Phosphoglycerate dehydrogenase inhibition induces p-mTOR-independent autophagy and promotes multilineage differentiation in embryonal carcinoma stem-like cells

Cell Death Dis. 2018 Sep 24;9(10):990. doi: 10.1038/s41419-018-0997-8.

Abstract

Cancer cells with a less differentiated stem-like phenotype are more resistant to therapeutic manipulations than their differentiated counterparts, and are considered as one of the main causes of cancer persistence and relapse. As such, induction of differentiation in cancer stem-like cells (CSLCs) has emerged as an alternative strategy to enhance the efficacy of anticancer therapies. CSLCs are metabolically distinct from differentiated cells, and any aberration from the intrinsic metabolic state can induce differentiation of CSLCs. Therefore, metabolism-related molecular targets, with a capacity to promote differentiation within CSLCs, are of therapeutic importance. Here, we demonstrate that phosphoglycerate dehydrogenase (PHGDH), an essential enzyme catalyzing the synthesis of amino acid serine, is important for maintaining the poorly differentiated, stem-like state of CSLCs. Our data shows that PHGDH deficiency impairs the tumorsphere formation capacity in embryonal carcinoma stem-like cells (ECSLCs), breast cancer stem-like cells (BCSLCs) and patient-derived brain tumor-initiating cells (BTICs), which is accompanied by the reduced expression of characteristic stemness-promoting factors, such as Oct4, Nanog, Sox-2, and Bmi-1. Mechanistically, PHGDH deficiency in ECSLCs promotes differentiation to various lineages via degradation of Oct4 and by increasing the stability of differentiation marker β3-tubulin. Furthermore, PHGDH inhibition promotes p-mTOR independent but Beclin-1-dependent autophagy, independent of apoptosis. When studied in combination, the inhibition of both PHGDH and p-mTOR in ECSLCs causes further augmentation of autophagy, and additionally promotes apoptosis, demonstrating the clinical applicability of PHGDH-based manipulations in cancer therapies. Recapitulating these in vitro findings in CSLC models, the intratumoral PHGDH expression in patient-derived tumors is positively correlated with the mRNA levels of stemness factors, especially Oct4, and cancer patients co-expressing high levels of PHGDH and Oct4 display significantly lower survival than those with low PHGDH/Oct4 co-expression. Altogether, this study identifies a clinically-relevant role for PHGDH in the regulation of stemness-differentiation axis within CSLCs.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Autophagy*
  • Beclin-1 / metabolism
  • Brain Neoplasms / metabolism*
  • Brain Neoplasms / pathology
  • Carbohydrate Metabolism, Inborn Errors / metabolism
  • Carcinoma, Embryonal / metabolism*
  • Carcinoma, Embryonal / pathology
  • Cell Differentiation*
  • Cell Line, Tumor
  • Embryonal Carcinoma Stem Cells / metabolism*
  • Glioblastoma / metabolism*
  • Glioblastoma / pathology
  • Humans
  • Male
  • Microcephaly / metabolism
  • Octamer Transcription Factor-3 / metabolism
  • Phosphoglycerate Dehydrogenase / antagonists & inhibitors
  • Phosphoglycerate Dehydrogenase / deficiency
  • Phosphoglycerate Dehydrogenase / genetics
  • Phosphoglycerate Dehydrogenase / metabolism*
  • Proteolysis
  • Psychomotor Disorders / metabolism
  • Seizures / metabolism
  • TOR Serine-Threonine Kinases / metabolism*
  • Testicular Neoplasms / metabolism*
  • Testicular Neoplasms / pathology
  • Transfection
  • Tubulin / metabolism
  • Ubiquitination

Substances

  • BECN1 protein, human
  • Beclin-1
  • Octamer Transcription Factor-3
  • POU5F1 protein, human
  • TUBB3 protein, human
  • Tubulin
  • Phosphoglycerate Dehydrogenase
  • MTOR protein, human
  • TOR Serine-Threonine Kinases

Supplementary concepts

  • Phosphoglycerate Dehydrogenase Deficiency

Grants and funding