1
|
Cho JH, Kim HG, Huang M, Wang S, Liu S, Lu A, McCrocklin K, Zhang Y, Fang Z, Wang J, Liu W, Wan J, Dong XC. The Patatin-Like Phospholipase Domain-Containing 3 148M Variant Exacerbates Alcohol-Induced Liver Injury and Tumorigenesis in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00154-3. [PMID: 40350061 DOI: 10.1016/j.ajpath.2025.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/02/2025] [Accepted: 04/22/2025] [Indexed: 05/14/2025]
Abstract
Patatin-like phospholipase domain-containing 3 (PNPLA3) protein 148M variant is strongly associated with cirrhosis and hepatocellular carcinoma (HCC); however, the underlying mechanisms remain elusive. This study aimed to elucidate the role of the PNPLA3148M variant in alcohol-related HCC development. Control and humanized PNPLA3148M transgenic mice were fed with an ethanol-containing diet for 12 weeks. The animals were examined for liver tumors. After the alcohol feeding, the PNPLA3148M mice had twofold higher liver cancer incidence rates and larger tumor sizes than those in the control mice. Cancer stem cell markers in the PNPLA3148M mouse livers were elevated relative to those in the control mouse livers. Alcohol detoxification was impaired in the PNPLA3148M mouse livers. Hepatic oxidative stress and DNA damage were elevated in the PNPLA3148M mice. Wnt/β-catenin and Yes-associated protein (YAP) and WW domain-containing transcription regulator 1 (TAZ) were activated in the PNPLA3148M mouse livers. The data suggest that the PNPLA3148M variant has a strong interaction with alcohol in HCC development through attenuation of alcohol detoxification and promotion of oncogenic pathways. Targeting the PNPLA3148M variant might be useful for the prevention or treatment of alcohol-associated HCC in patients carrying this variant.
Collapse
Affiliation(s)
- Jung-Hyo Cho
- Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, Indiana; Department of East and West Cancer Center, Daejeon Korean Medicine Hospital of Daejeon University, Daejeon, Republic of Korea
| | - Hyeong-Geug Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Menghao Huang
- Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, Indiana; Center for Diabetes and Metabolic Diseases, Indiana University, Indianapolis, Indiana; Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana
| | - Shen Wang
- Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Sheng Liu
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana; Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Alex Lu
- Park Tudor School, Indianapolis, Indiana
| | - Kyle McCrocklin
- Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, Indiana
| | - Yang Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Zhigang Fang
- Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Juexin Wang
- Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, Indiana; Center for Computational Biology and Bioinformatics, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Wanqing Liu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan
| | - Jun Wan
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana; Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, Indiana; Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, Indiana; Center for Computational Biology and Bioinformatics, School of Medicine, Indiana University, Indianapolis, Indiana
| | - X Charlie Dong
- Department of Biochemistry and Molecular Biology, School of Medicine, Indiana University, Indianapolis, Indiana; Center for Diabetes and Metabolic Diseases, Indiana University, Indianapolis, Indiana; Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis, Indiana; Luddy School of Informatics, Computing, and Engineering, Indiana University, Indianapolis, Indiana; Center for Computational Biology and Bioinformatics, School of Medicine, Indiana University, Indianapolis, Indiana.
| |
Collapse
|
2
|
Kondo Y, Ohashi S, Katada C, Nakai Y, Yamamoto Y, Tamaoki M, Kikuchi O, Yamada A, Hirohashi K, Mitani Y, Kataoka S, Saito T, Vu THN, Kondo T, Uneno Y, Sunami T, Yokoyama A, Matsubara J, Matsuda T, Naganuma S, Oryu K, Flashner S, Shimonosono M, Nakagawa H, Muto M. Aldh2 and the tumor suppressor Trp53 play important roles in alcohol-induced squamous field cancerization. J Gastroenterol 2025; 60:546-560. [PMID: 39909947 PMCID: PMC12014750 DOI: 10.1007/s00535-024-02210-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/30/2024] [Indexed: 02/07/2025]
Abstract
BACKGROUND Field cancerization defined by multiple development of squamous cell carcinomas (SCCs) in upper aerodigestive tract was explained by excessive alcohol intake. A dysfunctional mitochondrial aldehyde dehydrogenase 2 (Aldh2) delays the clearance of acetaldehyde, a genotoxic alcohol metabolite, and increases SCC risks. TP53 plays key roles in squamous carcinogenesis. However, the mechanism of alcohol-mediated squamous field cancerization has not been clearly elucidated. METHODS We developed a novel genetically engineered mouse strain KTPA-/- (Krt5CreERT2; Trp53loxp/loxp; Aldh2-/-) featuring Aldh2-loss concurrent with epithelial-specific Trp53 deletion. These mice were given 10%-EtOH, and we evaluated the development of squamous cell carcinogenesis histologically and genetically. RESULTS Widespread multifocal rete ridges (RRs), characterized by downward growth of proliferative preneoplastic cells, were found only in Aldh2+/- and Aldh2-/- mice with keratin5-specific Trp53 deletion (KTPA+/- and KTPA-/- mice, respectively), and alcohol drinking apparently increased RR formation rate. SCC occurred only in KTPA-/- (Aldh2 loss/TP53 loss) mice with alcohol drinking (15/18: 83%). Total alcohol consumption volume was significantly higher in KTPA-/- (Aldh2 loss/TP53 loss) mice with SCCs than those without SCCs. Further, target sequence revealed the occurrence of genetic abnormalities including Trp53 mutations in the esophageal epithelium of Aldh2-/- mice with alcohol drinking, suggesting direct mutagenic effects of alcohol drinking to the esophageal epithelium. CONCLUSION This study provides for the first time the evidence that alcohol drinking, Aldh2 dysfunction and Trp53 loss cooperate in squamous field cancerization. Alcohol consumption volume affects the SCCs development, even in the same genotype.
Collapse
Affiliation(s)
- Yuki Kondo
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
- Department of Gastroenterology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Shinya Ohashi
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan.
- Preemptive Medicine and Lifestyle Disease Research Center, Kyoto University Hospital, Kyoto, Japan.
| | - Chikatoshi Katada
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Yukie Nakai
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Yoshihiro Yamamoto
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Masashi Tamaoki
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Osamu Kikuchi
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Atsushi Yamada
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Kenshiro Hirohashi
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Yosuke Mitani
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Shigeki Kataoka
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Tomoki Saito
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Trang H Nguyen Vu
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Tomohiro Kondo
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Yu Uneno
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Tomohiko Sunami
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Akira Yokoyama
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Junichi Matsubara
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Tomonari Matsuda
- Environment Health Division, Kyoto University Graduate School of Engineering, Kyoto, Japan
| | - Seiji Naganuma
- Faculty of Health Sciences, Department of Medical Laboratory Science, Kochi Gakuen University, Kochi, Japan
| | - Kohei Oryu
- Faculty of Health Sciences, Department of Nutrition, Kochi Gakuen University, Kochi, Japan
| | - Samuel Flashner
- Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, USA
| | - Masataka Shimonosono
- Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, USA
| | - Hiroshi Nakagawa
- Division of Digestive and Liver Diseases, Department of Medicine and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, USA
| | - Manabu Muto
- Department of Medical Oncology, Kyoto University Graduate School of Medicine, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| |
Collapse
|
3
|
Maccioni L, Guan Y, Kim M, Parra MA, Peiffer B, Fu Y, Wang Y, Lin YH, Mackowiak B, Feng D, Cameron A, Sun Z, Kunos G, Stärkel P, Gao B. Opposite regulation of intestinal and intrahepatic CD8 + T cells controls alcohol-associated liver disease progression. Gut 2025:gutjnl-2024-334412. [PMID: 40199574 DOI: 10.1136/gutjnl-2024-334412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/15/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND Gut-liver crosstalk plays an important role in alcohol-associated liver disease (ALD) pathogenesis; but underlying mechanisms remain obscure. OBJECTIVE We examined the regulation of intestinal and intrahepatic CD8+ T lymphocytes and their contribution to ALD. DESIGN ALD patients were recruited for evaluation of intestinal and liver T cells. Single-cell RNA sequencing (scRNA seq) was performed to analyse intrahepatic and peripheral T cells in ALD. Wildtype, CD8-specific Bcl2 transgenic (Cd8 Bcl-2), and Cd8 -/- mice were subjected to chronic-plus-binge ethanol feeding. RESULTS In ALD patients, duodenal CD8+ T cells were selectively reduced and negatively correlated with liver injury and bacterial translocation markers, while intrahepatic CD8+ T cells were markedly increased. ScRNA seq analysis of ALD patient livers revealed several populations of CD8+ T cells expressing activation and survival genes (eg, Bcl2). Transcriptomics and functional studies revealed a key role of prosurvival BCL2 in this opposite regulation of CD8+ T cells. Mechanistically, chronic-plus-binge ethanol feeding reduced CD8+ T cells specifically in the duodenum where ethanol levels are high. Inducing BCL2 in CD8+ T cells reversed ethanol-induced loss of duodenal CD8+ T cells, improved gut barrier function and ameliorated ALD, while CD8 deficiency was linked to enhanced neutrophil and macrophage infiltration in the liver, exacerbating ALD in mice. CONCLUSIONS ALD is associated with loss of duodenal CD8+ T cells but elevation of intrahepatic CD8+ T cells, which aggravates and ameliorates ALD, respectively. Restoration of survival and functions of intestinal and intrahepatic CD8+ T cells may represent a novel therapeutic strategy for ALD patients.
Collapse
Affiliation(s)
- Luca Maccioni
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Yukun Guan
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Mariia Kim
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, District of Columbia, USA
| | - Maria A Parra
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Brandon Peiffer
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yaojie Fu
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Yang Wang
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Yu-Hong Lin
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Bryan Mackowiak
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Andrew Cameron
- Surgery - Division of Transplant Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zhaoli Sun
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - George Kunos
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Peter Stärkel
- Department of Hepato-Gastroenterology, Cliniques universitaires Saint-Luc, Bruxelles, Belgium
- Laboratory of Hepato-gastroenterology (GAEN), Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| |
Collapse
|
4
|
Lin YH, Lee YC, Liao JB, Yu PL, Chou CY, Yang YF. Alda‑1 restores ALDH2‑mediated alcohol metabolism to inhibit the NF‑κB/VEGFC axis in head and neck cancer. Int J Mol Med 2025; 55:55. [PMID: 39886980 PMCID: PMC11819766 DOI: 10.3892/ijmm.2025.5496] [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: 08/30/2024] [Accepted: 12/12/2024] [Indexed: 02/01/2025] Open
Abstract
The adaptation of cancer cells to hostile environments often necessitates metabolic pathway alterations to sustain proliferation and invasion. Head and neck cancer (HNC) has unfavorable outcomes. Therefore, elucidating the functional effects and molecular mechanisms underlying metabolic changes is key. Ingenuity Pathway Analysis identified 'ethanol degradation pathway II and IV' was consistently downregulated in tumor tissue, with aldehyde dehydrogenase 2 (ALDH2) emerging as a key prognostic gene among the top‑ranked differentially expressed metabolic pathways. Immunohistochemistry (IHC) of HNC specimens revealed significant downregulation of ALDH2 expression in tumor tissue, which was inversely correlated with T classification, overall stage, recurrence rate and independently predicted poor prognosis. Functional assays showed that ALDH2 knockdown enhanced HNC cell migration, invasion and colony formation, while ALDH2 overexpression attenuated these processes. Mechanistically, ALDH2 downregulation and subsequent reactive oxygen species (ROS) production in cells activated NF‑κB, upregulating vascular endothelial growth factor C (VEGFC) expression. ALDH2 overexpression inhibited ROS production and the NF‑κB/VEGFC oncogenic pathway, with pharmacological inhibition of NF‑κB and VEGFC mitigating the enhanced migration and invasion of ALDH2‑knockdown HNC cells. IHC and transcriptome analysis further highlighted an inverse association between ALDH2 and VEGFC, with the ALDH2high/VEGFClow profile predicting the most favorable survival outcome. Inhibition of ALDH2 with Daidzin increased VEGFC and phosphorylated NF‑κB levels, restoring the migration and invasion of ALDH2‑overexpressing HNC cells by enhancing the effects of VEGFC. Notably, modulating ALDH2 activity using Alda‑1 ameliorated NF‑kB/VEGFC axis upregulation following acetaldehyde treatment, aligning with the aforementioned alterations in alcohol metabolisms. These findings emphasize the key role of ALDH2 in influencing HNC progression and patient outcome, suggesting that targeting the ALDH2/NF‑κB/VEGFC pathway may represent a potential therapeutic strategy for HNC.
Collapse
Affiliation(s)
- Yu-Hsuan Lin
- Department of Otolaryngology, Head and Neck Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan, R.O.C
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung 804201, Taiwan, R.O.C
| | - Yi-Chen Lee
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan, R.O.C
| | - Jia-Bin Liao
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan, R.O.C
| | - Pei-Lun Yu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan, R.O.C
| | - Chih-Yu Chou
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan, R.O.C
| | - Yi-Fang Yang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan, R.O.C
| |
Collapse
|
5
|
Wang Q, Jia W, Liu J, Zhao Q, Yang Z. Global, regional, and national burden of liver cancer due to alcohol use, 1990-2021: results from the Global Burden of Disease study 2021. Eur J Gastroenterol Hepatol 2025; 37:466-476. [PMID: 39621868 DOI: 10.1097/meg.0000000000002899] [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] [Indexed: 01/03/2025]
Abstract
BACKGROUND Liver cancer is a major global health burden, with alcohol use being a well-established risk factor. This study aims to analyze the global, regional, and national incidence, prevalence, mortality, and disability-adjusted life years (DALYs) attributable to liver cancer due to alcohol use from 1990 to 2021. METHODS Data on liver cancer due to alcohol use were collected from the 2021 Global Burden of Disease (GBD) study. The changing trend of liver cancer among alcohol users was described using the linear regression model. In addition, we employed a hierarchical cluster analysis to study the evolving patterns across diverse GBD regions and conducted a frontier analysis to explore the nexus between the burden and sociodemographic progress. RESULTS In 2021, alcohol-related liver cancer globally accounted for 99 544 incidence cases, 132 033 prevalence cases, 92 228 death cases, and 2 316 027 DALYs cases. Males and middle-aged adults emerged as high-risk populations, while regions with a higher sociodemographic index (SDI) were identified as high-risk areas. From 1990 to 2021, both the number of cases and age-standardized rates (ASRs) increased. Our frontier analysis revealed unattained health gains between 1990 and 2021, highlighting disparities in disease burden among countries with varying SDI levels. This analysis further demonstrated an inverse correlation between SDI and alcohol-related liver cancer ASRs, with the ASRs stabilizing once the SDI exceeded 0.40. CONCLUSION Alcohol use is a significant contributor to the global burden of liver cancer. Comprehensive policies and interventions targeting alcohol use are needed to reduce the burden of alcohol-related liver cancer.
Collapse
Affiliation(s)
- Qihong Wang
- Department of Digestive Endoscopy, General Hospital of Northern Theater Command, Shenyang, Liaoning Province, China
| | | | | | | | | |
Collapse
|
6
|
Liu W, Hu K, Fu Y, Zhou T, Zhong Q, Wang W, Gui Y, Zhang P, Yao D, Yang X, Zhu W, Liu Z, Luo D, Xiao Y. Identification of methionine metabolism related prognostic model and tumor suppressive functions of BHMT in hepatocellular carcinoma. Sci Rep 2025; 15:9250. [PMID: 40102459 PMCID: PMC11920202 DOI: 10.1038/s41598-025-93650-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 03/07/2025] [Indexed: 03/20/2025] Open
Abstract
Given the resistance to conventional treatments and limitations of immune checkpoint blockade therapy in hepatocellular carcinoma (HCC), it is imperative to explore novel prognostic models and biomarkers. The dependence of cancer cell on exogenous methionine, known as Hoffman effect, is a hallmark of HCC, with numerous studies reporting a strong correlation between methionine metabolism and tumor development. Betaine-homocysteine S-methyltransferase (BHMT), a critical component of methionine metabolism pathway, has polymorphisms linking to poor prognosis in multiple cancers. Nevertheless, there is little literature regarding the relationship between methionine metabolism and incidence, mortality of HCC, as well as the function of BHMT in HCC progression. In this study, by analyzing multiple datasets, we constructed a methionine metabolism-related prognostic model and thoroughly investigated the influence of BHMT on the prognosis of HCC. Bioinformatics analysis revealed a marked decrease in BHMT expression in HCC, which was linked to adverse clinical outcomes. CIBERSORT results suggest that BHMT promotes infiltration of M1 macrophages. Our results suggest its potential as an ideal prognostic biomarker for anti PD-L1 immunotherapy. In summary, this study innovatively provides first methionine metabolism-related prognostic model and unveils the tumor suppressive function of BHMT in HCC, providing potential mechanism by which BHMT exert its function.
Collapse
Affiliation(s)
- Wenli Liu
- Department of Pathology, Infectious Diseases Hospital of Nanchang University, Nanchang, 330001, Jiangxi, China
| | - Kaiheng Hu
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yaqing Fu
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Tianmin Zhou
- Department of Pathology, Infectious Diseases Hospital of Nanchang University, Nanchang, 330001, Jiangxi, China
| | - Qingmei Zhong
- Department of Pathology, Infectious Diseases Hospital of Nanchang University, Nanchang, 330001, Jiangxi, China
| | - Wu Wang
- Department of Pathology, Infectious Diseases Hospital of Nanchang University, Nanchang, 330001, Jiangxi, China
| | - Yang Gui
- Department of Pathology, Infectious Diseases Hospital of Nanchang University, Nanchang, 330001, Jiangxi, China
| | - Ping Zhang
- Department of Pathology, Infectious Diseases Hospital of Nanchang University, Nanchang, 330001, Jiangxi, China
| | - Di Yao
- Department of Pathology, Infectious Diseases Hospital of Nanchang University, Nanchang, 330001, Jiangxi, China
| | - Xiaohong Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Weifeng Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Zhuoqi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Yingqun Xiao
- Department of Pathology, Infectious Diseases Hospital of Nanchang University, Nanchang, 330001, Jiangxi, China.
| |
Collapse
|
7
|
Liu J, Ma S, Deng D, Yang Y, Li J, Zhang Y, Yin P, Shang D. Multi-Omics Profiling Reveals Glycerolipid Metabolism-Associated Molecular Subtypes and Identifies ALDH2 as a Prognostic Biomarker in Pancreatic Cancer. Metabolites 2025; 15:207. [PMID: 40137171 PMCID: PMC11943634 DOI: 10.3390/metabo15030207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
Background: The reprogramming of lipid metabolism, especially glycerolipid metabolism (GLM), plays a key role in cancer progression and response to therapy. However, the role and molecular characterization of GLM in pancreatic cancer (PC) remain unclear. Methods: A pan-cancer analysis of glycerolipid metabolism-related genes (GMRGs) was first conducted to assess copy-number variants, single-nucleotide variations, methylation, and mRNA expression. Subsequently, GLM in PC was characterized using lipidomics, single-cell RNA sequencing (scRNA-seq), and spatial transcriptomic analysis. A cluster analysis based on bulk RNA sequencing data from 930 PC samples identified GLM-associated subtypes, which were then analyzed for differences in prognosis, biological function, immune microenvironment, and drug sensitivity. To prioritize prognostically relevant GMRGs in PC, we employed a random forest (RF) algorithm to rank their importance across 930 PC samples. Finally, the key biomarker of PC was validated using PCR and immunohistochemistry. Results: Pan-cancer analysis identified molecular features of GMRGs in cancers, while scRNA-seq, spatial transcriptomics, and lipidomics highlighted GLM heterogeneity in PC. Two GLM-associated subtypes with significant prognostic, biofunctional, immune microenvironmental, and drug sensitivity differences were identified in 930 PC samples. Finally, ALDH2 was identified as a novel prognostic biomarker in PC and validated in a large number of datasets and clinical samples. Conclusions: This study highlights the crucial role of GLM in PC and defines a new PC subtype and prognostic biomarker. These findings establish a novel avenue for studying prognostic prediction and precision medicine in PC patients.
Collapse
Affiliation(s)
- Jifeng Liu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (J.L.); (S.M.); (Y.Y.); (J.L.)
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China;
| | - Shurong Ma
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (J.L.); (S.M.); (Y.Y.); (J.L.)
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China;
| | - Dawei Deng
- Department of Hepato-Biliary-Pancreas, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China;
| | - Yao Yang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (J.L.); (S.M.); (Y.Y.); (J.L.)
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| | - Junchen Li
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (J.L.); (S.M.); (Y.Y.); (J.L.)
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| | - Yunshu Zhang
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China;
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| | - Peiyuan Yin
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (J.L.); (S.M.); (Y.Y.); (J.L.)
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China;
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| | - Dong Shang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China; (J.L.); (S.M.); (Y.Y.); (J.L.)
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116000, China;
- Institute of Integrative Medicine, Dalian Medical University, Dalian 116000, China
| |
Collapse
|
8
|
Liu T, Zhang F, Feng Y, Han P, Gao Y. Alcohol-Metabolizing Enzymes, Liver Diseases and Cancer. Semin Liver Dis 2025; 45:99-113. [PMID: 40157374 PMCID: PMC12031026 DOI: 10.1055/a-2551-3320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
Alcohol is generally believed to be metabolized in the liver by alcohol dehydrogenase (ADH), aldehyde dehydrogenase (ALDH), and to a much lesser extent cytochrome P450 2E1 (CYP2E1) and other enzymes. Recent studies suggest that gut also play important roles in the promotion of alcohol metabolism. ADH, ALDH, and CYP2E1 have several polymorphisms that markedly impact alcohol metabolism. These alcohol-metabolizing enzymes not only affect alcohol-associated liver disease (ALD), but may also modulate the pathogenesis of other liver diseases and cancer in the absence of alcohol consumption. In this review, we discuss alcohol metabolism and the roles of alcohol-metabolizing enzymes in the pathogenesis of ALD, metabolic dysfunction-associated steatotic liver disease, metabolic dysfunction and alcohol-associated liver disease, viral hepatitis, and liver cancer. We also discuss how alcohol-metabolizing enzymes may affect endogenous ethanol production, and how ethanol metabolism in the gut affects liver disease and cancer. Directions for future research on the roles of alcohol-metabolizing enzymes in liver disease and cancer are also elaborated.
Collapse
Affiliation(s)
- Tao Liu
- Department of Hepatology, Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, China
- China-Singapore Belt and Road Joint Laboratory on Liver Disease Research, Changchun, China
- Jilin Provincial Key Laboratory of Metabolic Liver Diseases, Jilin University, Changchun, China
| | - FeiYu Zhang
- Department of Hepatology, Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, China
- China-Singapore Belt and Road Joint Laboratory on Liver Disease Research, Changchun, China
- Jilin Provincial Key Laboratory of Metabolic Liver Diseases, Jilin University, Changchun, China
| | - Yue Feng
- Department of Hepatology, Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, China
- China-Singapore Belt and Road Joint Laboratory on Liver Disease Research, Changchun, China
- Jilin Provincial Key Laboratory of Metabolic Liver Diseases, Jilin University, Changchun, China
| | - PanShiLi Han
- Department of Hepatology, Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, China
- China-Singapore Belt and Road Joint Laboratory on Liver Disease Research, Changchun, China
- Jilin Provincial Key Laboratory of Metabolic Liver Diseases, Jilin University, Changchun, China
| | - YanHang Gao
- Department of Hepatology, Center of Infectious Diseases and Pathogen Biology, The First Hospital of Jilin University, Changchun, China
- China-Singapore Belt and Road Joint Laboratory on Liver Disease Research, Changchun, China
- Jilin Provincial Key Laboratory of Metabolic Liver Diseases, Jilin University, Changchun, China
| |
Collapse
|
9
|
Wu Q, Yang D, Liu C, Xu T. Alcohol Plus Additional Risk Factors: Rodent Model of Liver Injury. Semin Liver Dis 2025; 45:81-98. [PMID: 39719149 DOI: 10.1055/a-2490-4278] [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] [Indexed: 12/26/2024]
Abstract
Alcohol-associated liver disease (ALD), primarily caused by chronic excessive alcohol consumption, is a leading cause of chronic liver disease worldwide. ALD includes alcohol-associated steatotic liver, alcohol-associated hepatitis (AH), fibrosis, cirrhosis, and can even progress to hepatocellular carcinoma (HCC). Existing research indicates that the risk factors of ALD are quite numerous. In addition to drinking patterns, factors such as aldehyde dehydrogenase 2 (ALDH2) deficiency, smoking, medication administration, high-fat diet (HFD), hepatitis virus infection, and disruption of circadian rhythms can also increase susceptibility to ALD. However, there is limited understanding regarding the exacerbation of liver injury by alcohol plus additional risk factors. This review presents rodent models of EtOH + "X," which simulate the synergistic effects of alcohol and additional risk factors in causing liver injury. These models offer a further exploration of the interactions between alcohol and additional risk factors, advancing the simulation of human ALD and providing a more reliable platform for studying disease mechanisms and exploring therapeutic interventions. We summarize the modeling methods, relevant indicators of liver injury, and focus on the targets of the synergistic effects as well as the associated mechanisms.
Collapse
Affiliation(s)
- Qixiang Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, P.R. China
- Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Dashuai Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, P.R. China
- Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Chixiang Liu
- Department of Blood Transfusion, Southern Medical University, Nanfang Hospital, Guangzhou, P.R. China
- School of Laboratory and Biotechnology, Institute of Antibody Engineering, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, P.R. China
- Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, P.R. China
| |
Collapse
|
10
|
Luo X, McAndrews KM, Kalluri R. Natural and Bioengineered Extracellular Vesicles in Diagnosis, Monitoring and Treatment of Cancer. ACS NANO 2025; 19:5871-5896. [PMID: 39869032 PMCID: PMC12002402 DOI: 10.1021/acsnano.4c11630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Extracellular vesicles (EVs) are cell derived nanovesicles which are implicated in both physiological and pathological intercellular communication, including the initiation, progression, and metastasis of cancer. The exchange of biomolecules between stromal cells and cancer cells via EVs can provide a window to monitor cancer development in real time for better diagnostic and interventional strategies. In addition, the process of secretion and internalization of EVs by stromal and cancer cells in the tumor microenvironment (TME) can be exploited for delivering therapeutics. EVs have the potential to provide a targeted, biocompatible, and efficient delivery platform for the treatment of cancer and other diseases. Natural as well as engineered EVs as nanomedicine have immense potential for disease intervention. Here, we provide an overview of current knowledge of EVs' function in cancer progression, diagnostic and therapeutic applications for EVs in the cancer setting, as well as current EV engineering strategies.
Collapse
Affiliation(s)
- Xin Luo
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
- Department of Bioengineering, Rice University, Houston, Texas 77005, United States
| | - Kathleen M. McAndrews
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Raghu Kalluri
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
- Department of Bioengineering, Rice University, Houston, Texas 77005, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, United States
| |
Collapse
|
11
|
Liu M, Zhou M, Ren X, Xie Y. Establishment and application of murine models of alcoholic liver disease: A narrative review. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025; 49:271-284. [PMID: 39715699 DOI: 10.1111/acer.15520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/29/2024] [Indexed: 12/25/2024]
Abstract
In recent years, there have been significant advances in pathological research on alcoholic liver disease (ALD), with suitable animal models making a significant contribution. However, the currently established animal ALD models still have some significant drawbacks, especially the inability to induce the entire human ALD lineage, which may be related to physiological differences between animals and humans. This review comprehensively summarized the most widely used experimental models of ALD, including voluntary drinking, Lieber-DeCarli, Meadows-Cook, Tsukamoto-French, NIAAA, and the "second hit" model. "Second hit" refers to an additional factor that damages the liver. There are various "second hit" models that fall into two main categories: particular diets and drugs. These models can either simulate human drinking patterns more accurately or produce varying degrees of ALD without significantly increasing animal mortality. We introduced the established method of the original models, discussed the advantages and disadvantages of the existing models from the aspects of operability and practicality, and provided existing improvement methods, hoping to provide a reference for future researchers.
Collapse
Affiliation(s)
- Mengsi Liu
- Peking University People's Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Mingying Zhou
- Peking University People's Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Xueyi Ren
- Peking University People's Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Yandi Xie
- Peking University People's Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| |
Collapse
|
12
|
Xu B, Luo Z, Niu X, Li Z, Lu Y, Li J. Fungi, immunosenescence and cancer. Semin Cancer Biol 2025; 109:67-82. [PMID: 39788169 DOI: 10.1016/j.semcancer.2025.01.002] [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: 11/05/2024] [Revised: 12/22/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
Fungal microbes are a small but immunoreactive component of the human microbiome, which may influence cancer development, progression and therapeutic response. Immunosenescence is a process of immune dysfunction that occurs with aging, including lymphoid organ remodeling, contributing to alterations in the immune system in the elderly, which plays a critical role in many aspects of cancer. There is evidence for the interactions between fungi and immunosenescence in potentially regulating cancer progression and remodeling the tumor microenvironment (TME). In this review, we summarize potential roles of commensal and pathogenic fungi in modulating cancer-associated processes and provide more-detailed discussions on the mechanisms of which fungi affect tumor biology, including local and distant regulation of the TME, modulating antitumor immune responses and interactions with neighboring bacterial commensals. We also delineate the features of immunosenescence and its influence on cancer development and treatment, and highlight the interactions between fungi and immunosenescence in cancer. We discuss the prospects and challenges for harnessing fungi and immunosenescence in cancer diagnosis and/or treatment. Considering the limited understanding and techniques in conducting such research, we also provide our view on how to overcome challenges faced by the exploration of fungi, immunosenescence and their interactions on tumor biology.
Collapse
Affiliation(s)
- Bin Xu
- Jiangxi Health Committee Key (JHCK) Laboratory of Tumor Metastasis, Jiangxi Key Laboratory of Oncology, Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi 330029, China
| | - Zan Luo
- Jiangxi Health Committee Key (JHCK) Laboratory of Tumor Metastasis, Jiangxi Key Laboratory of Oncology, Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi 330029, China
| | - Xing Niu
- Experimental Center of BIOQGene, YuanDong International Academy of Life Sciences, 999077, China; Voylin Institute for Translation Medicine, Xiamen, Fujian 361000, China
| | - Zhi Li
- Jiangxi Health Committee Key (JHCK) Laboratory of Tumor Metastasis, Jiangxi Key Laboratory of Oncology, Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Gannan Medical University, Ganzhou, Jiangxi 341000, China
| | - Yeping Lu
- Department of Neurosurgery, The Fifth Hospital of Wuhan, Wuhan, Hubei 430050, China.
| | - Junyu Li
- Department of Radiation Oncology, Jiangxi Key Laboratory of Oncology, Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi 330029, China; The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
13
|
Xu Z, Gao Y, Yu Z, Zhang R, Wang R, Li S, Wang S, Du B, Tian Z, Huang L, Ding Z, Yuan J. Transcriptome analysis of liver injury of fatty liver disease induced by ALDH2 deficiency. Sci Rep 2025; 15:2487. [PMID: 39833331 PMCID: PMC11746932 DOI: 10.1038/s41598-025-86547-1] [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: 04/16/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
Aldehyde dehydrogenase 2 (Aldh2) Glu504Lys mutation, common in East Asians, is linked to various alcohol-related pathologies, notably fatty liver disease. Recent findings suggest that high ethanol-producing Klebsiella pneumoniae(HiAlc Kpn) exacerbates liver injury in non-alcoholic fatty liver disease (NAFLD). Our study investigated the combined effects of Aldh2 deficiency and HiAlc Kpn on NAFLD liver injury, transcriptome analyses to unearth potential mechanisms and therapeutic targets. In our controlled experiment with Aldh2-deficient mice, we induced fatty liver via alcohol and HiAlc Kpn gavage, followed by comprehensive analyses to detect gene expression and epigenetic changes. The results showed that Aldh2-deficient mice were particularly vulnerable to ethanol and HiAlc Kpn, with notable gene expression changes in key metabolic and liver injury pathways. Our analysis identified crucial differentially expressed genes (DEGs) and pathways, highlighting the significant roles of genes like Cyp8b1, Cyp7a1, and Ugt2b1 in liver metabolism and suggesting them as therapeutic targets. The study underscores the impact of Aldh2 deficiency and HiAlc Kpn on NAFLD progression, revealing potential therapeutic strategies. Despite these insights, further research is needed to clarify the systemic effects on aldehyde metabolism and the full implications of Aldh2 deficiency and HiAlc Kpn in liver injury.
Collapse
Affiliation(s)
- Ziying Xu
- Capital Institute of Pediatrics, Beijing, China
| | - Yagang Gao
- Capital Institute of Pediatrics, Beijing, China
| | - Zihui Yu
- Capital Institute of Pediatrics, Beijing, China
| | - Rui Zhang
- Clinical Laboratory Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ruikun Wang
- Capital Institute of Pediatrics, Beijing, China
- Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, China
| | - Shang Li
- Department of Orthopedics, the Fourth Medical Center of Chinese PLA General Hospital, Beijing, China
| | | | - Bing Du
- Capital Institute of Pediatrics, Beijing, China
| | - Ziyan Tian
- Capital Institute of Pediatrics, Beijing, China
| | | | - Zanbo Ding
- Capital Institute of Pediatrics, Beijing, China
| | - Jing Yuan
- Capital Institute of Pediatrics, Beijing, China.
| |
Collapse
|
14
|
Zhang J, Wang R, Xie L, Ren H, Luo D, Yang Y, Xie H, Shang Z, Liu C. Pharmacological activation of aldehyde dehydrogenase 2 inhibits ferroptosis via SLC7A11/GPX4 axis to reduce kidney stone formation. Eur J Pharmacol 2025; 986:177132. [PMID: 39547408 DOI: 10.1016/j.ejphar.2024.177132] [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: 05/02/2024] [Revised: 10/23/2024] [Accepted: 11/13/2024] [Indexed: 11/17/2024]
Abstract
Calcium oxalate (CaOx) kidney stones pose a global health challenge due to their high prevalence and recurrence rates. While cell death mechanisms such as ferroptosis are known to play a crucial role in stone formation, the precise underlying mechanisms remain enigmatic. Aldehyde dehydrogenase 2 (ALDH2) is a metabolic enzyme of the ferroptosis product 4-hydroxy-2-nonenal (4-HNE). However, the function of ALDH2 in kidney stones is poorly understood. In this study, we observed a downregulation of ALDH2 in the stone group. Significantly, the administration of Alda-1, an ALDH2 agonist, notably reduced crystal deposition in the kidneys and hindered crystal adhesion to cells. Furthermore, Alda-1 induced upregulation of SLC7A11 expression, promoting glutathione synthesis, reducing lipid peroxidation accumulation, and lowering Fe2+ levels. These collective effects attenuated crystal-induced ferroptosis. However, the renoprotective effects of Alda-1 were inhibited by SLC7A11 siRNA. In conclusion, our study explores the applications of Alda-1 and highlights the potential of targeting ALDH2 as a promising therapeutical strategy for urolithiasis.
Collapse
Affiliation(s)
- Jingdong Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Rui Wang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Linguo Xie
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Haotian Ren
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Di Luo
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yu Yang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Haijie Xie
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhiqun Shang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| | - Chunyu Liu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| |
Collapse
|
15
|
Chiu IJ, Ajay AK, Chen CH, Jadhav S, Zhao L, Cao M, Ding Y, Shah KM, Shah SI, Hsiao LL. Suppression of aldehyde dehydrogenase 2 in kidney proximal tubules contributes to kidney fibrosis through Transforming Growth Factor-β signaling. Kidney Int 2025; 107:84-98. [PMID: 39393529 DOI: 10.1016/j.kint.2024.09.010] [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: 11/20/2023] [Revised: 08/29/2024] [Accepted: 09/20/2024] [Indexed: 10/13/2024]
Abstract
Chronic kidney disease (CKD) is an increasingly prevalent disorder that poses a significant global health and socioeconomic burden. East Asian countries such as China, Taiwan, Japan, and South Korea have a higher incidence and prevalence of kidney failure when compared to Western nations, and the reasons for this discrepancy remain unclear. Aldehyde dehydrogenase 2 (ALDH2) is an essential detoxifying enzyme for exogenous and endogenous aldehyde metabolism in mitochondria. Inactivating mutations at E504K and E487K are found in 35-45% of East Asian populations and has been linked to a higher risk of various disorders, including cardiovascular diseases and cancer. However, little is known about the role of ALDH2 in CKD. Here, we characterized the expression pattern of ALDH2 in normal and CKD human and mouse kidneys and demonstrated that ALDH2 expression was significantly reduced, and that the protein level was inversely correlated with the degree of CKD and fibrosis. Further, we treated ALDH2∗2 knock-in mice, a loss of ALDH2 function model, with aristolochic acid and found that these mice showed enhanced fibrosis. Moreover, ALDH2 deficiency was associated with kidney fibrosis involving epithelial cell differentiation process in vivo and in vitro. However, ALDH2 overexpression protected proximal tubule epithelial cells from transforming growth factor-β-induced dedifferentiation or partial epithelial-mesenchymal transdifferentiation in vitro. Thus, our findings yield important clinical information regarding the development and progression of CKD involving ALDH2, especially among East Asian populations.
Collapse
Affiliation(s)
- I-Jen Chiu
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Taipei Medical University Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei, Taiwan
| | - Amrendra K Ajay
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Che-Hong Chen
- Department of Chemical and Systems Biology, School of Medicine, Stanford University, Stanford, California, USA
| | - Shreyas Jadhav
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Li Zhao
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Minghua Cao
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yan Ding
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Discovery Biology, Merck & Co, Inc., Rahway, New Jersey, USA
| | - Kavya M Shah
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, USA; Department of Statistics, Harvard University, Cambridge, Massachusetts
| | - Sujal I Shah
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Li-Li Hsiao
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
16
|
Mollazadeh S, Saeedi N, Al-Asady AM, Ghorbani E, Khazaei M, Ryzhikov M, Avan A, Hassanian SM. Exploring Hepatocellular Carcinoma Pathogenesis: The Influence of Genetic Polymorphisms. Curr Pharm Des 2025; 31:432-442. [PMID: 39297458 DOI: 10.2174/0113816128327773240827062719] [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: 05/25/2024] [Accepted: 07/23/2024] [Indexed: 02/20/2025]
Abstract
Hepatocellular carcinoma (HCC) is influenced by several factors, among which genetic polymorphisms play a key role. Polymorphisms in various genes affect key pathways involved in HCC development, including metabolism, expression of inflammatory cytokines, cell proliferation, and apoptosis regulation. These polymorphisms induce differential effects on susceptibility to HCC, disease progression, and treatment outcomes. Understanding the effect of genetic variations on HCC pathogenesis is essential to elucidate underlying mechanisms and identify potential therapeutic targets. This review explores the diverse roles of genetic polymorphisms in HCC, providing insights into the complex interplay between genetic factors and disease development.
Collapse
Affiliation(s)
- Samaneh Mollazadeh
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Nikoo Saeedi
- Student Research Committee, Islamic Azad University, Mashhad Branch, Mashhad, Iran
| | | | - Elnaz Ghorbani
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mikhail Ryzhikov
- School of Medicine, Saint Louis University, Saint Louis, MO 63103, USA
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Human Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
17
|
Li YT, Zeng XZ. Establishment and Validation of the Novel Necroptosis-related Genes for Predicting Stemness and Immunity of Hepatocellular Carcinoma via Machine-learning Algorithm. Comb Chem High Throughput Screen 2025; 28:146-165. [PMID: 39641162 DOI: 10.2174/0113862073271292231108113547] [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: 07/26/2023] [Revised: 08/21/2023] [Accepted: 09/21/2023] [Indexed: 12/07/2024]
Abstract
BACKGROUND Necroptosis, a recently identified mechanism of programmed cell death, exerts significant influence on various aspects of cancer biology, including tumor cell proliferation, stemness, metastasis, and immunosuppression. However, the role of necroptosis-related genes (NRGs) in Hepatocellular Carcinoma (HCC) remains elusive. METHODS In this study, we assessed the mutation signature, copy number variation, and expression of 37 NRGs in HCC using the TCGA-LIHC dataset. We further validated our results using the ICGC-LIRI-JP dataset. To construct our prognostic model, we utilized the least absolute shrinkage and selection operator (LASSO), and evaluated the predictive efficacy of the NRGs-score using various machine learning algorithms, including K-M curves, time-ROC curves, univariate and multivariate Cox regression, and nomogram. In addition, we analyzed immune infiltration using the CIBERSOFT and ssGSEA algorithms, calculated the stemness index through the one-class logistic regression (OCLR) algorithm, and performed anti-cancer stem cells (CSCs) drug sensitivity analysis using oncoPredict. Finally, we validated the expression of the prognostic NRGs through qPCR both in vitro and in vivo. RESULTS About 18 out of 37 NRGs were found to be differentially expressed in HCC and correlated with clinical outcomes. To construct a prognostic model, six signature genes (ALDH2, EZH2, PGAM5, PLK1, SQSTM1, and TARDBP) were selected using LASSO analysis. These genes were then employed to categorize HCC patients into two subgroups based on NRGs-score (low vs. high). A high NRGs score was associated with a worse prognosis. Furthermore, univariate and multivariate Cox regression analyses were performed to confirm the NRGs-score as an independent risk factor. These analyses revealed strong associations between NRGs-score and critical factors, such as AFP, disease stage, and tumor grade in the HCC cohort. NRGs-score effectively predicted the 1-, 3-, and 5-year survival of HCC patients. Immune infiltration analysis further revealed that the expression of immune checkpoint molecules was significantly enhanced in the high NRGs-score group. Stemness analysis in the HCC cohort showed that NRGs-score was positively correlated with mRNA stemness index, and patients with high NRGs-score were sensitive to CSCs inhibitors. The findings from the external validation cohort provided confirmation that the NRGs-score presented a trait with universal applicability in accurately predicting the survival of HCC. Additionally, the six prognostic genes were consistently differentially expressed in both the HCC cell line and the mouse HCC model. CONCLUSION Our study demonstrated the pivotal role of NRGs in promoting stemness and immune suppression in HCC and established a robust model which could successfully predict HCC prognosis.
Collapse
Affiliation(s)
- Yao-Ting Li
- Department of Forensic Science, Guangdong Police College, 500 Binjiang East Road, Guangzhou 510230, Guangdong, China
| | - Xue-Zhen Zeng
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| |
Collapse
|
18
|
Xiang H, Kasajima R, Azuma K, Tagami T, Hagiwara A, Nakahara Y, Saito H, Igarashi Y, Wei F, Ban T, Yoshihara M, Nakamura Y, Sato S, Koizume S, Tamura T, Sasada T, Miyagi Y. Multi-omics analysis-based clinical and functional significance of a novel prognostic and immunotherapeutic gene signature derived from amino acid metabolism pathways in lung adenocarcinoma. Front Immunol 2024; 15:1361992. [PMID: 39735553 PMCID: PMC11671776 DOI: 10.3389/fimmu.2024.1361992] [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: 12/27/2023] [Accepted: 07/30/2024] [Indexed: 12/31/2024] Open
Abstract
Background Studies have shown that tumor cell amino acid metabolism is closely associated with lung adenocarcinoma (LUAD) development and progression. However, the comprehensive multi-omics features and clinical impact of the expression of genes associated with amino acid metabolism in the LUAD tumor microenvironment (TME) are yet to be fully understood. Methods LUAD patients from The Cancer Genome Atlas (TCGA) database were enrolled in the training cohort. Using least absolute shrinkage and selection operator Cox regression analysis, we developed PTAAMG-Sig, a signature based on the expression of tumor-specific amino acid metabolism genes associated with overall survival (OS) prognosis. We evaluated its predictive performance for OS and thoroughly explored the effects of the PTAAMG-Sig risk score on the TME. The risk score was validated in two Gene Expression Omnibus (GEO) cohorts and further investigated against an original cohort of chemotherapy combined with immune checkpoint inhibitors (ICIs). Somatic mutation, chemotherapy response, immunotherapy response, gene set variation, gene set enrichment, immune infiltration, and plasma-free amino acids (PFAAs) profile analyses were performed to identify the underlying multi-omics features. Results TCGA datasets based PTAAMG-Sig model consisting of nine genes, KYNU, PSPH, PPAT, MIF, GCLC, ACAD8, TYRP1, ALDH2, and HDC, could effectively stratify the OS in LUAD patients. The two other GEO-independent datasets validated the robust predictive power of PTAAMG-Sig. Our differential analysis of somatic mutations in the high- and low-risk groups in TCGA cohort showed that the TP53 mutation rate was significantly higher in the high-risk group and negatively correlated with OS. Prediction from transcriptome data raised the possibility that PTAAMG-Sig could predict the response to chemotherapy and ICIs therapy. Our immunotherapy cohort confirmed the predictive ability of PTAAMG-Sig in the clinical response to ICIs therapy, which correlated with the infiltration of immune cells (e.g., T lymphocytes and nature killer cells). Corresponding to the concentrations of PFAAs, we discovered that the high PTAAMG-Sig risk score patients showed a significantly lower concentration of plasma-free α-aminobutyric acid. Conclusion In patients with LUAD, the PTAAMG-Sig effectively predicted OS, drug sensitivity, and immunotherapy outcomes. These findings are expected to provide new targets and strategies for personalized treatment of LUAD patients.
Collapse
Affiliation(s)
- Huihui Xiang
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| | - Rika Kasajima
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Center for Cancer Genome Medicine, Kanagawa Cancer Center, Yokohama, Japan
| | - Koichi Azuma
- Department of Internal Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Tomoyuki Tagami
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto Co., Inc., Kanagawa, Japan
| | - Asami Hagiwara
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto Co., Inc., Kanagawa, Japan
| | - Yoshiro Nakahara
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Haruhiro Saito
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Yuka Igarashi
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Japan
| | - Feifei Wei
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Japan
| | - Tatsuma Ban
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Mitsuyo Yoshihara
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Morphological Analysis Laboratory, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Yoshiyasu Nakamura
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Morphological Analysis Laboratory, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Shinya Sato
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
- Morphological Analysis Laboratory, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Shiro Koizume
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tetsuro Sasada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Japan
| | - Yohei Miyagi
- Molecular Pathology & Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| |
Collapse
|
19
|
Fu Y, Maccioni L, Wang XW, Greten TF, Gao B. Alcohol-associated liver cancer. Hepatology 2024; 80:1462-1479. [PMID: 38607725 DOI: 10.1097/hep.0000000000000890] [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] [Received: 01/31/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024]
Abstract
Heavy alcohol intake induces a wide spectrum of liver diseases ranging from steatosis, steatohepatitis, cirrhosis, and HCC. Although alcohol consumption is a well-known risk factor for the development, morbidity, and mortality of HCC globally, alcohol-associated hepatocellular carcinoma (A-HCC) is poorly characterized compared to viral hepatitis-associated HCC. Most A-HCCs develop after alcohol-associated cirrhosis (AC), but the direct carcinogenesis from ethanol and its metabolites to A-HCC remains obscure. The differences between A-HCC and HCCs caused by other etiologies have not been well investigated in terms of clinical prognosis, genetic or epigenetic landscape, molecular mechanisms, and heterogeneity. Moreover, there is a huge gap between basic research and clinical practice due to the lack of preclinical models of A-HCC. In the current review, we discuss the pathogenesis, heterogeneity, preclinical approaches, epigenetic, and genetic profiles of A-HCC, and discuss the current insights into and the prospects for future research on A-HCC. The potential effect of alcohol on cholangiocarcinoma and liver metastasis is also discussed.
Collapse
Affiliation(s)
- Yaojie Fu
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Luca Maccioni
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Xin Wei Wang
- Liver Carcinogenesis Section, Laboratory of Human Carcinogenesis, National Cancer Institute, NIH, Bethesda, Maryland, USA
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Tim F Greten
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
- Gastrointestinal Malignancies Section, Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
20
|
Hong X, Huang S, Jiang H, Ma Q, Qiu J, Luo Q, Cao C, Xu Y, Chen F, Chen Y, Sun C, Fu H, Liu Y, Li C, Chen F, Qiu P. Alcohol-related liver disease (ALD): current perspectives on pathogenesis, therapeutic strategies, and animal models. Front Pharmacol 2024; 15:1432480. [PMID: 39669199 PMCID: PMC11635172 DOI: 10.3389/fphar.2024.1432480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/22/2024] [Indexed: 12/14/2024] Open
Abstract
Alcohol-related liver disease (ALD) is a major cause of morbidity and mortality worldwide. It encompasses conditions such as fatty liver, alcoholic hepatitis, chronic hepatitis with liver fibrosis or cirrhosis, and hepatocellular carcinoma. Numerous recent studies have demonstrated the critical role of oxidative stress, abnormal lipid metabolism, endoplasmic reticulum stress, various forms of cell death (including apoptosis, necroptosis, and ferroptosis), intestinal microbiota dysbiosis, liver immune response, cell autophagy, and epigenetic abnormalities in the pathogenesis of ALD. Currently, abstinence, corticosteroids, and nutritional therapy are the traditional therapeutic interventions for ALD. Emerging therapies for ALD mainly include the blockade of inflammatory pathways, the promotion of liver regeneration, and the restoration of normal microbiota. Summarizing the advances in animal models of ALD will facilitate a more systematic investigation of the pathogenesis of ALD and the exploration of therapeutic targets. This review summarizes the latest insight into the pathogenesis and molecular mechanisms of ALD, as well as the pros and cons of ALD rodent models, providing a basis for further research on therapeutic strategies for ALD.
Collapse
Affiliation(s)
- Xiao Hong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuo Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - He Jiang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qing Ma
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiang Qiu
- Department of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Qihan Luo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chunlu Cao
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiyang Xu
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fuzhe Chen
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yufan Chen
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chunfeng Sun
- The First People’s Hospital of Xiaoshan District, Xiaoshan Affiliated Hospital of Wenzhou Medical University, Hangzhou, China
| | - Haozhe Fu
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiming Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Changyu Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangming Chen
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ping Qiu
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
21
|
Jeong M, Shin S, Lee G, Lee Y, Park SB, Kang J, Lee YS, Seo W, Lee H. Engineered lipid nanoparticles enable therapeutic gene silencing of GTSE1 for the treatment of liver fibrosis. J Control Release 2024; 374:337-348. [PMID: 39154935 DOI: 10.1016/j.jconrel.2024.08.012] [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: 06/16/2024] [Revised: 07/26/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024]
Abstract
Liver fibrosis is characterized by abnormal accumulation of extracellular matrix proteins, disrupting normal liver function. Despite its significant health impact, effective treatments remain limited. Here, we present the development of engineered lipid nanoparticles (LNPs) for targeted RNA therapeutic delivery in the liver. We investigated the therapeutic potential of modulating the G2 and S-phase expressed 1 (GTSE1) protein for treating liver fibrosis. Through screening, we identified P138Y LNP as a potent candidate with superior delivery efficiency and lower toxicity. Using these engineered LNPs, we successfully delivered siGTSE1 to hepatocytes, significantly reducing collagen accumulation and restoring liver function in a fibrosis animal model. Additionally, GTSE1 downregulation altered miRNA expression and upregulated hepatocyte nuclear factor 4 alpha (HNF4α). These findings suggest that therapeutic gene silencing of GTSE1 is a promising strategy for treating liver fibrosis by regenerating liver phenotypes and functions.
Collapse
Affiliation(s)
- Michaela Jeong
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Sumin Shin
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Gyeongseok Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Yeji Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Seo Bhin Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jisoo Kang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Young-Sun Lee
- Department of Internal Medicine, Korea University Medical Center, Seoul 08308, Republic of Korea
| | - Wonhyo Seo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| | - Hyukjin Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| |
Collapse
|
22
|
Yang X, Bhowmick K, Rao S, Xiang X, Ohshiro K, Amdur RL, Hassan MI, Mohammad T, Crandall K, Cifani P, Shetty K, Lyons SK, Merrill JR, Vegesna AK, John S, Latham PS, Crawford JM, Mishra B, Dasarathy S, Wang XW, Yu H, Wang Z, Huang H, Krainer AR, Mishra L. Aldehydes alter TGF-β signaling and induce obesity and cancer. Cell Rep 2024; 43:114676. [PMID: 39217614 PMCID: PMC11560041 DOI: 10.1016/j.celrep.2024.114676] [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: 01/24/2024] [Revised: 06/24/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
Obesity and fatty liver diseases-metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH)-affect over one-third of the global population and are exacerbated in individuals with reduced functional aldehyde dehydrogenase 2 (ALDH2), observed in approximately 560 million people. Current treatment to prevent disease progression to cancer remains inadequate, requiring innovative approaches. We observe that Aldh2-/- and Aldh2-/-Sptbn1+/- mice develop phenotypes of human metabolic syndrome (MetS) and MASH with accumulation of endogenous aldehydes such as 4-hydroxynonenal (4-HNE). Mechanistic studies demonstrate aberrant transforming growth factor β (TGF-β) signaling through 4-HNE modification of the SMAD3 adaptor SPTBN1 (β2-spectrin) to pro-fibrotic and pro-oncogenic phenotypes, which is restored to normal SMAD3 signaling by targeting SPTBN1 with small interfering RNA (siRNA). Significantly, therapeutic inhibition of SPTBN1 blocks MASH and fibrosis in a human model and, additionally, improves glucose handling in Aldh2-/- and Aldh2-/-Sptbn1+/- mice. This study identifies SPTBN1 as a critical regulator of the functional phenotype of toxic aldehyde-induced MASH and a potential therapeutic target.
Collapse
Affiliation(s)
- Xiaochun Yang
- Institute for Bioelectronic Medicine, Divisions of Gastroenterology and Hepatology, Department of Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Krishanu Bhowmick
- Institute for Bioelectronic Medicine, Divisions of Gastroenterology and Hepatology, Department of Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Shuyun Rao
- Department of Surgery, George Washington University, Washington, DC 20037, USA
| | - Xiyan Xiang
- Institute for Bioelectronic Medicine, Divisions of Gastroenterology and Hepatology, Department of Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Kazufumi Ohshiro
- Institute for Bioelectronic Medicine, Divisions of Gastroenterology and Hepatology, Department of Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | - Richard L Amdur
- Quantitative Intelligence Unit, The Institutes for Health Systems Science & Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Keith Crandall
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, George Washington University, Washington, DC 20037, USA
| | - Paolo Cifani
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Kirti Shetty
- Department of Gastroenterology and Hepatology, the University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Scott K Lyons
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Joseph R Merrill
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Anil K Vegesna
- Institute for Bioelectronic Medicine, Divisions of Gastroenterology and Hepatology, Department of Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Sahara John
- Institute for Bioelectronic Medicine, Divisions of Gastroenterology and Hepatology, Department of Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Patricia S Latham
- Department of Pathology, George Washington University, Washington, DC 20037, USA
| | - James M Crawford
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra, Northwell Health, Manhasset, NY 11030, USA
| | - Bibhuti Mishra
- Institute for Bioelectronic Medicine, Divisions of Gastroenterology and Hepatology, Department of Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; Department of Neurology, Northwell Health, Manhasset, NY 11030, USA
| | - Srinivasan Dasarathy
- Division of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis, Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Herbert Yu
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Zhanwei Wang
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Hai Huang
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Donald and Barbara Zucker School of Medicine at Hofstra, Northwell Health, Manhasset, NY 11030, USA
| | - Adrian R Krainer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Lopa Mishra
- Institute for Bioelectronic Medicine, Divisions of Gastroenterology and Hepatology, Department of Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA; Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Department of Surgery, George Washington University, Washington, DC 20037, USA.
| |
Collapse
|
23
|
Chin YT, Wu MH, Shih HY, Tsai CW, Pei JS, Ke TW, Wang YC, Hung YC, Chen JC, Bau DAT, Chang WS. Diagnostic Impacts of Aldehyde Dehydrogenase 2 Genetic Variants on Hepatocellular Carcinoma Susceptibility. CANCER DIAGNOSIS & PROGNOSIS 2024; 4:579-585. [PMID: 39238625 PMCID: PMC11372694 DOI: 10.21873/cdp.10366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 09/07/2024]
Abstract
Background/Aim The role of alcohol consumption and aldehyde dehydrogenase 2 (ALDH2) genotype in hepatocellular carcinoma (HCC) development remains uncertain. Materials and Methods We conducted genotyping of the ALDH2 rs671 single nucleotide polymorphism in 298 patients with HCC and 889 non-cancerous healthy controls. We assessed associations stratified by sex and alcohol consumption status. Results Distribution of ALDH2 rs671 variant genotypes differed significantly between HCC patients and controls (ptrend=0.0311). Logistic regression analyses indicated that compared to the wild-type GG genotype, the heterozygous variant AG genotype and homozygous variant AA genotype conferred 1.22- and 1.77-fold increases in HCC risk (p=0.1794 and 0.0150, respectively). Allelic frequency analysis showed that the A allele was associated with a 1.29-fold increased HCC risk (p=0.0123). Additionally, AA genotype carriers had significantly higher HCC risk than GG genotype carriers among males (p=0.0145) and non-alcohol drinkers (p<0.001). Conclusion HCC risk is influenced by ALDH2 genotype, with effects modified by sex and alcohol consumption. Particularly, individuals with the ALDH2 rs671 AA genotype should avoid alcohol consumption, especially males.
Collapse
Affiliation(s)
- Yu-Ting Chin
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Ming-Hsien Wu
- Division of Colon and Rectal Surgery, Taichung Armed Forces General Hospital, Taichung, Taiwan, R.O.C
- National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Hou-Yu Shih
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Chia-Wen Tsai
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Jen-Sheng Pei
- Department of Pediatrics, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan, R.O.C
| | - Tao-Wei Ke
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Yun-Chi Wang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Yi-Chih Hung
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Jaw-Chyun Chen
- Department of Medicinal Botanicals and Foods on Health Applications, Da-Yeh University, Changhua, Taiwan, R.O.C
| | - DA-Tian Bau
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan, R.O.C
| | - Wen-Shin Chang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| |
Collapse
|
24
|
Gao H, Peng X, Li N, Gou L, Xu T, Wang Y, Qin J, Liang H, Ma P, Li S, Wu J, Qin X, Xue B. Emerging role of liver-bone axis in osteoporosis. J Orthop Translat 2024; 48:217-231. [PMID: 39290849 PMCID: PMC11407911 DOI: 10.1016/j.jot.2024.07.008] [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: 03/06/2024] [Revised: 06/19/2024] [Accepted: 07/16/2024] [Indexed: 09/19/2024] Open
Abstract
Background Increasing attention to liver-bone crosstalk has spurred interest in targeted interventions for various forms of osteoporosis. Liver injury induced by different liver diseases can cause an imbalance in bone metabolism, indicating a novel regulatory paradigm between the liver and bone. However, the role of the liver-bone axis in both primary and secondary osteoporosis remains inadequately elucidated. Therefore, exploring the exact regulatory mechanisms of the liver-bone axis may offer innovative clinical approaches for treating diseases associated with the liver and bone. Methods Here, we summarize the latest research on the liver-bone axis by searching the PubMed and Web of Science databases and discuss the possible mechanism of the liver-bone axis in different types of osteoporosis. The literature directly reporting the regulatory role of the liver-bone axis in different types of osteoporosis from the PubMed and Web of Science databases has been included in the discussion of this review (including but not limited to the definition of the liver-bone axis, clinical studies, and basic research). In addition, articles discussing changes in bone metabolism caused by different etiologies of liver injury have also been included in the discussion of this review (including but not limited to clinical studies and basic research). Results Several endocrine factors (IGF-1, FGF21, hepcidin, vitamin D, osteocalcin, OPN, LCAT, Fetuin-A, PGs, BMP2/9, IL-1/6/17, and TNF-α) and key genes (SIRT2, ABCB4, ALDH2, TFR2, SPTBN1, ZNF687 and SREBP2) might be involved in the regulation of the liver-bone axis. In addition to the classic metabolic pathways involved in inflammation and oxidative stress, iron metabolism, cholesterol metabolism, lipid metabolism and immunometabolism mediated by the liver-bone axis require more research to elucidate the regulatory mechanisms involved in osteoporosis. Conclusion During primary and secondary osteoporosis, the liver-bone axis is responsible for liver and bone homeostasis via several hepatokines and osteokines as well as biochemical signaling. Combining multiomics technology and data mining technology could further advance our understanding of the liver-bone axis, providing new clinical strategies for managing liver and bone-related diseases.The translational potential of this article is as follows: Abnormal metabolism in the liver could seriously affect the metabolic imbalance of bone. This review summarizes the indispensable role of several endocrine factors and biochemical signaling pathways involved in the liver-bone axis and emphasizes the important role of liver metabolic homeostasis in the pathogenesis of osteoporosis, which provides novel potential directions for the prevention, diagnosis, and treatment of liver and bone-related diseases.
Collapse
Affiliation(s)
- Hongliang Gao
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
- Jiangsu Key Laboratory of Early Development and Chronic Disease Prevention in Children,Nanjing, Jiangsu,PR China
- Core Laboratory, Department of Clinical Laboratory, Sir Run Run Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, PR China
- Department of pathophysiology, Wannan Medical College, Wuhu, Anhui, PR China
| | - Xing Peng
- Core Laboratory, Department of Clinical Laboratory, Sir Run Run Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Ning Li
- Core Laboratory, Department of Clinical Laboratory, Sir Run Run Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Liming Gou
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
- Jiangsu Key Laboratory of Early Development and Chronic Disease Prevention in Children,Nanjing, Jiangsu,PR China
| | - Tao Xu
- Core Laboratory, Department of Clinical Laboratory, Sir Run Run Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Yuqi Wang
- Core Laboratory, Department of Clinical Laboratory, Sir Run Run Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Jian Qin
- Department of Orthoprdics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu , PR China
| | - Hui Liang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Peiqi Ma
- Medical Imaging Center, Fuyang People's Hospital, Fuyang, Anhui, PR China
| | - Shu Li
- Department of pathophysiology, Wannan Medical College, Wuhu, Anhui, PR China
| | - Jing Wu
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
- Jiangsu Key Laboratory of Early Development and Chronic Disease Prevention in Children,Nanjing, Jiangsu,PR China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Xihu Qin
- Department of General Surgery, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, PR China
| | - Bin Xue
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
- Jiangsu Key Laboratory of Early Development and Chronic Disease Prevention in Children,Nanjing, Jiangsu,PR China
- Core Laboratory, Department of Clinical Laboratory, Sir Run Run Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, PR China
- Department of General Surgery, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, PR China
| |
Collapse
|
25
|
Ye BJ, Li DF, Li XY, Hao JL, Liu DJ, Yu H, Zhang CD. Methylation synthetic lethality: Exploiting selective drug targets for cancer therapy. Cancer Lett 2024; 597:217010. [PMID: 38849016 DOI: 10.1016/j.canlet.2024.217010] [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: 04/28/2024] [Revised: 05/26/2024] [Accepted: 05/30/2024] [Indexed: 06/09/2024]
Abstract
In cancer, synthetic lethality refers to the drug-induced inactivation of one gene and the inhibition of another in cancer cells by a drug, resulting in the death of only cancer cells; however, this effect is not present in normal cells, leading to targeted killing of cancer cells. Recent intensive epigenetic research has revealed that aberrant epigenetic changes are more frequently observed than gene mutations in certain cancers. Recently, numerous studies have reported various methylation synthetic lethal combinations involving DNA damage repair genes, metabolic pathway genes, and paralogs with significant results in cellular models, some of which have already entered clinical trials with promising results. This review systematically introduces the advantages of methylation synthetic lethality and describes the lethal mechanisms of methylation synthetic lethal combinations that have recently demonstrated success in cellular models. Furthermore, we discuss the future opportunities and challenges of methylation synthetic lethality in targeted anticancer therapies.
Collapse
Affiliation(s)
- Bing-Jie Ye
- Clinical Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Di-Fei Li
- Clinical Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Xin-Yun Li
- Clinical Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Jia-Lin Hao
- Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Di-Jie Liu
- Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Hang Yu
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Chun-Dong Zhang
- Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
26
|
Huang HX, Zhong PY, Li P, Peng SJ, Ding XJ, Cai XL, Chen JH, Zhu X, Lu ZH, Tao XY, Liu YY, Chen L. Development and Validation of a Carbohydrate Metabolism-Related Model for Predicting Prognosis and Immune Landscape in Hepatocellular Carcinoma Patients. Curr Med Sci 2024; 44:771-788. [PMID: 39096475 DOI: 10.1007/s11596-024-2886-y] [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: 01/17/2024] [Accepted: 03/30/2024] [Indexed: 08/05/2024]
Abstract
OBJECTIVE The activities and products of carbohydrate metabolism are involved in key processes of cancer. However, its relationship with hepatocellular carcinoma (HCC) is unclear. METHODS The cancer genome atlas (TCGA)-HCC and ICGC-LIRI-JP datasets were acquired via public databases. Differentially expressed genes (DEGs) between HCC and control samples in the TCGA-HCC dataset were identified and overlapped with 355 carbohydrate metabolism-related genes (CRGs) to obtain differentially expressed CRGs (DE-CRGs). Then, univariate Cox and least absolute shrinkage and selection operator (LASSO) analyses were applied to identify risk model genes, and HCC samples were divided into high/low-risk groups according to the median risk score. Next, gene set enrichment analysis (GSEA) was performed on the risk model genes. The sensitivity of the risk model to immunotherapy and chemotherapy was also explored. RESULTS A total of 8 risk model genes, namely, G6PD, PFKFB4, ACAT1, ALDH2, ACYP1, OGDHL, ACADS, and TKTL1, were identified. Moreover, the risk score, cancer status, age, and pathologic T stage were strongly associated with the prognosis of HCC patients. Both the stromal score and immune score had significant negative/positive correlations with the risk score, reflecting the important role of the risk model in immunotherapy sensitivity. Furthermore, the stromal and immune scores had significant negative/positive correlations with risk scores, reflecting the important role of the risk model in immunotherapy sensitivity. Eventually, we found that high-/low-risk patients were more sensitive to 102 drugs, suggesting that the risk model exhibited sensitivity to chemotherapy drugs. The results of the experiments in HCC tissue samples validated the expression of the risk model genes. CONCLUSION Through bioinformatic analysis, we constructed a carbohydrate metabolism-related risk model for HCC, contributing to the prognosis prediction and treatment of HCC patients.
Collapse
Affiliation(s)
- Hong-Xiang Huang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Pei-Yuan Zhong
- Department of Oncology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China
| | - Ping Li
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Su-Juan Peng
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xin-Jing Ding
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xiang-Lian Cai
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jin-Hong Chen
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xie Zhu
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Zhi-Hui Lu
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xing-Yu Tao
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yang-Yang Liu
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Li Chen
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
27
|
Rwere F, White JR, Hell RCR, Yu X, Zeng X, McNeil L, Zhou KN, Angst MS, Chen CH, Mochly-Rosen D, Gross ER. Uncovering newly identified aldehyde dehydrogenase 2 genetic variants that lead to acetaldehyde accumulation after an alcohol challenge. J Transl Med 2024; 22:697. [PMID: 39075523 PMCID: PMC11288122 DOI: 10.1186/s12967-024-05507-x] [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: 02/02/2024] [Accepted: 07/14/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Aldehyde dehydrogenase 2 (ALDH2) is critical for alcohol metabolism by converting acetaldehyde to acetic acid. In East Asian descendants, an inactive genetic variant in ALDH2, rs671, triggers an alcohol flushing response due to acetaldehyde accumulation. As alcohol flushing is not exclusive to those of East Asian descent, we questioned whether additional ALDH2 genetic variants can drive facial flushing and inefficient acetaldehyde metabolism using human testing and biochemical assays. METHODS After IRB approval, human subjects were given an alcohol challenge (0.25 g/kg) while quantifying acetaldehyde levels and the physiological response (heart rate and skin temperature) to alcohol. Further, by employing biochemical techniques including human purified ALDH2 proteins and transiently transfected NIH 3T3 cells, we characterized two newly identified ALDH2 variants for ALDH2 enzymatic activity, ALDH2 dimer/tetramer formation, and reactive oxygen species production after alcohol treatment. RESULTS Humans heterozygous for rs747096195 (R101G) or rs190764869 (R114W) had facial flushing and a 2-fold increase in acetaldehyde levels, while rs671 (E504K) had facial flushing and a 6-fold increase in acetaldehyde levels relative to wild type ALDH2 carriers. In vitro studies with recombinant R101G and R114W ALDH2 enzyme showed a reduced efficiency in acetaldehyde metabolism that is unique when compared to E504K or wild-type ALDH2. The effect is caused by a lack of functional dimer/tetramer formation for R101G and decreased Vmax for both R101G and R114W. Transiently transfected NIH-3T3 cells with R101G and R114W also had a reduced enzymatic activity by ~ 50% relative to transfected wild-type ALDH2 and when subjected to alcohol, the R101G and R114W variants had a 2-3-fold increase in reactive oxygen species formation with respect to wild type ALDH2. CONCLUSIONS We identified two additional ALDH2 variants in humans causing facial flushing and acetaldehyde accumulation after alcohol consumption. As alcohol use is associated with a several-fold higher risk for esophageal cancer for the E504K variant, the methodology developed here to characterize ALDH2 genetic variant response to alcohol can lead the way precision medicine strategies to further understand the interplay of alcohol consumption, ALDH2 genetics, and cancer.
Collapse
Affiliation(s)
- Freeborn Rwere
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Joseph R White
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, 94305, USA
- Present address: Department of Anesthesiology, University of California Davis, Davis, CA, USA
| | - Rafaela C R Hell
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Xuan Yu
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Xiaocong Zeng
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, 94305, USA
- Present address: Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Leslie McNeil
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Kevin N Zhou
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Martin S Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Che-Hong Chen
- Department of Chemical and Systems Biology, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Eric R Gross
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
28
|
Li T, Wang A, Zhang Y, Chen W, Guo Y, Yuan X, Liu Y, Geng Y. Chemoproteomic Profiling of Signaling Metabolite Fructose-1,6-Bisphosphate Interacting Proteins in Living Cells. J Am Chem Soc 2024; 146:15155-15166. [PMID: 38775806 DOI: 10.1021/jacs.4c01335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Fructose-1,6-bisphosphate (FBP), a cellular endogenous sugar metabolite in the glycolytic pathway, has recently been reported to act as a signaling molecule to regulate various cellular events through the engagement of important proteins. Though tremendous progress has been made in identifying specific FBP-protein interactions, the comprehensive identification of FBP-interacting proteins and their regulatory mechanisms remains largely unexplored. Here, we describe a concise synthetic approach for the scalable preparation of a photoaffinity FBP probe that enables the quantitative chemoproteomic profiling of FBP-protein interactions based on photoaffinity labeling (PAL) directly in living cells. Using such a protocol, we captured known FBP targets including PKM2 and MDH2. Furthermore, among unknown FBP-interacting proteins, we identified a mitochondrial metabolic enzyme aldehyde dehydrogenase 2 (ALDH2), against which FBP showed inhibitory activity and resulted in cellular ROS upregulation accompanied by mitochondrial fragmentation. Our findings disclosed a new mode of glucose signaling mediating by the FBP-ALDH2-ROS axis.
Collapse
Affiliation(s)
- Tian Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Anhui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yanling Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wei Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yanshen Guo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xia Yuan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yuan Liu
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yiqun Geng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
29
|
Fan H, Lei S, Zhao Z, Huang Y, Wang H, Liu X, Li X, Xu M, Zhang W, Sun K, Xing H, Mei Y, Huang J, Zhu C, Zhang K, Zong Y, Shen X, Xie Q, Liu C. Beneficial Effects of Traditional Chinese Medicine Fuzheng Huayu on the Occurrence of Hepatocellular Carcinoma in Patients with Compensated Chronic Hepatitis B Cirrhosis Receiving Entecavir: A Multicenter Retrospective Cohort Study. J Clin Transl Hepatol 2024; 12:505-515. [PMID: 38779515 PMCID: PMC11106348 DOI: 10.14218/jcth.2023.00521] [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: 11/21/2023] [Revised: 02/26/2024] [Accepted: 03/07/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND AND AIMS The application of antifibrotic drugs to treat patients with chronic liver diseases who are receiving antiviral therapies for hepatocellular carcinoma (HCC) has not been established. Here, we aimed to assess the impact of the Traditional Chinese Medicine Fuzheng Huayu (FZHY) on the occurrence of HCC in patients with hepatitis B virus-related compensated cirrhosis receiving the antiviral drug entecavir (ETV). METHODS A multicenter retrospective cohort study was performed. Compensated liver cirrhosis patients were divided into the ETV+FZHY group or the ETV group according to treatment. The cumulative incidence of HCC was analyzed using Kaplan-Meier and log-rank tests. Propensity score matching was used for confounding factors. Stratified analysis and Cox regression were used to determine the effects of FZHY on the occurrence of HCC and liver function decompensation. RESULTS Out of 910 chronic hepatitis B patients, 458 were in the ETV+FZHY group and 452 were in the ETV group. After propensity score matching, the 5-year cumulative incidence of HCC was 9.8% in the ETV+FZHY group and 21.8% in the ETV group (p<0.01). The adjusted hazard ratio for HCC was 0.216 (0.108, 0.432) when FZHY treatment was >36 months. Age, diabetes, alanine aminotransferase, γ-glutamyl transpeptidase, albumin, hepatitis B e-antigen, and fibrosis 4 score were associated with the occurrence of HCC. FZHY decreased the risk of HCC in patients aged >45 years with a hepatitis B virus DNA level of ≥2,000 IU/l. CONCLUSION Adjunctive FZHY treatment reduced HCC occurrence in patients with hepatitis B virus cirrhosis who were treated with ETV, possibly due to the antifibrotic properties of FZHY.
Collapse
Affiliation(s)
- Haina Fan
- Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shujuan Lei
- Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhimin Zhao
- Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Huang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xudong Liu
- Department of Infectious Diseases, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xiaodong Li
- Institute of Liver Diseases, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei, China
| | - Min Xu
- Department of Infectious Diseases, Huaian Fourth People’s Hospital, Huai’an, Jiangsu, China
| | - Wei Zhang
- Institute of Liver Diseases, Shijiazhuang Fifth Hospital, Shijiazhuang, Hebei, China
| | - Kewei Sun
- Department of Liver Diseases, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Huichun Xing
- Center of Hepatology, Capital Medical University Affiliated Beijing Ditan Hospital, Beijing, China
| | - Yang Mei
- Department of Hepatology, The Fifth People’s Hospital of Anyang, Anyang, Henan, China
| | - Jiaquan Huang
- Department and Institute of Infectious Disease, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chuanwu Zhu
- Department of Hepatology, The Fifth People’s Hospital of Suzhou, Suzhou, Jiangsu, China
| | - Kejun Zhang
- Department of Gastroenterology, The First People’s Hospital of Jingmen, Jingmen, Hubei, China
| | - Yali Zong
- Department of Integrated Traditional and Western Medicine, The Ninth Hospital of Nanchang, Nanchang, Jiangxi, China
| | - Xizhong Shen
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing Xie
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenghai Liu
- Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
30
|
Tian Z, Song J, She J, He W, Guo S, Dong B. Constructing a disulfidptosis-related prognostic signature of hepatocellular carcinoma based on single-cell sequencing and weighted co-expression network analysis. Apoptosis 2024:10.1007/s10495-024-01968-z. [PMID: 38760515 DOI: 10.1007/s10495-024-01968-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2024] [Indexed: 05/19/2024]
Abstract
Hepatocellular carcinoma (HCC) ranks as the second leading cause of cancer-related deaths globally. Disulfidptosis is a newly identified form of regulated cell death that is induced by glucose starvation. However, the clinical prognostic characteristics of disulfidptosis-associated genes in HCC remain poorly understood. We conducted an analysis of the single-cell datasets GSE149614 and performed weighted co-expression network analysis (WGCNA) on the Cancer Genome Atlas (TCGA) datasets to identify the genes related to disulfidptosis. A prognostic model was constructed using univariate COX and Lasso regression. Survival analysis, immune microenvironment analysis, and mutation analysis were performed. Additionally, a nomogram associated with disulfidptosis-related signature was constructed to identify the prognosis of HCC patients. Patients with HCC in the TCGA and GSE14520 datasets were categorized using a disulfidptosis-related model, revealing significant differences in survival times between the high- and low-disulfidptosis groups. High-disulfidptosis patients exhibited increased expression of immune checkpoint-related genes, implying that immunotherapy and certain chemotherapies may be beneficial for them. Meanwhile, the ROC and decision curves analysis (DCA) indicated that the nomogram has satisfying prognostic efficacy. Moreover, the experimental results of GATM in this prognostic model indicated that GATM is low expressed in HCC tissues, and GATM knockdown promotes the proliferation and migration of HCC cells. By analyzing single-cell and bulk multi-omics sequencing data, we developed a prognostic signature related to disulfidptosis and explored the relationship between high- and low-disulfidptosis groups in HCC. This study offers a novel reference for gaining a deeper understanding of the role of disulfidptosis in HCC.
Collapse
Affiliation(s)
- Zelin Tian
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Junbo Song
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Jiang She
- Department of Orthopedics, Ninth Hospital of Xi'an, Xi'an, 710000, Shaanxi, China
| | - Weixiang He
- Department of Urology, Air Force Medical University, Xi'an, China
| | - Shanshan Guo
- Department of Physiology and Pathophysiology, Air Force Medical University, Xi'an, China
| | - Bingchen Dong
- Department of Orthopedics, Ninth Hospital of Xi'an, Xi'an, 710000, Shaanxi, China.
| |
Collapse
|
31
|
Liu ZY, Lin XH, Guo HY, Shi X, Zhang DY, Sun JL, Zhang GC, Xu RC, Wang F, Yu XN, Wang D, Weng SQ, Shen XZ, Liu TT, Dong L, Zhu JM. Multi-Omics profiling identifies aldehyde dehydrogenase 2 as a critical mediator in the crosstalk between Treg-mediated immunosuppression microenvironment and hepatocellular carcinoma. Int J Biol Sci 2024; 20:2763-2778. [PMID: 38725845 PMCID: PMC11077362 DOI: 10.7150/ijbs.93075] [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: 12/09/2023] [Accepted: 04/25/2024] [Indexed: 05/12/2024] Open
Abstract
Dysregulation of the aldehyde dehydrogenase (ALDH) family has been implicated in various pathological conditions, including cancer. However, a systematic evaluation of ALDH alterations and their therapeutic relevance in hepatocellular carcinoma (HCC) remains lacking. Herein, we found that 15 of 19 ALDHs were transcriptionally dysregulated in HCC tissues compared to normal liver tissues. A four gene signature, including ALDH2, ALDH5A1, ALDH6A1, and ALDH8A1, robustly predicted prognosis and defined a high-risk subgroup exhibiting immunosuppressive features like regulatory T cell (Tregs) infiltration. Single-cell profiling revealed selective overexpression of tumor necrosis factor receptor superfamily member 18 (TNFRSF18) on Tregs, upregulated in high-risk HCC patients. We identified ALDH2 as a tumor suppressor in HCC, with three novel phosphorylation sites mediated by protein kinase C zeta that enhanced enzymatic activity. Mechanistically, ALDH2 suppressed Tregs differentiation by inhibiting β-catenin/TGF-β1 signaling in HCC. Collectively, our integrated multi-omics analysis defines an ALDH-Tregs-TNFRSF18 axis that contributes to HCC pathogenesis and represents potential therapeutic targets for this aggressive malignancy.
Collapse
Affiliation(s)
- Zhi-Yong Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai 200030, China
- Shanghai Institute of Liver Diseases, Shanghai 200030, China
| | - Xia-Hui Lin
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai 200030, China
- Shanghai Institute of Liver Diseases, Shanghai 200030, China
| | - Hong-Ying Guo
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai 200030, China
- Shanghai Institute of Liver Diseases, Shanghai 200030, China
| | - Xuan Shi
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai 200030, China
- Shanghai Institute of Liver Diseases, Shanghai 200030, China
| | - Dan-Ying Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai 200030, China
- Shanghai Institute of Liver Diseases, Shanghai 200030, China
| | - Jia-Lei Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai 200030, China
- Shanghai Institute of Liver Diseases, Shanghai 200030, China
| | - Guang-Cong Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai 200030, China
- Shanghai Institute of Liver Diseases, Shanghai 200030, China
| | - Ru-Chen Xu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai 200030, China
- Shanghai Institute of Liver Diseases, Shanghai 200030, China
| | - Fu Wang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai 200030, China
- Shanghai Institute of Liver Diseases, Shanghai 200030, China
| | - Xiang-Nan Yu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai 200030, China
- Shanghai Institute of Liver Diseases, Shanghai 200030, China
| | - Dou Wang
- Department of Gastroenterology, Shigatse People's Hospital, Shigatse, Tibet 857000, China
| | - Shu-Qiang Weng
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai 200030, China
- Shanghai Institute of Liver Diseases, Shanghai 200030, China
| | - Xi-Zhong Shen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai 200030, China
- Shanghai Institute of Liver Diseases, Shanghai 200030, China
- Key Laboratory of Medical Molecular Virology, Shanghai Medical College of Fudan University, Shanghai 200030, China
| | - Tao-Tao Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai 200030, China
- Shanghai Institute of Liver Diseases, Shanghai 200030, China
| | - Ling Dong
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai 200030, China
- Shanghai Institute of Liver Diseases, Shanghai 200030, China
| | - Ji-Min Zhu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai 200030, China
- Shanghai Institute of Liver Diseases, Shanghai 200030, China
| |
Collapse
|
32
|
Danpanichkul P, Ng CH, Muthiah M, Suparan K, Tan DJH, Duangsonk K, Sukphutanan B, Kongarin S, Harinwan N, Panpradist N, Takahashi H, Kawaguchi T, Vichitkunakorn P, Chaiyakunapruk N, Nathisuwan S, Huang D, Arab JP, Noureddin M, Mellinger JL, Wijarnpreecha K. From Shadows to Spotlight: Exploring the Escalating Burden of Alcohol-Associated Liver Disease and Alcohol Use Disorder in Young Women. Am J Gastroenterol 2024; 119:893-909. [PMID: 38147513 DOI: 10.14309/ajg.0000000000002642] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 12/06/2023] [Indexed: 12/28/2023]
Abstract
INTRODUCTION The burden of alcohol-related complications is considerable, particularly alcohol-associated liver disease and alcohol use disorder (AUD). However, there are deficiencies in comprehensive epidemiological research focusing on these issues, especially among young women who display higher susceptibility to such complications compared with their male counterparts. We thus aimed to determine the global burden of these conditions in this vulnerable group. METHODS Leveraging data from the Global Burden of Disease Study 2019, we analyzed the prevalence, mortality, and disability-adjusted life years of alcohol-associated cirrhosis (AC), liver cancer from alcohol, and AUD in young women. The findings were categorized by region, nation, and sociodemographic index. RESULTS The highest age-standardized prevalence rates were observed in AUD (895.96 [95% uncertainty interval (UI) 722.6-1,103.58]), followed by AC (65.33 [95% UI 48.37-86.49]) and liver cancer from alcohol (0.13 [95% UI 0.09-0.19]) per 100,000 people. The highest age-standardized mortality rates were observed in AC (0.75 [95% UI 0.55-0.97]), followed by AUD (0.48 [95% UI 0.43-0.53]) and liver cancer from alcohol (0.06 [95% UI 0.04-0.09]). The highest burdens of AC and AUD were observed in Central Europe, whereas the high-income Asia Pacific had the highest burden of liver cancer from alcohol. DISCUSSION Throughout the past decade, the trend of AUD varied among regions while the impact of alcohol-associated liver disease has increased, requiring urgent public health strategy to mitigate these complications, particularly in female patients in Europe and the Asia-Pacific region.
Collapse
Affiliation(s)
- Pojsakorn Danpanichkul
- Immunology Unit, Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Cheng Han Ng
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore
| | - Mark Muthiah
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kanokphong Suparan
- Immunology Unit, Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Darren Jun Hao Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kwanjit Duangsonk
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | | | - Nateeluck Harinwan
- Department of Surgery, School of Medicine, Mae Fah Luang University, Chiang Rai, Thailand
| | - Nuttada Panpradist
- Global Center for Integrated Health for Women, Adolescents, and Children (Global WACh), Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Hirokazu Takahashi
- Division of Metabolism and Endocrinology, Faculty of Medicine, Saga University, Saga, Japan
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Polathep Vichitkunakorn
- Department of Family and Preventive Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Nathorn Chaiyakunapruk
- Department of Pharmacotherapy, College of Pharmacy, University of Utah, Salt Lake City, Utah, USA
| | - Surakit Nathisuwan
- Clinical Pharmacy Division, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Daniel Huang
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- NAFLD Research Center, Division of Gastroenterology, University of California at San Diego, La Jolla, California, USA
| | - Juan Pablo Arab
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University & London Health Sciences Centre, London, Ontario, Canada
- Department of Epidemiology and Biostatistics, Schulich School of Medicine, Western University, London, Ontario, Canada
| | - Mazen Noureddin
- Houston Methodist and Houston Research Institute, Houston, Texas, USA
| | - Jessica Leigh Mellinger
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Karn Wijarnpreecha
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Arizona College of Medicine and Division of Gastroenterology and Hepatology, Phoenix, Arizona, USA
- Department of Internal Medicine, Banner University Medical Center, Phoenix, Arizona, USA
| |
Collapse
|
33
|
Yang D, Hu Y, Yang J, Tao L, Su Y, Wu Y, Yao Y, Wang S, Ye S, Xu T. Research Progress on the Correlation between Acetaldehyde Dehydrogenase 2 and Hepatocellular Carcinoma Development. J Pharmacol Exp Ther 2024; 389:163-173. [PMID: 38453527 DOI: 10.1124/jpet.123.001898] [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: 08/24/2023] [Revised: 02/03/2024] [Accepted: 02/23/2024] [Indexed: 03/09/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the predominant pathologic type of primary liver cancer. It is a malignant tumor of liver epithelial cells. There are many ways to treat HCC, but the survival rate for HCC patients remains low. Therefore, understanding the underlying mechanisms by which HCC occurs and develops is critical to explore new therapeutic targets. Aldehyde dehydrogenase 2 (ALDH2) is an important player in the redox reaction of ethanol with endogenous aldehyde products released by lipid peroxidation. Increasing evidence suggests that ALDH2 is a crucial regulator of human tumor development, including HCC. Therefore, clarifying the relationship between ALDH2 and HCC is helpful for formulating rational treatment strategies. This review highlights the regulatory roles of ALDH2 in the development of HCC, elucidates the multiple potential mechanisms by which ALDH2 regulates the development of HCC, and summarizes the progress of research on ALDH2 gene polymorphisms and HCC susceptibility. Meanwhile, we envision viable strategies for targeting ALDH2 in the treatment of HCC SIGNIFICANCE STATEMENT: Numerous studies have aimed to explore novel therapeutic targets for HCC, and ALDH2 has been reported to be a critical regulator of HCC progression. This review discusses the functions, molecular mechanisms, and clinical significance of ALDH2 in the development of HCC and examines the prospects of ALDH2-based therapy for HCC.
Collapse
Affiliation(s)
- Dashuai Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China (Y.H.); Department of Pediatric orthopedics, Anhui Children's Hospital, Hefei, China (J.Y.); Bengbu Medical University, Bengbu, Anhui, China (Y.S.); and School of Materials and Chemistry and School of Plant Protection, Anhui Agricultural University, Hefei, Anhui, China (S.Y.)
| | - Ying Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China (Y.H.); Department of Pediatric orthopedics, Anhui Children's Hospital, Hefei, China (J.Y.); Bengbu Medical University, Bengbu, Anhui, China (Y.S.); and School of Materials and Chemistry and School of Plant Protection, Anhui Agricultural University, Hefei, Anhui, China (S.Y.)
| | - Junfa Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China (Y.H.); Department of Pediatric orthopedics, Anhui Children's Hospital, Hefei, China (J.Y.); Bengbu Medical University, Bengbu, Anhui, China (Y.S.); and School of Materials and Chemistry and School of Plant Protection, Anhui Agricultural University, Hefei, Anhui, China (S.Y.)
| | - Liangsong Tao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China (Y.H.); Department of Pediatric orthopedics, Anhui Children's Hospital, Hefei, China (J.Y.); Bengbu Medical University, Bengbu, Anhui, China (Y.S.); and School of Materials and Chemistry and School of Plant Protection, Anhui Agricultural University, Hefei, Anhui, China (S.Y.)
| | - Yue Su
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China (Y.H.); Department of Pediatric orthopedics, Anhui Children's Hospital, Hefei, China (J.Y.); Bengbu Medical University, Bengbu, Anhui, China (Y.S.); and School of Materials and Chemistry and School of Plant Protection, Anhui Agricultural University, Hefei, Anhui, China (S.Y.)
| | - Yincui Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China (Y.H.); Department of Pediatric orthopedics, Anhui Children's Hospital, Hefei, China (J.Y.); Bengbu Medical University, Bengbu, Anhui, China (Y.S.); and School of Materials and Chemistry and School of Plant Protection, Anhui Agricultural University, Hefei, Anhui, China (S.Y.)
| | - Yan Yao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China (Y.H.); Department of Pediatric orthopedics, Anhui Children's Hospital, Hefei, China (J.Y.); Bengbu Medical University, Bengbu, Anhui, China (Y.S.); and School of Materials and Chemistry and School of Plant Protection, Anhui Agricultural University, Hefei, Anhui, China (S.Y.)
| | - Shuxian Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China (Y.H.); Department of Pediatric orthopedics, Anhui Children's Hospital, Hefei, China (J.Y.); Bengbu Medical University, Bengbu, Anhui, China (Y.S.); and School of Materials and Chemistry and School of Plant Protection, Anhui Agricultural University, Hefei, Anhui, China (S.Y.)
| | - Sheng Ye
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China (Y.H.); Department of Pediatric orthopedics, Anhui Children's Hospital, Hefei, China (J.Y.); Bengbu Medical University, Bengbu, Anhui, China (Y.S.); and School of Materials and Chemistry and School of Plant Protection, Anhui Agricultural University, Hefei, Anhui, China (S.Y.)
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, China (D.Y., L.T., Y.W., Y.Y., S.W., T.X.); State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China (Y.H.); Department of Pediatric orthopedics, Anhui Children's Hospital, Hefei, China (J.Y.); Bengbu Medical University, Bengbu, Anhui, China (Y.S.); and School of Materials and Chemistry and School of Plant Protection, Anhui Agricultural University, Hefei, Anhui, China (S.Y.)
| |
Collapse
|
34
|
Wang S, Xie Y, Liu F, Wang J, Yang Y, Wang J, Wang Y, Hang X. miR-3188 inhibits hepatitis B virus transcription by targeting Bcl-2. Arch Virol 2024; 169:88. [PMID: 38565755 DOI: 10.1007/s00705-024-05992-x] [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: 10/18/2023] [Accepted: 12/30/2023] [Indexed: 04/04/2024]
Abstract
Transcription of the covalently closed circular DNA (cccDNA) of hepatitis B virus (HBV) is subject to dual regulation by host factors and viral proteins. MicroRNAs (miRNAs) can regulate the expression of target genes at the post-transcriptional level. Systematic investigation of miRNA expression in HBV infection and the interaction between HBV and miRNAs may deepen our understanding of the transcription mechanisms of HBV cccDNA, thereby providing opportunities for intervention. miRNA sequencing and real-time quantitative PCR (qRT-PCR) were used to analyze miRNA expression after HBV infection of cultured cells. Clinical samples were analyzed for miRNAs and HBV transcription-related indicators, using qRT-PCR, enzyme-linked immunoassay (ELISA), and Western blot. miRNA mimics or inhibitors were used to study their effects on the HBV life cycle. The target genes of miR-3188 and their roles in HBV cccDNA transcription were also identified. The expression of 10 miRNAs, including miR-3188, which was significantly decreased after HBV infection, was measured in clinical samples from patients with chronic HBV infection. Overexpression of miR-3188 inhibited HBV transcription, whereas inhibition of miR-3188 expression promoted HBV transcription. Further investigation confirmed that miR-3188 inhibited HBV transcription by targeting Bcl-2. miR-3188 is a key miRNA that regulates HBV transcription by targeting the host protein Bcl-2. This observation provides insights into the regulation of cccDNA transcription and suggests new targets for anti-HBV treatment.
Collapse
Affiliation(s)
- Shijie Wang
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ying Xie
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Fufei Liu
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jianrong Wang
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yinghao Yang
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, China
- Department of Infectious Diseases, The 988th Hospital of the Joint Logistic Support Force, Jiaozuo, China
| | - Junxue Wang
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yongxiang Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Xiaofeng Hang
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
35
|
Ferreira IC, Torrejón E, Abecasis B, Alexandre BM, Gomes RA, Verslype C, van Pelt J, Barbas A, Simão D, Bandeiras TM, Bortoluzzi A, Rebelo SP. Aldehyde Dehydrogenase 2 (ALDH2): A novel sorafenib target in hepatocellular carcinoma unraveled by the proteome-wide cellular thermal shift assay. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100154. [PMID: 38521503 DOI: 10.1016/j.slasd.2024.100154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/13/2024] [Accepted: 03/19/2024] [Indexed: 03/25/2024]
Abstract
Sorafenib is a multikinase inhibitor indicated for first-line treatment of unresectable hepatocellular carcinoma. Despite its widespread use in the clinic, the existing knowledge of sorafenib mode-of-action remains incomplete. To build upon the current understanding, we used the Cellular Thermal Shift Assay (CETSA) coupled to Mass Spectrometry (CETSA-MS) to monitor compound binding to its target proteins in the cellular context on a proteome-wide scale. Among the potential sorafenib targets, we identified aldehyde dehydrogenase 2 (ALDH2), an enzyme that plays a major role in alcohol metabolism. We validated the interaction of sorafenib with ALDH2 by orthogonal methods using pure recombinant protein, proving that this interaction is not mediated by other cellular components. Moreover, we showed that sorafenib inhibits ALDH2 activity, supporting a functional role for this interaction. Finally, we were able to demonstrate that both ALDH2 protein expression and activity were reduced in sorafenib-resistant cells compared to the parental cell line. Overall, our study allowed the identification of ALDH2 as a novel sorafenib target and sheds light on its potential role in both hepatocellular carcinoma and sorafenib resistance condition.
Collapse
Affiliation(s)
- Inês C Ferreira
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
| | - Estefania Torrejón
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; ITQB, ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Bernardo Abecasis
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
| | - Bruno M Alexandre
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; ITQB, ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Ricardo A Gomes
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; ITQB, ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Chris Verslype
- Department of Gastroenterology and Hepatology, KU Leuven, Leuven, Belgium
| | - Jos van Pelt
- Department of Oncology, Laboratory of Clinical Digestive Oncology, KU, Leuven, Belgium
| | - Ana Barbas
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; Bayer Portugal, Carnaxide, Portugal
| | - Daniel Simão
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
| | - Tiago M Bandeiras
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; ITQB, ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Alessio Bortoluzzi
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; ITQB, ITQB-NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal.
| | - Sofia P Rebelo
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal.
| |
Collapse
|
36
|
Flashner S, Shimonosono M, Tomita Y, Matsuura N, Ohashi S, Muto M, Klein-Szanto AJ, Alan Diehl J, Chen CH, Mochly-Rosen D, Weinberg KI, Nakagawa H. ALDH2 dysfunction and alcohol cooperate in cancer stem cell enrichment. Carcinogenesis 2024; 45:95-106. [PMID: 37978873 PMCID: PMC10859731 DOI: 10.1093/carcin/bgad085] [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: 05/08/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 11/19/2023] Open
Abstract
The alcohol metabolite acetaldehyde is a potent human carcinogen linked to esophageal squamous cell carcinoma (ESCC) initiation and development. Aldehyde dehydrogenase 2 (ALDH2) is the primary enzyme that detoxifies acetaldehyde in the mitochondria. Acetaldehyde accumulation causes genotoxic stress in cells expressing the dysfunctional ALDH2E487K dominant negative mutant protein linked to ALDH2*2, the single nucleotide polymorphism highly prevalent among East Asians. Heterozygous ALDH2*2 increases the risk for the development of ESCC and other alcohol-related cancers. Despite its prevalence and link to malignant transformation, how ALDH2 dysfunction influences ESCC pathobiology is incompletely understood. Herein, we characterize how ESCC and preneoplastic cells respond to alcohol exposure using cell lines, three-dimensional organoids and xenograft models. We find that alcohol exposure and ALDH2*2 cooperate to increase putative ESCC cancer stem cells with high CD44 expression (CD44H cells) linked to tumor initiation, repopulation and therapy resistance. Concurrently, ALHD2*2 augmented alcohol-induced reactive oxygen species and DNA damage to promote apoptosis in the non-CD44H cell population. Pharmacological activation of ALDH2 by Alda-1 inhibits this phenotype, suggesting that acetaldehyde is the primary driver of these changes. Additionally, we find that Aldh2 dysfunction affects the response to cisplatin, a chemotherapeutic commonly used for the treatment of ESCC. Aldh2 dysfunction facilitated enrichment of CD44H cells following cisplatin-induced oxidative stress and cell death in murine organoids, highlighting a potential mechanism driving cisplatin resistance. Together, these data provide evidence that ALDH2 dysfunction accelerates ESCC pathogenesis through enrichment of CD44H cells in response to genotoxic stressors such as environmental carcinogens and chemotherapeutic agents.
Collapse
Affiliation(s)
- Samuel Flashner
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Masataka Shimonosono
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Yasuto Tomita
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Norihiro Matsuura
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Shinya Ohashi
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Shogoin, Kyoto 606-8507, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Shogoin, Kyoto 606-8507, Japan
| | | | - J Alan Diehl
- Case Comprehensive Cancer Center, Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Che-Hong Chen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kenneth I Weinberg
- Division of Stem Cell Biology and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hiroshi Nakagawa
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| |
Collapse
|
37
|
Wang Y, Wang DY, Bu KN, Gao JD, Zhang BL. Prognosis prediction and risk stratification of breast cancer patients based on a mitochondria-related gene signature. Sci Rep 2024; 14:2859. [PMID: 38310106 PMCID: PMC10838276 DOI: 10.1038/s41598-024-52981-w] [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: 04/02/2023] [Accepted: 01/25/2024] [Indexed: 02/05/2024] Open
Abstract
As the malignancy with the highest global incidence, breast cancer represents a significant threat to women's health. Recent advances have shed light on the importance of mitochondrial function in cancer, particularly in metabolic reprogramming within tumors. Recognizing this, we developed a novel risk signature based on mitochondrial-related genes to improve prognosis prediction and risk stratification in breast cancer patients. In this study, transcriptome data and clinical features of breast cancer samples were extracted from two sources: the TCGA, serving as the training set, and the METABRIC, used as the independent validation set. We developed the signature using LASSO-Cox regression and assessed its prognostic efficacy via ROC curves. Furthermore, the signature was integrated with clinical features to create a Nomogram model, whose accuracy was validated through clinical calibration curves and decision curve analysis. To further elucidate prognostic variations between high and low-risk groups, we conducted functional enrichment and immune infiltration analyses. Additionally, the study encompassed a comparison of mutation landscapes and drug sensitivity, providing a comprehensive understanding of the differing characteristics in these groups. Conclusively, we established a risk signature comprising 8 mitochondrial-related genes-ACSL1, ALDH2, MTHFD2, MRPL13, TP53AIP1, SLC1A1, ME3, and BCL2A1. This signature was identified as an independent risk predictor for breast cancer patient survival, exhibiting a significant high hazard ratio (HR = 3.028, 95%CI 2.038-4.499, P < 0.001). Patients in the low-risk group showed a more favorable prognosis, with enhanced immune infiltration, distinct mutation landscapes, and greater sensitivity to anti-tumor drugs. In contrast, the high-risk group exhibited an adverse trend in these aspects. This risk signature represents a novel and effective prognostic indicator, suggesting valuable insights for patient stratification in breast cancer.
Collapse
Affiliation(s)
- Yang Wang
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ding-Yuan Wang
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ke-Na Bu
- Xingyuan Hospital of Yulin City, Yulin City, 719051, Shanxi Province, China
| | - Ji-Dong Gao
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union College, Shenzhen, 518116, China.
| | - Bai-Lin Zhang
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
38
|
Zhang L, Chen ZY, Wei XX, Li JD, Chen G. What are the changes in the hotspots and frontiers of microRNAs in hepatocellular carcinoma over the past decade? World J Clin Oncol 2024; 15:145-158. [PMID: 38292666 PMCID: PMC10823937 DOI: 10.5306/wjco.v15.i1.145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/08/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Emerging research suggests that microRNAs (miRNAs) play an important role in the development of hepatocellular carcinoma (HCC). A comprehensive analysis of recent research concerning miRNAs in HCC development could provide researchers with a valuable reference for further studies. AIM To make a comprehensive analysis of recent studies concerning miRNAs in HCC. METHODS All relevant publications were retrieved from the Web of Science Core Collection database. Bibliometrix software, VOSviewer software and CiteSpace software were used to visually analyze the distribution by time, countries, institutions, journals, and authors, as well as the keywords, burst keywords and thematic map. RESULTS A total of 9426 publications on this topic were found worldwide. According to the keywords analysis, we found that the studies of miRNAs focused on their expression level, effects, and mechanisms on the biological behaviour of HCC. Keywords bursting analysis showed that in the early years (2013-2017), "microRNA expression", "gene expression", "expression profile", "functional polymorphism", "circulating microRNA", "susceptibility" and "mir 21" started to attract attention. In the latest phase (2018-2022), the hot topics turned to "sorafenib resistance", "tumor microenvironment" and so on. CONCLUSION This study provides a comprehensive overview of the role of miRNAs in HCC development based on bibliometric analysis. The hotspots in this field focus on miRNAs expression level, effects, and mechanisms on the biological behavior of HCC. The frontiers turned to sorafenib resistance, tumor microenvironment and so on.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Zu-Yuan Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Xiao-Xian Wei
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Jian-Di Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
39
|
Zhang N, Zhu Y, Zhang X, Yang K, Yang X, An M, Tian C, Li J. Based on network pharmacology and experiments to explore the underlying mechanism of Mahonia bealei (Fortune) Carrière for treating alcoholic hepatocellular carcinoma. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116919. [PMID: 37453621 DOI: 10.1016/j.jep.2023.116919] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Mahonia bealei (Fortune) Carrière (M. bealei) is a traditional medicine widely used by the Hmong community in Guizhou. It possesses diverse biological activities and shows promise in cancer treatment; however, contemporary pharmacological research in this area is lacking. AIMS OF THE STUDY This study aimed to investigate the effects and underlying mechanisms of M. bealei on alcoholic hepatocellular carcinoma (HCC). MATERIALS AND METHODS We initially employed the LC-MS/MS method to identify the compounds present in M. bealei serum. Subsequently, its potential targets were predicted using public databases. Bioinformatics and network pharmacology approaches, such as univariate Cox regression and random forest (RF) algorithms, were utilized to identify differentially expressed genes (DEGs) associated with the prognosis of alcoholic HCC. Survival curve and receiver operating characteristic (ROC) analyses were conducted using alcoholic HCC-related data from TCGA and GEO to determine the diagnostic value of the identified DEGs. Molecular docking using the CDOCKER approach based on CHARMm was performed to validate the affinity between the predictive compounds and targets. Additionally, we evaluated the impact of M. bealei on cell proliferation, migration, and conducted western blot assays. RESULTS The LC-MS/MS approach identified 17 therapeutic components and predicted 483 component-related targets, of which 63 overlapped with alcoholic HCC targets and were considered potential therapeutic targets. GO and KEGG pathway analysis revealed significant associations between the 63 overlapping targets and alcoholic HCC progression. Through various approaches in the Cytoscape 3.9.0 software, we confirmed 9 hub genes (CDK1, CXCR4, DNMT1, ESR1, KIT, PDGFRB, SERPINE1, TOP2A, and TYMS) as core targets. TOP2A and CDK1 genes were identified as advantageous for diagnosing alcoholic HCC using univariate Cox regression, RF, survival curve, and ROC analysis. Molecular docking analysis demonstrated strong binding affinity between key bioactive components cyclamic acid, perfluoroalkyl carboxylic acid, perfluorosulfonic acid, alpha-linolenic acid, adenosine receptor antagonist (CGS 15943), and Prodigiosin and TOP2A and CDK1. In vitro experiments confirmed that M. bealei significantly suppressed cell proliferation and migration of HepG2 cells, while downregulating TOP2A and CDK1 expression. CONCLUSION This study highlights the potential of M. bealei as a natural medicine for the treatment of alcoholic HCC. Six compounds (cyclamic acid, perfluoroalkylic carboxylic acids, perfluorosulfonic acid, alpha-linolenic acid, adenosine receptor antagonist (CGS 15943), and Prodigiosin) present in M. bealei serum may exhibit therapeutic effects against alcoholic HCC by downregulating CDK1 and TOP2A expression levels in vitro.
Collapse
Affiliation(s)
- Nannan Zhang
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Yi Zhu
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Xuewu Zhang
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Kaiping Yang
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Xia Yang
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Mingyu An
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Changlin Tian
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Jun Li
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China.
| |
Collapse
|
40
|
Yoladı FB, Burmaoğlu E, Palabıyık ŞS. Experimental In Vivo Toxicity Models for Alcohol Toxicity. Eurasian J Med 2023; 55:82-90. [PMID: 39109811 PMCID: PMC11075036 DOI: 10.5152/eurasianjmed.2023.23345] [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: 11/28/2023] [Accepted: 12/26/2023] [Indexed: 08/11/2024] Open
Abstract
Alcohol consumption poses a significant risk for the development of chronic illnesses, one of the leading causes of "preventable" disease and death worldwide. Harmful consumption of alcohol is thought to result in approximately 2.5-3 million deaths each year, the majority of which are caused by alcohol-related liver diseases. Hepatocellular carcinoma, cirrhosis, fibrosis, steatosis, and steatohepatitis are among the liver illnesses caused by alcohol. The mechanisms behind human diseases are often mimicked and understood through the use of animal models. Rodents are the ideal animals to study alcohol-related liver diseases. In these experimental models using rodents, the ethanol ratio, method of administration, and diet to be applied vary. Within the scope of this review, it is aimed at providing information about the experimental models used today for alcohol toxicity and the advantages and disadvantages of these models.
Collapse
Affiliation(s)
- Fatma Betül Yoladı
- Department of Pharmaceutical Toxicology, Atatürk University Faculty of Pharmacy, Erzurum, Turkey
- Department of Pharmaceutical Toxicology, Hacettepe University Faculty of Pharmacy, Ankara, Turkey
| | | | - Şaziye Sezin Palabıyık
- Department of Pharmaceutical Toxicology, Atatürk University Faculty of Pharmacy, Erzurum, Turkey
- Clinical Research, Development and Design Application and Research Center, Atatürk University, Erzurum, Turkey
| |
Collapse
|
41
|
Dai X, Shi X, Luo M, Li P, Gao Y. Integrative analysis of transcriptomic and metabolomic profiles reveals enhanced arginine metabolism in androgen-independent prostate cancer cells. BMC Cancer 2023; 23:1241. [PMID: 38104097 PMCID: PMC10724921 DOI: 10.1186/s12885-023-11707-3] [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: 08/25/2023] [Accepted: 12/02/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND Prostate cancer is a common solid tumor that affects a significant number of men worldwide. Conventional androgen deprivation therapy (ADT) increases the risk of developing castration-resistant prostate cancer (CRPC). Effective clinical management of patients with CRPC is challenging due to the limited understanding. METHODS In this study, transcriptomic and metabolomic profiles of androgen-dependent prostate cancer cell line LNCaP and the androgen-independent cells developed from LNCaP cells (LNCaP-ADR) were investigated using RNA-sequencing and LC-MS/MS, respectively. The differentially expressed genes and metabolites were analyzed, and integrative analysis of transcriptomic and metabolomic data was further conducted to obtain a comprehensive understanding of the metabolic characteristics in LNCaP-ADR cells. Quantitative real-time PCR (QPCR) was employed to ascertain the mRNA expression levels of the selected differentially expressed genes. RESULTS The arginine and proline metabolism pathway was identified as a commonly altered pathway at both the transcriptional and metabolic levels. In the LNCaP-ADR cells, significant upregulation was observed for metabolites including 5-Aminopentanoic acid, L-Arginine, L-Glutamic acid, N-Acetyl-L-alanine, and Pyrrole-2-carboxylic acid at the metabolic level. At the transcriptional level, MAOA, ALDH3A2, ALDH2, ARG1, CKMT2, and CNDP1 were found to be significantly upregulated in the LNCaP-ADR cells. Gene set enrichment analysis (GSEA) identified various enriched gene sets in the LNCaP-ADR cells, encompassing inflammatory response, 9plus2 motile cilium, motile cilium, ciliary plasm, cilium or flagellum-dependent cell motility, cilium movement, cilium, response to endoplasmic reticulum stress, PTEN DN.V1 DN, SRC UP.V1 UP, IL15 UP.V1 DN, RB DN.V1 DN, AKT UP MTOR DN.V1 UP, VEGF A UP.V1 UP, and KRAS.LUNG.BREAST UP.V1 UP. CONCLUSIONS These findings highlight the substantial association between the arginine and proline metabolism pathway and CRPC, emphasizing the need to prioritize strategies that target dysregulated metabolites and differentially expressed genes as essential interventions in the clinical management of CRPC.
Collapse
Affiliation(s)
- Xingchen Dai
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Department of Nephrology, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xinyi Shi
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Ankang Central Hospital, Ankang, China
| | - Mingxiu Luo
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Pu Li
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yujing Gao
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China.
- National Health Commission Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
42
|
Aasarey R, Yadav K, Kashyap BK, Prabha S, Kumar P, Kumar A, Ruokolainen J, Kesari KK. Role of Immunological Cells in Hepatocellular Carcinoma Disease and Associated Pathways. ACS Pharmacol Transl Sci 2023; 6:1801-1816. [PMID: 38093838 PMCID: PMC10714437 DOI: 10.1021/acsptsci.3c00216] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 03/28/2024]
Abstract
Hepatocellular carcinoma (HCC) remains one of the predominant causes of cancer-related mortality across the globe. It is attributed to obesity, excessive alcohol consumption, smoking, and infection by the hepatitis virus. Early diagnosis of HCC is essential, and local treatments such as surgical excision and percutaneous ablation are effective. Palliative systemic therapy, primarily with the tyrosine kinase inhibitor Sorafenib, is used in advanced cases. However, the prognosis for advanced HCC remains poor. This Review additionally describes the pathophysiological mechanisms of HCC, which include aberrant molecular signaling, genomic instability, persistent inflammation, and the paradoxical position of the immune system in promoting and suppressing HCC. The paper concludes by discussing the growing body of research on the relationship between mitochondria and HCC, suggesting that mitochondrial dysfunction may contribute to the progression of HCC. This Review focuses on immunological interactions between different mechanisms of HCC progression, including obesity, viral infection, and alcohol consumption.
Collapse
Affiliation(s)
- Ram Aasarey
- Department
of Laboratory Medicine, All India Institute
of Medical Science, New Delhi-11029, India
| | - Kajal Yadav
- Department
of Biotechnology, All India Institute of
Medical Science, New Delhi-11029, India
| | - Brijendra Kumar Kashyap
- Department
of Biotechnology Engineering, Institute of Engineering and Technology, Bundelkhand University, Jhansi-284128, Uttar Pradesh, India
| | - Sarit Prabha
- Department
of Biological Science and Engineering, Maulana
Azad National Institute of Technology, Bhopal-462003, Madhya Pradesh,India
| | - Pramod Kumar
- Indian
Council of Medical Research, National Institute
of Cancer Prevention and Research (NICPR), l-7, Sector-39, Noida-201301, National Capital Region, India
| | - Anil Kumar
- Department
of Life Sciences, School of Natural Sciences, Central University of Jharkhand, Cheri-Manatu, Karmre, Kanke-835222, Ranchi, India
| | - Janne Ruokolainen
- Department
of Applied Physics, School of Science, Aalto
University, FI-00076 Espoo, Finland
| | - Kavindra Kumar Kesari
- Department
of Applied Physics, School of Science, Aalto
University, FI-00076 Espoo, Finland
- Research
and Development Cell, Lovely Professional
University, Phagwara-144411, Punjab, India
| |
Collapse
|
43
|
He W, Wang X, Chen M, Li C, Chen W, Pan L, Cui Y, Yu Z, Wu G, Yang Y, Xu M, Dong Z, Ma K, Wang J, He Z. Metformin reduces hepatocarcinogenesis by inducing downregulation of Cyp26a1 and CD8 + T cells. Clin Transl Med 2023; 13:e1465. [PMID: 37997519 PMCID: PMC10668005 DOI: 10.1002/ctm2.1465] [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: 04/14/2023] [Revised: 10/12/2023] [Accepted: 10/19/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a highly heterogeneous cancer with major challenges in both prevention and therapy. Metformin, adenosine monophosphate-activated protein kinase (AMPK) activator, has been suggested to reduce the incidence of HCC when used for patients with diabetes in preclinical and clinical studies. However, the possible effects of metformin and their mechanisms of action in non-diabetic HCC have not been adequately investigated. METHODS Fah-/- mice were used to construct a liver-injury-induced non-diabetic HCC model for exploring hepatocarcinogenesis and therapeutic potential of metformin. Changes in relevant tumour and biochemical indicators were measured. Bulk and single-cell RNA-sequencing analyses were performed to validate the crucial role of proinflammatory/pro-tumour CD8+ T cells. In vitro and in vivo experiments were performed to confirm Cyp26a1-related antitumour mechanisms of metformin. RESULTS RNA-sequencing analysis showed that chronic liver injury led to significant changes in AMPK-, glucose- and retinol metabolism-related pathways in Fah-/- mice. Metformin prevented the formation of non-diabetic HCC in Fah-/- mice with chronic liver injury. Cyp26a1 ddexpression in hepatocytes was significantly suppressed after metformin treatment. Moreover, downregulation of Cyp26a1 occurred in conjunction with increased levels of all-trans-retinoic acid (atRA), which is involved in the activation of metformin-suppressed hepatocarcinogenesis in Fah-/- mice. In contrast, both CD8+ T-cell infiltration and proinflammatory/pro-tumour cytokines in the liver were significantly upregulated in Fah-/- mice during chronic liver injury, which was notably reversed by either metformin or atRA treatment. Regarding mechanisms, metformin regulated the decrease in Cyp26a1 enzyme expression and increased atRA expression via the AMPK/STAT3/Gadd45β/JNK/c-Jun pathway. CONCLUSIONS Metformin inhibits non-diabetic HCC by upregulating atRA levels and downregulating CD8+ T cells. This is the first reporting that the traditional drug metformin regulates the metabolite atRA via the Cyp26a1-involved pathway. The present study provides a potential application of metformin and atRA in non-diabetic HCC.
Collapse
Affiliation(s)
- Weizhi He
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
- Fudan University Shanghai Cancer Center, International Co‐Laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Shanghai Medical College of Fudan University, Institutes of Biomedical SciencesShanghai Key Laboratory of Medical EpigeneticsShanghaiChina
| | - Xicheng Wang
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Miaomiao Chen
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Chong Li
- Zhoupu Community Health Service Center of Pudong New AreaShanghaiChina
| | - Wenjian Chen
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Lili Pan
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Yangyang Cui
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Postgraduate Training Base of Shanghai East HospitalJinzhou Medical UniversityJinzhouLiaoningChina
| | - Zhao Yu
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Guoxiu Wu
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Yang Yang
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Mingyang Xu
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Zhaoxuan Dong
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Keming Ma
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| | - Jinghan Wang
- Department of Hepatobiliary and Pancreatic SurgeryShanghai East Hospital, Tongji UniversityShanghaiChina
| | - Zhiying He
- Institute for Regenerative Medicine, Ji'an Hospital, Shanghai East HospitalSchool of Life Sciences and TechnologyTongji University School of MedicineShanghaiChina
- Shanghai Engineering Research Center of Stem Cells Translational MedicineShanghaiChina
| |
Collapse
|
44
|
Yang Y, Liang Q, Chen Y, Cao Y, Zhuo Q, Liu B, Wang S. Aldehyde dehydrogenase 2 gene rs671 G>A polymorphism is associated with an increased risk of digestive tract cancer. J Int Med Res 2023; 51:3000605231206257. [PMID: 37871625 PMCID: PMC10594971 DOI: 10.1177/03000605231206257] [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: 04/15/2023] [Accepted: 09/21/2023] [Indexed: 10/25/2023] Open
Abstract
OBJECTIVE Acetaldehyde can accumulate in cells and form acetaldehyde-DNA adducts that result in digestive tract cancer development. Acetaldehyde dehydrogenase 2 (ALDH2) enzymatic activity is involved in this process. Here, we aimed to analyze the relationship between an ALDH2 gene polymorphism and the digestive tract cancer risk in the Hakka population in China. METHODS This was a retrospective study, with the ALDH2 rs671 genotype and medical record information collected from all subjects. The relationships between these factors, including various blood cell parameters, and digestive tract cancer susceptibility were analyzed. RESULTS Overall, 307 cancer patients and 317 controls were included. The cancer patients had significantly higher percentages with a history of smoking and drinking alcohol, as well as an increased platelet to lymphocyte ratio and lower lymphocyte to monocyte ratio, compared with the controls. The ALDH2 rs671 genotype and allele distributions were significantly different between the cancer patients and controls. Logistic regression analysis showed that the ALDH2 G/A genotype (G/A vs. G/G) and A/A genotype (A/A vs. G/G) in the co-dominant mode were risk factors for digestive tract cancer susceptibility. CONCLUSIONS ALDH2 rs671 G/A or A/A genotype carriers may have an increased risk of developing digestive tract cancers among the Hakka people.
Collapse
Affiliation(s)
- Yang Yang
- Department of Gastroenterology, Meizhou People's Hospital, Meizhou, China
| | - Qun Liang
- Department of Gastroenterology, Meizhou People's Hospital, Meizhou, China
| | - Yijin Chen
- Department of Gastroenterology, Meizhou People's Hospital, Meizhou, China
| | - Yu Cao
- Department of Gastrointestinal Surgery, Meizhou People's Hospital, Meizhou, China
| | - Qingqing Zhuo
- Department of Gastroenterology, Meizhou People's Hospital, Meizhou, China
| | - Boying Liu
- Department of Gastroenterology, Meizhou People's Hospital, Meizhou, China
| | - Shengbing Wang
- Department of Gastroenterology, Meizhou People's Hospital, Meizhou, China
| |
Collapse
|
45
|
Wen Y, Ma L, Ju C. Recent insights into the pathogenesis and therapeutic targets of chronic liver diseases. EGASTROENTEROLOGY 2023; 1:e100020. [PMID: 38074919 PMCID: PMC10704956 DOI: 10.1136/egastro-2023-100020] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 05/05/2023] [Indexed: 01/03/2025]
Abstract
Viral hepatitis, alcohol-associated liver disease (ALD) and non-alcoholic fatty liver disease (NAFLD) are the three major causes of chronic liver diseases, which account for approximately 2 million deaths per year worldwide. The current direct-acting antiviral drugs and vaccinations have effectively reduced and ameliorated viral hepatitis infection, but there are still no effective drug treatments for ALD, NAFLD and liver cancer due to the poor understanding of their pathogenesis. To better understand the pathogenesis, the fifth Chinese American Liver Society/Society of Chinese Bioscientists in America Hepatology Division Annual Symposium, which was held virtually on 21-22 October 2022, focused on the topics related to ALD, NAFLD and liver cancer. Here, we briefly highlight the presentations that focus on the current progress in basic and translational research in ALD, NAFLD and liver cancer. The roles of non-coding RNA, autophagy, extrahepatic signalling, macrophages, etc in liver diseases are deliberated, and the application of single-cell RNA sequencing in the study of liver disease is also discussed.
Collapse
Affiliation(s)
- Yankai Wen
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas McGovern Medical School, Houston, Texas, USA
| | - Lichun Ma
- Cancer Data Science Laboratory, National Cancer Institute Center for Cancer Research, Bethesda, Maryland, USA
- Liver Cancer Program, National Cancer Institute Center for Cancer Research, Bethesda, Maryland, USA
| | - Cynthia Ju
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas McGovern Medical School, Houston, Texas, USA
| |
Collapse
|
46
|
Manea I, Iacob R, Iacob S, Cerban R, Dima S, Oniscu G, Popescu I, Gheorghe L. Liquid biopsy for early detection of hepatocellular carcinoma. Front Med (Lausanne) 2023; 10:1218705. [PMID: 37809326 PMCID: PMC10556479 DOI: 10.3389/fmed.2023.1218705] [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: 05/08/2023] [Accepted: 08/30/2023] [Indexed: 10/10/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly prevalent and lethal cancer globally. Over 90% of HCC cases arise in the context of liver cirrhosis, and the severity of the underlying liver disease or advanced tumor stage at diagnosis significantly limits treatment options. Early diagnosis is crucial, and all guidelines stress the importance of screening protocols for HCC early detection as a public health objective. As serum biomarkers are not optimal for early diagnosis, liquid biopsy has emerged as a promising tool for diagnosis, prognostication, and patients' stratification for personalized therapy in various solid tumors, including HCC. While circulating tumor cells (CTCs) are better suited for personalized therapy and prognosis, cell-free DNA (cfDNA) and extracellular vesicle-based technologies show potential for early diagnosis, HCC screening, and surveillance protocols. Evaluating the added value of liquid biopsy genetic and epigenetic biomarkers for HCC screening is a key goal in translational research. Somatic mutations commonly found in HCC can be investigated in cfDNA and plasma exosomes as genetic biomarkers. Unique methylation patterns in cfDNA or cfDNA fragmentome features have been suggested as innovative tools for early HCC detection. Likewise, extracellular vesicle cargo biomarkers such as miRNAs and long non-coding RNAs may serve as potential biomarkers for early HCC detection. This review will explore recent findings on the utility of liquid biopsy for early HCC diagnosis. Combining liquid biopsy methods with traditional serological biomarkers could improve the overall diagnostic accuracy for early HCC detection.
Collapse
Affiliation(s)
- Ioana Manea
- “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
- Digestive Diseases and Liver Transplantation Center, Fundeni Clinical Institute, Bucharest, Romania
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, Bucharest, Romania
| | - Razvan Iacob
- “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
- Digestive Diseases and Liver Transplantation Center, Fundeni Clinical Institute, Bucharest, Romania
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, Bucharest, Romania
| | - Speranta Iacob
- “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
- Digestive Diseases and Liver Transplantation Center, Fundeni Clinical Institute, Bucharest, Romania
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, Bucharest, Romania
| | - Razvan Cerban
- “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
- Digestive Diseases and Liver Transplantation Center, Fundeni Clinical Institute, Bucharest, Romania
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, Bucharest, Romania
| | - Simona Dima
- “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
- Digestive Diseases and Liver Transplantation Center, Fundeni Clinical Institute, Bucharest, Romania
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, Bucharest, Romania
| | - Gabriel Oniscu
- Transplant Division, Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden
| | - Irinel Popescu
- Digestive Diseases and Liver Transplantation Center, Fundeni Clinical Institute, Bucharest, Romania
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, Bucharest, Romania
| | - Liliana Gheorghe
- “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
- Digestive Diseases and Liver Transplantation Center, Fundeni Clinical Institute, Bucharest, Romania
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, Bucharest, Romania
| |
Collapse
|
47
|
Liu Y, Liu T, Zhang F, Gao Y. Unraveling the Complex Interplay between Epigenetics and Immunity in Alcohol-Associated Liver Disease: A Comprehensive Review. Int J Biol Sci 2023; 19:4811-4830. [PMID: 37781509 PMCID: PMC10539712 DOI: 10.7150/ijbs.87975] [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: 07/11/2023] [Accepted: 08/26/2023] [Indexed: 10/03/2023] Open
Abstract
The mechanisms of immune dysfunction in alcohol-associated liver disease (ALD) have garnered growing research interest in recent times. Alcohol-mediated immune dysfunction has been implicated as a potential cause of ALD-associated microbial infection and inflammatory response. The immune microenvironment of an organism is essentially a complex network of interactions between immune cells, cytokines, extracellular matrix, and other immune-related molecules. This microenvironment is highly adaptive and responsive to environmental cues. Epigenetic reprogramming of the immune microenvironment has recently emerged as a key driver of ALD progression, particularly in the context of endotoxin tolerance and immune disorders. Although epigenetic modifications are known to play an important role in the regulation of the immune microenvironment in ALD, the specific mechanisms and molecular processes by which this regulation is achieved are yet to be fully understood. This paper aims to provide an overview of the current knowledge on the effects of alcohol consumption on epigenetics, with special focus on summarizing the data on the epigenetic regulatory mechanisms involved in the effects of alcohol consumption on the immune microenvironment. In addition, this paper aims to present a review of the epigenetic modifications involved in different forms of ALD. This review is expected to offer new perspectives for the diagnosis, treatment, monitoring, and prognostic assessment of ALD from an epigenetic perspective.
Collapse
Affiliation(s)
| | | | | | - Yanhang Gao
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, 130021, China
| |
Collapse
|
48
|
Xia P, Liu DH, Wang D, Wen GM, Zhao ZY. SLC3A2, as an indirect target gene of ALDH2, exacerbates alcohol-associated liver cancer via the sphingolipid biosynthesis pathway. Free Radic Biol Med 2023; 206:125-133. [PMID: 37406742 DOI: 10.1016/j.freeradbiomed.2023.07.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/07/2023]
Abstract
Excessive drinking is one of the main causes of liver cancer. In the process of alcohol metabolism, aldehyde dehydrogenase 2 (ALDH2) is the key enzyme of acetaldehyde metabolism. ALDH2 gene deficiency is positively associated with the risk of hepatocellular carcinoma (HCC). However, no studies have shown a connection between ALDH2 and another metabolic regulatory gene, SLC3A2. In this study, we analyzed the expression levels of ALDH2 and SLC3A2 in liver cancer tissues based on the TCGA database. Subsequently, we constructed ALDH2 knockout and SLC3A2 knock-in transgenic mice to check the roles of ALDH2 and SLC3A2 in tumorigenesis in vivo. In addition, we examined the mechanisms of ALDH2 and SLC3A2 in HCC cells using small RNA interference technology. Consistent with previous studies, we also confirmed the functions of ALDH2 in inhibiting hepatocarcinogenesis, while SLC3A2 had the opposite effect. The main finding of this study is that ALDH2 inhibited BSG expression through the TGF-β1 pathway, which indirectly inhibited SLC3A2 expression; subsequently, the sphingolipid metabolism pathway was also inhibited in HCC cells. Therefore, SLC3A2 is a novel target for HCC treatment.
Collapse
Affiliation(s)
- Pu Xia
- Biological Anthropology Institute, Jinzhou Medical University, Jinzhou, Liaoning, PR China; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.
| | - Da-Hua Liu
- Biological Anthropology Institute, Jinzhou Medical University, Jinzhou, Liaoning, PR China
| | - Dan Wang
- College of Human Kinesiology, Shenyang Sport University, Shenyang, Liaoning, PR China
| | - Gui-Min Wen
- Department of Community Nursing, College of Nursing, Jinzhou Medical University, Jinzhou, Liaoning, PR China
| | - Zhen-Ying Zhao
- Department of Pharmacy, Tianjin Union Medical Center, Tianjin, PR China
| |
Collapse
|
49
|
Chong ML, Knight J, Peng G, Ji W, Chai H, Lu Y, Wu S, Li P, Hu Q. Integrated exome sequencing and microarray analyses detected genetic defects and underlying pathways of hepatocellular carcinoma. Cancer Genet 2023; 276-277:30-35. [PMID: 37418972 DOI: 10.1016/j.cancergen.2023.06.002] [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: 02/23/2023] [Revised: 05/25/2023] [Accepted: 06/26/2023] [Indexed: 07/09/2023]
Abstract
We performed whole exome sequencing (WES) and microarray analysis to detect somatic variants and copy number alterations (CNAs) for underlying mechanisms in a case series of hepatocellular carcinoma (HCC) with paired DNA samples from tumor and adjacent nontumor tissues. Clinicopathologic findings based on Edmondson-Steiner (E-S) grading, Barcelona-Clinic Liver Cancer (BCLC) stages, recurrence, and survival status and their associations with tumor mutation burden (TMB) and CNA burden (CNAB) were evaluated. WES from 36 cases detected variants in the TP53, AXIN1, CTNNB1, and SMARCA4 genes, amplifications of the AKT3, MYC, and TERT genes, and deletions of the CDH1, TP53, IRF2, RB1, RPL5, and PTEN genes. These genetic defects affecting the p53/cell cycle control, PI3K/Ras, and β-catenin pathways were observed in approximately 80% of cases. A germline variant in the ALDH2 gene was detected in 52% of the cases. Significantly higher CNAB in patients with poor prognosis by E-S grade III, BCLC stage C, and recurrence than patients with good prognosis by grade III, stage A, grade III and nonrecurrence was noted. Further analysis on a large case series to correlate genomic profiling with clinicopathologic classifications could provide evidence for diagnostic interpretation, prognostic prediction, and target intervention on involved genes and pathways.
Collapse
Affiliation(s)
- Mei Ling Chong
- Department of Genetics, School of Medicine, Yale University, New Haven, CT, USA
| | - James Knight
- Department of Genetics, School of Medicine, Yale University, New Haven, CT, USA; Yale Center for Genome Analysis, School of Medicine, Yale University, New Haven, CT, USA
| | - Gang Peng
- Department of Genetics, School of Medicine, Yale University, New Haven, CT, USA; Department of Medical and Molecular Genomics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Weizhen Ji
- Department of Genetics, School of Medicine, Yale University, New Haven, CT, USA
| | - Hongyan Chai
- Department of Genetics, School of Medicine, Yale University, New Haven, CT, USA
| | - Yufei Lu
- Department of Cell Biology and Genetics, School of Pre-Clinical Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Shengming Wu
- Department of Pathology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Peining Li
- Department of Genetics, School of Medicine, Yale University, New Haven, CT, USA; Yale Center for Genome Analysis, School of Medicine, Yale University, New Haven, CT, USA
| | - Qiping Hu
- Department of Cell Biology and Genetics, School of Pre-Clinical Medicine, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
50
|
Wu YJ, Wang J, Zhang P, Yuan LX, Ju LL, Wang HX, Chen L, Cao YL, Cai WH, Ni Y, Li M. PIWIL1 interacting RNA piR-017724 inhibits proliferation, invasion, and migration, and inhibits the development of HCC by silencing PLIN3. Front Oncol 2023; 13:1203821. [PMID: 37503320 PMCID: PMC10369847 DOI: 10.3389/fonc.2023.1203821] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/26/2023] [Indexed: 07/29/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) accounts for the majority of primary liver cancers. Worldwide, liver cancer is the fourth most common cause of cancer-related death. Recent studies have found that PIWI-interacting RNAs (piRNAs) participate in the occurrence and development of various tumors and are closely related to the growth, invasion, metastasis and prognosis of malignant tumors. Studies on the role and functional mechanism of piRNAs in HCC development and progression are limited. Methods Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) were used to detect the expression of piR-017724 in both HCC tissues and cells. Based on the clinical data of HCC patients, the clinical and prognostic value of piR-017724 was further analyzed. Then, targeted silencing and overexpressing of piR-017724 in HCC cells was further used to examine the biological functions of piR-017724. In addition, the downstream target protein of piR-017724 was predicted and validated through high-throughput sequencing and public databases. Results The piR-017724 was significantly downregulated in HCC tissues and cells, and the downregulation of piR-017724 was associated with tumor stage and poor prognosis in HCC. The piR-017724 inhibitor promoted the proliferation, migration and invasion of HCC cells, while the piR-017724 mimic had the opposite effect. However, the piR-017724 did not affect apoptosis of HCC cells. High-throughput sequencing and qRT-PCR confirmed a reciprocal relationship between piR-017724 and PLIN3. Therefore, we speculate that piR-017724 may inhibit the development and progression of HCC by affecting the downstream protein PLIN3. Conclusions Our study shows that piR-017724, which is lowly expressed in HCC, inhibits the proliferation, migration and invasion of HCC cells and may affect the development of hepatocellular liver cancer through PLIN3, which provides new insights into the clinical application of piR-017724 in the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yi-Jing Wu
- Medical School of Nantong University, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Jie Wang
- Medical School of Nantong University, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Peng Zhang
- Nantong Institute of Liver Disease, Department of Hepatobiliary Surgery, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Liu-Xia Yuan
- Nantong Institute of Liver Disease, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Lin-Ling Ju
- Nantong Institute of Liver Disease, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Hui-Xuan Wang
- Nantong Institute of Liver Disease, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Lin Chen
- Nantong Institute of Liver Disease, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Ya-Li Cao
- Preventive Health Department, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Wei-Hua Cai
- Department of Hepatobiliary Surgery, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Yi Ni
- Thyroid and Breast Surgery, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Min Li
- Integrating Traditional Chinese Medicine with Hepatology, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| |
Collapse
|