factor , Sex Differentiation
20.
Gene ; 330: 9-18, 2004 Apr 14.
Article in English | MEDLINE | ID: mdl-15087119

ABSTRACT

TBX5 is a member of the T-box gene family and encodes a transcription factor involved in cardiac and limb development. Mutations of TBX5 cause Holt-Oram syndrome (HOS), an autosomal-dominant condition with congenital cardiac defects and forelimb anomalies. Here, we used a GAL4-TBX5 fusion protein in a modified yeast-one hybrid system to elucidate the TBX5 transactivating domain. Using a series of deletion mutations of TBX5, we narrowed down its functional domain to amino acids 339-379 of its C-terminal half; point mutagenesis analysis then showed that the loss of amino acids 349-351 abolished transactivation. This result was confirmed in mammalian cells. Furthermore, wild-type TBX5, but not TBX5 with mutations at the amino acids 349-351, has ability to inhibit NCI-H1299 cell growth also suggesting that these amino acids are crucial for the TBX5 function in mammalian cells. In addition, to identify the nuclear localization signal of TBX5, we searched for cluster of basic amino acids. We found that the deletion of the KRK sequence at amino acids 325-327 mislocalizes TBX5 to cytoplasm, suggesting that these amino acids serve as a nuclear localization signal. These studies enhance our understanding of the structure-function relationship of TBX5 and suggest that truncation mutations of TBX5 could cause HOS through the loss of its transactivating domain and/or the nuclear localization signal.


Subject(s)
Nuclear Localization Signals/genetics , T-Box Domain Proteins/genetics , Transcriptional Activation/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Binding Sites/genetics , Cell Division/genetics , Cell Division/physiology , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Microscopy, Fluorescence , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Plasmids/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Sequence Homology, Amino Acid , T-Box Domain Proteins/physiology , Trans-Activators/genetics , Trans-Activators/physiology , Transcriptional Activation/physiology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...