Your browser doesn't support javascript.
loading
LMNA-Related Dilated Cardiomyopathy: Single-Cell Transcriptomics during Patient-Derived iPSC Differentiation Support Cell Type and Lineage-Specific Dysregulation of Gene Expression and Development for Cardiomyocytes and Epicardium-Derived Cells with Lamin A/C Haploinsufficiency.
Zaragoza, Michael V; Bui, Thuy-Anh; Widyastuti, Halida P; Mehrabi, Mehrsa; Cang, Zixuan; Sha, Yutong; Grosberg, Anna; Nie, Qing.
Affiliation
  • Zaragoza MV; UCI Cardiogenomics Program, Pediatrics and Biological Chemistry, UC Irvine School of Medicine, Irvine, CA 92697, USA.
  • Bui TA; Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA.
  • Widyastuti HP; UCI Cardiogenomics Program, Pediatrics and Biological Chemistry, UC Irvine School of Medicine, Irvine, CA 92697, USA.
  • Mehrabi M; UCI Cardiogenomics Program, Pediatrics and Biological Chemistry, UC Irvine School of Medicine, Irvine, CA 92697, USA.
  • Cang Z; Biomedical Engineering and Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, University of California, Irvine, Irvine, CA 92697, USA.
  • Sha Y; Mathematics and NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA.
  • Grosberg A; Mathematics and NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA.
  • Nie Q; Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA.
Cells ; 13(17)2024 Sep 03.
Article in En | MEDLINE | ID: mdl-39273049
ABSTRACT
LMNA-related dilated cardiomyopathy (DCM) is an autosomal-dominant genetic condition with cardiomyocyte and conduction system dysfunction often resulting in heart failure or sudden death. The condition is caused by mutation in the Lamin A/C (LMNA) gene encoding Type-A nuclear lamin proteins involved in nuclear integrity, epigenetic regulation of gene expression, and differentiation. The molecular mechanisms of the disease are not completely understood, and there are no definitive treatments to reverse progression or prevent mortality. We investigated possible mechanisms of LMNA-related DCM using induced pluripotent stem cells derived from a family with a heterozygous LMNA c.357-2A>G splice-site mutation. We differentiated one LMNA-mutant iPSC line derived from an affected female (Patient) and two non-mutant iPSC lines derived from her unaffected sister (Control) and conducted single-cell RNA sequencing for 12 samples (four from Patients and eight from Controls) across seven time points Day 0, 2, 4, 9, 16, 19, and 30. Our bioinformatics workflow identified 125,554 cells in raw data and 110,521 (88%) high-quality cells in sequentially processed data. Unsupervised clustering, cell annotation, and trajectory inference found complex heterogeneity ten main cell types; many possible subtypes; and lineage bifurcation for cardiac progenitors to cardiomyocytes (CMs) and epicardium-derived cells (EPDCs). Data integration and comparative analyses of Patient and Control cells found cell type and lineage-specific differentially expressed genes (DEGs) with enrichment, supporting pathway dysregulation. Top DEGs and enriched pathways included 10 ZNF genes and RNA polymerase II transcription in pluripotent cells (PP); BMP4 and TGF Beta/BMP signaling, sarcomere gene subsets and cardiogenesis, CDH2 and EMT in CMs; LMNA and epigenetic regulation, as well as DDIT4 and mTORC1 signaling in EPDCs. Top DEGs also included XIST and other X-linked genes, six imprinted genes (SNRPN, PWAR6, NDN, PEG10, MEG3, MEG8), and enriched gene sets related to metabolism, proliferation, and homeostasis. We confirmed Lamin A/C haploinsufficiency by allelic expression and Western blot. Our complex Patient-derived iPSC model for Lamin A/C haploinsufficiency in PP, CM, and EPDC provided support for dysregulation of genes and pathways, many previously associated with Lamin A/C defects, such as epigenetic gene expression, signaling, and differentiation. Our findings support disruption of epigenomic developmental programs, as proposed in other LMNA disease models. We recognized other factors influencing epigenetics and differentiation; thus, our approach needs improvement to further investigate this mechanism in an iPSC-derived model.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Cardiomyopathy, Dilated / Cell Differentiation / Myocytes, Cardiac / Lamin Type A / Induced Pluripotent Stem Cells / Haploinsufficiency / Transcriptome Limits: Adult / Female / Humans Language: En Journal: Cells Year: 2024 Document type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Cardiomyopathy, Dilated / Cell Differentiation / Myocytes, Cardiac / Lamin Type A / Induced Pluripotent Stem Cells / Haploinsufficiency / Transcriptome Limits: Adult / Female / Humans Language: En Journal: Cells Year: 2024 Document type: Article