1
|
Yeung CLS, Ng TH, Lai CJ, Xue T, Mao X, Tey SK, Lo RCL, Sin C, Ng KM, Wong DKH, Mak L, Yuen M, Ng IO, Cao P, Gao Y, Yun JP, Yam JWP. Small Extracellular Vesicle-Derived Nicotinamide Phosphoribosyltransferase (NAMPT) Induces Acyl-Coenzyme A Synthetase SLC27A4-Mediated Glycolysis to Promote Hepatocellular Carcinoma. J Extracell Vesicles 2025; 14:e70071. [PMID: 40237223 PMCID: PMC12000932 DOI: 10.1002/jev2.70071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Tumour-derived small extracellular vesicles (sEV) are critical mediators within the tumour microenvironment (TME) and are known to regulate various metabolic pathways. In metastatic hepatocellular carcinoma (HCC), mass spectrometry protein analysis of HCC-derived sEV (HCC-sEV) identified an upregulation of nicotinamide phosphoribosyltransferase (NAMPT), a key enzyme in maintaining cellular nicotinamide adenine dinucleotide (NAD+) levels. Our study demonstrates that sEV-NAMPT enhances glycolysis, tumorigenesis, and metastasis in HCC. Specifically, sEV-NAMPT activates the NF-κB transcription factor through toll-like receptor 4 (TLR4), leading to elevated SLC27A4 expression. SLC27A4 functions primarily as a long-chain fatty acid transporter and acyl-CoA synthetase. Lipidomic and metabolomic analyses revealed a positive correlation between SLC27A4 and intracellular levels of triacylglycerol (TG) and dihydroxyacetone phosphate (DHAP). Increased TG levels enhance lipolysis via hepatic lipase and facilitate the conversion of glycerol-3-P to DHAP, an intermediate that bridges lipid metabolism and glycolysis. This study uncovers a novel regulatory axis involving sEV-NAMPT and SLC27A4 in glycolysis, independent of traditional fatty acid metabolism pathways. Clinically, targeting sEV-NAMPT with the inhibitor FK866 significantly inhibited tumour growth in various HCC in vivo models, highlighting the potential of sEV-NAMPT as both a biomarker and therapeutic target in HCC.
Collapse
Affiliation(s)
- Cherlie Lot Sum Yeung
- Department of Pathology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Tung Him Ng
- Department of Pathology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Charlotte Jiaqi Lai
- Department of Pathology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Tingmao Xue
- Department of Pathology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
- Department of Hepatobiliary Surgery IIZhujiang Hospital, Southern Medical UniversityGuangzhouGuangdongChina
| | - Xiaowen Mao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauMacau
| | - Sze Keong Tey
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Regina Cheuk Lam Lo
- Department of Pathology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Chun‐Fung Sin
- Department of Pathology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Kwan Ming Ng
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science ParkHong Kong
| | - Danny Ka Ho Wong
- Department of Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Lung‐Yi Mak
- Department of Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
- State Key Laboratory of Liver ResearchThe University of Hong KongHong Kong
| | - Man‐Fung Yuen
- Department of Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
- State Key Laboratory of Liver ResearchThe University of Hong KongHong Kong
| | - Irene Oi‐Lin Ng
- Department of Pathology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
- State Key Laboratory of Liver ResearchThe University of Hong KongHong Kong
| | - Peihua Cao
- Clinical Research Center, Zhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Yi Gao
- Department of Hepatobiliary Surgery IIZhujiang Hospital, Southern Medical UniversityGuangzhouGuangdongChina
| | - Jing Ping Yun
- Department of PathologySun Yat‐sen University Cancer CenterGuangzhouGuangdongChina
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
- State Key Laboratory of Liver ResearchThe University of Hong KongHong Kong
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU‐SIRIShenzhenChina
| |
Collapse
|
2
|
Bannister ME, Chatterjee DA, Shetty S, Patten DA. The Role of Macrophages in Hepatocellular Carcinoma and Their Therapeutic Potential. Int J Mol Sci 2024; 25:13167. [PMID: 39684877 DOI: 10.3390/ijms252313167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Hepatocellular carcinoma (HCC) represents a significant clinical burden globally and is predicted to continue to increase in incidence for the foreseeable future. The treatment of HCC is complicated by the fact that, in the majority of cases, it develops on a background of advanced chronic inflammatory liver disease. Chronic inflammation can foster an immunosuppressive microenvironment that promotes tumour progression and metastasis. In this setting, macrophages make up a major immune component of the HCC tumour microenvironment, and in this review, we focus on their contribution to HCC development and progression. Tumour-associated macrophages (TAMs) are largely derived from infiltrating monocytes and their potent anti-inflammatory phenotype can be induced by factors that are found within the tumour microenvironment, such as growth factors, cytokines, hypoxia, and extracellular matrix (ECM) proteins. In general, experimental evidence suggest that TAMs can exhibit a variety of functions that aid HCC tumour progression, including the promotion of angiogenesis, resistance to drug therapy, and releasing factors that support tumour cell proliferation and metastasis. Despite their tumour-promoting profile, there is evidence that the underlying plasticity of these cells can be targeted to help reprogramme TAMs to drive tumour-specific immune responses. We discuss the potential for targeting TAMs therapeutically either by altering their phenotype within the HCC microenvironment or by cell therapy approaches by taking advantage of their infiltrative properties from the circulation into tumour tissue.
Collapse
Affiliation(s)
- Megan E Bannister
- Centre for Liver and Gastrointestinal Research, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham B15 2TT, UK
| | - Devnandan A Chatterjee
- Centre for Liver and Gastrointestinal Research, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham B15 2TT, UK
- National Institute for Health Research, Birmingham Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TH, UK
| | - Shishir Shetty
- Centre for Liver and Gastrointestinal Research, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham B15 2TT, UK
- National Institute for Health Research, Birmingham Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TH, UK
| | - Daniel A Patten
- Centre for Liver and Gastrointestinal Research, School of Infection, Inflammation and Immunology, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
3
|
Yang L, Pham K, Xi Y, Jiang S, Robertson KD, Liu C. Acyl-CoA Synthetase Medium-Chain Family Member 5-Mediated Fatty Acid Metabolism Dysregulation Promotes the Progression of Hepatocellular Carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1951-1966. [PMID: 39069168 PMCID: PMC11423759 DOI: 10.1016/j.ajpath.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/23/2024] [Accepted: 07/09/2024] [Indexed: 07/30/2024]
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer, with high incidence and mortality worldwide. Despite diagnostic and therapeutic advancements, HCC remains poorly responsive to treatment, with a poor prognosis. Understanding the molecular mechanisms driving HCC is crucial for developing effective therapies. Emerging evidence indicates that dysregulated fatty acid metabolism contributes to HCC. Acyl-CoA medium-chain synthetase 5 (ACSM5), involved in fatty acid metabolism, is down-regulated in HCC; however, its role is not well understood. This study was used to analyze ACSM5 expression in HCC patient samples and cell lines. The newly established ACSM5-overexpressing HCC cell lines, Huh7-ACSM5 and Hepa1-6-ACSM5, were used to investigate the effects and regulatory mechanisms of ACSM5. The results showed that ACSM5 was significantly down-regulated in HCC tumor tissues compared with non-tumor tissues. ACSM5 expression was regulated by DNA methylation, with a DNA methyltransferase 1 (DNMT1) inhibitor effectively increasing ACSM5 expression and reducing promoter region methylation. Overexpression of ACSM5 in Huh7 cells reduced fatty acid accumulation, decreased cell proliferation, migration, and invasion in vitro, and inhibited tumor growth in mouse xenografts. Furthermore, ACSM5 overexpression also decreased STAT3 phosphorylation, subsequently affecting downstream cytokine TGFB and FGF12 mRNA levels. These findings suggest that ACSM5 down-regulation contributes to HCC progression, providing insights into its oncogenic role and highlighting its potential as a biomarker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Lei Yang
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Kien Pham
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Yibo Xi
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Shaoning Jiang
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Keith D Robertson
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Chen Liu
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, Connecticut.
| |
Collapse
|
4
|
Al-Amodi HS, Kamel HF. Altered Metabolites in Hepatocellular Carcinoma (HCC) Paving the Road for Metabolomics Signature and Biomarkers for Early Diagnosis of HCC. Cureus 2024; 16:e71968. [PMID: 39569240 PMCID: PMC11576499 DOI: 10.7759/cureus.71968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2024] [Indexed: 11/22/2024] Open
Abstract
Globally, hepatocellular carcinoma (HCC) is one of the most commonly encountered cancers. Because the current early diagnostic tests for HCC are not very sensitive, most cases of the disease are discovered late when it is in its terminal stage. Cellular metabolism changes during carcinogenesis to enable cancer cells to adapt to the hypoxic milieu, boost anabolic synthesis, promote survival, and evade apoptotic death signals. Omic techniques represent a breakthrough in the field of diagnostic technology. For example, Metabolomics analysis could be used to identify these metabolite alterations. Understanding the metabolic alterations linked to HCC is crucial for improving high-risk patients' surveillance and understanding the illness's biology. This review highlights the metabolic alterations linked to energy production in cancer cells, as well as the significantly altered metabolites and pathways associated with hepatocarcinogenesis, including acylcarnitines (ACs), amino acids, proteins, lipids, carbohydrates, glucose, and lactate, which reflect the anabolic and catabolic changes occurring in these cells. Additionally, it discusses the clinical implications of recent metabolomics that may serve as potential biomarkers for early diagnosis and monitoring of the progression of HCC.
Collapse
Affiliation(s)
| | - Hala F Kamel
- Biochemistry, Umm Al-Qura University, Makkah, SAU
- Medical Biochemistry and Molecular Biology, Ain Shams University, Cairo, EGY
| |
Collapse
|
5
|
Peng X, He Z, Yuan D, Liu Z, Rong P. Lactic acid: The culprit behind the immunosuppressive microenvironment in hepatocellular carcinoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189164. [PMID: 39096976 DOI: 10.1016/j.bbcan.2024.189164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/05/2024]
Abstract
As a solid tumor with high glycolytic activity, hepatocellular carcinoma (HCC) produces excess lactic acid and increases extracellular acidity, thus forming a unique immunosuppressive microenvironment. L-lactate dehydrogenase (LDH) and monocarboxylate transporters (MCTs) play a very important role in glycolysis. LDH is the key enzyme for lactic acid (LA) production, and MCT is responsible for the cellular import and export of LA. The synergistic effect of the two promotes the formation of an extracellular acidic microenvironment. In the acidic microenvironment of HCC, LA can not only promote the proliferation, survival, transport and angiogenesis of tumor cells but also have a strong impact on immune cells, ultimately leading to an inhibitory immune microenvironment. This article reviews the role of LA in HCC, especially its effect on immune cells, summarizes the progress of LDH and MCT-related drugs, and highlights the potential of immunotherapy targeting lactate combined with HCC.
Collapse
Affiliation(s)
- Xiaopei Peng
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Molecular Imaging Research Center, Central South University, Changsha, Hunan 410013, China
| | - Zhenhu He
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Molecular Imaging Research Center, Central South University, Changsha, Hunan 410013, China
| | - Dandan Yuan
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Molecular Imaging Research Center, Central South University, Changsha, Hunan 410013, China
| | - Zhenguo Liu
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Pengfei Rong
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Molecular Imaging Research Center, Central South University, Changsha, Hunan 410013, China.
| |
Collapse
|
6
|
Li J, Su P, Li T, Hao Y, Wang T, Fu L, Liu X. The Role and Clinical Relevance of Glycolysis-Associated Genes on Immune Infiltration in Hepatocellular Carcinoma. J Cell Biochem 2024; 125:e30620. [PMID: 38923014 DOI: 10.1002/jcb.30620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
Hepatocellular carcinoma (HCC) poses a significant challenge with dismal survival rates, necessitating a deeper understanding of its molecular mechanisms and the development of improved therapies. Metabolic reprogramming, particularly heightened glycolysis, plays a crucial role in HCC progression. Glycolysis-associated genes (GAGs) emerge as key players in HCC pathogenesis, influencing the tumor microenvironment and immune responses. This study aims to investigate the intricate interplay between GAGs and the immune landscape within HCC, offering valuable insights into potential prognostic markers and therapeutic targets to enhance treatment strategies and patient outcomes. Through the exploration of GAGs, we have identified two distinct molecular glycolytic subtypes in HCC patients, each exhibiting significant differences in both the immune microenvironment and prognosis. A risk model comprising five key GAGs was formulated and subsequently evaluated for their predictive accuracy. Our findings underscore the diverse tumor microenvironment and immune responses associated with the varying glycolytic subtypes observed in HCC. The identified key GAGs hold promise as prognostic indicators for evaluating HCC risk levels, predicting patient outcomes, and guiding clinical treatment decisions, particularly in the context of anticipating responses to immunotherapy drugs.
Collapse
Affiliation(s)
- Jing Li
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Peng Su
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Ting Li
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Yang Hao
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Tianjun Wang
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Lei Fu
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Xin Liu
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| |
Collapse
|
7
|
Sim DY, Lee HJ, Ahn CH, Park J, Park SY, Kil BJ, Shim BS, Kim B, Kim SH. Negative Regulation of CPSF6 Suppresses the Warburg Effect and Angiogenesis Leading to Tumor Progression Via c-Myc Signaling Network: Potential Therapeutic Target for Liver Cancer Therapy. Int J Biol Sci 2024; 20:3442-3460. [PMID: 38993554 PMCID: PMC11234225 DOI: 10.7150/ijbs.93462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/08/2024] [Indexed: 07/13/2024] Open
Abstract
In this study, we explored the oncogenic mechanism of cleavage and polyadenylation-specific factor 6 (CPSF6) in hepatocellular carcinoma (HCC). CPSF6 was overexpressed in HCC tissues with poor survival rates compared to normal tissues. Hence, CPSF6 depletion suppressed cell viability and colony formation, induced apoptosis via PARP cleavage, and increased the sub-G1 population of Hep3B and Huh7 cells. In addition, CPSF6 enhanced the stability of c-Myc via their binding through nuclear co-localization by binding to c-Myc at the site of 258-360. Furthermore, c-Myc degradation by CPSF6 depletion was disturbed by FBW7 depletion or treatment with the proteasomal inhibitor MG132. Additionally, CPSF6 depletion suppressed the Warburg effect by inhibiting glucose, HK2, PKM2, LDH, and lactate; showed a synergistic effect with Sorafenib in Hep3B cells; and inhibited angiogenesis by tube formation and CAM assays, along with decreased expression and production of vascular endothelial growth factor (VEGF). Notably, CPSF6 depletion attenuated PD-L1 expression and increased Granzyme B levels, along with an increase in the percentage of CD4/CD8 cells in the splenocytes of BALB/c nude mice bearing Hep3B cells. Consistently, immunohistochemistry showed that CPSF6 depletion reduced the growth of Hep3B cells in BALB/c mice in orthotopic and xenograft tumor models by inhibiting tumor microenvironment-associated proteins. Overall, these findings suggest that CPSF6 enhances the Warburg effect for immune escape and angiogenesis, leading to cancer progression via c-Myc, mediated by the HK, PD-L1, and VEGF networks, with synergistic potential with sorafenib as a molecular target for liver cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Bonglee Kim
- Cancer Molecular Targeted Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447
| | - Sung-Hoon Kim
- Cancer Molecular Targeted Herbal Research Laboratory, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447
| |
Collapse
|
8
|
Lin S, Kuang M. RNA modification-mediated mRNA translation regulation in liver cancer: mechanisms and clinical perspectives. Nat Rev Gastroenterol Hepatol 2024; 21:267-281. [PMID: 38243019 DOI: 10.1038/s41575-023-00884-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/27/2023] [Indexed: 01/21/2024]
Abstract
Malignant liver cancer is characterized by rapid tumour progression and a high mortality rate, whereas the molecular mechanisms underlying liver cancer initiation and progression are still poorly understood. The dynamic and reversible RNA modifications have crucial functions in gene expression regulation by modulating RNA processing and mRNA translation. Emerging evidence has revealed that alterations in RNA modifications facilitate the selective translation of oncogenic transcripts and promote the diverse tumorigenic processes of liver cancer. In this Review, we first highlight the current progress on the functions and mechanisms underlying RNA modifications in the regulation of mRNA translation and then summarize the exciting discoveries on aberrant RNA modification-mediated mRNA translation in the regulation of tumour initiation, metastasis, metabolism, tumour microenvironment, and drug and radiotherapy resistance in liver cancer. Finally, we discuss the diagnostic and therapeutic potentials of targeting RNA modifications and mRNA translation for the clinical management of liver cancer.
Collapse
Affiliation(s)
- Shuibin Lin
- Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Ming Kuang
- Department of Liver Surgery, Center of Hepato-Pancreato-Biliary Surgery, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
9
|
Wang W, Li K, Xiao W. The pharmacological role of Ginsenoside Rg3 in liver diseases: A review on molecular mechanisms. J Ginseng Res 2024; 48:129-139. [PMID: 38465219 PMCID: PMC10920009 DOI: 10.1016/j.jgr.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/23/2023] [Accepted: 11/10/2023] [Indexed: 03/12/2024] Open
Abstract
Liver diseases are a significant global health burden and are among the most common diseases. Ginssennoside Rg3 (Rg3), which is one of the most abundant ginsenosides, has been found to have significant preventive and therapeutic effects against various types of diseases with minimal side effects. Numerous studies have demonstrated the significant preventive and therapeutic effects of Rg3 on various liver diseases such as viral hepatitis, acute liver injury, nonalcoholic liver diseases (NAFLD), liver fibrosis and hepatocellular carcinoma (HCC). The underlying molecular mechanism behind these effects is attributed to apoptosis, autophagy, antioxidant, anti-inflammatory activities, and the regulation of multiple signaling pathways. This review provides a comprehensive description of the potential molecular mechanisms of Rg3 in the development of liver diseases. The article focuses on the regulation of apoptosis, oxidative stress, autophagy, inflammation, and other related factors. Additionally, the review discusses combination therapy and liver targeting strategy, which can accelerate the translation of Rg3 from bench to bedside. Overall, this article serves as a valuable reference for researchers and clinicians alike.
Collapse
Affiliation(s)
- Wenhong Wang
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Ke Li
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Weihua Xiao
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
10
|
Kopsida M, Clavero AL, Khaled J, Balgoma D, Luna-Marco C, Chowdhury A, Nyman SS, Rorsman F, Ebeling Barbier C, Bergsten P, Lennernäs H, Hedeland M, Heindryckx F. Inhibiting the endoplasmic reticulum stress response enhances the effect of doxorubicin by altering the lipid metabolism of liver cancer cells. Mol Metab 2024; 79:101846. [PMID: 38030123 PMCID: PMC10755054 DOI: 10.1016/j.molmet.2023.101846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is characterized by a low and variable response to chemotherapeutic treatments. One contributing factor to the overall pharmacodynamics is the activation of endoplasmic reticulum (ER) stress pathways. This is a cellular stress mechanism that becomes activated when the cell's need for protein synthesis surpasses the ER's capacity to maintain accurate protein folding, and has been implicated in creating drug-resistance in several solid tumors. OBJECTIVE To identify the role of ER-stress and lipid metabolism in mediating drug response in HCC. METHODS By using a chemically-induced mouse model for HCC, we administered the ER-stress inhibitor 4μ8C and/or doxorubicin (DOX) twice weekly for three weeks post-tumor initiation. Histological analyses were performed alongside comprehensive molecular biology and lipidomics assessments of isolated liver samples. In vitro models, including HCC cells, spheroids, and patient-derived liver organoids were subjected to 4μ8C and/or DOX, enabling us to assess their synergistic effects on cellular viability, lipid metabolism, and oxygen consumption rate. RESULTS We reveal a pivotal synergy between ER-stress modulation and drug response in HCC. The inhibition of ER-stress using 4μ8C not only enhances the cytotoxic effect of DOX, but also significantly reduces cellular lipid metabolism. This intricate interplay culminates in the deprivation of energy reserves essential for the sustenance of tumor cells. CONCLUSIONS This study elucidates the interplay between lipid metabolism and ER-stress modulation in enhancing doxorubicin efficacy in HCC. This novel approach not only deepens our understanding of the disease, but also uncovers a promising avenue for therapeutic innovation. The long-term impact of our study could open the possibility of ER-stress inhibitors and/or lipase inhibitors as adjuvant treatments for HCC-patients.
Collapse
Affiliation(s)
- Maria Kopsida
- Department of Medical Cell Biology, Uppsala University, Sweden
| | | | - Jaafar Khaled
- Department of Medical Cell Biology, Uppsala University, Sweden
| | - David Balgoma
- Analytical Pharmaceutical Chemistry, Department of Medicinal Chemistry, Uppsala University, Sweden; Unidad de Excelencia, Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid - Consejo Superior de Investigaciones Científicas (CSIC), Valladolid, Spain
| | | | | | - Sofi Sennefelt Nyman
- Department of Surgical Sciences, Section of Radiology, Uppsala University, Uppsala, Sweden
| | - Fredrik Rorsman
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Peter Bergsten
- Department of Medical Cell Biology, Uppsala University, Sweden
| | - Hans Lennernäs
- Translational Drug Development and Discovery, Department of Pharmaceutical Biosciences, Uppsala University, Sweden
| | - Mikael Hedeland
- Analytical Pharmaceutical Chemistry, Department of Medicinal Chemistry, Uppsala University, Sweden
| | | |
Collapse
|
11
|
Hao W, Chen Z, Tang J, Yang R, Gao WQ, Xu H. hnRNPA2B1 promotes the occurrence and progression of hepatocellular carcinoma by downregulating PCK1 mRNA via a m6A RNA methylation manner. J Transl Med 2023; 21:861. [PMID: 38017546 PMCID: PMC10683354 DOI: 10.1186/s12967-023-04704-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/04/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND N6-methyladenosine (m6A) is the most prevalent RNA modification. Although hnRNPA2B1, as a reader of m6A modification, has been reported to promote tumorigenesis in a few types of tumors, its role in hepatocellular carcinoma (HCC) and the underlying molecular mechanism remains unclear. METHODS Multiple public databases were used to analyze the expression of hnRNPA2B1 in HCC and its correlation with survival prognosis. We employed a CRISPR-Cas9 sgRNA editing strategy to knockout hnRNPA2B1 expression in HCC cells. The biological function of hnRNPA2B1 in vitro in HCC cells was measured by CCK8, colony formation, migration, and invasion assay. The tumorigenic function of hnRNPA2B1 in vivo was determined by a subcutaneous tumor formation experiment and a HCC mouse model via tail injection of several plasmids into the mouse within 5s-7s. RNA binding protein immunoprecipitation (RIP) experiment using hnRNPA2B1 was performed to test the target genes of hnRNPA2B1 and methylated RNA immunoprecipitation (MeRIP) assay was performed to explore the m6A methylated mRNA of target genes. RESULTS hnRNPA2B1 highly expressed in HCC tissues, correlated with high grades and poor prognosis. Its knockout reduced HCC cell proliferation, migration, and invasion in vitro, while overexpression promoted these processes. hnRNPA2B1-knockout cells inhibited tumor formation in graft experiments. In HCC mice, endogenous knockout attenuated hepatocarcinogenesis. RNA-seq showed downregulated gluconeogenesis with high hnRNPA2B1 expression. hnRNPA2B1 negatively correlated with PCK1, a key enzyme. RIP assay revealed hnRNPA2B1 binding to PCK1 mRNA. hnRNPA2B1 knockout increased m6A-methylation of PCK1 mRNA. Interestingly, PCK1 knockout partially counteracted tumor inhibition by hnRNPA2B1 knockout in mice. CONCLUSION Our study indicated that hnRNPA2B1 is highly expressed in HCC and correlated with a poor prognosis. hnRNPA2B1 promotes the tumorigenesis and progression of HCC both in vitro and in vivo. Moreover, hnRNPA2B1 downregulates the expression of PCK1 mRNA via a m6A methylation manner. More importantly, the ability of hnRNPA2B1 to induce tumorigenesis and progression in HCC is dependent on its ability to decrease the expression of PCK1. Therefore, this study suggested that hnRNPA2B1 might be a diagnostic marker of poor prognosis of HCC and a potential therapeutic target for HCC patients.
Collapse
Affiliation(s)
- Weijie Hao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhimin Chen
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jingzhi Tang
- Department of Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ru Yang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Med-X Research Institute and School of Biological Medical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Huiming Xu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
12
|
Zhang YB, Yang G, Bu Y, Lei P, Zhang W, Zhang DY. Development of a machine learning-based model for predicting risk of early postoperative recurrence of hepatocellular carcinoma. World J Gastroenterol 2023; 29:5804-5817. [PMID: 38074914 PMCID: PMC10701309 DOI: 10.3748/wjg.v29.i43.5804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/07/2023] [Accepted: 11/03/2023] [Indexed: 11/20/2023] Open
Abstract
BACKGROUND Surgical resection is the primary treatment for hepatocellular carcinoma (HCC). However, studies indicate that nearly 70% of patients experience HCC recurrence within five years following hepatectomy. The earlier the recurrence, the worse the prognosis. Current studies on postoperative recurrence primarily rely on postoperative pathology and patient clinical data, which are lagging. Hence, developing a new pre-operative prediction model for postoperative recurrence is crucial for guiding individualized treatment of HCC patients and enhancing their prognosis. AIM To identify key variables in pre-operative clinical and imaging data using machine learning algorithms to construct multiple risk prediction models for early postoperative recurrence of HCC. METHODS The demographic and clinical data of 371 HCC patients were collected for this retrospective study. These data were randomly divided into training and test sets at a ratio of 8:2. The training set was analyzed, and key feature variables with predictive value for early HCC recurrence were selected to construct six different machine learning prediction models. Each model was evaluated, and the best-performing model was selected for interpreting the importance of each variable. Finally, an online calculator based on the model was generated for daily clinical practice. RESULTS Following machine learning analysis, eight key feature variables (age, intratumoral arteries, alpha-fetoprotein, pre-operative blood glucose, number of tumors, glucose-to-lymphocyte ratio, liver cirrhosis, and pre-operative platelets) were selected to construct six different prediction models. The XGBoost model outperformed other models, with the area under the receiver operating characteristic curve in the training, validation, and test datasets being 0.993 (95% confidence interval: 0.982-1.000), 0.734 (0.601-0.867), and 0.706 (0.585-0.827), respectively. Calibration curve and decision curve analysis indicated that the XGBoost model also had good predictive performance and clinical application value. CONCLUSION The XGBoost model exhibits superior performance and is a reliable tool for predicting early postoperative HCC recurrence. This model may guide surgical strategies and postoperative individualized medicine.
Collapse
Affiliation(s)
- Yu-Bo Zhang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan 750003, Ningxia Hui Autonomous Region, China
| | - Gang Yang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan 750003, Ningxia Hui Autonomous Region, China
| | - Yang Bu
- Department of Hepatobiliary Surgery, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750003, Ningxia Hui Autonomous Region, China
| | - Peng Lei
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan 750003, Ningxia Hui Autonomous Region, China
| | - Wei Zhang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan 750003, Ningxia Hui Autonomous Region, China
| | - Dan-Yang Zhang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan 750003, Ningxia Hui Autonomous Region, China
| |
Collapse
|
13
|
Zhang Y, Shen L, Wang B, Wu X. Ethanolamine-phosphate phospho-lyase (ETNPPL) contributes to the diagnosis, prognosis, and therapy of hepatocellular carcinoma. PeerJ 2023; 11:e15834. [PMID: 37637156 PMCID: PMC10448887 DOI: 10.7717/peerj.15834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 07/11/2023] [Indexed: 08/29/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is characterized by high mortality, difficulty in early screening, relapse, and poor prognosis. This study aimed to explore the expression of ethanolamine-phosphate phospho-lyase (ETNPPL) and its clinical significance in HCC. Methods Differentially expressed mRNAs were screened using microarray analysis. Functional enrichment was performed using GO (Gene Ontology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) analysis. We used qRT-PCR to measure the expression of ETNPPL in HCC tissues and paired paracarcinoma tissues. A receiver operating characteristic (ROC) curve and Kaplan-Meier curve were conducted to assess the diagnostic and prognostic values. Cell behaviors were evaluated using a scratch test and transwell assay. Results The results showed that numerous mRNAs are abnormally expressed in HCC. ETNPPL was decreased in HCC tissues and cells. The area under curve (AUC) of ETNPPL was 0.9089, demonstrating that ETNPPL had diagnostic value. Low expression of ETNPPL was related to poor prognosis for patients with HCC. Moreover, the over-expression of ETNPPL inhibited HCC cell migration and invasion. Conclusions In conclusion, downregulated ETNPPL was found in HCC and is related to poor patient prognosis and the promotion of cell metastasis. This suggests that ETNPPL serves both as a promising diagnosis and prognosis biomarker, and a therapy target of HCC.
Collapse
Affiliation(s)
- Yun Zhang
- Department of General Surgery, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, China
| | - Li Shen
- Disinfection Supply Center, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, China
| | - Bojun Wang
- Department of General Surgery, Yixing Fourth People’s Hospital, Yixing, Jiangsu, China
| | - Xiaohong Wu
- Department of General Surgery, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu, China
| |
Collapse
|
14
|
Peñuelas‐Haro I, Espinosa‐Sotelo R, Crosas‐Molist E, Herranz‐Itúrbide M, Caballero‐Díaz D, Alay A, Solé X, Ramos E, Serrano T, Martínez‐Chantar ML, Knaus UG, Cuezva JM, Zorzano A, Bertran E, Fabregat I. The NADPH oxidase NOX4 regulates redox and metabolic homeostasis preventing HCC progression. Hepatology 2023; 78:416-433. [PMID: 35920301 PMCID: PMC10344438 DOI: 10.1002/hep.32702] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS The NADPH oxidase NOX4 plays a tumor-suppressor function in HCC. Silencing NOX4 confers higher proliferative and migratory capacity to HCC cells and increases their in vivo tumorigenic potential in xenografts in mice. NOX4 gene deletions are frequent in HCC, correlating with higher tumor grade and worse recurrence-free and overall survival rates. However, despite the accumulating evidence of a protective regulatory role in HCC, the cellular processes governed by NOX4 are not yet understood. Accordingly, the aim of this work was to better understand the molecular mechanisms regulated by NOX4 in HCC in order to explain its tumor-suppressor action. APPROACH AND RESULTS Experimental models: cell-based loss or gain of NOX4 function experiments, in vivo hepatocarcinogenesis induced by diethylnitrosamine in Nox4 -deficient mice, and analyses in human HCC samples. Methods include cellular and molecular biology analyses, proteomics, transcriptomics, and metabolomics, as well as histological and immunohistochemical analyses in tissues. Results identified MYC as being negatively regulated by NOX4. MYC mediated mitochondrial dynamics and a transcriptional program leading to increased oxidative metabolism, enhanced use of both glucose and fatty acids, and an overall higher energetic capacity and ATP level. NOX4 deletion induced a redox imbalance that augmented nuclear factor erythroid 2-related factor 2 (Nrf2) activity and was responsible for MYC up-regulation. CONCLUSIONS Loss of NOX4 in HCC tumor cells induces metabolic reprogramming in a Nrf2/MYC-dependent manner to promote HCC progression.
Collapse
Affiliation(s)
- Irene Peñuelas‐Haro
- TGF‐β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, Barcelona, Spain
- CIBEREHD, ISCIII, Madrid, Spain
| | - Rut Espinosa‐Sotelo
- TGF‐β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, Barcelona, Spain
- CIBEREHD, ISCIII, Madrid, Spain
| | - Eva Crosas‐Molist
- TGF‐β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Macarena Herranz‐Itúrbide
- TGF‐β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, Barcelona, Spain
- CIBEREHD, ISCIII, Madrid, Spain
| | - Daniel Caballero‐Díaz
- TGF‐β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, Barcelona, Spain
- CIBEREHD, ISCIII, Madrid, Spain
| | - Ania Alay
- Unit of Bioinformatics for Precision Oncology, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Barcelona, Spain
- Preclinical and Experimental Research in Thoracic Tumors, Oncobell Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Xavier Solé
- Unit of Bioinformatics for Precision Oncology, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Barcelona, Spain
- Preclinical and Experimental Research in Thoracic Tumors, Oncobell Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- Molecular Biology CORE, Center for Biomedical Diagnostics, Hospital Clínic of Barcelona, Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain
| | - Emilio Ramos
- CIBEREHD, ISCIII, Madrid, Spain
- Department of Surgery, Liver Transplant Unit, University Hospital of Bellvitge, Barcelona, Spain
- Faculty of Medicine and Health Sciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Teresa Serrano
- CIBEREHD, ISCIII, Madrid, Spain
- Faculty of Medicine and Health Sciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Pathological Anatomy Service, University Hospital of Bellvitge, Barcelona, Spain
| | - María L. Martínez‐Chantar
- CIBEREHD, ISCIII, Madrid, Spain
- Liver Disease Lab, Center for Cooperative Research in Biosciences, Basque Research and Technology Alliance, Bizkaia Technology Park, Spain
| | - Ulla G. Knaus
- Conway Institute, University College Dublin, Dublin, Ireland
| | - José M. Cuezva
- Center for Molecular Biology “Severo Ochoa,” Autonoma University of Madrid, Madrid, Spain
- CIBERER, ISCIII, Madrid, Spain
| | - Antonio Zorzano
- Biochemistry and Molecular Biomedicine Department, University of Barcelona, Barcelona, Spain
- Institute of Research in Biomedicine, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- CIBERDEM, ISCIII, Madrid, Spain
| | - Esther Bertran
- TGF‐β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, Barcelona, Spain
- CIBEREHD, ISCIII, Madrid, Spain
| | - Isabel Fabregat
- TGF‐β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, Barcelona, Spain
- CIBEREHD, ISCIII, Madrid, Spain
- Physiological Sciences Department, University of Barcelona, Barcelona, Spain
| |
Collapse
|
15
|
Jeon AJ, Anene-Nzelu CG, Teo YY, Chong SL, Sekar K, Wu L, Chew SC, Chen J, Kendarsari RI, Lai H, Ling WH, Kaya NA, Lim JQ, Chung AYF, Cheow PC, Kam JH, Madhavan K, Kow A, Ganpathi IS, Lim TKH, Leow WQ, Loong S, Loh TJ, Wan WK, Soon GST, Pang YH, Yoong BK, Bee-Lan Ong D, Lim J, de Villa VH, dela Cruz RD, Chanwat R, Thammasiri J, Bonney GK, Goh BK, Foo RSY, Chow PKH. A genomic enhancer signature associates with hepatocellular carcinoma prognosis. JHEP Rep 2023; 5:100715. [PMID: 37168287 PMCID: PMC10165154 DOI: 10.1016/j.jhepr.2023.100715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/20/2023] [Accepted: 02/09/2023] [Indexed: 05/13/2023] Open
Abstract
Background & Aims Lifestyle and environmental-related exposures are important risk factors for hepatocellular carcinoma (HCC), suggesting that epigenetic dysregulation significantly underpins HCC. We profiled 30 surgically resected tumours and the matched adjacent normal tissues to understand the aberrant epigenetic events associated with HCC. Methods We identified tumour differential enhancers and the associated genes by analysing H3K27 acetylation (H3K27ac) chromatin immunoprecipitation sequencing (ChIP-seq) and Hi-C/HiChIP data from the resected tumour samples of 30 patients with early-stage HCC. This epigenome dataset was analysed with previously reported genome and transcriptome data of the overlapping group of patients from the same cohort. We performed patient-specific differential expression testing using multiregion sequencing data to identify genes that undergo both enhancer and gene expression changes. Based on the genes selected, we identified two patient groups and performed a recurrence-free survival analysis. Results We observed large-scale changes in the enhancer distribution between HCC tumours and the adjacent normal samples. Many of the gain-in-tumour enhancers showed corresponding upregulation of the associated genes and vice versa, but much of the enhancer and gene expression changes were patient-specific. A subset of the upregulated genes was activated in a subgroup of patients' tumours. Recurrence-free survival analysis revealed that the patients with a more robust upregulation of those genes showed a worse prognosis. Conclusions We report the genomic enhancer signature associated with differential prognosis in HCC. Findings that cohere with oncofoetal reprogramming in HCC were underpinned by genome-wide enhancer rewiring. Our results present the epigenetic changes in HCC that offer the rational selection of epigenetic-driven gene targets for therapeutic intervention or disease prognostication in HCC. Impact and Implications Lifestyle and environmental-related exposures are the important risk factors of hepatocellular carcinoma (HCC), suggesting that tumour-associated epigenetic dysregulations may significantly underpin HCC. We profiled tumour tissues and their matched normal from 30 patients with early-stage HCC to study the dysregulated epigenetic changes associated with HCC. By also analysing the patients' RNA-seq and clinical data, we found the signature genes - with epigenetic and transcriptomic dysregulation - associated with worse prognosis. Our findings suggest that systemic approaches are needed to consider the surrounding cellular environmental and epigenetic changes in HCC tumours.
Collapse
Affiliation(s)
- Ah-Jung Jeon
- Program in Clinical and Translational Liver Cancer Research, Division of Medical Science, National Cancer Center Singapore, Singapore
| | - Chukwuemeka George Anene-Nzelu
- Cardiovascular Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Montreal Heart Institute, Montreal, Quebec, Canada
- Department of Medicine, University of Montreal, Quebec, Canada
| | - Yue-Yang Teo
- Program in Clinical and Translational Liver Cancer Research, Division of Medical Science, National Cancer Center Singapore, Singapore
| | - Shay Lee Chong
- Program in Clinical and Translational Liver Cancer Research, Division of Medical Science, National Cancer Center Singapore, Singapore
| | - Karthik Sekar
- Program in Clinical and Translational Liver Cancer Research, Division of Medical Science, National Cancer Center Singapore, Singapore
| | - Lingyan Wu
- Program in Clinical and Translational Liver Cancer Research, Division of Medical Science, National Cancer Center Singapore, Singapore
| | - Sin-Chi Chew
- Program in Clinical and Translational Liver Cancer Research, Division of Medical Science, National Cancer Center Singapore, Singapore
| | - Jianbin Chen
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Raden Indah Kendarsari
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Hannah Lai
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Wen Huan Ling
- Program in Clinical and Translational Liver Cancer Research, Division of Medical Science, National Cancer Center Singapore, Singapore
| | - Neslihan Arife Kaya
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Jia Qi Lim
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Alexander Yaw Fui Chung
- Department of Hepatopancreatobiliary and Transplant Surgery, National Cancer Centre Singapore and Singapore General Hospital, Singapore
- Academic Clinical Programme for Surgery, Duke-NUS Medical School, Singapore
| | - Peng-Chung Cheow
- Department of Hepatopancreatobiliary and Transplant Surgery, National Cancer Centre Singapore and Singapore General Hospital, Singapore
- Academic Clinical Programme for Surgery, Duke-NUS Medical School, Singapore
| | - Juinn Huar Kam
- Department of Hepatopancreatobiliary and Transplant Surgery, National Cancer Centre Singapore and Singapore General Hospital, Singapore
- Academic Clinical Programme for Surgery, Duke-NUS Medical School, Singapore
| | - Krishnakumar Madhavan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University Surgical Cluster, National University Health System, Singapore
| | - Alfred Kow
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University Surgical Cluster, National University Health System, Singapore
| | - Iyer Shridhar Ganpathi
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University Surgical Cluster, National University Health System, Singapore
| | - Tony Kiat Hon Lim
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Wei-Qiang Leow
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Shihleone Loong
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Tracy Jiezhen Loh
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Wei Keat Wan
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | | | - Yin Huei Pang
- Department of Pathology, National University Hospital, Singapore
| | - Boon Koon Yoong
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Diana Bee-Lan Ong
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Jasmine Lim
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Vanessa H. de Villa
- Department of Surgery and Center for Liver Health and Transplantation, The Medical City, Pasig City, Philippines
| | - Rouchelle D. dela Cruz
- Department of Laboratory Medicine and Pathology, The Medical City, Pasig City, Philippines
| | - Rawisak Chanwat
- Hepato-Pancreato-Biliary Surgery Unit, Department of Surgery, National Cancer Institute, Bangkok, Thailand
| | | | - Glenn K. Bonney
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University Surgical Cluster, National University Health System, Singapore
| | - Brian K.P. Goh
- Department of Hepatopancreatobiliary and Transplant Surgery, National Cancer Centre Singapore and Singapore General Hospital, Singapore
- Academic Clinical Programme for Surgery, Duke-NUS Medical School, Singapore
| | - Roger Sik Yin Foo
- Cardiovascular Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Pierce Kah-Hoe Chow
- Program in Clinical and Translational Liver Cancer Research, Division of Medical Science, National Cancer Center Singapore, Singapore
- Department of Hepatopancreatobiliary and Transplant Surgery, National Cancer Centre Singapore and Singapore General Hospital, Singapore
- Academic Clinical Programme for Surgery, Duke-NUS Medical School, Singapore
- Corresponding author. Address: National Cancer Centre Singapore, 30 Hospital Boulevard, Singapore 168583, Singapore. Tel.: +65-63065424.
| |
Collapse
|
16
|
Wei D, Chen D, Mou H, Chakraborty S, Wei B, Tan L, Lorenzi PL, Qian X, Yang P. Targeting Glutamine Metabolism with a Novel Na+/K+-ATPase Inhibitor RX108 in Hepatocellular Carcinoma. Mol Cancer Ther 2023; 22:693-705. [PMID: 36780187 PMCID: PMC11817653 DOI: 10.1158/1535-7163.mct-22-0490] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/18/2022] [Accepted: 02/08/2023] [Indexed: 02/14/2023]
Abstract
The poor prognosis and limited therapeutic options for human hepatocellular carcinoma (HCC), the most common form of liver cancer, highlight the urgent need to identify novel therapeutic modalities. Here, we describe the antitumor activity and underlying molecular mechanisms of a novel Na+/K+-ATPase inhibitor RX108 in human HCC cells and its xenograft model. RX108 dose-dependently inhibited HCC cell proliferation in vitro and tumor growth in a xenograft mouse model, and that the inhibition was associated with induction of apoptosis. Mechanistically, RX108 significantly downregulated alanine serine cysteine transporter 2 (ASCT2) protein expression and reduced glutamine and glutamate concentration in HCC cells and tumors. In addition, RX108 exposure led to a significant decrease in cell energy metabolism in Huh7 and Hep3B cells, including decreased levels of glutathione, NADH, NADPH, and mitochondrial respiration oxygen consumption rate. Furthermore, HCC cells exhibited evidence of glutamine addiction; the antiproliferative effect of RX108 was dependent on glutamine transport. Clinically, elevated ASCT2 mRNA expression in HCCs was associated with unfavorable survival. Taken together, these findings reveal a novel approach to target glutamine metabolism through inhibiting Na+/K+-ATPase and provide a rationale for using RX108 to treat HCC in patients whose tumors express ASCT2 at high levels. RX108 is currently under clinical development.
Collapse
Affiliation(s)
- Daoyan Wei
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Authors contributed equally to the manuscript
| | - Dongmei Chen
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Authors contributed equally to the manuscript
| | | | - Sharmistha Chakraborty
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bo Wei
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, Houston, TX 77030, USA
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, Houston, TX 77030, USA
| | - Philip L. Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, Houston, TX 77030, USA
| | | | - Peiying Yang
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
17
|
Jiang L, Xu F, Li C, Liu T, Zhao Q, Liu Y, Zhao Y, Li Y, Zhang Z, Tang X, Zhang J. Sulfotransferase 1C2 promotes hepatocellular carcinoma progression by enhancing glycolysis and fatty acid metabolism. Cancer Med 2023; 12:10738-10754. [PMID: 36880364 PMCID: PMC10225225 DOI: 10.1002/cam4.5759] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 11/16/2022] [Accepted: 12/01/2022] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is aggressive liver cancer. Despite advanced imaging and other diagnostic measures, HCC in a significant portion of patients had reached the advanced stage at the first diagnosis. Unfortunately, there is no cure for advanced HCC. As a result, HCC is still a leading cause of cancer death, and there is a pressing need for new diagnostic markers and therapeutic targets. METHODS We investigated sulfotransferase 1C2 (SUTL1C2), which we recently showed was overexpressed in human HCC cancerous tissues. Specifically, we analyzed the effects of SULT1C2 knockdown on the growth, survival, migration, and invasiveness of two HCC cell lines, i.e., HepG2 and Huh7 cells. We also studied the transcriptomes and metabolomes in the two HCC cell lines before and after SULT1C2 knockdown. Based on the transcriptome and metabolome data, we further investigated the SULT1C2 knockdown-mediated shared changes, i.e., glycolysis and fatty acid metabolism, in the two HCC cell lines. Finally, we performed rescue experiments to determine whether the inhibitory effects of SULT1C2 knockdown could be rescued via overexpression. RESULTS We showed that SULT1C2 overexpression promoted the growth, survival, migration, and invasiveness of HCC cells. In addition, SULT1C2 knockdown resulted in a wide range of gene expression and metabolome changes in HCC cells. Moreover, analysis of shared alterations showed that SULT1C2 knockdown significantly suppressed glycolysis and fatty acid metabolism, which could be rescued via SULT1C2 overexpression. CONCLUSIONS Our data suggest that SULT1C2 is a potential diagnostic marker and therapeutic target for human HCC.
Collapse
Affiliation(s)
- Liya Jiang
- Henan Institute of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouHenanChina
- School of Life SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Fang Xu
- Henan Institute of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Chenglong Li
- School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Ting Liu
- BGI College & Henan Institute of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Qianwei Zhao
- Henan Institute of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & TreatmentZhengzhou UniversityZhengzhouHenanChina
| | - Yixian Liu
- Henan Institute of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Ying Zhao
- Henan Institute of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Yamei Li
- School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Zhendong Zhang
- BGI College & Henan Institute of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Xiaolei Tang
- Department of Veterinary Biomedical Sciences, College of Veterinary MedicineLong Island UniversityBrookvilleNew YorkUSA
- Division of Regenerative Medicine, Department of Medicine, Department of Basic Science, School of MedicineLoma Linda UniversityLoma LindaCaliforniaUSA
| | - Jintao Zhang
- Henan Institute of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & TreatmentZhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
18
|
Yang J, Zeng L, Chen R, Zheng S, Zhou Y, Chen R. Characterization of heterogeneous metabolism in hepatocellular carcinoma identifies new therapeutic target and treatment strategy. Front Immunol 2023; 14:1076587. [PMID: 37006288 PMCID: PMC10060979 DOI: 10.3389/fimmu.2023.1076587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/08/2023] [Indexed: 03/18/2023] Open
Abstract
BackgroundMetabolic reprogramming is a well-known hallmark of cancer. Systematical identification of clinically relevant metabolic subtypes of Hepatocellular carcinoma (HCC) is critical to understand tumor heterogeneity and develop efficient treatment strategies.MethodsWe performed an integrative analysis of genomic, transcriptomic, and clinical data from an HCC patient cohort in The Cancer Genome Atlas (TCGA).ResultsFour metabolic subtypes were defined: mHCC1, mHHC2, mHCC3, and mHCC4. These subtypes had distinct differences in mutations profiles, activities of metabolic pathways, prognostic metabolism genes, and immune features. The mHCC1 was associated with poorest outcome and was characterized by extensive metabolic alterations, abundant immune infiltration, and increased expression of immunosuppressive checkpoints. The mHHC2 displayed lowest metabolic alteration level and was associated with most significant improvement in overall survival in response to high CD8+ T cell infiltration. The mHHC3 was a “cold-tumor” with low immune infiltration and few metabolic alterations. The mHCC4 presented a medium degree of metabolic alteration and high CTNNB1 mutation rate. Based on our HCC classification and in vitro study, we identified palmitoyl-protein thioesterase 1 (PPT1) was a specific prognostic gene and therapeutic target for mHCC1.ConclusionOur study highlighted mechanistic differences among metabolic subtypes and identified potential therapeutic targets for subtype-specific treatment strategies targeting unique metabolic vulnerabilities. The immune heterogeneities across metabolic subtypes may help further clarify the association between metabolism and immune environment and guide the development of novel strategies through targeting both unique metabolic vulnerabilities and immunosuppressive triggers.
Collapse
Affiliation(s)
- Jiabin Yang
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Pancreatic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Liangtang Zeng
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Pancreatic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ruiwan Chen
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shangyou Zheng
- Department of Pancreatic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yu Zhou
- Department of Pancreatic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- *Correspondence: Rufu Chen, ; Yu Zhou,
| | - Rufu Chen
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Pancreatic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- *Correspondence: Rufu Chen, ; Yu Zhou,
| |
Collapse
|
19
|
Gao Y, Gong Y, Liu Y, Xue Y, Zheng K, Guo Y, Hao L, Peng Q, Shi X. Integrated analysis of transcriptomics and metabolomics in human hepatocellular carcinoma HepG2215 cells after YAP1 knockdown. Acta Histochem 2023; 125:151987. [PMID: 36473310 DOI: 10.1016/j.acthis.2022.151987] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 11/29/2022] [Accepted: 11/29/2022] [Indexed: 12/07/2022]
Abstract
Yes-associated protein 1 (YAP1) plays a critical role in hepatocellular carcinoma (HCC). Inhibition of YAP1 expression suppresses HCC progression, but the underlying mechanism is still unclear. In this study, we studied the effects and molecular mechanisms of YAP1 knockdown on the growth and metabolism in human HCC HepG2215 cells. Inhibition of YAP1 expression inhibits the proliferation and metastasis in HepG2215 cells, and differentially expressed genes (DEGs) and metabolites were identified in shYAP1-HepG2215 cells. Further, 805 DEGs, mainly associated with metabolism and particularly lipid metabolism, were identified by transcriptome sequencing analyses in shYAP1-HepG2215 cells. YAP1 knockdown increased albumin (ALB) levels by Protein-protein interaction (PPI) network analyses in HepG2215 cells. Metabolomic profiling identified 37 metabolites with significant differences in the shYAP1 group, and amino acid metabolism generally decreased in the shYAP1 group. Comprehensive analysis of transcriptomics and metabolomics revealed that the ATP-binding cassette (ABC) transporters play a central role after YAP1 knockdown in HepG2215 cells. Therefore, YAP1 knockdown inhibited HCC growth, which affected the metabolism of lipids and amino acids by regulating the expression of ALB and ABC transporters in HepG2215 cells.
Collapse
Affiliation(s)
- Yuting Gao
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; School of Basic Medical Sciences, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Yi Gong
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yiwei Liu
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yu Xue
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Kangning Zheng
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yinglin Guo
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Liyuan Hao
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Qing Peng
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Xinli Shi
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China.
| |
Collapse
|
20
|
Li S, Yin S, Ding H, Shao Y, Zhou S, Pu W, Han L, Wang T, Yu H. Polyphenols as potential metabolism mechanisms regulators in liver protection and liver cancer prevention. Cell Prolif 2023; 56:e13346. [PMID: 36229407 DOI: 10.1111/cpr.13346] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/19/2022] [Accepted: 09/29/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Liver cancer is one of the common malignancies. The dysregulation of metabolism is a driver of accelerated tumourigenesis. Metabolic changes are well documented to maintain tumour growth, proliferation and survival. Recently, a variety of polyphenols have been shown to have a crucial role both in liver disease prevention and metabolism regulation. METHODS We conducted a literature search and combined recent data with systematic analysis to comprehensively describe the molecular mechanisms that link polyphenols to metabolic regulation and their contribution in liver protection and liver cancer prevention. RESULTS Targeting metabolic dysregulation in organisms prevents and resists the development of liver cancer, which has important implications for identifying new therapeutic strategies for the management and treatment of cancer. Polyphenols are a class of complex compounds composed of multiple phenolic hydroxyl groups and are the main active ingredients of many natural plants. They mediate a broad spectrum of biological and pharmacological functions containing complex lipid metabolism, glucose metabolism, iron metabolism, intestinal flora imbalance, as well as the direct interaction of their metabolites with key cell-signalling proteins. A large number of studies have found that polyphenols affect the metabolism of organisms by interfering with a variety of intracellular signals, thereby protecting the liver and reducing the risk of liver cancer. CONCLUSION This review systematically illustrates that various polyphenols, including resveratrol, chlorogenic acid, caffeic acid, dihydromyricetin, quercetin, catechins, curcumin, etc., improve metabolic disorders through direct or indirect pathways to protect the liver and fight liver cancer.
Collapse
Affiliation(s)
- Shuangfeng Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Shuangshuang Yin
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Hui Ding
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingying Shao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Shiyue Zhou
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Weiling Pu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Lifeng Han
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Tao Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Haiyang Yu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| |
Collapse
|
21
|
Zhang X, Liu X, Zhu K, Zhang X, Li N, Sun T, Fan S, Dai L, Zhang J. CD5L-associated gene analyses highlight the dysregulations, prognostic effects, immune associations, and drug-sensitivity predicative potentials of LCAT and CDC20 in hepatocellular carcinoma. Cancer Cell Int 2022; 22:393. [PMID: 36494696 PMCID: PMC9733014 DOI: 10.1186/s12935-022-02820-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The dysregulation of CD5L has been reported in hepatocellular carcinoma (HCC). However, its functions in HCC were controversial. In this study, we aimed to identify CD5L-associated pathways and markers and explore their values in HCC diagnosis, prognosis and treatment. METHODS HCC datasets with gene expression profiles and clinical data in TCGA and ICGC were downloaded. The immune/stroma cell infiltrations were estimated with xCell. CD5L-associated pathways and CD5L-associated genes (CD5L-AGs) were identified with gene expression comparisons and gene set enrichment analysis (GSEA). Cox regression, Kaplan-Meier survival analysis, and least absolute shrinkage and selection operator (LASSO) regression analysis were performed. The correlations of the key genes with immune/stroma infiltrations, immunoregulators, and anti-cancer drug sensitivities in HCC were investigated. At protein level, the key genes dysregulations, their correlations and prognostic values were validated in clinical proteomic tumor analysis consortium (CPTAC) database. Serum CD5L and LCAT activity in 50 HCC and 30 normal samples were evaluated and compared. The correlations of serum LCAT activity with alpha-fetoprotein (AFP), albumin (ALB) and high-density lipoprotein (HDL) in HCC were also investigated. RESULTS Through systemic analyses, 14 CD5L-associated biological pathways, 256 CD5L-AGs and 28 CD5L-associated prognostic and diagnostic genes (CD5L-APDGs) were identified. A risk model consisting of LCAT and CDC20 was constructed for HCC overall survival (OS), which could discriminate HCC OS status effectively in both the training and the validation sets. CD5L, LCAT and CDC20 were shown to be significantly correlated with immune/stroma cell infiltrations, immunoregulators and 31 anti-cancer drug sensitivities in HCC. At protein level, the dysregulations of CD5L, LCAT and CDC20 were confirmed. LCAT and CDC20 were shown to be significantly correlated with proliferation marker MKI67. In serum, no significance of CD5L was shown. However, the lower activity of LCAT in HCC serum was obvious, as well as its significant positive correlations ALB and HDL concentrations. CONCLUSIONS CD5L, LCAT and CDC20 were dysregulated in HCC both at mRNA and protein levels. The LCAT-CDC20 signature might be new predicator for HCC OS. The associations of the three genes with HCC microenvironment and anti-cancer drug sensitivities would provide new clues for HCC immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Xiuzhi Zhang
- Department of Pathology, Henan Medical College, Zhengzhou, China
| | - Xiaoli Liu
- grid.414011.10000 0004 1808 090XLaboratory Department, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Keke Zhu
- Department of Pathology, Henan Medical College, Zhengzhou, China
| | - Xue Zhang
- grid.207374.50000 0001 2189 3846Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ningning Li
- Department of Pathology, Henan Medical College, Zhengzhou, China
| | - Tao Sun
- Department of Pathology, Henan Medical College, Zhengzhou, China
| | - Shasha Fan
- grid.477407.70000 0004 1806 9292Oncology Department, The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People’s Hospital, Changsha, China ,grid.411427.50000 0001 0089 3695Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University, Changsha, China
| | - Liping Dai
- grid.207374.50000 0001 2189 3846Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jinzhong Zhang
- Department of Pathology, Henan Medical College, Zhengzhou, China
| |
Collapse
|
22
|
Jamialahmadi O, Salehabadi E, Hashemi-Najafabadi S, Motamedian E, Bagheri F, Mancina RM, Romeo S. Cellular Genome-Scale Metabolic Modeling Identifies New Potential Drug Targets Against Hepatocellular Carcinoma. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2022; 26:671-682. [PMID: 36508280 DOI: 10.1089/omi.2022.0122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Genome-scale metabolic modeling (GEM) is one of the key approaches to unpack cancer metabolism and for discovery of new drug targets. In this study, we report the Transcriptional Regulated Flux Balance Analysis-CORE (TRFBA-), an algorithm for GEM using key growth-correlated reactions using hepatocellular carcinoma (HCC), an important global health burden, as a case study. We generated a HepG2 cell-specific GEM by integrating this cell line transcriptomic data with a generic human metabolic model to forecast potential drug targets for HCC. A total of 108 essential genes for growth were predicted by the TRFBA-CORE. These genes were enriched for metabolic pathways involved in cholesterol, sterol, and steroid biosynthesis. Furthermore, we silenced a predicted essential gene, 11-beta dehydrogenase hydroxysteroid type 2 (HSD11B2), in HepG2 cells resulting in a reduction in cell viability. To further identify novel potential drug targets in HCC, we examined the effect of nine drugs targeting the essential genes, and observed that most drugs inhibited the growth of HepG2 cells. Some of these drugs in this model performed better than Sorafenib, the first-line therapeutic against HCC. A HepG2 cell-specific GEM highlights sterol metabolism to be essential for cell growth. HSD11B2 downregulation results in lower cell growth. Most of the compounds, selected by drug repurposing approach, show a significant inhibitory effect on cell growth in a wide range of concentrations. These findings offer new molecular leads for drug discovery for hepatic cancer while also illustrating the importance of GEM and drug repurposing in cancer therapeutics innovation.
Collapse
Affiliation(s)
- Oveis Jamialahmadi
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Biotechnology and Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Ehsan Salehabadi
- Department of Biotechnology and Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Sameereh Hashemi-Najafabadi
- Department of Biomedical Engineering, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Ehsan Motamedian
- Department of Biotechnology and Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Bagheri
- Department of Biotechnology and Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Rosellina Margherita Mancina
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Stefano Romeo
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.,Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy.,Cardiology Department, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
23
|
Morine Y, Utsunomiya T, Yamanaka-Okumura H, Saito Y, Yamada S, Ikemoto T, Imura S, Kinoshita S, Hirayama A, Tanaka Y, Shimada M. Essential amino acids as diagnostic biomarkers of hepatocellular carcinoma based on metabolic analysis. Oncotarget 2022; 13:1286-1298. [PMID: 36441784 PMCID: PMC11623405 DOI: 10.18632/oncotarget.28306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 10/20/2022] [Indexed: 11/23/2022] Open
Abstract
Metabolomics, defined as the comprehensive identification of all small metabolites in a biological sample, has the power to shed light on phenotypic changes associated with various diseases, including cancer. To discover potential metabolomic biomarkers of hepatocellular carcinoma (HCC), we investigated the metabolomes of tumor and non-tumor tissue in 20 patients with primary HCC using capillary electrophoresis-time-of-flight mass spectrometry. We also analyzed blood samples taken immediately before and 14 days after hepatectomy to identify associated changes in the serum metabolome. Marked changes were detected in the different quantity of 61 metabolites that could discriminate between HCC tumor and paired non-tumor tissue and additionally between HCC primary tumors and colorectal liver metastases. Among the 30 metabolites significantly upregulated in HCC tumors compared with non-tumor tissues, 10 were amino acids, and 7 were essential amino acids (leucine, valine, tryptophan, isoleucine, methionine, lysine, and phenylalanine). Similarly, the serum metabolomes of HCC patients before hepatectomy revealed a significant increase in 16 metabolites, including leucine, valine, and tryptophan. Our results reveal striking differences in the metabolomes of HCC tumor tissue compared with non-tumor tissue, and identify the essential amino acids leucine, valine, and tryptophan as potential metabolic biomarkers for HCC.
Collapse
Affiliation(s)
- Yuji Morine
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Tohru Utsunomiya
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Hisami Yamanaka-Okumura
- Department of Clinical Nutrition and Food Management, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Yu Saito
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Shinichiro Yamada
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Tetsuya Ikemoto
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Satoru Imura
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| | - Shohei Kinoshita
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
- Systems Biology Program, Graduate School of Media and Governance, Keio University, Fujisawa, Kanagawa 252-0882, Japan
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
- Systems Biology Program, Graduate School of Media and Governance, Keio University, Fujisawa, Kanagawa 252-0882, Japan
| | - Yasuhito Tanaka
- Department of Gastroenterology and Hepatology, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Mitsuo Shimada
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| |
Collapse
|
24
|
Wang Y, Guan T, Zhou G, Zhao H, Gao J. SOJNMF: Identifying Multidimensional Molecular Regulatory Modules by Sparse Orthogonality-Regularized Joint Non-Negative Matrix Factorization Algorithm. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:3695-3703. [PMID: 34546925 DOI: 10.1109/tcbb.2021.3114146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Cancer is not only a very aggressive but also a very diverse disease. Recent advances in high-throughput omics technologies of cancer have enabled biomedical researchers to have more opportunities for studying its multi-level biological regulatory mechanism. However, there are few methods to explore the underlying mechanism of cancer by identifying its multidimensional molecular regulatory modules from the multidimensional omics data of cancer. In this paper, we propose a sparse orthogonality-regularized joint non-negative matrix factorization (SOJNMF) algorithm which can integratively analyze multidimensional omics data. This method can not only identify multidimensional molecular regulatory modules, but reduce the overlap rate of features among the multidimensional modules while ensuring the sparsity of the coefficient matrix after decomposition. Gene expression data, miRNA expression data and gene methylation data of liver cancer are integratively analyzed based on SOJNMF algorithm. Then, we obtain 238 multidimensional molecular regulatory modules. The results of permutation test indicate that different omics features within these modules are significantly correlated in statistics. Meanwhile, the results of functional enrichment analysis show that these multidimensional modules are significantly related to the underlying mechanism of the occurrence and development of liver cancer.
Collapse
|
25
|
Multi-Omics Analysis Revealed a Significant Alteration of Critical Metabolic Pathways Due to Sorafenib-Resistance in Hep3B Cell Lines. Int J Mol Sci 2022; 23:ijms231911975. [PMID: 36233276 PMCID: PMC9569810 DOI: 10.3390/ijms231911975] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/16/2022] [Accepted: 09/25/2022] [Indexed: 11/09/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the second prominent cause of cancer-associated death worldwide. Usually, HCC is diagnosed in advanced stages, wherein sorafenib, a multiple target tyrosine kinase inhibitor, is used as the first line of treatment. Unfortunately, resistance to sorafenib is usually encountered within six months of treatment. Therefore, there is a critical need to identify the underlying reasons for drug resistance. In the present study, we investigated the proteomic and metabolomics alterations accompanying sorafenib resistance in hepatocellular carcinoma Hep3B cells by employing ultra-high-performance liquid chromatography quadrupole time of flight mass spectrometry (UHPLC-QTOF-MS). The Bruker Human Metabolome Database (HMDB) library was used to identify the differentially abundant metabolites through MetaboScape 4.0 software (Bruker). For protein annotation and identification, the Uniprot proteome for Homo sapiens (Human) database was utilized through MaxQuant. The results revealed that 27 metabolites and 18 proteins were significantly dysregulated due to sorafenib resistance in Hep3B cells compared to the parental phenotype. D-alanine, L-proline, o-tyrosine, succinic acid and phosphatidylcholine (PC, 16:0/16:0) were among the significantly altered metabolites. Ubiquitin carboxyl-terminal hydrolase isozyme L1, mitochondrial superoxide dismutase, UDP-glucose-6-dehydrogenase, sorbitol dehydrogenase and calpain small subunit 1 were among the significantly altered proteins. The findings revealed that resistant Hep3B cells demonstrated significant alterations in amino acid and nucleotide metabolic pathways, energy production pathways and other pathways related to cancer aggressiveness, such as migration, proliferation and drug-resistance. Joint pathway enrichment analysis unveiled unique pathways, including the antifolate resistance pathway and other important pathways that maintain cancer cells' survival, growth, and proliferation. Collectively, the results identified potential biomarkers for sorafenib-resistant HCC and gave insights into their role in chemotherapeutic drug resistance, cancer initiation, progression and aggressiveness, which may contribute to better prognosis and chemotherapeutic outcomes.
Collapse
|
26
|
Gjorgjieva M, Ay AS, Correia de Sousa M, Delangre E, Dolicka D, Sobolewski C, Maeder C, Fournier M, Sempoux C, Foti M. MiR-22 Deficiency Fosters Hepatocellular Carcinoma Development in Fatty Liver. Cells 2022; 11:cells11182860. [PMID: 36139435 PMCID: PMC9496902 DOI: 10.3390/cells11182860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 12/24/2022] Open
Abstract
MiR-22 is mostly considered as a hepatic tumor-suppressor microRNA based on in vitro analyses. Yet, whether miR-22 exerts a tumor-suppressive function in the liver has not been investigated in vivo. Herein, in silico analyses of miR-22 expression were performed in hepatocellular carcinomas from human patient cohorts and different mouse models. Diethylnitrosamine-induced hepatocellular carcinomas were then investigated in lean and diet-induced obese miR-22-deficient mice. The proteome of liver tissues from miR-22-deficient mice prior to hepatocellular carcinoma development was further analyzed to uncover miR-22 regulated factors that impact hepatocarcinogenesis with miR-22 deficiency. MiR-22 downregulation was consistently observed in hepatocellular carcinomas from all human cohorts and mouse models investigated. The time of appearance of the first tumors was decreased and the number of tumoral foci induced by diethylnitrosamine was significantly increased by miR-22-deficiency in vivo, two features which were further drastically exacerbated with diet-induced obesity. At the molecular level, we provide evidence that the loss of miR-22 significantly affects the energetic metabolism and mitochondrial functions of hepatocytes, and the expression of tumor-promoting factors such as thrombospondin-1. Our study demonstrates that miR-22 acts as a hepatic tumor suppressor in vivo by restraining pro-carcinogenic metabolic deregulations through pleiotropic mechanisms and the overexpression of relevant oncogenes.
Collapse
Affiliation(s)
- Monika Gjorgjieva
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Anne-Sophie Ay
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Marta Correia de Sousa
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Etienne Delangre
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Dobrochna Dolicka
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Cyril Sobolewski
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Christine Maeder
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Margot Fournier
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Christine Sempoux
- Service of Clinical Pathology, Institute of Pathology, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Michelangelo Foti
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
- Translational Research Centre in Onco-Haematology, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
- Correspondence:
| |
Collapse
|
27
|
Tuszynski JA, Costa F. Low-energy amplitude-modulated radiofrequency electromagnetic fields as a systemic treatment for cancer: Review and proposed mechanisms of action. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:869155. [PMID: 36157082 PMCID: PMC9498185 DOI: 10.3389/fmedt.2022.869155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
Exposure to Low-Energy Amplitude-Modulated Radiofrequency Electromagnetic Fields (LEAMRFEMF) represents a new treatment option for patients with advanced hepatocellular carcinoma (AHCC). We focus on two medical devices that modulate the amplitude of a 27.12 MHz carrier wave to generate envelope waves in the low Hz to kHz range. Each provides systemic exposure to LEAMRFEMF via an intrabuccal antenna. This technology differs from so-called Tumour Treating Fields because it uses different frequency ranges, uses electromagnetic rather than electric fields, and delivers energy systemically rather than locally. The AutemDev also deploys patient-specific frequencies. LEAMRFEMF devices use 100-fold less power than mobile phones and have no thermal effects on tissue. Tumour type-specific or patient-specific treatment frequencies can be derived by measuring haemodynamic changes induced by exposure to LEAMRFEMF. These specific frequencies inhibited growth of human cancer cell lines in vitro and in mouse xenograft models. In uncontrolled prospective clinical trials in patients with AHCC, minorities of patients experienced complete or partial tumour responses. Pooled comparisons showed enhanced overall survival in treated patients compared to historical controls. Mild transient somnolence was the only notable treatment-related adverse event. We hypothesize that intracellular oscillations of charged macromolecules and ion flows couple resonantly with LEAMRFEMF. This resonant coupling appears to disrupt cell division and subcellular trafficking of mitochondria. We provide an estimate of the contribution of the electromagnetic effects to the overall energy balance of an exposed cell by calculating the power delivered to the cell, and the energy dissipated through the cell due to EMF induction of ionic flows along microtubules. We then compare this with total cellular metabolic energy production and conclude that energy delivered by LEAMRFEMF may provide a beneficial shift in cancer cell metabolism away from aberrant glycolysis. Further clinical research may confirm that LEAMRFEMF has therapeutic value in AHCC.
Collapse
Affiliation(s)
- Jack A. Tuszynski
- Division of Experimental Oncology, Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
- Dipartimento di Ingegneria Meccanica e Aerospaziale, Politecnico di Torino, Turin, Italy
- Autem Therapeutics, Hanover, NH, United States
| | - Frederico Costa
- Autem Therapeutics, Hanover, NH, United States
- Oncology Department, Hospital Sírio-Libanês, São Paulo, Brazil
| |
Collapse
|
28
|
Li J, Guan Y, Zhu R, Wang Y, Zhu H, Wang X. Identification of metabolic genes for the prediction of prognosis and tumor microenvironment infiltration in early-stage non-small cell lung cancer. Open Life Sci 2022; 17:881-892. [PMID: 36045718 PMCID: PMC9372707 DOI: 10.1515/biol-2022-0091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/03/2022] [Accepted: 05/03/2022] [Indexed: 11/15/2022] Open
Abstract
Early-stage non-small cell lung cancer (NSCLC) patients are at substantial risk of poor prognosis. We attempted to develop a reliable metabolic gene-set-based signature that can predict prognosis accurately for early-stage patients. Least absolute shrinkage and selection operator method Cox regression models were performed to filter the most useful prognostic genes, and a metabolic gene-set-based signature was constructed. Forty-two metabolism-related genes were finally identified, and with specific risk score formula, patients were classified into high-risk and low-risk groups. Overall survival was significantly different between the two groups in discovery (HR: 5.050, 95% CI: 3.368-7.574, P < 0.001), internal validation series (HR: 6.044, 95% CI: 3.918-9.322, P < 0.001), GSE30219 (HR: 2.059, 95% CI: 1.510-2.808, P < 0.001), and GSE68456 (HR: 2.448, 95% CI: 1.723-3.477, P < 0.001). Survival receiver operating characteristic curve at the 5 years suggested that the metabolic signature (area under the curve [AUC] = 0.805) had better prognostic accuracy than any other clinicopathological factors. Further analysis revealed the distinct differences in immune cell infiltration and tumor purity reflected by an immune and stromal score between high- and low-risk patients. In conclusion, the novel metabolic signature developed in our study shows robust prognostic accuracy in predicting prognosis for early-stage NSCLC patients and may function as a reliable marker for guiding more effective immunotherapy strategies.
Collapse
Affiliation(s)
- Jing Li
- Department of CyberKnife Center, Huashan Hospital, Fudan University, No. 525, Hongfeng Road, Pudong District, Shanghai 200040, China
| | - Yun Guan
- Department of CyberKnife Center, Huashan Hospital, Fudan University, No. 525, Hongfeng Road, Pudong District, Shanghai 200040, China
| | - Rongrong Zhu
- Department of Rehabilitation, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Yang Wang
- Department of CyberKnife Center, Huashan Hospital, Fudan University, No. 525, Hongfeng Road, Pudong District, Shanghai 200040, China
| | - Huaguang Zhu
- Department of CyberKnife Center, Huashan Hospital, Fudan University, No. 525, Hongfeng Road, Pudong District, Shanghai 200040, China
| | - Xin Wang
- Department of CyberKnife Center, Huashan Hospital, Fudan University, No. 525, Hongfeng Road, Pudong District, Shanghai 200040, China
| |
Collapse
|
29
|
Xia H, Huang Z, Xu Y, Yam JWP, Cui Y. Reprogramming of central carbon metabolism in hepatocellular carcinoma. Biomed Pharmacother 2022; 153:113485. [DOI: 10.1016/j.biopha.2022.113485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/20/2022] [Accepted: 07/27/2022] [Indexed: 11/02/2022] Open
|
30
|
Critical Investigation of the Usability of Hepatoma Cell Lines HepG2 and Huh7 as Models for the Metabolic Representation of Resectable Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:cancers14174227. [PMID: 36077764 PMCID: PMC9454736 DOI: 10.3390/cancers14174227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/16/2022] [Accepted: 08/26/2022] [Indexed: 12/24/2022] Open
Abstract
Metabolic alterations in hepatocellular carcinoma (HCC) are fundamental for the development of diagnostic screening and therapeutic intervention since energy metabolism plays a central role in differentiated hepatocytes. In HCC research, hepatoma cell lines (HCLs) like HepG2 and Huh7 cells are still the gold standard. In this study, we characterized the metabolic profiles of primary human hepatoma cells (PHCs), HCLs and primary human hepatocytes (PHHs) to determine their differentiation states. PHCs and PHHs (HCC-PHHs) were isolated from surgical specimens of HCC patients and their energy metabolism was compared to PHHs from non-HCC patients and the HepG2 and Huh7 cells at different levels (transcript, protein, function). Our analyses showed successful isolation of PHCs with a purity of 50–73% (CK18+). The transcript data revealed that changes in mRNA expression levels had already occurred in HCC-PHHs. While many genes were overexpressed in PHCs and HCC-PHHs, the changes were mostly not translated to the protein level. Downregulated metabolic key players of PHCs revealed a correlation with malign transformation and were predominantly pronounced in multilocular HCC. Therefore, HCLs failed to reflect these expression patterns of PHCs at the transcript and protein levels. The metabolic characteristics of PHCs are closer to those of HCC-PHHs than to HCLs. This should be taken into account for future optimized tumor metabolism research.
Collapse
|
31
|
Xia JK, Qin XQ, Zhang L, Liu SJ, Shi XL, Ren HZ. Roles and regulation of histone acetylation in hepatocellular carcinoma. Front Genet 2022; 13:982222. [PMID: 36092874 PMCID: PMC9452893 DOI: 10.3389/fgene.2022.982222] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Hepatocellular Carcinoma (HCC) is the most frequent malignant tumor of the liver, but its prognosis is poor. Histone acetylation is an important epigenetic regulatory mode that modulates chromatin structure and transcriptional status to control gene expression in eukaryotic cells. Generally, histone acetylation and deacetylation processes are controlled by the opposing activities of histone acetyltransferases (HATs) and histone deacetylases (HDACs). Dysregulation of histone modification is reported to drive aberrant transcriptional programmes that facilitate liver cancer onset and progression. Emerging studies have demonstrated that several HDAC inhibitors exert tumor-suppressive properties via activation of various cell death molecular pathways in HCC. However, the complexity involved in the epigenetic transcription modifications and non-epigenetic cellular signaling processes limit their potential clinical applications. This review brings an in-depth view of the oncogenic mechanisms reported to be related to aberrant HCC-associated histone acetylation, which might provide new insights into the effective therapeutic strategies to prevent and treat HCC.
Collapse
Affiliation(s)
- Jin-kun Xia
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Hepatobiliary Institute Nanjing University, Nanjing, China
| | - Xue-qian Qin
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Lu Zhang
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Shu-jun Liu
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiao-lei Shi
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Hepatobiliary Institute Nanjing University, Nanjing, China
| | - Hao-zhen Ren
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Hepatobiliary Institute Nanjing University, Nanjing, China
| |
Collapse
|
32
|
Hang J, Chen Y, Liu L, Chen L, Fang J, Wang F, Wang M. Antitumor effect and metabonomics of niclosamide micelles. J Cell Mol Med 2022; 26:4814-4824. [PMID: 35923077 PMCID: PMC9465187 DOI: 10.1111/jcmm.17509] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 07/13/2022] [Accepted: 07/19/2022] [Indexed: 01/12/2023] Open
Abstract
Polymer micelles now have promising applications in the treatment of cancer, increasing the water solubility and bioavailability of drugs. Previous studies have found that micelles of niclosamide have good anti‐liver cancer effect. In view of the poor water solubility of niclosamide (NIC), we decided to prepare niclosamide micelles. However, its therapeutic mechanism is not clear, so this paper conducted a preliminary study on its vitro anti‐tumour mechanism and metabonomics to find out its impact. It was found that the drug‐loaded micelles (PEG2K‐FIbu/NIC) had an inhibitory effect on HepG2 cells. Moreover, it can promote apoptosis of HepG2 cells and block S and G2/M phase of cell cycle. The plasma and liver metabolomics of mice in normal group, model group and administration group were studied by UPLC‐MS and 1H‐NMR. Principal component analysis (PCA) and orthogonal partial least squares discriminant analysis (OPLS‐DA) were used to process the data and find the relevant metabolites. metaboanalyst 5.0 was used to integrate the relevant metabolites to find the main related metabolic pathways. Thus, the anti‐tumour mechanism of PEG2K‐FIbu/NIC was analysed. Fifty‐one biomarkers were detected in plasma, and 43 biomarkers were detected in liver. After comprehensive biomarker and metabolic pathway analysis, it was found that PEG2K‐FIbu/NIC micelles could affect the changes of many metabolites, mainly affecting amino acid metabolism. This article is an in‐depth study based on the published Preparation and pharmacodynamics of niclosamide micelles (DOI: 10.1016/j.jddst.2021.103088).
Collapse
Affiliation(s)
- Jiarong Hang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Yu Chen
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Lukuan Liu
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Liwen Chen
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Jiqin Fang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Fei Wang
- Faculty of Robot Science and Engineering, Northeastern University, Shenyang, China
| | - Miao Wang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
33
|
Hang D, Yang X, Lu J, Shen C, Dai J, Lu X, Jin G, Hu Z, Gu D, Ma H, Shen H. Untargeted plasma metabolomics for risk prediction of hepatocellular carcinoma: A prospective study in two Chinese cohorts. Int J Cancer 2022; 151:2144-2154. [PMID: 35904854 DOI: 10.1002/ijc.34229] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/30/2022] [Accepted: 07/20/2022] [Indexed: 11/08/2022]
Abstract
Characterization of metabolic perturbation prior to hepatocellular carcinoma (HCC) may deepen the understanding of causal pathways and identify novel biomarkers for early prevention. We conducted two 1:1 matched nested case-control studies (108 and 55 pairs) to examine the association of plasma metabolome (profiled using LC-MS) with the risk of HCC based on two prospective cohorts in China. Differential metabolites were identified by paired t-tests and orthogonal partial least-squares discriminant analysis (OPLS-DA). Weighted gene co-expression network analysis (WGCNA) was performed to classify metabolites into modules for identifying biological pathways involved in hepatocarcinogenesis. We assessed the risk predictivity of metabolites using multivariable logistic regression models. Among 612 named metabolites, 44 differential metabolites were identified between cases and controls, including 12 androgenic/progestin steroid hormones, 8 bile acids, 10 amino acids, 6 phospholipids, and 8 others. These metabolites were associated with HCC in the multivariable logistic regression analyses, with odds ratios ranging from 0.19 (95% CI: 0.11-0.35) to 5.09 (95% CI: 2.73-9.50). WGCNA including 612 metabolites showed 8 significant modules related to HCC risk, including those representing metabolic pathways of androgen and progestin, primary and secondary bile acids, and amino acids. A combination of 18 metabolites of independent effects showed the potential to predict HCC risk, with an AUC of 0.87 (95% CI: 0.82-0.92) and 0.86 (95% CI: 0.80-0.93) in the training and validation sets, respectively. In conclusion, we identified a panel of plasma metabolites that could be implicated in hepatocellular carcinogenesis and have the potential to predict HCC risk.
Collapse
Affiliation(s)
- Dong Hang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine and International Joint Research Center on Environment and Human Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine and China International Cooperation Center for Environment and Human Health, Gusu School, Nanjing Medical University, Nanjing, China
| | - Xiaolin Yang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Epidemiology, School of Public Health, Southeast University, Nanjing, China
| | - JiaYi Lu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chong Shen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Juncheng Dai
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiangfeng Lu
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing, China.,Research Units of Cohort Study on Cardiovascular Diseases and Cancers, Chinese Academy of Medical Sciences
| | - Guangfu Jin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine and International Joint Research Center on Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Zhibin Hu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine and International Joint Research Center on Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Dongfeng Gu
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing, China.,Research Units of Cohort Study on Cardiovascular Diseases and Cancers, Chinese Academy of Medical Sciences
| | - Hongxia Ma
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine and International Joint Research Center on Environment and Human Health, Nanjing Medical University, Nanjing, China.,Research Units of Cohort Study on Cardiovascular Diseases and Cancers, Chinese Academy of Medical Sciences
| | - Hongbing Shen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine and International Joint Research Center on Environment and Human Health, Nanjing Medical University, Nanjing, China.,Research Units of Cohort Study on Cardiovascular Diseases and Cancers, Chinese Academy of Medical Sciences
| |
Collapse
|
34
|
Whyte SS, Karns R, Min K, Cho J, Lee S, Lake C, Bondoc A, Yoon J, Shin S. Integrated analysis using ToppMiR uncovers altered miRNA- mRNA regulatory networks in pediatric hepatocellular carcinoma-A pilot study. Cancer Rep (Hoboken) 2022; 6:e1685. [PMID: 35859536 PMCID: PMC9875636 DOI: 10.1002/cnr2.1685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/23/2022] [Accepted: 07/12/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pediatric hepatocellular carcinoma (HCC) is a group of liver cancers whose mechanisms behind their pathogenesis and progression are poorly understood. AIM We aimed to identify alterations in the expression of miRNAs and their putative target mRNAs in not only tumor tissues of patients with pediatric HCC but also in corresponding non-tumorous background livers by using liver tissues without underlying liver disease as a control. METHODS AND RESULTS We performed a small-scale miRNA and mRNA profiling of pediatric HCC (consisting of fibrolamellar carcinoma [FLC] and non-FLC HCC) and paired liver tissues to identify miRNAs whose expression levels differed significantly from control livers without underlying liver disease. ToppMiR was used to prioritize both miRNAs and their putative target mRNAs in a gene-annotation network, and the mRNA profile was used to refine the prioritization. Our analysis generated prioritized lists of miRNAs and mRNAs from the following three sets of analyses: (a) pediatric HCC versus control; (b) FLC versus control; and (c) corresponding non-tumorous background liver tissues from the same patients with pediatric HCC versus control. No liver disease liver tissues were used as the control group for all analyses. Many miRNAs whose expressions were deregulated in pediatric HCC were consistent with their roles in adult HCC and/or other non-hepatic cancers. Our gene ontology analysis of target mRNAs revealed enrichment of biological processes related to the sustenance and propagation of cancer and significant downregulation of metabolic processes. CONCLUSION Our pilot study indicates that alterations in miRNA-mRNA networks were detected in not only tumor tissues but also corresponding non-tumorous liver tissues from patients with pediatric HCC, suggesting multi-faceted roles of miRNAs in disease progression. Our results may lead to novel hypotheses for future large-scale studies.
Collapse
Affiliation(s)
- Senyo S. Whyte
- Division of Pediatric General and Thoracic SurgeryCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Rebekah Karns
- Division of Gastroenterology, Hepatology & NutritionCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Kyung‐Won Min
- Department of BiologyGangneung‐Wonju National UniversityGangneungRepublic of Korea
| | - Jung‐Hyun Cho
- Department of Biochemistry and Molecular BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Sanghoon Lee
- Division of Pediatric General and Thoracic SurgeryCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Charissa Lake
- Division of Pediatric General and Thoracic SurgeryCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Alexander Bondoc
- Division of Pediatric General and Thoracic SurgeryCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA,Department of SurgeryUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Je‐Hyun Yoon
- Department of Biochemistry and Molecular BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Soona Shin
- Division of Pediatric General and Thoracic SurgeryCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA,Department of SurgeryUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| |
Collapse
|
35
|
Gene Expression Analysis Reveals Prognostic Biomarkers of the Tyrosine Metabolism Reprogramming Pathway for Prostate Cancer. JOURNAL OF ONCOLOGY 2022; 2022:5504173. [PMID: 35847355 PMCID: PMC9279037 DOI: 10.1155/2022/5504173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/23/2022] [Indexed: 11/30/2022]
Abstract
Background Tyrosine metabolism pathway-related genes were related to prostate cancer progression, which may be used as potential prognostic markers. Aims To dissect the dysregulation of tyrosine metabolism in prostate cancer and build a prognostic signature based on tyrosine metabolism-related genes for prostate cancer. Materials and Method. Cross-platform gene expression data of prostate cancer cohorts were collected from both The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). Based on the expression of tyrosine metabolism-related enzymes (TMREs), an unsupervised consensus clustering method was used to classify prostate cancer patients into different molecular subtypes. We employed the least absolute shrinkage and selection operator (LASSO) Cox regression analysis to evaluate prognostic characteristics based on TMREs to obtain a prognostic effect. The nomogram model was established and used to synthesize molecular subtypes, prognostic characteristics, and clinicopathological features. Kaplan–Meier plots and logrank analysis were used to clarify survival differences between subtypes. Results Based on the hierarchical clustering method and the expression profiles of TMREs, prostate cancer samples were assigned into two subgroups (S1, subgroup 1; S2, subgroup 2), and the Kaplan–Meier plot and logrank analysis showed distinct survival outcomes between S1 and S2 subgroups. We further established a four-gene-based prognostic signature, and both in-group testing dataset and out-group testing dataset indicated the robustness of this model. By combining the four gene-based signatures and clinicopathological features, the nomogram model achieved better survival outcomes than any single classifier. Interestingly, we found that immune-related pathways were significantly concentrated on S1-upregulated genes, and the abundance of memory B cells, CD4+ resting memory T cells, M0 macrophages, resting dendritic cells, and resting mast cells were significantly different between S1 and S2 subgroups. Conclusions Our results indicate the prognostic value of genes related to tyrosine metabolism in prostate cancer and provide inspiration for treatment and prevention strategies.
Collapse
|
36
|
Xia H, Huang Z, Wang Z, Liu S, Zhao X, You J, Xu Y, Yam JWP, Cui Y. Glucometabolic reprogramming: From trigger to therapeutic target in hepatocellular carcinoma. Front Oncol 2022; 12:953668. [PMID: 35912218 PMCID: PMC9336635 DOI: 10.3389/fonc.2022.953668] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/27/2022] [Indexed: 12/11/2022] Open
Abstract
Glucose, the central macronutrient, releases energy as ATP through carbon bond oxidation and supports various physiological functions of living organisms. Hepatocarcinogenesis relies on the bioenergetic advantage conferred by glucometabolic reprogramming. The exploitation of reformed metabolism induces a uniquely inert environment conducive to survival and renders the hepatocellular carcinoma (HCC) cells the extraordinary ability to thrive even in the nutrient-poor tumor microenvironment. The rewired metabolism also confers a defensive barrier which protects the HCC cells from environmental stress and immune surveillance. Additionally, targeted interventions against key players of HCC metabolic and signaling pathways provide promising prospects for tumor therapy. The active search for novel drugs based on innovative mutation targets is warranted in the future for effectively treating advanced HCC and the preoperative downstage. This article aims to review the regulatory mechanisms and therapeutic value of glucometabolic reprogramming on the disease progression of HCC, to gain insights into basic and clinical research.
Collapse
Affiliation(s)
- Haoming Xia
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ziyue Huang
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhensheng Wang
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuqiang Liu
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xudong Zhao
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Junqi You
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yi Xu
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- *Correspondence: Yi Xu, ; Judy Wai Ping Yam, ; Yunfu Cui,
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- *Correspondence: Yi Xu, ; Judy Wai Ping Yam, ; Yunfu Cui,
| | - Yunfu Cui
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Yi Xu, ; Judy Wai Ping Yam, ; Yunfu Cui,
| |
Collapse
|
37
|
Sharifi H, Safarpour H, Moossavi M, Khorashadizadeh M. Identification of Potential Prognostic Markers and Key Therapeutic Targets in Hepatocellular Carcinoma Using Weighted Gene Co-Expression Network Analysis: A Systems Biology Approach. IRANIAN JOURNAL OF BIOTECHNOLOGY 2022; 20:e2968. [PMID: 36381283 PMCID: PMC9618018 DOI: 10.30498/ijb.2022.269817.2968] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND As the most prevalent form of liver cancer, hepatocellular carcinoma (HCC) ranks the fifth highest cause of cancer-related death worldwide. Despite recent advancements in diagnostic and therapeutic techniques, the prognosis for HCC is still unknown. OBJECTIVES This study aimed to identify potential genes contributing to HCC pathogenicity. MATERIALS AND METHODS To this end, we examined the GSE39791 microarray dataset, which included 72 HCC samples and 72 normal samples. An investigation of co-expression networks using WGCNA found a highly conserved blue module with 665 genes that were strongly linked to HCC. RESULTS APOF, NAT2, LCAT, TTC36, IGFALS, ASPDH, and VIPR1 were the blue module's top 7 hub genes. According to the results of hub gene enrichment, the most related issues in the biological process and KEGG were peroxisome organization and metabolic pathways, respectively. In addition, using the drug-target network, we discovered 19 FDA-approved medication candidates for different reasons that might potentially be employed to treat HCC patients through the modulation of 3 hub genes of the co-expression network (LCAT, NAT2, and VIPR1). Our findings also demonstrated that the 3 scientifically validated miRNAs regulated the co-expression network by the VIPR1 hub gene. CONCLUSION We found co-expressed gene modules and hub genes linked with HCC advancement, offering insights into the mechanisms underlying HCC progression as well as some potential HCC treatments.
Collapse
Affiliation(s)
- Hengameh Sharifi
- Department of Molecular Medicine, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Hossein Safarpour
- Cellular & Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Maryam Moossavi
- Department of Molecular Medicine, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohsen Khorashadizadeh
- Department of Molecular Medicine, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran,
Cellular & Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran,
3Department of Medical Biotechnology, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
38
|
Xu J, Cao W, Shao A, Yang M, Andoh V, Ge Q, Pan HW, Chen KP. Metabolomics of Esophageal Squamous Cell Carcinoma Tissues: Potential Biomarkers for Diagnosis and Promising Targets for Therapy. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7819235. [PMID: 35782075 PMCID: PMC9246618 DOI: 10.1155/2022/7819235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 12/24/2022]
Abstract
Background The incidence of esophageal squamous cell carcinoma in China ranks first in the world. The early diagnosis technology is underdeveloped, and the prognosis is poor, which seriously threatens the quality of life of the Chinese people. Epidemiological findings are related to factors such as diet, living habits, and age. The specific mechanism is not clear yet. Metabolomics is a kind of omics that simultaneously and quantitatively analyzes the comprehensive profile of metabolites in living systems. It has unique advantages in the study of the diagnosis and pathogenesis of tumor-related diseases, especially in the search for biomarkers. Therefore, it is desirable to perform metabolic profiling analysis of cancer tissues through metabolomics to find potential biomarkers for the diagnosis and treatment of esophageal squamous cell carcinoma. Methods HPLC-TOF-MS/MS technology and Illumina Hiseq Xten Sequencing was used for the analysis of 210 pairs of matched esophageal squamous cell carcinoma tissues and normal tissues in Zhenjiang City, Jiangsu Province, a high-incidence area of esophageal cancer in China. Bioinformatics analysis was also performed. Results Through metabolomic and transcriptomic analysis, this study found that a total of 269 differential metabolites were obtained in esophageal squamous cell carcinoma and normal tissues, and 48 differential metabolic pathways were obtained through KEGG enrichment analysis. After further screening and identification, 12 metabolites with potential biomarkers to differentiate esophageal squamous cell carcinoma from normal tissues were obtained. Conclusions From the metabolomic data, 4 unknown compounds were found to be abnormally expressed in esophageal squamous cell carcinoma for the first time, such as 9,10-epoxy-12,15-octadecadienoate; 3 metabolites were found in multiple abnormal expression in another tumor, but upregulation or downregulation was found for the first time in esophageal cancer, such as oleoyl glycine; at the same time, it was further confirmed that five metabolites were abnormally expressed in esophageal squamous cell carcinoma, which was similar to the results of other studies, such as PE.
Collapse
Affiliation(s)
- Jia Xu
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Weiping Cao
- The Fourth People's Hospital of Zhenjiang, Zhenjiang, Jiangsu 212001, China
| | - Aizhong Shao
- Department of Cardiothorac Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Ming Yang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Vivian Andoh
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Qi Ge
- School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Hui-wen Pan
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Ke-ping Chen
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| |
Collapse
|
39
|
Differential response of hepatocellular carcinoma glycolytic metabolism and oxidative stress markers after exposure to human amniotic membrane proteins. Mol Biol Rep 2022; 49:7731-7741. [PMID: 35716291 DOI: 10.1007/s11033-022-07598-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 04/16/2022] [Accepted: 05/12/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND The human Amniotic Membrane (hAM) has been studied as a potential therapeutic option in cancer, namely in hepatocellular carcinoma. Previously, our research group evaluated the effect of human Amniotic Membrane Protein Extracts (hAMPE) in cancer therapy, demonstrating that hAMPE inhibit the metabolic activity of human hepatocellular carcinoma cell lines: Hep3B2.1-7, HepG2 and Huh7. Therefore, and considering the close relationship between metabolic activity and oxidative stress, the aim of this study was to evaluate the effect of hAMPE treatment in glucose metabolism and its role in oxidative stress of hepatocellular carcinoma. METHODS AND RESULTS Glucose uptake and lactate production was assessed by 1 H-NMR, and the expression of several mediators of the glycolytic pathway was evaluated by Western blot or fluorescence. Total antioxidant capacity (TAC) and biomarkers of oxidative stress effects in proteins were detected. Our results showed that hAMPE treatment increased glucose consumption on Hep3B2.1-7, HepG2, and Huh7 through the increase of GLUT1 in Hep3B2.1-7 and Huh7, and GLUT3 in HepG2 cells. It was observed an increased expression of 6-phosphofrutokinase (PFK-1L) in all cell lines though glucose was not converted to lactate on HepG2 and Huh7 cells, suggesting that hAMPE treatment may counteract the Warburg effect observed in carcinogenesis. In Hep3B2.1-7, hAMPE treatment induced an increase in expression of lactate dehydrogenase (LDH) and monocarboxylate transporter isoform 4 (MCT4). We further detected that hAMPE enhances the TAC of culture media after 2 and 8 h. This was followed by a degree of protection against proteins nitration and carbonylation. CONCLUSIONS Overall, this work highlights the potential usefulness of hAMPE as anticancer therapy through the modulation of the glycolytic and oxidative profile in human hepatocellular carcinoma.
Collapse
|
40
|
Vishnoi K, Kumar S, Ke R, Rana A, Rana B. Dysregulation of immune checkpoint proteins in hepatocellular carcinoma: Impact on metabolic reprogramming. Curr Opin Pharmacol 2022; 64:102232. [DOI: 10.1016/j.coph.2022.102232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/11/2022] [Accepted: 03/23/2022] [Indexed: 11/15/2022]
|
41
|
Hydroxyacid Oxidase 2 (HAO2) Inhibits the Tumorigenicity of Hepatocellular Carcinoma and Is Negatively Regulated by miR-615-5p. J Immunol Res 2022; 2022:5003930. [PMID: 35528616 PMCID: PMC9071856 DOI: 10.1155/2022/5003930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/09/2022] [Indexed: 12/30/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the sixth most common kind of cancer worldwide and the third leading cause of cancer mortality. Although a few studies have shown that hydroxyacid oxidase 2 (HAO2) may prevent HCC development, the molecular mechanism is unclear. Methods We examined the levels of HAO2 expression in 23 pairs of HCC/paracancerous tissues by quantitative real-time polymerase chain reaction (qRT-PCR) and evaluated HAO2's expression in The Cancer Genome Atlas (TCGA) database. Furthermore, we examined the biological activity of HAO2 utilizing cell-based functional assays. Additionally, we evaluated the relationship between miR-615-5p and HAO2 in Hep3B cells using a dual-luciferase reporter system and assessed the downstream regulatory mechanisms of miR-615-5p on HAO2. Finally, the nude mice tumor formation experiment was used to determine the impact of HAO2 on the tumorigenicity of HCC cells. Results HAO2 expression was considerably underexpression in HCC tissues and cells, and patients with low HAO2 expression had poorer disease-free survival. Inhibition of cell proliferation, migration, and invasion was observed when HAO2 was overexpressed. miR-615-5p had a negative relation with HAO2, and miR-615-5p restored HAO2's biological activity in HCC cells. Additionally, the tumor volume and weight were considerably reduced in the OV-HAO2 group compared to the OV-NC group. Conclusion HAO2 was found to be underexpressed in HCC tissues and cells, and HAO2 overexpression inhibited HCC cell motility, which was negatively regulated by miR-615-5p. Exogenous expression of HAO2 reduced the tumorigenicity of HCC cells in vivo in nude mice.
Collapse
|
42
|
Ye Z, Song B, Lee PM, Ohliger MA, Laustsen C. Hyperpolarized carbon 13 MRI in liver diseases: Recent advances and future opportunities. Liver Int 2022; 42:973-983. [PMID: 35230742 PMCID: PMC9313895 DOI: 10.1111/liv.15222] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 01/20/2022] [Accepted: 02/04/2022] [Indexed: 02/05/2023]
Abstract
Hyperpolarized carbon-13 magnetic resonance imaging (HP 13 C MRI) is a recently translated metabolic imaging technique. With dissolution dynamic nuclear polarization (d-DNP), more than 10 000-fold signal enhancement can be readily reached, making it possible to visualize real-time metabolism and specific substrate-to-metabolite conversions in the liver after injecting carbon-13 labelled probes. Increasing evidence suggests that HP 13 C MRI is a potential tool in detecting liver abnormalities, predicting disease progression and monitoring response treatment. In this review, we will introduce the recent progresses of HP 13 C MRI in diffuse liver diseases and liver malignancies and discuss its future opportunities from a clinical perspective, hoping to provide a comprehensive overview of this novel technique in liver diseases and highlight its scientific and clinical potential in the field of hepatology.
Collapse
Affiliation(s)
- Zheng Ye
- Department of RadiologyWest China Hospital, Sichuan UniversityChengduSichuanChina
- The MR Research Center, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Bin Song
- Department of RadiologyWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Philip M. Lee
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Michael A. Ohliger
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Christoffer Laustsen
- The MR Research Center, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| |
Collapse
|
43
|
Ning Z, Guo X, Liu X, Lu C, Wang A, Wang X, Wang W, Chen H, Qin W, Liu X, Zhou L, Ma C, Du J, Lin Z, Luo H, Otkur W, Qi H, Chen D, Xia T, Liu J, Tan G, Xu G, Piao HL. USP22 regulates lipidome accumulation by stabilizing PPARγ in hepatocellular carcinoma. Nat Commun 2022; 13:2187. [PMID: 35449157 PMCID: PMC9023467 DOI: 10.1038/s41467-022-29846-9] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 03/30/2022] [Indexed: 12/13/2022] Open
Abstract
Elevated de novo lipogenesis is considered to be a crucial factor in hepatocellular carcinoma (HCC) development. Herein, we identify ubiquitin-specific protease 22 (USP22) as a key regulator for de novo fatty acid synthesis, which directly interacts with deubiquitinates and stabilizes peroxisome proliferator-activated receptor gamma (PPARγ) through K48-linked deubiquitination, and in turn, this stabilization increases acetyl-CoA carboxylase (ACC) and ATP citrate lyase (ACLY) expressions. In addition, we find that USP22 promotes de novo fatty acid synthesis and contributes to HCC tumorigenesis, however, this tumorigenicity is suppressed by inhibiting the expression of PPARγ, ACLY, or ACC in in vivo tumorigenesis experiments. In HCC, high expression of USP22 positively correlates with PPARγ, ACLY or ACC expression, and associates with a poor prognosis. Taken together, we identify a USP22-regulated lipogenesis mechanism that involves the PPARγ-ACLY/ACC axis in HCC tumorigenesis and provide a rationale for therapeutic targeting of lipogenesis via USP22 inhibition. Different deubiquitinases are associated to cancer development. Here, the authors show that PPARgamma is stabilized by USP22-mediated deubiquitination leading to lipid accumulation and promoting hepatocellular carcinoma.
Collapse
Affiliation(s)
- Zhen Ning
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.,The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116000, China.,Liaoning Key Laboratory of Molecular Targeted Drugs in Hepatobiliary and Pancreatic Cancer, Dalian, 116000, China
| | - Xin Guo
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.,The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116000, China
| | - Xiaolong Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Chang Lu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.,The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116000, China
| | - Aman Wang
- The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116000, China.,Liaoning Key Laboratory of Molecular Targeted Drugs in Hepatobiliary and Pancreatic Cancer, Dalian, 116000, China
| | - Xiaolin Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Wen Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Huan Chen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Wangshu Qin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Xinyu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Lina Zhou
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Chi Ma
- The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116000, China.,Liaoning Key Laboratory of Molecular Targeted Drugs in Hepatobiliary and Pancreatic Cancer, Dalian, 116000, China
| | - Jian Du
- The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116000, China.,Liaoning Key Laboratory of Molecular Targeted Drugs in Hepatobiliary and Pancreatic Cancer, Dalian, 116000, China
| | - Zhikun Lin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.,The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116000, China.,Liaoning Key Laboratory of Molecular Targeted Drugs in Hepatobiliary and Pancreatic Cancer, Dalian, 116000, China
| | - Haifeng Luo
- The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116000, China.,Liaoning Key Laboratory of Molecular Targeted Drugs in Hepatobiliary and Pancreatic Cancer, Dalian, 116000, China
| | - Wuxiyar Otkur
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Huan Qi
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Di Chen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Tian Xia
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Jiwei Liu
- The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116000, China.,Liaoning Key Laboratory of Molecular Targeted Drugs in Hepatobiliary and Pancreatic Cancer, Dalian, 116000, China
| | - Guang Tan
- The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116000, China. .,Liaoning Key Laboratory of Molecular Targeted Drugs in Hepatobiliary and Pancreatic Cancer, Dalian, 116000, China.
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.
| | - Hai-Long Piao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
44
|
Wang W, Yang C, Wang T, Deng H. Complex roles of nicotinamide N-methyltransferase in cancer progression. Cell Death Dis 2022; 13:267. [PMID: 35338115 PMCID: PMC8956669 DOI: 10.1038/s41419-022-04713-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/23/2022] [Accepted: 03/08/2022] [Indexed: 02/07/2023]
Abstract
Nicotinamide N-methyltransferase (NNMT) is an intracellular methyltransferase, catalyzing the N-methylation of nicotinamide (NAM) to form 1-methylnicotinamide (1-MNAM), in which S-adenosyl-l-methionine (SAM) is the methyl donor. High expression of NNMT can alter cellular NAM and SAM levels, which in turn, affects nicotinamide adenine dinucleotide (NAD+)-dependent redox reactions and signaling pathways, and remodels cellular epigenetic states. Studies have revealed that NNMT plays critical roles in the occurrence and development of various cancers, and analysis of NNMT expression levels in different cancers from The Cancer Genome Atlas (TCGA) dataset indicated that NNMT might be a potential biomarker and therapeutic target for tumor diagnosis and treatment. This review provides a comprehensive understanding of recent advances on NNMT functions in different tumors and deciphers the complex roles of NNMT in cancer progression.
Collapse
Affiliation(s)
- Weixuan Wang
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, People's Republic of China
| | - Changmei Yang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Tianxiang Wang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing, People's Republic of China.
| |
Collapse
|
45
|
Park Y, Han Y, Kim D, Cho S, Kim W, Hwang H, Lee HW, Han DH, Kim KS, Yun M, Lee M. Impact of Exogenous Treatment with Histidine on Hepatocellular Carcinoma Cells. Cancers (Basel) 2022; 14:cancers14051205. [PMID: 35267513 PMCID: PMC8909034 DOI: 10.3390/cancers14051205] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Sorafenib (Nexavar@) is the only currently approved anti-cancer drug for patients with advanced hepatocellular carcinoma (HCC). However, despite the development of strategies combining sorafenib with other cytotoxic chemotherapeutic agents to overcome sorafenib resistance, clinical trial results are still disappointing. In this study, we examined the enhancement of tumor responses to sorafenib by exogenous histidine treatment. Abstract Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths worldwide. Sorafenib, a multi-kinase inhibitor, is the first-line therapy for advanced HCC. However, long-term exposure to sorafenib often results in reduced sensitivity and the development of resistance. Although various amino acids have been shown to contribute to cancer initiation and progression, little is known about the effects of histidine, a dietary essential amino acid that is partially taken up via histidine/large neutral amino acid transporter (LAT1), on cancer cells. In this study, we evaluated the effects of histidine on HCC cells and sensitivity to sorafenib. Remarkably, we found that exogenous histidine treatment induced a reduction in the expression of tumor markers related to glycolysis (GLUT1 and HK2), inflammation (STAT3), angiogenesis (VEGFB and VEGFC), and stem cells (CD133). In addition, LAT1 expression was downregulated in HCC tumor regions with high expression of GLUT1, CD133, and pSTAT3, which are known to induce sorafenib resistance. Finally, we demonstrated that combined treatment with sorafenib and histidine could be a novel therapeutic strategy to enhance the sensitivity to sorafenib, thereby improving long-term survival in HCC.
Collapse
Affiliation(s)
- Yusun Park
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Korea; (Y.P.); (Y.H.); (S.C.); (W.K.); (H.H.)
| | - Yeonju Han
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Korea; (Y.P.); (Y.H.); (S.C.); (W.K.); (H.H.)
| | - Dongwoo Kim
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Sua Cho
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Korea; (Y.P.); (Y.H.); (S.C.); (W.K.); (H.H.)
| | - WonJin Kim
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Korea; (Y.P.); (Y.H.); (S.C.); (W.K.); (H.H.)
| | - Hyemin Hwang
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Korea; (Y.P.); (Y.H.); (S.C.); (W.K.); (H.H.)
| | - Hye Won Lee
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Dai Hoon Han
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (D.H.H.); (K.S.K.)
| | - Kyung Sik Kim
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (D.H.H.); (K.S.K.)
| | - Mijin Yun
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Korea;
- Correspondence: (M.Y.); (M.L.)
| | - Misu Lee
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Korea; (Y.P.); (Y.H.); (S.C.); (W.K.); (H.H.)
- Institute for New Drug Development, College of Life Science and Bioengineering, Incheon National University, Incheon 22012, Korea
- Correspondence: (M.Y.); (M.L.)
| |
Collapse
|
46
|
Zhao N, Zhang Y, Cheng R, Zhang D, Li F, Guo Y, Qiu Z, Dong X, Ban X, Sun B, Zhao X. Spatial maps of hepatocellular carcinoma transcriptomes highlight an unexplored landscape of heterogeneity and a novel gene signature for survival. Cancer Cell Int 2022; 22:57. [PMID: 35109839 PMCID: PMC8812006 DOI: 10.1186/s12935-021-02430-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 12/24/2021] [Indexed: 01/07/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) often presents with satellite nodules, rendering current curative treatments ineffective in many patients. The heterogeneity of HCC is a major challenge in personalized medicine. The emergence of spatial transcriptomics (ST) provides a powerful strategy for delineating the complex molecular landscapes of tumours. Methods In this study, the heterogeneity of tissue-wide gene expression in tumour and adjacent nonneoplastic tissues using ST technology were investigated. The transcriptomes of nearly 10,820 tissue regions and identified the main gene expression clusters and their specific marker genes (differentially expressed genes, DEGs) in patients were analysed. The DEGs were analysed from two perspectives. First, two distinct gene profiles were identified to be associated with satellite nodules and conducted a more comprehensive analysis of both gene profiles. Their clinical relevance in human HCC was validated with Kaplan–Meier (KM) Plotter. Second, DEGs were screened with The Cancer Genome Atlas (TCGA) database to divide the HCC cohort into high- and low-risk groups according to Cox analysis. HCC patients from the International Cancer Genome Consortium (ICGC) cohort were used for validation. KM analysis was used to compare the overall survival (OS) between the high- and low-risk groups. Univariate and multivariate Cox analyses were applied to determine the independent predictors for OS. Results Novel markers for the prediction of satellite nodules were identified and a tumour clusters-specific marker gene signature model (6 genes) for HCC prognosis was constructed. Conclusion The establishment of marker gene profiles may be an important step towards an unbiased view of HCC, and the 6-gene signature can be used for prognostic prediction in HCC. This analysis will help us to clarify one of the possible sources of HCC heterogeneity and uncover pathogenic mechanisms and novel antitumour drug targets. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02430-9.
Collapse
Affiliation(s)
- Nan Zhao
- Department of Pathology, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Yanhui Zhang
- Department of Pathology, Cancer Hospital of Tianjin Medical University, Tianjin, 300060, China
| | - Runfen Cheng
- Department of Pathology, Cancer Hospital of Tianjin Medical University, Tianjin, 300060, China
| | - Danfang Zhang
- Department of Pathology, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Fan Li
- Department of Pathology, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Yuhong Guo
- Department of Pathology, Cancer Hospital of Tianjin Medical University, Tianjin, 300060, China
| | - Zhiqiang Qiu
- Department of Pathology, Cancer Hospital of Tianjin Medical University, Tianjin, 300060, China
| | - Xueyi Dong
- Department of Pathology, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Xinchao Ban
- Department of Pathology, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China.,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, 300052, China
| | - Baocun Sun
- Department of Pathology, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China. .,Department of Pathology, Cancer Hospital of Tianjin Medical University, Tianjin, 300060, China. .,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, 300052, China.
| | - Xiulan Zhao
- Department of Pathology, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, China. .,Department of Pathology, General Hospital of Tianjin Medical University, Tianjin, 300052, China.
| |
Collapse
|
47
|
Wang W, Deng Z, Jin Z, Wu G, Wang J, Zhu H, Xu B, Wen Z, Guo Y. Bioinformatics analysis and experimental verification of five metabolism-related lncRNAs as prognostic models for hepatocellular carcinoma. Medicine (Baltimore) 2022; 101:e28694. [PMID: 35089224 PMCID: PMC8797488 DOI: 10.1097/md.0000000000028694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 01/04/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The number of patients with hepatocellular carcinoma (HCC) is showing a growing trend all over the world. The metabolic microenvironment has been shown to play a key role in the pathogenesis of HCC in recent studies. The expression of metabolites and metabolic processes in tumor cells can be regulated by gene regulation mediated by long non-coding RNAs (lncRNAs), the abnormal expression of which is closely related to tumor occurrence and metastasis. However, the fundamental mechanism of applying metabolism-related lncRNAs to predicting HCC is still unclear. METHODS With the complete RNA sequence data and clinical data obtained from The Cancer Genome Atlas database and metabolism-related genes downloaded from the Kyoto Encyclopedia of Genes and Genomes database, with false discovery rate < 0.001, log fold change > 1.5 selected as the screening criteria for lncRNA, the relationship between the expression level of metabolism-related LncRNAs (MRLs) and the overall survival rate was determined by the Univariate Cox regression analyses with the establishment of the metabolic prognosis model by the application of Multivariate Cox regression analyses, revealing the different biological processes and signaling pathways in both high-risk groups and low-risk groups by Gene Ontology, Kyoto Encyclopedia of Genes and Genomes enrichment analysis, and gene set enrichment analysis, leading the expression of lncRNA to be assessed by the reverse transcription-polymerase chain reaction results. RESULTS The overall survival rate of HCC patients is significantly correlated with signature of 5-MRLs. The prognosis characteristics of lncRNA reveal the relatively high death rate of patients in the high-risk groups, with the predicted signals by functional and pathway enrichment analysis related to biosynthesis, metabolic process, and metabolic pathway, with the prognostic characteristics of 5-MRLs by the combined analysis showing that it is an independent factor of HCC superior to the traditional clinical indicators in predicting the prognosis. A trend of high-expression was shown in MRLs in tumors by reverse transcription-polymerase chain reaction. CONCLUSION The new 5-MRLs as potential biomarkers provide more powerful prognostic information for HCC patients. In the future clinical treatment of HCC, it will provide doctors with more methods.
Collapse
Affiliation(s)
- Wei Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhenfeng Deng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zongrui Jin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Guolin Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jilong Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hai Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Banghao Xu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhang Wen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ya Guo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
48
|
Gouda G, Gupta MK, Donde R, Behera L, Vadde R. Metabolic pathway-based target therapy to hepatocellular carcinoma: a computational approach. THERANOSTICS AND PRECISION MEDICINE FOR THE MANAGEMENT OF HEPATOCELLULAR CARCINOMA, VOLUME 2 2022:83-103. [DOI: 10.1016/b978-0-323-98807-0.00003-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
|
49
|
Zhang Y, Zhai Z, Duan J, Wang X, Zhong J, Wu L, Li A, Cao M, Wu Y, Shi H, Zhong J, Guo Z. Lactate: The Mediator of Metabolism and Immunosuppression. Front Endocrinol (Lausanne) 2022; 13:901495. [PMID: 35757394 PMCID: PMC9218951 DOI: 10.3389/fendo.2022.901495] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/27/2022] [Indexed: 11/23/2022] Open
Abstract
The Warburg effect, one of the hallmarks of tumors, produces large amounts of lactate and generates an acidic tumor microenvironment via using glucose for glycolysis. As a metabolite, lactate not only serves as a substrate to provide energy for supporting cell growth and development but also acts as an important signal molecule to affect the biochemical functions of intracellular proteins and regulate the biological functions of different kinds of cells. Notably, histone lysine lactylation (Kla) is identified as a novel post-modification and carcinogenic signal, which provides the promising and potential therapeutic targets for tumors. Therefore, the metabolism and functional mechanism of lactate are becoming one of the hot fields in tumor research. Here, we review the production of lactate and its regulation on immunosuppressive cells, as well as the important role of Kla in hepatocellular carcinoma. Lactate and Kla supplement the knowledge gap in oncology and pave the way for exploring the mechanism of oncogenesis and therapeutic targets. Research is still needed in this field.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Zhao Zhai
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jiali Duan
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xiangcai Wang
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jinghua Zhong
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Longqiu Wu
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - An Li
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Miao Cao
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yanyang Wu
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Huaqiu Shi
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- *Correspondence: Huaqiu Shi, ; Jianing Zhong, ; Zhenli Guo,
| | - Jianing Zhong
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, China
- *Correspondence: Huaqiu Shi, ; Jianing Zhong, ; Zhenli Guo,
| | - Zhenli Guo
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- *Correspondence: Huaqiu Shi, ; Jianing Zhong, ; Zhenli Guo,
| |
Collapse
|
50
|
Jia W, Liang S, Cheng B, Ling C. The Role of Cancer-Associated Fibroblasts in Hepatocellular Carcinoma and the Value of Traditional Chinese Medicine Treatment. Front Oncol 2021; 11:763519. [PMID: 34868982 PMCID: PMC8636329 DOI: 10.3389/fonc.2021.763519] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/28/2021] [Indexed: 01/10/2023] Open
Abstract
Invasion and metastasis are the main reasons for the high mortality of liver cancer, which involve the interaction of tumor stromal cells and malignant cells. Cancer-associated fibroblasts (CAFs) are one of the major constituents of tumor stromal cells affecting tumor growth, invasion, and metastasis. The heterogeneous properties and sources of CAFs make both tumor-supporting and tumor-suppression effects possible. The mechanisms for CAFs in supporting hepatocellular carcinoma (HCC) progression can be categorized into upregulated aggressiveness and stemness, transformed metabolism toward glycolysis and glutamine reductive carboxylation, polarized tumor immunity toward immune escape of HCC cells, and increased angiogenesis. The tumor-suppressive effect of fibroblasts highlights the functional heterogenicity of CAF populations and provides new insights into tumor–stromal interplay mechanisms. In this review, we introduced several key inflammatory signaling pathways in the transformation of CAFs from normal stromal cells and the heterogeneous biofunctions of activated CAFs. In view of the pleiotropic regulation properties of traditional Chinese medicine (TCM) and heterogeneous effects of CAFs, we also introduced the application and values of TCM in the treatment of HCC through targeting CAFs.
Collapse
Affiliation(s)
- Wentao Jia
- School of Traditional Chinese Medicine, Naval Medical University, Shanghai, China
| | - Shufang Liang
- Department of Traditional Chinese Medicine, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Binbin Cheng
- School of Traditional Chinese Medicine, Naval Medical University, Shanghai, China
| | - Changquan Ling
- School of Traditional Chinese Medicine, Naval Medical University, Shanghai, China
| |
Collapse
|