Epigenetic modification facilitates proline synthase PYCR1 aberrant expression in gastric cancer

https://doi.org/10.1016/j.bbagrm.2022.194829Get rights and content

Highlights

  • PYCR1 overexpression is heterogenous among gastric cancer subtypes.

  • Upregulation of PYCR1 is an early event in the multistep of gastric carcinogenesis.

  • Epigenetic modification is responsible for PYCR1 overexpression in gastric cancer.

Abstract

Background & aims

Pyrroline-5-carboxylate reductase 1 (PYCR1) upregulation contributes to the progression of gastric cancer (GC) and indicates poor survival. However, PYCR1 expression profile in GC subtypes and the mechanism behind its upregulation are not well-studied.

Methods

PYCR1 expression profiles in GC subtypes and different stages of gastric carcinogenesis were assessed in different GC cohorts. Genetic alterations and epigenetic modulation in PYCR1 regulation were further investigated using bioinformatics analysis and in vitro experiments.

Results

PYCR1 expression was significantly higher in intestinal-type GC and associated molecular subtypes in TCGA and ACRG GC cohorts. During the cascade of intestinal-type GC, PYCR1 was continuously increased from normal gastric tissues through to atrophic gastritis, to intraepithelial neoplasia, and to GC. Copy number alterations in PYCR1 were associated with PYCR1 transcript expression. One CpG island was observed in PYCR1 promoter region, and the hypomethylation occurred at this region could contribute to PYCR1 transcriptional activation in GC. Besides, H3K27ac combination was found in PYCR1 promoter, and acetyltransferase p300 induced H3K27ac could promote PYCR1 expression in GC.

Conclusions

PYCR1 expression varies across GC subtypes, with intestinal-type GC and associated molecular subtypes having the highest expression. Hypomethylation at CpG sites and p300-induced H3K27ac modification within PYCR1 promoter could contribute to maintaining PYCR1 overexpression in GC. These results provide us with a new insight into epigenetic modulation in mitochondrial proline metabolism.

Introduction

Metabolic reprogramming is one of the hallmarks of cancer [1]. Targeting cancer metabolism and related metabolic enzymes may provide new insights for cancer treatment. Recently, increasing evidence has demonstrated that rewiring of mitochondrial proline metabolism is emerging as a key pathway in sustaining cancer cell proliferation, survival, and metastasis [2]. Increased proline levels in tumor tissues or body fluid samples have been confirmed by metabolomic technology in gastric cancer (GC), and higher level of proline was also an indicator of GC metastasis [3]. Thus, targeting proline metabolism and related enzymes could be beneficial for cancer treatment [4], [5].

The key enzyme catalyzing the last step of proline biosynthesis, pyrroline-5-carboxylate reductase 1 (PYCR1), is overexpressed in several malignancies including GC . Our previous study has confirmed that PYCR1 upregulation contributes to GC progression and might be a potential biomarker for predicting poor prognosis [6]. We also identified that PYCR1 expression varied according to WHO (World Health Organization) histopathological classification of GC. In addition to histological classification, the Lauren classification, which distinguishes intestinal-type, diffuse-type and mixed-type GC, is the most commonly used [7]. More importantly, molecular subtypes proposed by The Cancer Genome Atlas (TCGA) and Asian Cancer Research Group (ACRG) in recent years that associate molecular features with histological phenotypes and clinical characteristics have deepened our understanding of GC heterogeneity [8], [9]. In this regard, we seek to know whether reprogramming of proline metabolism via PYCR1 in GC is cancer-subtype specific; however, there is little evidence.

On the other hand, the mechanism underlying PYCR1 overexpression in GC remains unknown. Studies including ours have reported that PYCR1 could be regulated by the oncogenic transcription factor c-MYC [10] and PI3K signaling [6], [11]. c-MYC amplification and PIK3CA (coding PI3K) mutation are common molecular events in gastric carcinogenesis [12], [13], which might partially account for PYCR1 upregulation. Apart from transcriptional factors regulation, epigenetic modifications, such as methylation and acetylation, play a critical role in gene expression regulation. Several studies have confirmed that epigenetic alterations, such as aberrant DNA methylation, histone modification, non-coding RNAs and RNA editing are common in gastric malignancies [14]. Therefore, it is necessary to study whether epigenetic modification affects PYCR1 upregulation in GC.

The current study applied bioinformatics analysis combined with histologic and molecular biological experiments to investigate PYCR1 expression patterns in GC subtypes and potential mechanisms underlying PYCR1 overexpression. The findings of this study might contribute to a better understanding of the expression profile and epigenetic regulation of the key enzyme in proline synthesis in carcinogenesis.

Section snippets

Cell lines, antibodies and chemical reagents

The human gastric cancer cell lines AGS (adenocarcinoma/primary, Lauren intestinal type), HGC27 (undifferentiated carcinoma/metastatic lymph node), MKN45 (poorly-differentiated carcinoma/liver metastasis, Lauren diffuse type), and NCI-N87 (well-differentiated adenocarcinoma/liver metastasis, Lauren intestinal type) were purchased from National Infrastructure of Cell Line Resource (Beijing, China). KATO III (signet cell carcinoma/pleural effusion, Lauren diffuse type) was obtained from Procell

PYCR1 is overexpressed in intestinal-type gastric adenocarcinoma and associated molecular subtypes

Using a bioinformatics approach based on patient-derived data, we found that according to Lauren classification, intestinal-type gastric adenocarcinoma presented higher PYCR1 mRNA expression than normal tissues either in TCGA cohort or ACRG cohort (Fig. 1a–b). IHC staining with GC tissues indicated that PYCR1 staining score was higher in intestinal-type GC compared to diffuse-type GC (Fig. 1c). The representative images of PYCR1 expression pattern in three subtypes of GC in Lauren

Discussion

Evidence showed that metabolic shift towards proline synthesis might be a general metabolic rewiring signature for most cancer types [2], [21]. PYCR1, being one of the key enzymes catalyzing proline synthesis, has recently received high emphasis in terms of tumor development and progression [22]. Based on our previous study [6], the current study provides new information on the expression profile of PYCR1 during gastric carcinogenesis and the epigenetic mechanism behind PYCR1 aberrant

Conclusions

Overall, this study found that PYCR1 expression is heterogeneous across GC subtypes, with intestinal-type GC and associated molecular subtypes having the highest expression. The coordinated effects of CpG sites hypomethylation and p300-induced H3K27ac modification occurring within the PYCR1 gene promoter partially contribute to maintaining the higher PYCR1 expression in GC (Fig. 6). The current study combined our previous work to further highlight the importance of proline metabolism catalyzed

Ethics approval and consent to participate

All procedures followed were in accordance with the ethical standards of the responsible committee on human experimentation (institutional and national) and with the Helsinki Declaration of 1964 and later versions. Informed consent to be included in the study, or the equivalent, was obtained from all patients.

Availability of data and materials

All data supporting the findings of this study are available within the article and its supplementary information files. Raw data can be obtained from the corresponding author on reasonable request.

Funding

This study was funded by Special funds for Beijing Key laboratory of Helicobacter pylori infection and upper gastrointestinal diseases from Peking University Third Hospital (Y57405-32) and National Natural Science Foundation of China (81672410).

CRediT authorship contribution statement

Conception and design: LYZ, SYX; Experiment, analysis and interpretation of the data: SYX, XYY, JXY, XLT, ZHY; Acquisition of data: SYX, XYY, JXY; Statistical analysis: SYX, XYY; Drafting of the article: SYX; Manuscript revision and supervision: LYZ, SYX.

Declaration of competing interest

The authors declare that they have no competing interests.

Acknowledgments

We thank the whole staff in the Medical Research Center of Peking University Third Hospital for their excellent technical assistance on experiments. We thank all anonymous reviewers helping us improve the quality of our manuscript.

References (46)

  • B. Faubert et al.

    Metabolic reprogramming and cancer progression

    Science

    (2020)
  • C. D'Aniello et al.

    Proline metabolism in tumor growth and metastatic progression

    Front. Oncol.

    (2020)
  • S. Xiao et al.

    Gastric cancer: Metabolic and metabolomics perspectives (review)

    Int. J. Oncol.

    (2017)
  • K. Milne et al.

    A fragment-like approach to PYCR1 inhibition

    Bioorg. Med. Chem. Lett.

    (2019)
  • E.M. Christensen et al.

    In crystallo screening for proline analog inhibitors of the proline cycle enzyme PYCR1

    J. Biol. Chem.

    (2020)
  • S. Xiao et al.

    Pyrroline-5-carboxylate reductase 1 (PYCR1) upregulation contributes to gastric cancer progression and indicates poor survival outcome

    Ann. Transl. Med.

    (2020)
  • P. Lauren

    The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma

    Acta Pathol. Microbiol. Scand.

    (1965)
  • Comprehensive molecular characterization of gastric adenocarcinoma

    Nature

    (2014)
  • R. Cristescu et al.

    Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes

    Nat. Med.

    (2015)
  • W. Liu et al.

    Reprogramming of proline and glutamine metabolism contributes to the proliferative and metabolic responses regulated by oncogenic transcription factor c-MYC

    Proc. Natl. Acad. Sci.

    (2012)
  • W. Liu et al.

    Proline biosynthesis augments tumor cell growth and aerobic glycolysis: involvement of pyridine nucleotides

    Sci. Rep.

    (2015)
  • D.Q. Calcagno et al.

    MYC and gastric adenocarcinoma carcinogenesis

    World J. Gastroenterol.

    (2008)
  • T. Matsuoka et al.

    The role of PI3K/Akt/mTOR signaling in gastric carcinoma

    Cancers

    (2014)
  • N. Padmanabhan et al.

    How to stomach an epigenetic insult: the gastric cancer epigenome

    Nat. Rev. Gastroenterol. Hepatol.

    (2017)
  • H.E. Suchiman et al.

    Design, measurement and processing of region-specific DNA methylation assays: the mass spectrometry-based method EpiTYPER

    Front. Genet.

    (2015)
  • E. Cerami et al.

    The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data

    Cancer Discov.

    (2012)
  • J. Gao et al.

    Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal

    Sci. Signal.

    (2013)
  • Z. Lei et al.

    Identification of molecular subtypes of gastric cancer with different responses to PI3-kinase inhibitors and 5-fluorouracil

    Gastroenterology

    (2013)
  • P. Correa

    Human gastric carcinogenesis: a multistep and multifactorial process–first American Cancer Society award lecture on cancer epidemiology and prevention

    Cancer Res.

    (1992)
  • Q. Jin et al.

    Distinct roles of GCN5/PCAF-mediated H3K9ac and CBP/p300-mediated H3K18/27ac in nuclear receptor transactivation

    EMBO J.

    (2011)
  • L. Burke et al.

    The janus-like role of proline metabolism in cancer

    Cell Death Discov.

    (2020)
  • A.N. Bogner et al.

    Structure, biochemistry, and gene expression patterns of the proline biosynthetic enzyme pyrroline-5-carboxylate reductase (PYCR), an emerging cancer therapy target

    Amino Acids

    (2021)
  • S.E. Jackson et al.

    Personalised cancer medicine

    Int. J. Cancer

    (2015)
  • Cited by (0)

    View full text