File Download
  Links for fulltext
     (May Require Subscription)
Supplementary

Article: PFKFB4 Drives the Oncogenicity in TP53-Mutated Hepatocellular Carcinoma in a Phosphatase-Dependent Manner

TitlePFKFB4 Drives the Oncogenicity in TP53-Mutated Hepatocellular Carcinoma in a Phosphatase-Dependent Manner
Authors
KeywordsGlycolysis
Hypoxia
Next-Generation Sequencing
PFKFB
Issue Date1-Apr-2023
PublisherElsevier
Citation
Cellular and Molecular Gastroenterology and Hepatology, 2023, v. 15, n. 6, p. 1325-1350 How to Cite?
Abstract

Background & aims: Metabolic reprogramming is recognized as a cancer hallmark intimately linked to tumor hypoxia, which supports rapid tumor growth and mitigates the consequential oxidative stress. Phosphofructokinase-fructose bisphosphatase (PFKFB) is a family of bidirectional glycolytic enzymes possessing both kinase and phosphatase functions and has emerged as important oncogene in multiple types of cancer. However, its clinical relevance, functional significance, and underlying mechanistic insights in hepatocellular carcinoma (HCC), the primary malignancy that develops in the most important metabolic organ, has never been addressed.

Methods: PFKFB4 expression was examined by RNA sequencing in The Cancer Genome Atlas and our in-house HCC cohort. The up-regulation of PFKFB4 expression was confirmed further by quantitative polymerase chain reaction in an expanded hepatitis B virus-associated HCC cohort followed by clinicopathologic correlation analysis. Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated protein 9 (Cas9)-mediated PFKFB4 knockout cells were generated for functional characterization in vivo, targeted metabolomic profiling, as well as RNA sequencing analysis to comprehensively examine the impact of PFKFB4 loss in HCC.

Results: PFKFB4 expression was up-regulated significantly in HCC and correlated positively with TP53 and TSC2 loss-of-function mutations. In silico transcriptome-based analysis further revealed PFKFB4 functions as a critical hypoxia-inducible gene. Clinically, PFKFB4 up-regulation was associated with more aggressive tumor behavior. Functionally, CRISPR/Cas9-mediated PFKFB4 knockout significantly impaired in vivo HCC development. Targeted metabolomic profiling revealed that PFKFB4 functions as a phosphatase in HCC and its ablation caused an accumulation of metabolites in downstream glycolysis and the pentose phosphate pathway. In addition, PFKFB4 loss induced hypoxia-responsive genes in glycolysis and reactive oxygen species detoxification. Conversely, ectopic PFKFB4 expression conferred sorafenib resistance.

Conclusions: PFKFB4 up-regulation supports HCC development and shows therapeutic implications.


Persistent Identifierhttp://hdl.handle.net/10722/331714
ISSN
2021 Impact Factor: 8.797
2020 SCImago Journal Rankings: 3.417
ISI Accession Number ID

 

DC FieldValueLanguage
dc.contributor.authorKam, Charles Shing-
dc.contributor.authorHo, Daniel Wai-Hung-
dc.contributor.authorMing, Vanessa Sheung-In-
dc.contributor.authorTian, Lu-
dc.contributor.authorSze, Karen Man-Fong-
dc.contributor.authorZhang, Vanilla Xin-
dc.contributor.authorTsui, Yu-Man-
dc.contributor.authorHusain, Abdullah-
dc.contributor.authorLee, Joyce Man-Fong-
dc.contributor.authorWong, Carmen Chak-Lui-
dc.contributor.authorChan, Albert Chi-Yan-
dc.contributor.authorCheung, Tan-To-
dc.contributor.authorChan, Lo-Kong-
dc.contributor.authorNg, Irene Oi-Lin-
dc.date.accessioned2023-09-21T06:58:15Z-
dc.date.available2023-09-21T06:58:15Z-
dc.date.issued2023-04-01-
dc.identifier.citationCellular and Molecular Gastroenterology and Hepatology, 2023, v. 15, n. 6, p. 1325-1350-
dc.identifier.issn2352-345X-
dc.identifier.urihttp://hdl.handle.net/10722/331714-
dc.description.abstract<p><strong>Background & aims: </strong>Metabolic reprogramming is recognized as a cancer hallmark intimately linked to tumor hypoxia, which supports rapid tumor growth and mitigates the consequential oxidative stress. Phosphofructokinase-fructose bisphosphatase (PFKFB) is a family of bidirectional glycolytic enzymes possessing both kinase and phosphatase functions and has emerged as important oncogene in multiple types of cancer. However, its clinical relevance, functional significance, and underlying mechanistic insights in hepatocellular carcinoma (HCC), the primary malignancy that develops in the most important metabolic organ, has never been addressed.</p><p><strong>Methods: </strong>PFKFB4 expression was examined by RNA sequencing in The Cancer Genome Atlas and our in-house HCC cohort. The up-regulation of PFKFB4 expression was confirmed further by quantitative polymerase chain reaction in an expanded hepatitis B virus-associated HCC cohort followed by clinicopathologic correlation analysis. Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated protein 9 (Cas9)-mediated PFKFB4 knockout cells were generated for functional characterization in vivo, targeted metabolomic profiling, as well as RNA sequencing analysis to comprehensively examine the impact of PFKFB4 loss in HCC.</p><p><strong>Results: </strong>PFKFB4 expression was up-regulated significantly in HCC and correlated positively with TP53 and TSC2 loss-of-function mutations. In silico transcriptome-based analysis further revealed PFKFB4 functions as a critical hypoxia-inducible gene. Clinically, PFKFB4 up-regulation was associated with more aggressive tumor behavior. Functionally, CRISPR/Cas9-mediated PFKFB4 knockout significantly impaired in vivo HCC development. Targeted metabolomic profiling revealed that PFKFB4 functions as a phosphatase in HCC and its ablation caused an accumulation of metabolites in downstream glycolysis and the pentose phosphate pathway. In addition, PFKFB4 loss induced hypoxia-responsive genes in glycolysis and reactive oxygen species detoxification. Conversely, ectopic PFKFB4 expression conferred sorafenib resistance.</p><p><strong>Conclusions: </strong>PFKFB4 up-regulation supports HCC development and shows therapeutic implications.</p>-
dc.languageeng-
dc.publisherElsevier-
dc.relation.ispartofCellular and Molecular Gastroenterology and Hepatology-
dc.rightsThis work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.-
dc.subjectGlycolysis-
dc.subjectHypoxia-
dc.subjectNext-Generation Sequencing-
dc.subjectPFKFB-
dc.titlePFKFB4 Drives the Oncogenicity in TP53-Mutated Hepatocellular Carcinoma in a Phosphatase-Dependent Manner-
dc.typeArticle-
dc.description.naturepublished_or_final_version-
dc.identifier.doi10.1016/j.jcmgh.2023.02.004-
dc.identifier.scopuseid_2-s2.0-85152674589-
dc.identifier.volume15-
dc.identifier.issue6-
dc.identifier.spage1325-
dc.identifier.epage1350-
dc.identifier.eissn2352-345X-
dc.identifier.isiWOS:001029188400001-
dc.identifier.issnl2352-345X-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats