101
|
Li B, Gao H, Xiao H, He H, Ni Q, Li Q, Wang H, Chen L. Abnormal chenodexycholic acid metabolism programming promotes cartilage matrix degradation in male adult offspring rats induced by prenatal caffeine exposure. Toxicol Res (Camb) 2025; 14:tfaf063. [PMID: 40331087 PMCID: PMC12051868 DOI: 10.1093/toxres/tfaf063] [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: 11/18/2024] [Revised: 04/01/2025] [Accepted: 04/18/2025] [Indexed: 05/08/2025] Open
Abstract
Epidemiological evidence links osteoarthritis to fetal origins. Our study shows prenatal caffeine exposure (PCE) in rats predisposes adult offspring to osteoarthritis, associated with elevated intrauterine glucocorticoid levels. Previous research indicates that chenodeoxycholic acid (CDCA), a bile acid, can slow osteoarthritis progression when administered intra-articularly. This study explored if disrupted bile acid metabolism in cartilage affects osteoarthritis risk in adult offspring with PCE. Our findings indicate that the expression of MMP3/MMP13 was upregulated, while endogenous CDCA levels were reduced in the cartilage of PCE-exposed offspring. Furthermore, we observed a persistent reduction in H3K27ac levels at the CYP7B1 promoter and its expression in the cartilage of PCE offspring from fetus to adulthood. Moreover, a sub-physiological level of CDCA promoted NF-κB phosphorylation and the expression of MMP3/MMP13 in chondrocytes in vitro. High levels of glucocorticoids reduced H3K27ac levels and CYP7B1 expression in the promoter region of CYP7B1 through the glucocorticoid receptor and histone deacetylase 4, consequently leading to decreased CDCA levels. In summary, our findings suggest that intrauterine low-expression programming of CYP7B1, induced by elevated glucocorticoid levels, reduces local CDCA levels in the cartilage of PCE offspring, ultimately leading to increased matrix degradation and susceptibility to osteoarthritis.
Collapse
Affiliation(s)
- Bin Li
- Division of Joint Surgery and Sports Medicine, Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| | - Hui Gao
- Division of Joint Surgery and Sports Medicine, Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hao Xiao
- Division of Joint Surgery and Sports Medicine, Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| | - Hangyuan He
- Division of Joint Surgery and Sports Medicine, Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Qubo Ni
- Division of Joint Surgery and Sports Medicine, Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Qingxian Li
- Division of Joint Surgery and Sports Medicine, Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hui Wang
- Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
- Department of Pharmacology, Wuhan University Taikang Medical School (School of Basic Medical Sciences), Wuhan 430071, China
| | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopaedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Hubei Provincial Key Laboratory of Developmentally Originated Diseases, Wuhan 430071, China
| |
Collapse
|
102
|
Lin M, Wang Y, Zhao Y, Zhai X, Hu Y, Luan Q, Lv L, Wang Z, Yao J. Protective effects of albiflorin on acetaminophen-induced hepatotoxicity: Regulation of blood-biliary barrier integrity by ATF3. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156722. [PMID: 40250002 DOI: 10.1016/j.phymed.2025.156722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 03/19/2025] [Accepted: 04/01/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Acetaminophen-induced hepatotoxicity remains a clinical challenge with limited targeted therapeutic options. While recent advances have identified blood‒biliary barrier disruption as a critical pathogenic mechanism, effective interventions preserving this barrier are notably lacking. Albiflorin, a key bioactive constituent of Paeonia lactiflora Pall., exhibits unique bile acid modulation along with antioxidant and anti-inflammatory activities, suggesting its potential efficacy in preventing acetaminophen-induced liver injury. However, its specific role and underlying mechanisms in alleviating acetaminophen-induced hepatotoxicity remain unclear. OBJECTIVE This study's objective was to examine the pharmacological effects and primary molecular mechanisms of albiflorin in alleviating acetaminophen-induced liver injury. METHODS An acetaminophen-induced liver injury mouse model was created using a 300 mg/kg dose of acetaminophen. The hepatoprotective effects of albiflorin were assessed through histological and biochemical analyses. Blood‒biliary barrier integrity was evaluated via Evans blue dye tests, immunofluorescence, and bile acid assays. Transcriptomic analysis, gene overexpression and interference techniques, and ChIP‒qPCR were employed to explore the molecular mechanisms underlying the protective effects of albiflorin. RESULTS Albiflorin significantly reduced acetaminophen-induced liver injury, as evidenced by improved biochemical profiles and hepatocyte morphology. It also prevented increases in blood‒biliary barrier permeability and bile acid levels. RNA sequencing identified 3,184 differentially expressed genes, revealing critical pathways involved in maintaining blood‒biliary barrier integrity. AF reversed the acetaminophen-induced changes in the expression of genes related to the blood‒biliary barrier, particularly occludin, claudin 5, ABCG5, and ABCG8. Albiflorin protected the blood‒biliary barrier and mitigated acetaminophen-induced liver injury by enhancing ATF3 protein stability, with ATF3 identified as a critical mediator of these protective effects. CONCLUSION This study provides pioneering evidence that albiflorin protects against acetaminophen-induced liver injury by interacting with ATF3 to regulate blood-biliary barrier proteins and maintain the integrity of the blood-biliary barrier. These findings deepen our understanding of the role of the blood‒biliary barrier in liver diseases and suggest a therapeutic strategy for drug overdose-induced liver injury.
Collapse
Affiliation(s)
- Musen Lin
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning, 116000, China; Department of Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Yue Wang
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Yan Zhao
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Xiaohan Zhai
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Yan Hu
- Department of Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Qinrong Luan
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Li Lv
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Zhecheng Wang
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning, 116000, China.
| | - Jihong Yao
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning, 116000, China.
| |
Collapse
|
103
|
Niaki NM, Hatefnia F, Heidari MM, Tabean M, Mobed A. Alpha-Fetoprotein (AFP) biosensors. Clin Chim Acta 2025; 573:120293. [PMID: 40216053 DOI: 10.1016/j.cca.2025.120293] [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: 03/15/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/25/2025]
Abstract
Alpha-fetoprotein (AFP) is a glycoprotein mainly produced during fetal development, and elevated levels in adults are frequently associated with liver diseases, especially hepatocellular carcinoma (HCC), as well as certain germ cell tumors. Measuring AFP in biological samples is crucial for early diagnosis, monitoring disease progression, and evaluating treatment efficacy. While traditional detection methods like enzyme-linked immunosorbent assay (ELISA) and radioimmunoassay are dependable, they often face limitations such as lengthy processes, complexity, and the need for specialized equipment. In recent years, biosensing technologies have emerged as promising alternatives for detecting AFP, offering advantages like increased sensitivity, real-time monitoring, and ease of use. Various biosensing platforms, including electrochemical, optical, and piezoelectric sensors, have been developed to enable quick and specific detection of AFP. These sensors employ molecular recognition elements, such as antibodies, aptamers, or nanoparticles, to selectively bind AFP, producing a measurable signal. This article explores the structure and mechanisms of action of AFP, the diseases linked to it, and describes several biosensing technologies. It also reviews recent advancements in AFP biosensing, discussing their principles, performance, and potential applications in clinical settings. Furthermore, the article highlights the challenges and future prospects for developing cost-effective, portable, and multiplexed AFP biosensors, underscoring their potential to revolutionize early disease detection and personalized healthcare.
Collapse
Affiliation(s)
- Nava Moghadasian Niaki
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faezeh Hatefnia
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Mahdi Heidari
- Department of Pediatrics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Tabean
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ahmad Mobed
- Social Determinants of Health Research Center, Health Management and Safety Promotion, Iran.
| |
Collapse
|
104
|
Skvarce J, Bui A, Oro P, Sachar S, Harnegie MP, Kapoor A, Lindenmeyer CC, Siuba MT. Multisystem hemodynamic effects of terlipressin in cirrhosis: A scoping review. J Crit Care 2025; 87:155038. [PMID: 39955856 DOI: 10.1016/j.jcrc.2025.155038] [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: 09/15/2024] [Revised: 01/05/2025] [Accepted: 02/04/2025] [Indexed: 02/18/2025]
Abstract
INTRODUCTION Hepatorenal syndrome (HRS-AKI) is a serious complication of advanced liver disease. Pharmacologic options are limited in effectiveness, therefore liver transplantation is the definitive therapy. Early investigation into terlipressin as a first-line therapy for HRS-AKI has been promising but expected hemodynamic changes across organ systems in patients with cirrhosis have not been thoroughly examined. METHODS We conducted a scoping review of the literature including adult patients with cirrhosis who received terlipressin and hemodynamic parameters were recorded. Non-human studies, case reports, conference abstracts, and review articles were excluded. Searches were performed up to December 2024 in the following databases: MEDLINE, Embase, Cochrane Library, Scopus, Web of Science, and CINAHL. RESULTS Of 2022 studies retrieved, 56 studies met inclusion criteria. Heart rate, mean arterial pressure, and cardiac output were the most reported parameters. Pulmonary arterial pressure and wedge pressure were the next most common. Systemic vascular resistance, hepatic and renal measures such as resistive indices and portal pressure gradients had fewer studies. Studies reported decreased heart rate, increased mean arterial pressure, decreased cardiac output/index, and increased systemic vascular resistance. Other hemodynamic outcomes were more varied across studies. CONCLUSIONS Terlipressin exerts a variety of hemodynamic effects across organ systems and vascular beds. More studies are required to understand if any hemodynamic parameters might predict terlipressin response or adverse events.
Collapse
Affiliation(s)
- Jeremy Skvarce
- Department of Internal Medicine, Community Care Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Albert Bui
- Department of Critical Care Medicine, Integrated Hospital Care Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Peter Oro
- Department of Internal Medicine, Community Care Institute, Cleveland Clinic South Pointe, Cleveland, OH, USA
| | - Saloni Sachar
- Department of Internal Medicine, Community Care Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | | | - Aanchal Kapoor
- Department of Critical Care Medicine, Integrated Hospital Care Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Christina C Lindenmeyer
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Matthew T Siuba
- Department of Critical Care Medicine, Integrated Hospital Care Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
| |
Collapse
|
105
|
Alhalabi H, Korsmeier L, Thomas A, Thevis M. Investigations Into the Urinary Metabolite Elimination Profile of the Selective Androgen Receptor Modulator S-23 in Studies Mimicking Contaminated Product Ingestion for Doping Control Purposes. Biomed Chromatogr 2025; 39:e70090. [PMID: 40277337 PMCID: PMC12023825 DOI: 10.1002/bmc.70090] [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: 03/21/2025] [Revised: 03/26/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025]
Abstract
Selective androgen receptor modulators (SARMs) have repeatedly been reason of adverse analytical findings (AAFs) in routine doping controls. Among these, S-23 has been identified in five AAFs reported in 2022. In addition to intentional doping, inadvertent exposure through contaminated dietary supplements has emerged as a significant concern, purportedly as well as evidently contributing to AAFs involving SARMs. Thus, the differentiation of inadvertent intake and intentional abuse of S-23 is of growing relevance. This study aimed at investigating the urinary concentration profile of microdosed S-23 and to characterize the elimination pattern. Single and multidose administration studies with 1, 10, and 50 μg of S-23 were conducted, and collected urine samples were analyzed by LC-MS/MS following enzymatic hydrolysis and solid-phase extraction. The analytical method was validated for a semiquantitative detection of S-23 and characterized by a limit of detection of 1 pg/mL. A total of 18 metabolites was detected in human in vivo samples following oral administration of microdosed S-23. Moreover, the study demonstrated that a single dose of 1 μg can be detected for an average of up to 253 h, while a single dose of 50 μg can be detected up to 544 h on average.
Collapse
Affiliation(s)
- Hana Alhalabi
- Center for Preventive Doping Research, Institute of BiochemistryGerman Sport University CologneCologneGermany
| | - Linus Korsmeier
- Center for Preventive Doping Research, Institute of BiochemistryGerman Sport University CologneCologneGermany
| | - Andreas Thomas
- Center for Preventive Doping Research, Institute of BiochemistryGerman Sport University CologneCologneGermany
| | - Mario Thevis
- Center for Preventive Doping Research, Institute of BiochemistryGerman Sport University CologneCologneGermany
- European Monitoring Center for Emerging Doping Agents (EUMoCEDA)CologneGermany
| |
Collapse
|
106
|
Mohammed MJ, Kadhim HM. The hepatoprotective effects of the polyphenol-enriched n-butanol fraction of Cnicus benedictus against carbon tetrachloride-induced liver fibrosis in rats: In vivo study. Toxicol Rep 2025; 14:101850. [PMID: 39758800 PMCID: PMC11697782 DOI: 10.1016/j.toxrep.2024.101850] [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: 10/11/2024] [Revised: 11/21/2024] [Accepted: 12/03/2024] [Indexed: 01/07/2025] Open
Abstract
Liver fibrosis is a continuous wound-healing response to chronic injury caused by various chemical, virus, and pathological disorders; the lack of approved drugs or methods to reverse or prevent liver fibrosis makes it an interesting area of research. This study investigates the potential hepatoprotective effects of the phenolic extract of Cnicus benedictus in rat's module of liver fibrosis. Liver fibrosis was induced by intraperitoneal injection of carbon tetrachloride (CCl4) for six consecutive weeks; the butanol fraction of Cnicus and silymarin was administered orally concurrently with CCl4. After six weeks, all animals were euthanized. Rat liver tissue levels of malondialdehyde (MDA) and glutathione (GSH) were measured, and serum liver enzymes and protein were measured using the ELISA technique. Histopathological study and immunohistochemistry of liver tissue for transforming growth factor (TGF-β1), alpha-smooth muscle actin (α-SMA), and hydroxyproline were assessed. In HPLC analysis, Cnicus extract showed several components, including quercetin, gallic acid, rutin, kaempferol, silibinin, and apigenin. Treatment with Cnicus butanol extract reduces serum ALT, AST, bilirubin, and albumin levels compared to induction. Additionally, Cnicus extract increases liver GSH levels and decreases liver MDA levels compared to induction. Liver tissue of TGF-β1, α-SMA, and hydroxyproline expression was downregulated in rats receiving Cnicus extract. Liver tissue histopathology showed improvement in its features compared to the induction group. In conclusion, oral administration of the polyphenol-enriched n-butanol fraction of Cnicus benedictus showed a protective effect on liver fibrosis caused by CCl4, possibly through antioxidant and anti-inflammatory mechanisms.
Collapse
Affiliation(s)
- Mohammed Jasim Mohammed
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad, Iraq
- Ministry of Health and Environment, Kirkuk Health Directorate, Kirkuk, Iraq
| | - Haitham Mahmood Kadhim
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Nahrain University, Baghdad, Iraq
| |
Collapse
|
107
|
Liang G, Ma Y, Deng P, Li S, He C, He H, Liu H, Fan Y, Li Z. Role of cell-based therapies in digestive disorders: Obstacles and opportunities. Regen Ther 2025; 29:1-18. [PMID: 40124469 PMCID: PMC11925584 DOI: 10.1016/j.reth.2025.02.009] [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: 12/22/2024] [Revised: 02/01/2025] [Accepted: 02/20/2025] [Indexed: 03/25/2025] Open
Abstract
Stem cell-based therapies have emerged as a promising frontier in the treatment of gastrointestinal disorders, offering potential solutions for challenges posed by conventional treatments. This review comprehensively examines recent advancements in cell-based therapeutic strategies, particularly focusing on stem cell applications, immunotherapy, and cellular therapies for digestive diseases. It highlights the successful differentiation of enteric neural progenitors from pluripotent stem cells and their application in animal models, such as Hirschsprung disease. Furthermore, the review evaluates clinical trials and experimental studies demonstrating the potential of stem cells in regenerating damaged tissues, modulating immune responses, and promoting healing in conditions like Crohn's disease and liver failure. By addressing challenges, such as scalability, immunogenicity, and ethical considerations, the review underscores the translational opportunities and obstacles in realizing the clinical potential of these therapies. Concluding with an emphasis on future directions, the study provides insights into optimizing therapeutic efficacy and fostering innovations in personalized medicine for digestive disorders.
Collapse
Affiliation(s)
- Guodong Liang
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| | - Yuehan Ma
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| | - Ping Deng
- Medical Department, Jilin Cancer Hospital, Changchun 130012, China
| | - Shufeng Li
- First Department of Gynecological Tumor, Jilin Cancer Hospital, Changchun 130012, China
| | - Chunyan He
- Department of Anaesthesia, Jilin Cancer Hospital, Changchun 130012, China
| | - Haihang He
- Department of Otorhinolaryngology, Oral Maxillofacial, Head and Neck, Jilin Cancer Hospital, Changchun 130012, China
| | - Hairui Liu
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| | - Yunda Fan
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| | - Ze Li
- First Surgery Department of Colorectal, Gastric and Abdominal Tumors, Jilin Cancer Hospital, Changchun 130012, China
| |
Collapse
|
108
|
Panganiban J, Kehar M, Ibrahim SH, Hartmann P, Sood S, Hassan S, Ramirez CM, Kohli R, Censani M, Mauney E, Cuda S, Karjoo S. Metabolic dysfunction-associated steatotic liver disease (MASLD) in children with obesity: An Obesity Medicine Association (OMA) and expert joint perspective 2025. OBESITY PILLARS 2025; 14:100164. [PMID: 40230708 PMCID: PMC11995806 DOI: 10.1016/j.obpill.2025.100164] [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: 11/27/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 04/16/2025]
Abstract
Introduction This Obesity Medicine Association (OMA) Expert Joint Perspective examines steatotic liver disease (SLD), which is composed of metabolic dysfunction-associated steatotic liver disease (MASLD), and metabolic dysfunction-associated steatohepatitis (MASH) in children with obesity. The prevalence of obesity is increasing, rates have tripled since 1963 from 5 % to now 19 % of US children affected in 2018. MASLD, is the most common liver disease seen in children, can be a precursor to the development of Type 2 Diabetes (T2DM) and is the primary reason for liver transplant listing in young adults. We must be vigilant in prevention and treatment of MASLD in childhood to prevent further progression. Methods This joint clinical perspective is based upon scientific evidence, peer and clinical expertise. The medical literature was reviewed via PubMed search and appropriate articles were included in this review. This work was formulated from the collaboration of eight hepatologists/gastroenterologists with MASLD expertise and two physicians from the OMA. Results The authors who are experts in the field, determined sentinel questions often asked by clinicians regarding MASLD in children with obesity. They created a consensus and clinical guideline for clinicians on the screening, diagnosis, and treatment of MASLD associated with obesity in children. Conclusions Obesity and the comorbidity of MASLD is increasing in children, and this is a medical problem that needs to be addressed urgently. It is well known that children with metabolic associated chronic disease often continue to have these chronic diseases as adults, which leads to reduced life expectancy, quality of life, and increasing healthcare needs and financial burden. The authors of this paper recommend healthy weight reduction not only through lifestyle modification but through obesity pharmacotherapy and bariatric surgery. Therefore, this guidance reviews available therapies to achieve healthy weight reduction and reverse MASLD to prevent progressive liver fibrosis, and metabolic disease.
Collapse
Affiliation(s)
| | - Mohit Kehar
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Children's Hospital of Eastern Ontario, Ottawa, Canada
| | - Samar H. Ibrahim
- Division of Pediatric Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Phillipp Hartmann
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Division of Gastroenterology, Hepatology & Nutrition, Rady Children’s Hospital San Diego, San Diego, CA, USA
| | - Shilpa Sood
- Division of Pediatric Gastroenterology, Boston Children's Health Physicians, New York Medical College, Valhalla, NY, USA
| | - Sara Hassan
- University of Texas Southwestern, Dallas, TX, United States
| | | | - Rohit Kohli
- Children's Hospital Los Angeles, CA, United States
| | - Marisa Censani
- Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY, United States
| | - Erin Mauney
- Tufts Medical Center, Boston, MA, United States
| | - Suzanne Cuda
- Alamo City Healthy Kids and Families, San Antonio, TX, United States
| | - Sara Karjoo
- Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
- University of South Florida, Tampa, FL, United States
- Florida State University, Tallahassee, FL, United States
| |
Collapse
|
109
|
Dong R, Luo Z, Xue H, Shao J, Chen L, Jin W, Yang L, Shen C, Xu M, Wu M, Wang J. Development and Validation of an Explainable Machine Learning Model for Warning of Hepatitis E Virus-Related Acute Liver Failure. Liver Int 2025; 45:e70129. [PMID: 40344287 DOI: 10.1111/liv.70129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 03/22/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND AND AIMS Early identification of patients with acute hepatitis E (AHE) who are at high risk of progressing to hepatitis E virus-related acute liver failure (HEV-ALF) is crucial for enabling timely monitoring and intervention. This multicentre retrospective cohort study aimed to develop and validate an interpretable machine learning (ML) model for predicting the risk of HEV-ALF in hospitalised patients with AHE in tertiary care settings. METHODS The study cohort included patients admitted to seven tertiary medical centers in Jiangsu, China, between 01 January 2018 and 31 December 2024. Multiple ML algorithms were applied for feature selection and model training. The predictive performance of the models was evaluated in terms of discrimination, calibration and clinical net benefit. The interpretability of the final model was enhanced using the SHapley Additive exPlanations. RESULTS A total of 1912 participants were included in the study. Ten ML models were developed based on seven consensus-selected baseline features, with the survival gradient boosting machine (GBM) demonstrating superior performance compared to the traditional Cox proportional hazards regression model and other relevant models or scores. The GBM model achieved a Harrell's concordance index of 0.853 (95% CI: 0.791-0.914) in the external validation set. To facilitate clinical application, the GBM model was interpreted globally and locally and deployed as a web-based tool using the Streamlit-Python framework. CONCLUSIONS The GBM model demonstrated excellent performance in predicting HEV-ALF risk in hospitalised patients with AHE, offering a promising tool for clinical decision-making.
Collapse
Affiliation(s)
- Rui Dong
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, China
| | - Zhenghan Luo
- Department of Infectious Disease Prevention and Control, Huadong Research Institute for Medicine and Biotechniques, Nanjing, China
| | - Hong Xue
- Department of Liver Disease, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Jianguo Shao
- Nantong Institute of Liver Disease, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Lin Chen
- Nantong Institute of Liver Disease, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Wenjuan Jin
- Department of Infectious Disease, The Affiliated Suzhou Ninth Hospital of Soochow University, Suzhou, China
| | - Lingmei Yang
- Nantong Institute of Liver Disease, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Chao Shen
- Department of Immunization Program, Nanjing Municipal Center for Disease Control and Prevention, Nanjing, China
| | - Minzhi Xu
- Department of Infectious Disease Prevention and Control, Huadong Research Institute for Medicine and Biotechniques, Nanjing, China
| | - Mengping Wu
- Department of Big Data Center, The Affiliated Lianyungang Hospital of Xuzhou Medical University/The First People's Hospital of Lianyungang, Lianyungang, China
| | - Jie Wang
- Department of Fundamental and Community Nursing, School of Nursing, Nanjing Medical University, Nanjing, China
- Department of Nursing, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, Taizhou, China
| |
Collapse
|
110
|
Dong T, Zhang C, Wu Z, Shuai L, Fu N, Zhang Y, Zhang L, Xiong X. A biomimetic nanomedicine alleviates liver transplant-related biliary injury by sequentially inhibiting oxidative stress and regulating macrophage polarization via Nrf-2/HO-1 and JNK pathways. Mater Today Bio 2025; 32:101797. [PMID: 40343167 PMCID: PMC12059350 DOI: 10.1016/j.mtbio.2025.101797] [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: 03/02/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025] Open
Abstract
Liver transplantation is an effective method for treating end-stage liver disease. However, 10-20 % of liver transplantation patients develop biliary injury, the main cause of which is ischemia-reperfusion injury (IRI), which consists of oxidative stress injury in the early stage and inflammatory injury in the advanced stage. Biliary injury seriously affects patient outcomes and even leads to mortality, and there are few effective treatments for IRI. Herein, nanoparticles containing quercetin (QR) and rapamycin (RP) coated with poly (lactic-co-glycolic acid) (PLGA) and encapsulated by platelet membrane (PM) were designed to treat IRI in the liver transplantation. The specific binding of ICAM-1 expressed on the PM to integrins (e.g., LFA-1 and Mac-1) in damaged vascular endothelial cells, as well as the interaction between P-selectin on the platelet surface and PSGL-1 on the macrophage surface, allows the accumulation of these biomimetic cell membrane-encapsulated nanoparticles, and subsequently, the delivery of both drugs, to ischemia-reperfusion sites in the liver. The encapsulated QR alleviated oxidative stress injury by activating the Nrf-2/HO-1 signaling pathway in the early stage in model rats with IRI and liver transplantation models. Moreover, RP alleviated inflammatory damage in the advanced stage by suppressing the JNK signaling pathway in M1 macrophages. Thus, these biomimetic nanoparticles that intervene in IRI to alleviate both the early oxidative stress and the advanced inflammatory response constitute a novel delivery system for managing biliary injury after liver transplantation.
Collapse
Affiliation(s)
| | | | - Zhaoyi Wu
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ling Shuai
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Nengsheng Fu
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yujun Zhang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Leida Zhang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xiang Xiong
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| |
Collapse
|
111
|
Saluja S, Ganguly S, Singh J, Jain A, Sharma G, Chaudhary S, Pethusamy K, Chattopadhyay P, Chopra A, Singh A, Karmakar S, Bakhshi S, Palanichamy JK. Aberrant overexpression of m6A writer and reader genes in pediatric B-Cell Acute Lymphoblastic Leukemia (B-ALL). Transl Oncol 2025; 56:102403. [PMID: 40288000 PMCID: PMC12059321 DOI: 10.1016/j.tranon.2025.102403] [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: 11/26/2024] [Revised: 03/18/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND m6A modification, regulated by writers (METTL3, METTL14), erasers (ALKBH5, FTO), and readers (IGF2BPs), is implicated in various cancers, including leukemias. METHODS In our study, we examined a cohort of 227 pediatric B-ALL patients (152 primary and 75 relapsed) and assessed the expression profiles of m6A machinery genes, including both writers and erasers, as well as the IGF2BP RNA-binding proteins, which are known as m6A readers. We also quantified the absolute percentage of m6A (m6A%). The correlation between m6A machinery gene expression and patient prognosis was studied using univariate and multivariate analyses. RESULTS Our analysis revealed a significant upregulation of m6A writers (METTL3 and METTL14), erasers (FTO), and m6A readers (IGF2BPs 1 and 3) in B-ALL patients, both in the primary and relapsed groups. m6A% levels were markedly higher in B-ALL samples than in controls. Multivariate analysis revealed that the expression of IGF2BP3, METTL3, and FTO genes, independently predicted lower overall survival and event-free survival in primary B-ALL patients. CONCLUSIONS Despite the collective dysregulation of the m6A machinery, the writers and readers appear to have a more dominant phenotype, as evidenced by the significantly elevated m6A% levels. This is the first study to analyze and establish the role of m6A machinery gene expression and its correlation with survival outcomes in a large group of B-ALL patients. These findings could aid in the development of new therapeutics targeting the m6A machinery and help predict relapse in pediatric B-ALL patients.
Collapse
Affiliation(s)
- Sumedha Saluja
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi
| | - Shuvadeep Ganguly
- Department of Medical Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital (Dr BRAIRCH), All India Institute of Medical Sciences, New Delhi
| | - Jay Singh
- Department of Laboratory Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital (Dr BRAIRCH), All India Institute of Medical Sciences, New Delhi
| | - Ayushi Jain
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi
| | - Gunjan Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi
| | - Shilpi Chaudhary
- Department of Medical Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital (Dr BRAIRCH), All India Institute of Medical Sciences, New Delhi
| | | | | | - Anita Chopra
- Department of Laboratory Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital (Dr BRAIRCH), All India Institute of Medical Sciences, New Delhi
| | - Archna Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital (Dr BRAIRCH), All India Institute of Medical Sciences, New Delhi
| | | |
Collapse
|
112
|
Cao T, Ni X, Halengbieke A, Tang J, Han Y, Sun F, Gao B, Zheng D, Yan Y, Yang X. Effects of the triglyceride-glucose index on non-alcoholic fatty liver disease: Causal evidence from longitudinal cohort studies. Arch Gerontol Geriatr 2025; 133:105813. [PMID: 40073798 DOI: 10.1016/j.archger.2025.105813] [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: 12/16/2024] [Revised: 02/08/2025] [Accepted: 03/02/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Insulin resistance (IR) is strongly related to non-alcoholic fatty liver disease (NAFLD). Triglyceride-glucose (TyG) index serves as a novel substitute indicator for IR. However, research on the effect of TyG index on NAFLD remains sparse. This study aims to investigate the causal association between TyG index and incident NAFLD. METHODS The primary cohort consisted of 27,052 participants from the Beijing Health Management Cohort, while the external validation cohort included 75,023 participants from the Taiwan MJ Cohort. Entropy balancing for continuous treatments (EBCT) combined with logistic regression and targeted maximum likelihood estimation (TMLE) were used to evaluate the causal association between the TyG index and incident NAFLD. RESULTS During a median follow-up of 2.49 years in the primary cohort, 6,168 participants (median age: 36.0 years) developed incident NAFLD. EBCT combined with logistic regression revealed the odds ratio (95 % CI) of NAFLD risk was 1.742 (1.478-2.054) for each 1-unit increase in the baseline TyG index. In the TMLE model, the risk ratio (95 % CI) for NAFLD was 1.540 (1.406-1.687) in the Q4 (quartile 4) group compared with the Q1 group. These findings were consistent with those from the external validation cohort, reinforcing the robustness of the causal relationship between the TyG index and NAFLD incidence. CONCLUSIONS The advanced double-robust estimation method suggests that a higher baseline TyG index may be causally associated with an increased NAFLD risk, providing more reliable evidence for its role as a simple biomarker and demonstrating the utility of double-robust estimation causal inference models in epidemiology.
Collapse
Affiliation(s)
- Tengrui Cao
- School of Public Health, Capital Medical University, NO. 10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China.
| | - Xuetong Ni
- School of Public Health, Capital Medical University, NO. 10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China.
| | - Aheyeerke Halengbieke
- School of Public Health, Capital Medical University, NO. 10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China.
| | - Jianmin Tang
- School of Public Health, Capital Medical University, NO. 10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China.
| | - Yumei Han
- Science and Education Section, Beijing Physical Examination Center, No. 59, Beiwei Road, Xicheng District, Beijing 100050, China.
| | - Feng Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China.
| | - Bo Gao
- School of Public Health, Capital Medical University, NO. 10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China.
| | - Deqiang Zheng
- School of Public Health, Capital Medical University, NO. 10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China.
| | - Yuxiang Yan
- School of Public Health, Capital Medical University, NO. 10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China.
| | - Xinghua Yang
- School of Public Health, Capital Medical University, NO. 10 Xitoutiao, Youanmenwai, Fengtai District, Beijing 100069, China.
| |
Collapse
|
113
|
Kushiro K, Hirono H, Ohkoshi S. Platelet-activating cytokines potentially associated with MASLD-induced liver injury significantly decreased following CPAP therapy: A translational study using a fatty liver mouse model. Sleep Med 2025; 130:15-24. [PMID: 40112616 DOI: 10.1016/j.sleep.2025.03.011] [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: 11/20/2024] [Revised: 02/10/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND AND AIM Patients with obstructive sleep apnea (OSA) and metabolic dysfunction associated steatotic liver disease (MASLD) frequently overlap due to the high prevalence of obesity. This translational study aimed to identify cytokines linking these conditions, beginning with an analysis of fatty liver in mice. Serum cytokine levels upregulated in the fatty liver mice were subsequently examined in human OSA serum samples. METHODS Mice were fed a high-fat diet to induce fatty liver. Liver proteins were analyzed using cytokine arrays. Serum samples from seventy (70) OSA patients (with 20 non-MASLD and 50 MASLD, pre- and 6-month post-continuous positive airway pressure [CPAP] therapy) were analyzed for the cytokines identified in the mouse experiment using enzyme-linked immunosorbent assays. RESULTS Four platelet-activation chemokines/cytokines (CCL5/RANTES, P-selectin, CXCL4/PF4, and CXCL5/LIX) were upregulated in mice with fatty liver. While serum levels of these factors were not significantly higher in MASLD-OSA compared to non-MASLD-OSA patients, their levels significantly decreased 6 months after the initiation of CPAP therapy, along with a reduction in mean platelet volume. CPAP compliance was significantly associated with a reduction in CCL5 levels. Additionally, a decrease in ALT levels following 6 months of CPAP therapy was significantly associated with CPAP compliance in MASLD-OSA patients. CONCLUSIONS While platelet-activation cytokines were not directly implicated in liver injury in MASLD-OSA patients, they decreased with CPAP therapy. CPAP compliance may play a key role in ALT reduction in MASLD-OSA patients independently of body weight changes. CCL5/RANTES may be indirectly associated with liver injury in MASLD-OSA, potentially induced through intermittent hypoxia.
Collapse
Affiliation(s)
- Kosuke Kushiro
- Clinical Examination, Graduate School of Life Dentistry at Niigata, The Nippon Dental University, Niigata, Japan
| | - Haruka Hirono
- Clinical Examination, Graduate School of Life Dentistry at Niigata, The Nippon Dental University, Niigata, Japan
| | - Shogo Ohkoshi
- Clinical Examination, Graduate School of Life Dentistry at Niigata, The Nippon Dental University, Niigata, Japan.
| |
Collapse
|
114
|
Buttler L, Stange J, Pyrsopoulos N, Hassanein T, Wedemeyer H, Maasoumy B, Busch M. Bacterial Infections in Patients With Severe Alcohol-Associated Hepatitis: Drivers of Organ Failure and Mortality. Liver Int 2025; 45:e70111. [PMID: 40332100 PMCID: PMC12057653 DOI: 10.1111/liv.70111] [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: 02/04/2025] [Revised: 04/03/2025] [Accepted: 04/17/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND Severe alcohol-associated hepatitis (sAH) is a life-threatening condition with limited treatment options. Although corticosteroids offer some benefit in short-term survival, their use remains controversial due to concerns about increased infection risk. Infections are a major cause of mortality in sAH; however, the reasons for this remain unclear. METHODS A post hoc analysis of the prospective VTL-308 multicentre trial on 151 patients with sAH was performed. Competing-risk models evaluated predictors of infections, the influence of corticosteroids on infection risk, and the impact of infections on the clinical outcomes up to 1 year of follow-up. RESULTS Among 151 patients, 90 (59.6%) developed infections. The most frequent were urinary tract (34.4%) and bloodstream (30%) infections. The causative pathogen was isolated in 40 patients, with Enterococcus spp. being the most common (35%). Fungal infections were detected in 19 (12.6%) patients. Corticosteroid use was not associated with increased bacterial (subdistribution-hazard ratio [sHR] =0.74; 95% Confidence Interval (CI): 0.42-1.33; p = 0.32) or fungal infection risk (sHR = 1.74; 95% CI: 0.59-5.15; p = 0.31). Infections significantly increased multi-organ failure (MOF) in the univariate (sHR = 2.31; 95% CI: 1.03-5.17; p = 0.04) and multivariate models (sHR = 2.46; 95% CI: 1.12-5.39; p = 0.03). 37.8% of infected patients died versus 13.1% of non-infected patients. Bacterial infections strongly predicted mortality, with sHRs ranging from 5.22 to 7.78, indicating a five- to eight-fold increased risk of death (p < 0.001). CONCLUSIONS Infections in sAH are central drivers of MOF and mortality. Our findings highlight infections as an independent risk factor unaffected by corticosteroid use, addressing previous concerns about the safety of this treatment.
Collapse
Affiliation(s)
- Laura Buttler
- Department of Gastroenterology, Hepatology, Infectious Diseases and EndocrinologyHannover Medical SchoolHannoverGermany
| | - Jan Stange
- Center for Extracorporeal Organ Support (CEOS), Biomedical Research Center, Department of NephrologyUniversity of RostockRostockGermany
| | - Nikolaos Pyrsopoulos
- Liver Disease in New JerseyNYU Grossman School of Medicine, NYU Langone Transplant InstituteNew YorkUSA
| | | | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology, Infectious Diseases and EndocrinologyHannover Medical SchoolHannoverGermany
- German Center for Infection Research (DZIF)Hannover‐BraunschweigGermany
| | - Benjamin Maasoumy
- Department of Gastroenterology, Hepatology, Infectious Diseases and EndocrinologyHannover Medical SchoolHannoverGermany
- German Center for Infection Research (DZIF)Hannover‐BraunschweigGermany
| | - Markus Busch
- Department of Gastroenterology, Hepatology, Infectious Diseases and EndocrinologyHannover Medical SchoolHannoverGermany
| | | |
Collapse
|
115
|
Sheng L, Chen Y, Wei H, Che F, Wu Y, Qin Q, Yang C, Wang Y, Peng J, Bashir MR, Ronot M, Song B, Jiang H. Large Language Models for Diagnosing Focal Liver Lesions From CT/MRI Reports: A Comparative Study With Radiologists. Liver Int 2025; 45:e70115. [PMID: 40347005 DOI: 10.1111/liv.70115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/29/2025] [Accepted: 04/19/2025] [Indexed: 05/12/2025]
Abstract
BACKGROUND & AIMS Whether large language models (LLMs) could be integrated into the diagnostic workflow of focal liver lesions (FLLs) remains unclear. We aimed to investigate two generic LLMs (ChatGPT-4o and Gemini) regarding their diagnostic accuracies referring to the CT/MRI reports, compared to and combined with radiologists of different experience levels. METHODS From April 2022 to April 2024, this single-center retrospective study included consecutive adult patients who underwent contrast-enhanced CT/MRI for single FLL and subsequent histopathologic examination. The LLMs were prompted by clinical information and the "findings" section of radiology reports three times to provide differential diagnoses in the descending order of likelihood, with the first considered the final diagnosis. In the research setting, six radiologists (three junior and three middle-level) independently reviewed the CT/MRI images and clinical information in two rounds (first alone, then with LLM assistance). In the clinical setting, diagnoses were retrieved from the "impressions" section of radiology reports. Diagnostic accuracy was investigated against histopathology. RESULTS 228 patients (median age, 59 years; 155 males) with 228 FLLs (median size, 3.6 cm) were included. Regarding the final diagnosis, the accuracy of two-step ChatGPT-4o (78.9%) was higher than single-step ChatGPT-4o (68.0%, p < 0.001) and single-step Gemini (73.2%, p = 0.004), similar to real-world radiology reports (80.0%, p = 0.34) and junior radiologists (78.9%-82.0%; p-values, 0.21 to > 0.99), but lower than middle-level radiologists (84.6%-85.5%; p-values, 0.001 to 0.02). No incremental diagnostic value of ChatGPT-4o was observed for any radiologist (p-values, 0.63 to > 0.99). CONCLUSION Two-step ChatGPT-4o showed matching accuracies to real-world radiology reports and junior radiologists for diagnosing FLLs but was less accurate than middle-level radiologists and demonstrated little incremental diagnostic value.
Collapse
Affiliation(s)
- Liuji Sheng
- Department of Radiology and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yidi Chen
- Department of Radiology and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Hong Wei
- Department of Radiology and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Che
- Department of Radiology and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yingyi Wu
- Department of Radiology and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qin Qin
- Department of Radiology and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chongtu Yang
- Department of Radiology and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanshu Wang
- Department of Radiology and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jingwen Peng
- College of Nursing, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Mustafa R Bashir
- Division of Gastroenterology, Department of Radiology, Center for Advanced Magnetic Resonance in Medicine, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Maxime Ronot
- Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP. Nord, Clichy, France
| | - Bin Song
- Department of Radiology and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Radiology, Sanya People's Hospital, Sanya, Hainan, China
| | - Hanyu Jiang
- Department of Radiology and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
116
|
Geng Y, Chen Z, Luo T, Liu Y, Kong S, Yan X, Bai H, Wang Y. Innovative construction and application of bile duct organoids: Unraveling the complexity of bile duct diseases and potential therapeutic strategies. Cancer Lett 2025; 618:217619. [PMID: 40074068 DOI: 10.1016/j.canlet.2025.217619] [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: 01/13/2025] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
The biliary system is crucial for liver function, regulating bile production, secretion, and transport. Dysfunctions within this system can lead to various diseases, such as cholangiopathies and biliary fibrosis, which may progress from benign to malignant states like cholangiocarcinoma. While liver organoid research is well-established and technologically advanced, bile duct organoids (BDOs) offer significant potential. BDOs can accurately simulate the physiological structure and function of bile ducts, making them valuable tools for in-vitro biliary disease research. Here, we review the development of BDO models, focusing on stem cell-derived organoids and tissue-derived organoids. We also illustrate the role of cultivation strategies and extracellular scaffolds in supporting organoid growth and stability, including the influence of cellular components of the microenvironment and physicochemical factors. Furthermore, we discuss the applications of BDOs in biliary development, disease modeling, regenerative medicine, and drug screening. Additionally, we emphasize the transformative potential in BDO biobanks and personalized medicine, which helps to pave the way for innovative therapeutic strategies and personalized medicine. Finally, we summarize the current and prospective advancements in BDO technologies, highlighting the integration of emerging technologies such as artificial intelligence, 3D bioprinting, and organoid-on-chip systems. These technologies hold great promise for significantly enhancing both clinical and research applications in the field of biliary diseases.
Collapse
Affiliation(s)
- Yadi Geng
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China; School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
| | - Ziye Chen
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
| | - Tianzi Luo
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 102218, China
| | - Yakun Liu
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 102218, China
| | - Siming Kong
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 102218, China
| | - Xinlong Yan
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China.
| | - Hui Bai
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 102218, China.
| | - Yunfang Wang
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China; Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 102218, China; Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
117
|
Yao F, Liu C, Luo D, Zhou Y, Li Q, Huang H, Xu H. Metabolites of Microbiota: A Novel Therapy for Heart Disease. FOOD REVIEWS INTERNATIONAL 2025; 41:1099-1115. [DOI: 10.1080/87559129.2024.2437410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Fei Yao
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Medical University
| | | | - Duo Luo
- Guangzhou Medical University
| | | | | | | | | |
Collapse
|
118
|
Chen HF, Chang YY, Chen P, Shen XH, Chang CH, Hsu WL. Risks of liver cirrhosis, hepatocellular carcinoma, hepatic-related complications, and mortality in patients with type 2 diabetes in Taiwan. World J Diabetes 2025; 16:104576. [DOI: 10.4239/wjd.v16.i5.104576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/25/2025] [Accepted: 03/21/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Hepatitis B and C and alcoholic liver disease are the principal causes of hepatic-related morbidity and mortality. However, evidence of the associations between diabetes without the above risk factors and hepatic-related study endpoints is not well understood. In addition, the effects of associated metabolic dysfunction and exercise on hepatic outcomes are still not clear.
AIM To investigate the incidence and relative hazards of cirrhosis of the liver, hepatocellular carcinoma (HCC), hepatic-related complications and mortality in patients with type 2 diabetes (T2D) who were nonalcoholic and serologically negative for hepatitis B and C in Taiwan.
METHODS A total of 33184 T2D patients and 648746 nondiabetic subjects selected from Taiwan’s adult preventive health care service were linked to various National Health Insurance databases, cancer registry, and death registry to identify cirrhosis of the liver, HCC, hepatic-related complications, and mortality. The Poisson assumption and Cox proportional hazard regression model were used to estimate the incidences and relative hazards of all hepatic-related study endpoints, respectively. We also compared the risk of hepatic outcomes stratified by age, sex, associated metabolic dysfunctions, and regular exercise between T2D patients and nondiabetic subjects.
RESULTS Compared with nondiabetic subjects, T2D patients had a significantly greater incidence (6.32 vs 17.20 per 10000 person-years) and greater risk of cirrhosis of the liver [adjusted hazard ratio (aHR) 1.45; 95%CI: 1.30-1.62]. The aHRs for HCC, hepatic complications, and mortality were 1.81, 1.87, and 2.08, respectively. An older age, male sex, obesity, hypertension, and dyslipidemia further increased the risks of all hepatic-related study endpoints, and regular exercise decreased the risk, irrespective of diabetes status.
CONCLUSION Patients with T2D are at increased risk of cirrhosis of the liver, HCC, hepatic-related complications, and mortality, and associated metabolic dysfunctions provide additional hazard. Coordinated interprofessional care for high-risk T2D patients and diabetes education, with an emphasis on the importance of physical activity, are crucial for minimizing hepatic outcomes.
Collapse
Affiliation(s)
- Hua-Fen Chen
- Department of Endocrinology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
- Department of Public Health, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Yung-Yueh Chang
- Department of Endocrinology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei City 100, Taiwan
| | - Peter Chen
- Department of Gastroenterology, Choninn Hospital, Choninn Medical Group, New Taipei City 220, Taiwan
| | - Xiao-Han Shen
- Department of Endocrinology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
- Master Program of Big Data in Medical Healthcare Industry, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
- Data Science Center, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Chin-Huan Chang
- Department of Endocrinology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
| | - Wan-Lun Hsu
- Master Program of Big Data in Medical Healthcare Industry, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
- Data Science Center, Fu Jen Catholic University, New Taipei City 242, Taiwan
| |
Collapse
|
119
|
Ye Z, Yan B, Li H, Tang Q, Yuan K, Hou J, Xu L, Yuan J, Wang S, Jiao W, Fan H, Lyu Y, Wang B, Liu X. Dual-responsive magnetic vortex nanorings co-deliver lenvatinib and localized heat for synergistic activation of antitumor immunity. Acta Biomater 2025; 198:389-400. [PMID: 40204172 DOI: 10.1016/j.actbio.2025.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 04/11/2025]
Abstract
Hepatocellular carcinoma (HCC) presents significant treatment challenges, primarily due to its ability to suppress immune responses. Lenvatinib (LT), approved as a first-line therapy for HCC, modulates the immune microenvironment by reducing PD-L1 expression and decreasing the infiltration of regulatory T cells (Tregs) within the tumor. However, the low immunogenicity of HCC and high toxicity of LT often undermine its effectiveness. To address these challenges, polydopamine (PDA)-coated ferrimagnetic vortex-domain iron oxide nanorings (FVIO@PDA) were engineered to respond to both acidic conditions and magnetic fields, facilitating the simultaneous delivery of the drug (LT) and a physio-therapeutic heat modality. The dual-responsive nature of FVIO@PDA ensures a controlled and synergistic release of LT, activated by acidic tumor microenvironments and the heat produced by an alternating magnetic field (AMF). In a subcutaneous Hepa1-6 HCC model, LT-loaded FVIO@PDA-PEG (denoted as LT-loaded FPP)-mediated magnetic hyperthermia significantly increased the levels of cytotoxic T lymphocytes, showing an approximate 3.86-fold increase compared to the control groups. This combination of LT and magnetic hyperthermia also reduced Treg populations to 1.4 %, synergistically triggering a robust antitumor immune response. Additionally, it altered cytokine profiles, reducing the secretion of the immunosuppressive cytokine IL-10 to 0.41 times that of control levels, while increasing the secretion of pro-inflammatory cytokines IFN-γ and TNF-α by 3.25 and 4.34 times, respectively. Furthermore, LT-loaded FPP-mediated magnetic hyperthermia exhibits superior anti-tumor activity compared to either treatment alone. These results highlight the promise of combining LT with FPP-mediated immunogenic magnetic hyperthermia as a potent therapeutic strategy for HCC, offering a more effective approach to modulate the immune environment and enhance antitumor efficacy. STATEMENT OF SIGNIFICANCE: Lenvatinib (LT) is a selective multi-targeted tyrosine kinase inhibitor used for patients with unresectable HCC who have not previously undergone systemic therapy. LT's immunomodulatory effects alone are often insufficient to induce an effective immune response, and treatment outcomes continue to be unsatisfactory. We developed FVIO@PDA for the delivery of LT and localized heat. FVIO@PDA allowed for controlled release of LT, triggered by the acidic tumor microenvironment and the heat generated under an AMF. LT combined with magnetic hyperthermia increased CTLs, reduced Tregs, decreased immunosuppressive cytokines, and elevated pro-inflammatory ones, collectively initiating a strong antitumor immune response. LT combined with magnetic hyperthermia showed superior antitumor effect compared to either treatment alone.
Collapse
Affiliation(s)
- Zirui Ye
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Province Center for Regenerative Medicine and Surgery Engineering Research, Shaanxi Provincial Key Laboratory of Magnetic Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Bin Yan
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Province Center for Regenerative Medicine and Surgery Engineering Research, Shaanxi Provincial Key Laboratory of Magnetic Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Hugang Li
- School of Future Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Qianqian Tang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Province Center for Regenerative Medicine and Surgery Engineering Research, Shaanxi Provincial Key Laboratory of Magnetic Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Kexin Yuan
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Province Center for Regenerative Medicine and Surgery Engineering Research, Shaanxi Provincial Key Laboratory of Magnetic Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Jingjing Hou
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Province Center for Regenerative Medicine and Surgery Engineering Research, Shaanxi Provincial Key Laboratory of Magnetic Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Lexuan Xu
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Province Center for Regenerative Medicine and Surgery Engineering Research, Shaanxi Provincial Key Laboratory of Magnetic Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Jianlan Yuan
- Center for Nanomedicine and Engineering, Northwest University, Xi'an, 710127, China
| | - Siyao Wang
- Center for Nanomedicine and Engineering, Northwest University, Xi'an, 710127, China
| | - Wangbo Jiao
- Center for Nanomedicine and Engineering, Northwest University, Xi'an, 710127, China
| | - Haiming Fan
- Center for Nanomedicine and Engineering, Northwest University, Xi'an, 710127, China
| | - Yi Lyu
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Province Center for Regenerative Medicine and Surgery Engineering Research, Shaanxi Provincial Key Laboratory of Magnetic Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Bo Wang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Province Center for Regenerative Medicine and Surgery Engineering Research, Shaanxi Provincial Key Laboratory of Magnetic Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
| | - Xiaoli Liu
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Province Center for Regenerative Medicine and Surgery Engineering Research, Shaanxi Provincial Key Laboratory of Magnetic Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; School of Future Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
| |
Collapse
|
120
|
Mittal G, A P, Dhali A, Prasad R, S Y, Nurani KM, Găman MA. Plant extracts with antioxidant and hepatoprotective benefits for liver health: A bibliometric analysis of drug delivery systems. World J Gastroenterol 2025; 31:105836. [DOI: 10.3748/wjg.v31.i18.105836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/07/2025] [Accepted: 04/21/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND The rising global burden of liver diseases, such as non-alcoholic fatty liver disease and liver fibrosis, has necessitated innovative therapeutic approaches. Plant-based therapies, recognized for their anti-inflammatory and antioxidant properties, have shown promising effects. However, poor bioavailability limits their clinical application.
AIM To map global research trends, key contributors, and emerging themes in plant-based therapies combined with advanced drug delivery systems for liver health.
METHODS Using the Scopus database, 645 documents were retrieved and analyzed using bibliometric tools Biblioshiny and VOSviewer. Analysis focused on publication trends, geographical contributions, and advancements in drug delivery technologies, including nanoparticles, liposomes, and polymeric micelles. Metrics such as publication growth rate, authorship collaboration, and thematic clustering were assessed.
RESULTS The dataset spans 43 years (1981-2024), with an annual growth rate of 11.09% in the number of publications. Research output is dominated by China (33%), followed by the United States (24%) and India (18%). Collaborative studies accounted for 24.34% of publications, with an average of 5.81 co-authors per document. Key innovations include nanoparticle encapsulation of curcumin and silymarin, improving bioavailability by up to 85%. Highly cited studies demonstrated the antioxidant, anti-inflammatory, and anti-fibrotic properties of these compounds. For instance, curcumin nanoparticles showed a 70% improvement in solubility, and silymarin liposomal formulations enhanced therapeutic efficiency by 62%. Thematic analysis revealed a transition from basic clinical observations to molecular and pharmacokinetic research, with a focus on oxidative stress mitigation and hepatoprotection.
CONCLUSION This study highlights the growing synergy between plant-based therapies and advanced drug delivery systems, with significant contributions from Asian and Western countries. Future efforts should prioritize clinical trials, standardization of plant extract formulations, and interdisciplinary approaches to maximize therapeutic outcomes. The findings provide a foundation for integrating plant-derived compounds into evidence-based hepatological therapies, addressing critical challenges in bioavailability and safety.
Collapse
Affiliation(s)
- Gaurav Mittal
- MBBS Final Year StudentMahatma Gandhi Institute of Medical Sciences, Maharashtra 442102, India
| | - Prashanth A
- Department of Physiology, Mahatma Gandhi Institute of Medical Sciences, Maharashtra 442102, India
| | - Arkadeep Dhali
- Academic Unit of Gastroenterology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield S5 7AU, United Kingdom
| | - Roshan Prasad
- Department of Medicine, Datta Meghe Institute of Higher Education and Research, Sawangi 442107, India
| | - Yogesh S
- Department of Medicine, Madras Medical College, Chennai 600003, India
| | | | - Mihnea-Alexandru Găman
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, Bucharest 050474, Romania
- Department of Hematology, Centre of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, Bucharest 022328, Romania
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, Bucharest 010001, Romania
| |
Collapse
|
121
|
Patel J, Roy H, Chintamaneni PK, Patel R, Bohara R. Advanced Strategies in Enhancing the Hepatoprotective Efficacy of Natural Products: Integrating Nanotechnology, Genomics, and Mechanistic Insights. ACS Biomater Sci Eng 2025; 11:2528-2549. [PMID: 40211874 DOI: 10.1021/acsbiomaterials.5c00004] [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] [Indexed: 05/13/2025]
Abstract
Liver disorders like hepatitis, cirrhosis, and hepatocellular carcinoma present a significant global health challenge, with high morbidity and mortality rates. Key factors contributing to liver disorders include inflammation, oxidative stress, and apoptosis. Due to their multifaceted action, natural compounds are promising candidates for mitigating liver-related disorders. Research studies revealed the antioxidant, anti-inflammatory, and detoxifying properties of natural compounds like curcumin, glycyrrhizin, and silymarin and their potential for liver detoxification and protection. With advancements in nanotechnology in drug delivery, natural compounds have improved stability and targetability, thereby enhancing their bioavailability and therapeutic efficiency. Further, recent advancements in genomics and an increased understanding of genetic factors influencing liver disorders and the hepatoprotective effects of natural agents made way for personalized medicine. Moreover, combinatorial therapy with natural products, synthetic drugs, or other natural agents has improved therapeutic outcomes. Even though clinical trials have confirmed the efficiency of natural compounds as hepatoprotective agents, several challenges remain unanswered in their translation to clinical practice. Therefore, it is logical to integrate natural compounds with nanotechnology and genomics to further advance hepatoprotection. This review gives an overview of the substantial progress made in the field of hepatoprotection, with specific emphasis on natural compounds and their integration with nanotechnology and genomics. This provides valuable insights for future research and innovations in developing therapeutic strategies for liver disorders.
Collapse
Affiliation(s)
- Jitendra Patel
- Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education (Deemed to be University), Sawangi (Meghe), Wardha 442001, Maharashtra, India
| | - Harekrishna Roy
- Department of Pharmaceutics, Nirmala College of Pharmacy, Mangalagiri 522503, Andhra Pradesh, India
| | - Pavan Kuma Chintamaneni
- Department of Pharmaceutics, GITAM School of Pharmacy, GITAM Deemed to be University, Hyerabad 502329, Telangana, India
| | - Rukmani Patel
- Department of Chemistry, Bharati University Durg, Durg 491001, Chhattisgarh, India
| | | |
Collapse
|
122
|
Girisa S, Aswani BS, Manickasamy MK, Hegde M, Alqahtani MS, Abbas M, Sethi G, Kunnumakkara AB. Restoring FXR expression as a novel treatment strategy in liver cancer and other liver disorders. Expert Opin Ther Targets 2025:1-29. [PMID: 40169227 DOI: 10.1080/14728222.2025.2487465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 03/28/2025] [Indexed: 04/03/2025]
Abstract
INTRODUCTION Liver cancer is a leading cause of cancer-associated mortality and is often linked to preexisting liver conditions. Emerging research demonstrates FXR dysregulation, particularly its reduced expression, in the pathogenesis of liver diseases, including inflammation, fibrosis, cholestatic disorders, metabolic dysregulation, and liver cancer. Therefore, this review explores the role of FXR and its agonists in mitigating these conditions. AREAS COVERED This article summarizes FXR's involvement in liver disorders, primarily emphasizing on hepatic neoplasms, and examines the potential of FXR agonists in restoring FXR activity in liver diseases, thereby preventing their progression to liver cancer. The information presented is drawn from existing preclinical and clinical studies specific to each liver disorder, sourced from PubMed. EXPERT OPINION It is well established that FXR expression is downregulated in liver disorders, contributing to disease progression. Notably, FXR agonists have demonstrated therapeutic potential in ameliorating liver diseases, including hepatocellular carcinoma. We believe that activating or restoring FXR expression with agonists offers significant promise for the treatment of liver cancer and other liver conditions. Therefore, FXR modulation by agonists, particularly in combination with other therapeutic agents, could lead to more targeted treatments, improving efficacy while reducing side effects.
Collapse
Affiliation(s)
- Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, India
| | - Babu Santha Aswani
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, India
| | - Mukesh Kumar Manickasamy
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, India
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
- BioImaging Unit, Space Research Centre, University of Leicester, Leicester, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, Saudi Arabia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, India
| |
Collapse
|
123
|
Jaiswal J, Srivastav AK, Kushwaha M, Teotia A, Singh R, Mohan A, Makharia G, Kumar A. Gut Microbial Metabolite 4-Ethylphenylsulfate Is Selectively Deleterious and Anticancer to Colon Cancer Cells. J Med Chem 2025. [PMID: 40347180 DOI: 10.1021/acs.jmedchem.5c00609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2025]
Abstract
Gut microbiota-derived metabolites have emerged as promising candidates in cancer therapeutics. Among these metabolites, 4-ethylphenyl sulfate (4-EPS), produced through dietary metabolism, is linked to chronic diseases but remains unexplored as a therapeutic agent for colorectal cancer (CRC) treatment. This study investigates the selective anticancer activity of 4-EPS using HCT-116 human colorectal adenocarcinoma cells and CCD 841 normal colon epithelial cells. Treatment with 4-EPS significantly reduced cell proliferation, viability, ATP levels, and colony-forming ability while increased apoptosis rate. Morphological changes included cell shrinkage, intracellular vesicle formation, and loss of membrane integrity. Mechanistically, 4-EPS upregulated Bax, downregulated Bcl2, and induced G2/M phase cell cycle arrest. In silico investigations revealed strong interactions with HDAC isoforms, suggesting epigenetic modulation. Markedly, 4-EPS treatment showed no deleterious effect on CCD 841 normal colon epithelial cells, which proved its selective anticancer role for colon cancer cells. These findings highlight 4-EPS as a promising therapeutic agent for treating CRC.
Collapse
Affiliation(s)
- Jyoti Jaiswal
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Amit Kumar Srivastav
- Department of Forensic Science, UP State Institute of Forensic Sciences, Lucknow 226401, Uttar Pradesh, India
| | - Manish Kushwaha
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi 110067, India
- School of Life Science, Department of Biotechnology, Mahatma Gandhi Central University, Motihari 845401, Bihar, India
| | - Anupam Teotia
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi 110067, India
- Department of Environmental Science, Jamia Millia Islamia (Central University), New Delhi 110025, India
| | - Rajeev Singh
- Department of Environmental Science, Jamia Millia Islamia (Central University), New Delhi 110025, India
| | - Anand Mohan
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, India
| | - Govind Makharia
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi 110016, India
| | - Anil Kumar
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi 110067, India
| |
Collapse
|
124
|
Tan HY, Liu SQ, Liu YH, Dai GH, Zheng JL, Feng HG. Radiotherapy, tyrosine kinase inhibitors, immune checkpoint inhibitors combined with hepatic arterial infusion chemotherapy of RALOX versus FOLFOX for hepatocellular carcinoma with portal vein tumor thrombus: a propensity score-matching cohort study. Discov Oncol 2025; 16:717. [PMID: 40347357 DOI: 10.1007/s12672-025-02553-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 05/02/2025] [Indexed: 05/12/2025] Open
Abstract
BACKGROUND This retrospective study aimed to evaluate the safety and effectiveness of hepatic arterial infusion chemotherapy with raltitrexed and oxaliplatin (RALOX-HAIC) combined with radiotherapy, tyrosine kinase inhibitors, and immune checkpoint inhibitors in patients with hepatocellular carcinoma (HCC) with portal vein tumor thrombus (PVTT). METHODS A propensity score-matching (PSM) cohort study was conducted. The tumor response, treatment-related adverse events, survival outcomes were compared. Univariate and multivariate analyses were conducted to explore the risk factors of overall survival (OS). RESULTS Twenty-one pairs of patients were evaluated after PSM. No statistical differences were found in the tumor response, objective response rate, disease control rate, PVTT response, surgical resection rate, metastasis, and mortality between the two groups before and after PSM. Compared with the FOLFOX-HAIC group, the incidences of abdominal pain and fever was lower in the RALOX-HAIC group (P = 0.028, P = 0.029). These differences still had statistical significance after PSM (P = 0.013, P = 0.014). No statistical differences were found in OS and progression-free survival (PFS) between the two groups before and after PSM (Before [OS: hazard ratio(HR) = 1.138; 95%CI 0.569-2.276, P = 0.715; PFS: HR = 0.549; 95%CI 0.195-1.548, P = 0.257; After [OS: HR = 0.998; 95%CI 0.438-2.274, P = 0.995; PFS: HR = 0.792; 95%CI 0.359-1.748, P = 0.564]). The prealbumin < 170 mg/L before therapy was an independent risk factor for OS (HR = 2.234; 95%CI 1.051-4.751; P = 0.037). CONCLUSIONS The RALOX-HAIC combined radiotherapy, TKI, and ICI may provide similar survival advantages with fewer treatment-related abdominal pain and fever compared to FOLFOX-HAIC for HCC patients with PVTT. The prealbumin < 170 mg/L before therapy is an independent risk factor for OS.
Collapse
Affiliation(s)
- Hao-Yang Tan
- Department of Hepatobiliary Surgery, the Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Shuang-Quan Liu
- Department of Hepatobiliary Surgery, the Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Yan-Han Liu
- Department of Radiology, the Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Guo-Hua Dai
- Department of Hepatobiliary Surgery, the Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Jiu-Ling Zheng
- Department of Hepatobiliary Surgery, the Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, Chongqing, China.
| | - Hua-Guo Feng
- Department of Hepatobiliary Surgery, the Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, Chongqing, China.
| |
Collapse
|
125
|
Xu Q, Li X, Yuan Y, Hu Z, Zhang W, Wang Y, Shen A, Lei H. Development and validation of a nomogram model for predicting immune-mediated hepatitis in cancer patients treated with immune checkpoint inhibitors. Biosci Trends 2025; 19:202-210. [PMID: 39894525 DOI: 10.5582/bst.2024.01351] [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] [Indexed: 02/04/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have been widely used in various types of cancer, but they have also led to a significant number of adverse events, including ICI-induced immune-mediated hepatitis (IMH). This study aimed to explore the risk factors for IMH in patients treated with ICIs and to develop and validate a new nomogram model to predict the risk of IMH. Detailed information was collected between January 1, 2020, and December 31, 2023. Univariate logistic regression analysis was used to assess the impact of each clinical variable on the occurrence of IMH, followed by stepwise multivariate logistic regression analysis to determine independent risk factors for IMH. A nomogram model was constructed based on the results of the multivariate analysis. The performance of the nomogram model was evaluated via the area under the receiver operating characteristic curve (AUC), calibration curves, decision curve analysis (DCA), and clinical impact curve (CIC) analysis. A total of 216 (8.82%) patients developed IMH. According to stepwise multivariate logistic analysis, hepatic metastasis, the TNM stage, the WBC count, LYM, ALT, TBIL, ALB, GLB, and ADA were identified as risk factors for IMH. The AUC for the nomogram model was 0.817 in the training set and 0.737 in the validation set. The calibration curves, DCA results, and CIC results indicated that the nomogram model had good predictive accuracy and clinical utility. The nomogram model is intuitive and straightforward, making it highly suitable for rapid assessment of the risk of IMH in patients receiving ICI therapy in clinical practice. Implementing this model enables early adoption of preventive and therapeutic strategies, ultimately reducing the likelihood of immune-related adverse events (IRAEs), and especially IMH.
Collapse
Affiliation(s)
- Qianjie Xu
- Chongqing Cancer Multi-omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaosheng Li
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Yuliang Yuan
- Chongqing Cancer Multi-omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Zuhai Hu
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Wei Zhang
- Chongqing Cancer Multi-omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Ying Wang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Ai Shen
- Hepatobiliary Pancreatic Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Haike Lei
- Chongqing Cancer Multi-omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
126
|
Yang J, Yang Y, Tan X, Du H, Zhou Z, Chen L, Tian X, Zheng G, Hu J, Zhang C, Qiu Z. Unlocking the potential of the ACE2/Ang-(1-7)/Mas Axis in liver diseases: From molecular mechanisms to translational applications. Diabetes Obes Metab 2025. [PMID: 40344459 DOI: 10.1111/dom.16435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 04/11/2025] [Accepted: 04/21/2025] [Indexed: 05/11/2025]
Abstract
Over the past two decades, the identification of new functions within the renin-angiotensin system (RAS) has extended beyond its traditional roles, with the emergence of the angiotensin-converting enzyme 2 (ACE2)/Ang-(1-7)/Mas axis being particularly significant. This axis is hypothesized to balance or modulate the effects of the traditional ACE/Ang II/AT1 axis in various physiological and pathological contexts. ACE2, a membrane-bound carboxypeptidase and an ancient homologue of ACE converts Angiotensin II (Ang II) into Angiotensin 1-7 (Ang-(1-7)). The Mas receptor is a G-protein-coupled receptor that specifically binds Ang-(1-7). Recent research has increasingly focused on the local expression of RAS in different tissues. Ang-(1-7) produces a variety of biological effects by binding to the Mas receptor, including anti-inflammatory, antioxidant, anti-apoptotic and anti-fibrotic actions, thereby influencing a range of mechanisms in the heart, kidneys, brain and other tissues. Preclinical animal model studies indicate that manipulating the protective RAS can significantly alter the progression of multiple liver diseases. Hepatic overexpression of ACE2 or administration of Ang-(1-7) and its analogues has been shown to be therapeutically effective against drug-induced liver injury, metabolic-associated fatty liver disease, liver fibrosis and hepatocellular carcinoma progression. These effects are achieved through various pathways, including the regulation of lipid metabolism, inhibition of epithelial-mesenchymal transition (EMT) and extracellular matrix (ECM) production, as well as suppression of aerobic glycolysis. In current clinical trials, while recombinant human ACE2 (Rh-ACE2) has demonstrated safety and good tolerance in most studies, research on the relevance of activating the ACE2/Ang-(1-7) axis in the mechanisms and evolution of human diseases remains in its early stages. Therefore, further elucidation of the complex interactions between the classical and counter-regulatory RAS axes in clinical settings is crucial. This review will summarize the roles of selective activation of the ACE2/Ang-(1-7)/Mas axis, with a focus on its mechanisms in the treatment of liver diseases. Additionally, we will discuss the safety concerns regarding selective activation of the ACE2/Ang-(1-7)/Mas axis in clinical applications and the challenges of tissue-specific activation of this axis, providing effective therapeutic strategies for targeted activation of the hepatic ACE2/Ang-(1-7)/Mas axis in clinical practice.
Collapse
Affiliation(s)
- Jun Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Yuan Yang
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiangyun Tan
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Hongzhi Du
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Zhongshi Zhou
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Liang Chen
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Xianxiang Tian
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Guohua Zheng
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Junjie Hu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
| | - Cong Zhang
- College of Basic Medical Sciences, China Three Gorges University, Yichang, China
| | - Zhenpeng Qiu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
- Center of Traditional Chinese Medicine Modernization for Liver Diseases, Hubei University of Chinese Medicine, Wuhan, People's Republic of China
- Hubei Shizhen Laboratory, Wuhan, People's Republic of China
| |
Collapse
|
127
|
Verdan S, Torri GB, Marcos VN, Moreira MHS, Defante MLR, Fagundes MDC, de Barros EMJ, Dias AB, Shen L, Altmayer S. Ultrasound-derived fat fraction for diagnosing hepatic steatosis: a systematic review and meta-analysis. Eur Radiol 2025:10.1007/s00330-025-11652-8. [PMID: 40346257 DOI: 10.1007/s00330-025-11652-8] [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: 01/27/2025] [Revised: 03/06/2025] [Accepted: 04/05/2025] [Indexed: 05/11/2025]
Abstract
OBJECTIVE To perform a systematic review and meta-analysis to evaluate the diagnostic performance of Ultrasound-Derived Fat Fraction (UDFF) in detecting hepatic steatosis using Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF) as the reference standard. MATERIALS AND METHODS Relevant databases were searched through November 2024. Studies that evaluated the UDFF to detect hepatic steatosis using MRI-PDFF as the reference standard met the inclusion criteria. Our primary outcome was the sensitivity and specificity of UDFF compared to MRI-PDFF in distinguishing steatosis from non-steatosis. Analyses were performed using a bivariate random-effects approach, and heterogeneity was considered substantial if I2 > 50%. A sensitivity analysis was performed to detect potential studies that contribute to heterogeneity. RESULTS Nine studies comprising 1150 patients (mean age range, 14-62 years; 51.2% women) were included. Eight studies were performed using the same vendor platform. The pooled sensitivity of UDFF for detecting hepatic steatosis was 90.4% (95% CI: 84.0%, 94.4%), and the pooled specificity was 83.8% (95% CI: 75.1%, 89.8%). The AUC for the summary receiver-operating characteristic curve was 0.93 (95% CI: 0.83, 0.95). Heterogeneity among the studies was low (I² = 22.2%). CONCLUSION UDFF demonstrates high sensitivity and specificity for detecting hepatic steatosis, supporting its value as a noninvasive tool for screening. KEY POINTS Question Small individual studies suggest that US-Derived Fat Fraction (UDFF) may effectively detect hepatic steatosis compared to MRI, but no meta-analysis has been performed. Findings In nine studies including 1150 patients, UDFF demonstrated high pooled sensitivity (90.4%) and specificity (83.8%) relative to MRI with low between-study heterogeneity. Clinical relevance UDFF demonstrates high diagnostic accuracy compared with MRI, supporting its use as a noninvasive tool with potentially lower cost and wider availability for large-scale screening of hepatic steatosis.
Collapse
Affiliation(s)
- Sarah Verdan
- Department of Radiology and Diagnostic Imaging, University Hospital of Juiz de Fora - UFJF, Juiz de Fora, Brazil.
| | - Giovanni B Torri
- Department of Radiology and Diagnostic Imaging, Hospital Universitário de Santa Maria, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Vinícius Neves Marcos
- Department of Radiology and Diagnostic Imaging, University Hospital of Juiz de Fora - UFJF, Juiz de Fora, Brazil
| | - Maria Helena Silva Moreira
- Department of Radiology and Diagnostic Imaging, University Hospital of Juiz de Fora - UFJF, Juiz de Fora, Brazil
| | | | | | | | - Adriano B Dias
- University Medical Imaging Toronto, Joint Department of Medical Imaging, University Health Network-Sinai Health System-Women's College Hospital, University of Toronto, Toronto, ON, Canada
| | - Luyao Shen
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Stephan Altmayer
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
128
|
Brown GE, Bodke VV, Ware BR, Khetani SR. Liver portal fibroblasts induce the functions of primary human hepatocytes in vitro. Commun Biol 2025; 8:721. [PMID: 40346200 DOI: 10.1038/s42003-025-08135-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 04/25/2025] [Indexed: 05/11/2025] Open
Abstract
In vitro human liver models are critical to mitigate species-specific differences observed for toxicology, disease modeling, and regenerative medicine. Interactions with mesenchyme (i.e., fibroblasts) can promote phenotypic functions of primary human hepatocytes (PHHs) in culture; however, using liver-derived fibroblasts remains elusive. Portal fibroblasts (PFs) around the portal triad influence bile duct formation during development, but their role in regulating homeostatic hepatic functions remains unknown. Here, we show that human liver PFs induce long-term phenotypic functions in PHHs at higher levels than activated hepatic stellate cells across 2-dimensional and 3-dimensional culture formats. While PF-conditioned media induces some hepatic functions, partly via insulin-like growth factor binding protein-5 signaling, direct contact is necessary to induce optimal functional levels. Inhibiting Notch signaling reduces progenitor-like characteristics of PHHs and further enhances functionality. Overall, this work demonstrates a unique role for PFs in modulating hepatic functions and provides all-human and all-liver coculture strategies for downstream applications.
Collapse
Affiliation(s)
- Grace E Brown
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, USA
| | - Vedant V Bodke
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, USA
| | - Brenton R Ware
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, USA
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Salman R Khetani
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
129
|
Noureddin M, Rinella ME, Chalasani NP, Neff GW, Lucas KJ, Rodriguez ME, Rudraraju M, Patil R, Behling C, Burch M, Chan DC, Tillman EJ, Zari A, de Temple B, Shringarpure R, Jain M, Rolph T, Cheng A, Yale K. Efruxifermin in Compensated Liver Cirrhosis Caused by MASH. N Engl J Med 2025. [PMID: 40341827 DOI: 10.1056/nejmoa2502242] [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] [Indexed: 05/11/2025]
Abstract
BACKGROUND In phase 2 trials involving patients with stage 2 or 3 fibrosis caused by metabolic dysfunction-associated steatohepatitis (MASH), efruxifermin, a bivalent fibroblast growth factor 21 (FGF21) analogue, reduced fibrosis and resolved MASH. Data are needed on the efficacy and safety of efruxifermin in patients with compensated cirrhosis (stage 4 fibrosis) caused by MASH. METHODS In this phase 2b, randomized, placebo-controlled, double-blind trial, we assigned patients with MASH who had biopsy-confirmed compensated cirrhosis (stage 4 fibrosis) to receive subcutaneous efruxifermin (at a dose of 28 mg or 50 mg once daily) or placebo. The primary outcome was a reduction of at least one stage of fibrosis without worsening of MASH at week 36. Secondary outcomes included the same criterion at week 96. RESULTS A total of 181 patients underwent randomization and received at least one dose of efruxifermin or placebo. Of these patients, liver biopsy was performed in 154 patients at 36 weeks and in 134 patients at 96 weeks. At 36 weeks, a reduction in fibrosis without worsening of MASH occurred in 8 of 61 patients (13%) in the placebo group, in 10 of 57 patients (18%) in the 28-mg efruxifermin group (difference from placebo after adjustment for stratification factors, 3 percentage points; 95% confidence interval [CI], -11 to 17; P = 0.62), and in 12 of 63 patients (19%) in the 50-mg efruxifermin group (difference from placebo, 4 percentage points; 95% CI, -10 to 18; P = 0.52). At week 96, a reduction in fibrosis without worsening of MASH occurred in 7 of 61 patients (11%) in the placebo group, in 12 of 57 patients (21%) in the 28-mg efruxifermin group (difference from placebo, 10 percentage points; 95% CI, -4 to 24), and in 18 of 63 patients (29%) in the 50-mg efruxifermin group (difference from placebo, 16 percentage points; 95% CI, 2 to 30). Gastrointestinal adverse events were more common with efruxifermin; most events were mild or moderate. CONCLUSIONS In patients with compensated cirrhosis caused by MASH, efruxifermin did not significantly reduce fibrosis at 36 weeks. (Funded by Akero Therapeutics; SYMMETRY ClinicalTrials.gov number, NCT05039450.).
Collapse
Affiliation(s)
- Mazen Noureddin
- Houston Methodist Hospital, Houston
- Houston Research Institute, Houston
| | - Mary E Rinella
- University of Chicago Pritzker School of Medicine, Chicago
| | | | - Guy W Neff
- Covenant Metabolic Specialists, Fort Myers, FL
| | | | | | | | | | | | - Mark Burch
- Akero Therapeutics, South San Francisco, CA
| | | | | | - Arian Zari
- Akero Therapeutics, South San Francisco, CA
| | | | | | - Meena Jain
- Akero Therapeutics, South San Francisco, CA
| | | | | | - Kitty Yale
- Akero Therapeutics, South San Francisco, CA
| |
Collapse
|
130
|
Luo Y, Gao S, Chen J, Lv D, Mai W, Wang W, Zhu Y. Delivering integrated and standardized services to eliminate mother-to-child transmission of sexually transmitted infections: experience of the E-Clinic model in a Chinese pilot district. Health Policy Plan 2025; 40:578-584. [PMID: 40176734 PMCID: PMC12063579 DOI: 10.1093/heapol/czaf006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/21/2025] [Accepted: 04/02/2025] [Indexed: 04/04/2025] Open
Abstract
The elimination of mother-to-child transmission (EMTCT) of syphilis and hepatitis B virus (HBV) is a crucial strategy for preventing and reducing new pediatric infections. Timely and standardized EMTCT interventions for maternal infections have been demonstrated to be highly cost-effective and efficient. Regrettably, healthcare providers have encountered challenges in delivering these interventions due to the complexity of multidepartment referrals. Baoan district in Shenzhen, Guangdong Province, which serves as one of the six national pilot districts tasked with exploring effective EMTCT strategies, has pioneered the integration of infection management into antenatal care by establishing the E-Clinic (short for the Elimination of mother-to-child transmission Clinic) within the Obstetrics Department. The E-Clinic, through its provision of integrated services, has witnessed increased coverage of syphilis treatment for pregnant women living with syphilis, as well as increased coverage of antiviral treatment for high-risk pregnant women living with HBV. The E-Clinic has been recognized as a best practice for EMTCT by the World Health Organization and has been widely adopted and adapted in over 100 maternal and child healthcare institutions across Guangdong Province. The success of the E-Clinic underscores the importance of government leadership, innovation, and multilevel engagement in promoting standardized EMTCT services, providing insights for implementing EMTCT strategies in other regions worldwide.
Collapse
Affiliation(s)
- Yali Luo
- Department of Health Care, Shenzhen Baoan Women’s and Children’s Hospital, No. 56 Yulv Road, Xin’an Street, Guangdong 518101, China
| | - Shuang Gao
- Department of Maternal Health Care, Guangdong Women and Children’s Hospital and Health Institute, 521 Xingnan Avenue, Panyu District, Guangzhou 511400, China
| | - Jiahong Chen
- Department of Health Care, Shenzhen Baoan Women’s and Children’s Hospital, No. 56 Yulv Road, Xin’an Street, Guangdong 518101, China
| | - Dingyan Lv
- Department of Health Care, Shenzhen Baoan Women’s and Children’s Hospital, No. 56 Yulv Road, Xin’an Street, Guangdong 518101, China
| | - Weizhen Mai
- Department of Health Care, Shenzhen Baoan Women’s and Children’s Hospital, No. 56 Yulv Road, Xin’an Street, Guangdong 518101, China
| | - Wei Wang
- Department of Health Care, Shenzhen Baoan Women’s and Children’s Hospital, No. 56 Yulv Road, Xin’an Street, Guangdong 518101, China
| | - Yuanfang Zhu
- Maternal-Fetal Medicine Institute, Shenzhen Baoan Women’s and Children’s Hospital, No. 56 Yulv Road, Xin’an Street, Guangdong 518101, China
| |
Collapse
|
131
|
Li M, Zhang R, Huang X, Jian M, Qi H, Chen X, Deng J, Chen W, Feng C, Dong Q, Su B. Orthogonal DNA self-assembly technology enables rapid and accurate analysis of circulating tumor cells in breast cancer. Anal Chim Acta 2025; 1350:343871. [PMID: 40155170 DOI: 10.1016/j.aca.2025.343871] [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: 01/01/2025] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND As a non-invasive liquid biopsy technology, the detection of circulating tumor cells (CTCs) overcomes the limitations of traditional tissue biopsy methods, enabling continuous sample collection and long-term dynamic monitoring. However, current CTCs analysis methods typically rely on cell size to separate and identify tumor cells, which fails to effectively distinguish tumor cells from different sources. In addition, existing methods are often constrained by limited antibody species, typically detecting only 2-3 molecular phenotypes. This narrow detection scope does not fully capture the heterogeneity of CTCs at the single-cell level, thus limiting its utility in precision diagnosis and personalized treatment. To address these challenges, it is urgent to develop CTCs detection methods that can simultaneously integrate comprehensive target and cell morphology information. RESULTS Using breast cancer as a research model, we developed a computer-aided design-based hybridization chain reaction (CAD-HCR) by combining DNA encoding and antibody coupling technologies with orthogonal DNA self-assembly to achieve multiple detection and heterogeneity analysis of breast cancer mimic samples. This technology overcomes the limitation of antibody species in traditional CTCs detection and utilizes antibody-trigger strand coupling to convert target protein signals into DNA signals, thereby circumventing throughput limitation of existing detection methods. By utilizing the signal amplification effect of DNA self-assembly, this technology enhances sensitivity significantly, allowing for accurate single-cell level detection of CTCs. SIGNIFICANCE This technology provides spatial positioning and cell morphological characteristics information for CTCs analysis of breast cancer, which is expected to provide a more accurate basis for diagnosis and treatment decision-making for in-depth understanding of tumor heterogeneity and clinical applications.
Collapse
Affiliation(s)
- Min Li
- School of Medicine, Anhui University of Science & Technology, Huainan, 232001, China; Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Runchi Zhang
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Xiaozhi Huang
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Meng Jian
- School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Haipeng Qi
- School of Medicine, Anhui University of Science & Technology, Huainan, 232001, China; Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Xiaolong Chen
- Shanghai Chrona Life Science Technology Co., Ltd., Shanghai, 201306, China
| | - Jie Deng
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Wen Chen
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Chang Feng
- Shanghai Key Laboratory of Pathogenic Fungi Medical Testing, Shanghai Pudong New Area People's Hospital, Shanghai, 201299, China.
| | - Qiongzhu Dong
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission (SMHC), Minhang Hospital, Fudan University, Shanghai, 200040, China.
| | - Bin Su
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
132
|
Wang J, Wang H, Yang W, Zhao D, Liu D, Tang L, Chen XP. GPNMB regulates the differentiation and transformation of monocyte-derived macrophages during MASLD. Int Immunopharmacol 2025; 154:114554. [PMID: 40186908 DOI: 10.1016/j.intimp.2025.114554] [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: 12/03/2024] [Revised: 03/22/2025] [Accepted: 03/23/2025] [Indexed: 04/07/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is an increasingly concerning global health issue characterized by pronounced hepatic steatosis and liver fibrosis. Hepatic monocyte-derived macrophages (MDMs) are crucial in the pathogenesis of liver fibrosis under MASLD. Nevertheless, the precise functions of MDMs and the underlying mechanisms governing their differentiation remain inadequately elucidated. In this study, we revealed an orchestrator of this process: Glycoprotein Non-Metastatic Melanoma Protein B (GPNMB), one of the characteristic genes of MDMs. Notably, myeloid-specific Gpnmb-knockout contributed to the retention of resident Kupffer cells (KCs) and rerouted monocyte differentiation towards a monocyte-derived macrophage subset that occupies the Kupffer cell niche (MoKC subset, resembling resident KCs), thereby impeding the formation of hepatic lipid-associated macrophages (LAMs). This transition has a profound impact, manifested in significantly reduced steatosis and modestly decreased liver fibrosis in myeloid-specific Gpnmb-knockout mice. In conclusion, our research clarifies the complex interactions between Gpnmb and MDMs and underscores the therapeutic potential of targeting Gpnmb within MDMs to manage MASLD.
Collapse
Affiliation(s)
- Junqi Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China.; State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing. Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, PR China; Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha 410008, Hunan, PR China
| | - Huan Wang
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing. Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, PR China
| | - Wenting Yang
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing. Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, PR China
| | - Dianyuan Zhao
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing. Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, PR China
| | - Di Liu
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing. Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, PR China
| | - Li Tang
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing. Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, PR China; Institute of Future Agriculture, Northwest A&F University, Yangling 712100, Shaanxi, PR China..
| | - Xiao-Ping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China.; Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha 410008, Hunan, PR China.; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China.; Furong Laboratory, Changsha, Hunan, China.
| |
Collapse
|
133
|
Ling F, Feng H, Wu S, Zhu D, Chen Y, Zhou J, Lai J, Huang X, Hou T, Li Y. Role of m7G modification regulators as biomarkers in gastric cancer subtyping and precision immunotherapy. Int Immunopharmacol 2025; 154:114594. [PMID: 40194456 DOI: 10.1016/j.intimp.2025.114594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/17/2025] [Accepted: 03/28/2025] [Indexed: 04/09/2025]
Abstract
This study investigated the role of N7-methylguanosine (m7G) modification regulators as biomarkers in subtyping and precision immunotherapy of gastric cancer (GC). Through multi-omics analyses, including RNA sequencing, proteomics, and single-cell measurement, the study revealed heterogeneity in the m7G regulatory landscape among GC patients. Three m7G subtypes were identified, each with distinct pathways and phenotypes. Patients with low m7Gscores, based on an established scoring system, showed better survival outcomes and increased antitumor immune cell infiltration, as well as higher tumor mutation loads and lower PD-L1 expression. The predictive value of m7Gscore was confirmed in two immunotherapy cohorts. These findings highlight the potential of m7G modification in shaping the tumor microenvironment and provide new insights for immunotherapeutic strategies in GC patients.
Collapse
Affiliation(s)
- Fa Ling
- Department of gastrointestinal surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Huolun Feng
- Department of gastrointestinal surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China; School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Sifan Wu
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China; Guangdong Center for Clinical Laboratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Dandan Zhu
- Guangdong Center for Clinical Laboratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Yinfeng Chen
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China; MOE Joint International Research Laboratory of Pancreatic Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Jianlong Zhou
- Department of gastrointestinal surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Jiayi Lai
- School of Medical Information Engineering, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Xing Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China; MOE Joint International Research Laboratory of Pancreatic Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China.
| | - Tieying Hou
- Medical Experimental Center, Shenzhen Nanshan People's Hospital, Shenzhen, Guangdong, 518052, China; Shenzhen University Medical School, Shenzhen, Guangdong, 518073, China.
| | - Yong Li
- Department of gastrointestinal surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China; School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China; Department of Gastrointestinal Surgery, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, Jiangsu, 210000, China.
| |
Collapse
|
134
|
Flatley S, Dixon S, Pilsworth E, Dube A, Hoeroldt B, Harrison L, Gleeson D. Diabetes Mellitus in Patients With Autoimmune Hepatitis: Frequency, Risk Factors and Effect on Outcome. Aliment Pharmacol Ther 2025. [PMID: 40342076 DOI: 10.1111/apt.70188] [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: 08/04/2024] [Revised: 09/02/2024] [Accepted: 04/30/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND Treatment for autoimmune hepatitis (AIH) includes corticosteroids, which are associated with the development of diabetes mellitus (DM). Reported new-onset DM rates in patients with AIH have varied, and predisposing factors and prognostic implications are inadequately characterised. AIM To identify the frequency and predisposing factors for DM in AIH and its association with disease progression and mortality. METHODS Retrospective/prospective single-centre study of 494 patients with AIH presenting 1987-2023, 466 receiving corticosteroids (454 prednisolone, 12 budesonide) and followed for (median (range) 9 (0-36) years). RESULTS Forty-seven patients (10%) already had DM at AIH diagnosis. New-onset DM subsequently developed in another 59 (13%). In those receiving prednisolone, new-onset DM incidence was 8% ± 1% after 1 year and 14% ± 2% after 10 years (14- and 3-fold higher than expected population rate), and was independently associated with older age, non-Caucasian ethnicity, higher initial prednisolone dose, higher BMI at diagnosis and more weight gain after 2 years of follow-up. New-onset DM usually persisted despite stopping prednisolone. New-onset DM and DM at any time were independently associated with all-cause death/transplantation rate, along with previously established risk factors (older age, cirrhosis, lower ALT at diagnosis and failure of early ALT normalisation). New-onset DM and DM at any time were also independently associated with cirrhosis development. Similar associations of new-onset DM and DM at any time with liver-related death/transplantation were significant on univariate but not multivariate analysis. CONCLUSION New-onset DM occurred in 13% of patients with AIH, was related to older age, non-Caucasian ethnicity, higher prednisolone dose, higher BMI at diagnosis and weight gain; and was an independent predictor of all-cause death/transplantation and of cirrhosis development, underlining the need to minimise steroid burden in AIH.
Collapse
Affiliation(s)
- Sarah Flatley
- Liver Unit, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, University of Sheffield Medical School, Sheffield, UK
| | - Selena Dixon
- Liver Unit, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Eleanor Pilsworth
- Liver Unit, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Asha Dube
- Department of Histopathology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Barbara Hoeroldt
- Liver Unit, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Laura Harrison
- Liver Unit, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Dermot Gleeson
- Liver Unit, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, University of Sheffield Medical School, Sheffield, UK
| |
Collapse
|
135
|
Byun J, Han Y, Choi J, Sun R, Shaw VR, Zhu C, Xiao X, Lusk C, Badr H, Lee HS, Jang HJ, Li Y, Lim H, Long E, Liu Y, Kachuri L, Walsh KM, Wiencke JK, Albanes D, Lam S, Tardon A, Neuhouser ML, Barnett MJ, Chen C, Bojesen S, Brenner H, Landi MT, Johansson M, Risch A, Wichmann HE, Bickeböller H, Christiani DC, Rennert G, Arnold S, Field JK, Shete S, Le Marchand L, Liu G, Andrew AS, Zienolddiny S, Grankvist K, Johansson M, Caporaso N, Taylor F, Lazarus P, Schabath MB, Aldrich MC, Patel A, Lin X, Zanetti KA, Harris CC, Chanock S, McKay J, Schwartz AG, Hung RJ, Amos CI. Genome-wide association study for lung cancer in 6531 African Americans reveals new susceptibility loci. Hum Mol Genet 2025:ddaf059. [PMID: 40341939 DOI: 10.1093/hmg/ddaf059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 03/31/2025] [Accepted: 04/09/2025] [Indexed: 05/11/2025] Open
Abstract
Despite lung cancer affecting all races and ethnicities, disparities are observed in incidence and mortality rates among different ethnic groups in the United States. Non-Hispanic African Americans had a high incidence rate of lung cancer at 55.8 per 100 000 people, as well as the highest death rate at 37.2 per 100 000 people from 2016 to 2020. While previous genome-wide association studies (GWAS) have identified over 45 susceptibility risk loci that influence lung cancer development, few GWAS have investigated the etiology of lung cancer in African Americans. To address this gap in knowledge, we conducted GWAS of lung cancer focused on studying African Americans, comprising 2267 lung cancer cases and 4264 controls. We identified three loci associated with lung cancer, one with lung adenocarcinoma, and four with lung squamous cell carcinoma in this population at the genomic-wide significance level. Among them, three novel loci were identified near VWF at 12p13.31 for overall lung cancer and GACAT3 at 2p24.3 and LMAN1L at 15q24.1 for lung squamous cell carcinoma. In addition, we confirmed previously reported risk loci with known or new lead variants near CHRNA5 at 15q25.1 and CYP2A6 at 19q13.2 associated with lung cancer and TRIP13 at 5p15.33 and ERC1 at 12p13.33 associated with lung squamous cell carcinoma. Further multi-step functional analyses shed light on biological mechanisms underlying these associations of lung cancer in this population. Our study highlights the importance of ancestry-specific studies for the potential alleviation of lung cancer burden in African Americans.
Collapse
Affiliation(s)
- Jinyoung Byun
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- University of New Mexico Comprehensive Cancer Center, 1201 Camino de Salud NE, Albuquerque, NM, 87102, United States
| | - Younghun Han
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- University of New Mexico Comprehensive Cancer Center, 1201 Camino de Salud NE, Albuquerque, NM, 87102, United States
| | - Jiyeon Choi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9615 Medical Center Drive, Rockville, MD, 20850, United States
| | - Ryan Sun
- Department of Biostatistics, University of Texas, M.D. Anderson Cancer Center, 7007 Bertner Ave, Houston, TX, 77030, United States
| | - Vikram R Shaw
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Catherine Zhu
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Xiangjun Xiao
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Christine Lusk
- Department of Oncology, Wayne State University School of Medicine, 4100 John R, Detroit, MI, 48201, United States
- Karmanos Cancer Institute, 4100 John R Street, Detroit, MI, 48201, United States
| | - Hoda Badr
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Hyun-Sung Lee
- Systems Onco-Immunology Lab, David Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Hee-Jin Jang
- Systems Onco-Immunology Lab, David Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Yafang Li
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- University of New Mexico Comprehensive Cancer Center, 1201 Camino de Salud NE, Albuquerque, NM, 87102, United States
| | - Hyeyeun Lim
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Erping Long
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Yanhong Liu
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Linda Kachuri
- Department of Epidemiology and Population Health, Stanford University, 300 Pasteur Drive, Stanford, CA, 94305, United States
| | - Kyle M Walsh
- Duke Cancer Institute, Duke University Medical Center, 20 Duke Medicine Cir, Durham, NC, 27701, United States
| | - John K Wiencke
- Department of Neurological Surgery, The University of California, San Francisco, 400 Parnassus Ave, San Francisco, CA, 94143, United States
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9615 Medical Center Drive, Rockville, MD, 20850, United States
| | - Stephen Lam
- Department of Integrative Oncology, University of British Columbia, 675 West 10th Ave, Vancouver, BC V5Z 1L3, Canada
| | - Adonina Tardon
- Public Health Department, University of Oviedo, and Health Research Institute of Asturias, ISPA, Av. del Hospital Universitario, s/n, 33011 Oviedo, Asturias, Spain
| | - Marian L Neuhouser
- Program in Cancer Prevention, Public Health Sciences Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, United States
| | - Matt J Barnett
- Program in Cancer Prevention, Public Health Sciences Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, United States
| | - Chu Chen
- Program in Cancer Prevention, Public Health Sciences Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, United States
| | - Stig Bojesen
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9615 Medical Center Drive, Rockville, MD, 20850, United States
| | - Mattias Johansson
- Section of Genetics, International Agency for Research on Cancer, World Health Organization, 25 avenue Tony Garnier, CS 90627, 69366 LYON CEDEX 07, France
| | - Angela Risch
- Translational Lung Research Center Heidelberg (TLRC-H), German Center for Lung Research (DZL), Im Neuenheimer Feld 156, 69120, Heidelberg, Germany
- Division of Cancer Epigenomics, DKFZ-German Cancer Research Center, Im Neuenheimer Feld 280, D-69120, Heidelberg, Germany
- Department of Biosciences and Medical Biology, Center for Tumor Biology and Immunology, University of Salzburg and Cancer Cluster Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - H-Erich Wichmann
- Helmholtz-Munich Institute of Epidemiology, Ingolstädter Landstraße 1, Neuherberg, 85764, Germany
| | - Heike Bickeböller
- University Medical Center Göttingen, Institute of Genetic Epidemiology, Humboldtallee 32, 37073 Göttingen, Germany
| | - David C Christiani
- Department of Environmental Health and Epidemiology, Harvard T.H.Chan School of Public Health, 665 Huntington Avenue, Building 1, Boston, MA, 02115, United States
| | - Gad Rennert
- Clalit National Cancer Control Center at Carmel Medical Center and Technion Faculty of Medicine, Mikhal St 7, Haifa, 3436212, Israel
| | - Susanne Arnold
- University of Kentucky, Markey Cancer Center, 800 Rose Street, Lexington, KY, 40536, United States
| | - John K Field
- Institute of Translational Medicine, University of Liverpool, the Sherrington Building, Ashton St, Liverpool, L69 3GE, United Kingdom
| | - Sanjay Shete
- Department of Biostatistics, University of Texas, M.D. Anderson Cancer Center, 7007 Bertner Ave, Houston, TX, 77030, United States
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, United States
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, United States
| | - Geoffrey Liu
- University Health Network- The Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON M5G 2M9, Canada
| | - Angeline S Andrew
- Departments of Epidemiology and Community and Family Medicine, Dartmouth College, 1 Rope Ferry Road, Hanover, NH, 03755, United States
| | | | - Kjell Grankvist
- Department of Medical Biosciences, Umeå University, 901 87 Umeå, Sweden
| | - Mikael Johansson
- Department of Radiation Sciences, Oncology, Umeå University, 901 87 Umeå, Sweden
| | - Neil Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9615 Medical Center Drive, Rockville, MD, 20850, United States
| | - Fiona Taylor
- Sheffield Teaching Hospitals Foundation Trust, 8 Beech Hill Road, Sheffield, S10 2SB, United Kingdom
| | - Philip Lazarus
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, 412 East Spokane Falls Blvd, PBS 130, Spokane, WA, 99202, United States
| | - Matthew B Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, United States
| | - Melinda C Aldrich
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, 1161 21st Ave S, Nashville, TN, 37232, United States
| | - Alpa Patel
- American Cancer Society, Inc., 270 Peachtree Street NW, Atlanta, GA, 30303, United States
| | - Xihong Lin
- Department of Biostatistics, Harvard TH Chan School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, United States
| | - Krista A Zanetti
- Office of Nutrition Research, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, 6705 Rockledge Drive, Bethesda, MD, 20817, United States
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, 37 Convent Dr, Bethesda, MD, 20892, United States
| | - Stephen Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9615 Medical Center Drive, Rockville, MD, 20850, United States
| | - James McKay
- Section of Genetics, International Agency for Research on Cancer, World Health Organization, 25 avenue Tony Garnier, CS 90627, 69366 LYON CEDEX 07, France
| | - Ann G Schwartz
- Department of Oncology, Wayne State University School of Medicine, 4100 John R, Detroit, MI, 48201, United States
- Karmanos Cancer Institute, 4100 John R Street, Detroit, MI, 48201, United States
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 600 University Ave, Toronto, ON M5G 1X5, Canada
- Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, 155 College Street, Toronto, Ontario, M5T 3M7, Canada
| | - Christopher I Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- University of New Mexico Comprehensive Cancer Center, 1201 Camino de Salud NE, Albuquerque, NM, 87102, United States
| |
Collapse
|
136
|
Mouzaki M. Early burden, enduring risk: The alarming outcomes of pediatric MASLD. Hepatology 2025:01515467-990000000-01248. [PMID: 40338026 DOI: 10.1097/hep.0000000000001362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2025] [Accepted: 04/11/2025] [Indexed: 05/09/2025]
Affiliation(s)
- Marialena Mouzaki
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
137
|
Lu H, Zhang M, Hu Y, Sun X, Zhang R, Zhang X, Zhang M, Tang C, Cui Q, Zhang Z, Wu Z, Wang W, Song S, Cui L, Zhu J, Yang X, Yang Z. Short-chain fatty acids alleviate cholestatic liver injury by improving gut microbiota and bile acid metabolism. Int Immunopharmacol 2025; 154:114564. [PMID: 40186906 DOI: 10.1016/j.intimp.2025.114564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
Cholestasis, characterized by the obstruction of bile flow and the accumulation of bile acids, can lead to severe liver damage. Current treatments, such as ursodeoxycholic acid (UDCA) and obeticholic acid (OCA), are limited in effectiveness and have significant side effects, underscoring the need for new therapies. In our study, we investigated the effects of short-chain fatty acids (SCFAs) as a treatment in a mouse model of cholestasis induced by α-naphthylisothiocyanate (ANIT). Our findings demonstrated that SCFAs improved liver function, as indicated by reductions in liver function markers, decreased necrosis, and reduced bile duct proliferation and inflammation. Furthermore, SCFAs enhanced intestinal barrier function and increased the abundance of beneficial gut bacteria, such as Akkermansia muciniphila (A. muciniphila). SCFAs also triggered the FXR-Fgf15-Cyp7a1 pathway, reducing bile acid synthesis and improving bile acid metabolism. These findings indicate that SCFAs could offer a viable new treatment strategy for cholestatic liver conditions by improving gut-liver interactions, stabilizing bile acid metabolism, and alleviating inflammation.
Collapse
Affiliation(s)
- Han Lu
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210003, China; Huadong Medical Institute of Biotechniques, Nanjing 210018, China
| | - Mingmin Zhang
- Ili & Jiangsu Joint Institute of Health, Ili 835000, China; The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Ili 835000, China
| | - Yanan Hu
- The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing 211800, China
| | - Xuewei Sun
- Huadong Medical Institute of Biotechniques, Nanjing 210018, China
| | - Ruonan Zhang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing 211100, China
| | - Xinrui Zhang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Mingyan Zhang
- Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Chengliang Tang
- Huadong Medical Institute of Biotechniques, Nanjing 210018, China
| | - Qian Cui
- Air Force Hospital of Eastern Theater, Nanjing 210008, China
| | - Zhuohan Zhang
- Huadong Medical Institute of Biotechniques, Nanjing 210018, China
| | - Zihan Wu
- Huadong Medical Institute of Biotechniques, Nanjing 210018, China
| | - Wenjing Wang
- Department of Infectious Disease, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Shuang Song
- Department of Infectious Disease, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Lunbiao Cui
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Medical Key Laboratory of Pathogenic Microbiology in Emerging Major Infectious Diseases, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| | - Jin Zhu
- Huadong Medical Institute of Biotechniques, Nanjing 210018, China
| | - Xiaojun Yang
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210003, China.
| | - Zhan Yang
- Huadong Medical Institute of Biotechniques, Nanjing 210018, China.
| |
Collapse
|
138
|
Jiang M, Bu W, Wang X, Ruan J, Shi W, Yu S, Huang L, Xue P, Tang J, Zhao X, Su L, Cheng D. Pulmonary fibrosis: from mechanisms to therapies. J Transl Med 2025; 23:515. [PMID: 40340941 PMCID: PMC12063347 DOI: 10.1186/s12967-025-06514-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/18/2025] [Indexed: 05/10/2025] Open
Abstract
Pulmonary fibrosis (PF) is a chronic, progressive interstitial lung disease characterized by excessive deposition of extracellular matrix (ECM) and abnormal fibroblast proliferation, which is mainly caused by air pollution, smoking, aging, occupational exposure, environmental pollutants exposure, and microbial infections. Although antifibrotic agents such as pirfenidone and nintedanib, approved by the United States (US) Food and Drug Administration (FDA), can slow the decline in lung function and disease progression, their side effects and delivery inefficiency limit the overall prognosis of PF. Therefore, there is an urgent need to develop effective therapeutic targets and delivery approaches for PF in clinical settings. This review provides an overview of the pathogenic mechanisms, therapeutic drug targeting signaling pathways, and promising drug delivery strategies for treating PF.
Collapse
Affiliation(s)
- Mengna Jiang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Wenxia Bu
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Xuehai Wang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Jialing Ruan
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Weijian Shi
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Siqi Yu
- Department of Clinical Medicine, Jiangxi Medical College, Shangrao, 334000, China
| | - Lizhen Huang
- Department of Clinical Medicine, Jiangxi Medical College, Shangrao, 334000, China
| | - Peng Xue
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Juan Tang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China
| | - Xinyuan Zhao
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China.
| | - Liling Su
- Department of Clinical Medicine, Jiangxi Medical College, Shangrao, 334000, China.
| | - Demin Cheng
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, 226019, China.
| |
Collapse
|
139
|
Chen TT, Shan S, Chen YN, Li MQ, Zhang HJ, Li L, Gao PP, Li N, Huang Y, Li XL, Wei W, Sun WY. Deficiency of β-arrestin2 ameliorates MASLD in mice by promoting the activation of TAK1/AMPK signaling. Arch Pharm Res 2025:10.1007/s12272-025-01544-2. [PMID: 40341987 DOI: 10.1007/s12272-025-01544-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 04/24/2025] [Indexed: 05/11/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a liver manifestation of metabolic syndrome characterized by excessive hepatic lipid accumulation and lipid metabolism disorders. It has become the most common chronic liver disease worldwide. β-arrestin2 is a multifunctional scaffold protein that is among the most important regulatory molecules, and it exerts key roles in regulating various cellular processes, such as immune response, cellular collagen production, and inflammation. In the current study, we aimed to explore the function of β-arrestin2 in the development and progression of MASLD. Firstly, we observed that the expression of β-arrestin2 was upregulated in liver samples from patients with MASLD. Then, the western diet (WD) combined with CCl4 injection-induced MASLD was established in wild-type mice, and showed that liver β-arrestin2 expression was also gradually increased, and positively correlated with the degree of lipid metabolism disorder during MASLD progression. Ulteriorly, β-arrestin2 knockout (Arrb2 KO) mice were utilized to induce the MASLD model and found that β-arrestin2 deficiency significantly ameliorated lipid accumulation and inflammatory response in the liver of MASLD mice. Furthermore, the in vitro depletion and overexpression experiments showed that increased β-arrestin2 aggravated lipid accumulation via inhibiting the activation of the TAK1/AMPK pathway, which may be mediated by competitively binding to TAB1 with TAK1. These findings suggest that β-arrestin2 is essential to regulate intrahepatic lipid metabolism. Here, we provide a novel insight in understanding of the expression and function of β-arrestin2 in MASLD, demonstrating that it may be a potential therapeutic target for MASLD treatment.
Collapse
Affiliation(s)
- Ting-Ting Chen
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Shan Shan
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Ya-Ning Chen
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Meng-Qi Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Hui-Juan Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Ling Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Ping-Ping Gao
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Nan Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yan Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Xiao-Lei Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Hefei, 230032, Anhui, China.
| | - Wu-Yi Sun
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Hefei, 230032, Anhui, China.
| |
Collapse
|
140
|
Liu B, Tian X, Li L, Jiang N, Cheng M, Zhu J, Wu Z. SFTSV induces liver ferroptosis through m6A-related ferritinophagy. Autophagy 2025. [PMID: 40340535 DOI: 10.1080/15548627.2025.2503564] [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: 08/14/2024] [Revised: 04/15/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) is a widely prevalent infectious disease caused by severe fever with thrombocytopenia syndrome virus (SFTSV). SFTSV infection carries a high mortality rate and has emerged to be a public health concern. SFTSV infection could induce many classical cell death pathways. Ferroptosis, a novel iron-dependent form of regulated cell death, is shown to participate in various biological processes and is considered as a new therapeutic target. In the current study, we reported that SFTSV infection perturbed the classical redox cycle by downregulating the expression of GPX4, SLC7A11 and GSH, and increasing the level of reactive oxygen species (ROS) and malondialdehyde (MDA). Interestingly, we observed that the elevation of ATG5 mRNA m6A modification after SFTSV infection and mutation of the m6A-sites significantly rescued SFTSV infection-induced ferritinophagy. We further found that the NSs protein of SFTSV played a major role in driving the ferritinophagy. Finally, we found that ferroptosis inhibitor ferrostatin-1 prevented ferroptosis and suppressed SFTSV infection both in vitro and in vivo models. In summary, our study demonstrated that SFTSV infection could induce ferroptosis in liver, and m6A modified ATG5 mediated ferritinophagy to facilitate this process. Targeting ferroptosis may serve as a potential therapy for the treatment of SFTS.
Collapse
Affiliation(s)
- Bingxin Liu
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Xiaoyan Tian
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Linrun Li
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Na Jiang
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Min Cheng
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Jin Zhu
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, People's Republic of China
| | - Zhiwei Wu
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, People's Republic of China
- Huadong Medical Institute of Biotechnique, Nanjing, China
- School of Pharmacy, Dali University, Dali, Yunnan, P.R. China
| |
Collapse
|
141
|
Iommelli P, Musco N, Lombardi P, Spina AA, Morittu VM, Sarubbi F, Tufarelli V, Ceci E, Infascelli F, Tudisco R. Dietary fennel (Foeniculum vulgare Mill) seeds supplementation affects yield, fatty acid composition and flavour profile of milk and cheese in grazing goats. Trop Anim Health Prod 2025; 57:211. [PMID: 40335862 PMCID: PMC12058878 DOI: 10.1007/s11250-025-04456-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 04/23/2025] [Indexed: 05/09/2025]
Abstract
Fennel (Foeniculum vulgare Mill) is an annual plant belonging to the family of Apiaceae, widely used in Mediterranean areas for its aromatic and medical properties, especially for carminative, digestive and galactagogue effects. In this trial, 20 multiparous goats homogeneous for body weight (BW: 50.0 ± 2 kg), parity (3rd) and milk yield (1940 ± 120 g/head/day), were randomly allocated into two groups (C: control; F: fennel) fed on a permanent pasture (9:00 am to 4:00 pm). In the pen both groups received 400 g of concentrate mixture (barley and corn meals) and group F diet was supplemented with 15 g/head/day of organic fennel seeds. From the beginning of May until September, milk yield was measured daily, and samples of milk and pasture were collected monthly and analysed, along with concentrate, for their chemical composition and fatty acid profile. Cheese samples were obtained at the beginning and at the end of the trial and analysed for chemical composition, fatty acid and VOCs profile. Milk yield was significantly higher in group F (1809.6 g vs 1418.3 g for group F and C respectively), whereas the solid content did not differ between groups. Milk fatty acid profile differed between groups, especially for the content of MUFA, PUFA, and SFA. Cheese production and composition also was different for yield, fatty acid profile and VOCs composition between the groups. Indeed, the cheese of group F had higher antioxidant capacity and 4 aromatic compounds which were completely absent in the cheese of group C. These results confirm the galactagogue activity of fennel seeds in dairy goats and suggest their potential role as feed additive in grazing system to enhance production in terms of yield and antioxidant activity.
Collapse
Affiliation(s)
- Piera Iommelli
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, 80100, Naples, Italy
| | - Nadia Musco
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, 80100, Naples, Italy.
| | - Pietro Lombardi
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, 80100, Naples, Italy
| | - Anna Antonella Spina
- Department of Health Science, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Valeria Maria Morittu
- Department of Medical and Surgical Sciences, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Fiorella Sarubbi
- Institute for the Animal Production System in the Mediterranean Environment, National Research Council, 80055, Portici, Italy
| | - Vincenzo Tufarelli
- Department of Precision and Regenerative Medicine and Jonian Area, Section of Veterinary Science and Animal Production, University of Bari Aldo Moro, 70010, Valenzano, Italy
| | - Edmondo Ceci
- Department of Veterinary Medicine, University of Bari Aldo Moro, 70010, Valenzano, Italy
| | - Federico Infascelli
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, 80100, Naples, Italy
| | - Raffaella Tudisco
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, 80100, Naples, Italy
| |
Collapse
|
142
|
Lian S, Lu M, Jiajing L, Zhang B, Fang Y, Wang X, Zheng M, Ni Y, Xu G, Yang Y, Jiang R. Conjugated Lithocholic Acid Activates Hepatic TGR5 to Promote Lipotoxicity and MASLD-MASH Transition by Disrupting Carnitine Biosynthesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2410602. [PMID: 40344326 DOI: 10.1002/advs.202410602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 04/22/2025] [Indexed: 05/11/2025]
Abstract
Conjugated lithocholic acid (LCA) plays a critical role in the development of metabolic dysfunction-associated steatotic liver disease (MASLD). In this process, hepatocyte inflammation-caused upregulation of its receptor, Takeda G protein-coupled receptor 5 (TGR5) is a crucial factor. Serum bile acid profiling shows an increase in conjugated LCA, which correlates with disease severity. Depletion of Gpbar1 in hepatocytes significantly protects against the progression from MASLD to metabolic dysfunction-associated steatohepatitis (MASH) that is related to conjugated LCA. In vivo and in vitro experiments indicate that TGR5 activation in hepatocytes promotes lipotoxicity-induced cell death and inflammation by suppressing de novo carnitine biosynthesis. Mechanistically, TGR5 binding to CD36 facilitates E3 ubiquitin ligase TRIM21 recruitment, leading to the degradation of BBOX1, a crucial enzyme in de novo carnitine biosynthesis. Targeting TGR5 therapeutically can restore carnitine biosynthesis, which may offer a potent strategy to prevent or reverse the transition from MASLD to MASH.
Collapse
Affiliation(s)
- Senlin Lian
- Department of Lab Medicine, The First Affiliated Hospital of Anhui Medical University. MOE Innovation Center for Basic Research in Tumor Immunotherapy, and Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, 230022, China
| | - Meixi Lu
- Medical School of Nanjing University, Nanjing, Jiangsu Province, 210993, China
| | - Luo Jiajing
- Medical School of Nanjing University, Nanjing, Jiangsu Province, 210993, China
| | - Bin Zhang
- Department of Gastroenterology, Affiliated Nanjing Drum Tower Hospital, and Medical School of Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Yi Fang
- Department of Gastroenterology, Affiliated Nanjing Drum Tower Hospital, and Medical School of Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Xuran Wang
- Medical School of Nanjing University, Nanjing, Jiangsu Province, 210993, China
| | - Minghua Zheng
- NAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Yan Ni
- The Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Guifang Xu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Yonglin Yang
- Department of Infectious Diseases, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, China
| | - Runqiu Jiang
- Department of Lab Medicine, The First Affiliated Hospital of Anhui Medical University. MOE Innovation Center for Basic Research in Tumor Immunotherapy, and Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, 230022, China
| |
Collapse
|
143
|
Zhou S, Zhu Y, Wu Y, Zhang X, Kong X, Zhao X, Xiang H, Shang D. New insights on metabolic reprogramming in macrophage plasticity. Int Immunopharmacol 2025; 157:114797. [PMID: 40339492 DOI: 10.1016/j.intimp.2025.114797] [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: 02/12/2025] [Revised: 04/18/2025] [Accepted: 04/30/2025] [Indexed: 05/10/2025]
Abstract
Macrophages are the first line of defense in the innate immune system. Macrophages have two subtypes: classically activated macrophages (M1) and alternatively activated macrophages (M2), with different phenotypes and functions. They play a critical role in defending against pathogens and maintaining internal homeostasis. Macrophages have great plasticity in their biological characteristics. Although the regulation of macrophage plasticity has not been fully elucidated, accumulated evidence supports that microenvironmental differences are the root cause for macrophage differentiation into different subtypes. These differences alter macrophage plasticity by modulating key metabolites, activating downstream gene transcription, and influencing phagocytosis, cytokine secretion, and immune regulation. Herein, we systematically summarize metabolic reprogramming, including glucose, lipid, amino acid, ion, vitamin, nucleotide, and butyrate metabolism, as key regulators affecting macrophage polarization, providing new insights for developing targeted drugs that modulate macrophage plasticity.
Collapse
Affiliation(s)
- Siyu Zhou
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Yutong Zhu
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Yu Wu
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Xiaonan Zhang
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Xin Kong
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; College of Pharmacy, Dalian Medical University, Dalian 116011, China
| | - Xinya Zhao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; College of Pharmacy, Dalian Medical University, Dalian 116011, China
| | - Hong Xiang
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Dong Shang
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China; Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
144
|
Zhang Y, Xu P, Yu X, Zhuang K, Gui X, Yang R. Plasma proteomic analysis reveals altered protein abundances in HIV/HBV co-infection individuals with HCC and with liver cirrhosis. Sci Rep 2025; 15:15871. [PMID: 40335608 PMCID: PMC12059059 DOI: 10.1038/s41598-025-99072-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/16/2025] [Indexed: 05/09/2025] Open
Abstract
To develop a risk prediction model for hepatocellular carcinoma (HCC) by screening differentially expressed proteins (DEPs) in HIV/HBV coinfected patients with HCC and liver cirrhosis using proteomic techniques. DEPs were identified in plasma from HIV/HBV co-infected patients with HCC and liver cirrhosis using quantitative liquid chromatography-mass spectrometry (LC-MS). Mapping discovered proteins to the Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Disease Ontology (DO) databases yielded annotation information for DEPs. Differential plasma apolipoprotein A-1(APOA1) and transthyretin (TTR) expression levels were validated in 88 HIV/HBV co-infected individuals with HCC and liver cirrhosis. In total, 150 DEPs were discovered. The GO entries were primarily enriched for cutaneous immunological response mediated by circulating immunoglobulin and complement activation, as well as lipoprotein particle. The KEGG pathway enrichment was dominated by complement and coagulation cascades. Six of the 15 items enriched in the DO entries were related to lipid metabolism. APOA1, TTR, Prothrombin (F2), Antithrombin-III (SERPINC1), Alpha-2-HS-glycoprotein (AHSG), Alpha-2-macroglobulin (A2M) and Haptoglobin-related protein (HPR) were finally identified as hub proteins. Finally, a visual logistic model using immunoglobulin heavy variable 3-13 (IGHV3-13) and A2M to predict HCC were constructed. Significant variations in plasma APOA1 and TTR levels were found in HIV/HBV co-infected patients with HCC and liver cirrhosis. The screened hub proteins from DEPs can be employed as possible markers for early HCC detection. The developed HCC prediction model can be used to assess the risk of HCC in HIV/HBV co-infected cirrhotic individuals.
Collapse
Affiliation(s)
- Yongxi Zhang
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Center for AIDS Research, Wuhan University, Wuhan, Hubei, China
| | - Ping Xu
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xingxia Yu
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ke Zhuang
- ABSL-III Laboratory at the Center for Animal Experiment, State Key Laboratory of Virology, Wuhan University, Wuhan, Hubei, China
| | - Xien Gui
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Center for AIDS Research, Wuhan University, Wuhan, Hubei, China
| | - Rongrong Yang
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
- Center for AIDS Research, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
145
|
Deng H, Liang X, Dang S. Strengthening hepatitis B follow-up: Insights and experiences from China. Infect Dis Now 2025:105082. [PMID: 40345492 DOI: 10.1016/j.idnow.2025.105082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Affiliation(s)
- Haohui Deng
- Department of Infectious Disease, The Fifth People's Hospital of Shunde District, Foshan City (Longjiang Hospital of Shunde District, Foshan City), China.
| | - Xiaoting Liang
- Department of Infectious Disease, The Fifth People's Hospital of Shunde District, Foshan City (Longjiang Hospital of Shunde District, Foshan City), China
| | - Shuyuan Dang
- Department of Infectious Disease, The Fifth People's Hospital of Shunde District, Foshan City (Longjiang Hospital of Shunde District, Foshan City), China
| |
Collapse
|
146
|
Jackson E, Dennis A, Alkhouri N, Samala N, Vuppalanchi R, Sanyal AJ, Muthiah M, Banerjee R, Banerjee A. Cardiac and liver impairment on multiorgan MRI and risk of major adverse cardiovascular and liver events. Nat Med 2025:10.1038/s41591-025-03654-2. [PMID: 40335668 DOI: 10.1038/s41591-025-03654-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 03/11/2025] [Indexed: 05/09/2025]
Abstract
Cardiovascular disease and metabolic dysfunction-associated steatotic liver disease are common conditions associated with high mortality and morbidity, yet opportunities for integrated prevention are underinvestigated. We explored the association between impairment in the liver (defined by increased iron-corrected T1 (cT1) time) and/or heart (reduced left ventricular ejection fraction ≤ 50) and risk of experiencing cardiovascular- or liver-related events or all-cause mortality among 28,841 UK Biobank participants who underwent magnetic resonance imaging. Using Cox proportional hazard models, adjusted for age, sex, body mass index, type 2 diabetes and dyslipidaemia, we observed that cardiac impairment was associated with increased incidence of cardiovascular events (hazard ratio (HR) 2.3 (1.9-2.7)) and hospitalization (HR 2.1 (1.8-2.4)). Liver impairment was associated with incident cardiovascular hospitalization (cT1 ≥ 800 ms, HR 1.3 (1.1-1.5)), liver events (cT1 ≥ 875 ms, HR 9.2 (3.2-26) and hospitalization (cT1 ≥ 875 ms, HR 5.5 (3.2-9.3). Associations between cT1 and liver events were maintained in participants with metabolic dysfunction-associated steatotic liver disease (N = 6,223). Reduced left ventricular ejection fraction (≤50) combined with elevated cT1 (≥800 ms) were associated with earlier cardiovascular events (time to event 0.8 versus 2.4 years; P < 0.05). Cardiac and liver impairment are independently, or in combination, associated with cardiovascular or liver events, suggesting a dual role for magnetic resonance imaging in integrated prevention pathways.
Collapse
Affiliation(s)
| | | | - Naim Alkhouri
- Arizona Liver Health, Phoenix, AZ, USA
- Summit Clinical Research, San Antonio, AZ, USA
| | | | | | | | - Mark Muthiah
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Rajarshi Banerjee
- Perspectum Ltd, Oxford, UK
- Oxford University Hospitals NHS Trust, Oxford, UK
| | | |
Collapse
|
147
|
Lee M, Hong S, Cho Y, Rhee H, Yu MH, Bae J, Lee YH, Lee BW, Kang ES, Cha BS. Synergistic benefit of thiazolidinedione and sodium-glucose cotransporter 2 inhibitor for metabolic dysfunction-associated steatotic liver disease in type 2 diabetes: a 24-week, open-label, randomized controlled trial. BMC Med 2025; 23:266. [PMID: 40336058 PMCID: PMC12060414 DOI: 10.1186/s12916-025-04017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 03/18/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND The close interplay between metabolic dysfunction-associated steatotic liver disease (MASLD) and type 2 diabetes supports the need to identify beneficial combination therapies of antidiabetic medications targeted for the treatment of MASLD. This study aimed to investigate the complementary effects of combination therapy with pioglitazone (PIO) and empagliflozin (EMPA) on MASLD in individuals with type 2 diabetes. METHODS In a randomized, open-label trial, 50 participants with type 2 diabetes and MASLD were assigned 1:1:1 to receive PIO 15 mg, EMPA 10 mg, or a combination (PIO 15 mg plus EMPA 10 mg) daily for 24 weeks. Liver fat fraction and stiffness were evaluated using magnetic resonance imaging-proton density fat fraction (MRI-PDFF) and magnetic resonance elastography (MRE), respectively. RESULTS Combination therapy resulted in the largest reduction in liver fat and stiffness among treatment groups. Participants experiencing a relative reduction ≥ 30% or an absolute reduction ≥ 5% in liver fat were the most prevalent in the combination group (100.0% vs. 57.1% in PIO and 87.5% in EMPA, p = 0.010). In addition, the combination group showed the highest proportion of individuals with a relative reduction ≥ 30% in liver fat and ≥ 20% in liver stiffness than the monotherapy groups (50.0% vs. 21.4% in PIO and 6.3% in EMPA, p = 0.029). Combination therapy did not induce the changes in subcutaneous fat deposition observed in the monotherapy groups, but it did show the most substantial reduction in visceral fat, concurrently showing the largest increase in adiponectin level across the three groups (p = 0.036). CONCLUSIONS Combination therapy of PIO with EMPA showed synergistic benefits for MASLD in individuals with type 2 diabetes, compensating for the inadequate or unfavorable effects of monotherapies; ClincialTrials.gov number, NCT03646292. TRIAL REGISTRATION The trial was registered at ClinicalTrials.gov (registration number: NCT03646292).
Collapse
Affiliation(s)
- Minyoung Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Korea
| | - Sukchul Hong
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yongin Cho
- Department of Internal Medicine, Inha University College of Medicine, Incheon, Korea
| | - Hyungjin Rhee
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Min Heui Yu
- SENTINEL Team, Division of Endocrinology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jaehyun Bae
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Yong-Ho Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Korea
| | - Byung-Wan Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Seok Kang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Korea
| | - Bong-Soo Cha
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
148
|
Ochi H, Tani J, Tomonari T, Tanaka H, Imai Y, Tanaka T, Ohama H, Tada F, Hiraoka A, Hirose A, Ogawa C, Morishita A, Moriya A, Nakamura Y, Hirooka M, Deguchi A. Impact of muscle volume changes following atezolizumab-bevacizumab therapy in patients with unresectable hepatocellular carcinoma. Hepatol Res 2025. [PMID: 40331468 DOI: 10.1111/hepr.14201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 04/14/2025] [Accepted: 04/19/2025] [Indexed: 05/08/2025]
Abstract
AIM This study aimed to evaluate the prognostic impact of muscle volume changes during atezolizumab-bevacizumab (AB) combination therapy (AB therapy) for unresectable hepatocellular carcinoma (u-HCC). Additionally, we evaluated whether changes in muscle volume relate to prognosis based on treatment response. METHODS The present retrospective, multicenter study included 165 patients with u-HCC treated with AB therapy at eight institutions. RESULTS The median albumin-bilirubin score in the entire cohort was -2.42. The objective response rate was 31.5%, and the disease control rate was 80.6%. The median overall survival (OS) was not reached, whereas the median progression-free survival (PFS) was 6.9 months (95% confidence interval [CI]: 5.3-8.5). At the first post-treatment computed tomography scan, patients were categorized into two groups: those with a nondecreased psoas muscle area index (PI) (group A) and those with a decreased PI (group B). The median OS was not reached in either group (p = 0.059). The median PFS was 10.5 months in group A and 5.5 months in group B (p = 0.025). Multivariate analysis using the Cox proportional hazards model identified alpha-fetoprotein ≥400 ng/mL (hazard ratio [HR]: 1.68; 95% CI: 1.11-2.54; p = 0.01) and decreased PI (HR: 1.59; 95% CI: 1.07-2.36; p = 0.02) as factors associated with PFS. CONCLUSIONS Nondecreased muscle volume after initiating AB therapy was associated with improved treatment efficacy and prognosis in patients with u-HCC.
Collapse
Affiliation(s)
- Hironori Ochi
- Center for Liver-Biliary-Pancreatic Disease, Matsuyama Red Cross Hospital, Matsuyama, Ehime, Japan
| | - Joji Tani
- Department of Gastroenterology and Neurology, Kagawa University Graduate School of Medicine, Miki-cho, Kagawa, Japan
| | - Tetsu Tomonari
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima, Japan
| | - Hironori Tanaka
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima, Japan
| | - Yusuke Imai
- Center for Liver-Biliary-Pancreatic Disease, Matsuyama Red Cross Hospital, Matsuyama, Ehime, Japan
| | - Takaaki Tanaka
- Center for Liver-Biliary-Pancreatic Disease, Matsuyama Red Cross Hospital, Matsuyama, Ehime, Japan
| | - Hideko Ohama
- Gastroenterology Center, Ehime Prefectural Central Hospital, Matsuyama, Ehime, Japan
| | - Fujimasa Tada
- Gastroenterology Center, Ehime Prefectural Central Hospital, Matsuyama, Ehime, Japan
| | - Atsushi Hiraoka
- Gastroenterology Center, Ehime Prefectural Central Hospital, Matsuyama, Ehime, Japan
| | - Akira Hirose
- Department of Gastroenterology and Hepatology, Kochi University Graduate School of Medicine, Kochi, Japan
| | - Chikara Ogawa
- Department of Gastroenterology and Hepatology, Takamatsu Red Cross Hospital, Takamatsu, Kagawa, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Kagawa University Graduate School of Medicine, Miki-cho, Kagawa, Japan
| | - Akio Moriya
- Department of Gastroenterology, Mitoyo General Hospital, Kanonji, Kagawa, Japan
| | - Yoshiko Nakamura
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Masashi Hirooka
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Akihiro Deguchi
- Department of Gastroenterology, Kagawa Rosai Hospital, Marugame, Kagawa, Japan
| |
Collapse
|
149
|
Sun H, Cao Z, Zhao B, Zhou D, Chen Z, Zhang B. An elevated percentage of CD4⁺CD25⁺CD127 low regulatory T cells in peripheral blood indicates a poorer prognosis in hepatocellular carcinoma after curative hepatectomy. BMC Gastroenterol 2025; 25:340. [PMID: 40335903 PMCID: PMC12060481 DOI: 10.1186/s12876-025-03940-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 04/25/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Previous studies suggest the percentage of CD4⁺CD25⁺CD127low regulatory T cells (Tregs) in peripheral blood of patients with hepatocellular carcinoma (HCC) was significantly higher than that in healthy, which may be a significant predictor of HCC clinical outcome, and we examined the utility of Tregs in predicting prognosis in HCC after curative hepatectomy. METHODS 77 diagnosed HCC patients from August 2018 to March 2023 were selected as research objects, we retrospectively analyzed whether the preoperative percentage of CD4⁺CD25⁺CD127low Tregs in peripheral blood predicts prognosis after curative hepatectomy in HCC patients. The percentage of CD4⁺CD25⁺CD127low Tregs was detected by flow cytometry. RESULTS The percentage of CD4⁺CD25⁺CD127low Tregs was significantly elevated in patients who developed recurrence and death (p < 0.050). X-tile software was used to calculate optimal cut-off value of Treg percentage (5.85%), and patients were divided into two groups with high and low Treg percentage. Patients with higher preoperative Treg percentage had a significantly poorer prognosis (p < 0.050). Cox regression demonstrated the percentage of CD4⁺CD25⁺CD127low Tregs was an independent indicator for poor prognosis after hepatectomy. The Recurrence-free survival (RFS) (the log-rank test, p < 0.001) and Overall survival (OS) (the log-rank test, p = 0.008) in patients with higher Treg percentage were significantly lower than that in patients with lower Treg percentage. The results were confirmed by the subgroup analysis. CONCLUSION The percentage of CD4⁺CD25⁺ CD127low Tregs in peripheral blood is associated with poor prognosis in HCC patients. It can be suggested as a potential prognostic indicator for HCC patients after hepatectomy and complement existing risk stratification tools. Measuring the percentage of CD4⁺CD25⁺ CD127low Tregs may contribute to the formulation of treatment strategies and the improvement of the prognosis for HCC patients.
Collapse
Affiliation(s)
- Haoran Sun
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui Province, People's Republic of China
| | - Zepeng Cao
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui Province, People's Republic of China
| | - Baochen Zhao
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui Province, People's Republic of China
| | - Dachen Zhou
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui Province, People's Republic of China
| | - Zhongbiao Chen
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui Province, People's Republic of China
| | - Bin Zhang
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui Province, People's Republic of China.
| |
Collapse
|
150
|
Mishra AK, Dhiman RK. Hepatic encephalopathy in cirrhosis: therapies and developments. Metab Brain Dis 2025; 40:198. [PMID: 40332628 DOI: 10.1007/s11011-025-01598-7] [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: 07/07/2024] [Accepted: 03/30/2025] [Indexed: 05/08/2025]
Abstract
Hepatic encephalopathy (HE) is a frequent decompensation in patients with cirrhosis, which significantly affects morbidity and mortality. Ammonia is a major neurotoxin implicated in the pathogenesis, progression, and severity of HE, and various organs including the gut, muscle, kidney, and brain are involved in its metabolism. Therefore, therapeutic management involves reducing ammonia production and increasing its elimination from the blood and the brain. Prevention of HE in patients at high risk of first and recurrent episodes is important for prolonging survival. Various anti-ammonia therapies with synergistic and complementary actions have been attempted for overt HE and for prophylaxis of the first and recurrent episodes of HE. In the current review, we summarize the currently used and under-development pharmacotherapies/procedure(s) for HE in cirrhosis and their mechanism of action. Primary and secondary prophylaxis with monotherapies and combination therapies are also discussed.
Collapse
Affiliation(s)
- Ajay Kumar Mishra
- Department of Hepatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Radha Krishan Dhiman
- Department of Hepatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India.
| |
Collapse
|