Your browser doesn't support javascript.
loading
Full-length dystrophin deficiency leads to contractile and calcium transient defects in human engineered heart tissues.
Bremner, Samantha B; Mandrycky, Christian J; Leonard, Andrea; Padgett, Ruby M; Levinson, Alan R; Rehn, Ethan S; Pioner, J Manuel; Sniadecki, Nathan J; Mack, David L.
Afiliação
  • Bremner SB; Department of Bioengineering, University of Washington, Seattle, WA, USA.
  • Mandrycky CJ; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
  • Leonard A; Department of Bioengineering, University of Washington, Seattle, WA, USA.
  • Padgett RM; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
  • Levinson AR; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
  • Rehn ES; Department of Mechanical Engineering, University of Washington, Seattle, WA, USA.
  • Pioner JM; Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.
  • Sniadecki NJ; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
  • Mack DL; Department of Mechanical Engineering, University of Washington, Seattle, WA, USA.
J Tissue Eng ; 13: 20417314221119628, 2022.
Article em En | MEDLINE | ID: mdl-36003954
ABSTRACT
Cardiomyopathy is currently the leading cause of death for patients with Duchenne muscular dystrophy (DMD), a severe neuromuscular disorder affecting young boys. Animal models have provided insight into the mechanisms by which dystrophin protein deficiency causes cardiomyopathy, but there remains a need to develop human models of DMD to validate pathogenic mechanisms and identify therapeutic targets. Here, we have developed human engineered heart tissues (EHTs) from CRISPR-edited, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) expressing a truncated dystrophin protein lacking part of the actin-binding domain. The 3D EHT platform enables direct measurement of contractile force, simultaneous monitoring of Ca2+ transients, and assessment of myofibril structure. Dystrophin-mutant EHTs produced less contractile force as well as delayed kinetics of force generation and relaxation, as compared to isogenic controls. Contractile dysfunction was accompanied by reduced sarcomere length, increased resting cytosolic Ca2+ levels, delayed Ca2+ release and reuptake, and increased beat rate irregularity. Transcriptomic analysis revealed clear differences between dystrophin-deficient and control EHTs, including downregulation of genes related to Ca2+ homeostasis and extracellular matrix organization, and upregulation of genes related to regulation of membrane potential, cardiac muscle development, and heart contraction. These findings indicate that the EHT platform provides the cues necessary to expose the clinically-relevant, functional phenotype of force production as well as mechanistic insights into the role of Ca2+ handling and transcriptomic dysregulation in dystrophic cardiac function, ultimately providing a powerful platform for further studies in disease modeling and drug discovery.
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Contexto em Saúde: 6_ODS3_enfermedades_notrasmisibles Base de dados: MEDLINE Tipo de estudo: Prognostic_studies Idioma: En Revista: J Tissue Eng Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Contexto em Saúde: 6_ODS3_enfermedades_notrasmisibles Base de dados: MEDLINE Tipo de estudo: Prognostic_studies Idioma: En Revista: J Tissue Eng Ano de publicação: 2022 Tipo de documento: Article