Cargando…

Glucose-6-phosphate dehydrogenase is critical for suppression of cardiac hypertrophy by H(2)S

Hydrogen Sulfide (H(2)S), recently identified as the third endogenously produced gaseous messenger, is a promising therapeutic prospect for multiple cardio-pathological states, including myocardial hypertrophy. The molecular niche of H(2)S in normal or diseased cardiac cells is, however, sparsely un...

Descripción completa

Detalles Bibliográficos
Autores principales: Chhabra, Aastha, Mishra, Shalini, Kumar, Gaurav, Gupta, Asheesh, Keshri, Gaurav Kumar, Bharti, Brij, Meena, Ram Niwas, Prabhakar, Amit Kumar, Singh, Dinesh Kumar, Bhargava, Kalpana, Sharma, Manish
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5841415/
https://www.ncbi.nlm.nih.gov/pubmed/29531803
http://dx.doi.org/10.1038/s41420-017-0010-9
Descripción
Sumario:Hydrogen Sulfide (H(2)S), recently identified as the third endogenously produced gaseous messenger, is a promising therapeutic prospect for multiple cardio-pathological states, including myocardial hypertrophy. The molecular niche of H(2)S in normal or diseased cardiac cells is, however, sparsely understood. Here, we show that β-adrenergic receptor (β-AR) overstimulation, known to produce hypertrophic effects in cardiomyocytes, rapidly decreased endogenous H(2)S levels. The preservation of intracellular H(2)S levels under these conditions strongly suppressed hypertrophic responses to adrenergic overstimulation, thus suggesting its intrinsic role in this process. Interestingly, unbiased global transcriptome sequencing analysis revealed an integrated metabolic circuitry, centrally linked by NADPH homeostasis, as the direct target of intracellular H(2)S augmentation. Within these gene networks, glucose-6-phosphate dehydrogenase (G6PD), the first and rate-limiting enzyme (producing NADPH) in pentose phosphate pathway, emerged as the critical node regulating cellular effects of H(2)S. Utilizing both cellular and animal model systems, we show that H(2)S-induced elevated G6PD activity is critical for the suppression of cardiac hypertrophy in response to adrenergic overstimulation. We also describe experimental evidences suggesting multiple processes/pathways involved in regulation of G6PD activity, sustained over extended duration of time, in response to endogenous H(2)S augmentation. Our data, thus, revealed H(2)S as a critical endogenous regulator of cardiac metabolic circuitry, and also mechanistic basis for its anti-hypertrophic effects.