1
|
Emerson JI, Shi W, Paredes-Larios J, Walker WG, Hutton JE, Cristea IM, Marzluff WF, Conlon FL. X-Chromosome-Linked miRNAs Regulate Sex Differences in Cardiac Physiology. Circ Res 2025; 136:258-275. [PMID: 39772608 PMCID: PMC11781965 DOI: 10.1161/circresaha.124.325447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Males and females exhibit distinct anatomic and functional characteristics of the heart, predisposing them to specific disease states. METHODS We identified microRNAs (miRNAs/miR) with sex-differential expression in mouse hearts. RESULTS Four conserved miRNAs are present in a single locus on the X-chromosome and are expressed at higher levels in females than males. We show miRNA, miR-871, is responsible for decreased expression of the protein SRL (sarcalumenin) in females. SRL is involved in calcium signaling, and we show it contributes to differences in electrophysiology between males and females. miR-871 overexpression mimics the effects of the cardiac physiology of conditional cardiomyocyte-specific Srl-null mice. Inhibiting miR-871 with an antagomir in females shortened ventricular repolarization. The human orthologue of miR-871, miR-888, coevolved with the SRL 3' untranslated region and regulates human SRL. CONCLUSIONS These data highlight the importance of sex-differential miRNA mechanisms in mediating sex-specific functions and their potential relevance to human cardiac diseases.
Collapse
Affiliation(s)
- James I. Emerson
- Department of Biochemistry & Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Wei Shi
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jose Paredes-Larios
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - William G. Walker
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Josiah E. Hutton
- Department of Molecular Biology, Princeton University, Lew Thomas Laboratory, Princeton, NJ 08544, USA
| | - Ileana M. Cristea
- Department of Molecular Biology, Princeton University, Lew Thomas Laboratory, Princeton, NJ 08544, USA
| | - William F. Marzluff
- Department of Biochemistry & Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Integrative Program for Biological and Genome Science, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Frank L. Conlon
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Integrative Program for Biological and Genome Science, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
2
|
Uthayakumaran G, Asakai H, Gnanappa G. Pharmacological resynchronisation with flecainide in an infant with Ebstein anomaly and Wolff-Parkinson-White pattern: a case report. Eur Heart J Case Rep 2024; 8:ytae442. [PMID: 39258014 PMCID: PMC11384896 DOI: 10.1093/ehjcr/ytae442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/07/2024] [Accepted: 08/19/2024] [Indexed: 09/12/2024]
Abstract
Background Conduction abnormalities are frequently encountered in patients with Ebstein anomaly. The following case describes the safe use of flecainide in an infant with accessory-pathway mediated left ventricular dysfunction in the setting of Ebstein anomaly. Case Summary An infant with an antenatal diagnosis of Ebstein anomaly developed progressive left ventricular dilatation and dysfunction over the first 2 months of life. ECG demonstrated persistent Wolff-Parkinson-White pattern with delta-wave polarity suggesting a right-sided septal accessory pathway. In the absence of SVT, accessory-pathway mediated dyssynchrony was suspected as the cause for left ventricular dilatation and dysfunction. He was commenced on flecainide which successfully blocked antegrade conduction via the accessory pathway resulting in a reduction in left ventricular volume and improvement in left ventricular systolic function. He remains asymptomatic at 12 months of age. Discussion There is a known association between Ebstein anomaly and Wolff-Parkinson-White pattern. Right-sided septal accessory pathways can cause cardiomyopathy secondary to dyssynchronous left ventricular contraction. In patients who are unsuitable for accessory pathway ablation, flecainide can be used to block antegrade conduction via the accessory pathway resulting in improved left ventricular function, which was successful on this occasion.
Collapse
Affiliation(s)
- Gajon Uthayakumaran
- The Heart Centre for Children, The Children's Hospital at Westmead, Corner Hawkesbury Road and Hainsworth St, Westmead, NSW 2145, Australia
| | - Hiroko Asakai
- The Heart Centre for Children, The Children's Hospital at Westmead, Corner Hawkesbury Road and Hainsworth St, Westmead, NSW 2145, Australia
| | - Ganesh Gnanappa
- The Heart Centre for Children, The Children's Hospital at Westmead, Corner Hawkesbury Road and Hainsworth St, Westmead, NSW 2145, Australia
| |
Collapse
|
3
|
Thareja SK, Anfinson M, Cavanaugh M, Kim MS, Lamberton P, Radandt J, Brown R, Liang HL, Stamm K, Afzal MZ, Strande J, Frommelt MA, Lough JW, Fitts RH, Mitchell ME, Tomita-Mitchell A. Altered contractility, Ca 2+ transients, and cell morphology seen in a patient-specific iPSC-CM model of Ebstein's anomaly with left ventricular noncompaction. Am J Physiol Heart Circ Physiol 2023; 325:H149-H162. [PMID: 37204873 PMCID: PMC10312315 DOI: 10.1152/ajpheart.00658.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 05/21/2023]
Abstract
Patients with two congenital heart diseases (CHDs), Ebstein's anomaly (EA) and left ventricular noncompaction (LVNC), suffer higher morbidity than either CHD alone. The genetic etiology and pathogenesis of combined EA/LVNC remain largely unknown. We investigated a familial EA/LVNC case associated with a variant (p.R237C) in the gene encoding Kelch-like protein 26 (KLHL26) by differentiating induced pluripotent stem cells (iPSCs) generated from affected and unaffected family members into cardiomyocytes (iPSC-CMs) and assessing iPSC-CM morphology, function, gene expression, and protein abundance. Compared with unaffected iPSC-CMs, CMs containing the KLHL26 (p.R237C) variant exhibited aberrant morphology including distended endo(sarco)plasmic reticulum (ER/SR) and dysmorphic mitochondria and aberrant function that included decreased contractions per minute, altered calcium transients, and increased proliferation. Pathway enrichment analyses based on RNASeq data indicated that the "structural constituent of muscle" pathway was suppressed, whereas the "ER lumen" pathway was activated. Taken together, these findings suggest that iPSC-CMs containing this KLHL26 (p.R237C) variant develop dysregulated ER/SR, calcium signaling, contractility, and proliferation.NEW & NOTEWORTHY We demonstrate here that iPSCs derived from patients with Ebstein's anomaly and left ventricular noncompaction, when differentiated into cardiomyocytes, display significant structural and functional changes that offer insight into disease pathogenesis, including altered ER/SR and mitochondrial morphology, contractility, and calcium signaling.
Collapse
Affiliation(s)
- Suma K Thareja
- Division of Congenital Heart Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Melissa Anfinson
- Division of Congenital Heart Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Matthew Cavanaugh
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, United States
| | - Min-Su Kim
- Division of Congenital Heart Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Peter Lamberton
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, United States
| | - Jackson Radandt
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, United States
| | - Ryan Brown
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Huan-Ling Liang
- Division of Congenital Heart Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Karl Stamm
- Division of Congenital Heart Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Muhammad Zeeshan Afzal
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Jennifer Strande
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Michele A Frommelt
- Division of Pediatric Cardiology, Department of Pediatrics, Children's Wisconsin, Milwaukee, Wisconsin, United States
- Herma Heart Institute, Children's Wisconsin, Milwaukee, Wisconsin, United States
| | - John W Lough
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Robert H Fitts
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, United States
| | - Michael E Mitchell
- Division of Congenital Heart Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Herma Heart Institute, Children's Wisconsin, Milwaukee, Wisconsin, United States
| | - Aoy Tomita-Mitchell
- Division of Congenital Heart Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Herma Heart Institute, Children's Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
4
|
Kudryavtseva MM, Kiseleva AV, Myasnikov RP, Kulikova OV, Meshkov AN, Mershina EA, Angarsky RK, Sotnikova ЕA, Divashuk MG, Zharikova AA, Koretsky SN, Filatova DA, Sinitsyn VE, Sdvigova NA, Barsky VI, Basargina EN, Drapkina OM. Nucleotide sequence variant of the TPM1 gene in a family with different phenotypes of left ventricular non-compaction. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2023. [DOI: 10.15829/1728-8800-2022-3471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Left ventricular non-compaction (LVNC) is a rare, genetically and phenotypically heterogeneous disease, which is often accompanied by diagnostic difficulties.Aim. To demonstrate several generations of a family with LVNC with various clinical and phenotypic manifestations of the disease (dilated and isolated types of LVNC) with an identified rs397516387 variant of the TPM1 gene.Material and methods. Based on the multicenter registry "Myocardial Non-compaction", a family with a familial form of LVNC was selected. Next generation sequencing (NGS) was performed on an Ion S5 system (Thermo Fisher Scientific, USA) using Ampliseq technology. Variant was verified using Sanger sequencing on an Applied Biosystem 3500 Genetic Analyzer (Thermo Fisher Scientific, USA). For clinical interpretation, variants in the genes associated with LVNC with a minor allele frequency <0,1% were selected in the gnomAD database (v2.1.1). Results. Variant rs397516387 was found in 5 family members, including the proband. Further examination revealed LVNC in 2 additional family members. The proband and the proband’s uncle had a dilated type of LVNC, and the proband’s mother had an isolated type.Conclusion. The paper presents several generations of a family with different phenotypic manifestations of LVNC and rs397516387 variant in the TPM1 gene. The beginning of genetic screening from a proband, a thorough collection of a family history and further detailed genetic screening of relatives led to the identification of rs397516387 variant in 4 more family members, which in turn made it possible to conduct an additional examination to confirm the diagnosis and prescribe timely drug therapy.
Collapse
Affiliation(s)
| | - A. V. Kiseleva
- National Medical Research Center for Therapy and Preventive Medicine
| | - R. P. Myasnikov
- National Medical Research Center for Therapy and Preventive Medicine
| | - O. V. Kulikova
- National Medical Research Center for Therapy and Preventive Medicine
| | - A. N. Meshkov
- National Medical Research Center for Therapy and Preventive Medicine
| | - E. A. Mershina
- Medical Scientific and Educational Center, Lomonosov Moscow State University
| | - R. K. Angarsky
- National Medical Research Center for Therapy and Preventive Medicine
| | - Е. A. Sotnikova
- National Medical Research Center for Therapy and Preventive Medicine
| | - M. G. Divashuk
- National Medical Research Center for Therapy and Preventive Medicine; 3 All-Russia Research Institute of Agricultural Biotechnology
| | - A. A. Zharikova
- National Medical Research Center for Therapy and Preventive Medicine; Lomonosov Moscow State University, Faculty of Bioengineering and Bioinformatics
| | - S. N. Koretsky
- National Medical Research Center for Therapy and Preventive Medicine
| | - D. A. Filatova
- Medical Scientific and Educational Center, Lomonosov Moscow State University
| | - V. E. Sinitsyn
- Medical Scientific and Educational Center, Lomonosov Moscow State University
| | | | - V. I. Barsky
- National Medical Research Center for Children’s Health
| | | | - O. M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine
| |
Collapse
|
5
|
Finsterer J, Stöllberger C, Laccone F. Noncompaction and the novel variant c.425A>T in TPM1. Acta Cardiol 2022; 77:758-759. [PMID: 34309477 DOI: 10.1080/00015385.2021.1954746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Josef Finsterer
- Messerli Institute, Klinik Landstrasse, Postfach 20, 1180 Vienna, Austria
| | - Claudia Stöllberger
- 2nd Medical Department with Cardiology and Intensive Care Medicine, Klinik Landstrasse, Vienna, Austria
| | - Franco Laccone
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
6
|
Thareja SK, Frommelt MA, Lincoln J, Lough JW, Mitchell ME, Tomita-Mitchell A. A Systematic Review of Ebstein’s Anomaly with Left Ventricular Noncompaction. J Cardiovasc Dev Dis 2022; 9:jcdd9040115. [PMID: 35448091 PMCID: PMC9031964 DOI: 10.3390/jcdd9040115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 03/31/2022] [Accepted: 04/07/2022] [Indexed: 11/16/2022] Open
Abstract
Traditional definitions of Ebstein’s anomaly (EA) and left ventricular noncompaction (LVNC), two rare congenital heart defects (CHDs), confine disease to either the right or left heart, respectively. Around 15–29% of patients with EA, which has a prevalence of 1 in 20,000 live births, commonly manifest with LVNC. While individual EA or LVNC literature is extensive, relatively little discussion is devoted to the joint appearance of EA and LVNC (EA/LVNC), which poses a higher risk of poor clinical outcomes. We queried PubMed, Medline, and Web of Science for all peer-reviewed publications from inception to February 2022 that discuss EA/LVNC and found 58 unique articles written in English. Here, we summarize and extrapolate commonalities in clinical and genetic understanding of EA/LVNC to date. We additionally postulate involvement of shared developmental pathways that may lead to this combined disease. Anatomical variation in EA/LVNC encompasses characteristics of both CHDs, including tricuspid valve displacement, right heart dilatation, and left ventricular trabeculation, and dictates clinical presentation in both age and severity. Disease treatment is non-specific, ranging from symptomatic management to invasive surgery. Apart from a few variant associations, mainly in sarcomeric genes MYH7 and TPM1, the genetic etiology and pathogenesis of EA/LVNC remain largely unknown.
Collapse
Affiliation(s)
- Suma K. Thareja
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (S.K.T.); (J.W.L.)
- Department of Surgery, Division of Congenital Heart Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Michele A. Frommelt
- Department of Pediatrics, Division of Pediatric Cardiology, Children’s Wisconsin, Milwaukee, WI 53226, USA; (M.A.F.); (J.L.)
- Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI 53226, USA
| | - Joy Lincoln
- Department of Pediatrics, Division of Pediatric Cardiology, Children’s Wisconsin, Milwaukee, WI 53226, USA; (M.A.F.); (J.L.)
- Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI 53226, USA
| | - John W. Lough
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (S.K.T.); (J.W.L.)
| | - Michael E. Mitchell
- Department of Surgery, Division of Congenital Heart Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- Department of Pediatrics, Division of Pediatric Cardiology, Children’s Wisconsin, Milwaukee, WI 53226, USA; (M.A.F.); (J.L.)
- Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI 53226, USA
| | - Aoy Tomita-Mitchell
- Department of Surgery, Division of Congenital Heart Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- Department of Pediatrics, Division of Pediatric Cardiology, Children’s Wisconsin, Milwaukee, WI 53226, USA; (M.A.F.); (J.L.)
- Herma Heart Institute, Children’s Wisconsin, Milwaukee, WI 53226, USA
- Correspondence:
| |
Collapse
|
7
|
Rahimi HR, Mojarrad M, Moghbeli M. MicroRNA-96: A therapeutic and diagnostic tumor marker. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:3-13. [PMID: 35656454 DOI: 10.22038/ijbms.2021.59604.13226] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 12/19/2021] [Indexed: 12/17/2022]
Abstract
Cancer has been always considered as one of the main human health challenges worldwide. One of the main causes of cancer-related mortality is late diagnosis in the advanced stages of the disease, which reduces the therapeutic efficiency. Therefore, novel non-invasive diagnostic methods are required for the early detection of tumors and improving the quality of life and survival in cancer patients. MicroRNAs (miRNAs) have pivotal roles in various cellular processes such as cell proliferation, motility, and neoplastic transformation. Since circulating miRNAs have high stability in body fluids, they can be suggested as efficient noninvasive tumor markers. MiR-96 belongs to the miR-183-96-182 cluster that regulates cell migration and tumor progression as an oncogene or tumor suppressor by targeting various genes in solid tumors. In the present review, we have summarized all of the studies that assessed the role of miR-96 during tumor progression. This review clarifies the molecular mechanisms and target genes recruited by miR-96 to regulate tumor progression and metastasis. It was observed that miR-96 mainly affects tumorigenesis by targeting the structural proteins and FOXO transcription factors.
Collapse
Affiliation(s)
- Hamid Reza Rahimi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Mojarrad
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Lin Y, Huang J, Zhu Z, Zhang Z, Xian J, Yang Z, Qin T, Chen L, Huang J, Huang Y, Wu Q, Hu Z, Lin X, Xu G. Overlap phenotypes of the left ventricular noncompaction and hypertrophic cardiomyopathy with complex arrhythmias and heart failure induced by the novel truncated DSC2 mutation. Orphanet J Rare Dis 2021; 16:496. [PMID: 34819141 PMCID: PMC8611834 DOI: 10.1186/s13023-021-02112-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/06/2021] [Indexed: 12/18/2022] Open
Abstract
Background The left ventricular noncompaction cardiomyopathy (LVNC) is a rare subtype of cardiomyopathy associated with a high risk of heart failure (HF), thromboembolism, arrhythmia, and sudden cardiac death. Methods The proband with overlap phenotypes of LVNC and hypertrophic cardiomyopathy (HCM) complicates atrial fibrillation (AF), ventricular tachycardia (VT), and HF due to the diffuse myocardial lesion, which were diagnosed by electrocardiogram, echocardiogram and cardiac magnetic resonance imaging. Peripheral blood was collected from the proband and his relatives. DNA was extracted from the peripheral blood of proband for high-throughput target capture sequencing. The Sanger sequence verified the variants. The protein was extracted from the skin of the proband and healthy volunteer. The expression difference of desmocollin2 was detected by Western blot. Results The novel heterozygous truncated mutation (p.K47Rfs*2) of the DSC2 gene encoding an important component of desmosomes was detected by targeted capture sequencing. The western blots showed that the expressing level of functional desmocollin2 protein (~ 94kd) was lower in the proband than that in the healthy volunteer, indicating that DSC2 p.K47Rfs*2 obviously reduced the functional desmocollin2 protein expression in the proband. Conclusion The heterozygous DSC2 p.K47Rfs*2 remarkably and abnormally reduced the functional desmocollin2 expression, which may potentially induce the overlap phenotypes of LVNC and HCM, complicating AF, VT, and HF.
Collapse
Affiliation(s)
- Yubi Lin
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Jiana Huang
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China.,Reproductive Center, The Six Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510000, China
| | - Zhiling Zhu
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Zuoquan Zhang
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Jianzhong Xian
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Zhe Yang
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Tingfeng Qin
- Department of Physiology, The School of Medicine of Jinan University, Guangzhou, 510000, China
| | - Linxi Chen
- Department of Physiology, The School of Medicine of Jinan University, Guangzhou, 510000, China
| | - Jingmin Huang
- Department of Physiology, The School of Medicine of Jinan University, Guangzhou, 510000, China
| | - Yin Huang
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Qiaoyun Wu
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Zhenyu Hu
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore
| | - Xiufang Lin
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China.
| | - Geyang Xu
- Department of Physiology, The School of Medicine of Jinan University, Guangzhou, 510000, China.
| |
Collapse
|
9
|
Samudrala SSK, North LM, Stamm KD, Earing MG, Frommelt MA, Willes R, Tripathi S, Dsouza NR, Zimmermann MT, Mahnke DK, Liang HL, Lund M, Lin C, Geddes GC, Mitchell ME, Tomita‐Mitchell A. Novel KLHL26 variant associated with a familial case of Ebstein's anomaly and left ventricular noncompaction. Mol Genet Genomic Med 2020; 8:e1152. [PMID: 31985165 PMCID: PMC7196453 DOI: 10.1002/mgg3.1152] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 01/11/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Ebstein's anomaly (EA) is a rare congenital heart disease of the tricuspid valve and right ventricle. Patients with EA often manifest with left ventricular noncompaction (LVNC), a cardiomyopathy. Despite implication of cardiac sarcomere genes in some cases, very little is understood regarding the genetic etiology of EA/LVNC. Our study describes a multigenerational family with at least 10 of 17 members affected by EA/LVNC. METHODS We performed echocardiography on all family members and conducted exome sequencing of six individuals. After identifying candidate variants using two different bioinformatic strategies, we confirmed segregation with phenotype using Sanger sequencing. We investigated structural implications of candidate variants using protein prediction models. RESULTS Exome sequencing analysis of four affected and two unaffected members identified a novel, rare, and damaging coding variant in the Kelch-like family member 26 (KLHL26) gene located on chromosome 19 at position 237 of the protein (GRCh37). This variant region was confirmed by Sanger sequencing in the remaining family members. KLHL26 (c.709C > T p.R237C) segregates only with EA/LVNC-affected individuals (FBAT p < .05). Investigating structural implications of the candidate variant using protein prediction models suggested that the KLHL26 variant disrupts electrostatic interactions when binding to part of the ubiquitin proteasome, specifically Cullin3 (CUL3), a component of E3 ubiquitin ligase. CONCLUSION In this familial case of EA/LVNC, we have identified a candidate gene variant, KLHL26 (p.R237C), which may have an important role in ubiquitin-mediated protein degradation during cardiac development.
Collapse
Affiliation(s)
- Sai Suma K. Samudrala
- Department of Cell Biology, Neurobiology and AnatomyMedical College of WisconsinMilwaukeeWIUSA
| | - Lauren M. North
- Department of Otolaryngology and Communication SciencesMedical College of WisconsinMilwaukeeWIUSA
| | - Karl D. Stamm
- Department of SurgeryDivision of Cardiothoracic SurgeryMedical College of WisconsinMilwaukeeWIUSA
| | - Michael G. Earing
- Department of PediatricsChildren’s Hospital of WisconsinMilwaukeeWIUSA
- Herma Heart InstituteChildren’s Hospital of WisconsinMilwaukeeWIUSA
| | - Michele A. Frommelt
- Department of PediatricsChildren’s Hospital of WisconsinMilwaukeeWIUSA
- Herma Heart InstituteChildren’s Hospital of WisconsinMilwaukeeWIUSA
| | - Richard Willes
- Department of PediatricsChildren’s Hospital of WisconsinMilwaukeeWIUSA
| | - Swarnendu Tripathi
- Bioinformatics Research and Developmental LabGenomic Sciences and Precision Medicine CenterMedical College of WisconsinMilwaukeeWIUSA
| | - Nikita R. Dsouza
- Bioinformatics Research and Developmental LabGenomic Sciences and Precision Medicine CenterMedical College of WisconsinMilwaukeeWIUSA
| | - Michael T. Zimmermann
- Bioinformatics Research and Developmental LabGenomic Sciences and Precision Medicine CenterMedical College of WisconsinMilwaukeeWIUSA
- Clinical and Translational Science InstituteMedical College of WisconsinMilwaukeeWIUSA
| | - Donna K. Mahnke
- Department of SurgeryDivision of Cardiothoracic SurgeryMedical College of WisconsinMilwaukeeWIUSA
| | - Huan Ling Liang
- Department of SurgeryDivision of Cardiothoracic SurgeryMedical College of WisconsinMilwaukeeWIUSA
| | - Michael Lund
- Department of Obstetrics and GynecologyMedical College of WisconsinMilwaukeeWIUSA
| | - Chien‐Wei Lin
- Division of BiostatisticsMedical College of WisconsinMilwaukeeWIUSA
| | | | - Michael E. Mitchell
- Department of SurgeryDivision of Cardiothoracic SurgeryMedical College of WisconsinMilwaukeeWIUSA
- Herma Heart InstituteChildren’s Hospital of WisconsinMilwaukeeWIUSA
| | - Aoy Tomita‐Mitchell
- Department of SurgeryDivision of Cardiothoracic SurgeryMedical College of WisconsinMilwaukeeWIUSA
- Herma Heart InstituteChildren’s Hospital of WisconsinMilwaukeeWIUSA
- Department of Biomedical EngineeringMedical College of WisconsinMilwaukeeWIUSA
| |
Collapse
|
10
|
Wang Z, Song HM, Wang F, Zhao CM, Huang RT, Xue S, Li RG, Qiu XB, Xu YJ, Liu XY, Yang YQ. A New ISL1 Loss-of-Function Mutation Predisposes to Congenital Double Outlet Right Ventricle. Int Heart J 2019; 60:1113-1122. [DOI: 10.1536/ihj.18-685] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Zhi Wang
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine
| | - Hao-Ming Song
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine
| | - Fei Wang
- Department of Neurosurgery, Tongji Hospital, Tongji University School of Medicine
| | - Cui-Mei Zhao
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine
| | - Ri-Tai Huang
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University
| | - Song Xue
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University
| | - Ruo-Gu Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Xing-Biao Qiu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University
| | - Ying-Jia Xu
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University
| | - Xing-Yuan Liu
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine
| | - Yi-Qing Yang
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University
- Department of Cardiovascular Research Laboratory, The Fifth People's Hospital of Shanghai, Fudan University
- Department of Central Laboratory, The Fifth People's Hospital of Shanghai, Fudan University
| |
Collapse
|
11
|
Liu G, Zhao X, Zhou J, Cheng X, Ye Z, Ji Z. Long non-coding RNA MEG3 suppresses the development of bladder urothelial carcinoma by regulating miR-96 and TPM1. Cancer Biol Ther 2018; 19:1039-1056. [PMID: 30461333 DOI: 10.1080/15384047.2018.1480279] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We aimed at investigating effects of long non-coding RNA maternally expressed 3 (MEG3) on the proliferation, cell cycle and apoptosis of bladder urothelial carcinoma cells and regulatory relationships among lncRNA MEG3, miR-96 and α-tropomyosin 1 (TPM1). Human clinical data from The Cancer Genome Atlas (TCGA) which contains bladder urothelial carcinoma tissues and adjacent tissues were used for analysis. The expression profiles of MEG3, miR-96, TPM1, cell cycle-related genes and apoptosis-related genes were examined by real-time quantitative polymerase chain reaction (RT-qPCR) and western blot. Regulating relationship among MEG3, miR-96 and TPM1 was confirmed by dual luciferase reporter assay. MTT assay and flow cytometry were performed to observe cell proliferation, cell cycle and apoptosis. The effects of lncRNA MEG3 on bladder urothelial carcinoma were confirmed both in vivo and in vitro. The mRNA expression and protein expression of MEG3, TPM1 were down-regulated in carcinoma tissues, whereas miR-96 expression was up-regulated. MEG3 overexpression resulted in miR-96 downregulation along with TPM1 upregulation, which inhibited cell proliferation and cell cycle but promoted cell apoptosis of bladder urothelial carcinoma cells in vitro, and at the same time inhibited tumor growth in vivo. In this process, expressions of apoptosis-related protein BCL2 associated X (Bax), cleaved-caspase 3 was up-regulated, whereas apoptosis regulator protein (Bcl-2) expression was suppressed when MEG3 was overexpressed, and cell cycle-related protein Cyclin D1 was down-regulated. LncRNA MEG3 low-expression promotes the proliferation and inhibits apoptosis of bladder urothelial carcinoma cells by regulating miR-96 along with TPM1.
Collapse
Affiliation(s)
- Guanghua Liu
- a Department of Urology , Peking Union Medical College Hospital, Chinese Academy of Medical Science , Beijing , China
| | - Xin Zhao
- a Department of Urology , Peking Union Medical College Hospital, Chinese Academy of Medical Science , Beijing , China
| | - Jingmin Zhou
- a Department of Urology , Peking Union Medical College Hospital, Chinese Academy of Medical Science , Beijing , China
| | - Xiangming Cheng
- a Department of Urology , Peking Union Medical College Hospital, Chinese Academy of Medical Science , Beijing , China
| | - Zixing Ye
- a Department of Urology , Peking Union Medical College Hospital, Chinese Academy of Medical Science , Beijing , China
| | - Zhigang Ji
- a Department of Urology , Peking Union Medical College Hospital, Chinese Academy of Medical Science , Beijing , China
| |
Collapse
|
12
|
Ma L, Wang J, Li L, Qiao Q, Di RM, Li XM, Xu YJ, Zhang M, Li RG, Qiu XB, Li X, Yang YQ. ISL1 loss-of-function mutation contributes to congenital heart defects. Heart Vessels 2018; 34:658-668. [PMID: 30390123 DOI: 10.1007/s00380-018-1289-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 10/26/2018] [Indexed: 02/08/2023]
Abstract
Congenital heart defect (CHD) is the most common form of birth deformity and is responsible for substantial morbidity and mortality in humans. Increasing evidence has convincingly demonstrated that genetic defects play a pivotal role in the pathogenesis of CHD. However, CHD is a genetically heterogeneous disorder and the genetic basis underpinning CHD in the vast majority of cases remains elusive. This study was sought to identify the pathogenic mutation in the ISL1 gene contributing to CHD. A cohort of 210 unrelated patients with CHD and a total of 256 unrelated healthy individuals used as controls were registered. The coding exons and splicing boundaries of ISL1 were sequenced in all study subjects. The functional effect of an identified ISL1 mutation was evaluated using a dual-luciferase reporter assay system. A novel heterozygous ISL1 mutation, c.409G > T or p.E137X, was identified in an index patient with congenital patent ductus arteriosus and ventricular septal defect. Analysis of the proband's pedigree revealed that the mutation co-segregated with CHD, which was transmitted in the family in an autosomal dominant pattern with complete penetrance. The nonsense mutation was absent in 512 control chromosomes. Functional analysis unveiled that the mutant ISL1 protein failed to transactivate the promoter of MEF2C, alone or in synergy with TBX20. This study firstly implicates ISL1 loss-of-function mutation with CHD in humans, which provides novel insight into the molecular mechanism of CHD, implying potential implications for genetic counseling and individually tailored treatment of CHD patients.
Collapse
Affiliation(s)
- Lan Ma
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.,Department of Ultrasound, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Juan Wang
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Li Li
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Qi Qiao
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China
| | - Ruo-Min Di
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China
| | - Xiu-Mei Li
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China
| | - Ying-Jia Xu
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China
| | - Min Zhang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Ruo-Gu Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xing-Biao Qiu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xun Li
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Yi-Qing Yang
- Department of Cardiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China. .,Department of Cardiovascular Research Laboratory, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China. .,Department of Central Laboratory, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, 200240, China.
| |
Collapse
|
13
|
Abstract
Abstract
Ebstein anomaly (EA) is a rare congenital tricuspid valve malformation, characterized by downward displacement of the septal leaflet and an atrialized right ventricle. About 80% of cases of EA are non-syndromic; in the other 20%, the anomaly is associated with a chromosomal or Mendelian syndrome. The prevalence of EA is estimated at about 1 per 20,000 live births, and accounts for less than 1% of all congenital heart defects. EA has autosomal dominant inheritance. Likely causative genes are: NKX2-5, MYH7 and TPM1. This Utility Gene Test was developed on the basis of an analysis of the literature and existing diagnostic protocols. It is useful for confirming diagnosis, as well as for differential diagnosis, potential risk assessment and access to clinical trials.
Collapse
|
14
|
Translating emerging molecular genetic insights into clinical practice in inherited cardiomyopathies. J Mol Med (Berl) 2018; 96:993-1024. [PMID: 30128729 DOI: 10.1007/s00109-018-1685-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 07/22/2018] [Accepted: 08/08/2018] [Indexed: 12/19/2022]
Abstract
Cardiomyopathies are primarily genetic disorders of the myocardium associated with higher risk of life-threatening cardiac arrhythmias, heart failure, and sudden cardiac death. The evolving knowledge in genomic medicine during the last decade has reshaped our understanding of cardiomyopathies as diseases of multifactorial nature and complex pathophysiology. Genetic testing in cardiomyopathies has subsequently grown from primarily a research tool into an essential clinical evaluation piece with important clinical implications for patients and their families. The purpose of this review is to provide with a contemporary insight into the implications of genetic testing in diagnosis, therapy, and prognosis of patients with inherited cardiomyopathies. Here, we summarize the contemporary knowledge on genotype-phenotype correlations in inherited cardiomyopathies and highlight the recent significant achievements in the field of translational cardiovascular genetics.
Collapse
|