Your browser doesn't support javascript.
loading
The redox-active defensive Selenoprotein T as a novel stress sensor protein playing a key role in the pathophysiology of heart failure.
De Bartolo, Anna; Pasqua, Teresa; Romeo, Naomi; Rago, Vittoria; Perrotta, Ida; Giordano, Francesca; Granieri, Maria Concetta; Marrone, Alessandro; Mazza, Rosa; Cerra, Maria Carmela; Lefranc, Benjamin; Leprince, Jérôme; Anouar, Youssef; Angelone, Tommaso; Rocca, Carmine.
Afiliación
  • De Bartolo A; Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy.
  • Pasqua T; Department of Health Science, University Magna Graecia of Catanzaro, 88100, Catanzaro, Italy.
  • Romeo N; Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy.
  • Rago V; Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy.
  • Perrotta I; Centre for Microscopy and Microanalysis (CM2), Department of Biology, E. and E. S. (DiBEST), University of Calabria, 87036, Rende, Italy.
  • Giordano F; Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy.
  • Granieri MC; Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy.
  • Marrone A; Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy.
  • Mazza R; Organ and System Physiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy.
  • Cerra MC; Organ and System Physiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy.
  • Lefranc B; UNIROUEN, Inserm U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication (NorDiC), Rouen Normandie University, 76000, Mont-Saint-Aignan, France.
  • Leprince J; UNIROUEN, UMS-UAR HERACLES, PRIMACEN, Cell Imaging Platform of Normandy, Institute for Research and Innovation in Biomedicine (IRIB), 76183, Rouen, France.
  • Anouar Y; UNIROUEN, Inserm U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication (NorDiC), Rouen Normandie University, 76000, Mont-Saint-Aignan, France.
  • Angelone T; UNIROUEN, UMS-UAR HERACLES, PRIMACEN, Cell Imaging Platform of Normandy, Institute for Research and Innovation in Biomedicine (IRIB), 76183, Rouen, France.
  • Rocca C; UNIROUEN, Inserm U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication (NorDiC), Rouen Normandie University, 76000, Mont-Saint-Aignan, France.
J Transl Med ; 22(1): 375, 2024 Apr 20.
Article en En | MEDLINE | ID: mdl-38643121
ABSTRACT
Maladaptive cardiac hypertrophy contributes to the development of heart failure (HF). The oxidoreductase Selenoprotein T (SELENOT) emerged as a key regulator during rat cardiogenesis and acute cardiac protection. However, its action in chronic settings of cardiac dysfunction is not understood. Here, we investigated the role of SELENOT in the pathophysiology of HF (i) by designing a small peptide (PSELT), recapitulating SELENOT activity via the redox site, and assessed its beneficial action in a preclinical model of HF [aged spontaneously hypertensive heart failure (SHHF) rats] and against isoproterenol (ISO)-induced hypertrophy in rat ventricular H9c2 and adult human AC16 cardiomyocytes; (ii) by evaluating the SELENOT intra-cardiomyocyte production and secretion under hypertrophied stimulation. Results showed that PSELT attenuated systemic inflammation, lipopolysaccharide (LPS)-induced macrophage M1 polarization, myocardial injury, and the severe ultrastructural alterations, while counteracting key mediators of cardiac fibrosis, aging, and DNA damage and restoring desmin downregulation and SELENOT upregulation in the failing hearts. In the hemodynamic assessment, PSELT improved the contractile impairment at baseline and following ischemia/reperfusion injury, and reduced infarct size in normal and failing hearts. At cellular level, PSELT counteracted ISO-mediated hypertrophy and ultrastructural alterations through its redox motif, while mitigating ISO-triggered SELENOT intracellular production and secretion, a phenomenon that presumably reflects the extent of cell damage. Altogether, these results indicate that SELENOT could represent a novel sensor of hypertrophied cardiomyocytes and a potential PSELT-based new therapeutic approach in myocardial hypertrophy and HF.
Asunto(s)
Palabras clave

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Reductasa de Tiorredoxina-Disulfuro / Selenoproteínas / Insuficiencia Cardíaca Límite: Adult / Aged / Animals / Humans Idioma: En Revista: J Transl Med Año: 2024 Tipo del documento: Article País de afiliación: Italia

Texto completo: 1 Bases de datos: MEDLINE Asunto principal: Reductasa de Tiorredoxina-Disulfuro / Selenoproteínas / Insuficiencia Cardíaca Límite: Adult / Aged / Animals / Humans Idioma: En Revista: J Transl Med Año: 2024 Tipo del documento: Article País de afiliación: Italia