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.

Highlights

  • A well-established feature of cancer cells is their enhanced capacity to proliferate[1]

  • Patient-derived were compared between brain tumor-initiating cells (BTICs) derived from three individual patients (BT935, BT954, and BT698) and NT2/D1 embryonal carcinoma stem-like cells (ECSLCs) by western blot (WB) analysis. (iv) Quantification of Phosphoglycerate dehydrogenase (PHGDH) protein levels normalized to Ponceau stain. b (i) IMR90, WI38, H1299, A549, MDA-MB-231, MDA-MB-468, HMLE, HMLE-Ras (HMLER), HMLER shE-Cadherin (HMLERshECad), and NT2/D1 cells were subjected to WB analysis for PHGDH and Oct4.(ii) Quantification of PHGDH

  • Using a characteristic flux analysis that monitors the dynamic changes of the autophagic degradation process[27], we found that treatment with the late-stage autophagy inhibitor chloroquine (CQ) further enhanced the accumulation of LC3A-II, LC3B-II and p62/SQSTM1 in PHGDH KD NT2/D1 ECSLCs compared to the scrambled control, suggesting PHGDH KD in NT2/D1 ECSLCs enhances autophagic

Read more

Summary

Introduction

A well-established feature of cancer cells is their enhanced capacity to proliferate[1]. Serine is a non-essential amino acid (NEAA) that is used in the synthesis of proteins and nucleic acids and is rapidly consumed by cancer cells[3,4]. The serine biosynthesis pathway is often upregulated in cancer cells. Phosphoglycerate dehydrogenase (PHGDH), the enzyme which catalyzes the first step of the serine biosynthesis pathway, has been shown to be genomically amplified in many breast cancers and melanomas[5,6]. High levels of PHGDH have been associated with enhanced proliferation and poor prognosis in various types of cancers, and cancer cells that harbor high levels of PHGDH have been shown to be more susceptible to PHGDH inhibition[5,7]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call