Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Heart Rhythm ; 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38885753

ABSTRACT

BACKGROUND: The electrocardiogram-based algorithm for predicting para-septal atrial tachycardia (PSAT) is limited by the significant overlaps in the P-wave morphology originating from various para-septal sites. OBJECTIVE: The aim of this study was to investigate the endocardial activation characteristics of PSAT and to seek an endocardial activation-derived predictor for the ablation site. METHODS: Forty-four patients (11 males, average 62.6±14.7 years) with PSAT ablation in four tertiary medical centers were assigned into three groups according to the ablation site: right atrial (RA) para-Hisian region (Group 1, n=10), the noncoronary cusp (NCC) (Group 2, n=13) and left atrial (LA) para-septal areas (Group 3, n=21). Multiple-chamber activation mapping was performed guided by a 3D navigation system. The discrepancies in the earliest activation time between two of three chambers (ΔRA-LA, ΔRA-NCC and ΔLA-NCC) were calculated in each group and used for pairwise comparisons. RESULTS: There was significant difference in ΔRA-LA, ΔRA-NCC, and ΔLA-NCC compared among three groups. ΔRA-LA was the only parameter which could consistently predict the ablation site of PSAT with good accuracy (AUC 1.000, sensitivity 100%/ specificity 100%, and cut-off value 7ms for predicting right para-Hisian or NCC ablation; AUC 0.974, sensitivity 92.3%/specificity 95.2%, and cut-off value -4 ms for predicting NCC or left para-septal ablation). Based on two cut-off values, A two-step algorithm was developed to predict the ablation site of PSAT with positive predictive value of 95.4% and negative predictive value of 97.0%. CONCLUSIONS: ΔRA-LA is a useful endocardial activation-derived parameter for predicting the successful ablation site of PSAT.

2.
Biology (Basel) ; 12(9)2023 Aug 30.
Article in English | MEDLINE | ID: mdl-37759586

ABSTRACT

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.
Diagnostics (Basel) ; 13(2)2023 Jan 09.
Article in English | MEDLINE | ID: mdl-36673052

ABSTRACT

Dilated cardiomyopathy (DCM), characterized by left ventricular or biventricular enlargement with systolic dysfunction, is the most common type of cardiac muscle disease. It is a major cause of congestive heart failure and the most frequent indication for heart transplantation. Aggregating evidence has convincingly demonstrated that DCM has an underlying genetic basis, though the genetic defects responsible for DCM in a larger proportion of cases remain elusive, motivating the ongoing research for new DCM-causative genes. In the current investigation, a multigenerational family affected with autosomal-dominant DCM was recruited from the Chinese Han population. By whole-exome sequencing and Sanger sequencing analyses of the DNAs from the family members, a new BMP10 variation, NM_014482.3:c.166C > T;p.(Gln56*), was discovered and verified to be in co-segregation with the DCM phenotype in the entire family. The heterozygous BMP10 variant was not detected in 268 healthy volunteers enrolled as control subjects. The functional measurement via dual-luciferase reporter assay revealed that Gln56*-mutant BMP10 lost the ability to transactivate its target genes NKX2.5 and TBX20, two genes that had been causally linked to DCM. The findings strongly indicate BMP10 as a new gene contributing to DCM in humans and support BMP10 haploinsufficiency as an alternative pathogenic mechanism underpinning DCM, implying potential implications for the early genetic diagnosis and precision prophylaxis of DCM.

4.
J Am Heart Assoc ; 11(22): e027578, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36346048

ABSTRACT

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.


Subject(s)
Cardiomyopathy, Dilated , Humans , Cardiomyopathy, Dilated/metabolism , Mutation , Pedigree , Repressor Proteins/genetics , Cell Cycle Proteins/genetics , Kruppel-Like Transcription Factors/genetics
5.
Genet Mol Biol ; 45(2): e20210378, 2022.
Article in English | MEDLINE | ID: mdl-35377386

ABSTRACT

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.

6.
J Am Heart Assoc ; 10(23): e023517, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34845933

ABSTRACT

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.


Subject(s)
Atrial Fibrillation , Homeodomain Proteins , Atrial Fibrillation/genetics , Genetic Predisposition to Disease , Homeodomain Proteins/genetics , Humans , Mutation
7.
BMC Cardiovasc Disord ; 21(1): 471, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34592933

ABSTRACT

BACKGROUND: The association between anxiety and atrial fibrillation (AF) remains unclear. Moreover, this association has rarely been studied in Chinese individuals aged 60 years or older. This study investigated the association between anxiety and AF in a community-based case-control study of older adult residents in urban China. METHODS: The cases and controls were from a community-based study conducted in the Jingansi community in Shanghai, China, between January 2010 and December 2012. A total of 3622 residents aged 60 years or older without severe vision, hearing, or speaking impairments were eligible to participate in the physical examinations and questionnaire survey. AF was assessed based on a previous physician's diagnosis, electrocardiogram, ambulatory electrocardiogram, or echocardiogram. Anxiety was evaluated using the Zung Self-Rating Anxiety Scale (ZSAS). Using the AF group as a reference, the control group consisted of randomly selected age- and sex-matched individuals in a 1:5 ratio (case:control = 1:5). The association between anxiety and AF in the AF group and the multifactor-matched control group was explored using logistic regression. RESULTS: In the AF and control groups, after adjusting for a history of coronary heart disease, valvular heart disease, hypertension, stroke, hyperlipidemia, and diabetes, as well as depression score, ZSAS scores (odds ratio 1.07; 95% confidence interval 1.02-1.12; p = 0.003), and anxiety symptoms (odds ratio 3.94; 95% confidence interval 1.06-14.70; p = 0.041) were associated with AF. CONCLUSIONS: Anxiety symptoms were associated with AF in a Chinese older population. This suggests that older adults who have anxiety symptoms may need psychological intervention or treatment in daily life and care.


Subject(s)
Anxiety/epidemiology , Atrial Fibrillation/epidemiology , Age Factors , Aged , Aged, 80 and over , Anxiety/diagnosis , Anxiety/physiopathology , Atrial Fibrillation/diagnosis , Atrial Fibrillation/psychology , Case-Control Studies , China/epidemiology , Female , Humans , Male , Middle Aged , Risk Assessment , Risk Factors
8.
World J Gastroenterol ; 27(6): 470-486, 2021 Feb 14.
Article in English | MEDLINE | ID: mdl-33642822

ABSTRACT

BACKGROUND: Liver cancer is a common cancer and the main cause of cancer-related deaths worldwide. Liver cancer is the sixth most common cancer in the world. Although miR-34a and palmitoyl membrane palmitoylated protein (MPP2) are reportedly involved in various cell processes, their precise roles in liver cancer are still unclear. AIM: To investigate the expression of micro RNA 34a (miR-34a), methylation of the miR-34a promoter and the expression of MPP2 in liver cancer cells and their related mechanisms. METHODS: Together, 78 cases of liver cancer tissues and 78 cases of adjacent tissues were collected. The methylation degree of miR-34a promoter in liver cancer/ paracancerous tissue and liver cancer cells/normal liver cells, and the expression levels of miR-34a and MPP2 in the above samples were detected. Demethylation of liver cancer cells or transfection of liver cancer cells with miR-34a mimetic was performed. The MPP2 overexpression vector was used to transfect liver cancer cells, and the changes in proliferation, invasion, apoptosis, migration, and other biological functions of liver cancer cells after the above interventions were observed. Double luciferase reporter genes were used to detect the targeting relationship between miR-34a and MPP2. RESULTS: Clinical samples showed that the expression levels of miR-34a and MPP2 in liver cancer tissues were lower than those in the normal tissues. The methylation degree of miR-34a promoter region in liver cancer cells was higher than that in normal liver cells. After miR-34a demethylation/mimetic transfection/MPP2 overexpression, the apoptosis of liver cancer cells was increased; the proliferation, invasion and migration capabilities were decreased; the expression levels of caspase 3, caspase 9, E-cadherin, and B-cell lymphoma 2 (Bcl-2)-associated X protein were increased; and the expression levels of Bcl-2, N-cadherin, and ß-catenin were decreased. Double luciferase reporter genes confirmed that MPP2 is targeted by miR-34a. Rescue experiments showed that small interfering MPP2 could counteract the promoting effect of miR-34a demethylation on apoptosis and the inhibitory effect on cell proliferation, invasion, and migration. CONCLUSION: miR-34a demethylation upregulates the expression level of MPP2 in liver cancer cells and promotes the apoptosis of liver cancer cells. miR-34a demethylation is a potential method for liver cancer treatment.


Subject(s)
Apoptosis , Demethylation , Liver Neoplasms , MicroRNAs , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Lipoylation , Liver Neoplasms/genetics , Membrane Proteins/genetics , MicroRNAs/genetics , MicroRNAs/metabolism
9.
Int Heart J ; 61(4): 761-768, 2020 Jul 30.
Article in English | MEDLINE | ID: mdl-32641638

ABSTRACT

Congenital heart defect (CHD) represents the most common birth deformity, afflicting 1% of all births worldwide, and accounts for substantial morbidity and mortality. Increasing evidence highlights the pivotal roles of genetic etiologies in the pathogenesis of CHD, and pathogenic mutations in multiple genes, including TBX5 encoding a cardiac core transcription factor key to cardiovascular morphogenesis, have been involved in CHD. However, due to pronounced genetic heterogeneity of CHD, the genetic determinants underlying CHD in most cases remain obscure. In this investigation, by sequencing analysis of the coding exons and flanking introns of the TBX5 gene in 198 unrelated patients affected with CHD, a novel heterozygous mutation, NM_000192.3: c.692C>T; p. (Pro231Leu), was identified in an index patient with familial double outlet right ventricle (DORV), ventricular septal defect (VSD), and atrioventricular block (AVB). Genetic analysis of the proband's pedigree showed that the mutation co-segregated with the diseases. The missense mutation, which altered the amino acid conserved evolutionarily, was absent from 266 unrelated healthy subjects. Functional analyses with a dual-luciferase reporter assay system unveiled that the Pro231Leu-mutant TBX5 was associated with significantly reduced transcriptional activity on its target genes MYH6 and NPPA. Furthermore, the mutation disrupted the synergistic transactivation between TBX5 and NKX2-5 as well as GATA4, two other transcription factors causally linked to CHD. This study firstly links TBX5 loss-of-function mutation to familial DORV, VSD, and AVB, which provides novel insight into the mechanism underpinning CHD and AVB, suggesting potential implications for genetic evaluation and individualized treatment of patients affected by CHD and AVB.


Subject(s)
Atrioventricular Block/genetics , Heart Defects, Congenital/genetics , T-Box Domain Proteins/genetics , Adolescent , Adult , Animals , Case-Control Studies , Cattle , Child , Child, Preschool , Dogs , Female , Humans , Infant , Male , Mice , Middle Aged , Mutation, Missense , Rats , Young Adult
10.
Atherosclerosis ; 274: 199-205, 2018 07.
Article in English | MEDLINE | ID: mdl-29800789

ABSTRACT

BACKGROUND AND AIMS: Agonists of peroxisome proliferator-activated receptor gamma (Pparγ) have been demonstrated to reduce the risk of myocardial infarction (MI) in clinical trials and animal experiments. However, the cellular and molecular mechanisms are not completely understood. We aimed to reveal the functions of myeloid Pparγ in MI and explore the potential mechanisms in this study. METHODS: Myeloid Pparγ knockout (MPGKO) mice (n = 12) and control mice (n = 8) underwent coronary artery ligation to induce MI. Another cohort of MPGKO mice and control mice underwent coronary artery ligation and were then treated with IgG or neutralizing antibodies against interleukin (IL)-1ß. Infarct size was determined by TTC staining and cardiac function was measured using echocardiography. Conditioned media from GW9662- or vehicle-treated macrophages were used to treat H9C2 cardiomyocyte cell line. Gene expression was analyzed using quantitative PCR. Reactive oxygen species were measured using flow cytometry. RESULTS: Myeloid Pparγ deficiency significantly increased myocardial infarct size. Cardiac hypertrophy was also exacerbated in MPGKO mice, with upregulation of ß-myosin heavy chain (Mhc) and brain natriuretic peptide (Bnp) and downregulation of α-Mhc in the non-infarcted zone. Conditioned media from GW9662-treated macrophages increased expression of ß-Mhc and Bnp in H9C2 cells. Echocardiographic measurements showed that MPGKO mice had worsen cardiac dysfunction after MI. Myeloid Pparγ deficiency increased gene expression of NADPH oxidase subunits (Nox2 and Nox4) in the non-infarcted zone after MI. Conditioned media from GW9662-treated macrophages increased reactive oxygen species in H9C2 cells. Expression of inflammatory genes such as IL-1ß and IL-6 was upregulated in the non-infarcted zone of MPGKO mice after MI. With the injection of neutralizing antibodies against IL-1ß, control mice and MPGKO mice had comparable cardiac function and expression of inflammatory genes after MI. CONCLUSIONS: Myeloid Pparγ deficiency exacerbates MI, likely through increased oxidative stress and cardiac inflammation.


Subject(s)
Macrophages, Peritoneal/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , PPAR gamma/deficiency , Animals , Cell Line , Disease Models, Animal , Disease Progression , Genetic Predisposition to Disease , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , NADPH Oxidase 2/genetics , NADPH Oxidase 2/metabolism , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Oxidative Stress , PPAR gamma/genetics , Phenotype , Signal Transduction , Time Factors , Ventricular Function, Left , Ventricular Remodeling
11.
Heart Vessels ; 33(7): 722-732, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29445930

ABSTRACT

Dilated cardiomyopathy (DCM) is a common primary myocardial disease leading to congestive heart failure, arrhythmia and sudden cardiac death. Increasing studies demonstrate substantial genetic determinants for DCM. Nevertheless, DCM is of substantial genetic heterogeneity, and the genetic basis for DCM in most patients remains unclear. The present study was sought to investigate the association of a genetic variant in the ZBTB17 gene with DCM. A cohort of 158 unrelated patients with idiopathic DCM and a total of 230 unrelated, ethnically matched healthy individuals used as controls were recruited. The coding exons and splicing boundaries of ZBTB17 were sequenced in all study participants. The functional effect of the mutant ZBTB17 was characterized by a dual-luciferase reporter assay system. A novel heterozygous ZBTB17 mutation, p.E243X, was discovered in an index patient. Genetic scan of the mutation carrier's available relatives showed that the mutation was present in all affected family members but absent in unaffected family members. Analysis of the proband's pedigree revealed that the mutation co-segregated with DCM, which was transmitted in an autosomal dominant pattern with complete penetrance. The nonsense mutation was absent in the 460 control chromosomes. Functional assays demonstrated that the truncated ZBTB17 protein had no transcriptional activity as compared with its wild-type counterpart. This study firstly associates ZBTB17 loss-of-function mutation with enhanced susceptibility to DCM in humans, which provides novel insight into the molecular mechanism underpinning DCM, implying potential implications for genetic counseling and personalized management of DCM.


Subject(s)
Cardiomyopathy, Dilated/genetics , DNA/genetics , Genetic Predisposition to Disease , Kruppel-Like Transcription Factors/genetics , Mutation , Cardiomyopathy, Dilated/metabolism , DNA Mutational Analysis , Exons , Female , Heterozygote , Humans , Kruppel-Like Transcription Factors/metabolism , Male , Middle Aged , Pedigree , Polymerase Chain Reaction , Zinc Fingers
12.
Clin Chem Lab Med ; 56(3): 502-511, 2018 02 23.
Article in English | MEDLINE | ID: mdl-28902616

ABSTRACT

BACKGROUND: The MADS-box transcription factor myocyte enhancer factor 2C (MEF2C) is required for the cardiac development and postnatal adaptation and in mice-targeted disruption of the MEF2C gene results in dilated cardiomyopathy (DCM). However, in humans, the association of MEF2C variation with DCM remains to be investigated. METHODS: The coding regions and splicing boundaries of the MEF2C gene were sequenced in 172 unrelated patients with idiopathic DCM. The available close relatives of the index patient harboring an identified MEF2C mutation and 300 unrelated, ethnically matched healthy individuals used as controls were genotyped for MEF2C. The functional effect of the mutant MEF2C protein was characterized in contrast to its wild-type counterpart by using a dual-luciferase reporter assay system. RESULTS: A novel heterozygous MEF2C mutation, p.Y157X, was detected in an index patient with adult-onset DCM. Genetic screen of the mutation carrier's family members revealed that the mutation co-segregated with DCM, which was transmitted as an autosomal dominant trait with complete penetrance. The non-sense mutation was absent in 300 control individuals. Functional analyses unveiled that the mutant MEF2C protein had no transcriptional activity. Furthermore, the mutation abolished the synergistic transactivation between MEF2C and GATA4 as well as HAND1, two other transcription factors that have been associated with DCM. CONCLUSIONS: This study indicates MEF2C as a new gene responsible for human DCM, which provides novel insight into the mechanism underpinning DCM, suggesting potential implications for development of innovative prophylactic and therapeutic strategies for DCM, the most prevalent form of primary myocardial disease.


Subject(s)
Cardiomyopathy, Dilated/genetics , Adult , Cardiomyopathy, Dilated/metabolism , Female , HeLa Cells , Humans , MEF2 Transcription Factors/deficiency , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Male , Middle Aged , Mutation , Tumor Cells, Cultured
13.
Clin Chem Lab Med ; 55(9): 1417-1425, 2017 Aug 28.
Article in English | MEDLINE | ID: mdl-28099117

ABSTRACT

BACKGROUND: The zinc finger transcription factor CASZ1 plays a key role in cardiac development and postnatal adaptation, and in mice, deletion of the CASZ1 gene leads to dilated cardiomyopathy (DCM). However, in humans whether genetically defective CASZ1 contributes to DCM remains unclear. METHODS: The coding exons and splicing junction sites of the CASZ1 gene were sequenced in 138 unrelated patients with idiopathic DCM. The available family members of the index patient harboring an identified CASZ1 mutation and 200 unrelated, ethnically matched healthy individuals used as controls were genotyped for CASZ1. The functional characteristics of the mutant CASZ1 were analyzed in contrast to its wild-type counterpart using a luciferase reporter assay system. RESULTS: A novel heterozygous CASZ1 mutation, p.K351X, was identified in an index patient with DCM. Genetic analysis of the mutation carrier's family showed that the mutation co-segregated with DCM, which was transmitted in an autosomal dominant pattern with complete penetrance. The nonsense mutation, which was absent in 400 referential chromosomes, altered the amino acid that was highly conserved evolutionarily. Biological investigations revealed that the mutant CASZ1 had no transcriptional activity. CONCLUSIONS: The current study reveals CASZ1 as a new gene responsible for human DCM, which provides novel mechanistic insight and potential therapeutic target for CASZ1-associated DCM, implying potential implications in improved prophylactic and therapeutic strategies for DCM, the most common type of primary myocardial disease.


Subject(s)
Cardiomyopathy, Dilated/genetics , DNA-Binding Proteins/genetics , Transcription Factors/genetics , Cardiomyopathy, Dilated/metabolism , Cells, Cultured , DNA-Binding Proteins/metabolism , Female , HEK293 Cells , Humans , Male , Middle Aged , Mutation , Transcription Factors/metabolism
14.
G3 (Bethesda) ; 6(4): 987-92, 2016 04 07.
Article in English | MEDLINE | ID: mdl-26865696

ABSTRACT

Congenital heart disease (CHD) is the most common developmental abnormality, and is the leading noninfectious cause of mortality in neonates. Increasing evidence demonstrates that genetic defects play an important role in the pathogenesis of CHD. However, CHD exhibits substantial heterogeneity, and the genetic determinants for CHD remain unknown in the overwhelming majority of cases. In the current study, the coding exons and flanking introns of the HAND2 gene, which encodes a basic helix-loop-helix transcription factor essential for normal cardiovascular development, were sequenced in 192 unrelated patients with CHD, and a novel heterozygous mutation, p.S65I, was identified in a patient with congenital ventricular septal defect (VSD). Genetic analysis of the index patient's pedigree revealed that the mutation was present in all seven affected family members available, but absent in the 13 unaffected family members examined. Besides, in addition to VSD, five of the proband's close relatives also had pulmonary stenosis (PS), and the proband's son also had double outlet right ventricle (DORV). The missense mutation, which altered an evolutionarily conserved amino acid, was absent in 300 unrelated, ethnically matched healthy individuals. Biological analyses using a dual-luciferase reporter assay system showed that the mutant HAND2 was associated with significantly diminished transcriptional activity. Furthermore, the mutation abolished the synergistic activation between HAND2 and GATA4, as well as NKX2.5-two other cardiac core transcriptional factors that have been causally linked to CHD. These findings indicate that HAND2 loss-of-function mutation contributes to human CHD, perhaps via its interaction with GATA4 and NKX2.5.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Genetic Association Studies , Heart Septal Defects, Ventricular/genetics , Mutation , Pulmonary Valve Stenosis/genetics , Adolescent , Alleles , Amino Acid Substitution , Atrial Natriuretic Factor/genetics , Cell Line , Child , Child, Preschool , Codon , DNA Mutational Analysis , Female , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Heart Septal Defects, Ventricular/diagnosis , Homeobox Protein Nkx-2.5/genetics , Homeobox Protein Nkx-2.5/metabolism , Humans , Infant , Infant, Newborn , Male , Pedigree , Promoter Regions, Genetic , Pulmonary Valve Stenosis/diagnosis , Transcription Factors/genetics , Transcriptional Activation , Young Adult
15.
Gene ; 577(2): 258-64, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26657035

ABSTRACT

Congenital heart disease (CHD) is the most prevalent developmental abnormality in humans and is the most common non-infectious cause of infant morbidity and mortality. Increasing evidence demonstrates that genetic defects are involved in the pathogenesis of CHD. However, CHD is genetically heterogeneous, and the genetic determinants underpinning CHD in most patients remain unknown. In this study, the whole coding region of the PITX2 gene (isoform c) was sequenced in 185 unrelated patients with CHD. The available relatives of a mutation carrier and 300 unrelated healthy individuals used as controls were also genotyped for PITX2. The functional characteristics of the mutation were delineated by using a dual-luciferase reporter assay system. As a result, a novel heterozygous PITX2 mutation, p.Q102L, was identified in a patient with tetralogy of Fallot (TOF). Genetic analysis of the index patient's pedigree showed that the mutation co-segregated with TOF. The mutation was absent in 600 reference chromosomes. Biochemical analysis revealed that the Q102L-mutant PITX2 is associated with significantly reduced transcriptional activity compared with its wild-type counterpart. Furthermore, the mutation markedly decreased the synergistic activation between PITX2 and NKX2-5. This study firstly associates PITX2 loss-of-function mutation with increased susceptibility to TOF, providing novel insight into the molecular mechanism of CHD.


Subject(s)
Homeodomain Proteins/genetics , Mutation, Missense , Tetralogy of Fallot/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , CHO Cells , Child, Preschool , Cricetinae , Cricetulus , Female , Homeodomain Proteins/metabolism , Humans , Infant , Male , Molecular Sequence Data , Pedigree , Penetrance , RNA, Messenger/genetics , Transcription Factors/metabolism , Homeobox Protein PITX2
16.
Int J Mol Med ; 34(6): 1581-90, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25319568

ABSTRACT

Atrial fibrillation (AF) is the most common form of sustained cardiac arrhythmia and is associated with substantially increased morbidity and mortality rates. Aggregating evidence demonstrates that genetic defects are involved in the pathogenesis of AF and a number of AF-associated genes have been identified. Nevertheless, AF is a genetically heterogeneous disorder and the genetic components underpinning AF in an overwhelming majority of patients remain unclear. In this study, the entire coding exons and splice junction sites of the NK2 homeobox 6 (NKX2-6) gene, which encodes a homeodomain transcription factor important for cardiovascular development, were sequenced in 150 unrelated patients with lone AF, and a novel heterozygous NKX2-6 mutation, p.Q175H, was identified in an index patient. Genetic analysis of the available family members of the mutation carrier revealed that the mutation co-segregated with AF transmitted in an autosomal dominant pattern. The missense mutation was absent in the 200 unrelated ethnically matched healthy individuals used as controls and the altered amino acid was completely conserved evolutionarily among species. Due to unknown transcriptional targets of NKX2-6, the functional characteristics of the mutation as regards transcriptional activity were analyzed using NKX2-5 as a surrogate. Alignment between human NKX2-6 and NKX2-5 proteins displayed that the Q175H-mutant NKX2-6 was equivalent to the Q181H-mutant NKX2-5, and the introduction of Q181H into NKX2-5 significantly decreased its transcriptional activity at the atrial natriuretic factor promoter. The present study firstly associates genetically defective NKX2-6 with enhanced susceptibility to AF, providing novel insight into the molecular mechanisms underlying AF and suggesting potential strategies for the antenatal prophylaxis and personalized treatment of AF.


Subject(s)
Atrial Fibrillation/genetics , Genetic Predisposition to Disease/genetics , Homeodomain Proteins/genetics , Mutation , Adult , Aged , Amino Acid Sequence , Atrial Fibrillation/physiopathology , Base Sequence , DNA Mutational Analysis , Electrocardiography , Family Health , Female , Genotype , Humans , Male , Middle Aged , Molecular Sequence Data , Pedigree , Phenotype , Sequence Homology, Amino Acid
17.
PLoS One ; 9(8): e104414, 2014.
Article in English | MEDLINE | ID: mdl-25141004

ABSTRACT

A suitable small animal model may help in the screening and evaluation of new drugs, especially those from natural products, which can be administered at lower dosages, fulfilling an urgent worldwide need. In this study, we explore whether zebrafish could be a model organism for carrageenan-induced abdominal edema. The research results showed that intraperitoneal (i.p.) administration of 1.5% λ-carrageenan in a volume of 20 µL significantly increased abdominal edema in adult zebrafish. Levels of the proinflammatory proteins tumor necrosis factor-α (TNF-α) and inducible nitric oxide synthase (iNOS) were increased in carrageenan-injected adult zebrafish during the development of abdominal edema. An associated enhancement was also observed in the leukocyte marker, myeloperoxidase (MPO). To support these results, we further observed that i.p. methylprednisolone (MP; 1 µg), a positive control, significantly inhibited carrageenan-induced inflammation 24 h after carrageenan administration. Furthermore, i.p. pretreatment with either an anti-TNF-α antibody (1∶5 dilution in a volume of 20 µL) or the iNOS-selective inhibitor aminoguanidine (AG; 1 µg) inhibited carrageenan-induced abdominal edema in adult zebrafish. This new animal model is uncomplicated, easy to develop, and involves a straightforward inducement of inflammatory edema for the evaluation of small volumes of drugs or test compounds.


Subject(s)
Edema/metabolism , Inflammation/metabolism , Nitric Oxide Synthase Type II/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Carrageenan , Disease Models, Animal , Edema/chemically induced , Edema/drug therapy , Inflammation/chemically induced , Inflammation/drug therapy , Male , Methylprednisolone/therapeutic use , Peroxidase/metabolism , Zebrafish
18.
Eur J Med Genet ; 57(1): 25-31, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24333117

ABSTRACT

Atrial fibrillation (AF) represents the most prevalent form of sustained cardiac arrhythmia and contributes substantially to cardiovascular morbidity and mortality. Aggregating evidence demonstrates that genetic risk factors play an important role in the pathogenesis of AF. However, AF is a genetically heterogeneous disease and the genetic defects responsible for AF in an overwhelming majority of patients remain unclear. In the present study, the whole coding region and splice junction sites of the PITX2c gene, which encodes a paired-like homeobox transcription factor essential for normal cardiovascular development, were sequenced in 160 unrelated patients with lone AF, and a novel heterozygous mutation, c.349C > T equivalent to p.P117S, was identified in a patient with positive family history of AF. The missense mutation, which co-segregated with AF in the family with complete penetrance and was absent in 700 unrelated ethnically matched healthy individuals, altered the amino acid completely conserved evolutionarily across species and was predicted to be pathogenic by MutationTaster and PolyPhen-2. Biological assays revealed that the mutant PITX2c protein was associated with significantly decreased transcriptional activity when compared with its wild-type counterpart. The findings implicate PITX2c loss-of-function mutation in familial AF for the first time, providing novel insight into the molecular pathology of AF.


Subject(s)
Atrial Fibrillation/genetics , Homeodomain Proteins/genetics , Transcription Factors/genetics , Adult , Amino Acid Sequence , Base Sequence , Case-Control Studies , DNA Mutational Analysis , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Molecular Sequence Data , Mutation, Missense , Pedigree , Homeobox Protein PITX2
19.
Cardiovasc Revasc Med ; 14(5): 302-4, 2013.
Article in English | MEDLINE | ID: mdl-24034863

ABSTRACT

Acute renal artery embolism (RAE) is a clinical rare condition and diagnosis of it is often delayed or missed due to both the rarity of the disease and its non-specific clinical presentation. The exact role and optimal timing of endovascular revascularization remain controversial and uncertain. This article discusses a case of acute renal artery embolism caused by atrial fibrillation. Endovascular renal thrombus aspiration combined with local low-dose thrombolysis reversed the renal ischemia with restoration of renal function despite prolonged ischemia.


Subject(s)
Atrial Fibrillation/complications , Catheterization , Embolism/therapy , Endovascular Procedures , Fibrinolytic Agents/administration & dosage , Ischemia/therapy , Renal Artery Obstruction/therapy , Thrombolytic Therapy , Acute Disease , Atrial Fibrillation/diagnosis , Combined Modality Therapy , Embolism/diagnosis , Embolism/etiology , Humans , Ischemia/diagnosis , Ischemia/etiology , Male , Middle Aged , Renal Artery Obstruction/diagnosis , Renal Artery Obstruction/etiology , Suction , Tomography, X-Ray Computed , Treatment Outcome
20.
Zhonghua Xin Xue Guan Bing Za Zhi ; 41(3): 233-8, 2013 Mar.
Article in Chinese | MEDLINE | ID: mdl-23879950

ABSTRACT

OBJECTIVE: To evaluate the acute and long-term effects of catheter radiofrequency ablation for the treatment of ventricular arrhythmia storm (VAS) post implantable cardioverter-defibrillators (ICD) implantation. METHODS: Acute and long-term effects of catheter radiofrequency ablation for the treatment of VAS post ICD implantation were retrospectively assessed in 11 patients from September 2008 to August 2011. RESULTS: A total of 15 ablation procedures were performed in 11 patients. Six ablation procedures were performed through epicardial approach. In 9 patients, 20 types of ventricular tachycardia (VT) (including 20% hemodynamically unstable VT) were induced during the procedures [mean cycle length (384 ± 141) ms] and polymorphic ventricular tachycardia were induced in 7 patients. The average X-ray fluoroscopy time and procedural time were (26 ± 17) min and (189 ± 60) min, respectively. Complete success, partial success, and failure rates immediately post catheter radiofrequency ablation were 46.7% (7/15), 26.7% (4/15) and 26.7% (4/15), respectively. All patients are alive at follow-up[(2.45 ± 9.6) months after the last catheter ablation] and the complete success, partial success, and failure rates during follow-up were 72.7% (8/11), 9.1% (1/11) and 18.2% (2/11), respectively. CONCLUSION: VAS can be effectively treated by catheter radiofrequency ablation in patients post ICD implantation.


Subject(s)
Catheter Ablation , Defibrillators, Implantable/adverse effects , Tachycardia, Ventricular/surgery , Adult , Aged , Follow-Up Studies , Humans , Male , Middle Aged , Retrospective Studies , Tachycardia, Ventricular/etiology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...