1
|
Chatham JC, Patel RP. Protein glycosylation in cardiovascular health and disease. Nat Rev Cardiol 2024; 21:525-544. [PMID: 38499867 DOI: 10.1038/s41569-024-00998-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/13/2024] [Indexed: 03/20/2024]
Abstract
Protein glycosylation, which involves the attachment of carbohydrates to proteins, is one of the most abundant protein co-translational and post-translational modifications. Advances in technology have substantially increased our knowledge of the biosynthetic pathways involved in protein glycosylation, as well as how changes in glycosylation can affect cell function. In addition, our understanding of the role of protein glycosylation in disease processes is growing, particularly in the context of immune system function, infectious diseases, neurodegeneration and cancer. Several decades ago, cell surface glycoproteins were found to have an important role in regulating ion transport across the cardiac sarcolemma. However, with very few exceptions, our understanding of how changes in protein glycosylation influence cardiovascular (patho)physiology remains remarkably limited. Therefore, in this Review, we aim to provide an overview of N-linked and O-linked protein glycosylation, including intracellular O-linked N-acetylglucosamine protein modification. We discuss our current understanding of how all forms of protein glycosylation contribute to normal cardiovascular function and their roles in cardiovascular disease. Finally, we highlight potential gaps in our knowledge about the effects of protein glycosylation on the heart and vascular system, highlighting areas for future research.
Collapse
Affiliation(s)
- John C Chatham
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Rakesh P Patel
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
2
|
Olcum M, Rouhi L, Fan S, Gonzales MM, Jeong HH, Zhao Z, Gurha P, Marian AJ. PANoptosis is a prominent feature of desmoplakin cardiomyopathy. THE JOURNAL OF CARDIOVASCULAR AGING 2023; 3:3. [PMID: 36818425 PMCID: PMC9933912 DOI: 10.20517/jca.2022.34] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction Arrhythmogenic cardiomyopathy (ACM) is hereditary cardiomyopathy caused by pathogenic variants (mutations) in genes encoding the intercalated disc (ID), particularly desmosome proteins. ACM caused by mutations in the DSP gene encoding desmoplakin (DSP) is characterized by the prominence of cell death, myocardial fibrosis, and inflammation, and is referred to as desmoplakin cardiomyopathy. Aim The aim of this article was to gain insight into the pathogenesis of DSP cardiomyopathy. Methods and Results The Dsp gene was exclusively deleted in cardiac myocytes using tamoxifen-inducible MerCreMer (Myh6-Mcm Tam) and floxed Dsp (Dsp F/F) mice (Myh6-Mcm Tam:Dsp F/F). Recombination was induced upon subcutaneous injection of tamoxifen (30 mg/kg/d) for 5 days starting post-natal day 14. Survival was analyzed by Kaplan-Meier plots, cardiac function by echocardiography, arrhythmias by rhythm monitoring, and gene expression by RNA-Seq, immunoblotting, and immunofluorescence techniques. Cell death was analyzed by the TUNEL assay and the expression levels of specific markers were by RT-PCR and immunoblotting. Myocardial fibrosis was assessed by picrosirius red staining of the myocardial sections, RT-PCR, and immunoblotting. The Myh6-Mcm Tam: Dsp F/F mice showed extensive molecular remodeling of the IDs and the differential expression of ~10,000 genes, which predicted activation of KDM5A, IRFs, and NFκB and suppression of PPARGC1A and RB1, among others in the DSP-deficient myocytes. Gene set enrichment analysis predicted activation of the TNFα/NFκB pathway, inflammation, cell death programs, and fibrosis. Analysis of cell death markers indicated PANoptosis, comprised of apoptosis (increased CASP3, CASP8, BAD and reduced BCL2), necroptosis (increased RIPK1, RIPK3, and MLKL), and pyroptosis (increased GSDMD and ASC or PYCARD) in the DSP-deficient myocytes. Transcript levels of the pro-inflammatory and pro-fibrotic genes were increased and myocardial fibrosis comprised ~25% of the myocardium in the DSP-deficient hearts. The Myh6-Mcm Tam:Dsp F/F mice showed severe cardiac systolic dysfunction and ventricular arrhythmias, and died prematurely with a median survival rate of ~2 months. Conclusion The findings identify PANoptosis as a prominent phenotypic feature of DSP cardiomyopathy and set the stage for delineating the specific molecular mechanisms involved in its pathogenesis. The model also provides the opportunity to test the effects of pharmacological and genetic interventions on myocardial fibrosis and cell death.
Collapse
Affiliation(s)
- Melis Olcum
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | - Leila Rouhi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | - Siyang Fan
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Maya M. Gonzales
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | - Hyun-Hwan Jeong
- Center for Precision Health, School of Biomedical Informatics and School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics and School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Priyatansh Gurha
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | - Ali J. Marian
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
3
|
Bidzimou MTK, Landstrom AP. From diagnostic testing to precision medicine: the evolving role of genomics in cardiac channelopathies and cardiomyopathies in children. Curr Opin Genet Dev 2022; 76:101978. [PMID: 36058060 PMCID: PMC9733798 DOI: 10.1016/j.gde.2022.101978] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 07/04/2022] [Accepted: 08/01/2022] [Indexed: 12/13/2022]
Abstract
Pediatric sudden cardiac death (SCD) is the sudden unexpected death of a child or adolescent due to a presumed cardiac etiology. Heritable causes of pediatric SCD are predominantly cardiomyopathies and cardiac ion channelopathies. This review illustrates recent advances in determining the genetic cause of established and emerging channelopathies and cardiomyopathies, and how broader genomic sequencing is uncovering complex interactions between genetic architecture and disease manifestation. We discuss innovative models and experimental platforms for resolving the variant of uncertain significance as both the variants and genes associated with disease continue to evolve. Finally, we highlight the growing problem of incidentally identified variants in cardiovascular disease-causing genes and review innovative methods to determining whether these variants may ultimately result in penetrant disease. Overall, we seek to illustrate both the promise and inherent challenges in bridging the traditional role for genetics in diagnosing cardiomyopathies and channelopathies to one of true risk-predictive precision medicine.
Collapse
Affiliation(s)
- Minu-Tshyeto K Bidzimou
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States. https://twitter.com/@MBidzimou
| | - Andrew P Landstrom
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States; Department of Pediatrics, Division of Pediatric Cardiology, Duke University School of Medicine, Durham, NC, United States.
| |
Collapse
|
4
|
Towards a Better Understanding of Genotype-Phenotype Correlations and Therapeutic Targets for Cardiocutaneous Genes: The Importance of Functional Studies above Prediction. Int J Mol Sci 2022; 23:ijms231810765. [PMID: 36142674 PMCID: PMC9503274 DOI: 10.3390/ijms231810765] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Genetic variants in gene-encoding proteins involved in cell−cell connecting structures, such as desmosomes and gap junctions, may cause a skin and/or cardiac phenotype, of which the combination is called cardiocutaneous syndrome. The cardiac phenotype is characterized by cardiomyopathy and/or arrhythmias, while the skin particularly displays phenotypes such as keratoderma, hair abnormalities and skin fragility. The reported variants associated with cardiocutaneous syndrome, in genes DSP, JUP, DSC2, KLHL24, GJA1, are classified by interpretation guidelines from the American College of Medical Genetics and Genomics. The genotype−phenotype correlation, however, remains poorly understood. By providing an overview of variants that are assessed for a functional protein pathology, we show that this number (n = 115) is low compared to the number of variants that are assessed by in silico algorithms (>5000). As expected, there is a mismatch between the prediction of variant pathogenicity and the prediction of the functional effect compared to the real functional evidence. Aiding to improve genotype−phenotype correlations, we separate variants into ‘protein reducing’ or ‘altered protein’ variants and provide general conclusions about the skin and heart phenotype involved. We conclude by stipulating that adequate prognoses can only be given, and targeted therapies can only be designed, upon full knowledge of the protein pathology through functional investigation.
Collapse
|
5
|
van Wijk SW, Su W, Wijdeveld LFJM, Ramos KS, Brundel BJJM. Cytoskeletal Protein Variants Driving Atrial Fibrillation: Potential Mechanisms of Action. Cells 2022; 11:416. [PMID: 35159226 PMCID: PMC8834312 DOI: 10.3390/cells11030416] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/15/2022] [Accepted: 01/20/2022] [Indexed: 11/16/2022] Open
Abstract
The most common clinical tachyarrhythmia, atrial fibrillation (AF), is present in 1-2% of the population. Although common risk factors, including hypertension, diabetes, and obesity, frequently underlie AF onset, it has been recognized that in 15% of the AF population, AF is familial. In these families, genome and exome sequencing techniques identified variants in the non-coding genome (i.e., variant regulatory elements), genes encoding ion channels, as well as genes encoding cytoskeletal (-associated) proteins. Cytoskeletal protein variants include variants in desmin, lamin A/C, titin, myosin heavy and light chain, junctophilin, nucleoporin, nesprin, and filamin C. These cytoskeletal protein variants have a strong association with the development of cardiomyopathy. Interestingly, AF onset is often represented as the initial manifestation of cardiac disease, sometimes even preceding cardiomyopathy by several years. Although emerging research findings reveal cytoskeletal protein variants to disrupt the cardiomyocyte structure and trigger DNA damage, exploration of the pathophysiological mechanisms of genetic AF is still in its infancy. In this review, we provide an overview of cytoskeletal (-associated) gene variants that relate to genetic AF and highlight potential pathophysiological pathways that drive this arrhythmia.
Collapse
Affiliation(s)
| | | | | | | | - Bianca J. J. M. Brundel
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (S.W.v.W.); (W.S.); (L.F.J.M.W.); (K.S.R.)
| |
Collapse
|
6
|
Rouhi L, Cheedipudi SM, Chen SN, Fan S, Lombardi R, Chen X, Coarfa C, Robertson MJ, Gurha P, Marian AJ. Haploinsufficiency of Tmem43 in cardiac myocytes activates the DNA damage response pathway leading to a late-onset senescence-associated pro-fibrotic cardiomyopathy. Cardiovasc Res 2021; 117:2377-2394. [PMID: 33070193 PMCID: PMC8861264 DOI: 10.1093/cvr/cvaa300] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/18/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023] Open
Abstract
AIMS Arrhythmogenic cardiomyopathy (ACM) encompasses a genetically heterogeneous group of myocardial diseases whose manifestations are sudden cardiac death, cardiac arrhythmias, heart failure, and in a subset fibro-adipogenic infiltration of the myocardium. Mutations in the TMEM43 gene, encoding transmembrane protein 43 (TMEM43) are known to cause ACM. The purpose of the study was to gain insights into the molecular pathogenesis of ACM caused by TMEM43 haploinsufficiency. METHODS AND RESULTS The Tmem43 gene was specifically deleted in cardiac myocytes by crossing the Myh6-Cre and floxed Tmem43 mice. Myh6-Cre:Tmem43W/F mice showed an age-dependent phenotype characterized by an increased mortality, cardiac dilatation and dysfunction, myocardial fibrosis, adipogenesis, and apoptosis. Sequencing of cardiac myocyte transcripts prior to and after the onset of cardiac phenotype predicted early activation of the TP53 pathway. Increased TP53 activity was associated with increased levels of markers of DNA damage response (DDR), and a subset of senescence-associated secretary phenotype (SASP). Activation of DDR, TP53, SASP, and their selected downstream effectors, including phospho-SMAD2 and phospho-SMAD3 were validated by alternative methods, including immunoblotting. Expression of SASP was associated with epithelial-mesenchymal transition and age-dependent expression of myocardial fibrosis and apoptosis in the Myh6-Cre:Tmem43W/F mice. CONCLUSION TMEM43 haploinsufficiency is associated with activation of the DDR and the TP53 pathways, which lead to increased expression of SASP and an age-dependent expression of a pro-fibrotic cardiomyopathy. Given that TMEM43 is a nuclear envelope protein and our previous data showing deficiency of another nuclear envelope protein, namely lamin A/C, activates the DDR/TP53 pathway, we surmise that DNA damage is a shared mechanism in the pathogenesis of cardiomyopathies caused by mutations involving nuclear envelope proteins.
Collapse
Affiliation(s)
- Leila Rouhi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, TX 77030, USA
| | - Sirisha M Cheedipudi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, TX 77030, USA
| | - Suet Nee Chen
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, TX 77030, USA
| | - Siyang Fan
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, TX 77030, USA
| | - Raffaella Lombardi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, TX 77030, USA
| | - Xiaofan Chen
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, TX 77030, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Matthew J Robertson
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Priyatansh Gurha
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, TX 77030, USA
| | - Ali J Marian
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, TX 77030, USA
| |
Collapse
|
7
|
Marian AJ, Asatryan B, Wehrens XHT. Genetic basis and molecular biology of cardiac arrhythmias in cardiomyopathies. Cardiovasc Res 2021; 116:1600-1619. [PMID: 32348453 DOI: 10.1093/cvr/cvaa116] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/09/2020] [Accepted: 04/21/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiac arrhythmias are common, often the first, and sometimes the life-threatening manifestations of hereditary cardiomyopathies. Pathogenic variants in several genes known to cause hereditary cardiac arrhythmias have also been identified in the sporadic cases and small families with cardiomyopathies. These findings suggest a shared genetic aetiology of a subset of hereditary cardiomyopathies and cardiac arrhythmias. The concept of a shared genetic aetiology is in accord with the complex and exquisite interplays that exist between the ion currents and cardiac mechanical function. However, neither the causal role of cardiac arrhythmias genes in cardiomyopathies is well established nor the causal role of cardiomyopathy genes in arrhythmias. On the contrary, secondary changes in ion currents, such as post-translational modifications, are common and contributors to the pathogenesis of arrhythmias in cardiomyopathies through altering biophysical and functional properties of the ion channels. Moreover, structural changes, such as cardiac hypertrophy, dilatation, and fibrosis provide a pro-arrhythmic substrate in hereditary cardiomyopathies. Genetic basis and molecular biology of cardiac arrhythmias in hereditary cardiomyopathies are discussed.
Collapse
Affiliation(s)
- Ali J Marian
- Department of Medicine, Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston, 6770 Bertner Street, Suite C900A, Houston, TX 77030, USA
| | - Babken Asatryan
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Xander H T Wehrens
- Department of Biophysics and Molecular Physiology, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
8
|
Yuan P, Cheedipudi SM, Rouhi L, Fan S, Simon L, Zhao Z, Hong K, Gurha P, Marian AJ. Single-Cell RNA Sequencing Uncovers Paracrine Functions of the Epicardial-Derived Cells in Arrhythmogenic Cardiomyopathy. Circulation 2021; 143:2169-2187. [PMID: 33726497 DOI: 10.1161/circulationaha.120.052928] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Arrhythmogenic cardiomyopathy (ACM) manifests with sudden death, arrhythmias, heart failure, apoptosis, and myocardial fibro-adipogenesis. The phenotype typically starts at the epicardium and advances transmurally. Mutations in genes encoding desmosome proteins, including DSP (desmoplakin), are major causes of ACM. METHODS To delineate contributions of the epicardium to the pathogenesis of ACM, the Dsp allele was conditionally deleted in the epicardial cells in mice upon expression of tamoxifen-inducible Cre from the Wt1 locus. Wild type (WT) and Wt1-CreERT2:DspW/F were crossed to Rosa26mT/mG (R26mT/mG) dual reporter mice to tag the epicardial-derived cells with the EGFP (enhanced green fluorescent protein) reporter protein. Tagged epicardial-derived cells from adult Wt1-CreERT2:R26mT/mG and Wt1-CreERT2: R26mT/mG:DspW/F mouse hearts were isolated by fluorescence-activated cell staining and sequenced by single-cell RNA sequencing. RESULTS WT1 (Wilms tumor 1) expression was progressively restricted postnatally and was exclusive to the epicardium by postnatal day 21. Expression of Dsp was reduced in the epicardial cells but not in cardiac myocytes in the Wt1-CreERT2:DspW/F mice. The Wt1-CreERT2:DspW/F mice exhibited premature death, cardiac dysfunction, arrhythmias, myocardial fibro-adipogenesis, and apoptosis. Single-cell RNA sequencing of ≈18 000 EGFP-tagged epicardial-derived cells identified genotype-independent clusters of endothelial cells, fibroblasts, epithelial cells, and a very small cluster of cardiac myocytes, which were confirmed on coimmunofluorescence staining of the myocardial sections. Differentially expressed genes between the paired clusters in the 2 genotypes predicted activation of the inflammatory and mitotic pathways-including the TGFβ1 (transforming growth factor β1) and fibroblast growth factors-in the epicardial-derived fibroblast and epithelial clusters, but predicted their suppression in the endothelial cell cluster. The findings were corroborated by analysis of gene expression in the pooled RNA-sequencing data, which identified predominant dysregulation of genes involved in epithelial-mesenchymal transition, and dysregulation of 146 genes encoding the secreted proteins (secretome), including genes in the TGFβ1 pathway. Activation of the TGFβ1 and its colocalization with fibrosis in the Wt1-CreERT2:R26mT/mG:DspW/F mouse heart was validated by complementary methods. CONCLUSIONS Epicardial-derived cardiac fibroblasts and epithelial cells express paracrine factors, including TGFβ1 and fibroblast growth factors, which mediate epithelial-mesenchymal transition, and contribute to the pathogenesis of myocardial fibrosis, apoptosis, arrhythmias, and cardiac dysfunction in a mouse model of ACM. The findings uncover contributions of the epicardial-derived cells to the pathogenesis of ACM.
Collapse
Affiliation(s)
- Ping Yuan
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine (P.Y., S.M.C., L.R., S.F., P.G., A.J.M.).,Department of Cardiovascular Medicine, Second Affiliated Hospital of Nanchang University, China (P.Y., K.H.)
| | - Sirisha M Cheedipudi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine (P.Y., S.M.C., L.R., S.F., P.G., A.J.M.)
| | - Leila Rouhi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine (P.Y., S.M.C., L.R., S.F., P.G., A.J.M.)
| | - Siyang Fan
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine (P.Y., S.M.C., L.R., S.F., P.G., A.J.M.)
| | - Lukas Simon
- Center for Precision Health, School of Biomedical Informatics and School of Public Health, University of Texas Health Science Center at Houston (L.S., Z.Z.)
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics and School of Public Health, University of Texas Health Science Center at Houston (L.S., Z.Z.)
| | - Kui Hong
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Nanchang University, China (P.Y., K.H.)
| | - Priyatansh Gurha
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine (P.Y., S.M.C., L.R., S.F., P.G., A.J.M.)
| | - Ali J Marian
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine (P.Y., S.M.C., L.R., S.F., P.G., A.J.M.)
| |
Collapse
|
9
|
Auguste G, Rouhi L, Matkovich SJ, Coarfa C, Robertson MJ, Czernuszewicz G, Gurha P, Marian AJ. BET bromodomain inhibition attenuates cardiac phenotype in myocyte-specific lamin A/C-deficient mice. J Clin Invest 2021; 130:4740-4758. [PMID: 32484798 DOI: 10.1172/jci135922] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 05/27/2020] [Indexed: 01/03/2023] Open
Abstract
Mutation in the LMNA gene, encoding lamin A/C, causes a diverse group of diseases called laminopathies. Cardiac involvement is the major cause of death and manifests as dilated cardiomyopathy, heart failure, arrhythmias, and sudden death. There is no specific therapy for LMNA-associated cardiomyopathy. We report that deletion of Lmna in cardiomyocytes in mice leads to severe cardiac dysfunction, conduction defect, ventricular arrhythmias, fibrosis, apoptosis, and premature death within 4 weeks. The phenotype is similar to LMNA-associated cardiomyopathy in humans. RNA sequencing, performed before the onset of cardiac dysfunction, led to identification of 2338 differentially expressed genes (DEGs) in Lmna-deleted cardiomyocytes. DEGs predicted activation of bromodomain-containing protein 4 (BRD4), a regulator of chromatin-associated proteins and transcription factors, which was confirmed by complementary approaches, including chromatin immunoprecipitation sequencing. Daily injection of JQ1, a specific BET bromodomain inhibitor, partially reversed the DEGs, including those encoding secretome; improved cardiac function; abrogated cardiac arrhythmias, fibrosis, and apoptosis; and prolonged the median survival time 2-fold in the myocyte-specific Lmna-deleted mice. The findings highlight the important role of LMNA in cardiomyocytes and identify BET bromodomain inhibition as a potential therapeutic target in LMNA-associated cardiomyopathy, for which there is no specific effective therapy.
Collapse
Affiliation(s)
- Gaelle Auguste
- Center for Cardiovascular Genetics, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| | - Leila Rouhi
- Center for Cardiovascular Genetics, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| | - Scot J Matkovich
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Cristian Coarfa
- Department of Cell Biology, Baylor College of Medicine, Houston, Texas, USA
| | | | - Grazyna Czernuszewicz
- Center for Cardiovascular Genetics, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| | - Priyatansh Gurha
- Center for Cardiovascular Genetics, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| | - Ali J Marian
- Center for Cardiovascular Genetics, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| |
Collapse
|
10
|
Cheedipudi SM, Hu J, Fan S, Yuan P, Karmouch J, Czernuszewicz G, Robertson MJ, Coarfa C, Hong K, Yao Y, Campbell H, Wehrens X, Gurha P, Marian AJ. Exercise restores dysregulated gene expression in a mouse model of arrhythmogenic cardiomyopathy. Cardiovasc Res 2021; 116:1199-1213. [PMID: 31350552 DOI: 10.1093/cvr/cvz199] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/16/2019] [Accepted: 07/19/2019] [Indexed: 12/19/2022] Open
Abstract
AIMS Arrhythmogenic cardiomyopathy (ACM) is a myocardial disease caused mainly by mutations in genes encoding desmosome proteins ACM patients present with ventricular arrhythmias, cardiac dysfunction, sudden cardiac death, and a subset with fibro-fatty infiltration of the right ventricle predominantly. Endurance exercise is thought to exacerbate cardiac dysfunction and arrhythmias in ACM. The objective was to determine the effects of treadmill exercise on cardiac phenotype, including myocyte gene expression in myocyte-specific desmoplakin (Dsp) haplo-insufficient (Myh6-Cre:DspW/F) mice. METHODS AND RESULTS Three months old sex-matched wild-type (WT) and Myh6-Cre:DspW/F mice with normal cardiac function, as assessed by echocardiography, were randomized to regular activity or 60 min of daily treadmill exercise (5.5 kJ work per run). Cardiac myocyte gene expression, cardiac function, arrhythmias, and myocardial histology, including apoptosis, were analysed prior to and after 3 months of routine activity or treadmill exercise. Fifty-seven and 781 genes were differentially expressed in 3- and 6-month-old Myh6-Cre:DspW/F cardiac myocytes, compared to the corresponding WT myocytes, respectively. Genes encoding secreted proteins (secretome), including inhibitors of the canonical WNT pathway, were among the most up-regulated genes. The differentially expressed genes (DEGs) predicted activation of epithelial-mesenchymal transition (EMT) and inflammation, and suppression of oxidative phosphorylation pathways in the Myh6-Cre:DspW/F myocytes. Treadmill exercise restored transcript levels of two-third (492/781) of the DEGs and the corresponding dysregulated transcriptional and biological pathways, including EMT, inflammation, and secreted inhibitors of the canonical WNT. The changes were associated with reduced myocardial apoptosis and eccentric cardiac hypertrophy without changes in cardiac function. CONCLUSION Treadmill exercise restored transcript levels of the majority of dysregulated genes in cardiac myocytes, reduced myocardial apoptosis, and induced eccentric cardiac hypertrophy without affecting cardiac dysfunction in a mouse model of ACM. The findings suggest that treadmill exercise has potential beneficial effects in a subset of cardiac phenotypes in ACM.
Collapse
Affiliation(s)
- Sirisha M Cheedipudi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | - Jinzhu Hu
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | - Siyang Fan
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | - Ping Yuan
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | - Jennifer Karmouch
- Department of Medicine, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Grace Czernuszewicz
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | - Matthew J Robertson
- Department of Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cristian Coarfa
- Department of Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kui Hong
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, PR China
| | - Yan Yao
- Fuwai Hospital, Peking Union Medical College, Beijing, PR China
| | - Hanna Campbell
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xander Wehrens
- Cardiovascular Research Institute, Departments of Molecular Physiology & Biophysics, Medicine, Neuroscience, Pediatrics, and Center for Space Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Priyatansh Gurha
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | - Ali J Marian
- Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
11
|
Priori SG, Mazzanti A. Warning: not all carriers of pathogenic mutations in desmosomal genes should follow the same medical advices! Cardiovasc Res 2021; 116:1085-1088. [PMID: 32129836 DOI: 10.1093/cvr/cvaa049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Affiliation(s)
- Silvia G Priori
- Molecular Cardiology, ICS Maugeri, IRCCS, Via Maugeri, 10, 27100 Pavia, Italy.,Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Molecular Cardiology, Fundación Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Andrea Mazzanti
- Molecular Cardiology, ICS Maugeri, IRCCS, Via Maugeri, 10, 27100 Pavia, Italy.,Department of Molecular Medicine, University of Pavia, Pavia, Italy
| |
Collapse
|
12
|
Sun Q, Wine Lee L, Hall EK, Choate KA, Elder RW. Hair and skin predict cardiomyopathies: Carvajal and erythrokeratodermia cardiomyopathy syndromes. Pediatr Dermatol 2021; 38:31-38. [PMID: 33275305 DOI: 10.1111/pde.14478] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Carvajal and erythrokeratodermia cardiomyopathy syndromes (EKC) are rare, inherited cardiocutaneous disorders with potentially fatal consequences in young children. Some patients display features of congestive heart failure and rapidly deteriorate; others exhibit no evident warning signs until sudden death reveals underlying heart disease. We present two patients to illustrate the characteristic hair, skin, teeth, and nail abnormalities, which-especially when distinct from that of other family members-should prompt cardiac evaluation and genetic analysis. In this article, we discuss established treatments as well as a promising, novel therapeutic that has led to nearly complete resolution of the cutaneous and cardiac pathology in EKC syndrome.
Collapse
Affiliation(s)
- Qisi Sun
- Departments of Dermatology, Genetics, and Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Lara Wine Lee
- Medical University of South Carolina Health, Charleston, SC, USA
| | - E Kevin Hall
- Section of Cardiology, Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Keith A Choate
- Departments of Dermatology, Genetics, and Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Robert W Elder
- Section of Cardiology, Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
13
|
Leopoulou M, Mattsson G, LeQuang JA, Pergolizzi JV, Varrassi G, Wallhagen M, Magnusson P. Naxos disease - a narrative review. Expert Rev Cardiovasc Ther 2020; 18:801-808. [PMID: 32966140 DOI: 10.1080/14779072.2020.1828064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Naxos disease is a rare entity that manifests with woolly hair, keratosis of extremities, and cardiac manifestations that resemble arrhythmogenic right ventricular cardiomyopathy. It is inherited in an autosomal recessive pattern and mutations affecting plakoglobin and desmoplakin have been identified. There is an increased risk of arrhythmias, including sudden cardiac death at a young age. Right ventricular systolic dysfunction often progresses and left ventricular involvement may also occur. AREAS COVERED This article reviews historic background, epidemiology, clinical characteristics, genetics, and pathogenesis as well as therapeutic management and future perspectives. EXPERT OPINION The principles of evaluation and treatment are based on arrhythmogenic right ventricular cardiomyopathy (ARVC) and general heart failure guidelines, because specific data on Naxos disease are limited. Therefore, larger registries on Naxos disease are welcome in order to gain more knowledge about clinical course and risk stratification. Translational research on pathophysiological mechanisms has evolved, including promising approaches using stem cells for novel targets.
Collapse
Affiliation(s)
| | - Gustav Mattsson
- Centre for Research and Development, Uppsala University/Region Gävleborg , Gävle, Sweden
| | | | - Joseph V Pergolizzi
- NEMA Research, Inc , Naples, Florida, USA.,Native Cardio, Inc , Naples, Florida, USA
| | - Giustino Varrassi
- Paolo Procacci Foundation , Rome, Italy.,President, World Institute of Pain , California, USA
| | - Marita Wallhagen
- Faculty of Engineering and Sustainable Development, University of Gävle , Gävle, Sweden
| | - Peter Magnusson
- Centre for Research and Development, Uppsala University/Region Gävleborg , Gävle, Sweden.,Cardiology Research Unit, Department of Medicine, Karolinska Institutet , Stockholm, Sweden
| |
Collapse
|
14
|
Shoykhet M, Trenz S, Kempf E, Williams T, Gerull B, Schinner C, Yeruva S, Waschke J. Cardiomyocyte adhesion and hyperadhesion differentially require ERK1/2 and plakoglobin. JCI Insight 2020; 5:140066. [PMID: 32841221 PMCID: PMC7526536 DOI: 10.1172/jci.insight.140066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/12/2020] [Indexed: 12/23/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a heart disease often caused by mutations in genes coding for desmosomal proteins, including desmoglein-2 (DSG2), plakoglobin (PG), and desmoplakin (DP). Therapy is based on symptoms and limiting arrhythmia, because the mechanisms by which desmosomal components control cardiomyocyte function are largely unknown. A new paradigm could be to stabilize desmosomal cardiomyocyte adhesion and hyperadhesion, which renders desmosomal adhesion independent from Ca2+. Here, we further characterized the mechanisms behind enhanced cardiomyocyte adhesion and hyperadhesion. Dissociation assays performed in HL-1 cells and murine ventricular cardiac slice cultures allowed us to define a set of signaling pathways regulating cardiomyocyte adhesion under basal and hyperadhesive conditions. Adrenergic signaling, activation of PKC, and inhibition of p38MAPK enhanced cardiomyocyte adhesion, referred to as positive adhesiotropy, and induced hyperadhesion. Activation of ERK1/2 paralleled positive adhesiotropy, whereas adrenergic signaling induced PG phosphorylation at S665 under both basal and hyperadhesive conditions. Adrenergic signaling and p38MAPK inhibition recruited DSG2 to cell junctions. In PG-deficient mice with an AC phenotype, only PKC activation and p38MAPK inhibition enhanced cardiomyocyte adhesion. Our results demonstrate that cardiomyocyte adhesion can be stabilized by different signaling mechanisms, which are in part offset in PG-deficient AC. Desmosome mediated cardiomyocyte adhesion, crucial in the pathology of arrhythmogenic cardiomyopathy, is differentially regulated by multiple signaling mechanisms that depend either on ERK1/2 or plakoglobin.
Collapse
Affiliation(s)
- Maria Shoykhet
- Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sebastian Trenz
- Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Ellen Kempf
- Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Tatjana Williams
- Comprehensive Heart Failure Center and Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Brenda Gerull
- Comprehensive Heart Failure Center and Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Camilla Schinner
- Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sunil Yeruva
- Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jens Waschke
- Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
15
|
Cheedipudi SM, Matkovich SJ, Coarfa C, Hu X, Robertson MJ, Sweet M, Taylor M, Mestroni L, Cleveland J, Willerson JT, Gurha P, Marian AJ. Genomic Reorganization of Lamin-Associated Domains in Cardiac Myocytes Is Associated With Differential Gene Expression and DNA Methylation in Human Dilated Cardiomyopathy. Circ Res 2020; 124:1198-1213. [PMID: 30739589 DOI: 10.1161/circresaha.118.314177] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
RATIONALE LMNA (Lamin A/C), a nuclear membrane protein, interacts with genome through lamin-associated domains (LADs) and regulates gene expression. Mutations in the LMNA gene cause a diverse array of diseases, including dilated cardiomyopathy (DCM). DCM is the leading cause of death in laminopathies. OBJECTIVE To identify LADs and characterize their associations with CpG methylation and gene expression in human cardiac myocytes in DCM. METHODS AND RESULTS LMNA chromatin immunoprecipitation-sequencing, reduced representative bisulfite sequencing, and RNA-sequencing were performed in 5 control and 5 LMNA-associated DCM hearts. LADs were identified using enriched domain detector program. Genome-wide 331±77 LADs with an average size of 2.1±1.5 Mbp were identified in control human cardiac myocytes. LADs encompassed ≈20% of the genome and were predominantly located in the heterochromatin and less so in the promoter and actively transcribed regions. LADs were redistributed in DCM as evidenced by a gain of 520 and loss of 149 genomic regions. Approximately, 4500 coding genes and 800 long noncoding RNAs, whose levels correlated with the transcript levels of coding genes in cis, were differentially expressed in DCM. TP53 (tumor protein 53) was the most prominent among the dysregulated pathways. CpG sites were predominantly hypomethylated genome-wide in controls and DCM hearts, but overall CpG methylation was increased in DCM. LADs were associated with increased CpG methylation and suppressed gene expression. Integrated analysis identified genes whose expressions were regulated by LADs or CpG methylation, or by both, the latter pertained to genes involved in cell death, cell cycle, and metabolic regulation. CONCLUSIONS LADs encompass ≈20% of the genome in human cardiac myocytes comprised several hundred coding and noncoding genes. LADs are redistributed in LMNA-associated DCM in association with markedly altered CpG methylation and gene expression. Thus, LADs through genomic alterations contribute to the pathogenesis of DCM in laminopathies.
Collapse
Affiliation(s)
- Sirisha M Cheedipudi
- From the Center for Cardiovascular Genetics, Department of Medicine, Institute of Molecular Medicine, Texas Heart Institute, University of Texas Health Sciences Center at Houston (S.M.C., P.G., A.J.M.)
| | - Scot J Matkovich
- Center for Cardiovascular Research, Washington University, School of Medicine, St Louis, MO (S.J.M.)
| | | | - Xin Hu
- MD Anderson Cancer Center, Houston, TX (X.H.)
| | | | - Mary Sweet
- Division of Cardiology (M.S., M.T., L.M.), University of Colorado, Denver
| | - Matthew Taylor
- Division of Cardiology (M.S., M.T., L.M.), University of Colorado, Denver
| | - Luisa Mestroni
- Division of Cardiology (M.S., M.T., L.M.), University of Colorado, Denver
| | - Joseph Cleveland
- Division of Cardiothoracic Surgery (J.C.), University of Colorado, Denver
| | | | - Priyatansh Gurha
- From the Center for Cardiovascular Genetics, Department of Medicine, Institute of Molecular Medicine, Texas Heart Institute, University of Texas Health Sciences Center at Houston (S.M.C., P.G., A.J.M.)
| | - Ali J Marian
- From the Center for Cardiovascular Genetics, Department of Medicine, Institute of Molecular Medicine, Texas Heart Institute, University of Texas Health Sciences Center at Houston (S.M.C., P.G., A.J.M.)
| |
Collapse
|
16
|
Abstract
Arrhythmogenic cardiomyopathy is a genetic disorder characterized by the risk of life-threatening arrhythmias, myocardial dysfunction and fibrofatty replacement of myocardial tissue. Mutations in genes that encode components of desmosomes, the adhesive junctions that connect cardiomyocytes, are the predominant cause of arrhythmogenic cardiomyopathy and can be identified in about half of patients with the condition. However, the molecular mechanisms leading to myocardial destruction, remodelling and arrhythmic predisposition remain poorly understood. Through the development of animal, induced pluripotent stem cell and other models of disease, advances in our understanding of the pathogenic mechanisms of arrhythmogenic cardiomyopathy over the past decade have brought several signalling pathways into focus. These pathways include canonical and non-canonical WNT signalling, the Hippo-Yes-associated protein (YAP) pathway and transforming growth factor-β signalling. These studies have begun to identify potential therapeutic targets whose modulation has shown promise in preclinical models. In this Review, we summarize and discuss the reported molecular mechanisms underlying the pathogenesis of arrhythmogenic cardiomyopathy.
Collapse
|
17
|
Chen SN, Lombardi R, Karmouch J, Tsai JY, Czernuszewicz G, Taylor MRG, Mestroni L, Coarfa C, Gurha P, Marian AJ. DNA Damage Response/TP53 Pathway Is Activated and Contributes to the Pathogenesis of Dilated Cardiomyopathy Associated With LMNA (Lamin A/C) Mutations. Circ Res 2019; 124:856-873. [PMID: 30696354 DOI: 10.1161/circresaha.118.314238] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
RATIONALE Mutations in the LMNA gene, encoding LMNA (lamin A/C), are responsible for laminopathies. Dilated cardiomyopathy (DCM) is a major cause of mortality and morbidity in laminopathies. OBJECTIVE To gain insights into the molecular pathogenesis of DCM in laminopathies. METHODS AND RESULTS We generated a tet-off bigenic mice expressing either a WT (wild type) or a mutant LMNA (D300N) protein in cardiac myocytes. LMNAD300N mutation is associated with DCM in progeroid syndromes. Expression of LMNAD300N led to severe myocardial fibrosis, apoptosis, cardiac dysfunction, and premature death. Administration of doxycycline suppressed LMNAD300N expression and prevented the phenotype. Whole-heart RNA sequencing in 2-week-old WT and LMNAD300N mice led to identification of ≈6000 differentially expressed genes. Gene Set Enrichment and Hallmark Pathway analyses predicted activation of E2F (E2F transcription factor), DNA damage response, TP53 (tumor protein 53), NFκB (nuclear factor κB), and TGFβ (transforming growth factor-β) pathways, which were validated by Western blotting, quantitative polymerase chain reaction of selected targets, and immunofluorescence staining. Differentially expressed genes involved cell death, cell cycle regulation, inflammation, and epithelial-mesenchymal differentiation. RNA sequencing of human hearts with DCM associated with defined LMNA pathogenic variants corroborated activation of the DNA damage response/TP53 pathway in the heart. Increased expression of CDKN2A (cyclin-dependent kinase inhibitor 2A)-a downstream target of E2F pathway and an activator of TP53-provided a plausible mechanism for activation of the TP53 pathway. To determine pathogenic role of TP53 pathway in DCM, Tp53 gene was conditionally deleted in cardiac myocytes in mice expressing the LMNAD300N protein. Deletion of Tp53 partially rescued myocardial fibrosis, apoptosis, proliferation of nonmyocyte cells, left ventricular dilatation and dysfunction, and slightly improved survival. CONCLUSIONS Cardiac myocyte-specific expression of LMNAD300N, associated with DCM, led to pathogenic activation of the E2F/DNA damage response/TP53 pathway in the heart and induction of myocardial fibrosis, apoptosis, cardiac dysfunction, and premature death. The findings denote the E2F/DNA damage response/TP53 axis as a responsible mechanism for DCM in laminopathies and as a potential intervention target.
Collapse
Affiliation(s)
- Suet Nee Chen
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston (S.N.C., R.L., J.K., J.-Y.T., G.C., P.G., A.J.M.).,Section of Cardiology, University of Colorado-Anschutz Medical Campus, Denver (S.N.C., M.R.G.T., L.M.)
| | - Raffaella Lombardi
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston (S.N.C., R.L., J.K., J.-Y.T., G.C., P.G., A.J.M.).,Division of Cardiology, Department of Advanced Biomedical Science, University of Naples Federico II, Italy (R.L.)
| | - Jennifer Karmouch
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston (S.N.C., R.L., J.K., J.-Y.T., G.C., P.G., A.J.M.).,MD Anderson Cancer Center, Houston, TX (J.K.)
| | - Ju-Yun Tsai
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston (S.N.C., R.L., J.K., J.-Y.T., G.C., P.G., A.J.M.).,Thermo Fisher Scientific, Taiwan (J.-Y.T.)
| | - Grace Czernuszewicz
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston (S.N.C., R.L., J.K., J.-Y.T., G.C., P.G., A.J.M.)
| | - Matthew R G Taylor
- Section of Cardiology, University of Colorado-Anschutz Medical Campus, Denver (S.N.C., M.R.G.T., L.M.)
| | - Luisa Mestroni
- Section of Cardiology, University of Colorado-Anschutz Medical Campus, Denver (S.N.C., M.R.G.T., L.M.)
| | - Cristian Coarfa
- Department of Cell Biology, Baylor College of Medicine, Houston, TX (C.C.)
| | - Priyatansh Gurha
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston (S.N.C., R.L., J.K., J.-Y.T., G.C., P.G., A.J.M.)
| | - Ali J Marian
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine, University of Texas Health Sciences Center at Houston (S.N.C., R.L., J.K., J.-Y.T., G.C., P.G., A.J.M.)
| |
Collapse
|
18
|
Pigolkin YI, Shilova MA, Berezovskiy DP, Egorov VN, Tayutina TV, Bachurin SS, Kolomoets IA. [Molecular genetic basis of sudden cardiac death in the young with cardiomyopathy of various origins]. Sud Med Ekspert 2019; 62:48-53. [PMID: 31198205 DOI: 10.17116/sudmed20196203148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This paper provides a review of the modern literature devoted to the problem of forensic medical interpretation of the molecular genetic research of the young who died suddenly. The authors attempted to draw a parallel between the morphological markers of different variants of cardiomyopathy as the most common disease in sudden death at a young age and the association with genetic mutations in the genes responsible for the synthesis of sarcomer proteins, desmos and membrane channels. Based on the results of the analysis, further research is proposed to improve the accuracy of forensic diagnosis in cases of young deaths.
Collapse
Affiliation(s)
- Yu I Pigolkin
- Department of Forensic Medicine of the Sechenov First Moscow State Medical University under Ministry of Health of the Russia, Moscow, Russia, 119991
| | - M A Shilova
- Department of Forensic Medicine of the Sechenov First Moscow State Medical University under Ministry of Health of the Russia, Moscow, Russia, 119991
| | - D P Berezovskiy
- Department of Forensic Medicine and Medical Law of the Rostov State Medical University of the Ministry of Health of the Russia, Rostov-on-Don, Russia, 344022
| | - V N Egorov
- Department of Forensic Medicine and Medical Law of the Rostov State Medical University of the Ministry of Health of the Russia, Rostov-on-Don, Russia, 344022
| | - T V Tayutina
- Department of Forensic Medicine and Medical Law of the Rostov State Medical University of the Ministry of Health of the Russia, Rostov-on-Don, Russia, 344022
| | - S S Bachurin
- Department of Forensic Medicine and Medical Law of the Rostov State Medical University of the Ministry of Health of the Russia, Rostov-on-Don, Russia, 344022
| | - I A Kolomoets
- Department of Forensic Medicine and Medical Law of the Rostov State Medical University of the Ministry of Health of the Russia, Rostov-on-Don, Russia, 344022
| |
Collapse
|
19
|
|
20
|
|
21
|
Auguste G, Gurha P, Lombardi R, Coarfa C, Willerson JT, Marian AJ. Suppression of Activated FOXO Transcription Factors in the Heart Prolongs Survival in a Mouse Model of Laminopathies. Circ Res 2018; 122:678-692. [PMID: 29317431 DOI: 10.1161/circresaha.117.312052] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/13/2017] [Accepted: 01/05/2018] [Indexed: 01/15/2023]
Abstract
RATIONALE Mutations in the LMNA gene, encoding nuclear inner membrane protein lamin A/C, cause distinct phenotypes, collectively referred to as laminopathies. Heart failure, conduction defects, and arrhythmias are the common causes of death in laminopathies. OBJECTIVE The objective of this study was to identify and therapeutically target the responsible mechanism(s) for cardiac phenotype in laminopathies. METHODS AND RESULTS Whole-heart RNA sequencing was performed before the onset of cardiac dysfunction in the Lmna-/- and matched control mice. Differentially expressed transcripts and their upstream regulators were identified, validated, and targeted by adeno-associated virus serotype 9-short hairpin RNA constructs. A total of 576 transcripts were upregulated and 233 were downregulated in the Lmna-/- mouse hearts (q<0.05). Forkhead box O (FOXO) transcription factors (TFs) were the most activated while E2 factors were the most suppressed transcriptional regulators. Transcript levels of FOXO targets were also upregulated in the isolated Lmna-/- cardiac myocytes and in the myocardium of human heart failure patients. Nuclear localization of FOXO1 and 3 was increased, whereas phosphorylated (inactive) FOXO1 and 3 levels were reduced in the Lmna-/- hearts. Gene set enrichment analysis and gene ontology showed activation of apoptosis and inflammation and suppression of cell cycle, adipogenesis, and oxidative phosphorylation in the Lmna-/- hearts. Adeno-associated virus serotype 9-short hairpin RNA-mediated suppression of FOXO TFs rescued selected molecular signatures, improved apoptosis, and prolonged survival by ≈2-fold. CONCLUSIONS FOXO TFs are activated and contribute to the pathogenesis of cardiac phenotype in laminopathies. Suppression of the FOXO TFs in cardiac myocytes partially rescues the phenotype and prolongs survival. The findings identify FOXO TFs as potential therapeutic targets for cardiac phenotype in laminopathies.
Collapse
Affiliation(s)
- Gaelle Auguste
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston (G.A., P.G., R.L., T.T.W., A.J.M.), Texas Heart Institute (J.T.W., A.J.M.); and Baylor College of Medicine, Houston, TX (C.C.)
| | - Priyatansh Gurha
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston (G.A., P.G., R.L., T.T.W., A.J.M.), Texas Heart Institute (J.T.W., A.J.M.); and Baylor College of Medicine, Houston, TX (C.C.)
| | - Raffaella Lombardi
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston (G.A., P.G., R.L., T.T.W., A.J.M.), Texas Heart Institute (J.T.W., A.J.M.); and Baylor College of Medicine, Houston, TX (C.C.)
| | - Cristian Coarfa
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston (G.A., P.G., R.L., T.T.W., A.J.M.), Texas Heart Institute (J.T.W., A.J.M.); and Baylor College of Medicine, Houston, TX (C.C.)
| | - James T Willerson
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston (G.A., P.G., R.L., T.T.W., A.J.M.), Texas Heart Institute (J.T.W., A.J.M.); and Baylor College of Medicine, Houston, TX (C.C.)
| | - Ali J Marian
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston (G.A., P.G., R.L., T.T.W., A.J.M.), Texas Heart Institute (J.T.W., A.J.M.); and Baylor College of Medicine, Houston, TX (C.C.).
| |
Collapse
|
22
|
Priori SG, Santiago DJ. Arrhythmogenic Cardiomyopathy: Pathophysiology Beyond Cardiac Myocytes. Circ Res 2017; 121:1296-1298. [PMID: 29217704 DOI: 10.1161/circresaha.117.312211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Silvia G Priori
- From the Molecular Cardiology, IRCCS ICS Maugeri, Pavia, Italy (S.G.P.); Department of Molecular Medicine, University of Pavia, Italy (S.G.P.); and Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (S.G.P., D.J.S.).
| | - Demetrio J Santiago
- From the Molecular Cardiology, IRCCS ICS Maugeri, Pavia, Italy (S.G.P.); Department of Molecular Medicine, University of Pavia, Italy (S.G.P.); and Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (S.G.P., D.J.S.)
| |
Collapse
|