Your browser doesn't support javascript.
loading
Genetic and Drug Inhibition of LDH-A: Effects on Murine Gliomas.
Maeda, Masatomo; Ko, Myat; Mane, Mayuresh M; Cohen, Ivan J; Shindo, Masahiro; Vemuri, Kiranmayi; Serganova, Inna; Blasberg, Ronald.
Afiliação
  • Maeda M; Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
  • Ko M; Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
  • Mane MM; Department of Neurosurgery, Nozaki Tokushukai Hospital, Osaka 5740074, Japan.
  • Cohen IJ; Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
  • Shindo M; Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
  • Vemuri K; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
  • Serganova I; Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
  • Blasberg R; Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
Cancers (Basel) ; 14(9)2022 May 06.
Article em En | MEDLINE | ID: mdl-35565435
ABSTRACT
The effects of the LDH-A depletion via shRNA knockdown on three murine glioma cell lines and corresponding intracranial (i.c.) tumors were studied and compared to pharmacologic (GNE-R-140) inhibition of the LDH enzyme complex, and to shRNA scrambled control (NC) cell lines. The effects of genetic-shRNA LDH-A knockdown and LDH drug-targeted inhibition (GNE-R-140) on tumor-cell metabolism, tumor growth, and animal survival were similar. LDH-A KD and GNE-R-140 unexpectedly increased the aggressiveness of GL261 intracranial gliomas, but not CT2A and ALTS1C1 i.c. gliomas. Furthermore, the bioenergetic profiles (ECAR and OCR) of GL261 NC and LDH-A KD cells under different nutrient limitations showed that (a) exogenous pyruvate is not a major carbon source for metabolism through the TCA cycle of native GL261 cells; and (b) the unique upregulation of LDH-B that occurs in GL261 LDH-A KD cells results in these cells being better able to (i) metabolize lactate as a primary carbon source through the TCA cycle, (ii) be a net consumer of lactate, and (iii) showed a significant increase in the proliferation rate following the addition of 10 mM lactate to the glucose-free media (only seen in GL261 KD cells). Our study suggests that inhibition of LDH-A/glycolysis may not be a general strategy to inhibit the i.c. growth of all gliomas, since the level of LDH-A expression and its interplay with LDH-B can lead to complex metabolic interactions between tumor cells and their environment. Metabolic-inhibition treatment strategies need to be carefully assessed, since the inhibition of glycolysis (e.g., inhibition of LDH-A) may lead to the unexpected development and activation of alternative metabolic pathways (e.g., upregulation of lipid metabolism and fatty-acid oxidation pathways), resulting in enhanced tumor-cell survival in a nutrient-limited environment and leading to increased tumor aggressiveness.
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Idioma: En Revista: Cancers (Basel) Ano de publicação: 2022 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Base de dados: MEDLINE Idioma: En Revista: Cancers (Basel) Ano de publicação: 2022 Tipo de documento: Article País de afiliação: Estados Unidos