1
|
Wu H, Chen J, Guo S, Deng J, Zhou Z, Zhang X, Qi T, Yu F, Yang Q. Advances in the acting mechanism and treatment of gut microbiota in metabolic dysfunction-associated steatotic liver disease. Gut Microbes 2025; 17:2500099. [PMID: 40394806 PMCID: PMC12101596 DOI: 10.1080/19490976.2025.2500099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/17/2025] [Accepted: 04/25/2025] [Indexed: 05/22/2025] Open
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease(MASLD) is increasing in prevalence worldwide and has become the greatest potential risk for cirrhosis and hepatocellular liver cancer. Currently, the role of gut microbiota in the development of MASLD has become a research hotspot. The development of MASLD can affect the homeostasis of gut microbiota, and significant changes in the composition or abundance of gut microbiota and its metabolite abnormalities can influence disease progression. The regulation of gut microbiota is an important strategy and novel target for the treatment of MASLD with good prospects. In this paper, we summarize the role of gut microbiota and its metabolites in the pathogenesis of MASLD, and describe the potential preventive and therapeutic efficacy of gut microbiota as a noninvasive marker to regulate the pathogenesis of MASLD based on the "gut-hepatic axis", which will provide new therapeutic ideas for the clinic.
Collapse
Affiliation(s)
- Huaying Wu
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - Jingjing Chen
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - Shuyuan Guo
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jinhao Deng
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - Zimeng Zhou
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - Xuan Zhang
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - TianTian Qi
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
| | - Fei Yu
- Department of Spine Surgery, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qi Yang
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
2
|
Rittavee Y, Ratanaprasert N, Ahmed S, Anekpuritanang T, Muanprasat C, Pongsapich W, Jinawath N. Biomarkers for predicting second primary malignancy risk in head and neck squamous cell carcinoma: An integrated molecular perspective. Crit Rev Oncol Hematol 2025; 210:104711. [PMID: 40157583 DOI: 10.1016/j.critrevonc.2025.104711] [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: 02/05/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Second primary malignancies (SPMs) threaten long-term survival in head and neck squamous cell carcinoma (HNSCC). Advancing our understanding of the molecular events driving these secondary tumors is essential. This review has explicated molecular drivers of SPM development, including epigenetic alterations such as DNA hypermethylation, genetic polymorphisms affecting detoxification pathways, and shifts in gene and protein expression profiles. Disruptions in the p53 signaling pathway, immune-related pathways, and impairments in glutathione S-transferase-mediated detoxification, emerge as central contributors to SPM risk. Additionally, direct comparisons of tumor specimens with adjacent or distant normal mucosa highlight field cancerization biomarkers, underscoring widespread carcinogen-induced damage. Loss of heterozygosity at chromosome arm 13q, p53 overexpression in tumor-distant epithelia, and proteomic abnormalities in ostensibly healthy mucosa collectively promote a tumor-prone field that encourages the formation of independent secondary tumors. This interplay underscores a multifactorial landscape of SPM pathogenesis, involving genetic susceptibility, environmental exposures, and intricate epigenetic and transcriptomic networks. By recognizing and validating reliable biomarkers, clinicians may pinpoint high-risk patients with greater precision, intervene earlier, and customize follow-up protocols and treatment regimens. Ultimately, translating these insights into routine practice promises a more proactive, individualized approach to preventing SPMs in HNSCC.
Collapse
Affiliation(s)
- Yutthana Rittavee
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand
| | - Narin Ratanaprasert
- Department of Otorhinolaryngology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Thailand
| | - Saad Ahmed
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand
| | | | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand
| | - Warut Pongsapich
- Department of Otorhinolaryngology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Thailand
| | - Natini Jinawath
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand; Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand; Integrative Computational Bioscience (ICBS) Center, Mahidol University, Nakhon Pathom, Thailand.
| |
Collapse
|
3
|
Seitz HK. A narrative review on alcohol and alimentary tract cancer with special emphasis on acetaldehyde and oxidative stress. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2025. [PMID: 40378880 DOI: 10.1055/a-2588-6849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2025]
Abstract
Approximately 4% of all cancer cases worldwide are caused by alcohol consumption (oropharynx, larynx, esophagus, stomach, colorectum, liver and the female breast). Various mechanisms contribute to ethanol-mediated carcinogenesis, including the action of acetaldehyde, the first metabolite of ethanol oxidation and oxidative stress primarily promoted through the induction of cytochrome P4502E1. Acetaldehyde is toxic and carcinogenic, binds to DNA and proteins, inhibits the oxidative defense- and the nuclear repair system, and prevents DNA methylation. High levels of acetaldehyde occur through increased production in the presence of a hyperactive alcohol dehydrogenase (ADH1C*1,1) or decreased degradation in the presence of low active aldehyde dehydrogenase (ALDH2*1,2). In addition, microbes of the upper alimentary tract and the colorectum effectively produce acetaldehyde from ethanol. In addition, ethanol induces cytochrome P4502E1 resulting in an enhanced ethanol metabolism and the generation of reactive oxygen species (ROS). ROS may cause lipid peroxidation (LPO) with the LPO-products 4-hydroxynonenal or malondialdehyde, which may form highly carcinogenic etheno DNA-adducts CYP2E1 is also involved in the activation of a variety of dietary and tobacco procarcinogens and in the degradation of retinoic acid. Alcohol also influences tumor promotion, such as epigenetics with a change in DNA methylation and histone modification, and affects a variety of cancer genes and signaling pathways. Preventive measures include reducing alcohol consumption, quitting smoking and keeping good oral hygiene. Alcohol consumers - especially when they smoke or belong to genetic risk groups - should be regularly checked for cancer of the upper alimentary tract, for alcohol- associated liver disease, and for breast cancer. Cessation or reduction of alcohol consumption definitively reduces cancer risk.
Collapse
Affiliation(s)
- Helmut Karl Seitz
- Centre of Liver- and Alcohol Diseases, ETHIANUM Klinik, Heidelberg, Germany
- Internal Medicine, Gastroenetrology, Alcohol Research, Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
4
|
Quammie S, Crooks CJ, Aliyu A, Aithal GP, Aravinthan AD. Chronic pancreatitis and extra pancreatic cancers- A systematic review and meta analysis. Pancreatology 2025:S1424-3903(25)00088-2. [PMID: 40382256 DOI: 10.1016/j.pan.2025.05.003] [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: 10/18/2024] [Revised: 03/11/2025] [Accepted: 05/10/2025] [Indexed: 05/20/2025]
Abstract
INTRODUCTION Chronic pancreatitis (CP) is a known risk factor for pancreatic cancer; however its association to extra pancreatic (EP) cancers remains inadequately explored. The aim of this systematic review is to investigate the evidence for CP as a risk factor for developing EP cancers. METHOD Electronic search was conducted on Ovid Medline, EMBASE and Scopus from inception to January 27, 2024 to identify patients with CP who developed EP cancers. Prevalence and incidence of each cancer were calculated where possible from the reported numbers. A random effects meta-analysis was used to pool prevalence, incidence and hazard ratios (HR) of each EP cancer. Heterogeneity was assessed using I2. PROSPERO registration: CRD42024543050. RESULTS Sixteen (16) studies consisting of 117,163 CP patients met the eligibility criteria. 4015 (3.4%) patients developed 4019 EP cancers. The overall annual prevalence and incidence of EP cancers were 7962 (95% CI 5044-10880) per 100,000 CP patients and 1039 (95% CI 649-1663) per 100,000 person years. Lung cancer had the highest annual prevalence - (1540 (95% CI 667-2413) per 100,000 CP patients) and incidence (260 (95% CI 120-390) per 100,000 person years). The pooled HR were 1.31 (95% CI 1.03-1.66) and 1.58 (95% CI 1.27-1.98) for adjusted lung cancer and crude liver cancer in patients with CP compared to patients without CP, respectively. CONCLUSION Patients with CP have an increased risk of developing EP cancers compared to patients without CP, in particular lung and liver cancer which had the highest relative risk. Shared risk factor modifications, such as smoking cessation and alcohol reduction, could lower the risk of common EP cancers. Further, implementing non-invasive screening measures may aid in early diagnosis in this high-risk group.
Collapse
Affiliation(s)
- Shauntelle Quammie
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, UK; Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Colin John Crooks
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, UK; Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Abdulsalam Aliyu
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Guruprasad P Aithal
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, UK; Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Aloysious D Aravinthan
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, UK; Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK.
| |
Collapse
|
5
|
Menghani SV. Carcinogenetic mechanisms employed by the oral microbiome: A narrative review. Am J Med Sci 2025; 369:556-561. [PMID: 39788425 DOI: 10.1016/j.amjms.2025.01.001] [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: 07/27/2024] [Revised: 12/15/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Cancers of the oral cavity, lip, salivary gland, and oropharynx cause substantial global disease burden. While tobacco-use and alcohol use are highly associated with oral cancers, the rising incidence of disease in patients who do not use tobacco or alcohol points to additional carcinogenic risk factors. Chronic inflammation, disruption of the oral microbiome, and dysbiosis are becoming more widely implicated in the pathogenesis of oral cancer. Several studies have identified specific bacterial species enriched in patients with oral cancer, including Porphyromonas gingivalis and Fusobacterium nucleatum. In this narrative review, we describe potential carcinogenic mechanisms exhibited by these species and other microbes in the development of oral cancer.
Collapse
Affiliation(s)
- Sanjay V Menghani
- University of Arizona College of Medicine - Tucson, AZ, USA; Medical Scientist Training MD-PhD Program, University of Arizona College of Medicine Tucson, AZ, USA.
| |
Collapse
|
6
|
Naudin S, Wang M, Dimou N, Ebrahimi E, Genkinger J, Adami HO, Albanes D, Babic A, Barnett M, Bogumil D, Cai H, Chen C, Eliassen AH, Freudenheim JL, Gierach G, Giovannucci EL, Gunter MJ, Håkansson N, Hirabayashi M, Hou T, Huang BZ, Huang WY, Jayasekara H, Jones ME, Katzke VA, Koh WP, Lacey JV, Lagerros YT, Larsson SC, Liao LM, Lo K, Loftfield E, MacInnis RJ, Männistö S, McCullough ML, Miller A, Milne RL, Moore SC, Mucci LA, Neuhouser ML, Patel AV, Platz EA, Prizment A, Robien K, Rohan TE, Sacerdote C, Sandin S, Sawada N, Schoemaker M, Shu XO, Sinha R, Snetselaar L, Stampfer MJ, Stolzenberg-Solomon R, Thomson CA, Tjønneland A, Um CY, van den Brandt PA, Visvanathan K, Wang SS, Wang R, Weiderpass E, Weinstein SJ, White E, Willett W, Woslk A, Wolpin BM, Yaun SSS, Yuan C, Yuan JM, Zheng W, Brennan P, Smith-Warner SA, Ferrari P. Alcohol intake and pancreatic cancer risk: An analysis from 30 prospective studies across Asia, Australia, Europe, and North America. PLoS Med 2025; 22:e1004590. [PMID: 40392909 PMCID: PMC12091891 DOI: 10.1371/journal.pmed.1004590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 03/25/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Alcohol is a known carcinogen, yet the evidence for an association with pancreatic cancer risk is considered as limited or inconclusive by international expert panels. We examined the association between alcohol intake and pancreatic cancer risk in a large consortium of prospective studies. METHODS AND FINDINGS Population-based individual-level data was pooled from 30 cohorts across four continents, including Asia, Australia, Europe, and North America. A total of 2,494,432 participants without cancer at baseline (62% women, 84% European ancestries, 70% alcohol drinkers [alcohol intake ≥ 0.1 g/day], 47% never smokers) were recruited between 1980 and 2013 at the median age of 57 years and 10,067 incident pancreatic cancer cases were recorded. In age- and sex-stratified Cox proportional hazards models adjusted for smoking history, diabetes status, body mass index, height, education, race and ethnicity, and physical activity, pancreatic cancer hazard ratios (HR) and 95% confidence intervals (CI) were estimated for categories of alcohol intake and in continuous for a 10 g/day increase. Potential heterogeneity by sex, smoking status, geographic regions, and type of alcoholic beverage was investigated. Alcohol intake was positively associated with pancreatic cancer risk, with HR30-to-<60 g/day and HR≥60 g/day equal to 1.12 (95% CI [1.03,1.21]) and 1.32 (95% CI [1.18,1.47]), respectively, compared to intake of 0.1 to <5 g/day. A 10 g/day increment of alcohol intake was associated with a 3% increased pancreatic cancer risk overall (HR: 1.03; 95% CI [1.02,1.04]; pvalue < 0.001) and among never smokers (HR: 1.03; 95% CI [1.01,1.06]; pvalue = 0.006), with no evidence of heterogeneity by sex (pheterogeneity = 0.274) or smoking status (pheterogeneity = 0.624). Associations were consistent in Europe-Australia (HR10 g/day = 1.03, 95% CI [1.00,1.05]; pvalue = 0.042) and North America (HR10 g/day = 1.03, 95% CI [1.02,1.05]; pvalue < 0.001), while no association was observed in cohorts from Asia (HR10 g/day = 1.00, 95% CI [0.96,1.03]; pvalue = 0.800; pheterogeneity = 0.003). Positive associations with pancreatic cancer risk were found for alcohol intake from beer (HR10 g/day = 1.02, 95% CI [1.00,1.04]; pvalue = 0.015) and spirits/liquor (HR10 g/day = 1.04, 95% CI [1.03,1.06]; pvalue < 0.001), but not wine (HR10 g/day = 1.00, 95% CI [0.98,1.03]; pvalue = 0.827). The differential associations across geographic regions and types of alcoholic beverages might reflect differences in drinking habits and deserve more investigations. CONCLUSIONS Findings from this large-scale pooled analysis support a modest positive association between alcohol intake and pancreatic cancer risk, irrespective of sex and smoking status. Associations were particularly evident for baseline alcohol intake of at least 15 g/day in women and 30 g/day in men.
Collapse
Grants
- 75N92021D00002 NHLBI NIH HHS
- U01 CA176726 NCI NIH HHS
- UM1 CA173640 NCI NIH HHS
- 75N92021D00001 NHLBI NIH HHS
- R01 AA024770 NIAAA NIH HHS
- R01 CA039742 NCI NIH HHS
- P30 CA033572 NCI NIH HHS
- U01 CA199277 NCI NIH HHS
- UM1 CA164917 NCI NIH HHS
- U01 HL145386 NHLBI NIH HHS
- 75N92021D00005 WHI NIH HHS
- U01 CA063673 NCI NIH HHS
- U01 CA167462 NCI NIH HHS
- P01 CA087969 NCI NIH HHS
- R01 CA144034 NCI NIH HHS
- U01 CA167552 NCI NIH HHS
- U01 CA086308 NCI NIH HHS
- 75N92021D00003 WHI NIH HHS
- UM1 CA186107 NCI NIH HHS
- P30 CA023100 NCI NIH HHS
- R01 CA077398 NCI NIH HHS
- UM1 CA167462 NCI NIH HHS
- U01 CA164973 NCI NIH HHS
- 75N92021D00004 WHI NIH HHS
- U01 AG018033 NIA NIH HHS
- UM1 CA182876 NCI NIH HHS
- National Institute on Alcohol Abuse and Alcoholism
- National Institutes of Health
- National Cancer Institute
- National Institute on Aging
- Centers for Disease Control and Prevention
- Canadian Cancer Society, the Department of National Health and Welfare
- National Cancer Institute of Canada
- Alberta Heritage Fund for Cancer Research
- Manitoba Health Services Commission
- Medical Research Council of Canada
- Ministry of Health and Social Services of Québec
- Nova Scotia Department of Health
- Ontario Ministry of Health.
- Swedish Research Council
- The American Cancer Society
- International Agency for Research on Cancer
- National Institute for Health and Care Research
- Danish Cancer Society
- Ligue Contre le Cancer, Institut Gustave Roussy, Mutuelle Générale de l’Education Nationale, and Institut National de la Santé et de la Recherche Médicale
- German Cancer Aid, German Cancer Research Center (DKFZ), German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), and Federal Ministry of Education and Research (BMBF)
- Associazione Italiana per la Ricerca sul Cancro (AIRC-Italy), Compagnia di San Paolo, and National Research Council
- Dutch Ministry of Public Health, Welfare and Sports (VWS), Netherlands Cancer Registry (NKR), LK Research Funds, Dutch Prevention Funds, Zorg Onderzoek Nederland (ZON), World Cancer Research Fund (WCRF), and Statistics Netherlands
- Health Research Fund (FIS)—Instituto de Salud Carlos III (ISCIII), Regional Governments of Andalucia, Asturias, Basque Country, Murcia and Navarra, and the Catalan Institute of Oncology—ICO
- Swedish Cancer Society, Swedish Research Council, and County Councils of Skane and Vasterbotten
- Cancer Research UK
- Medical Research Council
- Breast Cancer Now
- National Institute for Health and Care Research (NIHR) Biomedical Research Centre at The Royal Marsden NHS Foundation Trust and the Institute of Cancer Research
- National Cancer Center Research and Development Fund
- Ministry of Health, Labour and Welfare of Japan
- VicHealth and Cancer Council Victoria
- Australian National Health and Medical Research Council
- Intramural Research Program, Division of Cancer Epidemiology and Genetics
- Dutch Cancer Society and World Cancer Research Fund
- Division of Cancer Prevention, National Cancer Institute
- Center for Disease Control and Prevention, National Program for Central Registries
- Singapore Ministry of Health’s National Medical Research Council
Collapse
Affiliation(s)
- Sabine Naudin
- International Agency for Research on Cancer, World Health Organization, Lyon, France
- UPS, UVSQ, National Institute of Health and Medical Research, Gustave Roussy, Centre for research in epidemiology and population health, Villejuif, France
| | - Molin Wang
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, Massachusetts, United States of America
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Niki Dimou
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Elmira Ebrahimi
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Jeanine Genkinger
- Department of Epidemiology, Mailman School of Public Health, Columbia University Medical Center, New York, New York, United States of America
- Cancer Epidemiology Population Sciences Program, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York, United States of America
| | - Hans-Olov Adami
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Clinical Effectiveness Group, Institute of Health and Society, University of Oslo, Oslo, Norway
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, United States of America
| | - Ana Babic
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Matt Barnett
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - David Bogumil
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, California, United States of America
| | - Hui Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Chu Chen
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - A. Heather Eliassen
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Jo L. Freudenheim
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York, United States of America
| | - Gretchen Gierach
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, United States of America
| | - Edward L. Giovannucci
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Marc J. Gunter
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Niclas Håkansson
- Department of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mayo Hirabayashi
- Division of Prevention, National Cancer Center Institute for Cancer Control, Tokyo, Japan
| | - Tao Hou
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Brian Z. Huang
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, California, United States of America
| | - Wen-Yi Huang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, United States of America
| | - Harindra Jayasekara
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Michael E. Jones
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Verena A. Katzke
- Division of Cancer Epidemiology, Nutritional Epidemioloy, German Cancer Research Center, Heidelberg, Germany
| | - Woon-Puay Koh
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore, Singapore
| | - James V. Lacey
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope, Duarte, California, United States of America
| | - Ylva Trolle Lagerros
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Obesity, Academic Specialist Center, Stockholm, Sweden
| | - Susanna C. Larsson
- Department of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Linda M. Liao
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, United States of America
| | - Kenneth Lo
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Erikka Loftfield
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, United States of America
| | - Robert J. MacInnis
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Satu Männistö
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Marjorie L. McCullough
- Department of Population Science, American Cancer Society, Atlanta, Georgia, United States of America
| | - Anthony Miller
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Roger L. Milne
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Steven C. Moore
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, United States of America
| | - Lorelei A. Mucci
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, Massachusetts, United States of America
- Discovery Sciences, American Cancer Society, Atlanta, Georgia, United States of America
| | - Marian L. Neuhouser
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Alpa V. Patel
- Department of Population Science, American Cancer Society, Atlanta, Georgia, United States of America
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
| | - Anna Prizment
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, and the University of Minnesota Masonic Cancer Center, Minneapolis, Minnesota, United States of America
| | - Kim Robien
- Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, George Washington University, Washington District of Columbia, United States of America
| | - Thomas E. Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Città della Salute e della Scienza University-Hospital and Center for Cancer Prevention, Turin, Italy
| | - Sven Sandin
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Seaver Center for Autism Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Norie Sawada
- Division of Cohort Research, National Cancer Center Institute for Cancer Control, Tokyo, Japan
| | | | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Rashmi Sinha
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, United States of America
| | | | - Meir J. Stampfer
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, Massachusetts, United States of America
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Rachael Stolzenberg-Solomon
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, United States of America
| | - Cynthia A. Thomson
- Mel & Enid Zuckerman College of Public Health, University of Arizona Cancer Center, Tucson, Arizona, United States of America
| | - Anne Tjønneland
- Danish Cancer Institute, Diet, Cancer and Health, Copenhagen, Denmark
| | - Caroline Y. Um
- Department of Population Science, American Cancer Society, Atlanta, Georgia, United States of America
| | | | - Kala Visvanathan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland, United States of America
| | - Sophia S. Wang
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope, Duarte, California, United States of America
| | - Renwei Wang
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, Pennsylvania, United States of America
| | - Elisabete Weiderpass
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Stephanie J. Weinstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, United States of America
| | - Emily White
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Walter Willett
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Alicja Woslk
- Department of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Brian M. Wolpin
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Shiaw-Shyuan S. Yaun
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Chen Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jian-Min Yuan
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, Pennsylvania, United States of America
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Paul Brennan
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Stephanie A. Smith-Warner
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Pietro Ferrari
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| |
Collapse
|
7
|
Zhou XY, Zhang NC, Zhang XN, Sun XD, Ruan ZL, Yang Q, Hu MM, Shu HB. The carcinogenic metabolite acetaldehyde impairs cGAS activity to negatively regulate antiviral and antitumor immunity. Cancer Lett 2025; 617:217615. [PMID: 40056967 DOI: 10.1016/j.canlet.2025.217615] [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: 12/27/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
The cGAS-MITA/STING pathway plays critical roles in both host defense against DNA virus and intrinsic antitumor immunity by sensing viral genomic DNA or dis-located mitochondrial/cellular DNA. Whether carcinogenic metabolites can target the cGAS-MITA axis to promote tumorigenesis is unknown. In this study, we identified acetaldehyde, a carcinogenic metabolite, as a suppressor of the cGAS-MITA pathway. Acetaldehyde inhibits the DNA virus herpes simplex virus 1 (HSV-1)- and transfected DNA-triggered but not cGAMP-induced activation of downstream components and induction of downstream effector genes. Mechanistically, acetaldehyde impairs the binding of cGAS to DNA as well as the phase separation of the cGAS-DNA complex in cells. In mouse models, acetaldehyde inhibits antiviral cytokine production, promotes viral replication and lethality upon HSV-1 infection. In a colorectal tumor xenograft model, acetaldehyde promotes tumor growth and inhibits CD8+ T cell infiltration by targeting cGAS in both the tumor cells and immune cells in mice. Bioinformatic analysis indicates that expression of acetaldehyde dehydrogenase 2 (ALDH2), which converts acetaldehyde to acetic acid, is negatively correlated with stimulatory immune signatures in clinical colorectal tumors, and higher ALDH2 expression exhibits better prognosis of colorectal cancer patients. Collectively, our results suggest that acetaldehyde impairs cGAS activity to inhibit the cGAS-MITA axis, which contributes to its effects on carcinogenesis.
Collapse
Affiliation(s)
- Xiao-Yue Zhou
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology, State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei, China
| | - Nian-Chao Zhang
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology, State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei, China
| | - Xia-Nan Zhang
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology, State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei, China
| | - Xue-Dan Sun
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology, State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei, China
| | - Zi-Lun Ruan
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology, State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei, China
| | - Qing Yang
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology, State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei, China
| | - Ming-Ming Hu
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology, State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei, China.
| | - Hong-Bing Shu
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology, State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
8
|
Guo L, Li X. Nurse night shift work and risk of gastrointestinal cancers. Front Public Health 2025; 13:1532623. [PMID: 40356833 PMCID: PMC12066619 DOI: 10.3389/fpubh.2025.1532623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
The prevalence of night-shift employment is on the rise among full-time and part-time workers globally. Those engaged in night-shift work encounter various biological challenges, including exposure to artificial light during nighttime and disruptions to their circadian rhythms. These factors, along with changes in daily routines and activities, may pose significant risks to the health of night workers. Notably, the number of individuals working overtime or on night shifts has increased across various sectors, particularly in transportation, healthcare, and manufacturing. The International Agency for Research on Cancer (IARC) has classified night-shift work as probably carcinogenic to humans (IARC Group 2A). Subsequent research has identified several potential mechanisms through which night-shift work may contribute to carcinogenicity: (1) disruption of circadian rhythms, (2) suppression of melatonin levels due to nighttime light exposure, (3) physiological alterations, (4) lifestyle changes, and (5) reduced vitamin D levels resulting from inadequate sunlight exposure. Colorectal cancer (CRC) poses a significant public health challenge, ranking as the second leading cause of cancer-related death worldwide in 2020. Other than CRC, other gastrointestinal cancers are also creating a great global health issue because of their morbidity and mortality rates. In this review, we highlight the role of night shifts in disturbing circadian rhythm and how this action leads to carcinogenesis in the GI tract.
Collapse
Affiliation(s)
- Lin Guo
- Medical Simulated Center, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xiaojun Li
- School of Nursing, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| |
Collapse
|
9
|
Ko HH, Wu FY, Chen YS, Lin W, Fang CK, Bai CH, Lin HY, Kao HF, Cheng SJ. Sex differences in prognostic factors and genomic variations in oral squamous cell carcinoma: A 5-year retrospective study. J Dent Sci 2025; 20:1086-1094. [PMID: 40224086 PMCID: PMC11993083 DOI: 10.1016/j.jds.2024.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 12/17/2024] [Indexed: 04/15/2025] Open
Abstract
Background/purpose This study examined the prognostic factors and genomic variations in oral squamous cell carcinoma (OSCC) among male and female patients, focusing on the rising incidence of OSCC in women. Materials and methods Using data from 98 OSCC cases treated at National Taiwan University Hospital between 2013 and 2018, the study analyzed the patient cohort, clinical characteristics, and genomic profiles. Results The Female patients had a higher incidence of tongue cancer, while the male patients were prone to have buccal cancer. Key prognostic factors included age over 55 years, tongue cancer, alcohol use in female patients as well as the buccal cancer, betel chewing, and smoking in male patients. Notably, women with tongue OSCC or without oral habits had poorer 5-year survival rates. Genomic analysis revealed the males with high-risk habits had elevated antigen-processing and reactive oxygen gene sets, whereas the low-risk females showed dysregulation in metabolic pathways. Immunologically, the female patients had fewer naïve B cells and higher suppressive M2 macrophages. Conclusion Our findings highlight distinct sex-related OSCC prognosis differences and suggest that personalized treatments targeting specific risk factors and genomic characteristics may improve the clinical outcomes, particularly for the female OSCC patients.
Collapse
Affiliation(s)
- Hui-Hsin Ko
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
- School of Dentistry, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan
- National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Fang-Yu Wu
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, Jinshan Branch, New Taipei, Taiwan
| | - Ya-Syuan Chen
- Department of Health Care Administration, Chang Jung Christian University, Tainan, Taiwan
| | - Wender Lin
- Department of Health Care Administration, Chang Jung Christian University, Tainan, Taiwan
| | - Chun-Kai Fang
- Hospice and Palliative Care Center and Department of Psychiatry, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Chyi-Huey Bai
- Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Hung-Ying Lin
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
- School of Dentistry, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan
| | - Hsiang-Fong Kao
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- Institute of Oncology, National Taiwan University, Taipei, Taiwan
| | - Shih-Jung Cheng
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
- School of Dentistry, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
10
|
Zakhari S, Neuman M, Seitz HK. The role of cytochrome P4502E1 in ethanol mediated diseases: a narrative update. Alcohol Alcohol 2025; 60:agaf014. [PMID: 40192654 DOI: 10.1093/alcalc/agaf014] [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: 01/19/2025] [Revised: 02/21/2025] [Accepted: 03/08/2025] [Indexed: 05/17/2025] Open
Abstract
Cytochrome P450 (CYPs) superfamily of enzymes metabolize thousands of endogenous and exogenous substrates including ethanol. Results: Cytochrome P4502E1 (CYP2E1) is involved in ethanol metabolism as part of the so-called microsomal ethanol metabolizing system, in the metabolism of fatty acids and some drugs such as acetaminophen and isoniazid, and in the activation of a variety of procarcinogens (PCs). Chronic ethanol consumption induces CYP2E1 which may result in an enhanced metabolism of these drugs to their toxic intermediates, and in the generation of carcinogens. In addition, ethanol oxidation increases and is associated with the generation of reactive oxygen species (ROS). This oxidative stress is an important driver for the development of alcohol-associated liver disease (AALD) and alcohol-mediated cancer (AMC). ROS may bind directly to proteins and to DNA. ROS may also lead to lipid peroxidation (LPO) with the generation of LPO products. These LPO products may bind to DNA forming etheno-DNA adducts. Cell culture studies as well as animal experiments have shown that CYP2E1 knock-out animals or the inhibition of CYP2E1 by chemicals results in a significant improvement of liver histology. CYP2E1 is also involved in pathogenesis of hepatic steatosis and fibrosis. More recent studies in patients with AALD have demonstrated an improvement of serum transaminase activities when CYP2E1 was inhibited by clomethiazole. In addition to its role in the generation of ROS, CYP2E1 also enhances the activation of PCs and decreases the level of retinol and retinoic acid in the liver. Conclusion: Inhibition of CYP2E1 may improve AALD and may inhibit AMC.
Collapse
Affiliation(s)
- Samir Zakhari
- Independent Researcher, Washington, DC, University Park, 20782, USA
| | - Manuela Neuman
- In Vitro Drug Safety and Biotechnology, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Helmut K Seitz
- Centre of Liver and Alcohol Diseases, Ethianum Clinic, Heidelberg, Germany
- Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
11
|
Rubavathy SME, Hema G, Prakash M. Molecular mechanism behind the cholinium-taurate ionic liquid in stabilisation of HDAC2 for alcohol use disorders: insights from DFT and MD simulations. Phys Chem Chem Phys 2025; 27:6263-6277. [PMID: 40062365 DOI: 10.1039/d4cp04535a] [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: 03/20/2025]
Abstract
This study explores how an active pharmaceutical ingredient-ionic liquid (API-IL), cholinium taurate ([Cho]+[Tau]-) IL, may alter the structural and functional stability of histone deacetylase 2 (HDAC2), which is a crucial enzyme linked to alcohol use disorder (AUD). A particular hallmark of AUD, which is a worldwide health burden, is epigenetic dysregulation, in which HDAC2 plays a significant role in gene silencing and chronic neuroplastic alterations. Leveraging the unique physicochemical properties of [Cho]+[Tau]-IL, including hydrogen bond (H-bond) formation and structural reinforcement, we explored its therapeutic potential through comprehensive computational approaches. Density functional theory (DFT) analyses provided insights into the hydration and stability profiles of [Cho]+[Tau]-IL, while 200 ns molecular dynamics (MD) simulations elucidated its interaction with HDAC2 at the molecular level. Strikingly, the [Tau]- ion emerged as a key modulator of HDAC2 stability, facilitating conformational transitions in the enzyme's secondary structure, notably from turns to helices. This stabilisation was mediated by intricate hydration networks, water-mediated H-bonds, and diverse non-covalent interactions (NCIs). The rigorous nature of our structural analyses confirmed the potential of [Cho]+[Tau]-IL as a robust stabiliser of HDAC2, offering a novel therapeutic avenue for AUD treatment. This work underscores the promise of API-ILs in targeting epigenetic regulators and advancing strategies for AUD, providing a solid foundation for future research in this area.
Collapse
Affiliation(s)
- S M Esther Rubavathy
- Computational Chemistry Research Laboratory (CCRL), Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur-603 203, Chengalpattu, Tamil Nadu, India.
| | - Gopal Hema
- Computational Chemistry Research Laboratory (CCRL), Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur-603 203, Chengalpattu, Tamil Nadu, India.
| | - Muthuramalingam Prakash
- Computational Chemistry Research Laboratory (CCRL), Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur-603 203, Chengalpattu, Tamil Nadu, India.
| |
Collapse
|
12
|
Xia X, Cao X, Gong C, Liu Y, Zhang X, Liao L. Adherence to the Mediterranean diet is associated with lower cancer-related fatigue: a cross-sectional analysis from NHANES 2017-2020. Front Nutr 2025; 12:1506055. [PMID: 40177181 PMCID: PMC11961423 DOI: 10.3389/fnut.2025.1506055] [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: 10/04/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Background and objectives Cancer-related fatigue is a common and distressing symptom experienced by cancer patients, which may persist from the time of diagnosis to the end of life. This fatigue negatively affects patients' physical, emotional, and cognitive well-being. Nutrition plays a key role in managing cancer-related fatigue, and recently, the Mediterranean diet has gained attention as a potential intervention. The present study uses data from the National Health and Nutrition Examination Survey (NHANES) to investigate the association between cancer-related fatigue and the Mediterranean diet. Methods Data from the NHANES 2017-2020.03 cycle were selected for this cross-sectional study. The Alternative Mediterranean Diet Adherence (AMED) score was used to evaluate the participants' adherence to the Mediterranean diet. AMED scores were calculated based on data from 24-h dietary recall interviews conducted on both day one and day two. Multiple linear regression modeling was used to explore the association between AMED scores and cancer-related fatigue, as well as the relationship between AMED scores and fatigue in the general population. Results A total of 6,413 adults aged 20 years and older were included in the study, with 707 identified as cancer patients. There was a noteworthy inverse relationship found between AMED scores and fatigue, which was more pronounced in cancer patients: β = -0.121, 95% CI: -0.172, -0.071 (p < 0.001) in the unadjusted model. This correlation remained significant after adjusting for all variables in model 3: β = -0.074, 95% CI: -0.127, -0.021 (p = 0.007). A significant dose-dependent relationship was found when AMED scores were expressed in quartiles, with a more pronounced negative association as AMED increased across all models (p for trend <0.05). In the cancer population, the analysis of individual nutrients and fatigue revealed that alcohol was significantly negatively associated with cancer-related fatigue in all models, particularly in the unadjusted model: β = -0.710, 95% CI: -1.058, -0.362 (p < 0.001). Subgroup analyses indicated that diabetes, education level and type of cancer had a significant effect on the relationship between AMED and fatigue, with interaction p-values of 0.010, 0.023 and 0.049, respectively. Conclusion The present study suggests that higher adherence to the Mediterranean diet may contribute to reduce fatigue, especially in cancer patients; however, further research is necessary to validate this correlation.
Collapse
Affiliation(s)
- Xueqin Xia
- Department of Obstetrics and Gynecology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xuehua Cao
- Department of Obstetrics and Gynecology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Chen Gong
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yi Liu
- Department of Emergency Intensive Care Unit, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoyuan Zhang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Limei Liao
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
13
|
Adepu VK, Kumar HSS, Ravibabu K, Nagaraju R. Effect of Lead Exposure and Lifestyle Factors on Methylation Index Markers Among Pb-Exposed Workers. Biol Trace Elem Res 2025; 203:1342-1350. [PMID: 38935257 DOI: 10.1007/s12011-024-04270-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024]
Abstract
S-Adenosylmethionine (SAM) and S-adenosylhomocysteine (SAH) and the ratio of SAM and SAH in Pb-exposed workers need to be assessed. In this study, we investigated the effects of Pb exposure on SAM, SAH, and methylation index (MI) in Pb-exposed workers with contemplation of lifestyle factors. Blood lead levels (BLLs), SAM, SAH, MI, and lifestyle factors were assessed in 338 male Pb-exposed workers. BLLs are estimated by ICP-OES method. SAM and SAH levels in serum were determined by ELISA method. The MI was calculated using SAM and SAH individual values. The lifestyle factors were collected using standard questionnaire. Levels of SAM and MI were significantly decreased with increased age, experience > 5 years, habits of tobacco chewing, smoking, alcohol consumption, and BLLs 10-30, 30-50, and > 50 µg/dL. Levels of SAH were significantly increased with increased age, habits of tobacco chewing and smoking, and BLLs 10-30, 30-50, and > 50 µg/dL. The association between BLLs and methylation index markers (SAM and MI) was reported as negative and significant. The association between BLLs and SAH was noted positive and significant. The influence of BLLs and lifestyle factors on SAM was noted at 12%, SAH at 35%, and MI at 27%, respectively. The highest percentage of influence was noted in SAH, followed by MI and SAM. In the workers exposed to Pb, lifestyle factors resulted in decreased SAM and MI and increased SAH levels. Adaptation of healthy lifestyle factors, personal hygiene practices, and use of PPE were suggested to minimize the reduction of methylation index markers.
Collapse
Affiliation(s)
- Vinay Kumar Adepu
- Department of Biochemistry, Regional Occupational Health Centre (Southern), Indian Council of Medical Research, ICMR Complex, Kannamangala Post, Poojanahalli Road Devanahalli Taluk, Bengaluru, 562110, Karnataka, India
| | - H S Santosh Kumar
- Department of Biotechnology, Kuvempu University, Jnana Sahyadri, Shimoga, Karnataka, 577451, India
| | - Kalahasthi Ravibabu
- Department of Biochemistry, Regional Occupational Health Centre (Southern), Indian Council of Medical Research, ICMR Complex, Kannamangala Post, Poojanahalli Road Devanahalli Taluk, Bengaluru, 562110, Karnataka, India.
| | - Raju Nagaraju
- Department of Biochemistry, Regional Occupational Health Centre (Southern), Indian Council of Medical Research, ICMR Complex, Kannamangala Post, Poojanahalli Road Devanahalli Taluk, Bengaluru, 562110, Karnataka, India
| |
Collapse
|
14
|
Guo J, Xie H, Chen X. OXIDATIVE STRESS AND ANTIOXIDANT MARKERS IN ORAL SUBMUCOUS FIBROSIS: A SYSTEMATIC REVIEW AND META-ANALYSIS OF CASE-CONTROL STUDIES. J Evid Based Dent Pract 2025; 25:102073. [PMID: 39947776 DOI: 10.1016/j.jebdp.2024.102073] [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: 04/24/2024] [Revised: 11/11/2024] [Accepted: 11/23/2024] [Indexed: 05/09/2025]
Abstract
OBJECTIVE This meta-analysis systematically investigates the markers of oxidative stress (OS) and antioxidant systems in the saliva and serum of patients with oral submucous fibrosis (OSF) and healthy controls. METHODS Using a comprehensive search of databases from their inception period to December 31, 2023, articles related to OS or antioxidant markers in patients with OSF and healthy controls were retrieved. Literature screening and data extraction were performed by two reviewers according to the set criteria, and a third evaluator was involved in case of disagreements. The included studies were assessed with the Newcastle-Ottawa Scale, and the results were analyzed using Review Manager 5.4 software. RESULTS A total of 14 studies were included. Patients with OSF exhibited significantly higher levels of malondialdehyde (serum: SMD: 7.65, 95% confidence interval [CI]: 5.14-10.17, P < .00001, saliva: SMD: 3.07, 95% CI: 0.6-5.54, P < .00001) and 8-hydroxy-deoxyguanosine (saliva: SMD: 4.84, 95% CI: 3.49-6.18, P < .00001) than healthy controls. Levels of vitamin C (serum: SMD: -3.54, 95% CI: -4.10 to -2.99, P < .00001, saliva: SMD: -1.10, 95% CI: -1.39 to -0.80, P < .00001), vitamin E (saliva: SMD: -1.78, 95% CI: -2.1 to -1.45, P < .00001), and superoxide dismutase (serum: SMD: -9.77, 95% CI: -13.79 to -5.74, P < .00001) were significantly lower in patients with OSF than in healthy controls. CONCLUSIONS The results showed increased levels of OS markers and decreased levels of antioxidant markers in patients with OSF, providing valuable indicators that may aid in clinical diagnosis and management.
Collapse
Affiliation(s)
- Jincai Guo
- Department of Pharmacy, Changsha Stomatological Hospital, Changsha, China; School of Stomatology, Hunan University of Chinese Medicine, Changsha, China; School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Hui Xie
- Department of Pharmacy, Changsha Stomatological Hospital, Changsha, China; School of Stomatology, Hunan University of Chinese Medicine, Changsha, China
| | - Xueru Chen
- Department of Pharmacy, Changsha Stomatological Hospital, Changsha, China; School of Stomatology, Hunan University of Chinese Medicine, Changsha, China; School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China.
| |
Collapse
|
15
|
Wang L, Dong W, Fan L, Kong H, Liang S, Huang Z, Chen J, Zhi S, Xu S, Qiu Q, Yang M, Hou Y, Hu Y, Pan T, Zheng M, Li X, Huang Z, Song L. Repression of the ERRγ-CYP2E1 pathway by FGF4 mitigates alcohol-associated liver injury. Hepatology 2025:01515467-990000000-01184. [PMID: 40009617 DOI: 10.1097/hep.0000000000001282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/25/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND AND AIMS Alcohol-associated liver disease (ALD) represents a critical global health challenge characterized by liver damage resulting from excessive alcohol consumption. Early detection and timely intervention are essential for optimizing patient outcomes. However, the mechanisms underlying alcohol-induced liver injury have not been fully elucidated. Fibroblast growth factor 4 (FGF4) has been implicated in the progression of various liver diseases. This study aims to elucidate the role of FGF4 in the pathogenesis of ALD. APPROACH AND RESULTS We analyzed human liver specimens and observed significant upregulation of FGF4 mRNA and protein levels in patients with ALD. Consistent findings were noted in mouse models subjected to a Lieber-DeCarli liquid diet. Importantly, hepatic FGF4 expression exhibited a positive correlation with ALD severity in both human subjects and murine models. Hepatocyte-specific deletion of Fgf4 ( Fgf4 -LKO) exacerbated alcohol-induced liver injury through increased oxidative stress, inflammation, and apoptosis. Specifically, Fgf4 -LKO mice demonstrated heightened susceptibility to ethanol plus CCl 4 -induced fibrosis and liver injury. However, treatment with the ERRγ inverse agonist GSK5182 and CYP2E1 inhibitor chlormethiazole (CMZ) mitigated the exacerbated liver injury associated with Fgf4 deficiency. Mechanistic investigations revealed that FGFR4 phosphorylates ERRγ, promoting its ubiquitination and degradation in hepatocytes. Hepatic-specific knockout of Fgfr4 intensified alcohol-induced liver injury and nullified the protective conferred of recombinant FGF4 △NT . CONCLUSIONS Our study identifies FGF4 as a stress-responsive regulator in liver pathophysiology, operating through an FGFR4-mediated ERRγ-CYP2E1 signaling pathway. These results underscore the potential of FGF4 and its downstream pathways as therapeutic targets for ALD treatment.
Collapse
Affiliation(s)
- Luyao Wang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenliya Dong
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lei Fan
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hongru Kong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Siyu Liang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhuobing Huang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jie Chen
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Sisi Zhi
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Siyan Xu
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qiaoling Qiu
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Miaomiao Yang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yushu Hou
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yue Hu
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tongtong Pan
- Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Minghua Zheng
- Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaokun Li
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhifeng Huang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lintao Song
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
16
|
Mekonen H, Negesse A, Endalifer ML, Molla G, Aneley Z. Dietary factors associated with breast cancer among women in Ethiopia: a systematic review and meta-analysis of case-control studies. Front Nutr 2025; 12:1499634. [PMID: 40034736 PMCID: PMC11872731 DOI: 10.3389/fnut.2025.1499634] [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: 09/21/2024] [Accepted: 01/24/2025] [Indexed: 03/05/2025] Open
Abstract
Background Diet is the primary and largely modifiable factor associated with breast cancer risk. However, inconsistent findings were evidenced in many epidemiological studies and resulted in a lack of conclusiveness. Therefore, this systematic review and meta-analysis aimed to explore dietary risk factors that may predict breast cancer among Ethiopian women. Design data source and eligibility criteria A systematic review and meta-analysis was carried out. The articles were retrieved through electronic databases searching, including PubMed/Medline, Web of Science, Science Direct, EMBASE and Google Scholar. The Joanna Briggs Institute Meta-Analysis of Statistics Assessment and Review Instrument was applied for the critical appraisal. All case-control studies conducted in Ethiopia and reporting dietary factors of breast cancer in women were included in the final analysis. Data extraction Two independent reviewers extracted the data using a standardized data extraction format in Excel software. Stata version 17 software was used for the data analysis. Cochran's Q statistic with inverse variance (I2) was used to assess the presence of heterogeneity. A random effect model was used to estimate the odds ratio with a 95% confidence interval. Results Overall, eight eligible articles with 2,774 women were included to explore the dietary predictors of breast cancer in Ethiopia. As a result, alcohol consumption (OR: 1.26, 95% CI: 1.01, 1.57), packed food intake (OR: 6.83; 95% CI: 4.56, 10.24), saturated fat/oil intake (OR: 1.51; 95% CI: 1.13, 2.02), meat consumption (OR: 6.08, 95% CI: 3.62, 10.22), and vegetable consumption (OR: 0.75, 95% CI: 0.49, 0.89) were identified as significant predictors of breast cancer among women in Ethiopia. Conclusion The current study revealed a significant relationship between dietary factors and breast cancer. Avoiding the consumption of alcohol, saturated fats/oils, packed foods, and meat, coupled with promotion of vegetable consumption, could substantially contribute to reduce the burden of breast cancer among women in Ethiopia. Therefore, policymakers and other concerned bodies should provide routine community-based nutrition education to raise public awareness about the contribution of women's dietary practices on their breast cancer risk.
Collapse
Affiliation(s)
- Habitamu Mekonen
- Department of Human Nutrition, College of Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Ayenew Negesse
- Department of Human Nutrition, College of Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Melese Linger Endalifer
- Department of Human Nutrition, College of Health Science, Debre Markos University, Debre Markos, Ethiopia
| | - Gebeyaw Molla
- Department of Human Nutrition, Faculty of Chemical and Food Engineering, Bahir Dar Institute of Technology, Bahir Dar University, Bahir Dar, Ethiopia
| | - Zelalem Aneley
- Department of Human Nutrition, College of Health Science, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
17
|
Kojima S, Kuribayashi N, Goda H, Nakashiro KI, Uchida D. Oral cancer driver gene mutations in oral potentially malignant disorders: clinical significance and diagnostic implications. Discov Oncol 2025; 16:174. [PMID: 39946003 PMCID: PMC11825417 DOI: 10.1007/s12672-025-01923-7] [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: 10/15/2024] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
Oral potentially malignant disorders (OPMDs), such as oral lichen planus (OLP) and oral leukoplakia (OLK), are clinical conditions associated with the risk of developing oral squamous cell carcinoma; however, no standardized treatment guidelines exist. Mutations in specific genes, known as oral cancer driver gene mutations (ODGMs), are responsible for carcinogenesis. We aimed to analyze the ODGMs in OPMDs and investigate their clinical correlations. We investigated 41 cases of OPMDs, including OLP and OLK, and performed next-generation sequencing using a custom gene panel targeting whole exons of TP53, HRAS, PIK3CA, NOTCH1, CDKN2A, FBXW7, and BRAF. We detected ODGMs in four OLK cases and one OLP case. All ODGM-positive OLK cases were located in the tongue, a site associated with a higher risk of malignant transformation compared with those in other oral sites. Moreover, ODGMs were significantly associated with alcohol consumption. While there was a tendency for mutations to increase with higher grades, we found no significant correlation between the presence of ODGMs and oral epithelial dysplasia (OED) grade. Notably, in the case of OLP with ODGM, the lesion histopathologically developed OED during the follow-up period, indicating an increased risk of cancer development and that cases with identified ODGMs require early surgical excision. These findings suggest that ODGM analysis may predict the risk of cancer development in OPMDs that are difficult to diagnose using histopathological examination alone.
Collapse
Affiliation(s)
- Sayaka Kojima
- Department of Oral and Maxillofacial Surgery, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0295, Japan
| | - Nobuyuki Kuribayashi
- Department of Oral and Maxillofacial Surgery, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0295, Japan.
| | - Hiroyuki Goda
- Department of Oral and Maxillofacial Surgery, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0295, Japan
| | - Koh-Ichi Nakashiro
- Department of Oral and Maxillofacial Surgery, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0295, Japan
| | - Daisuke Uchida
- Department of Oral and Maxillofacial Surgery, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime, 791-0295, Japan
| |
Collapse
|
18
|
Xu S, Su Z, Jiang R, Wu X, Wang J, Wang Y, Cao X, Xia J, Shi H, Tan W. Electrochemical sensing system based on coordination bond connected porphyrin-MOFs@MXenes hybrids for in situ and real-time monitoring of H 2O 2 released from cells. Sci Rep 2025; 15:5235. [PMID: 39939370 PMCID: PMC11821885 DOI: 10.1038/s41598-025-89688-5] [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: 09/27/2024] [Accepted: 02/06/2025] [Indexed: 02/14/2025] Open
Abstract
Herein, the coordination bond connected porphyrin-MOFs/MXenes composites has been prepared to construct an electrochemical sensing system for in situ and real-time monitoring of H2O2 released by cells. The composites were synthesized by introducing 4-mercaptopyridine and utilizing its binding interaction with titanium in Mxenes and iron in MOFs. This composite was then transferred to the surface of ITO to construct an electrochemical sensing system. The unique properties of Mxenes and porphyrin MOFs endowed the sensing system with excellent electrocatalytic activity, good electrical conductivity and desirable biocompatibility. The electrochemical detections of hydroquinone verified the superior electrochemical performances of the sensing system. The constructed system achieved sensitive electrochemical detection of H2O2 with a detection limit of 3.1 µM and a linear range of 10 µM to 3 mM. Furthermore, the excellent biocompatibility of the composites ensured HeLa cells growth and proliferation on its surface. Based on these favorable properties, the sensing system successfully achieved in-situ and real-time monitoring of H2O2 released by HeLa cells.
Collapse
Affiliation(s)
- Shiquan Xu
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Zhaojie Su
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Rong Jiang
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Instrumental Analysis Center of Qingdao University, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Xia Wu
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Jie Wang
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Ying Wang
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Xiyue Cao
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Instrumental Analysis Center of Qingdao University, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Jianfei Xia
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Instrumental Analysis Center of Qingdao University, Qingdao University, Qingdao, 266071, People's Republic of China.
| | - He Shi
- Orthopedics and Sports Medicine Center, Qingdao Municipal Hospital, Qingdao, 266011, People's Republic of China.
| | - Weiqiang Tan
- Department of Organ Transplantation, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, 361102, People's Republic of China.
| |
Collapse
|
19
|
Zhang M, Ru M, Zhang J, Wang Z, Lu J, Butler KR, Chatterjee N, Couper DJ, Prizment AE, Soori MM, Visvanathan K, Zahnow CA, Joshu CE, Platz EA. Alcohol Consumption Does not Modify the Polygenic Risk Score-Based Genetic Risk of Breast Cancer in Postmenopausal Women: Atherosclerosis Risk in Communities Study. Cancer Prev Res (Phila) 2025; 18:73-83. [PMID: 39676351 PMCID: PMC11790378 DOI: 10.1158/1940-6207.capr-24-0208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/31/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
High genetic risk and alcohol consumption ≥1 drink/day are associated with increased breast cancer risk. However, the interaction between alcohol and genetics on breast cancer risk is poorly understood, including in populations not enriched with daily drinkers. We prospectively studied 5,651 White and Black postmenopausal women in the Atherosclerosis Risk in Communities study. Alcohol intake was assessed by a food frequency questionnaire. The 313-SNP polygenic risk score (PRS) was calculated. Breast cancer cases were ascertained primarily by cancer registry linkage through 2015. Multivariable Cox regression was used to estimate HRs and 95% confidence intervals (CI) for the association of PRS and current ethanol intake with breast cancer, and their interaction. Of these individuals, 50.6% were current drinkers, and of them, 50.8% drank <1 drink/week and 12.8% drank >7 drinks/week. A higher PRS was associated with a higher breast cancer risk among White (HR1-SD, 1.48; 95% CI, 1.34-1.65) and Black (HR1-SD, 1.15; 95% CI, 0.96-1.38) women. Positive associations were not observed between current ethanol intake and breast cancer risk (White: HR13 g/week, 1.00; 95% CI, 0.98-1.03; Black: HR, 0.83; 95% CI, 0.69-1.00). Among both White and Black women, PRS generally seemed to be positively associated with risk in drinkers and nondrinkers. There was no evidence of a PRS-ethanol intake interaction among White or Black women. Patterns in Black women were similar when using an 89-SNP PRS developed among African ancestry women. In conclusion, in a prospective analysis of White and Black postmenopausal women in a study population not enriched with daily drinkers, our findings suggest that alcohol drinking does not modify the PRS-based genetic risk of breast cancer. Prevention Relevance: Although our findings suggest that alcohol drinking does not modify the PRS-based genetic risk of breast cancer among White and Black women with lower alcohol intake, nevertheless, women should consider limiting alcohol consumption as a general cancer prevention strategy, as indicated in dietary guidelines.
Collapse
Affiliation(s)
- Minghui Zhang
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Meng Ru
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Jingning Zhang
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Ziqiao Wang
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Jiayun Lu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Kenneth R. Butler
- Department of Medicine: Division of Geriatrics, School of Medicine, University of Mississippi Medical Center, Jackson, MS
| | - Nilanjan Chatterjee
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - David J. Couper
- University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Anna E. Prizment
- Department of Laboratory Medicine & Pathology, University of Minnesota Medical School, and the University of Minnesota Masonic Cancer Center, Minneapolis, MN
| | - Mehrnoosh M. Soori
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Kala Visvanathan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Cynthia A. Zahnow
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Corinne E. Joshu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| |
Collapse
|
20
|
Baldari S, Antonini A, Di Rocco G, Toietta G. Expression pattern and prognostic significance of aldehyde dehydrogenase 2 in lung adenocarcinoma as a potential predictor of immunotherapy efficacy. CANCER INNOVATION 2025; 4:e149. [PMID: 39640071 PMCID: PMC11620833 DOI: 10.1002/cai2.149] [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: 01/03/2024] [Revised: 04/30/2024] [Accepted: 05/23/2024] [Indexed: 12/07/2024]
Abstract
Background The incidence of alcohol-associated cancers is higher within Asian populations having an increased prevalence of an inactivating mutation in aldehyde dehydrogenase 2 (ALDH2), a mitochondrial enzyme required for the clearance of acetaldehyde, a cytotoxic metabolite of ethanol. The role of alcohol consumption in promoting lung cancer is controversial, and little attention has been paid to the association between alcohol drinking and pulmonary ALDH2 expression. Methods We performed a comprehensive bioinformatic analysis of multi-omics data available in public databases to elucidate the role of ALDH2 in lung adenocarcinoma (LUAD). Results Transcriptional and proteomic data indicate a substantial pulmonary expression of ALDH2, which is functional for the metabolism of alcohol diffused from the bronchial circulation. ALDH2 expression is higher in healthy lung tissue than in LUAD and inhibits cell cycle, apoptosis, and epithelial-mesenchymal transition pathways. Moreover, low ALDH2 mRNA levels predict poor prognosis and low overall survival in LUAD patients. Interestingly, ALDH2 expression correlates with immune infiltration in LUAD. Conclusions A better understanding of the role of ALDH2 in lung tumor progression and immune infiltration might support its potential use as a prognostic marker and therapeutic target for improving immunotherapeutic response.
Collapse
Affiliation(s)
- Silvia Baldari
- Tumor Immunology and Immunotherapy UnitIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Annalisa Antonini
- Tumor Immunology and Immunotherapy UnitIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Giuliana Di Rocco
- Unit of Cellular Networks and Molecular Therapeutic TargetsIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Gabriele Toietta
- Tumor Immunology and Immunotherapy UnitIRCCS Regina Elena National Cancer InstituteRomeItaly
| |
Collapse
|
21
|
Lauri G, Mills K, Majumder S, Capurso G. The exposome as a target for primary prevention and a tool for early detection of pancreatic cancer. Best Pract Res Clin Gastroenterol 2025; 74:101991. [PMID: 40210335 DOI: 10.1016/j.bpg.2025.101991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/04/2025] [Accepted: 02/11/2025] [Indexed: 04/12/2025]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is a highly aggressive malignancy with limited survival due to late stage diagnosis and scarce therapeutic options. Emerging evidence highlights the role of the "exposome," which encompasses environmental, lifestyle, and metabolic exposures, as a crucial determinant in PDAC risk and a potential avenue for early detection. This review synthesizes findings on modifiable risk factors, including smoking, obesity, diabetes, diet, and alcohol consumption, and their interplay with genetic and metabolic profiles in PDAC development. Additionally, we explore cutting-edge approaches in exposomic research, such as biobanking, electronic health record analysis, and AI-driven predictive models, to identify early biomarkers and stratify high-risk populations. This integrated framework aims to inform prevention strategies and improve early detection of PDAC.
Collapse
Affiliation(s)
- Gaetano Lauri
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Krystal Mills
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Shounak Majumder
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Gabriele Capurso
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
22
|
Ding T, Liu C, Li Z. The mycobiome in human cancer: analytical challenges, molecular mechanisms, and therapeutic implications. Mol Cancer 2025; 24:18. [PMID: 39815314 PMCID: PMC11734361 DOI: 10.1186/s12943-025-02227-8] [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: 11/13/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025] Open
Abstract
The polymorphic microbiome is considered a new hallmark of cancer. Advances in High-Throughput Sequencing have fostered rapid developments in microbiome research. The interaction between cancer cells, immune cells, and microbiota is defined as the immuno-oncology microbiome (IOM) axis. Fungal microbes (the mycobiome), although representing only ∼ 0.1-1% of the microbiome, are a critical immunologically active component of the tumor microbiome. Accumulating evidence suggests a possible involvement of commensal and pathogenic fungi in cancer initiation, progression, and treatment responsiveness. The tumor-associated mycobiome mainly consists of the gut mycobiome, the oral mycobiome, and the intratumoral mycobiome. However, the role of fungi in cancer remains poorly understood, and the diversity and complexity of analytical methods make it challenging to access this field. This review aims to elucidate the causal and complicit roles of mycobiome in cancer development and progression while highlighting the issues that need to be addressed in executing such research. We systematically summarize the advantages and limitations of current fungal detection and analysis methods. We enumerate and integrate these recent findings into our current understanding of the tumor mycobiome, accompanied by the prospect of novel and exhilarating clinical implications.
Collapse
Affiliation(s)
- Ting Ding
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin South Road, Chengdu, Sichuan Province, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Chang Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin South Road, Chengdu, Sichuan Province, 610041, China
| | - Zhengyu Li
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin South Road, Chengdu, Sichuan Province, 610041, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China.
| |
Collapse
|
23
|
Papier K, Bradbury KE, Balkwill A, Barnes I, Smith-Byrne K, Gunter MJ, Berndt SI, Le Marchand L, Wu AH, Peters U, Beral V, Key TJ, Reeves GK. Diet-wide analyses for risk of colorectal cancer: prospective study of 12,251 incident cases among 542,778 women in the UK. Nat Commun 2025; 16:375. [PMID: 39779669 PMCID: PMC11711514 DOI: 10.1038/s41467-024-55219-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Uncertainty remains regarding the role of diet in colorectal cancer development. We examined associations of 97 dietary factors with colorectal cancer risk in 542,778 Million Women Study participants (12,251 incident cases over 16.6 years), and conducted a targeted genetic analysis in the ColoRectal Transdisciplinary Study, Colon Cancer Family Registry, and Genetics and Epidemiology of Colorectal Cancer Consortium (GECCO). Alcohol (relative risk per 20 g/day=1.15, 95% confidence interval 1.09-1.20) and calcium (per 300 mg/day=0.83, 0.77-0.89) intakes had the strongest associations, followed by six dairy-related factors associated with calcium. We showed a positive association with red and processed meat intake and weaker inverse associations with breakfast cereal, fruit, wholegrains, carbohydrates, fibre, total sugars, folate, and vitamin C. Genetically predicted milk consumption was inversely associated with risk of colorectal, colon, and rectal cancers. We conclude that dairy products help protect against colorectal cancer, and that this is driven largely or wholly by calcium.
Collapse
Affiliation(s)
- Keren Papier
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| | - Kathryn E Bradbury
- Department of Epidemiology and Biostatistics, School of Population Health, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Angela Balkwill
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Isobel Barnes
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Karl Smith-Byrne
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Marc J Gunter
- Cancer Epidemiology and Prevention Research Unit, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Anna H Wu
- University of Southern California, Department of Population and Public Health Sciences, Los Angeles, CA, USA
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Valerie Beral
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Timothy J Key
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Gillian K Reeves
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| |
Collapse
|
24
|
Thapa MJ, Chan K. The mutagenic properties of formaldehyde and acetaldehyde: Reflections on half a century of progress. Mutat Res 2025; 830:111886. [PMID: 39549522 DOI: 10.1016/j.mrfmmm.2024.111886] [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: 03/21/2024] [Revised: 10/20/2024] [Accepted: 10/28/2024] [Indexed: 11/18/2024]
Abstract
Formaldehyde and acetaldehyde are reactive, small compounds that humans are exposed to routinely, variously from endogenous and exogenous sources. Both small aldehydes are classified as human carcinogens. Investigation of the DNA damaging properties of these two compounds began some 50 years ago. In this review, we summarize progress in this field since its inception over half a century ago, distilling insights gained by the collective efforts of many research groups while highlighting areas for future directions. Over the decades, general consensus about aspects of the mutagenicity of formaldehyde and acetaldehyde has been reached. But other characteristics of formaldehyde and acetaldehyde remain incompletely understood and require additional investigation. These include crucial details about the mutational signature(s) induced and possible mechanistic role(s) during carcinogenesis.
Collapse
Affiliation(s)
- Mahanish Jung Thapa
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa Faculty of Medicine, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Kin Chan
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa Faculty of Medicine, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
25
|
Zhou J, Ge D, Liu Y, Chu Y, Zheng X, Lu Y, Chu Y. Developing Multiple Media Approach to Investigate Reproducible Characteristic VOCs of Lung Cancer Cells. Anal Chem 2024; 96:20398-20405. [PMID: 39692311 DOI: 10.1021/acs.analchem.4c03894] [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: 12/19/2024]
Abstract
Cellular volatile organic compound (VOC) detection is crucial for studying lung cancer biomarkers. However, the reported VOC biomarkers from the same cell line seem to be inconsistent across different research groups. It is possibly related to the variation in culture media, and the result obtained by a conventional single medium approach (SMA) depends on what medium is used in the cell experiment. This study proposes a multiple media approach (MMA) to investigate reproducible characteristic VOCs of lung cancer cells. Using solid-phase microextraction-gas chromatography-mass spectrometry (SPME-GC-MS) in combination with untargeted analysis, we conducted two independently repetitive experiments to compare lung cancer cells (A549) and normal lung cells (BEAS-2B) under three culture media conditions (RPMI 1640, DMEM, and Ham's F12). Both experiments indicated that, compared with 62-96 VOCs obtained by the SMA, only two VOCs (3-methyl-1-butanol and 2-methyl-1-butanol) were reproducibly achieved by the MMA. Moreover, their concentrations were significantly lower in lung cancer cells than in normal cells. Further targeted analysis confirmed the downregulation trend of both VOCs in subcutaneous and primary tumor tissues from the lung cancer model mouse. The present work demonstrated that the MMA cell experiment, just like the multicenter trials for cell lines, can facilitate the discovery of reproducible characteristic VOCs. This provides a cellular experimental basis and scientific evidence for lung cancer biomarker investigation and even breath biopsy technique development.
Collapse
Affiliation(s)
- Jijuan Zhou
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, P. R. China
- University of Science and Technology of China, Hefei 230026, P. R. China
| | - Dianlong Ge
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, P. R. China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, P. R. China
| | - Yue Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, P. R. China
- University of Science and Technology of China, Hefei 230026, P. R. China
| | - Yajing Chu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, P. R. China
- University of Science and Technology of China, Hefei 230026, P. R. China
| | - Xiangxue Zheng
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, P. R. China
- University of Science and Technology of China, Hefei 230026, P. R. China
| | - Yan Lu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, P. R. China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, P. R. China
| | - Yannan Chu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, P. R. China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, P. R. China
| |
Collapse
|
26
|
Yao J, Chen G. The global, regional, and national alcohol-related colorectal cancer burden and forecasted trends: results from the global burden of disease study 2021. Front Nutr 2024; 11:1520852. [PMID: 39777070 PMCID: PMC11704491 DOI: 10.3389/fnut.2024.1520852] [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: 10/31/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Background The mortality of colorectal cancer (CRC) is increasing year by year and poses a significant global health burden. Many studies have demonstrated that alcohol consumption is an important risk factor for CRC and is closely associated with malignant metastasis in CRC patients, which in turn leads to a poor prognosis. Methods This study aimed to quantify the global, regional, and national burden of alcohol-related CRC between 1990 and 2021. First, numbers and age-standardized rates of deaths and disability-adjusted life years (DALYs) for alcohol-related CRC in 2021 were analyzed at different levels. Temporal trends in the burden of disease from 1990 to 2021 were analyzed through linear regression models. Finally, both Age-Period-Cohort (APC) models and Bayesian Age-Period-Cohort (BAPC) models were utilized to project the future burden of the disease for 2022-2046. Results The global burden of disease for alcohol-related CRC is higher in 2021 compared to 1990. Male and older age groups are at high risk. Disease burden varies very much between Sociodemographic Index (SDI) regions, Global Burden of Disease (GBD) regions and countries. From 1990 to 2021, the number of cases increased, but the Age-Standardized Rate (ASR) decreased. The trends in disease burden predicted by the two models for 2022-2046 were not consistent. Conclusion This study describes the burden of disease in alcohol-related CRC and emphasizes that alcohol is a non-negligible risk factor for CRC. In order to mitigate harm, we need to strengthen disease surveillance, early prevention, timely detection, and improved treatment measures, with different approaches and responses for different regions.
Collapse
Affiliation(s)
| | - Guo Chen
- Department of Oncology, Shuguang Anhui Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (The First Affiliated Hospital West District of Anhui University of Chinese Medicine), Hefei, China
| |
Collapse
|
27
|
Fanfarillo F, Caronti B, Lucarelli M, Francati S, Tarani L, Ceccanti M, Piccioni MG, Verdone L, Caserta M, Venditti S, Ferraguti G, Fiore M. Alcohol Consumption and Breast and Ovarian Cancer Development: Molecular Pathways and Mechanisms. Curr Issues Mol Biol 2024; 46:14438-14452. [PMID: 39727994 DOI: 10.3390/cimb46120866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
Alcohol consumption has been consistently linked to an increased risk of several cancers, including breast and ovarian cancer. Despite substantial evidence supporting this association, the precise mechanisms underlying alcohol's contribution to cancer pathogenesis remain incompletely understood. This narrative review focuses on the key current literature on the biological pathways through which alcohol may influence the development of breast and ovarian cancer. Key mechanisms discussed include the modulation of estrogen levels, the generation of reactive oxygen species, the production of acetaldehyde, the promotion of chronic inflammation, and the induction of epigenetic changes. Alcohol's impact on estrogenic signaling, particularly in the regulation of estrogen and progesterone, is explored in the context of hormone-dependent cancers. Additionally, the role of alcohol-induced DNA damage, mutagenesis, and immune system modulation in tumor initiation and progression is examined. Overall, this review emphasizes the importance of alcohol as a modifiable risk factor for breast and ovarian cancer and highlights the need for further research to clarify its role in cancer biology.
Collapse
Affiliation(s)
- Francesca Fanfarillo
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Brunella Caronti
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Marco Lucarelli
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Silvia Francati
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Luigi Tarani
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Mauro Ceccanti
- SITAC, Società Italiana per il Trattamento dell'Alcolismo e le sue Complicanze, 00185 Rome, Italy
| | - Maria Grazia Piccioni
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Loredana Verdone
- Institute of Molecular Biology and Pathology (IBPM-CNR), 00161 Rome, Italy
| | - Micaela Caserta
- Institute of Molecular Biology and Pathology (IBPM-CNR), 00161 Rome, Italy
| | - Sabrina Venditti
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00161 Rome, Italy
| | - Giampiero Ferraguti
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Marco Fiore
- Institute of Biochemistry and Cell Biology (IBBC-CNR), Department of Sensory Organs, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
28
|
Todorovic N, Martinelli S, Nannini G, Weiskirchen R, Amedei A. Etiology-Dependent Microbiome Differences in Hepatocellular Carcinoma Development. Int J Mol Sci 2024; 25:13510. [PMID: 39769276 PMCID: PMC11677376 DOI: 10.3390/ijms252413510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
Chronic liver disease is characterised by persistent inflammation, tissue damage, and regeneration, which leads to steatosis, fibrosis, and, lastly, cirrhosis and hepatocellular carcinoma (HCC). HCC, the most prevalent form of primary liver cancer, is one of the leading causes of cancer-related mortality worldwide. The gut microbiota plays a fundamental role in human physiology, and disturbances in its critical balance are widely recognised as contributors to various pathological conditions, including chronic liver diseases, both infectious and non-infectious in nature. Growing interest in microbiota research has recently shifted the focus towards the study of intratumoural microbiota, referred to as the "oncobiome", which can significantly impact the development and progression of HCC. In this review, we discuss existing research and provide an overview of the microbiota influence on viral hepatitis, particularly in shaping the progression of liver disease caused by the hepatitis B and hepatitis C viruses. We also explore microbial dysbiosis and its contribution to the silent and dangerous progression of non-alcoholic fatty liver disease. Additionally, we address the impact of alcohol on the liver and its interaction with the microbiota, tracing the pathway from inflammation to cirrhosis and cancer. The review emphasises the most common etiologies of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Nevena Todorovic
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (N.T.); (S.M.); (G.N.)
- Clinic for Infectious and Tropical Diseases, University Clinical Centre of Serbia, 11000 Belgrade, Serbia
| | - Serena Martinelli
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (N.T.); (S.M.); (G.N.)
| | - Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (N.T.); (S.M.); (G.N.)
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (N.T.); (S.M.); (G.N.)
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 50139 Florence, Italy
| |
Collapse
|
29
|
Rodrigues C, Irving SC, Alves P, Dinis-Ribeiro M, Brandão C, Correia M. The Influence of Diet and Obesity in Lynch Syndrome: What Do We Know So Far. Nutrients 2024; 16:4352. [PMID: 39770972 PMCID: PMC11677193 DOI: 10.3390/nu16244352] [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: 11/03/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Of all new cases of colorectal cancer, Lynch syndrome (LS) accounts for approximately 3%. This syndrome is the most common hereditary cancer syndrome and is caused by pathogenic variants in the genes responsible for DNA mismatch repair. Although the relationship between colorectal cancer risk and diet is well established, little is known regarding the influence of diet and nutritional characteristics on LS's clinical evolution. There is some evidence suggesting that individuals living with LS should follow general guidelines for diet and alcohol restriction/moderation, so as to achieve and maintain a favorable weight status and overall health and quality of life. However, more research is needed, preferentially from clinical studies of a prospective nature with robust designs, to better inform diet and behavioral patterns targeting cancer prevention in LS.
Collapse
Affiliation(s)
- Cláudio Rodrigues
- Gastroenterology Department, Unidade Local de Saúde de Viseu Dão Lafões, 3504-509 Viseu, Portugal;
| | - Susana Couto Irving
- Nutrition–Medicine Department, Instituto Português de Oncologia, Francisco Gentil E.P.E., 4200-072 Porto, Portugal; (S.C.I.); (P.A.)
| | - Paula Alves
- Nutrition–Medicine Department, Instituto Português de Oncologia, Francisco Gentil E.P.E., 4200-072 Porto, Portugal; (S.C.I.); (P.A.)
| | - Mário Dinis-Ribeiro
- IRISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal;
- MEDCIDS-Department of Community Medicine, Information and Decision in Health, Faculty of Porto, University of Medicine, 4200-072 Porto, Portugal
- Department of Gastroenterology, Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal
| | - Catarina Brandão
- IRISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal;
- Department of Gastroenterology, Porto Comprehensive Cancer Center, 4200-072 Porto, Portugal
| | - Marta Correia
- Laboratório Associado, Escola Superior de Biotecnologia, Centro de Biotecnologia e Química Fina, Universidade Católica Portuguesa, CBQF, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal;
| |
Collapse
|
30
|
Zhang Z, Wu Z, Zeng Y, Li Y, Feng Y, Gao Z, Chen Y. Association of Methylenetetrahydrofolate Reductase Gene rs1801133 Polymorphism and Controlling Nutritional Status (CONUT) Score with Colorectal Cancer Susceptibility. Int J Gen Med 2024; 17:6281-6290. [PMID: 39712200 PMCID: PMC11662921 DOI: 10.2147/ijgm.s495139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/13/2024] [Indexed: 12/24/2024] Open
Abstract
Background Susceptibility to some cancers is linked to methylenetetrahydrofolate reductase (MTHFR) polymorphisms and the Controlling Nutritional Status (CONUT) score in some populations. However, their relationship with susceptibility to colorectal cancer (CRC) susceptibility in the Hakka Chinese population remains unclear. Methods In total, 620 CRC patients and 734 controls were enrolled. MTHFR rs1801133 was genotyped, medical records (age, sex, smoking history, alcohol consumption, hypertension, diabetes mellitus, and family history of cancer, and blood cell parameters) were collected, and the relationship between this information and CRC susceptibility was analyzed. Results There were significant differences in the distribution of CONUT classification (p=0.002), and proportions of history of smoking (p<0.001), hypertension (p<0.001), diabetes mellitus (p<0.001), and family history of cancer (p=0.002) between patients and controls. There were statistically significant differences in MTHFR rs1801133 genotypes distribution (58.7% C/C, 35.5% C/T, and 5.8% T/T in patients vs 65.5%, 31.2%, and 3.3% in controls, p=0.010) and allele distribution (76.5% C, and 23.5% T allele in patients vs 81.1%, and 18.9% in controls, p=0.003) between patients and controls. Logistic regression analysis indicated that non-normal CONUT range (non-normal vs normal, odds ratio (OR): 1.451, 95% confidence interval (CI): 1.119-1.882, p=0.005), and MTHFR rs1801133 variant (C/T + T/T vs C/C, OR: 1.373, 95% CI: 1.091-1.728, p=0.007), older age (≥65 vs <65 years, OR: 1.298, 95% CI: 1.023-1.646, p=0.032), male sex (OR: 1.354, 95% CI: 1.067-1.718, p=0.013), and history of alcohol drinking (OR: 2.232, 95% CI: 1.164-4.282, p=0.016) were independently associated with CRC risk. Conclusion Individuals carried MTHFR rs1801133 variant and with non-normal CONUT range, advanced age, history of alcohol consumption may be at increased CRC risk in the Hakka population.
Collapse
Affiliation(s)
- Zhuoxin Zhang
- Department of Gastrointestinal Surgery, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Zuguang Wu
- Department of Gastrointestinal Surgery, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Yuwen Zeng
- Department of Gastrointestinal Surgery, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Yunlin Li
- Department of Gastrointestinal Surgery, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Yingchuan Feng
- Department of Gastrointestinal Surgery, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Zhen Gao
- Department of Gastrointestinal Surgery, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Yijin Chen
- Department of Gastroenterology, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| |
Collapse
|
31
|
Locasale JW, Goncalves MD, Di Tano M, Burgos-Barragan G. Diet and Cancer Metabolism. Cold Spring Harb Perspect Med 2024; 14:a041549. [PMID: 38621831 PMCID: PMC11610756 DOI: 10.1101/cshperspect.a041549] [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: 04/17/2024]
Abstract
Diet and exercise are modifiable lifestyle factors known to have a major influence on metabolism. Clinical practice addresses diseases of altered metabolism such as diabetes or hypertension by altering these factors. Despite enormous public interest, there are limited defined diet and exercise regimens for cancer patients. Nevertheless, the molecular basis of cancer has converged over the past 15 years on an essential role for altered metabolism in cancer. However, our understanding of the molecular mechanisms that underlie the impact of diet and exercise on cancer metabolism is in its very early stages. In this work, we propose conceptual frameworks for understanding the consequences of diet and exercise on cancer cell metabolism and tumor biology and also highlight recent developments. By advancing our mechanistic understanding, we also discuss actionable ways that such interventions could eventually reach the mainstay of both medical oncology and cancer control and prevention.
Collapse
Affiliation(s)
- Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, Durham, Norh Carolina 27710, USA
| | - Marcus D Goncalves
- Division of Endocrinology, Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065, USA
| | - Maira Di Tano
- Division of Endocrinology, Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065, USA
| | - Guillermo Burgos-Barragan
- Department of Pharmacology, Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10056, USA
| |
Collapse
|
32
|
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
|
33
|
Ramachandran S. Oral cancer: Recent breakthroughs in pathology and therapeutic approaches. ORAL ONCOLOGY REPORTS 2024; 12:100678. [DOI: 10.1016/j.oor.2024.100678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
34
|
Huang HY, Li FR, Zhang YF, Lau HC, Hsueh CY, Zhou L, Zhang M. Metagenomic shotgun sequencing reveals the enrichment of Salmonella and Mycobacterium in larynx due to prolonged ethanol exposure. Comput Struct Biotechnol J 2024; 23:396-405. [PMID: 38235358 PMCID: PMC10792199 DOI: 10.1016/j.csbj.2023.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/04/2023] [Accepted: 12/18/2023] [Indexed: 01/19/2024] Open
Abstract
The exposure of ethanol increases the risk of head and neck inflammation and tumor progression. However, limited studies have investigated the composition and functionality of laryngeal microbiota under ethanol exposure. We established an ethanol-exposed mouse model to investigate the changes in composition and function of laryngeal microbiota using Metagenomic shotgun sequencing. In the middle and late stages of the experiment, the laryngeal microbiota of mice exposed to ethanol exhibited obvious distinguished from that of the control group on principal-coordinate analysis (PCoA) plots. Among the highly abundant species, Salmonella enterica and Mycobacterium marinum were likely to be most impacted. Our findings indicated that the exposure to ethanol significantly increased their abundance in larynxes in mice of the same age, which has been confirmed through FISH experiments. Among the species-related functions and genes, metabolism is most severely affected by ethanol. The difference was most obvious in the second month of the experiment, which may be alleviated later because the animal established tolerance. Notable enrichments concerning energy, amino acid, and carbohydrate metabolic pathways occurred during the second month under ethanol exposure. Finally, based on the correlation between species and functional variations, a network was established to investigate relationships among microbiota, functional pathways, and related genes affected by ethanol. Our data first demonstrated the continuous changes of abundance, function and their interrelationship of laryngeal microbiota under ethanol exposure by Metagenomic shotgun sequencing. Importance Ethanol may participate in the inflammation and tumor progression by affecting the composition of the laryngeal microbiota. Here, we applied the metagenomic shotgun sequencing instead of 16 S rRNA sequencing method to identify the laryngeal microbiota under ethanol exposure. Salmonella enterica and Mycobacterium marinum are two dominant species that may play a role in the reconstruction of the laryngeal microenvironment, as their local abundance increases following exposure to ethanol. The metabolic function is most evidently impacted, and several potential metabolic pathways could be associated with alterations in microbiota composition. These findings could help us better understand the impact of prolonged ethanol exposure on the microbial composition and functionality in the larynx.
Collapse
Affiliation(s)
| | | | | | - Hui-Ching Lau
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Chi-Yao Hsueh
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Liang Zhou
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Ming Zhang
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| |
Collapse
|
35
|
Chen HY, Por CR, Hong YK, Kong EQZ, Subramaniyan V. Molecular mechanisms underlying oesophageal cancer development triggered by chronic alcohol consumption. CLINICAL AND TRANSLATIONAL DISCOVERY 2024; 4. [DOI: 10.1002/ctd2.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/24/2024] [Indexed: 12/27/2024]
Abstract
AbstractThis review explores the mechanisms underlying alcohol‐induced oesophageal carcinogenesis, including DNA damage, oxidative stress, and nutritional deficiencies. Alcohol metabolism primarily involves alcohol dehydrogenase (ADH) converting ethanol to acetaldehyde, which can cause DNA damage, inhibit repair mechanisms, and form DNA adducts thus inhibiting DNA replication. Plus, it delves into the epidemiological evidence, genetic susceptibility, epigenetic modifications, biomarkers, and preventive strategies associated with alcohol‐related oesophageal cancers. Consumption of alcohol increases the risk of gastroesophageal reflux disease thus compromising mucosal integrity of the oesophagus as dysregulation of cytokines such as IL‐18, TNFA, GATA3, TLR4, and CD68 expands the intercellular spaces of epithelial cells. Genetic variants, such as ADH1B rs1229984 and ALDH2 rs671, significantly influence susceptibility to alcohol‐related oesophageal cancers, with these variations affecting acetaldehyde metabolism and cancer risk. Understanding these factors is crucial for early detection, effective treatment, and the development of targeted prevention strategies. Biomarkers, such as miRNA and metabolite markers, offer non‐invasive methods for early detection, while advanced endoscopic techniques provide better diagnostic accuracy. Pharmacological interventions, such as statins and proton pump inhibitors, also show potential for reducing cancer progression in high‐risk individuals. Despite advances, late‐stage oesophageal cancer diagnoses are still common, highlighting the need for better screening and prevention. Further research, including this study, should aim to improve early detection, personalise prevention, and explore new treatments to reduce cases and enhance outcomes in alcohol‐related oesophageal cancers.
Collapse
Affiliation(s)
- Huai Yi Chen
- Jeffrey Cheah School of Medicine and Health Sciences Monash University Jalan Lagoon Selatan Bandar Sunway Subang Jaya Malaysia
| | - Chia Rou Por
- Jeffrey Cheah School of Medicine and Health Sciences Monash University Jalan Lagoon Selatan Bandar Sunway Subang Jaya Malaysia
| | - Yong Kai Hong
- Jeffrey Cheah School of Medicine and Health Sciences Monash University Jalan Lagoon Selatan Bandar Sunway Subang Jaya Malaysia
| | - Eason Qi Zheng Kong
- Jeffrey Cheah School of Medicine and Health Sciences Monash University Jalan Lagoon Selatan Bandar Sunway Subang Jaya Malaysia
| | - Vetriselvan Subramaniyan
- Jeffrey Cheah School of Medicine and Health Sciences Monash University Jalan Lagoon Selatan Bandar Sunway Subang Jaya Malaysia
- School of Medical and Life Sciences Sunway University Jalan Lagoon Selatan Bandar Sunway Petaling Jaya Malaysia
| |
Collapse
|
36
|
Sun YQ, Wu Y, Li MR, Wei YY, Guo M, Zhang ZL. Elafibranor alleviates alcohol-related liver fibrosis by restoring intestinal barrier function. World J Gastroenterol 2024; 30:4660-4668. [PMID: 39575408 PMCID: PMC11572637 DOI: 10.3748/wjg.v30.i43.4660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/29/2024] [Accepted: 10/18/2024] [Indexed: 10/31/2024] Open
Abstract
We discuss the article by Koizumi et al published in the World Journal of Gastroenterology. Our focus is on the therapeutic targets for fibrosis associated with alcohol-related liver disease (ALD) and the mechanism of action of elafibranor (EFN), a dual agonist of peroxisome proliferator-activated receptor α (PPARα) and peroxisome PPAR δ (PPARδ). EFN is currently in phase III clinical trials for the treatment of metabolic dysfunction-associated fatty liver disease and primary biliary cholangitis. ALD progresses from alcoholic fatty liver to alcoholic steatohepatitis (ASH), with chronic ASH eventually leading to fibrosis, cirrhosis, and, in some cases, hepatocellular carcinoma. The pathogenesis of ALD is driven by hepatic steatosis, oxidative stress, and acetaldehyde toxicity. Alcohol consumption disrupts lipid metabolism by inactivating PPARα, exacerbating the progression of ALD. EFN primarily activates PPARα, promoting lipolysis and β-oxidation in ethanol-stimulated HepG2 cells, which significantly reduces hepatic steatosis, apoptosis, and fibrosis in an ALD mouse model. Additionally, alcohol disrupts the gut-liver axis at several interconnected levels, contributing to a proinflammatory environment in the liver. EFN helps alleviate intestinal hyperpermeability by restoring tight junction protein expression and autophagy, inhibiting apoptosis and inflammatory responses, and enhancing intestinal barrier function through PPARδ activation.
Collapse
Affiliation(s)
- Yu-Qi Sun
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Yang Wu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Meng-Ran Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Yu-Yao Wei
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Mei Guo
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Zi-Li Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| |
Collapse
|
37
|
Bouajila N, Domenighetti C, Aubin HJ, Naassila M. Alcohol consumption and its association with cancer, cardiovascular, liver and brain diseases: a systematic review of Mendelian randomization studies. FRONTIERS IN EPIDEMIOLOGY 2024; 4:1385064. [PMID: 39574800 PMCID: PMC11578756 DOI: 10.3389/fepid.2024.1385064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 10/14/2024] [Indexed: 11/24/2024]
Abstract
Background The health effects of alcohol consumption, particularly regarding potential protective benefits of light to moderate intake compared to abstinence, remain a subject of ongoing debate. However, epidemiological studies face limitations due to imprecise exposure measurements and the potential for bias through residual confounding and reverse causation. To address these limitations, we conducted a systematic review of Mendelian Randomization (MR) studies examining the causal relationship between alcohol consumption and cancers, cardiovascular, liver, and neurological diseases. Methodology We searched PubMed, ScienceDirect and Embase and Europe PMC up to 05/2024 for MR studies investigating the association of genetically predicted alcohol consumption with cancers, cardiovascular, liver and neurological diseases. We assessed methodological quality based on key elements of the MR design a genetic association studies tool. Results We included 70 MR studies that matched our inclusion criteria. Our review showed a significant association of alcohol consumption with multiple cancers such as oral and oropharyngeal, esophageal, colorectal cancers, hepatocellular carcinoma and cutaneous melanoma. While the available studies did not consistently confirm the adverse or protective effects of alcohol on other cancers, such as lung cancer, as suggested by observational studies. Additionally, MR studies confirmed a likely causal effect of alcohol on the risk of hypertension, atrial fibrillation, myocardial infraction and vessels disease. However, there was no evidence to support the protective effects of light to moderate alcohol consumption on cognitive function, Alzheimer's disease, and amyotrophic lateral sclerosis, as reported in observational studies while our review revealed an increased risk of epilepsy and multiple sclerosis. The available studies provided limited results on the link between alcohol consumption and liver disease. Conclusions Despite the valuable insights into the causal relationship between alcohol consumption and various health outcomes that MR studies provided, it is worth noting that the inconsistent ability of genetic instrumental variables to distinguish between abstainers, light and moderate drinkers makes it difficult to differentiate between U or J-shaped vs. linear relationships between exposure and outcome. Additional research is necessary to establish formal quality assessment tools for MR studies and to conduct more studies in diverse populations, including non-European ancestries. Systematic Review Registration www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021246154, Identifier: PROSPERO (CRD42021246154).
Collapse
Affiliation(s)
- Naouras Bouajila
- Inserm Unit UMRS 1247, University of Picardie Jules Verne, Amiens, France
| | - Cloé Domenighetti
- UVSQ, Univ. Paris-Sud, Inserm, Team “Exposome, Heredity, Cancer, and Health”, CESP, University of Paris-Saclay, Villejuif, France
| | - Henri-Jean Aubin
- Department of Psychiatry and Addictology, Paul-Brousse Hospital, AP-HP, Center for Epidemiology and Population Health Research (CESP), Inserm 1018, University of Paris-Saclay, Villejuif, France
| | - Mickael Naassila
- Inserm Unit UMRS 1247, University of Picardie Jules Verne, Amiens, France
| |
Collapse
|
38
|
Ha NB, Yao F. Alcohol and Hepatocellular Carcinoma. Clin Liver Dis 2024; 28:633-646. [PMID: 39362712 PMCID: PMC12037205 DOI: 10.1016/j.cld.2024.06.007] [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] [Indexed: 10/05/2024]
Abstract
Alcohol-associated liver disease (ALD) poses a significant risk for hepatocellular carcinoma (HCC), comprising various liver conditions from steatosis to cirrhosis. Despite accounting for a third of global HCC cases and deaths, ALD-related HCC lacks characterization compared to viral hepatitis-related HCC. Proposed mechanisms for ALD-related HCC include acetaldehyde toxicity, increased reactive oxygen species, and inflammation. This review examines ALD-associated HCC epidemiology, co-factors like viral hepatitis and metabolic syndrome, surveillance, and treatment challenges. Despite advances in screening and management, ALD-related HCC often presents at advanced stages, limiting treatment options and survival.
Collapse
Affiliation(s)
- Nghiem B Ha
- Hepatology, Liver Transplant, Division of Gastroenterology and Hepatology, Department of Medicine, University of California, San Francisco, 505 Parnassus Avenue, S-357, San Francisco, CA 94112, USA
| | - Francis Yao
- Hepatology, Liver Transplant, Division of Gastroenterology and Hepatology, Department of Medicine, University of California, San Francisco, 505 Parnassus Avenue, S-357, San Francisco, CA 94112, USA.
| |
Collapse
|
39
|
Matković K, Gerić M, Kazensky L, Milić M, Kašuba V, Cvitković A, Sanković M, Šumanovac A, Møller P, Gajski G. Comparison of DNA damage in fresh and frozen blood samples: implications for the comet assay in human biomonitoring studies. Arch Toxicol 2024; 98:3467-3476. [PMID: 39004639 DOI: 10.1007/s00204-024-03823-1] [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: 05/15/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024]
Abstract
The use of the comet assay in large biomonitoring studies may present logistical and technical challenges because of the processing of numerous samples. Proper sample preservation becomes imperative to prevent spurious DNA breakage. Previous research has shown the feasibility of conducting the comet assay on frozen blood samples, highlighting the potential of freezing at - 80 °C in preserving DNA integrity. Nonetheless, this approach presents challenges, including potential DNA damage during freezing and thawing, variability in processing, and the need for standardized protocols. Our objective was to evaluate whether there are comparable results in DNA migration assessed by the comet assay between fresh and frozen blood samples on a larger scale (N = 373). In our findings, elevated DNA migration was evident in frozen samples relative to fresh ones. Additionally, smoking, alcohol consumption, and season were linked to increased DNA damage levels in whole blood cells. Based on our results and available literature, conducting the comet assay on frozen blood samples emerges as a practical and efficient approach for biomonitoring and epidemiological research. This method enables the assessment of DNA damage in large populations over time, with samples, if properly cryopreserved, that may be used for years, possibly even decades. These observations hold significant implications for large-scale human biomonitoring and long-term epidemiological studies, particularly when samples are collected during fieldwork or obtained from biobanks. Continued method optimization and validation efforts are essential to enhance the utility of this approach in environmental and occupational health studies, emphasizing caution when comparing data obtained between fresh and frozen blood samples.
Collapse
Affiliation(s)
- Katarina Matković
- Division of Toxicology, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Marko Gerić
- Division of Toxicology, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Luka Kazensky
- Division of Toxicology, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Mirta Milić
- Division of Toxicology, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Vilena Kašuba
- Division of Toxicology, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Ante Cvitković
- Teaching Institute of Public Health Brod-Posavina County, Slavonski Brod, Croatia
- Faculty of Dental Medicine and Health, J. J. Strossmayer University of Osijek, Osijek, Croatia
- Faculty of Medicine, J. J. Strossmayer University of Osijek, Osijek, Croatia
| | - Mandica Sanković
- City of Vinkovci, Department of Physical Planning, Construction and Environmental Protection, Vinkovci, Croatia
| | - Antun Šumanovac
- Faculty of Medicine, J. J. Strossmayer University of Osijek, Osijek, Croatia
- County General Hospital Vinkovci, Vinkovci, Croatia
| | - Peter Møller
- Department of Public Health, Section of Environmental Health, University of Copenhagen, Copenhagen, Denmark
| | - Goran Gajski
- Division of Toxicology, Institute for Medical Research and Occupational Health, Zagreb, Croatia.
| |
Collapse
|
40
|
Tin Tin S, Smith-Byrne K, Ferrari P, Rinaldi S, McCullough ML, Teras LR, Manjer J, Giles G, Le Marchand L, Haiman CA, Wilkens LR, Chen Y, Hankinson S, Tworoger S, Eliassen AH, Willett WC, Ziegler RG, Fuhrman BJ, Sieri S, Agnoli C, Cauley J, Menon U, Fourkala EO, Rohan TE, Kaaks R, Reeves GK, Key TJ. Alcohol intake and endogenous sex hormones in women: Meta-analysis of cohort studies and Mendelian randomization. Cancer 2024; 130:3375-3386. [PMID: 38824654 DOI: 10.1002/cncr.35391] [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: 10/06/2023] [Revised: 03/12/2024] [Accepted: 04/12/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND The mechanisms underlying alcohol-induced breast carcinogenesis are not fully understood but may involve hormonal changes. METHODS Cross-sectional associations were investigated between self-reported alcohol intake and serum or plasma concentrations of estradiol, estrone, progesterone (in premenopausal women only), testosterone, androstenedione, dehydroepiandrosterone sulfate, and sex hormone binding globulin (SHBG) in 45 431 premenopausal and 173 476 postmenopausal women. Multivariable linear regression was performed separately for UK Biobank, European Prospective Investigation into Cancer and Nutrition, and Endogenous Hormones and Breast Cancer Collaborative Group, and meta-analyzed the results. For testosterone and SHBG, we also conducted Mendelian randomization and colocalization using the ADH1B (alcohol dehydrogenase 1B) variant (rs1229984). RESULTS Alcohol intake was positively, though weakly, associated with all hormones (except progesterone in premenopausal women), with increments in concentrations per 10 g/day increment in alcohol intake ranging from 1.7% for luteal estradiol to 6.6% for postmenopausal dehydroepiandrosterone sulfate. There was an inverse association of alcohol with SHBG in postmenopausal women but a small positive association in premenopausal women. Two-sample randomization identified positive associations of alcohol intake with total testosterone (difference per 10 g/day increment: 4.1%; 95% CI, 0.6-7.6) and free testosterone (7.8%; 4.1-11.5), and an inverse association with SHBG (-8.1%; -11.3% to -4.9%). Colocalization suggested a shared causal locus at ADH1B between alcohol intake and higher free testosterone and lower SHBG (posterior probability for H4, 0.81 and 0.97, respectively). CONCLUSIONS Alcohol intake was associated with small increases in sex hormone concentrations, including bioavailable fractions, which may contribute to its effect on breast cancer risk.
Collapse
Affiliation(s)
- Sandar Tin Tin
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Karl Smith-Byrne
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Pietro Ferrari
- International Agency for Research on Cancer (IARC/WHO), Nutrition and Metabolism Branch, Lyon, France
| | - Sabina Rinaldi
- International Agency for Research on Cancer (IARC/WHO), Nutrition and Metabolism Branch, Lyon, France
| | | | - Lauren R Teras
- Department of Population Science, American Cancer Society, Atlanta, Georgia, USA
| | - Jonas Manjer
- Department of Surgery, Skåne University Hospital Malmö, Lund University, Malmö, Sweden
| | - Graham Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
| | - Loïc Le Marchand
- University of Hawai'i Cancer Center, University of Hawai'i, Honolulu, Hawaii, USA
| | - Christopher A Haiman
- USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Lynne R Wilkens
- University of Hawai'i Cancer Center, University of Hawai'i, Honolulu, Hawaii, USA
| | - Yu Chen
- Division of Epidemiology, Department of Population Health, New York University Grossman School of Medicine, New York, New York, USA
| | - Sue Hankinson
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Shelley Tworoger
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - A Heather Eliassen
- Departments of Nutrition & Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Walter C Willett
- Departments of Nutrition & Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Regina G Ziegler
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | | | - Sabina Sieri
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Claudia Agnoli
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Jane Cauley
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Usha Menon
- MRC Clinical Trials Unit, Institute of Clinical Trials and Methodology, University College London, London, UK
| | | | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Gillian K Reeves
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Timothy J Key
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| |
Collapse
|
41
|
Afifah NN, Permatasari LI, Diantini A, Intania R, Wijaya I, Obinata H, Barliana MI. Exploring Genetic Variants and Platinum Chemotherapy Response in Indonesian Non-Small Cell Lung Cancer Patients: Insights from ERCC2 rs13181. Onco Targets Ther 2024; 17:767-776. [PMID: 39319218 PMCID: PMC11421434 DOI: 10.2147/ott.s475219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 09/08/2024] [Indexed: 09/26/2024] Open
Abstract
Purpose Individual responses to platinum-based treatment for Non-Small Cell Lung Cancer (NSCLC) are influenced by genetic polymorphisms, including Single Nucleotide Polymorphisms (SNPs). This study aimed to explore the role of ERCC2 in the Nucleotide Excision Repair (NER) pathway for platinum-based chemotherapy in NSCLC. While ERCC2 is widely studied, data for Southeast Asian populations are lacking. Addressing this gap could improve personalized treatment strategies for NSCLC in this demographic. Patients and Methods This study recruited 82 NSCLC patients with wildtype mutations of EGFR at Dr. H.A. Rotinsulu Lung Hospital, Bandung, and Dharmais Cancer Hospital, Jakarta. Data were collected prospectively from whole blood samples and medical records, while the effectiveness of chemotherapy was assessed by evaluating the response using RECIST 1.1 criteria on fourth cycle of chemotherapy. Results The results of this study showed the presence of genotype variation among the subjects, with frequency distribution as follows: AA genotype (82.9%), AC genotype (15.9%), and CC genotype (1.2%). The analysis of the association between ERCC2 rs13181 CC + AC versus AA with RECIST 1.1 yielded an odds ratio (OR) of 1.042 (95% CI: 0.292-3.715; p=0.950). A multivariate analysis that included cancer stage and chemotherapy regimen as additional variables produced an adjusted odds ratio (aOR) of 0.970 (95% CI: 0.263-3.568; p=0.963). Conclusion This study did not find statistically significant associations between ERCC2 rs13181 polymorphisms and chemotherapy responses. However, this research highlights the presence of genetic variation within the Indonesian population, with the AA genotype being the most prevalent, which may influence chemotherapy responses. The results provided preliminary data and lay the foundation for future comprehensive cohort observational investigations.
Collapse
Affiliation(s)
- Nadiya Nurul Afifah
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
- Department of Pharmacy, Faculty of Health Sciences, Universitas Esa Unggul, Jakarta, Indonesia
| | - Lanny Indah Permatasari
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
| | - Ajeng Diantini
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
| | - Ruri Intania
- Division of Pulmonology and Respiratory, Dr.H.A Rotinsulu, Lung Hospital, Bandung, Indonesia
| | - Indra Wijaya
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Hasan Sadikin General Hospital, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Hideru Obinata
- Education and Research Support Center, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Melisa Intan Barliana
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Jatinangor, Indonesia
| |
Collapse
|
42
|
Jee Y, Ryu M, Sull JW. Alcohol Consumption and Cancer Risk: Two Sample Mendelian Randomization. EPIDEMIOLOGIA 2024; 5:618-626. [PMID: 39311360 PMCID: PMC11417818 DOI: 10.3390/epidemiologia5030043] [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: 07/29/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 09/26/2024] Open
Abstract
Although numerous observational studies have reported on the association between alcohol consumption and cancer, insufficient studies have estimated the causality. Our study evaluated the causal relationship between various types of cancer according to the frequency of drinking and the amount of alcohol consumed. The research data were obtained from the publicly available MR-Base platform. The frequency and amount of drinking were selected as the exposure, and 16 cancer types were selected as the outcome. Two-sample summary data Mendelian randomization (2SMR) was conducted to examine the causality between alcohol consumption and cancer type. Additionally, for cancers suspected of pleiotropy, outliers were removed and re-analyzed through radial MR. The MR results using the inverse variance weighted (IVW) method were different before and after removing outliers. The biggest differences were found for esophageal cancer and biliary tract cancer. For esophageal cancer, after removing outliers (rs13102973, rs540606, rs650558), the OR (95% CI) was 3.44 (1.19-9.89), which was statistically significant (p = 0.02172). Even in biliary tract cancer, after removing outliers (rs13231886, rs58905411), the OR (95% CI) was 3.86 (0.89-16.859), which was of borderline statistical significance (p = 0.07223). The strongest association was found for esophageal cancer. For other cancers, the evidence was not sufficient to draw conclusions. More research is needed to understand the causality between drinking and cancer.
Collapse
Affiliation(s)
- Yongho Jee
- Advanced Biomedical Research Institute, Ewha Womans University Seoul Hospital, Seoul 07804, Republic of Korea;
| | - Mikyung Ryu
- Institute of Genetic Epidemiology, Basgenbio Inc., Seoul 04167, Republic of Korea
| | - Jae-Woong Sull
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Seongnam 13135, Republic of Korea;
| |
Collapse
|
43
|
Asonuma S, Hatta W, Koike T, Okata H, Uno K, Iwai W, Saito M, Yonechi M, Fukushi D, Kayaba S, Kikuchi R, Ito H, Fushiya J, Maejima R, Abe Y, Kawamura M, Honda J, Kondo Y, Dairaku N, Toda S, Watanabe K, Takahashi K, Echigo H, Abe Y, Endo H, Okata T, Hoshi T, Kinoshita K, Kisoi M, Nakamura T, Nakaya N, Iijima K, Masamune A. Risk stratification of synchronous gastric cancers including alcohol-related genetic polymorphisms. J Gastroenterol Hepatol 2024; 39:1554-1562. [PMID: 38628101 DOI: 10.1111/jgh.16570] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/30/2024] [Accepted: 03/26/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND AND AIM We previously identified that ever-smoking and severe gastric atrophy in pepsinogen are risk factors for synchronous gastric cancers (SGCs). This study aimed to determine the association of alcohol drinking status or alcohol-related genetic polymorphism with SGCs and also stratify their risk. METHODS This multi-center prospective cohort study included patients who underwent endoscopic submucosal dissection for the initial early gastric cancers at 22 institutions in Japan. We evaluated the association of alcohol drinking status or alcohol dehydrogenase 1B (ADH1B) and acetaldehyde dehydrogenase 2 (ALDH2) genotypes with SGCs. We then stratified the risk of SGCs by combining prespecified two factors and risk factors identified in this study. RESULTS Among 802 patients, 130 had SGCs. Both the ADH1B Arg and ALDH2 Lys alleles demonstrated a significant association with SGCs on multivariate analysis (odds ratio, 1.77), although alcohol drinking status showed no association. The rates of SGCs in 0-3 risk factors in the combined evaluation of three risk factors (ever-smoking, severe gastric atrophy in pepsinogen, and both the ADH1B Arg and ALDH2 Lys alleles) were 7.6%, 15.0%, 22.0%, and 32.1%, respectively. The risk significantly increased from 0 to 3 risk factors on multivariate analysis (P for trend <0.001). CONCLUSIONS Both the ADH1B Arg and ALDH2 Lys alleles were at high risk for SGCs. The risk stratification by these three factors may be a less invasive and promising tool for predicting their risk.
Collapse
Affiliation(s)
- Sho Asonuma
- Department of Gastroenterology, South Miyagi Medical Center, Ogawara-machi, Japan
| | - Waku Hatta
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomoyuki Koike
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hideki Okata
- Department of Gastroenterology, South Miyagi Medical Center, Ogawara-machi, Japan
| | - Kaname Uno
- Department of Gastroenterology, South Miyagi Medical Center, Ogawara-machi, Japan
| | - Wataru Iwai
- Department of Gastroenterology, Miyagi Cancer Center, Natori, Japan
| | - Masashi Saito
- Department of Gastroenterology, Miyagi Cancer Center, Natori, Japan
| | - Makoto Yonechi
- Division of Gastroenterology, Tohoku Medical and Pharmaceutical University School of Medicine, Sendai, Japan
| | - Daisuke Fukushi
- Division of Gastroenterology, Tohoku Medical and Pharmaceutical University School of Medicine, Sendai, Japan
| | - Shoichi Kayaba
- Department of Gastroenterology, Iwate Prefectural Isawa Hospital, Ohshu, Japan
| | - Ryosuke Kikuchi
- Department of Gastroenterology, JR Sendai Hospital, Sendai, Japan
| | - Hirotaka Ito
- Department of Gastroenterology, Osaki Citizen Hospital, Osaki, Japan
| | - Jun Fushiya
- Department of Gastroenterology, Iwate Prefectural Central Hospital, Morioka, Japan
| | - Ryuhei Maejima
- Department of Gastroenterology, Red Cross Ishinomaki Hospital, Ishinomaki, Japan
| | - Yasuhiko Abe
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Masashi Kawamura
- Department of Gastroenterology, Sendai City Hospital, Sendai, Japan
| | - Junya Honda
- Department of Gastroenterology, Iwate Prefectural Iwai Hospital, Ichinoseki, Japan
| | - Yutaka Kondo
- Department of Gastroenterology, Tohoku Rosai Hospital, Sendai, Japan
| | - Naohiro Dairaku
- Department of Gastroenterology, Japanese Red Cross Sendai Hospital, Sendai, Japan
| | - Shusuke Toda
- Department of Gastroenterology, Obihiro Daiichi Hospital, Obihiro, Japan
| | - Kenta Watanabe
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita, Japan
| | - Kiichi Takahashi
- Department of Gastroenterology, Hachinohe City Hospital, Hachinohe, Japan
| | - Hiroharu Echigo
- Department of Gastroenterology, Iwaki City Medical Center, Iwaki, Japan
| | - Yasuaki Abe
- Department of Gastroenterology, Yamagata City Hospital Saiseikan, Yamagata, Japan
| | - Hiroyuki Endo
- Department of Gastroenterology, Japan Community Health Care Organization Sendai Hospital, Sendai, Japan
| | - Tomoki Okata
- Department of Gastroenterology, Iwate Prefectural Chubu Hospital, Kitakami, Japan
| | - Tatsuya Hoshi
- Department of Gastroenterology, Kesennuma City Hospital, Kesennuma, Japan
| | | | | | | | - Naoki Nakaya
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Katsunori Iijima
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita, Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
44
|
Meijnikman AS, Nieuwdorp M, Schnabl B. Endogenous ethanol production in health and disease. Nat Rev Gastroenterol Hepatol 2024; 21:556-571. [PMID: 38831008 DOI: 10.1038/s41575-024-00937-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/23/2024] [Indexed: 06/05/2024]
Abstract
The gut microbiome exerts metabolic actions on distal tissues and organs outside the intestine, partly through microbial metabolites that diffuse into the circulation. The disruption of gut homeostasis results in changes to microbial metabolites, and more than half of the variance in the plasma metabolome can be explained by the gut microbiome. Ethanol is a major microbial metabolite that is produced in the intestine of nearly all individuals; however, elevated ethanol production is associated with pathological conditions such as metabolic dysfunction-associated steatotic liver disease and auto-brewery syndrome, in which the liver's capacity to metabolize ethanol is surpassed. In this Review, we describe the mechanisms underlying excessive ethanol production in the gut and the role of ethanol catabolism in mediating pathogenic effects of ethanol on the liver and host metabolism. We conclude by discussing approaches to target excessive ethanol production by gut bacteria.
Collapse
Affiliation(s)
| | - Max Nieuwdorp
- Department of Internal Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, Netherlands
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, Netherlands
- Diabeter Centrum Amsterdam, Amsterdam, Netherlands
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA.
- Center for Innovative Phage Applications and Therapeutics, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
45
|
Pallozzi M, De Gaetano V, Di Tommaso N, Cerrito L, Santopaolo F, Stella L, Gasbarrini A, Ponziani FR. Role of Gut Microbial Metabolites in the Pathogenesis of Primary Liver Cancers. Nutrients 2024; 16:2372. [PMID: 39064815 PMCID: PMC11280141 DOI: 10.3390/nu16142372] [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: 05/29/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Hepatobiliary malignancies, which include hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), are the sixth most common cancers and the third leading cause of cancer-related death worldwide. Hepatic carcinogenesis is highly stimulated by chronic inflammation, defined as fibrosis deposition, and an aberrant imbalance between liver necrosis and nodular regeneration. In this context, the gut-liver axis and gut microbiota have demonstrated a critical role in the pathogenesis of HCC, as dysbiosis and altered intestinal permeability promote bacterial translocation, leading to chronic liver inflammation and tumorigenesis through several pathways. A few data exist on the role of the gut microbiota or bacteria resident in the biliary tract in the pathogenesis of CCA, and some microbial metabolites, such as choline and bile acids, seem to show an association. In this review, we analyze the impact of the gut microbiota and its metabolites on HCC and CCA development and the role of gut dysbiosis as a biomarker of hepatobiliary cancer risk and of response during anti-tumor therapy. We also discuss the future application of gut microbiota in hepatobiliary cancer management.
Collapse
Affiliation(s)
- Maria Pallozzi
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Valeria De Gaetano
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Natalia Di Tommaso
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Lucia Cerrito
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Francesco Santopaolo
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Leonardo Stella
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Antonio Gasbarrini
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Liver Unit, Centro Malattie dell’Apparato Digerente (CEMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, 00168 Rome, Italy; (M.P.); (V.D.G.); (N.D.T.); (L.C.); (F.S.); (L.S.); (A.G.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
46
|
Yu M, Li Q, Dolios G, Tu P, Teitelbaum S, Chen J, Petrick L. Active Molecular Network Discovery Links Lifestyle Variables to Breast Cancer in the Long Island Breast Cancer Study Project. ENVIRONMENT & HEALTH (WASHINGTON, D.C.) 2024; 2:401-410. [PMID: 38932753 PMCID: PMC11197006 DOI: 10.1021/envhealth.3c00218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 06/28/2024]
Abstract
A healthy lifestyle has been associated with decreased risk of developing breast cancer. Using untargeted metabolomics profiling, which provides unbiased information regarding lifestyle choices such as diet and exercise, we aim to identify the molecular mechanisms connecting lifestyle and breast cancer through network analysis. A total of 100 postmenopausal women, 50 with breast cancer and 50 cancer-free controls, were selected from the Long Island Breast Cancer Study Project (LIBCSP). We measured untargeted plasma metabolomics using liquid chromatography-high-resolution mass spectrometry (LC-HRMS). Using the "enet" package, we retained highly correlated metabolites representing active molecular network (AMN) clusters for analysis. LASSO was used to examine associations between cancer status and AMN metabolites and covariates such as BMI, age, and reproductive factors. LASSO was then repeated to examine associations between AMN metabolites and 10 lifestyle-related variables including smoking, physical activity, alcohol consumption, meat consumption, fruit and vegetable consumption, and supplemental vitamin use. Results were displayed as a network to uncover biological pathways linking lifestyle factors to breast cancer status. After filtering, 851 "active" metabolites out of 1797 metabolomics were retained in 197 correlation AMN clusters. Using LASSO, breast cancer status was associated with 71 "active" metabolites. Several of these metabolites were associated with lifestyle variables including meat consumption, alcohol consumption, and supplemental β-carotene, B12, and folate use. Those metabolites could potentially serve as molecular-level biological intermediaries connecting healthy lifestyle factors to breast cancer, even though direct associations between breast cancer and the investigated lifestyles at the phenotype level are not evident. In particular, DiHODE, a metabolite linked with inflammation, was associated with breast cancer status and connected to β-carotene supplement usage through an AMN. We found several plasma metabolites associated with lifestyle factors and breast cancer status. Future studies investigating the mechanistic role of inflammation in linking supplement usage to breast cancer status are warranted.
Collapse
Affiliation(s)
- Miao Yu
- Department
of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- The
Jackson Laboratory, Farmington, Connecticut 06032, United States
| | - Qian Li
- Department
of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Georgia Dolios
- Department
of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Peijun Tu
- Department
of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Susan Teitelbaum
- Department
of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- The
Institute for Exposomics Research, Icahn
School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Jia Chen
- Department
of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- The
Institute for Exposomics Research, Icahn
School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Lauren Petrick
- Department
of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- The
Institute for Exposomics Research, Icahn
School of Medicine at Mount Sinai, New York, New York 10029, United States
- The
Bert Strassburger Metabolic Center, Sheba
Medical Center, Tel-Hashomer 5266202, Israel
| |
Collapse
|
47
|
Eloranta R, Vilén ST, Keinänen A, Salo T, Qannam A, Bello IO, Snäll J. Oral squamous cell carcinoma: Effect of tobacco and alcohol on cancer location. Tob Induc Dis 2024; 22:TID-22-112. [PMID: 38895166 PMCID: PMC11185050 DOI: 10.18332/tid/189303] [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: 12/07/2023] [Revised: 05/12/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
INTRODUCTION The underlying factors of oral squamous cell cancers (OSCC) have been elucidated, but studies have focused little on etiological differences in affected oral cavity sites. The aim of this retrospective study was to clarify the role of carcinogen exposure in OSCC of different oral cavity areas. METHODS A cross-sectional study of patients with primary OSCC was conducted retrospectively, based on patient records from Helsinki University Hospital, Finland, between January 2016 and December 2020. The patients' self-reported history of tobacco smoking and alcohol use was explained by tumor site, age, sex, tumor size, and lymph node status in a logistic regression model. The information on smoking and alcohol use was compiled from a patient background form. RESULTS In 519 patients, tumors occurred most often in the tongue (51%), gingiva (21%), or floor of the mouth (FOM; 15%). FOM had 26-fold greater odds for a history of smoking and alcohol use than other tumor sites (OR=25.78; 95% CI: 8.02-82.95; p<0.001). Gingival and buccal sites were associated significantly less with smoking and alcohol use (OR=0.43, 95% CI: 0.28-0.67; p<0.001 and OR=0.47; 95% CI: 0.25-0.92; p<0.026, respectively). Patients of older age were less likely to have a history of smoking and alcohol use (AOR=0.95; 95% CI: 0.94-0.97; p<0.001) than younger patients. Tumor size (T3-4) and FOM increased the odds for history of smoking and alcohol use (AOR=1.73; 95% CI: 1.15-2.60; p=0.009 and AOR=26.15; 95% CI: 8.01-84.84; p<0.001, respectively). CONCLUSIONS OSCC of oral cavity sites has notable differences in etiology. FOM seems to be related almost exclusively to conventional smoking and heavy alcohol use.
Collapse
Affiliation(s)
- Riikka Eloranta
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Suvi-Tuuli Vilén
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
- Department of Oral and Maxillofacial Diseases, Kymenlaakso Central Hospital Kotka, Kotka, Finland
| | - Arvi Keinänen
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
- Translational Immunology Research Program (TRIMM), University of Helsinki, Helsinki, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
- Department of Pathology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Ahmed Qannam
- Department of Oral Medicine and Diagnostic Sciences, King Saud University College of Dentistry, Riyadh, Saudi Arabia
| | - Ibrahim O. Bello
- Department of Oral Medicine and Diagnostic Sciences, King Saud University College of Dentistry, Riyadh, Saudi Arabia
| | - Johanna Snäll
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
48
|
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
|
49
|
Papadakos SP, Chatzikalil E, Arvanitakis K, Vakadaris G, Stergiou IE, Koutsompina ML, Argyrou A, Lekakis V, Konstantinidis I, Germanidis G, Theocharis S. Understanding the Role of Connexins in Hepatocellular Carcinoma: Molecular and Prognostic Implications. Cancers (Basel) 2024; 16:1533. [PMID: 38672615 PMCID: PMC11048329 DOI: 10.3390/cancers16081533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Connexins, a family of tetraspan membrane proteins forming intercellular channels localized in gap junctions, play a pivotal role at the different stages of tumor progression presenting both pro- and anti-tumorigenic effects. Considering the potential role of connexins as tumor suppressors through multiple channel-independent mechanisms, their loss of expression may be associated with tumorigenic activity, while it is hypothesized that connexins favor the clonal expansion of tumor cells and promote cell migration, invasion, and proliferation, affecting metastasis and chemoresistance in some cases. Hepatocellular carcinoma (HCC), characterized by unfavorable prognosis and limited responsiveness to current therapeutic strategies, has been linked to gap junction proteins as tumorigenic factors with prognostic value. Notably, several members of connexins have emerged as promising markers for assessing the progression and aggressiveness of HCC, as well as the chemosensitivity and radiosensitivity of hepatocellular tumor cells. Our review sheds light on the multifaceted role of connexins in HCC pathogenesis, offering valuable insights on recent advances in determining their prognostic and therapeutic potential.
Collapse
Affiliation(s)
- Stavros P. Papadakos
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.P.P.); (E.C.)
| | - Elena Chatzikalil
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.P.P.); (E.C.)
| | - Konstantinos Arvanitakis
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (K.A.); (G.V.)
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Georgios Vakadaris
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (K.A.); (G.V.)
| | - Ioanna E. Stergiou
- Pathophysiology Department, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.E.S.); (M.-L.K.)
| | - Maria-Loukia Koutsompina
- Pathophysiology Department, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.E.S.); (M.-L.K.)
| | - Alexandra Argyrou
- Academic Department of Gastroenterology, Laikon General Hospital, Athens University Medical School, 11527 Athens, Greece; (A.A.); (V.L.)
| | - Vasileios Lekakis
- Academic Department of Gastroenterology, Laikon General Hospital, Athens University Medical School, 11527 Athens, Greece; (A.A.); (V.L.)
| | | | - Georgios Germanidis
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (K.A.); (G.V.)
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.P.P.); (E.C.)
| |
Collapse
|
50
|
Schreurs M, Piampongsant S, Roncoroni M, Cool L, Herrera-Malaver B, Vanderaa C, Theßeling FA, Kreft Ł, Botzki A, Malcorps P, Daenen L, Wenseleers T, Verstrepen KJ. Predicting and improving complex beer flavor through machine learning. Nat Commun 2024; 15:2368. [PMID: 38531860 PMCID: PMC10966102 DOI: 10.1038/s41467-024-46346-0] [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: 10/30/2023] [Accepted: 02/21/2024] [Indexed: 03/28/2024] Open
Abstract
The perception and appreciation of food flavor depends on many interacting chemical compounds and external factors, and therefore proves challenging to understand and predict. Here, we combine extensive chemical and sensory analyses of 250 different beers to train machine learning models that allow predicting flavor and consumer appreciation. For each beer, we measure over 200 chemical properties, perform quantitative descriptive sensory analysis with a trained tasting panel and map data from over 180,000 consumer reviews to train 10 different machine learning models. The best-performing algorithm, Gradient Boosting, yields models that significantly outperform predictions based on conventional statistics and accurately predict complex food features and consumer appreciation from chemical profiles. Model dissection allows identifying specific and unexpected compounds as drivers of beer flavor and appreciation. Adding these compounds results in variants of commercial alcoholic and non-alcoholic beers with improved consumer appreciation. Together, our study reveals how big data and machine learning uncover complex links between food chemistry, flavor and consumer perception, and lays the foundation to develop novel, tailored foods with superior flavors.
Collapse
Affiliation(s)
- Michiel Schreurs
- VIB-KU Leuven Center for Microbiology, Gaston Geenslaan 1, B-3001, Leuven, Belgium
- CMPG Laboratory of Genetics and Genomics, KU Leuven, Gaston Geenslaan 1, B-3001, Leuven, Belgium
- Leuven Institute for Beer Research (LIBR), Gaston Geenslaan 1, B-3001, Leuven, Belgium
| | - Supinya Piampongsant
- VIB-KU Leuven Center for Microbiology, Gaston Geenslaan 1, B-3001, Leuven, Belgium
- CMPG Laboratory of Genetics and Genomics, KU Leuven, Gaston Geenslaan 1, B-3001, Leuven, Belgium
- Leuven Institute for Beer Research (LIBR), Gaston Geenslaan 1, B-3001, Leuven, Belgium
| | - Miguel Roncoroni
- VIB-KU Leuven Center for Microbiology, Gaston Geenslaan 1, B-3001, Leuven, Belgium
- CMPG Laboratory of Genetics and Genomics, KU Leuven, Gaston Geenslaan 1, B-3001, Leuven, Belgium
- Leuven Institute for Beer Research (LIBR), Gaston Geenslaan 1, B-3001, Leuven, Belgium
| | - Lloyd Cool
- VIB-KU Leuven Center for Microbiology, Gaston Geenslaan 1, B-3001, Leuven, Belgium
- CMPG Laboratory of Genetics and Genomics, KU Leuven, Gaston Geenslaan 1, B-3001, Leuven, Belgium
- Leuven Institute for Beer Research (LIBR), Gaston Geenslaan 1, B-3001, Leuven, Belgium
- Laboratory of Socioecology and Social Evolution, KU Leuven, Naamsestraat 59, B-3000, Leuven, Belgium
| | - Beatriz Herrera-Malaver
- VIB-KU Leuven Center for Microbiology, Gaston Geenslaan 1, B-3001, Leuven, Belgium
- CMPG Laboratory of Genetics and Genomics, KU Leuven, Gaston Geenslaan 1, B-3001, Leuven, Belgium
- Leuven Institute for Beer Research (LIBR), Gaston Geenslaan 1, B-3001, Leuven, Belgium
| | - Christophe Vanderaa
- Laboratory of Socioecology and Social Evolution, KU Leuven, Naamsestraat 59, B-3000, Leuven, Belgium
| | - Florian A Theßeling
- VIB-KU Leuven Center for Microbiology, Gaston Geenslaan 1, B-3001, Leuven, Belgium
- CMPG Laboratory of Genetics and Genomics, KU Leuven, Gaston Geenslaan 1, B-3001, Leuven, Belgium
- Leuven Institute for Beer Research (LIBR), Gaston Geenslaan 1, B-3001, Leuven, Belgium
| | - Łukasz Kreft
- VIB Bioinformatics Core, VIB, Rijvisschestraat 120, B-9052, Ghent, Belgium
| | - Alexander Botzki
- VIB Bioinformatics Core, VIB, Rijvisschestraat 120, B-9052, Ghent, Belgium
| | | | - Luk Daenen
- AB InBev SA/NV, Brouwerijplein 1, B-3000, Leuven, Belgium
| | - Tom Wenseleers
- Laboratory of Socioecology and Social Evolution, KU Leuven, Naamsestraat 59, B-3000, Leuven, Belgium
| | - Kevin J Verstrepen
- VIB-KU Leuven Center for Microbiology, Gaston Geenslaan 1, B-3001, Leuven, Belgium.
- CMPG Laboratory of Genetics and Genomics, KU Leuven, Gaston Geenslaan 1, B-3001, Leuven, Belgium.
- Leuven Institute for Beer Research (LIBR), Gaston Geenslaan 1, B-3001, Leuven, Belgium.
| |
Collapse
|