1
|
Wang M, Li Y, Cao Y, Yang MM, Liu FJ, Jiao J, Wang SY, Song B, Wang L, Wu YQ, Kang HJ. Aspartate aminotransferase-to-platelet ratio index as a novel predictor of early mortality in heat stroke patients: a multi-centre retrospective study. Ann Med 2025; 57:2478485. [PMID: 40089314 PMCID: PMC11912296 DOI: 10.1080/07853890.2025.2478485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 02/25/2025] [Accepted: 03/02/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND The aspartate aminotransferase-to-platelet ratio index (APRI) is an effective non-invasive marker for chronic liver dysfunction. Given that heat stroke patients often suffer from poor prognosis due to multi-organ involvement, with liver injury and coagulation dysfunction being of particular concern, this study aims to investigate whether APRI can comprehensively reflect liver injury and coagulation dysfunction in heat stroke patients and explore its relationship with 28-day mortality. METHODS This retrospective study analysed electronic medical records from patients treated at 57 grade A tertiary hospitals in China from May 2005 to May 2024. The primary outcome was 28-day mortality, and the secondary outcome was 7-day mortality. Restricted cubic splines (RCS) were utilized to visualize the relationship between APRI and 28-day mortality risk. The independent association between APRI and outcomes was assessed using Cox proportional hazards models, with multivariable analyses controlling for confounding factors. The predictive ability of APRI for outcomes was evaluated using receiver operating characteristic (ROC) curves. RESULTS A total of 450 eligible patients were included, with 71 deaths occurring within 28 days. RCS analysis showed a positive correlation between APRI and 28-day mortality. Participants were divided into higher (APRI ≥ 15.14) and lower (APRI < 15.14) APRI groups. Cox proportional hazards models indicated that individuals with higher APRI had a significantly increased 28-day mortality rate (HR 5.322, 95% confidence interval [CI] 2.642-10.720, p < 0.0001). Subgroup and interaction analyses confirmed the robustness of the core findings. Additionally, the areas under the ROC (AUROC) for APRI predicting 28-day mortality was 0.823 (95% CI 0.772-0.875), significantly higher than the AST to ALT ratio (0.526, 95% CI 0.448-0.605) and total bilirubin (0.694, 95% CI 0.623-0.765). CONCLUSION APRI is an independent predictor of early mortality risk in heat stroke.
Collapse
Affiliation(s)
- Min Wang
- Medical School of Chinese PLA, Beijing, China
- Department of Critical Care Medicine, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yun Li
- Medical School of Chinese PLA, Beijing, China
- Department of Critical Care Medicine, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yuan Cao
- Medical School of Chinese PLA, Beijing, China
- Department of Critical Care Medicine, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Meng-Meng Yang
- Department of Critical Care Medicine, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Fu-Jing Liu
- Department of Emergency, The Affiliated Changzhou NO.2 People’s Hospital of Nanjing Medical University, Jiangsu, China
| | - Jie Jiao
- Department of Critical Care Medicine, Hainan Hospital of Chinese PLA General Hospital, Sanya, China
| | - Sheng-Yuan Wang
- The Sixth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Bin Song
- The Seventh Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Lu Wang
- Medical School of Chinese PLA, Beijing, China
- Department of Critical Care Medicine, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yi-Qi Wu
- Medical School of Chinese PLA, Beijing, China
- Department of Critical Care Medicine, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Hong-Jun Kang
- Department of Critical Care Medicine, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| |
Collapse
|
2
|
Yao S, Yang Z, Li J, Peng B, Wang H, Liang J, Sun C. Prevalence and prognostic significance of cachexia diagnosed by novel definition for Asian population among Chinese cirrhotic patients. Arch Gerontol Geriatr 2025; 133:105833. [PMID: 40120202 DOI: 10.1016/j.archger.2025.105833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/10/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND & AIMS Cachexia is a multifaceted metabolic disorder often linked to chronic illnesses, characterized by substantial weight reduction, inflammatory states, and loss of appetite. The novel diagnostic criteria concerning cachexia established by the Asian Working Group for Cachexia (AWGC) have not been fully validated in Chinese populations with cirrhosis. To assess the prognostic impact of AWGC-defined cachexia among hospitalized cirrhotic patients and explore the synergistic impact of Model for End-Stage Liver Disease 3.0 (MELD 3.0) scores with cachexia status on prognosis. METHODS We retrospectively analyzed clinical data from patients with decompensated cirrhosis admitted to our tertiary hospital between January 2021 and December 2023. Cachexia was identified according to AWGC criteria, and disease severity was assessed using MELD 3.0 scores. The study's primary outcome was all-cause mortality within one year. RESULTS A total of 368 patients were included in the analyses. The prevalence of cachexia was 61.7 %, and patients with cachexia had a significantly higher one-year all-cause mortality rate (26.4 % vs. 7.8 %, P < 0.001). Multivariate Cox regression analysis showed that cachexia (HR 2.68, 95 %CI 1.40-5.13, P = 0.003), along with MELD 3.0 (HR 1.18, 95 %CI 1.13-1.23, P < 0.001), were independent predictors of one-year mortality. The combined assessment of cachexia and MELD 3.0 scores yielded a higher discriminative ability for predicting one-year mortality compared to either metric alone. CONCLUSIONS AWGC-defined cachexia is a significant prognostic factor in hospitalized patients with cirrhosis. The integration of cachexia with MELD 3.0 scoring enhances prognostic prediction, underscoring the importance to introduce cachexia evaluation during clinical practice for this vulnerable setting.
Collapse
Affiliation(s)
- Shuangzhe Yao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, PR China
| | - Ziyi Yang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, PR China
| | - Jia Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, PR China
| | - Binbin Peng
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, PR China
| | - Han Wang
- Department of Health Management, Tianjin Hospital, No. 406 Jiefang South Road, Hexi District, Tianjin 300211, PR China
| | - Jing Liang
- Department of Gastroenterology and Hepatology, The Third Central Hospital of Tianjin, No.83 Jintang Road, Hedong District, Tianjin 300170, PR China.
| | - Chao Sun
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, PR China; Department of Gastroenterology, Tianjin Medical University General Hospital Airport Hospital, East Street 6, Tianjin Airport Economic Area, Tianjin 300308, PR China.
| |
Collapse
|
3
|
Huang R, Cui H, Yahya Ali Alshami MA, Fu C, Jiang W, Cai M, Zhou S, Zhu X, Hu C. LOX-1 rewires glutamine ammonia metabolism to drive liver fibrosis. Mol Metab 2025; 96:102132. [PMID: 40180177 PMCID: PMC12004974 DOI: 10.1016/j.molmet.2025.102132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025] Open
Abstract
OBJECTIVE Liver fibrosis is a crucial condition for evaluating the prognosis of chronic liver disease. Lectin-1ike oxidized low density lipoprotein receptor-1 (LOX-1) has been shown potential research value and therapeutic targeting possibilities in different fibrotic diseases. However, the role of LOX-1 and the underlying mechanisms in liver fibrosis progression remain unclear. METHODS LOX-1 expression was detected in liver tissues from patients and rodents with liver fibrosis. LOX-1 knockout rats were subjected to CCl4 or methionine and choline-deficient diet (MCD) to induce liver fibrosis. Transcriptomic and metabolomics analysis were used to investigate the involvement and mechanism of LOX-1 on liver fibrosis. RESULTS We found that LOX-1 exacerbated liver fibrosis by promoting hepatic stellate cells (HSCs) activation. LOX-1 deletion reversed the development of liver fibrosis. We further verified that LOX-1 drove liver fibrosis by reprogramming glutamine metabolism through mediating isoform switching of glutaminase (GLS). Mechanistically, we revealed the crucial role of the LOX-1/OCT1/GLS1 axis in the pathogenesis of liver fibrosis. Moreover, LOX-1 rewired ammonia metabolism by regulating glutamine metabolism-urea cycle to drive the progression of liver fibrosis. CONCLUSIONS Our findings uncover the pivotal role of LOX-1 in the progression of liver fibrosis, enrich the pathological significance of LOX-1 regulation of hepatic ammonia metabolism, and provide an insight into promising targets for the therapeutic strategy of liver fibrosis, demonstrating the potential clinical value of targeting LOX-1 in antifibrotic therapy.
Collapse
Affiliation(s)
- Ruihua Huang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Hanyu Cui
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | | | - Chuankui Fu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Wei Jiang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Mingyuan Cai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Shuhan Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Xiaoyun Zhu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, China.
| | - Changping Hu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; School of Pharmacy, Changzhi Medical College, Changzhi 046000, Shanxi, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Changsha 410013 China.
| |
Collapse
|
4
|
Xuan Y, Wang B, Xie B, Cen Y, Yu S, Yao Q. Nonlinear relationship between serum high sensitivity C reactive protein to high density lipoprotein cholesterol ratio with non-alcoholic fatty liver disease. Sci Rep 2025; 15:18579. [PMID: 40425766 PMCID: PMC12116901 DOI: 10.1038/s41598-025-03528-0] [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: 01/13/2025] [Accepted: 05/21/2025] [Indexed: 05/29/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become an increasing public health concern. We examined the association between serum high-sensitivity C-reactive protein(hs-CRP)to high-density lipoprotein cholesterol (HDL-C) ratio (HCHR) and the prevalence of NAFLD, extent of hepatic steatosis and fibrosis in the general US population. This cross-sectional analysis included 4039 adult participants from the 2017 to 2018 National Health and Nutrition Examination Survey. Multivariable logistic regression and multivariable linear regression analyses were used to assess the association between HCHR levels and NAFLD, fatty liver degree, and liver fibrosis. Generalized additive models examined the nonlinear relationship between the HCHR and NAFLD. In the three models, HCHR was positively associated with NAFLD, model 1 (OR 1.315, 95% CI 1.273, 1.359), model 2 (OR 1.364, 95% CI 1.317, 1.412) and model 3 (OR 1.120, 95% CI 1.074, 1.168). Stratified analyses showed that the association was more prominent in women, those younger than 40 years, Mexican-Americans, and those with a 25-30 kg/m2 BMI. Nonlinear association analysis revealed a threshold effect between HCHR and NAFLD, with a threshold inflection point of 2.598. We also found a significant link between HCHR and liver steatosis and the risk of liver fibrosis. In the US adult population, the increased risk of NAFLD, liver fibrosis, and severity of hepatic steatosis are independently associated with increased HCHR, and the HCHR may serve as a potential marker for NAFLD and liver fibrosis.
Collapse
Affiliation(s)
- Yanyan Xuan
- Department of Hospital Infection, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China.
- Department of Hepatology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China.
- Department of Geriatrics Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China.
| | - Bujiang Wang
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Binhua Xie
- Department of Rheumatology Immunology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Yuanyuan Cen
- Department of Respiratory and Critical Care Medicine, Cixi Longshan Hospital, Ningbo, Zhejiang, China
| | - Songping Yu
- Department of Geriatrics Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Qi Yao
- Department of Geriatrics Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China.
| |
Collapse
|
5
|
Huang CL, Qi-En S, Cen XF, Ye T, Qu HS, Chen SJ, Liu D, Xia HG, Xu CF, Zhu JS. TJ0113 attenuates fibrosis in metabolic dysfunction-associated steatohepatitis by inducing mitophagy. Int Immunopharmacol 2025; 156:114678. [PMID: 40252468 DOI: 10.1016/j.intimp.2025.114678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/01/2025] [Accepted: 04/13/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatohepatitis (MASH) fibrosis is a liver disease accompanied by inflammatory cell infiltration. There is growing evidence that insufficient mitophagy can exacerbate inflammation and liver fibrosis (LF). TJ0113 is a novel mitophagy inducer. The study aimed to explore the role of TJ0113 in ameliorating fibrosis in MASH and its mechanisms. METHODS A high-fat diet (HFD)-induced MASH mice model and a transforming growth factor (TGF)-β1-induced LX-2 cells model were used, and then they were treated with TJ0113. Changes in hepatocyte damage were observed using electron microscopy. Expression of key molecules related to mitophagy, mitochondrial damage and inflammation in liver was detected by immunofluorescence staining (IF), immunohistochemistry (IHC) and western blotting (WB). RESULT TJ0113 induces mitophagy through parkin/PINK1 and ATG5 signaling pathways and reduces lipid accumulation, inflammation and fibrosis in the liver of MASH mice. TJ0113 attenuated hepatic injury and lowered serum ALT, AST, TC and TG levels. TJ0113 reduced pro-inflammatory factors (IL-1β, IL-6, TNF-α), TGF-β1/Smad pathway activation and typical fibrosis-related molecules (α-SMA, Collagen-1) expression. In addition, NF-κB/NLRP3 signaling pathway activation after MASH was significantly attenuated by enhanced Mitophagy. We found that TJ0113 was able to effectively and safely induce mitophagy in vitro and reduce TGF-β1/Smad signaling and downstream pro-fibrotic responses in TGF-β1-treated LX-2 cells. CONCLUSION TJ0113 enhances mitophagy to inhibit lipid accumulation, inflammation and fibrosis formation in MASH, and is a candidate for MASH treatment.
Collapse
Affiliation(s)
- Chun-Lian Huang
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China
| | - Shen Qi-En
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Xu-Feng Cen
- Research Center of Clinical Pharmacy of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Ting Ye
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China
| | - Hang-Shuai Qu
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China.
| | - Si-Jia Chen
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China
| | - Dong Liu
- Hangzhou PhecdaMed Co., Ltd., Third Floor, Building 2, No. 2626. Yuhangtang Road, Yuhang District, Hangzhou 310003, China.
| | - Hong-Guang Xia
- Research Center of Clinical Pharmacy of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Cheng-Fu Xu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Jian-Sheng Zhu
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China.
| |
Collapse
|
6
|
Shen H, Liangpunsakul S, Iwakiri Y, Szabo G, Wang H. Immunological mechanisms and emerging therapeutic targets in alcohol-associated liver disease. Cell Mol Immunol 2025:10.1038/s41423-025-01291-w. [PMID: 40399593 DOI: 10.1038/s41423-025-01291-w] [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: 03/09/2025] [Accepted: 04/19/2025] [Indexed: 05/23/2025] Open
Abstract
Alcohol-associated liver disease (ALD) is a major global health challenge, with inflammation playing a central role in its progression. As inflammation emerges as a critical therapeutic target, ongoing research aims to unravel its underlying mechanisms. This review explores the immunological pathways of ALD, highlighting the roles of immune cells and their inflammatory mediators in disease onset and progression. We also examine the complex interactions between inflammatory cells and non-parenchymal liver cells, as well as their crosstalk with extra-hepatic organs, including the gut, adipose tissue, and nervous system. Furthermore, we summarize current clinical research on anti-inflammatory therapies and discuss promising therapeutic targets. Given the heterogeneity of ALD-associated inflammation, we emphasize the need for precision medicine to optimize treatment strategies and improve patient outcomes.
Collapse
Affiliation(s)
- Haiyuan Shen
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| | - Yasuko Iwakiri
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Gyongyi Szabo
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China.
| |
Collapse
|
7
|
Li Y, Gao W, Qiu Y, Pan J, Guo Q, Liu X, Geng L, Shen Y, Deng Y, Hu Z, Li S, Liu S, Idris A, Huang J, Yang H, Ge B, Fan X, Chen X, Li J. RING1 dictates GSDMD-mediated inflammatory response and host susceptibility to pathogen infection. Cell Death Differ 2025:10.1038/s41418-025-01527-2. [PMID: 40369166 DOI: 10.1038/s41418-025-01527-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 04/27/2025] [Accepted: 05/06/2025] [Indexed: 05/16/2025] Open
Abstract
RING1 is an E3 ligase component of the polycomb repressive complex 1 (PRC1) with known roles in chromatin regulation and cellular processes such as apoptosis and autophagy. However, its involvement in inflammation and pyroptosis remains elusive. Here, we demonstrate that human RING1, not RING2, promotes K48-linked ubiquitination of Gasdermin D (GSDMD) and acts as a negative regulator of pyroptosis and bacterial infection. Indeed, we showed that loss of Ring1 increased S. typhimurium infectious load and mortality in vivo. Though RING1 deletion initially reduced M. tuberculosis (Mtb) infectious load in vivo, increased lung inflammation and impaired immune defense responses were later observed. Moreover, Ring1 knockout exacerbated acute sepsis induced by lipopolysaccharide (LPS) in vivo. Mechanistically, RING1 directly interacts with GSDMD and ubiquitinates the K51 and K168 sites of GSDMD for K48-linked proteasomal degradation, thereby inhibiting pyroptosis. Inhibition of RING1 E3 ligase activity by direct mutation or with the use of small molecule inhibitors increased GSDMD level and cell death during pyroptosis. Our findings reveal that RING1 dictates GSDMD-mediated inflammatory response and host susceptibility to pathogen infection, highlighting RING1 as a potential therapeutic target for combating infectious diseases.
Collapse
Affiliation(s)
- Yuanyuan Li
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Wenqing Gao
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Yuxin Qiu
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Jiasong Pan
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Qingqing Guo
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Xuehe Liu
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Lu Geng
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Yajie Shen
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Yifan Deng
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Zhidong Hu
- Shanghai Institute of Infectious Diseases and Biosecurity & Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Suhua Li
- Division of Natural Science, Duke Kunshan University, Suzhou, China
| | - Shanshan Liu
- Shanghai Key Laboratory of Tuberculosis, Department of Microbiology and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Adi Idris
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Jinqing Huang
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Hua Yang
- Shanghai Key Laboratory of Tuberculosis, Department of Microbiology and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Baoxue Ge
- Shanghai Key Laboratory of Tuberculosis, Department of Microbiology and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoyong Fan
- Shanghai Institute of Infectious Diseases and Biosecurity & Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xiangjun Chen
- Department of Neurology, Huashan Hospital and Institute of Neurology, Fudan University, Shanghai, China
| | - Jixi Li
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
8
|
Yu R, Shi R, Chen J, Zheng X, Yu R. Investigating the Mechanism of the Fuzheng Huayu Formula in Treating Cirrhosis through Network Pharmacology, Molecular Docking, and Experimental Verification. ACS OMEGA 2025; 10:19019-19032. [PMID: 40385224 PMCID: PMC12079202 DOI: 10.1021/acsomega.5c01225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 05/20/2025]
Abstract
Cirrhosis, characterized by liver fibrosis and structural remodeling, is a leading cause of liver cancer. The Fuzheng Huayu formula (FZHY) has been approved for treating liver fibrosis in China since 2002, but its effects and mechanisms on cirrhosis remain largely unknown. This study employed network pharmacology, molecular docking, and in vitro experiments to elucidate the specific mechanisms of FZHY against liver cirrhosis. First, intersecting genes between FZHY and cirrhosis were obtained from the Chinese Medicine System Pharmacology Database, the Swiss Target Prediction online platform, UniProt, GeneCards, DisGeNET, and OMIM. The STRING database was used to construct a protein-protein interaction network. Subsequently, Gene Ontology functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed, followed by molecular docking analysis to verify binding affinities between active ingredients and candidate targets. These analyses provided a theoretical basis for subsequent experimental research. Finally, we identified 117 FZHY target genes associated with cirrhosis and constructed a drug-component-target-cirrhosis-pathway network. Enrichment analysis revealed the AGE-RAGE signaling pathway in diabetic complications as a key pathway. Molecular docking showed that Isotanshinone II had the highest affinity for CHUK, IKBKB, and MAPK14. In vitro experiments demonstrated that Isotanshinone II dose-dependently reduced the mRNA expression of COL1A1 and α-SMA, as well as the protein levels of MAPK p38, IKKβ, and NF-κB p65 in LX-2 cells. These results revealed the underlying mechanism by which Isotanshinone II in FZHY inhibited LX-2 cell activation and collagen production through suppression of the MAPK/NF-κB signaling pathway. These findings support Isotanshinone II as a promising compound for cirrhosis targeting the MAPK/NF-κB pathway. Further research is warranted to explore the bioavailability of Isotanshinone II and to optimize its structure for clinical applications.
Collapse
Affiliation(s)
- Ruixue Yu
- Medicine
College of Pingdingshan University, Pingdingshan 467000, China
| | - Run Shi
- Medicine
College of Pingdingshan University, Pingdingshan 467000, China
| | - Jinghua Chen
- Northwest
Institute of Plateau Biology, Chinese Academy
of Sciences, Xining 810008, China
| | - Xinhua Zheng
- Medicine
College of Pingdingshan University, Pingdingshan 467000, China
| | - Ruitao Yu
- Qinghai
Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy
of Sciences, Xining 810008, China
| |
Collapse
|
9
|
Aldaba-Muruato LR, Escalante-Hipólito B, Alarcón-López AY, Martínez-Soriano PA, Angeles E, Macías-Pérez JR. Preclinical Research on Cinnamic Acid Derivatives for the Prevention of Liver Damage: Promising Therapies for Liver Diseases. Biomedicines 2025; 13:1094. [PMID: 40426923 PMCID: PMC12109523 DOI: 10.3390/biomedicines13051094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/27/2025] [Accepted: 04/27/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Liver diseases are a global health issue with an annual mortality of 80,000 patients, mainly due to complications that arise during disease progression, as effective treatments are lacking. Objectives: This study evaluated the hepatoprotective effects of two derivatives of cinnamic acid, LQM717 and LQM755, in a murine model of acute liver damage induced by carbon tetrachloride (CCl4, 4 g/kg, single dose p.o.). Methods: Male Wistar rats were pretreated with five doses of LQM717 (20 mg/kg i.p.) or LQM755 (equimolar dose), starting 2 days before inducing hepatotoxic damage with CCl4. Results: The key parameters of hepatocellular function and damage showed significant increases in ALT, ALP, GGT, and total and direct bilirubin in rats intoxicated with CCl4, with decreased liver glycogen and serum albumin. Macroscopic and microscopic liver examinations revealed reduced inflammation, necrosis, and steatosis in animals pretreated with LQM717 or LQM755. Hepatomegaly was observed only in the LQM717 + CCl4 group. LQM755 statistically provided partial protection against increases in ALT and ALP and completely prevented elevations in GGT and total and direct bilirubin. LQM755 completely prevented albumin reduction, while LQM717 only partially prevented it. Both compounds partially prevented glycogen depletion. Bioinformatic analysis identified 32 potential liver protein targets for LQM717 and 36 for LQM755. Conclusions: These findings suggest that LQM717 and LQM755 have significant hepatoprotective effects against CCl4-induced acute liver injury, providing information for future studies in other acute and chronic models, as well as to elucidate their mechanisms of action.
Collapse
Affiliation(s)
- Liseth Rubí Aldaba-Muruato
- Laboratorio de Ciencias Biomédicas, Facultad de Estudios Profesionales Zona Huasteca, Universidad Autónoma de San Luis Potosí, Ciudad Valles 79060, Mexico; (L.R.A.-M.); (B.E.-H.)
| | - Brayan Escalante-Hipólito
- Laboratorio de Ciencias Biomédicas, Facultad de Estudios Profesionales Zona Huasteca, Universidad Autónoma de San Luis Potosí, Ciudad Valles 79060, Mexico; (L.R.A.-M.); (B.E.-H.)
| | - Aldo Yoshio Alarcón-López
- Laboratorio de Química Teórica y Medicinal, Departamento de Ciencias Químicas, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54750, Mexico; (A.Y.A.-L.); (P.A.M.-S.); (E.A.)
| | - Pablo A. Martínez-Soriano
- Laboratorio de Química Teórica y Medicinal, Departamento de Ciencias Químicas, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54750, Mexico; (A.Y.A.-L.); (P.A.M.-S.); (E.A.)
| | - Enrique Angeles
- Laboratorio de Química Teórica y Medicinal, Departamento de Ciencias Químicas, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54750, Mexico; (A.Y.A.-L.); (P.A.M.-S.); (E.A.)
| | - José Roberto Macías-Pérez
- Laboratorio de Ciencias Biomédicas, Facultad de Estudios Profesionales Zona Huasteca, Universidad Autónoma de San Luis Potosí, Ciudad Valles 79060, Mexico; (L.R.A.-M.); (B.E.-H.)
| |
Collapse
|
10
|
Chen GY, Ji XY, Li Y, Zheng SS, Jin Q, Tao QW. Mechanisms of Total Glucosides of Paeony in Alleviating Methotrexate-Induced Liver Injury. Drug Des Devel Ther 2025; 19:3407-3423. [PMID: 40322039 PMCID: PMC12049130 DOI: 10.2147/dddt.s521740] [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: 02/19/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025] Open
Abstract
Objective Total glycoside of peony (TGP) enhances methotrexate efficacy and attenuates its hepatotoxicity in rheumatoid arthritis, but the mechanisms remain unclear. This study investigates the mechanisms of TGP against methotrexate-induced liver injury through a network pharmacology-based approach. Methods A liver injury model was established in CD-1 mice by intraperitoneal injection of 20 mg/kg methotrexate. TGP and the positive control drug silybin were used to intervene in the methotrexate-induced liver injury model in mice. Serum ALT and AST activities, liver index test and histopathology was detected to evaluate the effects of the treatment on methotrexate-induced liver injury. Additionally, network pharmacology and serum metabolomics were employed to predict the mechanisms of TGP in treating methotrexate-induced liver injury. Experimental validation was conducted by RT-PCR, ELISA and Western blot. Results TGP effectively alleviated the liver index and pathological liver damage induced by methotrexate and reduced the liver injury markers, serum ALT and AST, showing effects comparable to those of the positive control drug silybin. Network pharmacology predicted that the key targets and key signaling pathways of TGP in treating methotrexate-induced liver injury are closely associated with inflammatory response. Furthermore, serum metabolomics and network pharmacology analysis indicated a close association between effects of TGP on methotrexate-induced liver injury and arachidonic acid pathway. Experimental validation results confirmed that the expression levels of IL-6, TNF and COX-2 in liver tissues were significantly elevated, with the activation of the PI3K/AKT, MAPK, and NFκB pathways. TGP intervention can reverse these changes to a certain extent. Conclusion TGP treatment effectively mitigates methotrexate-induced liver injury, and its mechanism is closely associated with the inhibition of hepatic inflammatory responses.
Collapse
Affiliation(s)
- Guang-Yao Chen
- Department of TCM Rheumatology, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Xiang-Yu Ji
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, People’s Republic of China
| | - Ying Li
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
| | - Si-Si Zheng
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, 100088, People’s Republic of China
| | - Qi Jin
- Department of Dermatology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
| | - Qing-Wen Tao
- Department of TCM Rheumatology, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| |
Collapse
|
11
|
Shang DF, Xu WQ, Zhao Q, Zhao CL, Wang SY, Han YL, Li HG, Liu MH, Zhao WX. Molecular mechanisms of pyroptosis in non-alcoholic steatohepatitis and feasible diagnosis and treatment strategies. Pharmacol Res 2025; 216:107754. [PMID: 40306603 DOI: 10.1016/j.phrs.2025.107754] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/11/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
Pyroptosis is a distinct form of cell death that plays a critical role in intensifying inflammatory responses. It primarily occurs via the classical pathway, non-classical pathway, caspase-3/6/7/8/9-mediated pathways, and granzyme-mediated pathways. Key effector proteins involved in the pyroptosis process include gasdermin family proteins and pannexin-1 protein. Pyroptosis is intricately linked to the onset and progression of non-alcoholic steatohepatitis (NASH). During the development of NASH, factors such as pyroptosis, innate immunity, lipotoxicity, endoplasmic reticulum stress, and gut microbiota imbalance interact and interweave, collectively driving disease progression. This review analyzes the molecular mechanisms of pyroptosis and its role in the pathogenesis of NASH. Furthermore, it explores potential diagnostic and therapeutic strategies targeting pyroptosis, offering new avenues for improving the diagnosis and treatment of NASH.
Collapse
Affiliation(s)
- Dong-Fang Shang
- Henan University of CM, Zhengzhou 450000, China; The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China
| | - Wen-Qian Xu
- Henan University of CM, Zhengzhou 450000, China
| | - Qing Zhao
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China
| | - Chen-Lu Zhao
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China
| | - Si-Ying Wang
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China
| | - Yong-Li Han
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China
| | - He-Guo Li
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China.
| | - Ming-Hao Liu
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China.
| | - Wen-Xia Zhao
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China.
| |
Collapse
|
12
|
Chen C, Bu X, Deng L, Xia J, Wang X, Chen L, Li W, Huang J, Chen Q, Wang C. Astragaloside IV as a promising therapeutic agent for liver diseases: current landscape and future perspectives. Front Pharmacol 2025; 16:1574154. [PMID: 40337517 PMCID: PMC12055773 DOI: 10.3389/fphar.2025.1574154] [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: 02/11/2025] [Accepted: 04/10/2025] [Indexed: 05/09/2025] Open
Abstract
Astragaloside IV (C41H68O14, AS-IV) is a naturally occurring saponin isolated from the root of Astragalus membranaceus, a widely used traditional Chinese botanical drug in medicine. In recent years, AS-IV has attracted considerable attention for its hepatoprotective properties, which are attributed to its low toxicity as well as its anti-inflammatory, antioxidant and antitumour effects. Numerous preclinical studies have demonstrated its potential in the prevention and treatment of various liver diseases, including multifactorial liver injury, metabolic-associated fatty liver disease, liver fibrosis and liver cancer. Given the promising hepatoprotective potential of AS-IV and the growing interest in its research, this review provides a comprehensive summary of the current state of research on the hepatoprotective effects of AS-IV, based on literature available in databases such as CNKI, PubMed, ScienceDirect, Google Scholar and Web of Science. The hepatoprotective mechanisms of AS-IV are multifaceted, encompassing the inhibition of inflammatory responses, reduction of oxidative stress, improvement of insulin and leptin resistance, modulation of the gut microbiota, suppression of hepatocellular carcinoma cell proliferation and induction of tumour cell apoptosis. Notably, key molecular pathways involved in these effects include Nrf2/HO-1, NF-κB, NLRP3/Caspase-1, JNK/c-Jun/AP-1, PPARα/FSP1 and Akt/GSK-3β/β-catenin. Toxicity studies indicate that AS-IV has a high level of safety. In addition, this review discusses the sources, physicochemical properties, and current challenges in the development and clinical application of AS-IV, providing valuable insights into its potential as a hepatoprotective agent in the pharmaceutical and nutraceutical industries.
Collapse
Affiliation(s)
- Chunyan Chen
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xiaolan Bu
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Liping Deng
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jiayan Xia
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xinming Wang
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Li Chen
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Wen Li
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jie Huang
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Qixiang Chen
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Cheng Wang
- School of Clinical Medical, Chengdu Medical College, Chengdu, China
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
13
|
Zhang H, Kong X, Wang W, Zhou H, Qu H, Guan Z, Wu H, Zhai X, Jin B. TRIM25-Mediated INSIG1 Ubiquitination Promotes MASH Progression Through Reprogramming Lipid Metabolism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2414646. [PMID: 40231613 DOI: 10.1002/advs.202414646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/30/2025] [Indexed: 04/16/2025]
Abstract
The global incidence of Metabolic dysfunction-associated steatohepatitis (MASH) is increasing, highlighting the urgent need for new treatment strategies. This study aimed to investigate the involvement of tripartite motif-containing 25 (TRIM25) in MASH progression and explore the therapeutic potential of the TRIM25 inhibitor, C27H26N2O4S. Functional studies reveal that TRIM25 promoted lipid accumulation and inflammation by ubiquitinating and degrading insulin-induced gene 1 (INSIG1), thereby enhancing the nuclear translocation of sterol regulatory element-binding protein 2 (SREBP2) and upregulating lipid biosynthesis genes. In vivo experiments using TRIM25 knockout mice demonstrated that TRIM25 deletion ameliorated MASH progression, reduced fibrosis, and decreased inflammatory cell infiltration. It identifies C27H26N2O4S as a specific inhibitor of TRIM25. C27H26N2O4S effectively decreased INSIG1 ubiquitination and attenuated lipid accumulation in the hepatocytes. To enhance the hepatic delivery of C27H26N2O4S, it utilizes exosomes derived from hepatic stellate cells (HSC-EVs). Biodistribution analysis confirmed that the HSC-EVs preferentially accumulated in the liver. In a MASH mouse model, HSC-EV-encapsulated C27H26N2O4S (C27H26N2O4S@HSC-EV) significantly reduced hepatic lipid accumulation and alleviated MASH severity and fibrosis. This study highlights the critical regulatory role of TRIM25 in MASH and presents C27H26N2O4S@HSC-EV as a promising therapeutic approach for MASH treatment.
Collapse
Affiliation(s)
- Hao Zhang
- Organ Transplant Department, Qilu Hospital of Shandong University, Jinan, China
- Shandong Province Engineering Research Center for Multidisciplinary Research on Hepatobiliary and Pancreatic Malignant Tumors, Jinan, China
| | - Xiangxu Kong
- Shandong Province Engineering Research Center for Multidisciplinary Research on Hepatobiliary and Pancreatic Malignant Tumors, Jinan, China
- Department of Hepatobiliary Surgery, the Second Hospital of Shandong University, Beiyuan Street & 247 Jinan, Shandong, China
| | - Wei Wang
- Medical Integration and Practice Center, Shandong University, Jinan, China
| | - Huaxin Zhou
- Shandong Province Engineering Research Center for Multidisciplinary Research on Hepatobiliary and Pancreatic Malignant Tumors, Jinan, China
- Department of Hepatobiliary Surgery, the Second Hospital of Shandong University, Beiyuan Street & 247 Jinan, Shandong, China
| | - Haoran Qu
- Department of Hepatobiliary Surgery, the Second Hospital of Shandong University, Beiyuan Street & 247 Jinan, Shandong, China
| | - Zhengyao Guan
- Department of Hepatobiliary Surgery, the Second Hospital of Shandong University, Beiyuan Street & 247 Jinan, Shandong, China
| | - Honglei Wu
- Department of Gastroenterology, the Second Hospital of Shandong University, Beiyuan Street & 247 Jinan, Shandong, China
| | - Xiangyu Zhai
- Shandong Province Engineering Research Center for Multidisciplinary Research on Hepatobiliary and Pancreatic Malignant Tumors, Jinan, China
- Department of Hepatobiliary Surgery, the Second Hospital of Shandong University, Beiyuan Street & 247 Jinan, Shandong, China
| | - Bin Jin
- Organ Transplant Department, Qilu Hospital of Shandong University, Jinan, China
- Shandong Province Engineering Research Center for Multidisciplinary Research on Hepatobiliary and Pancreatic Malignant Tumors, Jinan, China
- Department of Hepatobiliary Surgery, the Second Hospital of Shandong University, Beiyuan Street & 247 Jinan, Shandong, China
| |
Collapse
|
14
|
Hong J, Kim YH. Cutting-edge biotherapeutics and advanced delivery strategies for the treatment of metabolic dysfunction-associated steatotic liver disease spectrum. J Control Release 2025; 380:433-456. [PMID: 39923856 DOI: 10.1016/j.jconrel.2025.02.008] [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: 09/30/2024] [Revised: 12/22/2024] [Accepted: 02/04/2025] [Indexed: 02/11/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), a condition with the potential to progress into liver cirrhosis or hepatocellular carcinoma, has become a significant global health concern due to its increasing prevalence alongside obesity and metabolic syndrome. Despite the promise of existing therapies such as thyroid hormone receptor-β (THR-β) agonists, PPAR agonists, FXR agonists, and GLP-1 receptor agonists, their effectiveness is limited by the complexity of the metabolic, inflammatory, and fibrotic pathways that drive MASLD progression, encompassing steatosis, metabolic dysfunction-associated steatohepatitis (MASH), and reversible liver fibrosis. Recent advances in targeted therapeutics, including RNA interference (RNAi), mRNA-based gene therapies, monoclonal antibodies, proteolysis-targeting chimeras (PROTAC), peptide-based strategies, cell-based therapies such as CAR-modified immune cells and stem cells, and extracellular vesicle-based approaches, have emerged as promising interventions. Alongside these developments, innovative drug delivery systems are being actively researched to enhance the stability, precision, and therapeutic efficacy of these biotherapeutics. These delivery strategies aim to optimize biodistribution, improve target-specific action, and reduce systemic exposure, thus addressing critical limitations of existing treatment modalities. This review provides a comprehensive exploration of the underlying biological mechanisms of MASLD and evaluates the potential of these cutting-edge biotherapeutics in synergy with advanced delivery approaches to address unmet clinical needs. By integrating fundamental disease biology with translational advancements, it aims to highlight future directions for the development of effective, targeted treatments for MASLD and its associated complications.
Collapse
Affiliation(s)
- Juhyeong Hong
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research Hanyang University, 04763 Seoul, South Korea; Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 04763 Seoul, South Korea
| | - Yong-Hee Kim
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research Hanyang University, 04763 Seoul, South Korea; Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 04763 Seoul, South Korea; Cursus Bio Inc., Icure Tower, Gangnam-gu, Seoul 06170, Republic of Korea.
| |
Collapse
|
15
|
Hu T, Chen M, Tian S, Luo P, Zhang J. Mechanistic Studies on the Role of IL-17/NLRP3 in Arsenic-Induced Activation of Hepatic Stellate Cells Through Hepatocyte Proptosis. TOXICS 2025; 13:287. [PMID: 40278603 PMCID: PMC12031523 DOI: 10.3390/toxics13040287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 04/26/2025]
Abstract
Long-term exposure to arsenic, a prevalent environmental contaminant, has been implicated in the pathogenesis of various hepatic conditions. Hepatic stellate cells (HSCs) are central to the development of liver fibrosis. Recently, the involvement of interleukin-17 (IL-17) and the NOD-like receptor protein 3 (NLRP3) inflammasome in hepatic pathologies has attracted significant research interest. Hepatocyte pyroptosis, a form of programmed cell death, is a critical factor in the occurrence of inflammation. The objective of this study was to investigate the specific roles of IL-17 and NLRP3 in the arsenic-induced activation of HSCs through hepatocyte pyroptosis. We pretreated MIHA cells with MCC950 (1 and 5 μM) and secukinumab (10 and 100 nM) for 4 h, then with NaAsO2 (25 μM) for 24 h at 37 °C under 5% CO2. After incubation, the cell-culture supernatant was collected and mixed with serum-free high-glucose DMEM medium in a 1:1 ratio to prepare the conditioned medium, which was subsequently used for the culture of LX-2 cells. The results showed that exposure to NaAsO2 induced hepatocellular pyroptosis, which led to the release of the inflammatory cytokines IL-18 and IL-1β and subsequent activation of HSCs. Treatment with the inhibitors MCC950 and secukinumab significantly reduced the secretion of Extracellular matrix (ECM) components and attenuated HSC activation. These results demonstrate that blocking the IL-17 and NLRP3 signaling pathways significantly reduces HSC activation and attenuates hepatic fibrogenesis. These results provide novel molecular targets for the prevention and treatment of arsenic-related liver fibrosis.
Collapse
Affiliation(s)
- Ting Hu
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China; (T.H.); (M.C.); (S.T.)
| | - Mei Chen
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China; (T.H.); (M.C.); (S.T.)
| | - Sai Tian
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China; (T.H.); (M.C.); (S.T.)
| | - Peng Luo
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China; (T.H.); (M.C.); (S.T.)
| | - Jiangping Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 561113, China; (T.H.); (M.C.); (S.T.)
- Guiyang Public Health Clinical Center, Guiyang 550003, China
| |
Collapse
|
16
|
Xiao P, Ye Z, Li X, Feng Q, Su Y. Ginseng and its functional components in non-alcoholic fatty liver disease: therapeutic effects and multi-target pharmacological mechanisms. Front Pharmacol 2025; 16:1540255. [PMID: 40271056 PMCID: PMC12014752 DOI: 10.3389/fphar.2025.1540255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/28/2025] [Indexed: 04/25/2025] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is a common type of chronic liver disease and its incidence is increasing. Its disease progression is closely related to non-alcoholic steatohepatitis and liver fibrosis. Effective treatment is currently lacking. The traditional Chinese medicine ginseng (Panax ginseng) shows unique advantages in NAFLD intervention, but its complex compositional system and molecular mechanism network still need to be systematically analyzed. Objective This paper systematically integrates evidence from nearly 20 years of research to elucidate the multi-target pharmacological mechanism of ginseng for the treatment of NAFLD. Methods Relevant information was sourced from Pubmed, Web of science, Embase and CNKI databases. Using BioRender and visio to draw biomedical illustrations. Results The active ingredients of ginseng contain 2 classes of saponins (tetracyclic triterpene saponins, pentacyclic triterpene saponins and other modified types) and non-saponins. Different cultivation methods, processing techniques and extraction sites have expanded the variety of ginseng constituents and demonstrated different pharmacological activities. Studies have shown that ginseng and its functional components have the ability to regulate lipid metabolism disorders, inflammation, oxidative stress, endoplasmic reticulum stress, insulin resistance, disruption of intestinal flora structure, cell death and senescence. Demonstrates the potential of ginseng for the treatment of NAFLD. Conclusion This study reveals for the first time the integrative mechanism of ginseng in the treatment of NAFLD through the tertiary mode of action of "multi-component multi-target multi-pathway". The multilevel modulatory ability of ginseng provides a new direction for the development of comprehensive therapeutic strategies for NAFLD.
Collapse
Affiliation(s)
| | | | | | - Quansheng Feng
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yue Su
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
17
|
Cheung J, Cheung BMY, Yiu KH, Tse HF, Chan YH. Role of metabolic dysfunction-associated fatty liver disease in atrial fibrillation and heart failure: molecular and clinical aspects. Front Cardiovasc Med 2025; 12:1573841. [PMID: 40264510 PMCID: PMC12011764 DOI: 10.3389/fcvm.2025.1573841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MASLD) is a rising global health concern. In addition to direct hepatic complications, extra-hepatic complications, including cardiovascular diseases (CVD), type 2 diabetes (T2D), gastroesophageal reflux disease, chronic kidney disease and some malignancies, are increasingly recognized. CVD, including atrial fibrillation (AF) and heart failure (HF), is the leading cause of death in patients with MASLD. External factors, including excess energy intake, sedentary lifestyle and xenobiotic use, induce inflammation-related complications. MASLD, AF, and HF are associated with immune system activation, including the reprogramming of immune cells and the establishment of immune memory. Emerging evidence suggests that the heart and the liver cross-talk with each other through the diverse spectrum of autocrine, paracrine and endocrine mechanisms. Pro-inflammatory cytokines produced from the liver and the heart circulate systemically to orchestrate metabolic derangements that promote the systematic immune dysregulation in the heart-liver axis and the development of end-organ complications. Cardio-hepatic syndrome describes the clinical and biochemical evidence of hepatic dysfunction and cardiac pathology due to the interaction between the heart and the liver. Activation of inflammatory cascades, oxidative stress and immune system dysregulation underlie key mechanisms in bringing about such pathological changes. This review focuses on the current clinical and molecular evidence about the heart-liver cross-talk. It summarizes the epidemiological and pathophysiological associations of MASLD, AF and HF. In addition, we will discuss how repurposing currently available and emerging pharmacotherapies may help tackle the cardiovascular risks resulting from MASLD.
Collapse
Affiliation(s)
- Jamie Cheung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Bernard Man-Yung Cheung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kai-Hang Yiu
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Medicine, Shenzhen Hong Kong University Hospital, Hong Kong SAR, China
- Institute of Cardiovascular Science and Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Hung-Fat Tse
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Medicine, Shenzhen Hong Kong University Hospital, Hong Kong SAR, China
| | - Yap-Hang Chan
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
- Institute of Cardiovascular Science and Medicine, The University of Hong Kong, Hong Kong SAR, China
- Division of Experimental Medicine and Immunotherapeutics, Cambridge University Hospitals NHS Foundation Trust/University of Cambridge, Cambridge, United Kingdom
- Department of Cardiology, Royal Papworth Hospital, Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
18
|
Hou W, Fang P, Liang J, Wei X, Ma C, Gao Y, Zhang Q, Li J. The Jieduan-Niwan Formula Reduces Inflammatory Responses in Acute-on-Chronic Liver Failure Rats by Inhibiting HMGB1-Induced Hepatocyte Pyroptosis. Drug Des Devel Ther 2025; 19:2503-2517. [PMID: 40190810 PMCID: PMC11972575 DOI: 10.2147/dddt.s488659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/29/2025] [Indexed: 04/09/2025] Open
Abstract
Background Acute-on-chronic liver failure (ACLF) is a global intractable disease. HMGB1-induced hepatocyte pyroptosis expanding inflammatory responses contributes to the pathogenesis of ACLF. The JDNW formula (JDNWF) has a significant clinical effect on ACLF, but its hepatoprotective mechanisms remain elusive. Purpose To explore the potential molecular mechanisms of the JDNWF in ACLF by HMGB1-induced hepatocyte pyroptosis. Methods Rats were divided into normal, ACLF, Caspase-1 inhibitor, HMGB1 inhibitor, JDNW, JDNW+Caspase-1 inhibitor and JDNW+HMGB1 inhibitor groups. The ACLF rat model was established by 40% carbon tetrachloride-induced liver fibrosis, followed by intraperitoneal injection of D-galactosamine and lipopolysaccharide. The liver function, coagulation function, liver pathological damage and ultrastructural changes of hepatocytes were evaluated. Triple-immunostaining of active Caspase-1, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and albumin were performed to evaluate the percentage of pyroptotic hepatocytes. Western blot, immunofluorescence, enzyme-linked immunosorbent assay (ELISA) and quantitative real-time PCR (RT-qPCR) were used to analyze the expressions of key genes and proteins in HMGB1-induced pyroptosis pathways and the level of inflammatory factors. Results The JDNWF improved liver function, coagulation function and liver pathological damage, reduced the percentage of pyroptotic hepatocytes and inflammatory responses, and down-regulated the expressions of key genes and proteins in the HMGB1-induced pyroptosis pathways in ACLF rats. The effect of the JDNWF was better than those of HMGB1 inhibitor (glycyrrhizin) and Caspase-1 inhibitor (VX-765). Compared with glycyrrhizin or VX-765, there were no significant differences in the above indicators after the JDNWF in combination with glycyrrhizin or VX-765. These results indicated that the JDNWF inhibited hepatocyte pyroptosis and liver inflammation in ACLF rats through the HMGB1-induced pyroptosis pathways. Conclusion The JDNWF protects the livers of ACLF rats by inhibiting HMGB1-induced hepatocyte pyroptosis reducing inflammatory responses, suggesting that HMGB1-induced hepatocyte pyroptosis may be a potential therapeutic target of ACLF.
Collapse
Affiliation(s)
- Weixin Hou
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- Key Laboratory of Gut Microbiota Translational Medicine Research, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Peng Fang
- Department of Infectious Diseases, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Jiajun Liang
- Department of Hepatology, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou, Guangdong province, People’s Republic of China
| | - Xiaoyi Wei
- Office of Science and Technology Administration, Beijing Friendship Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Chongyang Ma
- Department of Hepatology, School of Traditional Chinese Medicine, Capital Medical University, Beijing, People’s Republic of China
- Department of Hepatology, Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Capital Medical University, Beijing, People’s Republic of China
| | - Yanbin Gao
- Department of Endocrinology, School of Traditional Chinese Medicine, Capital Medical University, Beijing, People’s Republic of China
- Department of Endocrinology, Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Capital Medical University, Beijing, People’s Republic of China
| | - Qiuyun Zhang
- Department of Hepatology, School of Traditional Chinese Medicine, Capital Medical University, Beijing, People’s Republic of China
- Department of Hepatology, Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Capital Medical University, Beijing, People’s Republic of China
| | - Jingnan Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- Key Laboratory of Gut Microbiota Translational Medicine Research, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
19
|
Gao J, Wang X, Ye S, Zhang Y, Qin Y. Ghrelin alleviates inflammation and pyroptosis by inhibiting TNF-α /caspase-8/caspase-3/ GSDME signalling pathways in an in vitro model of high glucose induced liver injury. Tissue Cell 2025; 93:102672. [PMID: 39721419 DOI: 10.1016/j.tice.2024.102672] [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: 10/11/2024] [Revised: 11/17/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024]
Abstract
Diabetic liver injury (DLI) refers to liver injury resulting from prolonged chronic hyperglycemia and represents a significant complication associated with diabetes, The specific pathogenic mechanism of DLI remains incompletely understood. Tumor necrosis factor α (TNF-α) has been demonstrated to play a crucial role in diabetic complications through intricate signalling pathways, including pyroptosis. However, it remains uncertain whether TNF-α mediates pyroptosis in DLI, we initially established an in vitro model of DLI and confirmed the presence of an inflammatory state characterized by TNF-α in DLI. Furthermore, evidence of gasdermin E (GSDME)-mediated pyroptosis and the activation of cysteinyl aspartate specific proteinase (caspase)-8 was observed in AML-12 cell exposed to high glucose concentrations. We subsequently demonstrated that TNF-α can trigger caspase-8 activation, leading to GSDME-mediated cellular pyroptosis. Furthermore, treatment with ghrelin effectively suppressed hepatic cell pyroptosis induced by high glucose concentrations and provided protection against liver injury. Therefore, we propose that the TNF-α/caspase-8/caspase-3/GSDME pathway represents a novel mechanism underlying pyrodeath in DLI cells and to explore the protective role and molecular mechanisms underlying the effects of ghrelin on DLI by this special pathway, These findings may present potential therapeutic implications for the management of DLI.
Collapse
Affiliation(s)
- Jingwen Gao
- Department of Basic Medicine, Dali University, Dali, Yunnan 671000, China
| | - Xinrui Wang
- Department of Basic Medicine, Dali University, Dali, Yunnan 671000, China
| | - Shengying Ye
- Department of Basic Medicine, Dali University, Dali, Yunnan 671000, China
| | - Yixin Zhang
- Department of Basic Medicine, Dali University, Dali, Yunnan 671000, China
| | - Yan Qin
- Department of Basic Medicine, Dali University, Dali, Yunnan 671000, China.
| |
Collapse
|
20
|
Kornfehl A, Tiede A, Hemetsberger P, Kappel J, Müllner-Bucsics T, Stockhoff L, Rieland H, Reider L, Dominik N, Kramer G, Trauner M, Mandorfer M, Falk C, Maasoumy B, Reiberger T, Hartl L. Decreasing interleukin-6 levels after TIPS predict outcomes in decompensated cirrhosis. JHEP Rep 2025; 7:101308. [PMID: 40124165 PMCID: PMC11929062 DOI: 10.1016/j.jhepr.2024.101308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 03/25/2025] Open
Abstract
Background & Aims Transjugular intrahepatic portosystemic shunt (TIPS) effectively treats complications of cirrhosis. Systemic inflammation (SI) is linked to acute-on-chronic liver failure (ACLF) and liver-related death. We aimed to assess the trajectory and clinical impact of SI parameters after TIPS implantation. Methods Consecutive patients undergoing elective implantation of covered TIPS for recurrent/refractory ascites or portal-hypertensive bleeding at the Medical University Vienna (NCT03409263; n = 58) and at the Hannover Medical School (NCT04801290, n = 51) were included. IL-6 was assessed at baseline (BL), 3 months (M3) and up to 6 (M6; Hannover cohort) or 9 months (M9; Vienna cohort) of follow-up; C-reactive protein (CRP) and lipopolysaccharide-binding protein (LBP) were assessed in the Vienna cohort only. Results In 109 patients (66.1% male, median age 57 years) receiving TIPS mainly (72.4%) by indication ascites the median BL IL-6 levels were 10.5 pg/ml; and 41.3% (n = 45/109) patients exhibiting IL-6 ≥14 pg/ml. From BL to M3, IL-6 decreased in 63.8% (n = 37/58; Vienna cohort) and in 68.6% (n = 35/51; Hannover cohort) of patients, respectively. Similar rates of decreases were observed also for CRP (in 62.1%) and for LBP (in 77.4%). A considerable IL-6 reduction (≥50% of baseline) was noted in 41 (37.6%) patients during follow-up. Competing risk regression in the combined cohort adjusted for age, albumin, and model for end-stage liver disease revealed that IL-6 decrease at M3 was an independently protective factor for the development of ACLF (adjusted subdistribution hazard ratio [asHR]: 0.26; 95% CI: 0.09-0.77; p = 0.016) and liver-related death (asHR: 0.26; 95% CI: 0.07-0.95; p = 0.042). Conclusions TIPS leads to a sustained reduction of SI and bacterial translocation in patients with decompensated cirrhosis. Decreasing IL-6 levels three months after TIPS implantation indicate a lower risk of ACLF and liver-related death in patients with cirrhosis. Impact and implications Systemic inflammation is a major driver of disease progression in patients with decompensated advanced chronic liver disease (dACLD). This study demonstrates that systemic inflammation (i.e. interleukin-6 [IL-6]) effectively and sustainedly decreases after transjugular intrahepatic portosystemic shunt (TIPS) implantation. A decrease of IL-6 3 months after TIPS implantation is a protective factor for acute-on-chronic liver failure and liver-related death. Thus, our results suggest that TIPS reduces systemic inflammation in a clinically meaningful way.
Collapse
Affiliation(s)
- Andrea Kornfehl
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Anja Tiede
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Hannover/Braunschweig, Germany
| | - Paul Hemetsberger
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Julia Kappel
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Clinical Research Group MOTION, Medical University of Vienna, Vienna, Austria
| | - Theresa Müllner-Bucsics
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Clinical Research Group MOTION, Medical University of Vienna, Vienna, Austria
| | - Lena Stockhoff
- Department of Internal Medicine, Marienhospital Osnabrück, Germany
| | - Hannah Rieland
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Lukas Reider
- Division of Interventional Radiology, Department of Radiology, Medical University of Vienna, Vienna, Austria
| | - Nina Dominik
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Georg Kramer
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Clinical Research Group MOTION, Medical University of Vienna, Vienna, Austria
| | - Mattias Mandorfer
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Clinical Research Group MOTION, Medical University of Vienna, Vienna, Austria
| | - Christine Falk
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
| | - Benjamin Maasoumy
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Hannover/Braunschweig, Germany
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Clinical Research Group MOTION, Medical University of Vienna, Vienna, Austria
- Christian Doppler Lab for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria
| | - Lukas Hartl
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Clinical Research Group MOTION, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
21
|
Chen Y, Bian S, Le J. Molecular Landscape and Diagnostic Model of MASH: Transcriptomic, Proteomic, Metabolomic, and Lipidomic Perspectives. Genes (Basel) 2025; 16:399. [PMID: 40282358 PMCID: PMC12026639 DOI: 10.3390/genes16040399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH), a progressive form of fatty liver disease, presents a significant global health challenge. Despite extensive research, fully elucidating its complex pathogenesis and developing accurate non-invasive diagnostic tools remain key goals. Multi-omics approaches, integrating data from transcriptomics, proteomics, metabolomics, and lipidomics, offer a powerful strategy to achieve these aims. This review summarizes key findings from multi-omics studies in MASH, highlighting their contributions to our understanding of disease mechanisms and the development of improved diagnostic models. Transcriptomic studies have revealed widespread gene dysregulation affecting lipid metabolism, inflammation, and fibrosis, while proteomics has identified altered protein expression patterns and potential biomarkers. Metabolomic and lipidomic analyses have further uncovered significant changes in various metabolites and lipid species, including ceramides, sphingomyelins, phospholipids, and bile acids, underscoring the central role of lipid dysregulation in MASH. These multi-omics findings have been leveraged to develop novel diagnostic models, some incorporating machine learning algorithms, with improved accuracy compared to traditional methods. Further research is needed to validate these findings, explore the complex interplay between different omics layers, and translate these discoveries into clinically useful tools for improved MASH diagnosis and prognosis.
Collapse
Affiliation(s)
- Yilong Chen
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; (Y.C.); (S.B.)
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Shuixiu Bian
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; (Y.C.); (S.B.)
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Jiamei Le
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; (Y.C.); (S.B.)
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| |
Collapse
|
22
|
Zhao T, Chi Z, Wang D. Versatility of gasdermin D beyond pyroptosis. Trends Cell Biol 2025:S0962-8924(25)00061-3. [PMID: 40121145 DOI: 10.1016/j.tcb.2025.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/14/2025] [Accepted: 02/21/2025] [Indexed: 03/25/2025]
Abstract
Gasdermin D (GSDMD) has garnered significant attention primarily for the pore-forming role of its p30 N-terminal fragment (NT-p30) generated during pyroptosis, a proinflammatory form of cell death. However, emerging evidence suggests that the formation of GSDMD-NT pores is reversible, and the activation of GSDMD does not necessarily lead to pyroptosis. Instead, this process may take part either in other forms of cell death, or in various state changes of living cells, including (i) inflammation regulation, (ii) endolysosomal pathway rewiring, (iii) granule exocytosis, (iv) type II immunity, (v) food tolerance maintenance, and (vi) temporary permeability alteration. This review explores the latest insights into the involvement of GSDMD in cell death and homeostasis maintenance, aiming to underscore the pleiotropic nature of GSDMD.
Collapse
Affiliation(s)
- Tianming Zhao
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 310058, China
| | - Zhexu Chi
- Center for Regeneration and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Yiwu 322000, China.
| | - Di Wang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 310058, China.
| |
Collapse
|
23
|
Zhao Z, Zhu Y, Wan D. Exercise and tissue fibrosis: recent advances in therapeutic potential and molecular mechanisms. Front Endocrinol (Lausanne) 2025; 16:1557797. [PMID: 40182630 PMCID: PMC11965137 DOI: 10.3389/fendo.2025.1557797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Tissue fibrosis represents an aberrant repair process, occurring because of prolonged injury, sustained inflammatory response, or metabolic disorders. It is characterized by an excessive accumulation of extracellular matrix (ECM), resulting in tissue hardening, structural remodeling, and loss of function. This pathological phenomenon is a common feature in the end stage of numerous chronic diseases. Despite the advent of novel therapeutic modalities, including antifibrotic agents, these have only modest efficacy in reversing established fibrosis and are associated with adverse effects. In recent years, a growing body of research has demonstrated that exercise has significant benefits and potential in the treatment of tissue fibrosis. The anti-fibrotic effects of exercise are mediated by multiple mechanisms, including direct inhibition of fibroblast activation, reduction in the expression of pro-fibrotic factors such as transforming growth factor-β (TGF-β) and slowing of collagen deposition. Furthermore, exercise has been demonstrated to assist in maintaining the dynamic equilibrium of tissue repair, thereby indirectly reducing tissue damage and fibrosis. It can also help maintain the dynamic balance of tissue repair by improving metabolic disorders, exerting anti-inflammatory and antioxidant effects, regulating cellular autophagy, restoring mitochondrial function, activating stem cell activity, and reducing cell apoptosis, thereby indirectly alleviating tissue. This paper presents a review of the therapeutic potential of exercise and its underlying mechanisms for the treatment of a range of tissue fibrosis, including cardiac, pulmonary, renal, hepatic, and skeletal muscle. It offers a valuable reference point for non-pharmacological intervention strategies for the comprehensive treatment of fibrotic diseases.
Collapse
Affiliation(s)
- Zheng Zhao
- School of Physical Education, Anyang Normal University, Anyang, Henan, China
| | - Yongjia Zhu
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Dongfeng Wan
- School of Health, Shanghai Normal University Tianhua College, Shanghai, China
| |
Collapse
|
24
|
Wei R, Liu W, Yuan C, Zhang C, Sang Z, Ma Q. Editorial: Preventing and treating liver diseases: medicinal and food plants, their metabolites as potential options. Front Pharmacol 2025; 16:1577547. [PMID: 40170722 PMCID: PMC11959060 DOI: 10.3389/fphar.2025.1577547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 03/03/2025] [Indexed: 04/03/2025] Open
Affiliation(s)
- Rongrui Wei
- College of Medicine and Health Science & School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
| | - Wenmin Liu
- College of Chemistry and Pharmaceutical Engineering, Nanyang Normal University, Nanchang, China
| | - Chunsu Yuan
- Tang Center for Herbal Medicine Research, Department of Anesthesia and Critical Care, Committee On Clinical Pharmacology and Pharmacogenomics, Pritzker School of Medicine, The University of Chicago, Chicago, IL, United States
| | - Chunlei Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhipei Sang
- School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Qinge Ma
- College of Medicine and Health Science & School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
| |
Collapse
|
25
|
Nakamura T, Masuda A, Nakano D, Amano K, Sano T, Nakano M, Kawaguchi T. Pathogenic Mechanisms of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD)-Associated Hepatocellular Carcinoma. Cells 2025; 14:428. [PMID: 40136677 PMCID: PMC11941585 DOI: 10.3390/cells14060428] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/25/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer and the third leading cause of cancer deaths worldwide. The etiology of HCC has now dramatically changed from viral hepatitis to metabolic dysfunction-associated steatotic liver disease (MASLD). The main pathogenesis of MASLD-related HCC is the hepatic lipid accumulation of hepatocytes, which causes chronic inflammation and the subsequent progression of hepatic fibrosis. Chronic hepatic inflammation generates oxidative stress and DNA damage in hepatocytes, which contribute to genomic instability, resulting in the development of HCC. Several metabolic and molecular pathways are also linked to chronic inflammation and HCC in MASLD. In particular, the MAPK and PI3K-Akt-mTOR pathways are upregulated in MASLD, promoting the survival and proliferation of HCC cells. In addition, MASLD has been reported to enhance the development of HCC in patients with chronic viral hepatitis infection. Although there is no approved medication for MASLD besides resmetirom in the USA, there are some preventive strategies for the onset and progression of HCC. Sodium-glucose cotransporter-2 (SGLT2) inhibitor, a class of medications, has been reported to exert anti-tumor effects on HCC by regulating metabolic reprogramming. Moreover, CD34-positive cell transplantation improves hepatic fibrosis by promoting intrahepatic angiogenesis and supplying various growth factors. Furthermore, exercise improves MASLD through an increase in energy consumption as well as changes in chemokines and myokines. In this review, we summarize the recent progress made in the pathogenic mechanisms of MASLD-associated HCC. Furthermore, we introduced new therapeutic strategies for preventing the development of HCC based on the pathogenesis of MASLD.
Collapse
Affiliation(s)
- Toru Nakamura
- Division of Gastroenterology, Department of Medicine, School of Medicine, Kurume University, Kurume 830-0011, Japan; (T.N.); (A.M.); (D.N.); (K.A.); (T.S.); (M.N.)
- Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, 67 Asahi-Machi, Kurume 830-0011, Japan
| | - Atsutaka Masuda
- Division of Gastroenterology, Department of Medicine, School of Medicine, Kurume University, Kurume 830-0011, Japan; (T.N.); (A.M.); (D.N.); (K.A.); (T.S.); (M.N.)
- Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, 67 Asahi-Machi, Kurume 830-0011, Japan
| | - Dan Nakano
- Division of Gastroenterology, Department of Medicine, School of Medicine, Kurume University, Kurume 830-0011, Japan; (T.N.); (A.M.); (D.N.); (K.A.); (T.S.); (M.N.)
- Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, 67 Asahi-Machi, Kurume 830-0011, Japan
| | - Keisuke Amano
- Division of Gastroenterology, Department of Medicine, School of Medicine, Kurume University, Kurume 830-0011, Japan; (T.N.); (A.M.); (D.N.); (K.A.); (T.S.); (M.N.)
- Fukuoka Consulting and Support Center for Liver Diseases, Kurume 830-0011, Japan
| | - Tomoya Sano
- Division of Gastroenterology, Department of Medicine, School of Medicine, Kurume University, Kurume 830-0011, Japan; (T.N.); (A.M.); (D.N.); (K.A.); (T.S.); (M.N.)
- Fukuoka Consulting and Support Center for Liver Diseases, Kurume 830-0011, Japan
| | - Masahito Nakano
- Division of Gastroenterology, Department of Medicine, School of Medicine, Kurume University, Kurume 830-0011, Japan; (T.N.); (A.M.); (D.N.); (K.A.); (T.S.); (M.N.)
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, School of Medicine, Kurume University, Kurume 830-0011, Japan; (T.N.); (A.M.); (D.N.); (K.A.); (T.S.); (M.N.)
- Liver Cancer Research Division, Research Center for Innovative Cancer Therapy, Kurume University, 67 Asahi-Machi, Kurume 830-0011, Japan
| |
Collapse
|
26
|
Baumert BO, Maretti-Mira AC, Walker DI, Li Z, Stratakis N, Wang H, Zhao Y, Fischer FC, Jia Q, Valvi D, Bartell SM, Chen C, Inge T, Ryder J, Jenkins T, Sisley S, Xanthakos S, Kleiner DE, Kohli R, Rock S, Eckel SP, La Merrill MA, Aung MM, Salomon MP, McConnell R, Goodrich J, Conti DV, Golden-Mason L, Chatzi L. Integrated Spheroid-to-Population Framework for Evaluating PFHpA-Associated Metabolic Dysfunction and Steatotic Liver Disease. RESEARCH SQUARE 2025:rs.3.rs-5960979. [PMID: 40092438 PMCID: PMC11908348 DOI: 10.21203/rs.3.rs-5960979/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
The rising prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD), particularly among pediatric populations, requires identification of modifiable risk factors to control disease progression. Per- and polyfluoroalkyl substances (PFAS) have emerged as potential contributors to liver damage; however, their role in the etiology of MASLD remains underexplored. This study aimed to bridge the gap between human epidemiological data and in vitro experimental findings to elucidate the effect of perfluoroheptanoic acid (PFHpA), a short chain, unregulated PFAS congener on MASLD development. Our analysis of the Teen-LABS cohort, a national multi-site study on obese adolescents undergoing bariatric surgery, revealed that doubling of PFHpA plasma levels was associated with an 80% increase in MASLD risk (OR, 1.8; 95% CI: 1.3-2.5) based on liver biospies. To further investigate the underlying mechanisms, we used 3D human liver spheroids and single-cell transcriptomics to assess the effect of PFHpA on hepatic metabolism. Integrative analysis identified dysregulation of common pathways in both human and spheroid models, particularly those involved in innate immunity, inflammation, and lipid metabolism. We applied the latent unknown clustering with integrated data (LUCID) model to assess associations between PFHpA exposure, multiomic signatures, and MASLD risk. Our results identified a proteome profile with significantly higher odds of MASLD (OR = 7.1), whereas a distinct metabolome profile was associated with lower odds (OR = 0.51), highlighting the critical role of protein dysregulation in disease pathogenesis. A translational framework was applied to uncover the molecular mechanisms of PFAS-induced MASLD in a cohort of obese adolescents. Identifying key molecular mechanisms for PFAS-induced MASLD can guide the development of targeted prevention and treatment.
Collapse
Affiliation(s)
- Brittney O. Baumert
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ana C. Maretti-Mira
- USC Research Center for Liver Diseases, Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Douglas I. Walker
- Gangarosa Department of Environmental Health, Rollins School of Public Health, 1518 Clifton Road, NE, Atlanta, GA, United States
| | - Zhenjiang Li
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Nikos Stratakis
- Barcelona Institute for Global Health, ISGlobal, Dr. Aiguader 88, 08003, Barcelona, Spain
| | - Hongxu Wang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Yinqi Zhao
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Fabian Christoph Fischer
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI 02881, United States
| | - Qiran Jia
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Damaskini Valvi
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Scott M. Bartell
- Department of Environmental and Occupational Health, University of California, Irvine, Irvine, CA, United States
| | - Carmen Chen
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Thomas Inge
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
| | - Justin Ryder
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States
| | - Todd Jenkins
- Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Stephanie Sisley
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Stavra Xanthakos
- Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - David E. Kleiner
- National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, MD, United States
| | - Rohit Kohli
- Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Sarah Rock
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Sandrah P. Eckel
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Michele A. La Merrill
- Department of Environmental Toxicology, University of California, Davis, CA, United States
| | - Max M. Aung
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Matthew P. Salomon
- USC Research Center for Liver Diseases, Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Rob McConnell
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Jesse Goodrich
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - David V. Conti
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Lucy Golden-Mason
- USC Research Center for Liver Diseases, Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Lida Chatzi
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
27
|
Zhang S, Dong H, Jin X, Sun J, Li Y. The multifaceted roles of macrophages in the transition from hepatitis to hepatocellular carcinoma: From mechanisms to therapeutic strategies. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167676. [PMID: 39828046 DOI: 10.1016/j.bbadis.2025.167676] [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: 11/28/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Macrophages are central to the progression from hepatitis to hepatocellular carcinoma (HCC), with their remarkable plasticity and ability to adapt to the changing liver microenvironment. Chronic inflammation, fibrosis, and ultimately tumorigenesis are driven by macrophage activation, making them key regulators of liver disease progression. This review explores the diverse roles of macrophages in the transition from hepatitis to HCC. In the early stages of hepatitis, macrophages are essential for pathogen clearance and tissue repair. However, chronic activation leads to prolonged inflammation, which exacerbates liver damage and promotes fibrosis. As the disease progresses to liver fibrosis, macrophages interact with hepatic stellate cells, fostering a pro-tumorigenic microenvironment that supports HCC development. In hepatocarcinogenesis, macrophages contribute to tumor initiation, growth, metastasis, immune evasion, cancer stem cell maintenance, and angiogenesis. Their functional plasticity enables them to adapt to the tumor microenvironment, thereby promoting tumor progression and resistance to therapy. Targeting macrophages represents a promising strategy for preventing and treating HCC. Therapeutic approaches, including reprogramming macrophage phenotypes to enhance anti-tumor immunity, blocking macrophage recruitment and activation, and utilizing nanoparticle-based drug delivery systems, may provide new avenues for combating HCC by modulating macrophage functions and tumor microenvironment dynamics.
Collapse
Affiliation(s)
- Shuairan Zhang
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang, PR China
| | - Hang Dong
- Phase I Clinical Trials Center, The People's Hospital of China Medical University, Shenyang, PR China
| | - Xiuli Jin
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang, PR China
| | - Jing Sun
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang, PR China
| | - Yiling Li
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang, PR China.
| |
Collapse
|
28
|
Tepebaşi MY, Aşci H, Selçuk E, Kolay Ö, Milletsever A, Şahin M, Özmen Ö. Investigation of the Theranekron Ameliorative Effect on Sepsis-Induced Hepatotoxicity via Inflammation and Oxidative Stress Pathways. Cell Biochem Biophys 2025:10.1007/s12013-025-01699-1. [PMID: 39992615 DOI: 10.1007/s12013-025-01699-1] [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] [Accepted: 02/10/2025] [Indexed: 02/26/2025]
Abstract
Lipopolysaccharide (LPS)-induced inflammatory liver injury can cause significant tissue damage and apoptosis. Homeopathic formulations such as Tarantula cubensis venom show potential in regulating inflammation. This study's objective was to assess theranecron's (THE) impact on inflammation and oxidative stress in a model of liver injury caused by lipopolysaccharide (LPS). Wistar albino female rats were used in this investigation, and they were split up into four groups of eight each: Control, LPS, LPS+THE, and THE. Single-dose treatments were administered to the respective groups on the same day. Liver tissues were collected 6 h after LPS treatment for histopathological, immunohistochemical, biochemical, and genetic evaluations. Total antioxidant status (TAS) was lower, total oxidant status (TOS) and oxidative stress index (OSI) were higher, and the LPS group had higher levels of interleukin 6 (IL-6), tumor necrosis factor α (TNF-α), and macrophage antigen-1 (CD11B). Significant liver damage was also seen in this group, as evidenced by elevated levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) and decreased albumin. Nuclear factor erythroid 2-related factor 2 (Nrf2), Sirtuin 1 (SIRT1), heme oxygenase 1 (HO-1), kelch-like ECH-associated protein 1 (Keap1), and glutathione peroxidase 4 (GPx4) were all found to be downregulated by gene expression analysis. However, THE therapy was shown to reverse all of these findings in the LPS+THE group. The THE group similarly maintained baseline levels of these markers and showed no adverse effects. In conclusion, Theranekron showed hepatoprotective effects in LPS-induced liver injury by reducing oxidative stress and inflammation and regulating antioxidant gene expression, possibly through IL-6 and TNF-α.
Collapse
Affiliation(s)
| | - Halil Aşci
- Department of Pharmacology, Faculty of Medicine, Süleyman Demirel University, Isparta, Türkiye
| | - Esma Selçuk
- Department of Medical Biology, Faculty of Medicine, Süleyman Demirel University, Isparta, Türkiye
| | - Öznur Kolay
- Department of Medical Pharmacology, Institute of Health Sciences, Süleyman Demirel University, Isparta, Türkiye
| | - Adem Milletsever
- Department of Pathology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Türkiye
| | - Melda Şahin
- Department of Pathology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Türkiye
| | - Özlem Özmen
- Department of Pathology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Türkiye
| |
Collapse
|
29
|
Taru V, Reiberger T. Reply to: "Pyroptosis in liver failure: A twisted firestarter". J Hepatol 2025; 82:e93-e94. [PMID: 39442890 DOI: 10.1016/j.jhep.2024.10.021] [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: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024]
Affiliation(s)
- Vlad Taru
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Iuliu Hatieganu University of Medicine and Pharmacy, 4th Dept. of Internal Medicine and "Octavian Fodor" Regional Institute of Gastroenterology and Hepatology, Hepatology Department, Cluj-Napoca, Romania; Vienna Hepatic Experimental Hemodynamic (HEPEX) Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Christian Doppler Lab for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria.
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Vienna Hepatic Experimental Hemodynamic (HEPEX) Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Christian Doppler Lab for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|
30
|
Mehta G, Jalan R. Pyroptosis in liver failure: A twisted firestarter. J Hepatol 2025; 82:e91-e92. [PMID: 39218224 DOI: 10.1016/j.jhep.2024.08.023] [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: 07/25/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Affiliation(s)
- Gautam Mehta
- Institute for Liver and Digestive Health, University College London, UK; Royal Free London NHS Foundation Trust, UK.
| | - Rajiv Jalan
- Institute for Liver and Digestive Health, University College London, UK; Royal Free London NHS Foundation Trust, UK
| |
Collapse
|
31
|
Dan L, Li X, Chen S, You X, Wang D, Wang T, Li J, Liu W, Mu J, Feng Q. Protective role of ginsenoside Rg1 in the dynamic progression of liver injury to fibrosis: a preclinical meta-analysis. Front Pharmacol 2025; 16:1512184. [PMID: 39936090 PMCID: PMC11810943 DOI: 10.3389/fphar.2025.1512184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/02/2025] [Indexed: 02/13/2025] Open
Abstract
Background The pathological progression from liver injury to fibrosis is a hallmark of liver disease, with no effective strategies to halt this transition. Ginsenoside Rg1 has demonstrated a range of hepatoprotective properties; however, systematic preclinical evidence supporting its therapeutic potential for liver injury and fibrosis remains limited. Purpose. This study evaluated the efficacy and underlying mechanisms of ginsenoside Rg1 in animal models of liver injury and fibrosis, and providing a basis for future clinical investigation. Methods A systematic review was conducted on preclinical studies published in PubMed, Web of Science, and Embase databases up to 1 August 2024, adhereing to rigorous quality standards. The methodological quality was assessed using SYRCLE's risk of bias tool. Meta-analysis and subgroup analysis were performed using Revman 5.4 software, while publication bias was evaluated through funnel plots and Egger's test in STATA 15.0 software. Additionally, a time-dose interval curve was utilized to assess the dose-response relationship and identify the effective dose of ginsenoside Rg1 for treating liver injury and fibrosis. Results Twenty-four trials involving 423 animals were included. The findings indicated that ginsenoside Rg1 significantly improved liver function markers (ALT and AST), reduced pathological indicators associated with liver injury and fibrosis, and lowered liver fibrosis-related markers (α-SMA, HYP, and PCIII). Furthermore, it exhibited beneficial effects on mechanistic indicators of inflammation, oxidative stress, and apoptosis, compared to the control group (P < 0.05). Time-dose interval analysis revealed that the effective dose range of ginsenoside Rg1 was between 4 and 800 mg/kg/d. Conclusion Rg1 at a dose of 4-800 mg/kg/d mitigates the progression of liver injury to fibrosis via anti-inflammatory, antioxidative, and anti-apoptotic pathways. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD 42024557878.
Collapse
Affiliation(s)
- Lijuan Dan
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiuyan Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuanglan Chen
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaojie You
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dong Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tianyuan Wang
- Traditional Chinese Medicine Department, 363 Hospital of Chengdu, Chengdu, China
| | - Jia Li
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenping Liu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Mu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Quansheng Feng
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
32
|
Liu MK, Tang JJ, Li H, Chen XY, Cai JL, Lin GY, Chen KY, Liu ZP, Ji XF, Yang ZJ, Li Z. Artemisitene ameliorates carbon tetrachloride-induced liver fibrosis by inhibiting NLRP3 inflammasome activation and modulating immune responses. Int Immunopharmacol 2025; 146:113818. [PMID: 39681062 DOI: 10.1016/j.intimp.2024.113818] [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: 09/07/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024]
Abstract
Artemisitene (ATT), an artemisinin (ART) analog retaining the endoperoxide moiety and incorporating an additional α, β-unsaturated carbonyl structure, exhibits enhanced biological activities. However, its therapeutic effects on liver fibrosis remain unclear. In this study, we demonstrated that ATT significantly alleviated liver inflammation and fibrosis induced by carbon tetrachloride (CCL4) in mice. ATT treatment markedly reduced the count of neutrophils in the liver, as well as macrophages in both the liver and spleen. Additionally, the frequencies of Th2 and Th17 cells were significantly lowered, while Th1 cells frequency and the Th1/Th2 index were notably increased. The frequency of ILC2 cells, correlated with ST2 and IL-33 expression levels, was also significantly lowered. Consistently, ATT inhibited NLRP3 inflammasome activation, which was positively associated with AST and ALT levels, and with the count of Neutrophils, macrophages, and ILC2 cells, but negatively correlated with Th1frequeny. Furthermore, liver fibrosis severity showed a significant positive correlation with neutrophil and Th17 cell counts in the liver, and a negative correlation with Th1 cell count and the Th1/Th2 index. Therefore, ATT alleviated CCL4-induced mice liver fibrosis through NLRP3 inflammasome inhibition and immunomodulation.
Collapse
Affiliation(s)
- Meng-Ke Liu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Juan-Juan Tang
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Hao Li
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xu-Yang Chen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Jun-Ling Cai
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Gui-Ying Lin
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Kan-Yao Chen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; Department of Clinical Laboratory, Guangdong Provincial People's Hospital Zhuhai Hospital (Zhuhai Golden Bay Center Hospital), Zhuhai, China
| | - Zhi-Peng Liu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; The Affiliated Qingyuan Hospital of Guangzhou Medical University (Qingyuan People's Hospital), Qingyuan, China
| | - Xiao-Fang Ji
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Zhong-Jin Yang
- School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin 300384, China.
| | - Zi Li
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou 510260, China.
| |
Collapse
|
33
|
Ortega-Ribera M, Zhuang Y, Brezani V, Joshi RS, Zsengeller Z, Nagesh PT, Datta A, Szabo G. Gasdermin D deletion prevents liver injury and exacerbates extrahepatic damage in a murine model of alcohol-induced ACLF. EGASTROENTEROLOGY 2025; 3:e100151. [PMID: 40134717 PMCID: PMC11934631 DOI: 10.1136/egastro-2024-100151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 02/27/2025] [Indexed: 03/27/2025]
Abstract
Background Gasdermin D (GSDM-D), a key executor of pyroptosis, is increased in various liver diseases and contributes to disease progression. Alcohol induces inflammasome activation and cell death, which are both linked to GSDM-D activation. However, its role in alcohol-induced acute-on-chronic liver failure (ACLF) remains unclear. Methods ACLF was induced in GSDM-D-deficient or wild-type (WT) mice by 28-day bile duct ligation surgery plus a single 5 g/kg alcohol binge leading to acute decompensation. Nine hours after the alcohol binge, blood, liver, kidney and cerebellum specimens were collected for analysis. Results Active GSDM-D was significantly increased in humans and mice ACLF livers compared with both healthy controls and cirrhotic livers. GSDM-D-deficient mice with ACLF showed decreased inflammation, neutrophil infiltration and fibrosis in the liver, together with a reduction in pyroptotic, apoptotic and necroptotic death, compared with WT ACLF mice. Notably, GSDM-D-deficient mice also showed decreased liver regeneration and hepatocyte function. This was associated with an increase in senescence and expression of stem-like/cholangiocyte markers in the liver. Interestingly, in the kidney, GSDM-D-deficient mice showed an increase in histopathological damage score, decreased function and increased expression of necroptosis-related genes. In the cerebellum, GSDM-D deficiency increased the expression of neuroinflammation markers, astrocyte activation and apoptosis-related genes. Conclusion Our data indicate that GSDM-D deficiency has organ-specific effects in ACLF. While it reduces inflammation, neutrophil activation, cell death and fibrosis in the liver, GSDM-D deficiency impairs the synthetic function and increases senescence in hepatocytes. GSDM-D deficiency also increases kidney injury and neuroinflammation in ACLF.
Collapse
Affiliation(s)
- Martí Ortega-Ribera
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Yuan Zhuang
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Veronika Brezani
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Radhika S Joshi
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Zsuzsanna Zsengeller
- Department of Medicine, Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Prashanth Thevkar Nagesh
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Aditi Datta
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Gyongyi Szabo
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
34
|
Han J, Wu J, Kou WT, Xie LN, Tang YL, Zhi DL, Li P, Chen DQ. New insights into SUMOylation and NEDDylation in fibrosis. Front Pharmacol 2024; 15:1476699. [PMID: 39697538 PMCID: PMC11652140 DOI: 10.3389/fphar.2024.1476699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024] Open
Abstract
Fibrosis is the outcome of any abnormal tissue repair process that results in normal tissue replacement with scar tissue, leading to persistent tissue damage and cellular injury. During the process of fibrosis, many cytokines and chemokines are involved, and their activities are controlled by post-translational modifications, especially SUMOylation and NEDDylation. Both these modifications entail a three-step process of activation, conjugation, and ligation that involves three kinds of enzymes, namely, E1 activating, E2 conjugating, and E3 ligase enzymes. SUMOylation participates in organ fibrosis by modulating FXR, PML, TGF-β receptor I, Sirt3, HIF-1α, and Sirt1, while NEDDylation influences organ fibrosis by regulating cullin3, NIK, SRSF3, and UBE2M. Further investigations exhibit the therapeutic potentials of SUMOylation/NEDDylation activators and inhibitors against organ fibrosis, especially ginkgolic acid in SUMOylation and MLN4924 in NEDDylation. These results demonstrate the therapeutic effects of SUMOylation and NEDDylation against organ fibrosis and highlight their activators as well as inhibitors as potential candidates. In the future, deeper investigations of SUMOylation and NEDDylation are needed to identify novel substrates against organ fibrosis; moreover, clinical investigations are needed to determine the therapeutic effects of their activators and inhibitors that can benefit patients. This review highlights that SUMOylation and NEDDylation function as potential therapeutic targets for organ fibrosis.
Collapse
Affiliation(s)
- Jin Han
- Northwest University Chang An Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi’an, Shaanxi, China
- Department of Nephrology, Chang An District Hospital, Xi’an, Shaanxi, China
| | - Jun Wu
- School of Pharmacy, Shandong College of Traditional Chinese Medicine, Yantai, Shandong, China
| | - Wen-Tao Kou
- Northwest University Chang An Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi’an, Shaanxi, China
- Department of Nephrology, Chang An District Hospital, Xi’an, Shaanxi, China
| | - Li-Na Xie
- Northwest University Chang An Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi’an, Shaanxi, China
- Department of Nephrology, Chang An District Hospital, Xi’an, Shaanxi, China
| | - Ya-Li Tang
- Northwest University Chang An Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi’an, Shaanxi, China
- Department of Nephrology, Chang An District Hospital, Xi’an, Shaanxi, China
| | - Da-Long Zhi
- Northwest University Chang An Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi’an, Shaanxi, China
- Department of Nephrology, Chang An District Hospital, Xi’an, Shaanxi, China
| | - Ping Li
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Dan-Qian Chen
- Northwest University Chang An Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi’an, Shaanxi, China
- Department of Nephrology, Chang An District Hospital, Xi’an, Shaanxi, China
| |
Collapse
|
35
|
FAGOONEE S, PANELLA R. MASHing up molecular imaging and biomarkers for improved diagnosis of metabolic dysfunction-associated steatotic liver disease. MINERVA BIOTECHNOLOGY AND BIOMOLECULAR RESEARCH 2024; 36. [DOI: 10.23736/s2724-542x.24.03224-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/10/2025]
|
36
|
Sabnis RW. Novel Substituted Benzimidazole Compounds as TLR9 Inhibitors for Treating Fibrotic Diseases, Particularly Idiopathic Pulmonary Fibrosis. ACS Med Chem Lett 2024; 15:1804-1805. [PMID: 39563824 PMCID: PMC11571016 DOI: 10.1021/acsmedchemlett.4c00473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Indexed: 11/21/2024] Open
Abstract
Provided herein are novel substituted benzimidazole compounds as TLR9 inhibitors, pharmaceutical compositions, use of such compounds in treating fibrotic diseases, particularly idiopathic pulmonary fibrosis, and processes for preparing such compounds.
Collapse
Affiliation(s)
- Ram W Sabnis
- Smith, Gambrell & Russell LLP, 1105 W. Peachtree Street NE, Suite 1000, Atlanta, Georgia 30309, United States
| |
Collapse
|
37
|
Olivera-Salazar R, Sánchez A, Herrera B, García-Sáez J, Vega-Clemente L, Villarejo Campos P, García-Olmo D, García-Arranz M. A Protocol for the Isolation of Oval Cells without Preconditioning. Int J Mol Sci 2024; 25:10497. [PMID: 39408831 PMCID: PMC11477217 DOI: 10.3390/ijms251910497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Oval cells (OCs) is the name of hepatic progenitor cells (HPCs) in rodents. They are a small population of cells in the liver with the remarkable ability to proliferate and regenerate hepatocytes and cholangiocytes in response to acute liver damage. Isolating OCs generally requires a pretreatment with special diets, chemicals, and/or surgery to induce hepatic damage and OC proliferation in mice. Unfortunately, these pretreatments are not only painful for the mice but also increase the cost of the assays, and the effects on the different organs as well as on various liver cells are still unclear. Therefore, the search for a protocol to obtain OCs without prior liver damage is mandatory. In our study, we present a protocol to isolate murine OCs from healthy liver (HL-OCs) and compare them with OCs isolated from mice pretreated with 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC-OCs). Our results demonstrated that cells derived from untreated mice exhibited similar behavior to those from treated mice in terms of surface marker expression, proliferation, and differentiation capacity. Therefore, given the impracticability of isolating human cells with prior hepatotoxic treatment, our model holds promise for enabling the isolation of progenitor cells from human tissue in the future. This advancement could prove invaluable for translational medicine in the understanding and treatment of liver diseases.
Collapse
Affiliation(s)
- Rocío Olivera-Salazar
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Avda. Reyes Católicos, 2, 28040 Madrid, Spain; (R.O.-S.); (L.V.-C.); (D.G.-O.)
| | - Aránzazu Sánchez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the “Hospital Clínico San Carlos” (IdISSC), 28040 Madrid, Spain; (A.S.); (B.H.); (J.G.-S.)
- Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBEREHD-ISCIII), 28029 Madrid, Spain
| | - Blanca Herrera
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the “Hospital Clínico San Carlos” (IdISSC), 28040 Madrid, Spain; (A.S.); (B.H.); (J.G.-S.)
- Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBEREHD-ISCIII), 28029 Madrid, Spain
| | - Juan García-Sáez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the “Hospital Clínico San Carlos” (IdISSC), 28040 Madrid, Spain; (A.S.); (B.H.); (J.G.-S.)
- Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBEREHD-ISCIII), 28029 Madrid, Spain
| | - Luz Vega-Clemente
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Avda. Reyes Católicos, 2, 28040 Madrid, Spain; (R.O.-S.); (L.V.-C.); (D.G.-O.)
| | - Pedro Villarejo Campos
- Department of Surgery, Fundación Jiménez Díaz University Hospital (FJD), 28040 Madrid, Spain;
| | - Damián García-Olmo
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Avda. Reyes Católicos, 2, 28040 Madrid, Spain; (R.O.-S.); (L.V.-C.); (D.G.-O.)
- Department of Surgery, Fundación Jiménez Díaz University Hospital (FJD), 28040 Madrid, Spain;
- Department of Surgery, Universidad Autónoma de Madrid, 28034 Madrid, Spain
| | - Mariano García-Arranz
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Avda. Reyes Católicos, 2, 28040 Madrid, Spain; (R.O.-S.); (L.V.-C.); (D.G.-O.)
- Department of Surgery, Universidad Autónoma de Madrid, 28034 Madrid, Spain
| |
Collapse
|