Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Am J Transl Res ; 16(5): 2034-2048, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38883374

RESUMO

OBJECTIVE: Aggregating evidence highlights the strong genetic basis underpinning congenital heart disease (CHD). Here BMP4 was chosen as a prime candidate gene causative of human CHD predominantly because BMP4 was amply expressed in the embryonic hearts and knockout of Bmp4 in mice led to embryonic demise mainly from multiple cardiovascular developmental malformations. The aim of this retrospective investigation was to discover a novel BMP4 mutation underlying human CHD and explore its functional impact. METHODS: A sequencing examination of BMP4 was implemented in 212 index patients suffering from CHD and 236 unrelated non-CHD individuals as well as the family members available from the proband carrying a discovered BMP4 mutation. The impacts of the discovered CHD-causing mutation on the expression of NKX2-5 and TBX20 induced by BMP4 were measured by employing a dual-luciferase analysis system. RESULTS: A new heterozygous BMP4 mutation, NM_001202.6:c.318T>G;p.(Tyr106*), was found in a female proband affected with familial CHD. Genetic research of the mutation carrier's relatives unveiled that the truncating mutation was in co-segregation with CHD in the pedigree. The nonsense mutation was absent from 236 unrelated non-CHD control persons. Quantitative biologic measurement revealed that Tyr106*-mutant BMP4 failed to induce the expression of NKX2-5 and TBX20, two genes whose expression is lost in CHD. CONCLUSION: The current findings indicate BMP4 as a new gene predisposing to human CHD, allowing for improved prenatal genetic counseling along with personalized treatment of CHD patients.

2.
Biology (Basel) ; 12(9)2023 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-37759586

RESUMO

Atrial fibrillation (AF), the most prevalent type of sustained cardiac dysrhythmia globally, confers strikingly enhanced risks for cognitive dysfunction, stroke, chronic cardiac failure, and sudden cardiovascular demise. Aggregating studies underscore the crucial roles of inherited determinants in the occurrence and perpetuation of AF. However, due to conspicuous genetic heterogeneity, the inherited defects accounting for AF remain largely indefinite. Here, via whole-genome genotyping with genetic markers and a linkage assay in a family suffering from AF, a new AF-causative locus was located at human chromosome 7p14.2-p14.3, a ~4.89 cM (~4.43-Mb) interval between the markers D7S526 and D7S2250. An exome-wide sequencing assay unveiled that, at the defined locus, the mutation in the TBX20 gene, NM_001077653.2: c.695A>G; p.(His232Arg), was solely co-segregated with AF in the family. Additionally, a Sanger sequencing assay of TBX20 in another family suffering from AF uncovered a novel mutation, NM_001077653.2: c.862G>C; p.(Asp288His). Neither of the two mutations were observed in 600 unrelated control individuals. Functional investigations demonstrated that the two mutations both significantly reduced the transactivation of the target gene KCNH2 (a well-established AF-causing gene) and the ability to bind the promoter of KCNH2, while they had no effect on the nuclear distribution of TBX20. Conclusively, these findings reveal a new AF-causative locus at human chromosome 7p14.2-p14.3 and strongly indicate TBX20 as a novel AF-predisposing gene, shedding light on the mechanism underlying AF and suggesting clinical significance for the allele-specific treatment of AF patients.

3.
Front Cardiovasc Med ; 10: 1217069, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37600052

RESUMO

Introduction: In coronary bifurcation lesions treated with percutaneous coronary intervention (PCI) using a 1-stent strategy, the occurrence of side branch (SB) compromise may lead to long-term myocardial ischemia in the SB territory. Murray law-based quantitative flow ratio (µQFR) is a novel angiography-based approach estimating fractional flow reserve from a single angiographic view, and thus is more feasible to assess SB compromise in routine practice. However, its association with long-term SB coronary blood flow remains unknown. Methods: A total of 146 patients with 313 non-left main bifurcation lesions receiving 1-stent strategy with drug-eluting stents was included in this retrospective study. These lesions had post-procedural Thrombolysis in Myocardial Infarction (TIMI) flow grade 3 in SBs, and documented angiographic images of index procedure and 6- to 24-month angiographic follow-up. Post-procedural SB µQFR was calculated. Long-term SB coronary blood flow was quantified with the TIMI grading system using angiograms acquired at angiographic follow-up. Results: At follow-up, 8 (2.6%), 16 (5.1%), 61 (19.5%), and 228 (72.8%) SBs had a TIMI flow grade of 0, 1, 2, and 3, respectively. The incidences of long-term SB TIMI flow grade ≤1 and ≤2 both tended to decrease across the tertiles of post-procedural SB µQFR. The receiver operating characteristic curve analyses indicated the post-procedural SB µQFR ≤0.77 was the optimal cut-off value to identify long-term SB TIMI flow grade ≤1 (specificity, 37.50%; sensitivity, 87.20%; area under the curve, 0.6673; P = 0.0064), and it was independently associated with 2.57-fold increased risk (adjusted OR, 2.57; 95% CI, 1.02-7.25; P = 0.045) in long-term SB TIMI flow grade ≤1 after adjustment. Discussion: Post-procedural SB µQFR was independently associated with increased risk in impaired SB TIMI flow at long-term follow-up. Further investigations should focus on whether PCI optimization based on µQFR may contribute to improve SB flow in the long term.

4.
Biology (Basel) ; 12(3)2023 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-36979038

RESUMO

As the most prevalent type of birth malformation, congenital heart disease (CHD) gives rise to substantial mortality and morbidity as well as a socioeconomic burden. Although aggregating investigations highlight the genetic basis for CHD, the genetic determinants underpinning CHD remain largely obscure. In this research, a Chinese family suffering from autosomal dominant CHD (atrial septal defect) and arrhythmias was enrolled. A genome-wide genotyping with microsatellite markers followed by linkage assay as well as sequencing analysis was conducted. The functional effects of the discovered genetic mutation were characterized by dual patch-clamp electrophysiological recordings in N2A cells and propidium iodide uptake assays in HeLa cells. As a result, a novel genetic locus for CHD and arrhythmias was located on chromosome 17q21.31-q21.33, a 4.82-cM (5.12 Mb) region between two markers of D17S1861 and D17S1795. Sequencing assays of the genes at the mapped locus unveiled a novel heterozygous mutation in the GJC1 gene coding for connexin 45 (Cx45), NM_005497.4:c.550A>G;p.R184G, which was in co-segregation with the disease in the whole family and was not observed in 516 unrelated healthy individuals or gnomAD. Electrophysiological analyses revealed that the mutation significantly diminished the coupling conductance in homomeric cell pairs (R184G/R184G) and in cell pairs expressing either R184G/Cx45 or R184G/Cx43. Propidium iodide uptake experiments demonstrated that the Cx45 R184G mutation did not increase the Cx45 hemichannel function. This investigation locates a new genetic locus linked to CHD and arrhythmias on chromosome 17q21.31-q21.33 and indicates GJC1 as a novel gene predisposing to CHD and arrhythmias, implying clinical implications for prognostic risk assessment and personalized management of patients affected with CHD and arrhythmias.

5.
Eur J Med Genet ; 66(3): 104705, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36657711

RESUMO

Dilated cardiomyopathy (DCM), characteristic of left ventricular or biventricular dilation with systolic dysfunction, is the most common form of cardiomyopathy, and a leading cause of heart failure and sudden cardiac death. Aggregating evidence highlights the underlying genetic basis of DCM, and mutations in over 100 genes have been causally linked to DCM. Nevertheless, due to pronounced genetic heterogeneity, the genetic defects underpinning DCM in most cases remain obscure. Hence, this study was sought to identify novel genetic determinants of DCM. In this investigation, whole-exome sequencing and bioinformatics analyses were conducted in a family suffering from DCM, and a novel heterozygous mutation in the VEZF1 gene (coding for a zinc finger-containing transcription factor critical for cardiovascular development and structural remodeling), NM_007146.3: c.490A > T; p.(Lys164*), was identified. The nonsense mutation was validated by Sanger sequencing and segregated with autosome-dominant DCM in the family with complete penetrance. The mutation was neither detected in another cohort of 200 unrelated DCM patients nor observed in 400 unrelated healthy individuals nor retrieved in the Single Nucleotide Polymorphism database, the Human Gene Mutation Database and the Genome Aggregation Database. Biological analyses by utilizing a dual-luciferase reporter assay system revealed that the mutant VEZF1 protein failed to transactivate the promoters of MYH7 and ET1, two genes that have been associated with DCM. The findings indicate VEZF1 as a new gene responsible for DCM, which provides novel insight into the molecular pathogenesis of DCM, implying potential implications for personalized precisive medical management of the patients affected with DCM.


Assuntos
Cardiomiopatia Dilatada , Humanos , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/patologia , Proteínas de Ligação a DNA/genética , Heterozigoto , Mutação , Linhagem , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
6.
J Am Heart Assoc ; 11(22): e027578, 2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36346048

RESUMO

Background Dilated cardiomyopathy (DCM), characterized by progressive left ventricular enlargement and systolic dysfunction, is the most common type of cardiomyopathy and a leading cause of heart failure and cardiac death. Accumulating evidence underscores the critical role of genetic defects in the pathogenesis of DCM, and >250 genes have been implicated in DCM to date. However, DCM is of substantial genetic heterogeneity, and the genetic basis underpinning DCM remains elusive in most cases. Methods and Results By genome-wide scan with microsatellite markers and genetic linkage analysis in a 4-generation family inflicted with autosomal-dominant DCM, a new locus for DCM was mapped on chromosome 15q13.1-q13.3, a 4.77-cM (≈3.43 Mbp) interval between markers D15S1019 and D15S1010, with the largest 2-point logarithm of odds score of 5.1175 for the marker D15S165 at recombination fraction (θ)=0.00. Whole-exome sequencing analyses revealed that within the mapping chromosomal region, only the mutation in the KLF13 gene, c.430G>T (p.E144X), cosegregated with DCM in the family. In addition, sequencing analyses of KLF13 in another cohort of 266 unrelated patients with DCM and their available family members unveiled 2 new mutations, c.580G>T (p.E194X) and c.595T>C (p.C199R), which cosegregated with DCM in 2 families, respectively. The 3 mutations were absent from 418 healthy subjects. Functional assays demonstrated that the 3 mutants had no transactivation on the target genes ACTC1 and MYH7 (2 genes causally linked to DCM), alone or together with GATA4 (another gene contributing to DCM), and a diminished ability to bind the promoters of ACTC1 and MYH7. Add, the E144X-mutant KLF13 showed a defect in intracellular distribution. Conclusions This investigation indicates KLF13 as a new gene predisposing to DCM, which adds novel insight to the molecular pathogenesis underlying DCM, implying potential implications for prenatal prevention and precision treatment of DCM in a subset of patients.


Assuntos
Cardiomiopatia Dilatada , Humanos , Cardiomiopatia Dilatada/metabolismo , Mutação , Linhagem , Proteínas Repressoras/genética , Proteínas de Ciclo Celular/genética , Fatores de Transcrição Kruppel-Like/genética
7.
Front Cardiovasc Med ; 9: 965364, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36158840

RESUMO

Background: The optimum therapy for coronary slow flow phenomenon (CSFP) stays debatable. This study compared the effectiveness of alprostadil with isosorbide dinitrate in alleviating angina episodes in CSFP patients. Methods: In this prospective, randomized controlled study, 102 patients with CSFP without severe coronary artery stenosis that exhibited stable angina were allocated randomly in a ratio of 1:1 to either the alprostadil group (40 µg, three times per day, n = 51) or the isosorbide dinitrate group (5 mg, three times per day, n = 51). Frequency of angina events, intensity of suffering, and the Canadian Cardiovascular Society (CCS) grading of angina pectoris were evaluated at baseline and one month after. Additionally, the Seattle Angina Questionnaire (SAQ) was assessed. Results: Baseline characteristics were comparable between the two groups. At 1-month follow-up, patients administered with alprostadil experienced fewer angina episodes [episodes per week, 1 (2) vs. 2 (2), P < 0.001] and less pain intensity [self-evaluated pain score, 2 (3) vs. 3 (4), P < 0.001] than those with isosorbide dinitrate. In the alprostadil group, 78.4% of patients were classified as CCS class I, significantly higher than the 47.1% seen in the isosorbide dinitrate group (P = 0.001). Furthermore, treatment of alprostadil led to a significant improvement in the SAQ score (7.09 U, 95% CI: 4.22-9.96, P < 0.001) compared to isosorbide dinitrate. Additionally, fewer patients suffered headaches when receiving alprostadil (7.8% vs. 19.6%, P = 0.084). Conclusion: Alprostadil was more effective in ameliorating angina symptoms in CSFP patients than isosorbide dinitrate. Clinical trial registration: [www.chictr.org.cn], identifier [ChiCTR2000033233].

8.
Am J Transl Res ; 14(3): 1672-1684, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35422912

RESUMO

INTRODUCTION: As the most frequent type of birth defect in humans, congenital heart disease (CHD) leads to a large amount of morbidity and mortality as well as a tremendous socioeconomic burden. Accumulating studies have convincingly substantiated the pivotal roles of genetic defects in the occurrence of familial CHD, and deleterious variations in a great number of genes have been reported to cause various types of CHD. However, owing to pronounced genetic heterogeneity, the hereditary components underpinning CHD remain obscure in most cases. This investigation aimed to identify novel genetic determinants underlying CHD. METHODS AND RESULTS: A four-generation pedigree with high incidence of autosomal-dominant CHD was enrolled from the Chinese Han race population. Using whole-exome sequencing and Sanger sequencing assays of the family members available, a novel SOX7 variation in heterozygous status, NM_031439.4: c.310C>T; p.(Gln104*), was discovered to be in co-segregation with the CHD phenotype in the whole family. The truncating variant was absent in 500 unrelated healthy subjects utilized as control individuals. Functional measurements by dual-luciferase reporter analysis revealed that Gln104*-mutant SOX7 failed to transactivate its two important target genes, GATA4 and BMP2, which are both responsible for CHD. In addition, the nonsense variation invalidated the cooperative transactivation between SOX7 and NKX2.5, which is another recognized CHD-causative gene. CONCLUSION: The present study demonstrates for the first time that genetically defective SOX7 predisposes to CHD, which sheds light on the novel molecular mechanism underpinning CHD, and implies significance for precise prevention and personalized treatment in a subset of CHD patients.

9.
Genet Mol Biol ; 45(2): e20210378, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35377386

RESUMO

Atrial fibrillation (AF) represents the most common type of sustained cardiac arrhythmia in humans and confers a significantly increased risk for thromboembolic stroke, congestive heart failure and premature death. Aggregating evidence emphasizes the predominant genetic defects underpinning AF and an increasing number of deleterious variations in more than 50 genes have been involved in the pathogenesis of AF. Nevertheless, the genetic basis underlying AF remains incompletely understood. In the current research, by whole-exome sequencing and Sanger sequencing analysis in a family with autosomal-dominant AF and congenital patent ductus arteriosus (PDA), a novel heterozygous variation in the PRRX1 gene encoding a homeobox transcription factor critical for cardiovascular development, NM_022716.4:c.373G>T;p.(Glu125*), was identified to be in co-segregation with AF and PDA in the whole family. The truncating variation was not detected in 306 unrelated healthy individuals employed as controls. Quantitative biological measurements with a reporter gene analysis system revealed that the Glu125*-mutant PRRX1 protein failed to transactivate its downstream target genes SHOX2 and ISL1, two genes that have been causally linked to AF. Conclusively, the present study firstly links PRRX1 loss-of-function variation to AF and PDA, suggesting that AF and PDA share a common abnormal developmental basis in a proportion of cases.

10.
Coron Artery Dis ; 33(4): 284-294, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35085159

RESUMO

BACKGROUND: Coronary artery calcium (CAC) is a modifiable contributor of in-stent restenosis (ISR), but quantitative analyses using a noninvasive approach are limited. We aimed to investigate the associations between CAC score derived from ECG-gated coronary computed tomography angiography (CCTA) or non-gated non-contrast chest computed tomography (NCCT) and ISR. METHODS: We included 368 lesions in 194 patients with coronary drug-eluting stent implantations in final analyses. CAC was quantified using the Agatston score. Primary endpoint was ISR, defined as lumen diameter stenosis over 50% at the stent segment or its proximal or distal edges (5-mm segments adjacent to the stent), at angiographic follow-up. RESULTS: The CAC scores in either CCTA/2.5 mm group (r = 0.7702; P < 0.0001) or NCCT/5 mm group (r = 0.7105; P < 0.0001) were both correlated with in-stent diameter stenosis. The receiver-operating characteristic curve analysis identified a CAC score >245 in CCTA/2.5 mm group as the optimal ISR cutoff (sensitivity, 60.0%; specificity, 83.7%; area under the curve, 0.744; P < 0.001), and >209 in NCCT/5 mm group (sensitivity, 46.7%; specificity, 91.9%; area under the curve, 0.704; P < 0.001). Multivariable logistic regression models indicated a CAC score >245 in CCTA/2.5 mm group and >209 in NCCT/5 mm group independently associated with an 8.46- and 21.89-fold increase in ISR, respectively (all P < 0.01). CONCLUSIONS: Either a CAC score >245 in CCTA/2.5 mm or >209 in NCCT/5 mm was significantly associated with increased risk in ISR.


Assuntos
Reestenose Coronária , Stents Farmacológicos , Intervenção Coronária Percutânea , Cálcio , Constrição Patológica/complicações , Angiografia Coronária/métodos , Reestenose Coronária/diagnóstico por imagem , Reestenose Coronária/etiologia , Vasos Coronários/diagnóstico por imagem , Stents Farmacológicos/efeitos adversos , Humanos , Intervenção Coronária Percutânea/efeitos adversos
11.
Ann Vasc Surg ; 80: 325-332, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34780937

RESUMO

BACKGROUND: We assessed the safety as well as the efficacy of self-expanding stent placement for the treatment of malignant superior vena cava syndrome (SVCS), besides identifying the predictable probable factors for the clinical improvement of endovascular stent treatment in SVCS. METHODS: The study reviewed 112 patients (92 men) with malignant SVCS retrospectively from January 2015 to December 2020. RESULTS: Out of total 112 patients, 106 stents were successfully placed in 102 patients, however 4 patient's occlusions could not be passed and 6 patient's procedure was abandoned due to intraluminal thrombus as detected in venography. In 92 patients, complete resolution of syndrome was observed within 72 hrs but 10 patients did not to intervention. In 102 patients, procedure-related 8 complications were noted including stent migrations (n = 4), pulmonary embolism (n = 2), and pulmonary edemas (n = 2). Besides after stenting, 3 hemorrhages with anticoagulation therapy were observed with 4 recurrences at 22, 36, 51 and day 58 in 6 months. The pressure gradient across the lesion (≥ 20mmHg) was used as a predictor for clinical efficacy of stent therapy for SVCS. CONCLUSIONS: Endovascular stent insertion is a safe and effective intervention for malignant SVCS, especially for those with pressure gradient across the lesions ≥ 20mmHg.


Assuntos
Procedimentos Endovasculares/métodos , Stents , Síndrome da Veia Cava Superior/cirurgia , Angiografia , Feminino , Humanos , Modelos Logísticos , Neoplasias Pulmonares/complicações , Masculino , Pessoa de Meia-Idade , Desenho de Prótese , Estudos Retrospectivos , Síndrome da Veia Cava Superior/etiologia , Tempo para o Tratamento , Resultado do Tratamento
12.
J Am Heart Assoc ; 10(23): e023517, 2021 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-34845933

RESUMO

Background Atrial fibrillation (AF) is the most common form of clinical cardiac dysrhythmia responsible for thromboembolic cerebral stroke, congestive heart failure, and death. Aggregating evidence highlights the strong genetic basis of AF. Nevertheless, AF is of pronounced genetic heterogeneity, and in an overwhelming majority of patients, the genetic determinants underpinning AF remain elusive. Methods and Results By genome-wide screening with polymorphic microsatellite markers and linkage analysis in a 4-generation Chinese family affected with autosomal-dominant AF, a novel locus for AF was mapped to chromosome 1q24.2-q25.1, a 3.20-cM (≈4.19 Mbp) interval between markers D1S2851 and D1S218, with the greatest 2-point logarithm of odds score of 4.8165 for the marker D1S452 at recombination fraction=0.00. Whole-exome sequencing and bioinformatics analyses showed that within the mapping region, only the mutation in the paired related homeobox 1 (PRRX1) gene, NM_022716.4:c.319C>T;(p.Gln107*), cosegregated with AF in the family. In addition, sequencing analyses of PRRX1 in another cohort of 225 unrelated patients with AF revealed a new mutation, NM_022716.4:c.437G>T; (p.Arg146Ile), in a patient. The 2 mutations were absent in 908 control subjects. Biological analyses in HeLa cells demonstrated that the 2 mutants had significantly diminished transactivation on the target genes ISL1 and SHOX2 and markedly decreased ability to bind the promoters of ISL1 and SHOX2 (2 genes causally linked to AF), although with normal intracellular distribution. Conclusions This study first indicates that PRRX1 loss-of-function mutations predispose to AF, which provides novel insight into the molecular pathogenesis underpinning AF, implying potential implications for precisive prophylaxis and management of AF.


Assuntos
Fibrilação Atrial , Proteínas de Homeodomínio , Fibrilação Atrial/genética , Predisposição Genética para Doença , Proteínas de Homeodomínio/genética , Humanos , Mutação
13.
Genes (Basel) ; 12(3)2021 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-33809104

RESUMO

Atrial fibrillation (AF) represents the most common type of clinical cardiac arrhythmia and substantially increases the risks of cerebral stroke, heart failure and death. Accumulating evidence has convincingly demonstrated the strong genetic basis of AF, and an increasing number of pathogenic variations in over 50 genes have been causally linked to AF. Nevertheless, AF is of pronounced genetic heterogeneity, and the genetic determinants underpinning AF in most patients remain obscure. In the current investigation, a Chinese pedigree with AF as well as ventricular arrhythmias and hypertrophic cardiomyopathy was recruited. Whole exome sequencing and bioinformatic analysis of the available family members were conducted, and a novel heterozygous variation in the KLF15 gene (encoding Krüppel-like factor 15, a transcription factor critical for cardiac electrophysiology and structural remodeling), NM_014079.4: c.685A>T; p.(Lys229*), was identified. The variation was verified by Sanger sequencing and segregated with autosomal dominant AF in the family with complete penetrance. The variation was absent from 300 unrelated healthy subjects used as controls. In functional assays using a dual-luciferase assay system, mutant KLF15 showed neither transcriptional activation of the KChIP2 promoter nor transcriptional inhibition of the CTGF promoter, alone or in the presence of TGFB1, a key player in the pathogenesis of arrhythmias and cardiomyopathies. The findings indicate KLF15 as a new causative gene responsible for AF as well as ventricular arrhythmias and hypertrophic cardiomyopathy, and they provide novel insight into the molecular mechanisms underlying cardiac arrhythmias and hypertrophic cardiomyopathy.


Assuntos
Arritmias Cardíacas/genética , Fibrilação Atrial/genética , Cardiomiopatias/genética , Predisposição Genética para Doença/genética , Fatores de Transcrição Kruppel-Like/genética , Mutação/genética , Adolescente , Adulto , Idoso , Animais , Povo Asiático/genética , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Células HeLa , Heterozigoto , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Células NIH 3T3 , Linhagem , Regiões Promotoras Genéticas/genética , Ativação Transcricional/genética , Adulto Jovem
14.
Eur J Med Genet ; 64(5): 104211, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33794346

RESUMO

As the most prevalent form of human birth defect, congenital heart disease (CHD) contributes to substantial morbidity, mortality and socioeconomic burden worldwide. Aggregating evidence has convincingly demonstrated that genetic defects exert a pivotal role in the pathogenesis of CHD, and causative mutations in multiple genes have been causally linked to CHD. Nevertheless, CHD is of pronounced genetic heterogeneity, and the genetic components underpinning CHD in the overwhelming majority of patients remain obscure. In this research, a four-generation consanguineous family suffering from CHD transmitted in an autosomal dominant mode was recruited. By whole-exome sequencing and bioinformatics analyses as well as Sanger sequencing analyses of the family members, a new heterozygous SOX17 variation, NM_022454.4: c.553G > T; p.(Glu185*), was identified to co-segregate with CHD in the family, with complete penetrance. The nonsense variation was neither detected in 310 unrelated healthy volunteers used as controls nor retrieved in such population genetics databases as the Exome Aggregation Consortium database, Genome Aggregation Database, and the Single Nucleotide Polymorphism database. Functional assays by utilizing a dual-luciferase reporter assay system unveiled that the Glu185*-mutant SOX17 protein had no transcriptional activity on its two target genes NOTCH1 and GATA4, which have been reported to cause CHD. Furthermore, the mutation abrogated the synergistic transactivation between SOX17 and NKX2.5, another established CHD-causing transcription factor. These findings firstly indicate SOX17 loss-of-function mutation predisposes to familial CHD, which adds novel insight to the molecular mechanism of CHD, implying potential implications for genetic risk appraisal and individualized prophylaxis of the family members affected with CHD.


Assuntos
Cardiopatias Congênitas/genética , Mutação com Perda de Função , Fatores de Transcrição SOXF/genética , Adolescente , Adulto , Animais , Células COS , Criança , Chlorocebus aethiops , Códon sem Sentido , Feminino , Células HeLa , Cardiopatias Congênitas/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Penetrância , Fatores de Transcrição SOXF/metabolismo
15.
Medicine (Baltimore) ; 100(15): e25551, 2021 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-33847683

RESUMO

BACKGROUND: The aim of the study was to evaluate the efficacy of nicorandil and alprostadil on myocardial protection in patients undergoing elective percutaneous coronary intervention (PCI). METHODS: In this prospective, single-blinded, randomized controlled study, 90 consecutive patients scheduled for elective PCI for de novo coronary lesions were assigned to the nicorandil, alprostadil, and nitroglycerin groups in a 1:1:1 ratio. Drugs were administered intracoronary via a targeted perfusion microcatheter. The primary endpoint was the thrombolysis in myocardial infarction (TIMI) myocardial perfusion frame count (TMPFC). Additionally, the corrected TIMI frame count (cTFC), TIMI myocardial perfusion grade (TMPG), and incidence of periprocedural myocardial injury (PMI) were assessed. RESULTS: Both nicorandil and alprostadil were significantly effective in reducing TMPFC (114.6 ±â€Š33.7 vs 93.4 ±â€Š30.9, P = .016; 114.3 ±â€Š34.3 vs 94.7 ±â€Š33.3, P = .029, respectively). Similar findings were observed in the improvement of cTFC (20.3 ±â€Š10.5 vs 13.5 ±â€Š5.0, P = .003; 20.2 ±â€Š7.4 vs 15.2 ±â€Š5.2, P = .003, respectively) and percentage of TMPG 3 (100% vs 82.8%, P = .052; 83.3% vs 96.7%, P = .196, respectively); whereas, nitroglycerin produced a limited effect on TMPFC (114.4 ±â€Š30.9 vs 112.1 ±â€Š31.9, P = .739), cTFC (19.4 ±â€Š7.2 vs 19.3 ±â€Š7.2, P = .936), and percentage of TMPG 3 (86.7% vs 86.7%, P = 1.000). No significant difference was found in the incidence of PMI (16.7% vs 16.0% vs 27.6%, P = .537), though it was comparatively lower in the nicorandil and alprostadil groups. Furthermore, the intracoronary administration of nicorandil and alprostadil had a mild effect on blood pressure and heart rate. CONCLUSIONS: The intracoronary administration of nicorandil and alprostadil via a targeted perfusion microcatheter was more effective in improving myocardial perfusion in patients undergoing elective PCI than nitroglycerin.


Assuntos
Alprostadil/administração & dosagem , Cardiotônicos/administração & dosagem , Infarto do Miocárdio/terapia , Nicorandil/administração & dosagem , Nitroglicerina/administração & dosagem , Vasodilatadores/administração & dosagem , Idoso , Vias de Administração de Medicamentos , Procedimentos Cirúrgicos Eletivos/métodos , Feminino , Coração/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Intervenção Coronária Percutânea/métodos , Perfusão , Estudos Prospectivos , Método Simples-Cego , Resultado do Tratamento
16.
Clin Chem Lab Med ; 59(5): 955-963, 2021 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-33554560

RESUMO

OBJECTIVES: Dilated cardiomyopathy (DCM) represents the most frequent form of cardiomyopathy, leading to heart failure, cardiac arrhythmias and death. Accumulating evidence convincingly demonstrates the crucial role of genetic defects in the pathogenesis of DCM, and over 100 culprit genes have been implicated with DCM. However, DCM is of substantial genetic heterogeneity, and the genetic determinants underpinning DCM remain largely elusive. METHODS: Whole-exome sequencing and bioinformatical analyses were implemented in a consanguineous Chinese family with DCM. A total of 380 clinically annotated control individuals and 166 more DCM index cases then underwent Sanger sequencing analysis for the identified genetic variation. The functional characteristics of the variant were delineated by utilizing a dual-luciferase assay system. RESULTS: A heterozygous variation in the MEF2A gene (encoding myocyte enhancer factor 2A, a transcription factor pivotal for embryonic cardiogenesis and postnatal cardiac adaptation), NM_001365204.1: c.718G>T; p. (Gly240*), was identified, and verified by Sanger sequencing to segregate with autosome-dominant DCM in the family with complete penetrance. The nonsense variation was neither detected in 760 control chromosomes nor found in 166 more DCM probands. Functional analyses revealed that the variant lost transactivation on the validated target genes MYH6 and FHL2, both causally linked to DCM. Furthermore, the variation nullified the synergistic activation between MEF2A and GATA4, another key transcription factor involved in DCM. CONCLUSIONS: The findings firstly indicate that MEF2A loss-of-function variation predisposes to DCM in humans, providing novel insight into the molecular mechanisms of DCM and suggesting potential implications for genetic testing and prognostic evaluation of DCM patients.


Assuntos
Cardiomiopatia Dilatada , Cardiomiopatia Dilatada/genética , Análise Mutacional de DNA , Heterozigoto , Humanos , Proteínas com Homeodomínio LIM , Fatores de Transcrição MEF2/genética , Proteínas Musculares , Linhagem , Fatores de Transcrição
17.
Heart Rhythm ; 18(5): 684-693, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33429106

RESUMO

BACKGROUND: Atrial fibrillation (AF) represents the most common clinical cardiac arrhythmia and substantially increases the risk of cerebral stroke, heart failure, and death. Although causative genes for AF have been identified, the genetic determinants for AF remain largely unclear. OBJECTIVE: This study aimed to investigate the molecular basis of AF in a Chinese kindred. METHODS: A 4-generation family with autosomal-dominant AF and other arrhythmias (atrioventricular block, sinus bradycardia, and premature ventricular contractions) was recruited. Genome-wide scan with microsatellite markers and linkage analysis as well as whole-exome sequencing analysis were performed. Electrophysiological characteristics and subcellular localization of the AF-linked mutant were analyzed using dual whole-cell patch clamps and confocal microscopy, respectively. RESULTS: A novel genetic locus for AF was mapped to chromosome 17q21.3, a 3.23-cM interval between markers D17S951 and D17S931, with a maximum 2-point logarithm of odds score of 4.2144 at marker D17S1868. Sequencing analysis revealed a heterozygous mutation in the mapping region, NM_005497.4:c.703A>T;p.(M235L), in the GJC1 gene encoding connexin45 (Cx45). The mutation cosegregated with AF in the family and was absent in 632 control individuals. The mutation decreased the coupling conductance in cell pairs (M235L/M235L, M235L/Cx45, M235L/Cx43, and M235L/Cx40), likely because of impaired subcellular localization. CONCLUSION: This study defines a novel genetic locus for AF on chromosome 17q21.3 and reveals a loss-of-function mutation in GJC1 (Cx45) contributing to AF and other cardiac arrhythmias.


Assuntos
Fibrilação Atrial/genética , Doença do Sistema de Condução Cardíaco/genética , Conexinas/genética , DNA/genética , Eletrocardiografia/métodos , Adolescente , Adulto , Idoso , Fibrilação Atrial/fisiopatologia , Doença do Sistema de Condução Cardíaco/fisiopatologia , Criança , Conexinas/metabolismo , Análise Mutacional de DNA , Feminino , Sistema de Condução Cardíaco/fisiopatologia , Heterozigoto , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Linhagem , Adulto Jovem
18.
Genet Mol Biol ; 43(4): e20200142, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33306779

RESUMO

TBX5 has been linked to Holt-Oram syndrome, with congenital heart defect (CHD) and atrial fibrillation (AF) being two major cardiac phenotypes. However, the prevalence of a TBX5 variation in patients with CHD and AF remains obscure. In this research, by sequencing analysis of TBX5 in 178 index patients with both CHD and AF, a novel heterozygous variation, NM_000192.3: c.577G>T; p.(Gly193*), was identified in one index patient with CHD and AF as well as bicuspid aortic valve (BAV), with an allele frequency of approximately 0.28%. Genetic analysis of the proband's pedigree showed that the variation co-segregated with the diseases. The pathogenic variation was not detected in 292 unrelated healthy subjects. Functional analysis by using a dual-luciferase reporter assay system showed that the Gly193*-mutant TBX5 protein failed to transcriptionally activate its target genes MYH6 and NPPA. Moreover, the mutation nullified the synergistic transactivation between TBX5 and GATA4 as well as NKX2-5. Additionally, whole-exome sequencing analysis showed no other genes contributing to the diseases. This investigation firstly links a pathogenic variant in the TBX5 gene to familial CHD and AF as well as BAV, suggesting that CHD and AF as well as BAV share a common developmental basis in a subset of patients.

19.
Eur J Med Genet ; 63(11): 104029, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32771629

RESUMO

Atrial fibrillation (AF) represents the most frequent form of sustained cardiac rhythm disturbance, affecting approximately 1% of the general population worldwide, and confers a substantially enhanced risk of cerebral stroke, heart failure, and death. Increasing epidemiological studies have clearly demonstrated a strong genetic basis for AF, and variants in a wide range of genes, including those coding for ion channels, gap junction channels, cardiac structural proteins and transcription factors, have been identified to underlie AF. Nevertheless, the genetic pathogenesis of AF is complex and still far from completely understood. Here, whole-exome sequencing and bioinformatics analyses of a three-generation family with AF were performed, and after filtering variants by multiple metrics, we identified a heterozygous variant in the ISL1 gene (encoding a transcription factor critical for embryonic cardiogenesis and postnatal cardiac remodeling), NM_002202.2: c.481G > T; p.(Glu161*), which was validated by Sanger sequencing and segregated with autosome-dominant AF in the family with complete penetrance. The nonsense variant was absent from 284 unrelated healthy individuals used as controls. Functional assays with a dual-luciferase reporter assay system revealed that the truncating ISL1 protein lost transcriptional activation on the verified target genes MEF2C and NKX2-5. Additionally, the variant nullified the synergistic transactivation between ISL1 and TBX5 as well as GATA4, two other transcription factors that have been implicated in AF. The findings suggest ISL1 as a novel gene contributing to AF, which adds new insight to the genetic mechanisms underpinning AF, implying potential implications for genetic testing and risk stratification of the AF family members.


Assuntos
Fibrilação Atrial/genética , Proteínas com Homeodomínio LIM/genética , Mutação com Perda de Função , Fatores de Transcrição/genética , Adulto , Idoso , Fibrilação Atrial/patologia , Códon sem Sentido , Feminino , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Células HEK293 , Proteína Homeobox Nkx-2.5/genética , Proteína Homeobox Nkx-2.5/metabolismo , Humanos , Proteínas com Homeodomínio LIM/metabolismo , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Masculino , Pessoa de Meia-Idade , Linhagem , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/metabolismo
20.
J Cardiovasc Electrophysiol ; 31(3): 621-628, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31930545

RESUMO

BACKGROUND: Catheter ablation (CA) is a promising option in most patients with refractory atrial fibrillation (AF). However, data on over 5 years' outcomes with larger numbers in hypertrophic cardiomyopathy (HCM) patients with AF have not been reported. We assessed the outcome of 120 HCM patients following CA compared with a non-CA group and general patients without AF matched by HCM type with a 61.9 ± 31.6-month follow-up. METHODS AND RESULTS: A total of 120 patients (age 61 ± 9.8 years, female n = 43, 35.8%) with paroxysmal AF (n = 60, 50%) and persistent AF (n = 60, 50%) were enrolled. Of the 120 patients, 48 (40%) required redo procedures, and 82 (68.3%) were in sinus rhythm at the last evaluation. The composite clinical events rate following the initial CA was lower than that in the non-CA group (P = .023) and was also comparable to that in general patients without AF matched by HCM type (P = .729). Female (HR 2.358, 95% CI, 1.151-4.831; P = .019), NYHA functional class III-IV (HR 2.422, 95% CI, 1.032-5.685; P = .042) and left atrial diameter ≥50 mm (HR 3.319, 95% CI, 1.469-7.499; P = .004) were predictors of AF recurrence after multiple procedures. CONCLUSIONS: CA was successful in restoring long-term sinus rhythm and improving symptomatic status in most HCM patients with refractory AF especially for those patients with small atrial size and mild symptoms. In addition, CA may contribute to the prevention of major clinical adverse events in the long-term clinical course.


Assuntos
Fibrilação Atrial/cirurgia , Cardiomiopatia Hipertrófica/complicações , Ablação por Cateter , Frequência Cardíaca , Idoso , Fibrilação Atrial/diagnóstico , Fibrilação Atrial/etiologia , Fibrilação Atrial/fisiopatologia , Cardiomiopatia Hipertrófica/diagnóstico por imagem , Cardiomiopatia Hipertrófica/fisiopatologia , Ablação por Cateter/efeitos adversos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva , Reoperação , Estudos Retrospectivos , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA