Abstract
Dilated cardiomyopathy (DCM) is a group of heart muscle diseases that often lead to heart failure, with more than 50 causative genes have being linked to DCM. The heterogenous nature of the inherited DCMs suggest the need of precision medicine. Consistent with this emerging concept, transcriptome studies in human patients with DCM indicated distinct molecular signature for DCMs of different genetic etiology. To facilitate this line of research, we reviewed the status of transcriptome studies of inherited DCMs by focusing on three predominant DCM causative genes, TTN, LMNA, and BAG3. Besides studies in human patients, we summarized transcriptomic analysis of these inherited DCMs in a variety of model systems ranging from iPSCs to rodents and zebrafish. We concluded that the RNA-seq technology is a powerful genomic tool that has already led to the discovery of new modifying genes, signaling pathways, and related therapeutic avenues. We also pointed out that both temporal (different pathological stages) and spatial (different cell types) information need to be considered for future transcriptome studies. While an important bottle neck is the low throughput in experimentally testing differentially expressed genes, new technologies in efficient animal models such as zebrafish starts to be developed. It is anticipated that the RNA-seq technology will continue to uncover both unique and common pathological events, aiding the development of precision medicine for inherited DCMs.
Similar content being viewed by others
Data availability
Data sharing not applicable to this article as no datasets were generated or analyzed during the current study.
References
Abplanalp Wesley Tyler, Tucker Nathan, Dimmeler Stefanie (2022) Single-cell technologies to decipher cardiovascular diseases. Euro Heart J 43(43):4536–4547. https://doi.org/10.1093/eurheartj/ehac095
Ahmad F, Seidman JG, Seidman CE (2005) The genetic basis for cardiac remodeling. Annu Rev Genomics Hum Genet 6:185–216. https://doi.org/10.1146/annurev.genom.6.080604.162132
Bang ML et al (2001) The complete gene sequence of titin, expression of an unusual≈ 700-kDa titin isoform, and its interaction with obscurin identify a novel Z-line to I-band linking system. Circ Res 89(11):1065–1072. https://doi.org/10.1161/hh2301.100981
Belmonte T et al (2020) Peripheral microRNA panels to guide the diagnosis of familial cardiomyopathy. Transl Res 218:1–15. https://doi.org/10.1016/j.trsl.2020.01.003
Calderon-Dominguez M et al (2020) Emerging role of microRNAs in dilated cardiomyopathy: Evidence regarding etiology. Transl Res 215:86–101. https://doi.org/10.1016/j.trsl.2019.08.007
Chami N et al (2014) Nonsense mutations in BAG3 are associated with early-onset dilated cardiomyopathy in French Canadians. Can J Cardiol 30(12):1655–1661. https://doi.org/10.1016/j.cjca.2014.09.030
Chaudhry F, Isherwood J, Bawa T, Patel D, Gurdziel K, Lanfear DE, Ruden DM, Levy PD (2019) Single-cell RNA sequencing of the cardiovascular system: new looks for old diseases. Front Cardiovasc Med 6:173. https://doi.org/10.3389/fcvm.2019.00173
Chauveau C, Rowell J, Ferreiro A (2014) A rising titan: TTN review and mutation update. Hum Mutat 35(9):1046–1059. https://doi.org/10.1002/humu.22611
Cheedipudi SM et al (2019) Genomic reorganization of lamin-associated domains in cardiac myocytes is associated with differential gene expression and DNA methylation in human dilated cardiomyopathy. Circ Res 124(8):1198–1213. https://doi.org/10.1161/CIRCRESAHA.118.314177
Choi JC, Muchir A, Wu W, Iwata S, Homma S, Morrow JP, Worman HJ (2012) Temsirolimus activates autophagy and ameliorates cardiomyopathy caused by lamin A/C gene mutation. Sci Transl Med 4(144):144ra102-144ra102. https://doi.org/10.1126/scitranslmed.3003875
Costa MC et al (2021) Circulating circRNA as biomarkers for dilated cardiomyopathy etiology. J Mol Med 99(12):1711–1725. https://doi.org/10.1007/s00109-021-02119-6
Cresci Sharon et al (2019) Heart failure in the era of precision medicine: a scientific statement from the American Heart Association. Circu: Genom Precis Med 12(10):e000058. https://doi.org/10.1161/HCG.0000000000000058
Cui Y et al (2019) Single-cell transcriptome analysis maps the developmental track of the human heart. Cell Rep 26(7):1934–1950. https://doi.org/10.1016/j.celrep.2019.01.079
Ding Yonghe et al (2019) Haploinsufficiency of mechanistic target of rapamycin ameliorates bag3 cardiomyopathy in adult zebrafish. Dis Models Mech 12(10):dmm040154. https://doi.org/10.1242/dmm.040154
Ding Y, Bu H, Xu X (2020) Modeling inherited cardiomyopathies in adult zebrafish for precision medicine. Front Physiol 11:599244. https://doi.org/10.3389/fphys.2020.599244
Ding Y, Wang M, Bu H, Li J, Lin X, Xu X (2022) An F0-based genetic assay in adult zebrafish for discovering modifier genes of an inherited cardiomyopathy. Dis Model Mech. https://doi.org/10.1242/dmm.049427
Dvornikov AV, de Tombe PP, Xu X (2018) Phenotyping cardiomyopathy in adult zebrafish. Prog Biophys Mol Biol 138:116–125. https://doi.org/10.1016/j.pbiomolbio.2018.05.013
Eschenhagen T, Mummery C, Knollmann BC (2015) Modelling sarcomeric cardiomyopathies in the dish: from human heart samples to iPSC cardiomyocytes. Cardiovasc Res 105(4):424–438. https://doi.org/10.1093/cvr/cvv017
Fang Xi et al (2017) Loss-of-function mutations in co-chaperone BAG3 destabilize small HSPs and cause cardiomyopathy. J Clin Investig 127(8):3189–3200. https://doi.org/10.1172/JCI94310DS1
Fang Xi et al (2019) P209L mutation in Bag3 does not cause cardiomyopathy in mice. Am J Physiol-Heart Circu Physiol 316(2):H392–H399. https://doi.org/10.1152/ajpheart.00714.2018
Fatkin D, Huttner IG, Kovacic JC, Seidman JG, Seidman CE (2019) Precision medicine in the management of dilated cardiomyopathy: JACC state-of-the-art review. J Am Coll Cardiol 74(23):2921–2938. https://doi.org/10.1016/j.jacc.2019.10.011
Flores Ramirez, Ricardo O et al (2021) Consensus transcriptional landscape of human end-stage heart failure. J Am Heart Assoc 10(7):e019667. https://doi.org/10.1161/JAHA.120.019667
Franaszczyk M et al (2014) The BAG3 gene variants in Polish patients with dilated cardiomyopathy: four novel mutations and a genotype-phenotype correlation. J Transl Med 12(1):1–8. https://doi.org/10.1186/1479-5876-12-192
Gao C et al (2016) RBFox1-mediated RNA splicing regulates cardiac hypertrophy and heart failure. J Clin Investig 126(1):195–206. https://doi.org/10.1172/JCI84015
Garnier Sophie et al (2021) Genome-wide association analysis in dilated cardiomyopathy reveals two new players in systolic heart failure on chromosomes 3p25. 1 and 22q11. 23. Euro Heart J 42(20):2000–2011. https://doi.org/10.1093/eurheartj/ehab030
Gigli M et al (2016) A review of the giant protein titin in clinical molecular diagnostics of cardiomyopathies. Front Cardiovasc Medi 3:21. https://doi.org/10.3389/fcvm.2016.00021
Gramlich M et al (2009) Stress-induced dilated cardiomyopathy in a knock-in mouse model mimicking human titin-based disease. J Mol Cell Cardiol 47(3):352–358. https://doi.org/10.1016/j.yjmcc.2009.04.014
Harakalova Magdalena, Asselbergs Folkert W (2018) Systems analysis of dilated cardiomyopathy in the next generation sequencing era. Wiley Interdiscip Rev: Syst Biol Med 10(4):e1419. https://doi.org/10.1002/wsbm.1419
Herman DS et al (2012) Truncations of titin causing dilated cardiomyopathy. N Engl J Med 366(7):619–628. https://doi.org/10.1056/NEJMoa1110186
Hershberger RE, Morales A, Siegfried JD (2010) Clinical and genetic issues in dilated cardiomyopathy: a review for genetics professionals. Genet Med 12(11):655–667. https://doi.org/10.1097/GIM.0b013e3181f2481f
Hershberger, R. E., & Morales, A. (2016). LMNA-related dilated cardiomyopathy. PMID: 20301717.
Hinson JT et al (2015) Titin mutations in iPS cells define sarcomere insufficiency as a cause of dilated cardiomyopathy. Science 349(6251):982–986. https://doi.org/10.1126/science.aaa5458
Homma S, Iwasaki M, Shelton GD, Engvall E, Reed JC, Takayama S (2006) BAG3 deficiency results in fulminant myopathy and early lethality. Am J Pathol 169(3):761–773. https://doi.org/10.2353/ajpath.2006.060250
Huttner IG et al (2018) A-band titin truncation in zebrafish causes dilated cardiomyopathy and hemodynamic stress intolerance. Circu Genom Precis Med 11(8):e002135
Jiang He et al (2021) Functional analysis of a gene-edited mouse model to gain insights into the disease mechanisms of a titin missense variant. Basic Res Cardiol 116(1):1–18. https://doi.org/10.1007/s00395-021-00853-z
Jordan E et al (2021) Evidence-based assessment of genes in dilated cardiomyopathy. Circulation 144(1):7–19. https://doi.org/10.1161/CIRCULATIONAHA.120.053033
Judge Luke M et al (2017) A BAG3 chaperone complex maintains cardiomyocyte function during proteotoxic stress. JCI Insight. https://doi.org/10.1172/jci.insight.94623
Ke Yang, Jian-Yuan Huang, Ping Zhou, Yue Wang, Na Xing, Jian Yang, Kai-Xuan Lin, Yi-Fan Sun, Han-Bin Lin, Rong Li (2022) The progressive application of single-cell RNA sequencing technology in cardiovascular diseases. Biomed Pharmacother 154:113604. https://doi.org/10.1016/j.biopha.2022.113604
Kimura K et al (2021) Overexpression of human BAG3P209L in mice causes restrictive cardiomyopathy. Nat Commun 12(1):1–17. https://doi.org/10.1038/s41467-021-23858-7
Knezevic T et al (2015) BAG3: a new player in the heart failure paradigm. Heart Fail Rev 20(4):423–434. https://doi.org/10.1007/s10741-015-9487-6
Koenig AL, Shchukina I, Amrute J, Andhey PS, Zaitsev K, Lai L, Lavine KJ (2022) Single-cell transcriptomics reveals cell-type-specific diversification in human heart failure. Nat Cardiovasc Res 1(3):263–280. https://doi.org/10.1038/s44161-022-00028-6
Koentges C et al (2018) Gene expression analysis to identify mechanisms underlying heart failure susceptibility in mice and humans. Basic Res Cardiol 113(1):1–17. https://doi.org/10.1007/s00395-017-0666-6
Koshimizu, E., et al. (2011). Embryonic senescence and laminopathies in a progeroid zebrafish model. PLoS One, 6(3), e17688. https://doi.org/10.1371/journal.pone.0017688
Lau E, Paik DT, Wu JC (2019) Systems-wide approaches in induced pluripotent stem cell models. Annu Rev Pathol 14:395. https://doi.org/10.1146/annurev-pathmechdis-012418-013046
Lee JH, Gao C, Peng G, Greer C, Ren S, Wang Y, Xiao X (2011) Analysis of transcriptome complexity through RNA sequencing in normal and failing murine hearts. Circ Res 109(12):1332–1341. https://doi.org/10.1161/CIRCRESAHA.111.249433
Lee J et al (2019) Activation of PDGF pathway links LMNA mutation to dilated cardiomyopathy. Nature 572(7769):335–340. https://doi.org/10.1038/s41586-019-1406-x
Li J, Hong S, Chen W, Zuo E, Yang H (2019) Advances in detecting and reducing off-target effects generated by CRISPR-mediated genome editing. J Genet Genom 46(11):513–521. https://doi.org/10.1016/j.jgg.2019.11.002
Liao D et al (2019) Upregulation of Yy1 suppresses dilated cardiomyopathy caused by Ttn insufficiency. Sci Rep 9(1):1–12. https://doi.org/10.1038/s41598-019-52796-0
Martin TG et al (2021) Cardiomyocyte contractile impairment in heart failure results from reduced BAG3-mediated sarcomeric protein turnover. Nat Commun 12(1):1–1. https://doi.org/10.1038/s41467-021-23272-z
Mazzarotto F et al (2020) Reevaluating the genetic contribution of monogenic dilated cardiomyopathy. Circulation 141(5):387–398. https://doi.org/10.1161/CIRCULATIONAHA.119.037661
McDermott-Roe, et al (2019) Investigation of a dilated cardiomyopathy–associated variant in BAG3 using genome-edited iPSC-derived cardiomyocytes. JCI Insight. https://doi.org/10.1172/jci.insight.128799
McNally EM, Mestroni L (2017) Dilated cardiomyopathy: genetic determinants and mechanisms. Circ Res 121(7):731–748. https://doi.org/10.1161/CIRCRESAHA.116.309396
Mehdiabadi NR et al (2022) Defining the fetal gene program at single-cell resolution in pediatric dilated cardiomyopathy. Circulation 146(14):1105–1108. https://doi.org/10.1161/CIRCULATIONAHA.121.057763
Muchir A, Pavlidis P, Decostre V, Herron AJ, Arimura T, Bonne G, Worman HJ (2007) Activation of MAPK pathways links LMNA mutations to cardiomyopathy in Emery-Dreifuss muscular dystrophy. J Clin Investig 117(5):1282–1293. https://doi.org/10.1172/JCI29042
Muchir A, Shan J, Bonne G, Lehnart SE, Worman HJ (2009) Inhibition of extracellular signal-regulated kinase signaling to prevent cardiomyopathy caused by mutation in the gene encoding A-type lamins. Hum Mol Genet 18(2):241–247. https://doi.org/10.1093/hmg/ddn343
Myers VD et al (2018a) Association of variants in BAG3 with cardiomyopathy outcomes in African American individuals. JAMA Cardiol 3(10):929–938. https://doi.org/10.1001/jamacardio.2018.2541
Myers VD et al (2018b) Haplo-insufficiency of Bcl2-associated athanogene 3 in mice results in progressive left ventricular dysfunction, β-adrenergic insensitivity, and increased apoptosis. J Cell Physiol 233(9):6319–6326. https://doi.org/10.1002/jcp.26482
Nicin L et al (2021) Single nuclei sequencing reveals novel insights into the regulation of cellular signatures in children with dilated cardiomyopathy. Circulation 143(17):1704–1719. https://doi.org/10.1161/CIRCULATIONAHA.120.051391
Nicolas HA, Hua K, Quigley H, Ivare J, Tesson F, Akimenko MA (2022) A CRISPR/Cas9 zebrafish lamin A/C mutant model of muscular laminopathy. Dev Dyn 251(4):645–661. https://doi.org/10.1002/dvdy.427
Norton N et al (2011) Genome-wide studies of copy number variation and exome sequencing identify rare variants in BAG3 as a cause of dilated cardiomyopathy. Am J Hum Genet 88(3):273–282. https://doi.org/10.1016/j.ajhg.2011.01.016
Olson TM, Michels VV, Thibodeau SN, Tai YS, Keating MT (1998) Actin mutations in dilated cardiomyopathy, a heritable form of heart failure. Science 280(5364):750–752. https://doi.org/10.1126/science.280.5364.750
Paik DT et al (2018) Large-scale single-cell RNA-seq reveals molecular signatures of heterogeneous populations of human induced pluripotent stem cell-derived endothelial cells. Circ Res 123(4):443–450. https://doi.org/10.1161/CIRCRESAHA.118.312913
Park HY (2017) Hereditary dilated cardiomyopathy: recent advances in genetic diagnostics. Korean Circu J 47(3):291–298. https://doi.org/10.4070/kcj.2016.0017
Perrot A et al (2009) Identification of mutational hot spots in LMNA encoding lamin A/C in patients with familial dilated cardiomyopathy. Basic Res Cardiol 104(1):90–99. https://doi.org/10.1007/s00395-008-0748-6
Reichart D, Magnussen C, Zeller T, Blankenberg S (2019) Dilated cardiomyopathy: from epidemiologic to genetic phenotypes: a translational review of current literature. J Intern Med 286(4):362–372. https://doi.org/10.1111/joim.12944
Reuter H et al (2022) Aging activates the immune system and alters the regenerative capacity in the zebrafish heart. Cells 11(3):345. https://doi.org/10.3390/cells11030345
Roberts Angharad M et al (2015) Integrated allelic, transcriptional, and phenomic dissection of the cardiac effects of titin truncations in health and disease. Sci Transl Med 7(270):270ra6-270ra6. https://doi.org/10.1126/scitranslmed.3010134
Santiago CF, Huttner IG, Fatkin D (2021) Mechanisms of TTN tv-related dilated cardiomyopathy: insights from zebrafish models. J Cardiovasc Dev Dis 8(2):10. https://doi.org/10.3390/jcdd8020010
Sayed Nazish et al (2020) Clinical trial in a dish using iPSCs shows lovastatin improves endothelial dysfunction and cellular cross-talk in LMNA cardiomyopathy. Sci Transl Med 12(554):eaax9276. https://doi.org/10.1126/scitranslmed.aax9276
Schafer S et al (2017) Titin-truncating variants affect heart function in disease cohorts and the general population. Nat Genet 49(1):46–53. https://doi.org/10.1038/ng.3719
Seeley M, Huang W, Chen Z, Wolff WO, Lin X, Xu X (2007) Depletion of zebrafish titin reduces cardiac contractility by disrupting the assembly of Z-discs and A-bands. Circ Res 100(2):238–245. https://doi.org/10.1161/01.RES.0000255758.69821.b5
Sehnert AJ, Huq A, Weinstein BM, Walker C, Fishman M, Stainier DY (2002) Cardiac troponin T is essential in sarcomere assembly and cardiac contractility. Nat Genet 31(1):106–110. https://doi.org/10.1038/ng875
Sewanan LR, Campbell SG (2020) Modelling sarcomeric cardiomyopathies with human cardiomyocytes derived from induced pluripotent stem cells. J Physiol 598(14):2909–2922. https://doi.org/10.1113/JP276753
Shah PP et al (2021) Pathogenic LMNA variants disrupt cardiac lamina-chromatin interactions and de-repress alternative fate genes. Cell Stem Cell 28(5):938–954. https://doi.org/10.1016/j.stem.2020.12.016
Shao X, Fu Y, Ma J, Li X, Lu C, Zhang R (2020) Functional alterations and transcriptomic changes during zebrafish cardiac aging. Biogerontology 21(5):637–652. https://doi.org/10.1007/s10522-020-09881-z
Shi X, Zhang L, Li Y, Xue J, Liang F, Ni HW, He B (2022) Integrative analysis of bulk and single-cell RNA sequencing data reveals cell types involved in heart failure. Front Bioeng Biotechnol 9:779225. https://doi.org/10.3389/fbioe.2021.779225
Shih YH, Zhang Y, Ding Y, Ross CA, Li H, Olson TM, Xu X (2015) Cardiac transcriptome and dilated cardiomyopathy genes in zebrafish. Circu Cardiovasc Genet 8(2):261–269. https://doi.org/10.1161/CIRCGENETICS.114.000702
Sielemann K et al (2020) Distinct myocardial transcriptomic profiles of cardiomyopathies stratified by the mutant genes. Genes 11(12):1430. https://doi.org/10.3390/genes11121430
Siu Chung-Wah et al (2012) Modeling of lamin A/C mutation premature cardiac aging using patient-specific induced pluripotent stem cells. Aging (Albany NY) 4(11):803. https://doi.org/10.18632/aging.100503
Sun X, Hoage T, Bai P, Ding Y, Chen Z, Zhang R et al (2009) Cardiac hypertrophy involves both myocyte hypertrophy and hyperplasia in anemic zebrafish. PLoS One 4:e6596. https://doi.org/10.1371/journal.pone.0006596
Tabish AM, Azzimato V, Alexiadis A, Buyandelger B, Knöll R (2017) Genetic epidemiology of titin-truncating variants in the etiology of dilated cardiomyopathy. Biophys Rev 9(3):207–223. https://doi.org/10.1007/s12551-017-0265-7
Tesson F, Saj M, Uvaize MM, Nicolas H, Płoski R, Bilińska Z (2014) Lamin A/C mutations in dilated cardiomyopathy. Cardiol J 21(4):331–342. https://doi.org/10.5603/CJ.a2014.0037
Verdonschot JAJ et al (2018) Titin cardiomyopathy leads to altered mitochondrial energetics, increased fibrosis and long-term life-threatening arrhythmias. Eur Heart J 39(10):864–873. https://doi.org/10.1093/eurheartj/ehx808
Verdonschot JAJ et al (2020) Distinct cardiac transcriptomic clustering in titin and lamin A/C-associated dilated cardiomyopathy patients. Circulation 142(12):1230–1232. https://doi.org/10.1161/CIRCULATIONAHA.119.045118
Verdonschot JAJ et al (2021) Phenotypic clustering of dilated cardiomyopathy patients highlights important pathophysiological differences. Eur Heart J 42(2):162–174. https://doi.org/10.1093/eurheartj/ehaa841
Verma AD, Parnaik VK (2017) Heart-specific expression of laminopathic mutations in transgenic zebrafish. Cell Biol Int 41(7):809–819. https://doi.org/10.1002/cbin.10784
Villard E et al (2011) A genome-wide association study identifies two loci associated with heart failure due to dilated cardiomyopathy. Eur Heart J 32(9):1065–1076. https://doi.org/10.1093/eurheartj/ehr105
Weintraub RG, Semsarian C, Macdonald P (2017) Dilated cardiomyopathy. The Lancet 390(10092):400–414. https://doi.org/10.1016/S0140-6736(16)31713-5
Whiting A, Wardale J, Trinick J (1989) Does titin regulate the length of muscle thick filaments? J Mol Biol 205(1):263–268. https://doi.org/10.1016/0022-2836(89)90381-1
Wu W, Muchir A, Shan J, Bonne G, Worman HJ (2011) Mitogen-activated protein kinase inhibitors improve heart function and prevent fibrosis in cardiomyopathy caused by mutation in lamin A/C gene. Circulation 123(1):53–61. https://doi.org/10.1161/CIRCULATIONAHA.110.970673
Wu W, Iwata S, Homma S, Worman HJ, Muchir A (2014) Depletion of extracellular signal-regulated kinase 1 in mice with cardiomyopathy caused by lamin A/C gene mutation partially prevents pathology before isoenzyme activation. Hum Mol Genet 23(1):1–11. https://doi.org/10.1093/hmg/ddt387
Wyles SP et al (2016) Modeling structural and functional deficiencies of RBM20 familial dilated cardiomyopathy using human induced pluripotent stem cells. Hum Mol Genet 25(2):254–265. https://doi.org/10.1093/hmg/ddv468
Xu X, Meiler SE, Zhong TP, Mohideen M, Crossley DA, Burggren WW, Fishman MC (2002) Cardiomyopathy in zebrafish due to mutation in an alternatively spliced exon of titin. Nat Genet 30(2):205–209. https://doi.org/10.1038/ng816
Yamada S, Nomura S (2020) Review of single-cell RNA sequencing in the heart. Int J Mol Sci 21(21):8345. https://doi.org/10.3390/ijms21218345
Yang Jin et al (2022) Phenotypic screening with deep learning identifies HDAC6 inhibitors as cardioprotective in a BAG3 mouse model of dilated cardiomyopathy. Sci Transl Med 14(652):eabl5654. https://doi.org/10.1126/scitranslmed.abl5654
Zaunbrecher RJ et al (2019) Cronos titin is expressed in human cardiomyocytes and necessary for normal sarcomere function. Circulation 140(20):1647–1660. https://doi.org/10.1161/CIRCULATIONAHA.119.039521
Zhang X, Shao X, Zhang R, Zhu R, Feng R (2021) Integrated analysis reveals the alterations that LMNA interacts with euchromatin in LMNA mutation-associated dilated cardiomyopathy. Clin Epigenetics 13(1):1–13. https://doi.org/10.1186/s13148-020-00996-1
Zhang Yike et al (2022) Familial atrial myopathy in a large multigenerational heart-hand syndrome pedigree carrying an LMNA missense variant in rod 2B domain (p. R335W). Heart Rhythm 19(3):466–475. https://doi.org/10.1016/j.hrthm.2021.11.022
Zou Jun et al (2015) An internal promoter underlies the difference in disease severity between N-and C-terminal truncation mutations of Titin in zebrafish. Elife 4:e09406. https://doi.org/10.7554/eLife.09406.025
Acknowledgements
This work was supported in part by grants from NIH (HL107304, HL081753) and the Mayo Foundation to X.X., NIH (T32 HL007111) to M.K.
Funding
Mayo Foundation for Medical Education and Research, T32 HL007111, National Institutes of Health, HL107304.
Author information
Authors and Affiliations
Contributions
MK and XX designed and wrote the manuscript. DM-R contributed to sections that are related to BAG3 DCM.
Corresponding author
Ethics declarations
Conflict of interest
The authors do not have any conflicts of interest.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Koslow, M., Mondaca-Ruff, D. & Xu, X. Transcriptome studies of inherited dilated cardiomyopathies. Mamm Genome 34, 312–322 (2023). https://doi.org/10.1007/s00335-023-09978-z
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1007/s00335-023-09978-z