1
|
Wang D, Miao J, Zhang L, Zhang L. Research advances in the diagnosis and treatment of MASLD/MASH. Ann Med 2025; 57. [DOI: 10.1080/07853890.2024.2445780] [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: 11/21/2023] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 01/06/2025] Open
Affiliation(s)
- Dekai Wang
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jinxian Miao
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lihua Zhang
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lin Zhang
- Department of General Practice, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
2
|
Tanaka M, Akiyama Y, Mori K, Hosaka I, Endo K, Ogawa T, Sato T, Suzuki T, Yano T, Ohnishi H, Hanawa N, Furuhashi M. Machine learning-based analyses of contributing factors for the development of hypertension: a comparative study. Clin Exp Hypertens 2025; 47:2449613. [PMID: 39773295 DOI: 10.1080/10641963.2025.2449613] [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: 02/22/2024] [Revised: 11/25/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
OBJECTIVES Sufficient attention has not been given to machine learning (ML) models using longitudinal data for investigating important predictors of new onset of hypertension. We investigated the predictive ability of several ML models for the development of hypertension. METHODS A total of 15 965 Japanese participants (men/women: 9,466/6,499, mean age: 45 years) who received annual health examinations were randomly divided into a training group (70%, n = 11,175) and a test group (30%, n = 4,790). The predictive abilities of 58 candidates including fatty liver index (FLI), which is calculated by using body mass index, waist circumference and levels of γ-glutamyl transferase and triglycerides, were investigated by statistics analogous to the area under the curve (AUC) in receiver operating characteristic curve analyses using ML models including logistic regression, random forest, naïve Bayes, extreme gradient boosting and artificial neural network. RESULTS During a 10-year period (mean period: 6.1 years), 2,132 subjects (19.1%) in the training group and 917 subjects (19.1%) in the test group had new onset of hypertension. Among the 58 parameters, systolic blood pressure, age and FLI were identified as important candidates by random forest feature selection with 10-fold cross-validation. The AUCs of ML models were 0.765-0.825, and discriminatory capacity was significantly improved in the artificial neural network model compared to that in the logistic regression model. CONCLUSIONS The development of hypertension can be simply and accurately predicted by each ML model using systolic blood pressure, age and FLI as selected features. By building multiple ML models, more practical prediction might be possible.
Collapse
Affiliation(s)
- Marenao Tanaka
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Tanaka Medical Clinic, Yoichi, Japan
| | - Yukinori Akiyama
- Department of Neurosurgery, Sapporo Medical University, Sapporo, Japan
| | - Kazuma Mori
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Japan
| | - Itaru Hosaka
- Department of Cardiovascular Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Keisuke Endo
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshifumi Ogawa
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tatsuya Sato
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toru Suzuki
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Natori Toru Internal Medicine and Diabetes Clinic, Natori, Japan
| | - Toshiyuki Yano
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hirofumi Ohnishi
- Department of Public Health, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Nagisa Hanawa
- Department of Health Checkup and Promotion, Keijinkai Maruyama Clinic, Sapporo, Japan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
3
|
Yuan M, Yin Z, Wang Z, Xiong Z, Chen P, Yao L, Liu P, Sun M, Shu K, Li L, Jiang Y. Modification of MSCs with aHSCs-targeting peptide pPB for enhanced therapeutic efficacy in liver fibrosis. Biomaterials 2025; 321:123295. [PMID: 40188718 DOI: 10.1016/j.biomaterials.2025.123295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 02/22/2025] [Accepted: 03/26/2025] [Indexed: 05/04/2025]
Abstract
Mesenchymal stem cells (MSCs) hold significant therapeutic potential for liver fibrosis but face translational challenges due to suboptimal homing efficiency and poor retention at injury sites. Activated hepatic stellate cells (aHSCs), the primary drivers of fibrogenesis, overexpress platelet-derived growth factor receptor-beta (PDGFRB), a validated therapeutic target in liver fibrosis. Here, we engineered pPB peptide-functionalized MSCs (pPB-MSCs) via hydrophobic insertion of DMPE-PEG-pPB (DPP) into the MSC membrane, creating a targeted "MSC-pPB-aHSC" delivery system. Our findings demonstrated that pPB modification preserved MSC viability, differentiation potential, and paracrine functions. pPB-MSCs exhibited higher binding affinity to TGF-β1-activated HSCs in vitro and greater hepatic accumulation in TAA-induced fibrotic mice, as quantified by in vivo imaging. Moreover, pPB-MSCs attenuated collagen deposition, suppressed α-SMA+ HSCs, and restored serum ALT/AST levels to near-normal ranges. Mechanistically, pPB-MSCs promoted hepatocyte regeneration via HGF upregulation, inhibited epithelial-mesenchymal transition through TGF-β/Smad pathway suppression, and polarized macrophages toward an M2 phenotype, reducing pro-inflammatory IL-6/TNF-α while elevating anti-inflammatory IL-10. Overall, our study raised a non-genetic MSC surface engineering strategy that synergizes PDGFRB-targeted homing with multifactorial tissue repair, addressing critical barriers in cell therapy for liver fibrosis. By achieving enhanced spatial delivery without compromising MSC functionality, our approach provides a clinically translatable platform for enhancing regenerative medicine outcomes.
Collapse
Affiliation(s)
- Mengqin Yuan
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China; Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Research Center for Medical Imaging in Hubei Province, Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Zhengrong Yin
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zheng Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China
| | - Zhiyu Xiong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China
| | - Ping Chen
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China
| | - Lichao Yao
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China
| | - Pingji Liu
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China
| | - Muhua Sun
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China
| | - Kan Shu
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China
| | - Lanjuan Li
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China.
| | - Yingan Jiang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Wuhan 430000, China.
| |
Collapse
|
4
|
Zhang X, He Y, Long L, Jiang G, Yao T, Cheng X, Peng Z, Hu G, Tao L, Meng J. Mefunidone treats pulmonary fibrosis by targeting SDH to regulate fibro-promoting macrophages. Int Immunopharmacol 2025; 160:114971. [PMID: 40449267 DOI: 10.1016/j.intimp.2025.114971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 05/25/2025] [Accepted: 05/26/2025] [Indexed: 06/03/2025]
Abstract
BACKGROUND AND OBJECTIVE Pulmonary fibrosis, a pathological process where the extracellular matrix overly deposits in lung tissue because of various pathogenic factors, leads to lung structure damage and function decline. Idiopathic pulmonary fibrosis (IPF) has a poor prognosis and high mortality, lacking effective drug treatments. Mefunidone (MFD), a new small-molecule compound, showed therapeutic effects on it in previous studies, but its specific molecular target is unknown. This study aims to clarify MFD's target and its potential mechanism. By exploring this, we hope to offer new insights and potential solutions for treating IPF and improving patients' outcomes. METHODS Mice with pulmonary fibrosis induced by bleomycin (BLM) were used as experimental models. MFD was administered by gavage. The changes in inflammation and fibrosis were evaluated through histopathological examinations. Subsequently, single-cell sequencing technology was used to explore how MFD affects the phenotype of pro-fibrotic macrophages, and verification was carried out in vitro to prove that MFD treats pulmonary fibrosis by influencing the phenotype of pro-fibrotic macrophages. RESULT MFD can inhibit the generation of succinate by binding and inhibiting the activity of succinate dehydrogenase (SDH). MFD can also inhibit the transformation of MMP12+CCL2+ profibrotic macrophages in the BLM pulmonary fibrosis model. Treatment with succinate can induce the transformation of macrophages into MMP12+CCL2+ profibrotic macrophages, and this induction depends on the succinate-specific receptor GPR91. CONCLUSION Our research results have revealed for the first time that MFD can treat pulmonary fibrosis by targeting SDH and regulating the transformation of MMP12+CCL2+ profibrotic macrophages.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China; Central South University, Changsha, China
| | - Yijun He
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China; Central South University, Changsha, China
| | - Lingzhi Long
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China; Central South University, Changsha, China
| | - Guoliang Jiang
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China; Central South University, Changsha, China
| | - Tingting Yao
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China; Central South University, Changsha, China
| | - Xiaoyun Cheng
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China; Central South University, Changsha, China
| | - Zhangzhe Peng
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China; Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Gaoyun Hu
- Central South University, Changsha, China
| | - Lijian Tao
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China; Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Meng
- Department of Pulmonary and Critical Care Medicine, Third Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China; Central South University, Changsha, China.
| |
Collapse
|
5
|
Gu S, Chen C, Wang J, Wang Y, Zhao L, Xiong Z, Zhang H, Deng T, Pan Q, Zheng Y, Li Y. Camellia Japonica Radix modulates gut microbiota and 9(S)-HpODE-mediated ferroptosis to alleviate oxidative stress against MASLD. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156806. [PMID: 40334428 DOI: 10.1016/j.phymed.2025.156806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/04/2025] [Accepted: 04/25/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND Camellia japonica radix (CJR), derived from the root of Camellia japonica L., has the potential to function as an herbal tea substitute for the prevention and intervention of metabolic dysfunction-associated steatotic liver disease (MASLD). It can provide systemic therapeutic benefits, boast a favorable safety profile, facilitate convenient consumption, and support long-term applicability. Despite its potential, research on CJR remains limited. PURPOSE The aim of this study aims is to elucidate the therapeutic mechanisms of CJR in MASLD, thereby providing evidence to support its clinical application. METHODS The therapeutic effects of CJR were evaluated using a water-supplementation model in MASLD mice. Integrated microbiome, transcriptome, proteome, and metabolome analyses were employed to comprehensively explore the mechanisms involved. A drug-target pull-down assay was performed to identify specific protein targets of small molecule metabolites in vitro. Fecal microbiota transplantation in antibiotic-treated ABX mice was conducted to confirm the critical role of gut microbiota and its metabolites. Furthermore, customized medicated feed supplemented with linoleic acid was used to explore the intervention effect of its metabolite, 9(S)-HpODE, as well as to evaluate its dietary intervention potential. RESULTS This present study explicitly elucidates the efficacy of CJR extract in alleviating hepatic inflammation and steatosis in a MASLD model mice, with its pharmacological mechanism associated with gut microbiota, linoleic acid metabolism, and GPX4-mediated ferroptosis. Notably, 9(S)-HpODE was discovered to be a key metabolite of linoleic acid, which could target both KEAP1 and SLC7A11, bidirectionally regulating GPX4-mediated ferroptosis, while acting as a signaling molecule at low doses to induce redox adaptation via oxidative preconditioning, thus ameliorating oxidative stress in MASLD. CONCLUSION Our findings indicate that both CJR and linoleic acid exhibit significant potential as dietary interventions for the management of MASLD, offering promising avenues for future research and clinical application.
Collapse
Affiliation(s)
- Simin Gu
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chong Chen
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Junmin Wang
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanping Wang
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lina Zhao
- Department of Hepatobiliary Diseases, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou, China
| | - Zhekun Xiong
- Department of Spleen, Stomach and Hepatobiliary, Zhongshan Hospital of Traditional Chinese Medicine, Zhongshan, China
| | - Hui Zhang
- Department of Spleen, Stomach and Hepatobiliary, Zhongshan Hospital of Traditional Chinese Medicine, Zhongshan, China
| | - Taoying Deng
- Department of Spleen, Stomach and Hepatobiliary, Zhongshan Hospital of Traditional Chinese Medicine, Zhongshan, China
| | - Qihui Pan
- Department of Gastroenterology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiyuan Zheng
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yong Li
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
6
|
Zambrano-Vásquez OR, Cortés-Camacho F, Castañeda-Sánchez JI, Aréchaga-Ocampo E, Valle-Velázquez E, Cabrera-Angeles JC, Sánchez-Gloria JL, Sánchez-Muñoz F, Arellano-Buendia AS, Sánchez-Lozada LG, Osorio-Alonso H. Update in non-alcoholic fatty liver disease management: role of sodium-glucose cotransporter 2 inhibitors. Life Sci 2025; 372:123638. [PMID: 40246191 DOI: 10.1016/j.lfs.2025.123638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/28/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterized by excessive lipid accumulation in hepatocytes without significant alcohol consumption. It is closely associated with sedentarism, hypercaloric diets, obesity, dyslipidemia, insulin resistance, type 2 diabetes mellitus, and genetic predisposition. NAFLD comprises a spectrum of liver disorders, from simple steatosis to non-alcoholic (NASH) and liver cirrhosis. The complex etiological mechanisms include oxidative stress, inflammation, apoptosis, and fibrosis; therefore, its management is challenging. Sodium-glucose cotransporter type 2 inhibitors (SGLT2i), a class of antidiabetic drugs, have emerged as promising therapeutic agents due to their ability to improve key metabolic parameters, including obesity, dyslipidemia, insulin resistance, and hyperglycemia. This review explores the cellular mechanisms by which SGLT2i, either as monotherapy or combined with other treatments, modulate signaling pathways involved in lipid and carbohydrate metabolism. Additionally, we examine their effects on oxidative stress, inflammation, fibrosis, and apoptosis, which are critical drivers of NAFLD progression. This review is intended to summarize the multiple benefits of SGLT2 inhibitors and to educate healthcare providers on the therapeutic potential of these drugs in order to foster their incorporation into effective NAFLD management plans.
Collapse
Affiliation(s)
- Oscar R Zambrano-Vásquez
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Ciudad de México 04960, Mexico; Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| | - Fernando Cortés-Camacho
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Ciudad de México 04960, Mexico; Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| | - Jorge I Castañeda-Sánchez
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Unidad Xochimilco, México City 04960, Mexico
| | - Elena Aréchaga-Ocampo
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana, Unidad Cuajimalpa, México City 05348, Mexico
| | - Estefanía Valle-Velázquez
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| | - Juan C Cabrera-Angeles
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, Mexico
| | - José L Sánchez-Gloria
- Department of Internal Medicine, Division of Nephrology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Fausto Sánchez-Muñoz
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| | - Abraham S Arellano-Buendia
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| | - Laura G Sánchez-Lozada
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| | - Horacio Osorio-Alonso
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico.
| |
Collapse
|
7
|
Qian Y, Jia Y. Identification of Key Efferocytosis-Related Genes and Mechanisms in Diabetic Retinopathy. Mol Biotechnol 2025; 67:2785-2797. [PMID: 39085562 DOI: 10.1007/s12033-024-01239-x] [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: 09/08/2023] [Accepted: 07/09/2024] [Indexed: 08/02/2024]
Abstract
This study aimed to explore the key efferocytosis-related genes in diabetic retinopathy (DR) and their regulatory mechanisms. Public DR-related gene expression datasets, GSE160306 (training) and GSE60436 (validation), were downloaded. Differentially expressed efferocytosis-related genes (DEERGs) were analyzed using differential expression analysis and weighted gene co-expression network analysis. Functional enrichment analysis was conducted. Moreover, efferocytosis-related signature genes were identified using machine learning analysis, and their expression levels and diagnostic value were analyzed. Furthermore, nomograms were constructed; immune cell infiltration was analyzed; and gene set enrichment analysis, transcriptional regulation analysis, and small-molecule drug (SMD) prediction of efferocytosis-related signature genes were performed. In total, 36 DEERGs were identified in DR, and were markedly enriched in multiple functions, such as visual system development. Through further machine learning analysis, two efferocytosis-related signature genes, Ferritin Light Chain (FTL) and Fc Gamma Binding Protein (FCGBP), were identified, and were found to be upregulated in DR samples and showed high diagnostic performance for DR. A nomogram constructed using FTL and FCGBP accurately predicted the risk of DR. Moreover, the level of infiltration of immature B cells was positively correlated with FTL and FCGBP expression levels. Multiple transcription factors (TFs), such as CCCTC-Binding Factor (CTCF) and KLF Transcription Factor 9 (KLF9), were found to interact with both FTL and FCGBP. In addition, FTL can be targeted by miRNAs, such as miR-22-3p, and FCGBP can be targeted by miR-7973. In addition, both FTL and FCGBP can be targeted by SMDs, such as bisphenol A. Key efferocytosis-related genes, such as FTL and FCGBP, may promote DR development. Detecting or targeting FTL and FCGBP may aid in the prevention, diagnosis, and treatment of DR.
Collapse
Affiliation(s)
- Yu Qian
- Department of Ophthalmology, The First People's Hospital of Zhaoqing, 9 Donggang East Road, Zhaoqing, 526060, Guangdong, China.
| | - Yanwen Jia
- Department of Ophthalmology, Changzhou Second People's Hospital Affiliated Nanjing Medical University, Changzhou, 213004, Jiangsu, China
| |
Collapse
|
8
|
Zhang J, Xie Z, Zhu X, Xu C, Lin J, Zhao M, Cheng Y. New insights into therapeutic strategies for targeting hepatic macrophages to alleviate liver fibrosis. Int Immunopharmacol 2025; 158:114864. [PMID: 40378438 DOI: 10.1016/j.intimp.2025.114864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/29/2025] [Accepted: 05/09/2025] [Indexed: 05/18/2025]
Abstract
Liver fibrosis is a wound-healing response induced by persistent liver damage, resulting from complex multicellular interactions and multifactorial networks. Without intervention, it can progress to cirrhosis and even liver cancer. Current understanding suggests that liver fibrosis is reversible, making it crucial to explore effective therapeutic strategies for its alleviation. Chronic inflammation serves as the primary driver of liver fibrosis, with hepatic macrophages playing a dual role depending on their polarization state. This review summarizes various prevention and therapeutic strategies targeting hepatic macrophages in the context of liver fibrosis. These strategies include inhibition of macrophage recruitment, modulation of macrophage activation and polarization, regulation of macrophage metabolism, and induction of phagocytosis and autophagy in hepatic macrophages. Additionally, we discuss the communication between hepatic macrophages, hepatocytes, and hepatic stellate cells (HSCs), as well as the current clinical application of anti-fibrotic drugs targeting macrophages. The goal is to identify effective therapeutic targets at each stage of macrophage participation in liver fibrosis development, with the aim of using hepatic macrophages as a target for liver fibrosis treatment.
Collapse
Affiliation(s)
- Jialu Zhang
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Zhaojing Xie
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Xueyu Zhu
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Chenxi Xu
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Jiguo Lin
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Mingqi Zhao
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China
| | - Yunyun Cheng
- NHC Key Laboratory of Radiobiology, College of Public Health, Jilin University, Changchun 130021, China.
| |
Collapse
|
9
|
Xie K, Zhang Y, Ou X, Xiao Y, Luo J, Tan S. Taurine ameliorates liver fibrosis by repressing Fpr2-regulated macrophage M1 polarization. Eur J Pharmacol 2025; 997:177614. [PMID: 40216178 DOI: 10.1016/j.ejphar.2025.177614] [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: 10/30/2024] [Revised: 03/21/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025]
Abstract
Liver fibrosis is a reversible pathophysiological condition characterized by excessive extracellular matrix deposition that can progress to cirrhosis and liver failure if left untreated. Taurine, a sulfur-containing amino acid, protects the liver from damage. However, the effects of taurine on liver fibrogenesis have not been completely elucidated. In this study, we used amino acid metabolomics, gene expression microanalysis, and single-cell RNA sequencing (scRNA-seq) to investigate the roles of taurine, formyl peptide receptor 2 (Fpr2), and proinflammatory macrophages in liver fibrosis in human fibrotic sections and two distinct mouse models of liver fibrosis. Taurine transporter SLC6A6 wild-type and knockout littermate models and critical element inhibitors were also used. We found that taurine levels were significantly reduced in both human and murine fibrotic sections and that exogenous taurine supplementation alleviated fibrosis via SLC6A6. Furthermore, gene expression microarray analysis and scRNA-seq analyses demonstrated that exogenous taurine mitigated liver fibrosis, mainly by regulating Fpr2-related macrophage status. WRW4-mediated inhibition of Fpr2 ameliorated M1 macrophage polarization and alleviated liver fibrosis. Additionally, exogenous taurine suppressed Fpr2-modulated macrophage M1 polarization and the production of associated proinflammatory cytokines by repressing NF-κBp65 phosphorylation; moreover, SLC6A6 deficiency or treatment of liver fibrosis mouse models with an NF-κB inhibitor, BAY, impaired this protective effect of taurine. Therefore, taurine exerts a protective effect against liver fibrosis by repressing Fpr2/NF-κBp65-regulated macrophage M1 polarization, highlighting its potential therapeutic agent.
Collapse
Affiliation(s)
- Kaiduan Xie
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Yiwang Zhang
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Xingtong Ou
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Yuelin Xiao
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Jiajie Luo
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Siwei Tan
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China.
| |
Collapse
|
10
|
Mo J, Zhang Y, Cao C, Zheng W, Zheng L, Wang X, Wang S, Wu J, Bao Y. Activation of mechanosensitive ion channel Piezo1 linking metabolic reprogramming and pro-inflammatory responses in hepatocellular carcinoma. Cell Commun Signal 2025; 23:280. [PMID: 40514668 PMCID: PMC12164131 DOI: 10.1186/s12964-025-02289-7] [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: 02/14/2025] [Accepted: 06/04/2025] [Indexed: 06/16/2025] Open
Abstract
Piezo1 is identified as a novel mechanosensitive ion channel protein regulating a variety of physiological and pathological processes. However, the cellular effects of activation of Piezo1 protein on hepatocellular carcinoma (HCC) remains to be illuminated. In this study, we investigated the interaction between Piezo1 activation associated with metabolic reprogramming, pro-inflammatory cytokine release, and HCC progression. Our results indicated that the expression of Piezo1 gene and protein was up-regulated in the cancerous tissues of HCC patients, and the increased mRNA levels of Piezo1 were associated with unfavorable clinical outcomes. The activation of Piezo1 by its agonist Yoda1, facilitated HCC development by stimulating proliferation, migration, and invasion. Furthermore, Piezo1 channel activation by its agonist mediated pro-inflammatory responses mainly through its downstream molecule CXCL8 via ERK1/2 and AKT signaling pathways by up-regulating CXCL8 expression in transcriptional levels of HCC cells. The secretion of CXCL8 was induced by Piezo1 activation in HCC cells, improving immune cells trafficking and promoting angiogenesis, and therefore cross-talked with the tumor microenvironment (TME). Additionally, increased expression of MTHFD2, a metabolic checkpoint enzyme, was induced by the activation of Piezo1 channel and was likely to be a downstream target involved in mitochondrial stress followed by Piezo1 channel activation. In vitro data was further corroborated by the deceleration of tumor growth sizes in nude mice subcutaneously injected with Piezo1-depleted HCC cells in vivo. Finally, we showed synergistic anti-tumor effects of GsMTx4 and arachidonic acid, two Piezo1 antagonists, together with oxaliplatin in HCC cells. In summary, our findings suggest that the present linkage of metabolic reprogramming and pro-inflammatory responses after Piezo1 activation in HCC. Targeting Piezo1 appears to be a novel therapeutic strategy improving the treatment efficacy in HCC.
Collapse
Affiliation(s)
- Juanfen Mo
- The Key Laboratory, The Second Affiliated Hospital of Jiaxing University, 1518 Huancheng North Road, Jiaxing, Zhejiang, 314000, China
| | - YuXuan Zhang
- The Key Laboratory, The Second Affiliated Hospital of Jiaxing University, 1518 Huancheng North Road, Jiaxing, Zhejiang, 314000, China
| | - Chenxi Cao
- Department of Oncology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - WeiLi Zheng
- The Key Laboratory, The Second Affiliated Hospital of Jiaxing University, 1518 Huancheng North Road, Jiaxing, Zhejiang, 314000, China
| | - Li Zheng
- The Key Laboratory, The Second Affiliated Hospital of Jiaxing University, 1518 Huancheng North Road, Jiaxing, Zhejiang, 314000, China
| | - Xiaoguang Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Shiyue Wang
- The Key Laboratory, The Second Affiliated Hospital of Jiaxing University, 1518 Huancheng North Road, Jiaxing, Zhejiang, 314000, China
| | - Jiayuan Wu
- The Key Laboratory, The Second Affiliated Hospital of Jiaxing University, 1518 Huancheng North Road, Jiaxing, Zhejiang, 314000, China.
| | - Yi Bao
- The Key Laboratory, The Second Affiliated Hospital of Jiaxing University, 1518 Huancheng North Road, Jiaxing, Zhejiang, 314000, China.
- Department of Oncology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
- The Key Laboratory, Department of Oncology, The Second Affiliated Hospital of Jiaxing University, 1518 Huanchen North Road, Jiaxing, Zhejiang, 314000, China.
| |
Collapse
|
11
|
Lou C, Lan T, Xu S, Hu X, Li J, Xiang Z, Lin S, Fan X, Chen J, Xu X. Heterogeneity and plasticity of cholangiocytes in liver injury: a journey from pathophysiology to therapeutic utility. Gut 2025:gutjnl-2025-334763. [PMID: 40490318 DOI: 10.1136/gutjnl-2025-334763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 05/12/2025] [Indexed: 06/11/2025]
Abstract
Cholangiocytes are highly specialised cells participating in the pathobiology of various liver diseases and recognised to play a crucial role in response to liver injury. Cholangiocytes exhibit dramatic heterogeneity and plasticity, with distinct subtypes performing disparate functions during liver injury and regeneration. Acting as the liver progenitor cells, cholangiocytes can also convert to hepatocytes in the context of impaired hepatocyte proliferation. Harnessing the intrinsic regenerative ability of cholangiocytes is of great importance to alleviate liver injury and promote cholangiocyte-driven liver regeneration. Clinically, cholangiocytes and cholangiocyte organoids are expected to serve as favourable sources for cell therapy in cholangiopathies, which are known as a group of complex diseases involving the biliary system while lacking effective therapeutic options. A comprehensive understanding of the biological characteristics of cholangiocytes provides insights into developing cholangiocyte cell therapy for cholangiopathies. In this review, we discuss the critical role of cholangiocytes in liver injury and regeneration, reveal the underlying mechanism of cholangiocyte plasticity, and explore the prospects and challenges of using cholangiocytes as a source for cell therapy.
Collapse
Affiliation(s)
- Chengtao Lou
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Tianchen Lan
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shengjun Xu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Xinhao Hu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jiarui Li
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ze Xiang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shengda Lin
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaohui Fan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jian Chen
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Xiao Xu
- Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
Guan D, Huang P, Liu X, Li Q, Zhang X, Liu N, Wang Y, Wan Y, Chai J, Cai S, Chen R, Ye Z. Deficiency of myeloid NPC1 exacerbates liver injury and fibrosis by impairing macrophage efferocytosis. J Adv Res 2025; 72:213-227. [PMID: 39547438 DOI: 10.1016/j.jare.2024.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/31/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024] Open
Abstract
INTRODUCTION Niemann-Pick C1 (NPC1), a lysosomal cholesterol transport protein, is required for efficient efferocytosis. Patients with Npc1 mutation are frequently accompanied with hepatic symptoms, including hepatomegaly, elevated liver transaminases, or even acute liver failure, but the pathogenic mechanism remains unknown. OBJECTIVES Our work aims to characterize the functional role of myeloid NPC1 in liver injury and elucidate its underlying mechanism. METHODS Analyses of injured livers from patients with liver diseases and mouse models were conducted to examine NPC1 expression. Myeloid cell-specific Npc1 knockout mice were constructed to determine the functional role of macrophage NPC1 in liver injury. Isolated macrophages were subjected to in vitro mechanistical assays. RESULTS We found that NPC1 is mainly expressed in hepatic macrophages. Its mRNA and protein expression are significantly elevated in injured livers from both patients and mouse models. Tissue-specific deletion of myeloid Npc1 increased liver inflammation, levels of serum liver function enzymes, and liver fibrosis in mouse models of liver injury induced by carbon tetrachloride (CCl4) injection and methionine-and-choline-deficient (MCD) diets. Further analyses indicate that Npc1 deficiency in mouse models of liver injury resulted in increased levels of serum HMGB1 and mitochondrial DNA, promoted hepatic macrophage proinflammatory activation, M1 polarization, led to overproduction of hepatic inflammatory cytokines/chemokines, e.g. CCL2 and STING/NFκB pathway activation. In vitro mechanistical studies reveal that Npc1-deficient macrophages exhibited inefficient efferocytosis, partly due to impaired cargo degradation. CONCLUSIONS These findings indicate that elevated expression of myeloid NPC1 in liver diseases protects liver from injury by promoting macrophage efferocytosis of damaged cells. Dysfunction of NPC1 aggravates liver injury, suggesting that NPC1 may be a potential therapeutic target for treating liver diseases.
Collapse
Affiliation(s)
- Dongwei Guan
- Laboratory Animal Research Center, School of Medicine, Chongqing University, Chongqing 400044, China.
| | - Pengju Huang
- Laboratory Animal Research Center, School of Medicine, Chongqing University, Chongqing 400044, China
| | - Xinlei Liu
- Laboratory Animal Research Center, School of Medicine, Chongqing University, Chongqing 400044, China
| | - Qing Li
- Laboratory Animal Research Center, School of Medicine, Chongqing University, Chongqing 400044, China
| | - Xiaoxun Zhang
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center and Center for Metabolic-Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Nan Liu
- Department of Urology Oncological Surgery, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yong Wang
- Department of Laboratory Animal Science, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ying Wan
- Biomedical Analysis Center, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jin Chai
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center and Center for Metabolic-Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shiying Cai
- Department of Internal Medicine and Liver Center, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Rui Chen
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing 400030, China.
| | - Zhijia Ye
- Laboratory Animal Research Center, School of Medicine, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
13
|
Chen L, Guillot A, Tacke F. Reviewing the function of macrophages in liver disease. Expert Rev Gastroenterol Hepatol 2025; 19:621-637. [PMID: 40387555 DOI: 10.1080/17474124.2025.2508963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/10/2025] [Accepted: 05/08/2025] [Indexed: 05/20/2025]
Abstract
INTRODUCTION The liver is a central metabolic organ, but is also hosting a unique immune microenvironment to sustain homeostasis and proper defense measures against injury threats in healthy individuals. Liver macrophages, mostly represented by the tissue-resident Kupffer cells and bone marrow- or monocyte-derived macrophages, are intricately involved in various aspects of liver homeostasis and disease, including tissue injury, inflammation, fibrogenesis and repair mechanisms. AREAS COVERED We review recent findings on defining the liver macrophage landscape and their functions in liver diseases with the aim of highlighting potential targets for therapeutic interventions. A comprehensive literature search in PubMed and Google Scholar was conducted to identify relevant literature up to date. EXPERT OPINION Liver macrophages orchestrate key homeostatic and pathogenic processes in the liver. Thus, targeting liver macrophages represents an attractive strategy for drug development, e.g. to ameliorate liver inflammation, steatohepatitis or fibrosis. However, translation from fundamental research to therapies remains challenging due to the versatile nature of the liver macrophage compartment. Recent and major technical advances such as single-cell and spatially-resolved omics approaches deepened our understanding of macrophage biology at a molecular level. Yet, further studies are needed to identify suitable, etiology- and stage-dependent strategies for the treatment of liver diseases.
Collapse
Affiliation(s)
- Lanlan Chen
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Adrien Guillot
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
14
|
Wu Y, Tao Q, Xie J, Liu X, Zhou Y, Wei C, Zhang C, Wang J, Jin Y. Indole-3-carbinol inhibits PD-L1-mediated immune evasion in hepatocellular carcinoma via suppressing NF-κB p105 Ubiquitination. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156692. [PMID: 40215823 DOI: 10.1016/j.phymed.2025.156692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most common malignant tumors worldwide, and immunotherapy has demonstrated significant therapeutic benefit in HCC. Indole-3-carbinol (I3C), a naturally occurring ingredient of cruciferous vegetables, significantly inhibits the growth of a wide range of tumors. However, its mechanism of action has not been fully elucidated. PURPOSE This study aims to verify and explore the immunomodulatory effect of I3C in HCC models, and to investigate the specific role and mechanism by which I3C affects PD-L1 expression through the ubiquitination of NF-κB p105. METHODS In vitro, I3C was treated with HepG2 cells and relevant indicators were analyzed. In vivo, the mouse HCC model was established and the effect of I3C on tumors and immune function was evaluated. Subsequently, the downstream target of I3C was found through target prediction, molecular docking, and molecular dynamics simulation. Finally, combined therapy was used to further investigate the effect of I3C on mouse HCC tumors. RESULTS We observed that I3C resulted in decreased programmed cell death ligand 1 (PD-L1) expression in HepG2 cells and increased CD8 T cell infiltration in tissues. Subsequently, target prediction and molecular docking demonstrated that I3C was able to efficiently bind to NF-κB p105. In addition, overexpression of NF-κB p105 upregulated PD-L1 expression and almost completely eliminated the inhibitory effect of I3C. Notably, the combination of I3C and PD-L1 monoclonal antibodies showed synergistic anti-tumor effects in the mouse HCC model. CONCLUSION This study demonstrated that I3C inhibits PD-L1-mediated immune evasion in HCC via suppressing NF-κB p105 ubiquitination. The role of I3C in tumors deserves further investigation and provides the foundation for the future development of novel immunotherapeutic drugs.
Collapse
Affiliation(s)
- Yongkang Wu
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Qing Tao
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Jing Xie
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Xiao Liu
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Yuanzhi Zhou
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Chengyan Wei
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Chunwei Zhang
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Jingjing Wang
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Yong Jin
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China.
| |
Collapse
|
15
|
Xu J, Li Y, Wang G, Chen D, Hou C, Wang R, Dong W, Sun L, Yang K, Duan J, Pu G, Zhao X, Ai R, Xiu W, Wang Q, Wang C. Impaired mitochondrial degradation of CHCHD2 promotes metabolic dysfunction-associated steatohepatitis-related hepatocellular carcinoma by upregulating VEGFA. Oncogene 2025; 44:1475-1487. [PMID: 40025232 DOI: 10.1038/s41388-025-03321-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/01/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is the fastest-growing cause of liver cancer. The liver microenvironment of patients with MASH supports the development of hepatocellular carcinoma (HCC). Coiled-coil-helix-coiled-coil-helix domain-containing 2 (CHCHD2), which is located in both the mitochondria and nucleus, is increased in MASH liver. Its role in the development of MASH-HCC remain unknown. In this study, we found CHCHD2 protein levels were elevated in both tumor and para-tumor tissues of patients with MASH-HCC and diethylnitrosamine- and high-fat diet-induced MASH-HCC mice. Chchd2-knockout mice were generated. CHCHD2 was overexpressed in hepatocytes using AAV with TBG promoter. Chchd2 knockout inhibited the progression of MASH-HCC in mice. CHCHD2 protein-targeted ChIP-sequencing data revealed that CHCHD2 target genes encoding secretory proteins were enriched in cancer pathways. Among these genes, vascular endothelial growth factor A (VEGFA) level increased in CHCHD2-overexpressing livers and hepatocytes. Chchd2 knockdown reduced palmitate-induced VEGFA expression. Palmitate-treated hepatocyte increased the angiogenic activity of endothelial cells in a paracrine manner, and this was suppressed by Chchd2 knockdown in hepatocytes. CHCHD2-overexpressing hepatocytes promoted the angiogenic activity of endothelial cells. We futher employed an orthotopic murine model of HCC to demonstrate that elevated CHCHD2 protein levels in para-tumor tissues support HCC growth. In addition, we found that the degradation of CHCHD2 was primarily mediated by mitochondrial protease ClpXP, which was repressed in the MASH liver. In conclusion, the mitochondrial degradation of CHCHD2 is impaired in MASH, and elevated CHCHD2 levels in hepatocytes promote VEGFA transcription and support the growth of HCC.
Collapse
Affiliation(s)
- Jingwen Xu
- Department of Physiology and Pathophysiology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Yue Li
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Guangyan Wang
- Department of Physiology and Pathophysiology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Dapeng Chen
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, China
| | - Chenxue Hou
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Rui Wang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Wenhui Dong
- Department of Physiology and Pathophysiology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Lei Sun
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Kun Yang
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Jinjie Duan
- Department of Physiology and Pathophysiology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- School of Public Health, Tianjin Medical University, Tianjin, China
| | - Guangyin Pu
- Department of Physiology and Pathophysiology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Xiaoyu Zhao
- Department of Physiology and Pathophysiology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Rong Ai
- Department of Physiology and Pathophysiology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Wenjing Xiu
- Department of Physiology and Pathophysiology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China.
| | - Qi Wang
- Center of Liver Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China.
| | - Chunjiong Wang
- Department of Physiology and Pathophysiology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China.
- Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
16
|
Jiang Y, Xie F, Ling X, Zhang J, Yu Y, Huang Q, Zhang L, Ye L, Tao W, Hou M, Zhang C, Wang J. TCA Cycle Intermediate Mitigates Di(2-ethylhexyl) Phthalate-Induced Cholestatic Liver Injury Through Modulation of the Nrf2/NQO1 Signalling Axis. Basic Clin Pharmacol Toxicol 2025; 136:e70047. [PMID: 40370325 DOI: 10.1111/bcpt.70047] [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: 12/06/2024] [Revised: 04/17/2025] [Accepted: 04/22/2025] [Indexed: 05/16/2025]
Abstract
As a commonly used phthalate compound, di(2-ethylhexyl) phthalate (DEHP) has been shown to disrupt the tricarboxylic acid (TCA) cycle and aggravate tissue damage. However, whether the TCA cycle is involved in cholestatic liver injury (CLI) induced by DEHP and the protective effect of dimethyl fumarate (DMF), which is used to supplement TCA intermediate metabolites, remained unclear. Here, mice were randomized into five groups (n = 6/group): (1) Control, (2) DEHP (200 mg/kg/day), (3) DMF (100 mg/kg/day), (4) DEHP + DMF (30 mg/kg/day) and (5) DEHP + DMF (100 mg/kg/day). Our data demonstrated that DEHP exposure upregulated total bile acid (TBA) levels and broke the TCA cycle, resulting in reduced fumaric acid and malic acid. However, we further supplemented fumaric acid with DMF and found that DMF effectively reversed the high levels of TBA, alkaline phosphatase (ALP) and glutamyl transpeptidase (GGT) induced by DEHP in mice. Meanwhile, pathological results in the liver showed that DMF improved bile duct cell damage, inflammatory cell infiltration, collagen deposition and necrosis caused by DEHP. In addition, we found that DEHP elevated the level of interleukin (IL)-1β, IL-6, TNF-α and MDA and decreased the level of SOD in the mouse liver, which was effectively reversed by DMF treatment. Besides, DMF upregulated the expression of Nrf2 and NQO1 in the liver of DEHP-exposed mice. For in vitro validation, AML-12 cells were treated with (1) Control, (2) DEHP (250 μM), (3) DEHP + DMF (10 μM), (4) DEHP + DMF (25 μM) and (5) DEHP + DMF (50 μM). DEHP exposure increased the expression of IL-1β, IL-6 and TNF-α, which was mitigated by DMF, while ML385, an Nrf2 inhibitor, could counteract the anti-inflammatory effects of DMF. These findings indicate that DEHP broke the TCA cycle of the mouse liver, and DMF supplementation protects against DEHP-induced CLI by activating the Nrf2/NQO1 pathway.
Collapse
Affiliation(s)
- Yue Jiang
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Fang Xie
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xutao Ling
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jiayi Zhang
- School of Pharmacy, Anhui Medical University, Hefei, China
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Public Health Clinical Center, Hefei, China
| | - Yun Yu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Public Health Clinical Center, Hefei, China
| | - Qianqian Huang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Public Health Clinical Center, Hefei, China
| | - Lun Zhang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Public Health Clinical Center, Hefei, China
| | - Lu Ye
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Wenkang Tao
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Mengzhen Hou
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Cheng Zhang
- Department of Toxicology, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
- MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China
| | - Jianqing Wang
- School of Pharmacy, Anhui Medical University, Hefei, China
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Public Health Clinical Center, Hefei, China
- MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China
| |
Collapse
|
17
|
Wang J, Miao Z, Gao Y, Xie Z, Liu M, Zou W. Formyl peptide receptor 2: a potential therapeutic target for inflammation-related diseases. Pharmacol Rep 2025; 77:593-609. [PMID: 40102363 DOI: 10.1007/s43440-025-00704-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 03/20/2025]
Abstract
Formyl peptide receptor 2 (FPR2) is a G protein-coupled receptor with seven transmembrane domains, widely distributed in human cells. It plays a crucial role in inflammation-related diseases. Known for its "double-edged sword" nature, FPR2 can bind a variety of exogenous and endogenous ligands, mediating both pro-inflammatory and anti-inflammatory responses in tissues such as eyes, liver, joints, lungs, nerves, and blood vessels. FPR2's bioactivities are regulated by a complex network of genes and signaling pathways. However, the precise regulatory mechanisms governing its functions in different inflammatory conditions are still not well understood. This review summarizes the FPR2's activities in various inflammation-related diseases and looks into its potential as a therapeutic target. This review highlights recent advances in developing exogenous agonists for FPR2 and discusses receptor expression across species to support nonclinical research. Overall, this review aims to clarify FPR2's role in inflammation and provide insights for the development of new drugs against inflammatory diseases.
Collapse
Affiliation(s)
- Jiaying Wang
- School of Pharmacy, Hunan Provincial Maternal and Child Health Care Hospital, University of South China, Hengyang, 421001, China
| | - Zhishuo Miao
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yinhuang Gao
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - ZhiZhong Xie
- School of Pharmacy, Hunan Provincial Maternal and Child Health Care Hospital, University of South China, Hengyang, 421001, China
| | - Menghua Liu
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Wei Zou
- School of Pharmacy, Hunan Provincial Maternal and Child Health Care Hospital, University of South China, Hengyang, 421001, China.
| |
Collapse
|
18
|
Bozward AG, Davies SP, Morris SM, Kayani K, Oo YH. Cellular interactions in self-directed immune-mediated liver diseases. J Hepatol 2025; 82:1110-1124. [PMID: 39793614 DOI: 10.1016/j.jhep.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/18/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025]
Abstract
The lymphocyte population must traverse a complex path throughout their journey to the liver. The signals which these cells must detect, including cytokines, chemokines and other soluble factors, steer their course towards further crosstalk with other hepatic immune cells, hepatocytes and biliary epithelial cells. A series of specific chemokine receptors and adhesion molecules drive not only the recruitment, migration, and retention of these cells within the liver, but also their localisation. Perturbation of these interactions and failure of self-recognition drive the development of several autoimmune liver diseases. We also describe check point-induced liver injury. Immune cell internalisation into hepatocytes (emperipolesis) in autoimmune hepatitis and into biliary epithelial cells (intra-epithelial lymphocyte) in primary biliary cholangitis are typical features in autoimmune liver diseases. Finally, we describe emerging immune-based therapies, including regulatory T cell, anti-cytokine and anti-chemokine therapies, cytokine supplementation (e.g. interleukin-2), as well as co-inhibitory molecule manipulation, including T-cell engagers, and discuss their potential application in the treatment of autoimmune liver diseases.
Collapse
Affiliation(s)
- Amber G Bozward
- Centre for Liver and Gastroenterology research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK; National Institute of Health Research Biomedical Research Centre, University of Birmingham and University Hospital Birmingham NHS Foundation Trust, Birmingham, UK; Centre for Rare Diseases, European Reference Network on Hepatological Diseases (ERN-RARE-LIVER) Centre, University of Birmingham, Birmingham, UK.
| | - Scott P Davies
- Centre for Liver and Gastroenterology research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK; National Institute of Health Research Biomedical Research Centre, University of Birmingham and University Hospital Birmingham NHS Foundation Trust, Birmingham, UK; Centre for Rare Diseases, European Reference Network on Hepatological Diseases (ERN-RARE-LIVER) Centre, University of Birmingham, Birmingham, UK
| | - Sean M Morris
- Centre for Liver and Gastroenterology research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK; National Institute of Health Research Biomedical Research Centre, University of Birmingham and University Hospital Birmingham NHS Foundation Trust, Birmingham, UK; Centre for Rare Diseases, European Reference Network on Hepatological Diseases (ERN-RARE-LIVER) Centre, University of Birmingham, Birmingham, UK
| | - Kayani Kayani
- Centre for Liver and Gastroenterology research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK; National Institute of Health Research Biomedical Research Centre, University of Birmingham and University Hospital Birmingham NHS Foundation Trust, Birmingham, UK; Centre for Rare Diseases, European Reference Network on Hepatological Diseases (ERN-RARE-LIVER) Centre, University of Birmingham, Birmingham, UK
| | - Ye H Oo
- Centre for Liver and Gastroenterology research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK; National Institute of Health Research Biomedical Research Centre, University of Birmingham and University Hospital Birmingham NHS Foundation Trust, Birmingham, UK; Centre for Rare Diseases, European Reference Network on Hepatological Diseases (ERN-RARE-LIVER) Centre, University of Birmingham, Birmingham, UK; Liver Transplant and Hepatobiliary Department, Queen Elizabeth Hospital, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK.
| |
Collapse
|
19
|
Zhang Z, Zhang J, Yan X, Wang J, Huang H, Teng M, Liu Q, Han S. Dissecting the genetic basis and mechanisms underlying the associations between multiple extrahepatic factors and autoimmune liver diseases. J Transl Autoimmun 2025; 10:100260. [PMID: 39741931 PMCID: PMC11683281 DOI: 10.1016/j.jtauto.2024.100260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/02/2024] [Accepted: 11/03/2024] [Indexed: 01/03/2025] Open
Abstract
Background Autoimmune liver diseases (AILDs) encompass autoimmune hepatitis (AIH), primary biliary cholangitis (PBC), and primary sclerosing cholangitis (PSC). The onset of these diseases is fundamentally influenced by genetic susceptibility. Although various extrahepatic factors are potentially linked to AILDs, the genetic underpinnings and mechanisms of these associations remain unclear. Methods Utilizing large-scale genome-wide association study (GWAS) data, this study systematically investigated the relationships between extrahepatic autoimmune diseases (EHAIDs), immune cells, and various triggering factors with AILDs. Mendelian randomization (MR) was employed to assess the causal effects of these extrahepatic factors on AILDs, complemented by linkage disequilibrium score (LDSC) regression to uncover shared genetic architecture and causal effects underlying the associations between autoimmune diseases. We employed colocalization, enrichment analysis, and protein-protein interaction (PPI) network to identify the functions of shared loci. Additionally, we proposed that activated immune cells in the circulation may contribute to liver and biliary tract inflammation via migration, mediating the impact of extrahepatic factors on AILDs. This hypothesis was tested using two mediation analysis methods: two-step MR (TSMR) and multivariable MR (MVMR). Results Causal associations between multiple extrahepatic factors and AILDs were identified. Notably, CD27+ B cells were found to be a risk factor for PBC, while PSC progression was associated with CD28+ CD8+ T cells exhaustion and increased levels of CD28- CD8+ T cells. Mediation analyses revealed 64 pathways via TSMR and 15 pathways via MVMR, indicating that the effects of extrahepatic factors on AILDs may be mediated by circulating immune cells. The shared genetic architecture also contributed to these associations. Analysis of shared loci and gene functions identified ATXN2 as being shared between PBC and 9 EHAIDs, while SH2B3 and PSMG1 were shared with 6 and 5 EHAIDs, respectively, in PSC. Conclusions Our research compared three distinct AILDs, enhancing the understanding of their etiology and providing new evidence on risk factors, diagnostic markers, and potential therapeutic targets.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Jiayi Zhang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Xinyang Yan
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Jiachen Wang
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Haoxiang Huang
- Department of urology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Menghao Teng
- Department of Orthopedics, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Qingguang Liu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Shaoshan Han
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| |
Collapse
|
20
|
Guglielmi V, Colangeli L, Parrotta ME, Ciammariconi A, Milani I, D'Adamo M, Sbraccia P, Capoccia D. Social isolation and loneliness in non-communicable chronic diseases: Impact of COVID-19 pandemic, population aging and technological progress. Nutr Metab Cardiovasc Dis 2025; 35:104015. [PMID: 40189996 DOI: 10.1016/j.numecd.2025.104015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 04/29/2025]
Abstract
AIMS Social isolation and loneliness have increasingly emerged as closely linked to onset and progression of non-communicable chronic diseases (NCDs). The aim of this review is to highlight the importance of addressing social isolation in the prevention and management of NCDs such as obesity, type 2 diabetes, and cardiovascular diseases in order to hinder their development and improve their outcomes. DATA SYNTHESIS Social isolation and loneliness affect a significant portion of the older adult population, due to decrease in social interactions, chronic illnesses and sensory impairments. However, many other vulnerable populations may experience social isolation because of psychiatric or disabling health conditions, substances abuse, low socioeconomic status, unemployment and belonging to ethnic or marginalized minorities. The unprecedented COVID-19-related social distancing can be taken as a proof-of-concept of the detrimental role of poor interactions in NCDs prevention and management not only at individual level but also in a public health perspective. Indeed, social isolation has been linked to unhealthy lifestyle choices, disrupted sleep quality, low utilization of healthcare, preventive services and adherence to treatments. Underlying mechanisms like inflammation and stress responses may also play a role in the association between social isolation and worse NCDs outcomes. CONCLUSIONS Social isolation negatively impacts on the development, progression and management of NCDs. Effective interventions for social isolation should address both societal factors and healthcare-related needs. To counteract the detrimental effects of social distancing during COVID-19 pandemic, the use of telemedicine was implemented. However, telemedicine is not always available, and legislative and age-related barriers persist.
Collapse
Affiliation(s)
- Valeria Guglielmi
- Department of Systems Medicine, University of Rome Tor Vergata, Obesity Medical Center, University Hospital Policlinico Tor Vergata, Rome, Italy.
| | - Luca Colangeli
- Department of Systems Medicine, University of Rome Tor Vergata, Obesity Medical Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Maria Eugenia Parrotta
- Department of Systems Medicine, University of Rome Tor Vergata, Obesity Medical Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Azzurra Ciammariconi
- Department of Systems Medicine, University of Rome Tor Vergata, Obesity Medical Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Ilaria Milani
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Monica D'Adamo
- Department of Systems Medicine, University of Rome Tor Vergata, Obesity Medical Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Paolo Sbraccia
- Department of Systems Medicine, University of Rome Tor Vergata, Obesity Medical Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Danila Capoccia
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
21
|
Cai Y, Chen J, Deng X, Wang C, Huang J, Lian N. Sleep behaviors and metabolic-associated fatty liver disease. PLoS One 2025; 20:e0323715. [PMID: 40435271 PMCID: PMC12118868 DOI: 10.1371/journal.pone.0323715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 04/11/2025] [Indexed: 06/01/2025] Open
Abstract
OBJECTIVE This study aimed to comprehensively evaluate the association between various sleep behaviors and the risk of metabolic-associated fatty liver disease (MAFLD), particularly self-reported snoring. METHODS Multivariate logistic regression was used to explore independent factors associated with MAFLD. ROC curve and decision curve analyses were used to analyze and compare the different indicators. RESULTS A total of 3708 patients were enrolled, and 41.4% of them had MAFLD. According per multivariate logistic regression analysis, self-reported snoring was an independent predictor of MAFLD (p < 0.001), particularly the occasional and frequent snoring groups [OR (95% CI): 1.44 (1.12-1.87), 1.48 (1.15-1.91), p < 0.001]. In addition, the liver function levels and incidence rates of metabolic parameters were independently associated with the severity of self-reported snoring (all p < 0.05). Subgroup analyses suggested that the frequency of snoring was independently related to the risk of MAFLD in young and middle-aged patients (both p < 0.05), and was no longer associated with any frequency of self-reported snoring in the subgroup older than 60 years (p = 0.400). In both female and male subgroup, subjects who snored frequently had a higher odds risk of MAFLD than those who did not (both p < 0.05). The area under the ROC curve for snoring was 0.638, which was superior to that of the other indicators for MAFLD prediction (all p < 0.001). Meanwhile, decision curve analysis showed that snoring had a better clinical net benefit compared to other biomarkers, with a threshold probability (Pt) of approximately 0.3-0.6. CONCLUSION Self-reported snoring was an independent risk factor for MAFLD in young and middle-aged subjects with a moderate predictive value. Therefore, intense monitoring and evaluation of MAFLD in these patients is necessary.
Collapse
Affiliation(s)
- Yuqing Cai
- Department of respiratory and critical care medicine, Respiratory Disease Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of respiratory and critical care medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jia Chen
- Department of respiratory and critical care medicine, Respiratory Disease Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of respiratory and critical care medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiaoyu Deng
- Department of respiratory and critical care medicine, Respiratory Disease Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of respiratory and critical care medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Caiyun Wang
- Department of respiratory and critical care medicine, Respiratory Disease Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of respiratory and critical care medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jiefeng Huang
- Department of respiratory and critical care medicine, Respiratory Disease Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of respiratory and critical care medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ningfang Lian
- Department of respiratory and critical care medicine, Respiratory Disease Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of respiratory and critical care medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
22
|
Ma J, Xu R, Li R, Fu Y, Xu J, Zhou L, Qi Y. Integration of bioinformatics and identification of the role of m6A genes in NAFLD. PLoS One 2025; 20:e0321757. [PMID: 40435202 PMCID: PMC12119021 DOI: 10.1371/journal.pone.0321757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/11/2025] [Indexed: 06/01/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is prevalent worldwide and seriously affects health. M6A methylation is crucial in its pathogenesis. In this study, a thorough analysis of three gene expression datasets identified nine key differentially expressed genes DEGs associated with m6A methylation in NAFLD that are involved in important biological processes. Subsequently, functional enrichment analysis, weighted gene co-expression network analysis (WGCNA), gene set variation analysis (GSVA) and immune infiltration analysis were conducted to explore the molecular mechanism and gene expression patterns. The LASSO risk model contains a total of 5 m6A-related differentially expressed genes (m6A-RDEGs)(RBM15, IGF2BP2, EIF3B, YTHDC1, WTAP), and the diagnostic model based on these key genes has high accuracy. Among them, YTHDC1 and WTAP are used as prominent biomarkers. In addition, an interaction network between mRNA and miRNA, RNA-binding protein (RBP), transcription factor (TF) and drugs is also constructed. Finally, the animal model of NAFLD was successfully established and validated by RT-qPCR and western blot. This study provides a valuable tool for clinical diagnosis and drives the progress of NAFLD research.
Collapse
Affiliation(s)
- Jianguo Ma
- The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Rongyi Xu
- The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Renlin Li
- College of Acupuncture and Massage, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
| | - Yangyang Fu
- The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Jing Xu
- The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Lei Zhou
- The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Yan Qi
- The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, Kunming, China
| |
Collapse
|
23
|
Sacco R, Giannini EG, Tortora R, Di Costanzo GG, Mega A, Marzi L, Pieri G, Pasta A, Daniele B, Federico P, Cabibbo G, Russello M, Cocuzza C, Giacomelli L, Silletta M, Gallo P, Gentilucci UV, Casadei-Gardini A, Claar E, Pellicelli A, Bellini M, Morisco F, Tatali C, Pace Palitti V, Izzi A, Di Stefano M, Rinaldi L, Facciorusso A. Lenvatinib Is Highly Effective in Patients with Hepatocellular Carcinoma Related to Both Metabolic Dysfunction-Associated Steatohepatitis and Alcoholic Etiology: A Propensity Score Analysis. Cancers (Basel) 2025; 17:1808. [PMID: 40507289 PMCID: PMC12153599 DOI: 10.3390/cancers17111808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Revised: 04/30/2025] [Accepted: 05/19/2025] [Indexed: 06/16/2025] Open
Abstract
Background and aims: Metabolic dysfunction-associated steatotic liver disease (MASLD)-related hepatocellular carcinoma (HCC) may have distinct biological characteristics influencing systemic treatment response. However, the prognostic impact of MASLD vs. alcohol-related HCC in patients receiving lenvatinib remains unclear. This study aimed to assess lenvatinib's effectiveness and safety in these populations. Methods: A multicenter cohort of 378 HCC patients treated with lenvatinib (2019-2024) was analyzed. Propensity score matching was performed based on age, sex, tumoral stage, alpha-fetoprotein levels and Child-Pugh class. Survival was estimated using Kaplan-Meier analysis and compared with the log-rank test. Results were expressed as HR and 95% CI. Results: After matching, 115 patients per group were compared. Median OS was 21 months (95% CI: 20-23) in the group with metabolic dysfunction-associated steatohepatitis (MASH) and 19 months (95% CI: 18-21) in the group with alcohol etiology (p = 0.18). In multivariate analysis, only Child-Pugh class (HR 2.67, 95% CI: 1.84-5.41) and tumor stage (HR 2.18, 95% CI: 1.57-6.93) resulted as significant predictors of OS. Median PFS was 9 months (95% CI: 8-9) in patients with MASH and 9 months (95% CI: 7-10) in patients with alcohol etiology (p = 0.33). Only the Child-Pugh class was a significant predictor of PFS in univariate analysis (HR 1.56, 95% CI: 1.15-3.41; p = 0.03). No difference in terms of adverse event rate was observed between the two groups. Conclusions: Lenvatinib is effective in patients with both MASH- and alcohol-related HCC, with no difference in oncological outcomes between the two groups.
Collapse
Affiliation(s)
- Rodolfo Sacco
- Gastroenterology and Digestive Endoscopy Unit, Department of Surgical and Medical Sciences, University of Foggia, Viale Pinto 1, 71122 Foggia, Italy;
| | - Edoardo G. Giannini
- Gastroenterology Unit, Department of Internal Medicine, University of Genova, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, 16126 Genova, Italy; (E.G.G.); (G.P.); (A.P.)
| | - Raffaella Tortora
- Liver Unit, Department of Transplantation, Cardarelli Hospital, 80131 Naples, Italy; (R.T.); (G.G.D.C.)
| | | | - Andrea Mega
- Gastroenterology Unit, Bolzano Regional Hospital, 39100 Bolzano, Italy; (A.M.); (L.M.)
| | - Luca Marzi
- Gastroenterology Unit, Bolzano Regional Hospital, 39100 Bolzano, Italy; (A.M.); (L.M.)
| | - Giulia Pieri
- Gastroenterology Unit, Department of Internal Medicine, University of Genova, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, 16126 Genova, Italy; (E.G.G.); (G.P.); (A.P.)
| | - Andrea Pasta
- Gastroenterology Unit, Department of Internal Medicine, University of Genova, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, 16126 Genova, Italy; (E.G.G.); (G.P.); (A.P.)
| | - Bruno Daniele
- Medical Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (B.D.); (P.F.)
| | - Piera Federico
- Medical Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (B.D.); (P.F.)
| | - Giuseppe Cabibbo
- Section of Gastroenterology & Hepatology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, PROMISE, University of Palermo, 90127 Palermo, Italy;
| | - Maurizio Russello
- Liver Unit, ARNAS Garibaldi-Nesima, 95122 Catania, Italy; (M.R.); (C.C.)
| | - Caterina Cocuzza
- Liver Unit, ARNAS Garibaldi-Nesima, 95122 Catania, Italy; (M.R.); (C.C.)
| | | | - Marianna Silletta
- Division of Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy;
| | - Paolo Gallo
- Clinical Medicine and Hepatology Unit, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy; (P.G.); (U.V.G.)
| | - Umberto Vespasiani Gentilucci
- Clinical Medicine and Hepatology Unit, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy; (P.G.); (U.V.G.)
| | - Andrea Casadei-Gardini
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, 20132 Milan, Italy;
| | - Ernesto Claar
- Department of Medicine, Ospedale Evangelico Villa Betania, 80147 Naples, Italy;
| | - Adriano Pellicelli
- Liver Unit, Department of Liver Transplant, A.O. San Camillo Forlanini, 00152 Rome, Italy;
| | - Massimo Bellini
- Gastroenterology Unit, University of Pisa, 56124 Pisa, Italy;
| | - Filomena Morisco
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy;
| | - Concetta Tatali
- Gastroenterology and Digestive Endoscopy Unit, Department of Surgical and Medical Sciences, University of Foggia, Viale Pinto 1, 71122 Foggia, Italy;
| | | | - Antonio Izzi
- Emergency and Highly Contagious Infectious Diseases, A.O. dei Colli, P.O.D. Cotugno, 80131 Naples, Italy;
| | | | - Luca Rinaldi
- Department of Medicine and Health Sciences, University of Molise, 86100 Campobasso, Italy;
| | - Antonio Facciorusso
- Gastroenterology Unit, Department of Experimental Medicine, Università del Salento, 73100 Lecce, Italy;
| |
Collapse
|
24
|
Zeng X, Huang D, Zhu Z, Cai Q, Yang Y, Lu H, Chen J. Mechanism-guided drug development and treatment for liver fibrosis: a clinical perspective. Front Pharmacol 2025; 16:1574385. [PMID: 40492139 PMCID: PMC12146339 DOI: 10.3389/fphar.2025.1574385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 05/07/2025] [Indexed: 06/11/2025] Open
Abstract
Liver fibrosis is a common response to chronic liver injury due to multiple etiologies and plays a crucial in the progression of chronic liver disease to cirrhosis, hepatocellular carcinoma, and other liver-related clinical outcomes. Currently, available treatments to block liver fibrosis are designed to eliminate the underlying causes of liver disease. The lack of truly effective drugs to regress or reverse fibrosis is a major unmet clinical need. In this context, this article briefly describes the pathological process of hepatic fibrosis and focuses on reviewing the progress of clinical studies on mechanism-based anti-fibrotic drug development and therapy, highlighting that the positive effect of thyroid hormone receptor-β (THR-β) analogs, fibroblast growth factor 21 (FGF21) analogues, Glucagon-like peptide 1 receptor (GLP-1R) agonists, pan-peroxisome proliferator-activated receptor (pan-PPAR) agonists, fatty acid synthase (FASN) inhibitors, and hydronidone in reducing liver fibrosis caused by specific etiologies. Moreover, multi-pathway guided combination therapy or traditional Chinese medicine demonstrate significant advantages in combating liver fibrosis. Finally, new technologies and approaches affecting the clinical development of anti-hepatic fibrosis drugs were discussed.
Collapse
Affiliation(s)
- Xiangchang Zeng
- Department of Liver Diseases, Shenzhen Third People's Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Deliang Huang
- Department of Liver Diseases, Shenzhen Third People's Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Zhibin Zhu
- Department of Liver Diseases, Shenzhen Third People's Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Qingxian Cai
- Department of Liver Diseases, Shenzhen Third People's Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yang Yang
- Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Clinical Research Center for Infectious Disease, State Key Discipline of Infectious Disease, Shenzhen Third People's Hospital, The Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Hongzhou Lu
- Department of Liver Diseases, Shenzhen Third People's Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jun Chen
- Department of Liver Diseases, Shenzhen Third People's Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
25
|
Xiang Y, Mao W. Neutrophil-derived ratios as predictors of short-term mortality in HBV-associated decompensated cirrhosis. BMC Gastroenterol 2025; 25:404. [PMID: 40419938 PMCID: PMC12105264 DOI: 10.1186/s12876-025-03991-z] [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: 02/12/2025] [Accepted: 05/13/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Hepatitis B virus-associated decompensated cirrhosis (HBV-DC) is recognized as a critical illness with an increased risk of short-term mortality. Neutrophil-derived ratios, including neutrophil-to-lymphocyte ratio (NLR), neutrophil-to-albumin ratio, neutrophil-to-high-density lipoprotein-cholesterol ratio, neutrophil-to-hemoglobin ratio, and neutrophil-to-platelet ratio, have emerged as potential prognostic markers in various liver diseases. The present study aimed to determine the effectiveness of these neutrophil-derived ratios for prediction of mortality in patients with HBV-DC. METHODS We conducted a retrospective analysis of HBV-DC patients at our hospital between April 2022 and April 2024. The study endpoint was the 30-day mortality rate. These neutrophil-derived ratios were calculated from data obtained during routine laboratory tests on admission. Disease severity was assessed using the Model for End-Stage Liver Disease (MELD) score. Multivariate regression analyses and receiver operating characteristic (ROC) curve analyses were conducted. RESULTS The study investigated 160 HBV-DC patients, of whom 23 (14.4%) experienced mortality within 30 days. Non-survivors exhibited markedly higher values for neutrophil-derived ratios than survivors. All neutrophil-derived ratios were associated with mortality in univariate analyses, but only NLR and MELD score remained as independent predictors of mortality in multivariate analyses. In the ROC analyses, NLR showed a similar prognostic value to MELD score. Moreover, both NLR and MELD score had high specificity for prediction of mortality in HBV-DC patients. CONCLUSIONS Among neutrophil-derived ratios, NLR stands out as a simple and reliable predictor of mortality in HBV-DC patients.
Collapse
Affiliation(s)
- Yang Xiang
- Department of Endocrinology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - WeiLin Mao
- Department of Clinical Laboratory, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, 310003, Zhejiang Province, China.
| |
Collapse
|
26
|
Zhao M, Zhao J, Yang H, Ouyang Z, Lv C, Geng Z, Zhao J. The bile acid-gut microbiota axis: A central hub for physiological regulation and a novel therapeutic target for metabolic diseases. Biomed Pharmacother 2025; 188:118182. [PMID: 40413999 DOI: 10.1016/j.biopha.2025.118182] [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: 04/16/2025] [Revised: 05/18/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025] Open
Abstract
Bile acids are a family of signaling molecules synthesized in the liver and metabolized by gut bacteria. As metabolites of the intestinal microbiota, bile acids bind to various receptors, and affect the metabolism and immune function of the host, including glucose and lipid metabolism, energy homeostasis, and inflammatory response. Conversely, bile acids also shape the composition of the gut microbiota. Given their critical role in physiological regulation, disrupted bile acid signaling is closely linked to metabolic diseases. Consequently, therapeutic strategies targeting bile acids are increasingly being explored. The size, composition, and function of the bile acid pool can be modulated through direct treatments (e.g., bile acid replacement therapy, administration of bile acid receptor agonists/antagonists) or indirect treatments (e.g., gut microbiota modulation, probiotic supplementation), providing new ideas for preventing and treating metabolic diseases.
Collapse
Affiliation(s)
- Min Zhao
- Hebei Provincial Center for Clinical Laboratories, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jiafeng Zhao
- Hebei Provincial Center for Clinical Laboratories, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huimin Yang
- Hebei Provincial Center for Clinical Laboratories, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zirou Ouyang
- Hebei Provincial Center for Clinical Laboratories, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chang Lv
- Hebei Provincial Center for Clinical Laboratories, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zijun Geng
- Hebei Provincial Center for Clinical Laboratories, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jianhong Zhao
- Hebei Provincial Center for Clinical Laboratories, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
27
|
El-Sehrawy AAMA, Jafari M, Zwamel AH, Rashidian P, Ballal S, Kalia R, Nanda A, Maharana L, Javankiani S, Hashemi M, Amini-Salehi E. Neutrophil Percentage-to-Albumin Ratio and Neutrophil-to-Albumin Ratio as novel biomarkers for non-alcoholic fatty liver disease: a systematic review and meta-analysis. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2025; 44:167. [PMID: 40413495 PMCID: PMC12102820 DOI: 10.1186/s41043-025-00926-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 05/11/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a major global health concern, with rising prevalence linked to obesity, insulin resistance, and metabolic syndrome. Timely and accurate identification of individuals at risk is crucial for improving outcomes. Recently, systemic inflammatory and nutritional markers such as the Neutrophil Percentage-to-Albumin Ratio (NPAR) and the Neutrophil-to-Albumin Ratio (NAR) have emerged as promising non-invasive biomarkers for NAFLD. Both ratios reflect inflammation and hepatic nutritional status, offering potential utility in predicting disease presence and progression. This systematic review and meta-analysis aimed to evaluate the diagnostic value of NPAR and NAR in patients with NAFLD. METHODS A comprehensive search was performed across databases including PubMed, Embase, Scopus, and Web of Science from inception to December 28, 2024. Data extraction was carried out using a standardized form, and the methodological quality of included studies was assessed using the Newcastle-Ottawa Scale. Statistical analyses were performed using STATA version 18, employing a random-effects model. RESULTS The meta-analysis demonstrated that both the Neutrophil Percentage-to-Albumin Ratio (NPAR) and the Neutrophil-to-Albumin Ratio (NAR) were significantly higher in patients with NAFLD compared to healthy individuals. NPAR showed a standardized mean difference (SMD) of 0.28 (95% CI: 0.22-0.35, P < 0.01), while NAR had a higher effect size with an SMD of 0.69 (95% CI: 0.44-0.93, P < 0.01). The pooled diagnostic performance of NPAR yielded a sensitivity of 69.5% (95% CI: 56.3-82.6%), specificity of 63.1% (95% CI: 46.6-70.0%), and an area under the curve (AUC) of 76.05% (95% CI: 66.3-85.7%). For NAR, the pooled sensitivity was 65.0% (95% CI: 49.0-82.0%), specificity was 63.0% (95% CI: 47.0-79.0%), and AUC was 69.0% (95% CI: 48.0-89.0%). CONCLUSION In conclusion, both NPAR and NAR were found to be elevated in individuals with NAFLD, supporting their potential as non-invasive and accessible biomarkers. These ratios reflect key aspects of systemic inflammation and nutritional status, offering clinical value in early detection and risk stratification. However, given the limited number of studies available-particularly for NAR-further research is needed to confirm these findings, establish standardized thresholds, and assess their performance across diverse populations and clinical settings. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
| | - Maryam Jafari
- Student Research Committee, Anzali International Campus, Guilan University of Medical Sciences, Rasht, Iran
| | - Ahmed Hussein Zwamel
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Najaf, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College,, Al Diwaniyah, , The Islamic University of Babylon, Babylon, Iraq
| | - Pegah Rashidian
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN, Bangalore, Karnataka, India
| | - Rishiv Kalia
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, India
| | - Anima Nanda
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Laxmidhar Maharana
- Department of Pharmaceutical Sciences, Siksha 'O' Anusandhan, Bhubaneswar, Odisha, India
| | - Sepide Javankiani
- General Surgery Department, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hashemi
- Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Ehsan Amini-Salehi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
28
|
Zhou T, Xie C. Association between Triglyceride-Glucose index and onset of type 2 diabetes in metabolic Dysfunction-Associated steatotic liver disease population. Sci Rep 2025; 15:18044. [PMID: 40410296 PMCID: PMC12102177 DOI: 10.1038/s41598-025-03453-2] [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: 01/21/2025] [Accepted: 05/20/2025] [Indexed: 05/25/2025] Open
Abstract
Previous studies have demonstrated a strong correlation between the Triglyceride-Glucose Index(TyG) index and diabetes mellitus; however, limited research has focused on the relationship between the TyG index and the incidence of diabetes mellitus in the Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) population. This study aims to investigate the association between the TyG index and the occurrence of diabetes mellitus in the MASLD population in Japan. This retrospective cohort study included 2,741 patients with MASLD from Murakami Memorial Hospital in Japan, spanning the years 2004 to 2015. The primary method employed to investigate the relationship between the TyG index and the occurrence of diabetes was Cox proportional hazards regression. Additionally, a Generalized Additive Model and a two-piecewise linear regression model were utilized to assess whether a linear relationship exists between the TyG index and the development of diabetes, as well as to calculate threshold effects. After adjusting for variables that may potentially influence the occurrence of diabetes, we found that the TyG index was positively correlated with diabetes incidence (HR = 1.43, 95% CI: 1.03, 1.98) in MASLD population overall. Further in-depth analysis revealed a nonlinear relationship between the TyG index and the occurrence of diabetes. When the TyG index was less than 7.95, it exhibited a negative correlation (HR = 0.22, 95% CI: 0.07, 0.66); however, when the TyG index exceeded 7.95, it showed a positive correlation (HR = 1.68, 95% CI: 1.20, 2.36). Additionally, we conducted a subject working curve analysis on the TyG index and the two components used to calculate it, fasting blood glucose and triglycerides, and found that the TyG index demonstrated higher sensitivity and specificity in predicting the occurrence of diabetes in the MASLD population. TyG index exhibits a nonlinear relationship with the incidence of diabetes in the MASLD population, and its sensitivity and specificity are superior to those of fasting blood glucose and triglycerides.
Collapse
Affiliation(s)
- Tengchao Zhou
- Department of Interventional Vascular Surgery, Hunan Provincial People's Hospital (The First-affiliated Hospital of Hunan Normal University), Jiefang West Road, Changsha, Hunan, 410000, P. R. China
| | - Canbin Xie
- Department of Interventional Vascular Surgery, Hunan Provincial People's Hospital (The First-affiliated Hospital of Hunan Normal University), Jiefang West Road, Changsha, Hunan, 410000, P. R. China.
| |
Collapse
|
29
|
Grossini E, Ola Pour MM, Venkatesan S. The Role of Extracellular Vesicles in the Pathogenesis of Metabolic Dysfunction-Associated Steatotic Liver Disease and Other Liver Diseases. Int J Mol Sci 2025; 26:5033. [PMID: 40507843 PMCID: PMC12154092 DOI: 10.3390/ijms26115033] [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: 03/25/2025] [Revised: 05/09/2025] [Accepted: 05/19/2025] [Indexed: 06/16/2025] Open
Abstract
The increasing prevalence of liver diseases, such as metabolic dysfunction-associated steatotic liver disease (MASLD), presents considerable medical challenges, particularly given the absence of approved pharmacological treatments, which underscores the necessity to comprehend its underlying mechanisms. Extracellular vesicles (EVs), which are tiny particles released by cells, play a crucial role in facilitating communication and can transport harmful molecules that promote inflammation and tissue damage. These EVs are involved in the progression of various types of liver disorders since they aggravate inflammation and oxidative stress. Because of their critical role, it is believed that EVs are widely involved in the initiation and progression of MASLD, as well as in viral hepatitis, alcoholic liver disease, drug-induced liver injury, and hepatocellular carcinoma. This review emphasizes recent findings regarding the functions of EVs in the above liver pathologies and underscores their potential as new therapeutic targets, paving the way for innovative approaches to address those detrimental liver conditions.
Collapse
Affiliation(s)
- Elena Grossini
- Laboratory of Physiology, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (M.M.O.P.); (S.V.)
| | | | | |
Collapse
|
30
|
Zhang Q, Wang Y, Yang Z, Xin Z, Deng H, Chen W. Carrier Free 1,2,3,4,6- O-Pentagalloylglucose Nanoparticles for Treatment of Acute Lung Injury. Bioconjug Chem 2025; 36:1121-1131. [PMID: 40338811 DOI: 10.1021/acs.bioconjchem.5c00197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Acute lung injury (ALI) is a severe lung disease with a high mortality rate, and novel therapeutic strategies must be developed crucially. The amelioration of inflammation and oxidative stress is a promising strategy for alleviating ALI. 1,2,3,4,6-O-pentagalloylglucose (PGG) has a pronounced therapeutic effect on ALI, with excellent anti-inflammatory and antioxidant effects. However, poor solubility and low bioavailability have affected its clinical application. In this study, carrier-free PGG nanoparticles (PGG NPs) were prepared by antisolvent precipitation method. PGG NPs have been engineered to improve solubility, sustained release behavior, and higher bioavailability than free PGG. Moreover, the pharmacodynamic results showed that the remarkable protective effect of PGG NPs on ALI in rats is better than that of free PGG, which is related to the activation of Nrf2/Keap1/HO-1/NLRP3 pathway. Overall, this study not only demonstrates the efficacy and safety of PGG against ALI, but also holds promise as a carrier-free nanodrug system.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Natural Medicine, School of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, China
- Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Ying Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Zixuan Yang
- Department of Natural Medicine, School of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Zhiming Xin
- Fujian Research Center of Drug's Non-clinical Safety Evaluation, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Haohua Deng
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, China
| | - Wei Chen
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, Fujian, China
| |
Collapse
|
31
|
D’Amico T, Miglionico M, Cangemi R, Romiti GF, De Fabrizio B, Fasano S, Recchia F, Stefanini L, Raparelli V, Violi F, Basili S, P. R. O.-LIVER Collaborators, Palasciano G, D’Alitto F, Palmieri VO, Santovito D, Di Michele D, Croce G, Sacerdoti D, Brocco S, Fasolato S, Cecchetto L, Bombonato G, Bertoni M, Restuccia T, Andreozzi P, Liguori ML, Perticone F, Caroleo B, Perticone M, Staltari O, Manfredini R, De Giorgi A, Averna M, Giammanco A, Granito A, Pettinari I, Marinelli S, Bolondi L, Falsetti L, Salvi A, Durante-Mangoni E, Cesaro F, Farinaro V, Ragone E, Morana I, Andriulli A, Ippolito A, Iacobellis A, Niro G, Merla A, Raimondo G, Maimone S, Cacciola I, Varvara D, Drenaggi D, Staffolani S, Picardi A, Vespasiani-Gentilucci U, Galati G, Gallo P, Davì G, Schiavone C, Santilli F, Tana C, Licata A, Soresi M, Bianchi GB, Carderi I, Pinto A, Tuttolomondo A, Ferrari G, Gresele P, Fierro T, Morelli O, Laffi G, Romanelli RG, Arena U, Stasi C, Gasbarrini A, Gargovich M, Zocco MA, Riccardi L, Ainora ME, Capeci W, Martino GP, Nobili L, Cavallo M, Frugiuele P, Greco A, Pietrangelo A, Ventura P, Cuoghi C, Marcacci M, Serviddio G, Vendemiale G, Villani R, Gargano R, Vidili G, Di Cesare V, et alD’Amico T, Miglionico M, Cangemi R, Romiti GF, De Fabrizio B, Fasano S, Recchia F, Stefanini L, Raparelli V, Violi F, Basili S, P. R. O.-LIVER Collaborators, Palasciano G, D’Alitto F, Palmieri VO, Santovito D, Di Michele D, Croce G, Sacerdoti D, Brocco S, Fasolato S, Cecchetto L, Bombonato G, Bertoni M, Restuccia T, Andreozzi P, Liguori ML, Perticone F, Caroleo B, Perticone M, Staltari O, Manfredini R, De Giorgi A, Averna M, Giammanco A, Granito A, Pettinari I, Marinelli S, Bolondi L, Falsetti L, Salvi A, Durante-Mangoni E, Cesaro F, Farinaro V, Ragone E, Morana I, Andriulli A, Ippolito A, Iacobellis A, Niro G, Merla A, Raimondo G, Maimone S, Cacciola I, Varvara D, Drenaggi D, Staffolani S, Picardi A, Vespasiani-Gentilucci U, Galati G, Gallo P, Davì G, Schiavone C, Santilli F, Tana C, Licata A, Soresi M, Bianchi GB, Carderi I, Pinto A, Tuttolomondo A, Ferrari G, Gresele P, Fierro T, Morelli O, Laffi G, Romanelli RG, Arena U, Stasi C, Gasbarrini A, Gargovich M, Zocco MA, Riccardi L, Ainora ME, Capeci W, Martino GP, Nobili L, Cavallo M, Frugiuele P, Greco A, Pietrangelo A, Ventura P, Cuoghi C, Marcacci M, Serviddio G, Vendemiale G, Villani R, Gargano R, Vidili G, Di Cesare V, Masala M, Delitala G, Invernizzi P, Di Minno G, Tufano A, Purrello F, Privitera G, Forgione A, Curigliano V, Senzolo M, Rodríguez-Castro KI, Giannelli G, Serra C, Neri S, Rizzetto M, Debernardi Venon W, Svegliati Baroni G, Bergamaschi G, Masotti M, Costanzo F, Corazza GR, Caldwell SH, Angelico F, Del Ben M, Napoleone L, Polimeni L, Proietti M, Raparelli V, Romiti GF, Ruscio E, Severoni A, Talerico G, Toriello F, Vestri A, Stefanini L, Rumbolà L, Buoninfante G, Maiorca F, Sabetta A, Di Cola S. Neutrophil–lymphocyte ratio is associated with worse outcomes in patients with cirrhosis: insights from the PRO-LIVER Registry. Intern Emerg Med 2025. [DOI: 10.1007/s11739-025-03955-x] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/17/2025] [Indexed: 06/02/2025]
Abstract
Abstract
Background
Liver cirrhosis (LC) is a leading global cause of morbidity and mortality, with inflammation playing a key role in disease progression and clinical complications of LC. The Neutrophil/Lymphocyte Ratio (NLR), a readily available marker of systemic inflammation, has been linked to short-term adverse outcomes in LC, but data on long-term follow-up are limited. This study aimed to investigate the relationship between NLR and long-term all-cause mortality in an unselected cohort of LC patients.
Methods
Data were gathered from the Italian multicenter observational study “PRO-LIVER”. Patients with available data to calculate NLR at baseline were included. Baseline clinical determinants of NLR and the association of NRL with all-cause mortality at 2-year follow-up were evaluated.
Results
From the overall cohort (n = 753), 506 patients with LC (31% female, mean age 64.8 ± 11.9 years) were included in the analysis. Median value of NLR was 2.42 (Interquartile Range [IQR]: 1.61–3.52). At baseline, patients with NLR ≥ 2.42 were more likely to have Child–Pugh B or C, hepatocellular carcinoma (HCC), or portal vein thrombosis (PVT). After a median follow-up of 21 months, 129 patients died: 44 (17%) with NLR < 2.42 and 85 (34%) with NLR ≥ 2.42 (p < 0.001). At multiple-adjusted Cox regression analysis, NLR ≥ 2.42 was independently associated with all-cause mortality (HR: 1.65; 95% CI: 1.12–2.44; p = 0.012), along with age, Child–Pugh C class, HCC and PVT.
Conclusions
NLR is associated with long-term all-cause mortality in LC. NLR may serve as a potentially easily available tool to aid risk refinement in LC.
Trial registration number
ClinicalTrials.gov Identifier: NCT01470547.
Collapse
|
32
|
Kong J, Han X, Wei C. Causal relationship between metabolic dysfunction-associated fatty liver disease and endotoxin biomarkers: A Mendelian randomization study. Medicine (Baltimore) 2025; 104:e42311. [PMID: 40388727 PMCID: PMC12091621 DOI: 10.1097/md.0000000000042311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 04/14/2025] [Indexed: 05/21/2025] Open
Abstract
Although the relationship among lipopolysaccharides (LPS), LPS-binding proteins, and metabolic dysfunction-associated fatty liver disease (MAFLD) is widely studied, no conclusive evidence is available. In this study, we used mendelian randomization (MR) to study the causal relationship of LPS, LPS-binding proteins, and MAFLD. Using bidirectional two-sample MR method, we evaluated data from the genome wide association study; for this analysis, nonalcoholic fatty liver disease (NAFLD), liver fat percentage, and other metabolic syndromes were employed as outcomes. Furthermore, MR analysis mainly involved the inverse variance weighted method. Heterogeneity and pleiotropy tests were also conducted. LPS was found to have a causal relationship with NAFLD, obesity, high density lipoprotein cholesterol, and TG levels. Furthermore, TG levels and LBP had significant causal relationships. This study mainly concluded that LPS is a risk factor for NAFLD, obesity, high density lipoprotein cholesterol, and TG, corroborating it's the LPS role in MAFLD pathogenesis. Hence, optimizing the gut microbiota using proper diet, probiotics, or fecal microbiota transplantation may help to reduce inflammation and (IR), thereby improving lipid and glucose metabolism disorders. Although a causal relationship between TG and LBP was observed, further studies are required to determine a specific mechanism.
Collapse
Affiliation(s)
- Jingwen Kong
- Jining Medical University, Jining, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Xixi Han
- Jining Medical University, Jining, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Chao Wei
- Jining Medical University, Jining, China
| |
Collapse
|
33
|
El-Kassas M, Al-Naamani K, Elbadry M, Awad A, Tharwat M, Debzi N, Zemmouchi S, Abdulla M, Zakaria D, Esmat G, El-Karaksy H, Waked I, Shaltout I, Medhat MA, El-Shabrawi M, Abdeen N, Al-Khairalla M, Akroush MW, Alali AA, Almattooq M, Yaghi C, Tumi A, Elmehdawi R, Benazzouz M, Attia MF, Sanai F, Idlbi S, Labidi A, Houni AE, Beshyah S, Lakhdar A, Atef Z, Abdel Rahman AG, Saleh R, Al-Rifai A, Alqahtani S, Elzouki AN, Alswat K. Establishing consensus on Arabic medical terminology for steatotic liver disease: a mixed-methods approach. Arab J Gastroenterol 2025:S1687-1979(25)00009-7. [PMID: 40379545 DOI: 10.1016/j.ajg.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/03/2024] [Accepted: 01/11/2025] [Indexed: 05/19/2025]
Affiliation(s)
- Mohamed El-Kassas
- Endemic Medicine Department, Faculty of Medicine, Helwan University, Cairo, Egypt; Steatotic Liver Disease Study Foundation in the Middle East and North Africa (SLMENA), Cairo, Egypt.
| | - Khalid Al-Naamani
- Steatotic Liver Disease Study Foundation in the Middle East and North Africa (SLMENA), Cairo, Egypt; Department of Medicine, Division of Gastroenterology and Hepatology, The Medical City for Military and Security Services, Muscat, Oman
| | - Mohamed Elbadry
- Endemic Medicine Department, Faculty of Medicine, Helwan University, Cairo, Egypt; Steatotic Liver Disease Study Foundation in the Middle East and North Africa (SLMENA), Cairo, Egypt
| | - Abeer Awad
- Department of Internal Medicine, Hepatogastroenterology Unit, Kasr Al-Ainy School of Medicine, Cairo, Egypt
| | - Mina Tharwat
- Steatotic Liver Disease Study Foundation in the Middle East and North Africa (SLMENA), Cairo, Egypt; Department of Tropical Medicine and Gastroenterology, Aswan University, Aswan, Egypt
| | - Nabil Debzi
- Steatotic Liver Disease Study Foundation in the Middle East and North Africa (SLMENA), Cairo, Egypt; Hepatology Department, Mustapha Bacha University Hospital, Algeria, Algeria
| | | | - Maheeba Abdulla
- Internal Medicine Department, Ibn AlNafees Hospital, Manama, Bahrain
| | - Doaa Zakaria
- Tropical Medicine Department, Ain Shams University, Cairo, Egypt
| | - Gamal Esmat
- Steatotic Liver Disease Study Foundation in the Middle East and North Africa (SLMENA), Cairo, Egypt; Endemic Medicine Department, Cairo University, Cairo, Egypt; Badr University in Cairo Research Center, Badr University in Cairo, Badr City, Cairo, Egypt
| | - Hanaa El-Karaksy
- Pediatrics Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Imam Waked
- Steatotic Liver Disease Study Foundation in the Middle East and North Africa (SLMENA), Cairo, Egypt; Hepatology Department, National Liver Institute, Menoufia University, Shebeen ElKom, Egypt
| | - Inass Shaltout
- Internal Medicine and Diabetes Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohammed A Medhat
- Steatotic Liver Disease Study Foundation in the Middle East and North Africa (SLMENA), Cairo, Egypt; Tropical Medicine and Gastroenterology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | | | - Nermeen Abdeen
- Tropical Medicine Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | | | - Maisam W Akroush
- Steatotic Liver Disease Study Foundation in the Middle East and North Africa (SLMENA), Cairo, Egypt; Digestive and Liver Disease Clinic, Private Sector, Faculty of Medicine, Jordan University, Amman, Jordan
| | - Ali A Alali
- Department of Medicine, Faculty of Medicine, Kuwait University, Jabriyah, Kuwait; Thunayan Alghanim Gastroenterology and Hepatology Center, Amiri Hospital, Sharq, Kuwait
| | - Maen Almattooq
- Steatotic Liver Disease Study Foundation in the Middle East and North Africa (SLMENA), Cairo, Egypt; Gastroenterology & Hepatology Department, Jaber Hospital, Kuwait
| | - Cesar Yaghi
- Gastroenterology and Hepatology Department, Hotel-Dieu de France University Hospital, Beirut, Lebanon
| | - Ali Tumi
- Steatotic Liver Disease Study Foundation in the Middle East and North Africa (SLMENA), Cairo, Egypt; Medical Department, Central Hospital, Tripoli, Libya
| | - Rafik Elmehdawi
- Internal Medicine Department, University of Benghazi, Benghazi, Libya
| | - Mustapha Benazzouz
- Steatotic Liver Disease Study Foundation in the Middle East and North Africa (SLMENA), Cairo, Egypt; Hepatogastroenterology Department, Rabat International University, Rabat, Morocco
| | - Mona F Attia
- Department of English Language and Literature, Helwan University, Cairo, Egypt
| | - Faisal Sanai
- Steatotic Liver Disease Study Foundation in the Middle East and North Africa (SLMENA), Cairo, Egypt; Gastroenterology Section, Department of Medicine, King Abdulaziz Medical City, King Abdullah International Medical Research Center, Ministry of National Guard - Health Affairs, Jeddah, Saudi Arabia
| | - Sahar Idlbi
- General Diseases Department, University's Children Hospital, Damascus, Syria
| | - Asma Labidi
- Steatotic Liver Disease Study Foundation in the Middle East and North Africa (SLMENA), Cairo, Egypt; Gastroenterology "A" Department, Rabta Hospital, Tunisia
| | - Ali El Houni
- Medicine Department, Dubai Medical University, Dubai, United Arab Emirates
| | - Salem Beshyah
- Endocrinology Department, Yas Clinic Khalifa City, Abu Dhabi, United Arab Emirates
| | - Abdul Lakhdar
- Endocrinology Department, Barts Health NHS Trust, London, UK
| | - Zayed Atef
- Internal Medicine Department, Faculty of Medicine, Sana'a University, Sana'a, Yemen
| | - Amira G Abdel Rahman
- Department of Public Health and Community, Environmental and Occupational Medicine, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Rasha Saleh
- Arabic Language and Literature Department, Helwan University, Cairo, Egypt
| | - Ahmad Al-Rifai
- Division of Gastroenterology, Sheikh Shakhbout Medical City, Abu Dhabi, United Arab Emirates
| | - Saleh Alqahtani
- Organ Transplant Center of Excellence, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Abdel-Naser Elzouki
- Steatotic Liver Disease Study Foundation in the Middle East and North Africa (SLMENA), Cairo, Egypt; Medicine Department, Hamad Medical Corporation and College of Medicine, Qatar University, Doha, Qatar
| | - Khalid Alswat
- Steatotic Liver Disease Study Foundation in the Middle East and North Africa (SLMENA), Cairo, Egypt; Liver Disease Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
34
|
Carbone F, Després JP, Ioannidis JPA, Neeland IJ, Garruti G, Busetto L, Liberale L, Ministrini S, Vilahur G, Schindler TH, Macedo MP, Di Ciaula A, Krawczyk M, Geier A, Baffy G, Faienza MF, Farella I, Santoro N, Frühbeck G, Yárnoz-Esquiroz P, Gómez-Ambrosi J, Chávez-Manzanera E, Vázquez-Velázquez V, Oppert JM, Kiortsis DN, Sbraccia P, Zoccali C, Portincasa P, Montecucco F. Bridging the gap in obesity research: A consensus statement from the European Society for Clinical Investigation. Eur J Clin Invest 2025:e70059. [PMID: 40371883 DOI: 10.1111/eci.70059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/12/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Most forms of obesity are associated with chronic diseases that remain a global public health challenge. AIMS Despite significant advancements in understanding its pathophysiology, effective management of obesity is hindered by the persistence of knowledge gaps in epidemiology, phenotypic heterogeneity and policy implementation. MATERIALS AND METHODS This consensus statement by the European Society for Clinical Investigation identifies eight critical areas requiring urgent attention. Key gaps include insufficient long-term data on obesity trends, the inadequacy of body mass index (BMI) as a sole diagnostic measure, and insufficient recognition of phenotypic diversity in obesity-related cardiometabolic risks. Moreover, the socio-economic drivers of obesity and its transition across phenotypes remain poorly understood. RESULTS The syndemic nature of obesity, exacerbated by globalization and environmental changes, necessitates a holistic approach integrating global frameworks and community-level interventions. This statement advocates for leveraging emerging technologies, such as artificial intelligence, to refine predictive models and address phenotypic variability. It underscores the importance of collaborative efforts among scientists, policymakers, and stakeholders to create tailored interventions and enduring policies. DISCUSSION The consensus highlights the need for harmonizing anthropometric and biochemical markers, fostering inclusive public health narratives and combating stigma associated with obesity. By addressing these gaps, this initiative aims to advance research, improve prevention strategies and optimize care delivery for people living with obesity. CONCLUSION This collaborative effort marks a decisive step towards mitigating the obesity epidemic and its profound impact on global health systems. Ultimately, obesity should be considered as being largely the consequence of a socio-economic model not compatible with optimal human health.
Collapse
Affiliation(s)
- Federico Carbone
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Genoa, Italy
| | - Jean-Pierre Després
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Québec, Québec, Canada
- VITAM - Centre de Recherche en santé Durable, Centre intégré Universitaire de santé et de Services Sociaux de la Capitale-Nationale, Québec, Québec, Canada
| | - John P A Ioannidis
- Department of Medicine, Stanford Cardiovascular Institute, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, USA
- Department of Epidemiology and Population Health, Stanford Cardiovascular Institute, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, USA
- Department of Biomedical Science, Stanford Cardiovascular Institute, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, USA
| | - Ian J Neeland
- Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Cardiovascular Disease, Harrington Heart and Vascular Institute, Cleveland, Ohio, USA
| | - Gabriella Garruti
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Luca Busetto
- Department of Medicine, University of Padua, Padua, Italy
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Genoa, Italy
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Cardiology Department, Luzerner Kantonspital, Lucerne, Switzerland
| | - Gemma Vilahur
- Research Institute, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, IIB-Sant Pau, Barcelona, Spain
- CiberCV, Institute Carlos III, Madrid, Spain
| | - Thomas H Schindler
- Washington University in St. Louis, Mallinckrodt Institute of Radiology, Division of Nuclear Medicine, Cardiovascular Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Maria Paula Macedo
- APDP - Diabetes Portugal, Education and Research Center, Lisbon, Portugal
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS | FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Agostino Di Ciaula
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Marcin Krawczyk
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Essen, Germany
- Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver Surgery, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Andreas Geier
- Interdisciplinary Amyloidosis Center of Northern Bavaria, University Hospital of Würzburg, Würzburg, Germany
- Department of Internal Medicine II, Hepatology, University Hospital of Würzburg, Würzburg, Germany
| | - Gyorgy Baffy
- Department of Medicine, VA Boston Healthcare System, Harvard Medical School, Boston, Massachusetts, USA
| | - Maria Felicia Faienza
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Ilaria Farella
- Department of Medicine and Surgery, LUM University, Casamassima, Italy
| | - Nicola Santoro
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Medicine and Health Sciences, "V. Tiberio" University of Molise, Campobasso, Italy
| | - Gema Frühbeck
- Department of Endocrinology and Nutrition, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBERObn (CIBER Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Patricia Yárnoz-Esquiroz
- Department of Endocrinology and Nutrition, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBERObn (CIBER Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Gómez-Ambrosi
- Department of Endocrinology and Nutrition, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBERObn (CIBER Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Emma Chávez-Manzanera
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Jean-Michel Oppert
- Department of Nutrition, Pitié-Salpêtrière Hospital (AP-HP), Human Nutrition Research Center Ile-de-France (CRNH IdF), Sorbonne University, Paris, France
| | - Dimitrios N Kiortsis
- Atherothrombosis Research Centre, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Paolo Sbraccia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carmine Zoccali
- Renal Research Institute, New York, New York, USA
- Institute of Molecular Biology and Genetics (Biogem), Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia Trapianto Renale (IPNET), c/o Nefrologia, Grande Ospedale Metropolitano, Reggio Calabria, Italy
| | - Piero Portincasa
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Genoa, Italy
| |
Collapse
|
35
|
Losasso MR, Parussolo MLC, Oliveira Silva A, Direito R, Quesada K, Penteado Detregiachi CR, Bechara MD, Méndez-Sánchez N, Abenavoli L, Araújo AC, de Alvares Goulart R, Guiger EL, Fornari Laurindo L, Maria Barbalho S. Unraveling the Metabolic Pathways Between Metabolic-Associated Fatty Liver Disease (MAFLD) and Sarcopenia. Int J Mol Sci 2025; 26:4673. [PMID: 40429815 PMCID: PMC12111209 DOI: 10.3390/ijms26104673] [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: 04/03/2025] [Revised: 05/10/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Metabolic-Associated Fatty Liver Disease (MAFLD) is a public health concern that is constantly expanding, with a fast-growing prevalence, and it affects about a quarter of the world's population. This condition is a significant risk factor for cardiovascular, hepatic, and oncologic diseases, such as hypertension, hepatoma, and atherosclerosis. Sarcopenia was long considered to be an aging-related syndrome, but today, it is acknowledged to be secondarily related to chronic diseases such as metabolic syndrome, cardiovascular conditions, and liver diseases, among other comorbidities associated with insulin resistance and chronic inflammation, besides inactivity and poor nutrition. The physiopathology involving MAFLD and sarcopenia has still not been solved. Inflammation, oxidative stress, mitochondrial dysfunction, and insulin resistance seem to be some of the keys to this relationship since this hormone target is mainly the skeletal muscle. This review aimed to comprehensively discuss the main metabolic and physiological pathways involved in these conditions. MAFLD and sarcopenia are interconnected by a complex network of pathophysiological mechanisms, such as insulin resistance, skeletal muscle tissue production capacity, chronic inflammatory state, oxidative stress, and mitochondrial dysfunction, which are the main contributors to this relationship. In addition, in a clinical analysis, patients with sarcopenia and MAFLD manifest more severe hepatitis fibrosis when compared to patients with only MAFLD. These patients, with both disorders, also present clinical improvement in their MAFLD when treated for sarcopenia, reinforcing the association between them. Lifestyle changes accompanied by non-pharmacological interventions, such as dietary therapy and increased physical activity, undoubtedly improve this scenario.
Collapse
Affiliation(s)
- Marina Ribas Losasso
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Maria Luiza Cesto Parussolo
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Antony Oliveira Silva
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Rosa Direito
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines, Universidade de Lisboa (iMed.ULisboa), Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Karina Quesada
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Claudia Rucco Penteado Detregiachi
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Marcelo Dib Bechara
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Nahum Méndez-Sánchez
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico
- Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Ludovico Abenavoli
- Department of Health Sciences, University “Magna Graecia”, Viale Europa, 88100 Catanzaro, Italy
| | - Adriano Cressoni Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Ricardo de Alvares Goulart
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Elen Landgraf Guiger
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, SP, Brazil
- Research Coordinator, UNIMAR Charity Hospital, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| |
Collapse
|
36
|
Fan X, Tang Q, Xia N, Wang J, Zhao W, Jin M, Lu Q, Hu J, Zhang R, Zhang L, Jiang Z, Yu Q. Immune-endothelial cell crosstalk in hepatic endothelial injury of liver fibrotic mice. Eur J Pharmacol 2025; 1000:177730. [PMID: 40374060 DOI: 10.1016/j.ejphar.2025.177730] [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: 12/27/2024] [Revised: 05/09/2025] [Accepted: 05/13/2025] [Indexed: 05/17/2025]
Abstract
INTRODUCTION Liver fibrosis is a common pathological process in chronic liver disease, reflecting the advanced stage of the disease. Liver endothelial cells (ECs), especially liver sinusoidal endothelial cells (LSECs), are recognized as critical modulators of liver homeostasis and play essential roles in the recruitment and function of liver immune cells. In this study, we aimed to explore the mechanism of hepatic EC injury and the potential regulatory pathways of intercellular communication in liver fibrosis. METHODS In this study, C57BL/6 male mice were treated with CCl4 for 6 weeks to establish a liver fibrosis model. Masson staining and immunohistochemistry were performed to assess the extent of liver fibrosis. Hepatic endothelial injury was detected by using scanning electron microscopy (SEM) and PCR technology. Single-cell RNA sequencing (scRNA-seq) was performed to analyze phenotypic changes in nonparenchymal cells and dissect intercellular crosstalk. RESULTS A total of 24,534 cells were clustered into 10 main cell subsets. The LSEC fenestrae and surface receptor expression were reduced, and the expression of Cd34 was upregulated. Liver ECs exhibited dense cellular crosstalk with immune cells (macrophages, T and B cells). The analysis of intercellular signaling pathways revealed that immune cells targeted liver ECs through the Ptprc-Mrc1 and Sell-Podxl signaling pathways to maintain cellular interactions during liver fibrosis. CONCLUSION We revealed apparent damage and capillarization of liver ECs and demonstrated the cell-cell communications among liver immune cells and ECs during the development of liver fibrosis. The Ptprc-Mrc1 and Sell-Podxl signaling pathways exerted prominent roles in liver immune cell-EC interactions.
Collapse
Affiliation(s)
- Xue Fan
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Qianhui Tang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Ninglin Xia
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Jiwei Wang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Wen Zhao
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Ming Jin
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Qian Lu
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Jinyu Hu
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Rongmi Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Luyong Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Zhenzhou Jiang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China.
| | - Qinwei Yu
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
37
|
Wang X, Shi SR, Sun MM, Zhang XY, Zhang XH, Song SL, Yin F, Guo SD. Mechanisms of action of Fucus vesiculosus-derived fucoidan on improving dyslipidemia in New Zealand rabbits fed a high-fat diet. Int J Biol Macromol 2025; 314:144148. [PMID: 40368205 DOI: 10.1016/j.ijbiomac.2025.144148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/30/2025] [Accepted: 05/10/2025] [Indexed: 05/16/2025]
Abstract
Dyslipidemia is a major contributor to various diseases, including atherosclerotic cardiovascular disease and obesity. Treatment strategies for dyslipidemia continue to evolve as our understanding of this metabolic disorder and potential therapeutic candidates advance. Notably, fucoidan demonstrates promising effects in ameliorating dyslipidemia in rodents, although their lipid metabolism differs significantly from humans. This study, investigates the lipid-regulatory effects of Fucus vesiculosus-derived fucoidan (FvF) and elucidates the underlying mechanisms of action using New Zealand rabbits fed a high-fat diet, whose lipid profiles closely resemble those of patients with dyslipidemia. The results demonstrate that FvF intervention ameliorates dyslipidemia and lipid deposition in a dose-dependent manner. Mechanistically, FvF intervention modulates the expression levels of multiple molecules involved in lipid transport, fatty acid synthesis and beta-oxidation, and redox balance, as revealed by quantitative reverse transcription polymerase chain reaction, western blotting, and proteomic analysis. This study is the first to report that FvF, consisting of alternating [→4)-α-L-Fucp(1 → 3)-α-L-Fucp(1→] glycosyls ameliorates dyslipidemia by directly modulating lipid metabolism and indirectly attenuating oxidative stress. These findings suggest that FvF holds significant potential as a candidate for the treatment of lipid disorder-related diseases.
Collapse
Affiliation(s)
- Xue Wang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Shan-Rui Shi
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Min-Min Sun
- School of Stomatology, Shandong Second Medical University, Weifang 261053, China
| | - Xue-Ying Zhang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Xu-Hang Zhang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Shi-Lin Song
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Fan Yin
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
38
|
Li Q, Meng Q, Niu N, Li Y, Lu W, Wang T, Li Y, Ju W, Ma Y, Wu J. Yinchenzhufu decoction ameliorates cholestatic liver injury through decreasing the formation of neutrophil extracellular traps in mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119720. [PMID: 40174730 DOI: 10.1016/j.jep.2025.119720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/24/2025] [Accepted: 03/29/2025] [Indexed: 04/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Neutrophil extracellular traps (NETs) are important driving factors to liver disease. Yinchenzhufu decoction (YCZFD) is a classical traditional herbal medicine used to treat cholestatic liver injury (CLI); however, its effect on NETs in CLI remains unknown. AIM OF THE STUDY To elucidate the potential molecular mechanism of YCZFD against CLI through multiple omics techniques. MATERIALS AND METHODS The protective effects of YCZFD against cholestatic liver injury and fibrosis were investigated in a 3, 5-Diethoxycarbonyl-1, 4-Dihydro-2, 4, 6-Collidine (DDC) induced cholestatic mouse model. Its effects on blood biochemical indicators, liver tissue morphology, neutrophil infiltration, and fibrosis in cholestatic mice were determined. Then RNA sequencing, quantitative proteomics, and molecular biology were used to analyze the mechanism of YCZFD against CLI. RESULTS Treatment different doses of YCZFD significantly ameliorated neutrophil infiltration, alleviating CLI in cholestatic mice. RNA sequencing showed that neutrophil recruitment genes, such as CXCL5 and CXCR2, were upregulated in the liver tissues of cholestatic mice, but YCZFD treatment decreased this expression. Additionally, quantitative proteomics revealed that liver myeloperoxidase and S100A9 expression, key marker proteins of NETs, was significantly increased in cholestatic mice and decreased by YCZFD. Similarly, molecular biology confirmed that YCZFD reduced NETs proteins expression and ameliorated NETs formation in the liver of cholestatic mice. As NETs promotes inflammation, further results showed that YCZFD treatment inhibited the NF-κB signaling pathway and collagen formation, alleviating liver inflammation and fibrosis of cholestatic mice. CONCLUSIONS YCZFD decreases neutrophil recruitment and formation of NETs, in turn alleviating cholestatic liver injury and fibrosis.
Collapse
Affiliation(s)
- Qi Li
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qian Meng
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ningning Niu
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiqian Li
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenye Lu
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianming Wang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanyuan Li
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Wanjun Ju
- Department of Endocrinology, Shanghai Pudong New Area Hospital of Traditional Chinese Medicine, Shanghai, China.
| | - Yueming Ma
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jiasheng Wu
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
39
|
He Y, Ke H, Zhu J, Yuan X, Li H, Wu W, Yang S, Yu H. Construction and validation of a meropenem-induced liver injury risk prediction model: a multicenter case-control study. Front Pharmacol 2025; 16:1542554. [PMID: 40417215 PMCID: PMC12098429 DOI: 10.3389/fphar.2025.1542554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/25/2025] [Indexed: 05/27/2025] Open
Abstract
Objective To construct and validate a risk prediction model for patients with meropenem-induced liver injury (MiLI). Methods A retrospective case-control study was conducted to collect data on inpatients treated with meropenem at Shiyan People's Hospital, Hubei, China from January 2018 to December 2022; this study served as the model construction dataset. Univariate analysis and multiple logistic regression analysis were employed to identify the related factors for MiLI, and a nomogram risk prediction model for MiLI was constructed. The recognition ability and prediction accuracy of the model were evaluated using the receiver operating characteristic (ROC) and calibration curves. The clinical efficacy was assessed via the decision curve analysis (DCA). The internal validation was performed using the bootstrap method, and external validation was conducted based on an external dataset from Shiyan Taihe Hospital between October 2021 and December 2023. Results A total of 1,625 individuals were included in the model construction dataset, of which 62 occurred MiLI. The external validation dataset included 1,032 cases, with 74 patients developing liver injury. Six variables were independent factors for MiLI and included in the final prediction model: being male (OR = 2.080, 95% CI: 1.050-4.123, P = 0.036), ICU admission (OR = 8.207, 95% CI: 4.094-16.453, P < 0.001), gallbladder disease (OR = 8.240, 95% CI: 3.605-18.832, P < 0.001), baseline ALP (OR = 1.012, 95% CI: 1.004-1.019, P = 0.004), GGT (OR = 1.010, 95% CI: 1.005-1.015, P < 0.001), and PLT (OR = 0.997, 95% CI: 0.994-0.999, P = 0.020). The c-statistic value for internal validation of the prediction model was 0.821; the sensitivity and specificity were 0.997 and 0.924, respectively. The c-statistic value of the prediction model in the model construction dataset was 0.837 (95% CI, 0.789-0.885), while in the external validation dataset was 0.851 (95% CI, 0.802-0.901). The P-values of the calibration curve in the two datasets were 0.935 and 0.084, respectively. Conclusion Being male, ICU admission, gallbladder disease, higher levels of baseline ALP and GGT, and lower levels of baseline PLT were the risk factors for MiLI. The nomogram model built based on these factors demonstrated favorable performance in discrimination, calibration, clinical applicability, and internal-external validation. The nomogram model can assist clinicians in early identification of high-risk patients receiving meropenem, predicting the risk of MiLI, and ensuring safe medication practices.
Collapse
Affiliation(s)
- Yan He
- Department of Pharmacy, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, Hubei, China
| | - Hongqin Ke
- Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Jianyong Zhu
- Department of Respiratory Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xin Yuan
- Department of Pharmacy, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Hongliang Li
- Department of Pharmacy, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Wenwen Wu
- Department of Preventive Medicine, School of Public Health, Hubei University of Medicine, Shiyan, Hubei, China
| | - Shuman Yang
- Department of Endocrinology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Huibin Yu
- Department of Pharmacy, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
40
|
Li HX, Fei J, Xu W, Peng Y, Yan PJ, Xu Y, Qin G, Teng FY. The characterization and validation of regulated cell death-related genes in chronic rhinosinusitis with nasal polyps. Int Immunopharmacol 2025; 154:114509. [PMID: 40158428 DOI: 10.1016/j.intimp.2025.114509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/20/2025] [Accepted: 03/16/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Regulated cell death (RCD), a genetically controlled process mediated by specialized molecular pathways (commonly termed programmed cell death), plays pivotal roles in diverse pathophysiological processes. However, the landscape and functional implications of RCD subtypes in chronic rhinosinusitis with nasal polyps (CRSwNP) remain poorly characterized. This study aimed to systematically investigate the involvement of RCD mechanisms in the pathogenesis and progression of CRSwNP. METHODS Transcriptomic datasets (GSE136825, GSE23552, GSE198950, GSE196169, GSE156285) related to CRSwNP were retrieved from the Gene Expression Omnibus (GEO) database. A comprehensive panel of 18 RCD-associated gene sets was compiled through a systematic literature review. Gene set variation analysis (GSVA) was employed to profile RCD activation patterns in CRSwNP. Integrative bioinformatics approaches including weighted gene co-expression network analysis (WGCNA) and least absolute shrinkage and selection operator (LASSO) regression were implemented to identify hub RCD-related genes and construct a cell death index (CDI). Single-cell RNA sequencing (scRNA-seq) data were analyzed to map RCD dynamics across cellular subpopulations. Clinical validation was performed using qRT-PCR quantification of key genes in nasal polyp/inferior turbinate tissues, with the concurrent assessment of symptom severity via visual analogue scale (VAS) scores. RESULTS GSVA revealed significant upregulation of 8 RCD subtypes in CRSwNP: apoptosis, ferroptosis, necroptosis, entotic cell death, lysosome-dependent cell death, NETosis, immunogenic cell death, and anoikis. Pathway enrichment analysis demonstrated that RCD-related differentially expressed genes were predominantly involved in epithelial-mesenchymal transition (EMT) and immune-inflammatory regulation. Furthermore, the WGCNA algorithm and LASSO analysis identified 8 key cell death genes (PTHLH, GRINA, S100A9, SCG2, HMOX1, RNF183, TYROBP, SEMA7A), which were utilized to construct the cell death-related index (CDI). In training and validation cohorts, the CDI was significantly elevated in CRSwNP compared to control and exhibited high diagnostic performance, with elevated scores correlating with enhanced immune cell infiltration. Single-cell resolution analysis uncovered cell type-specific RCD activation patterns. Clinical validation confirmed significantly higher expression of S100A9, PTHLH, and HMOX1 in eosinophilic versus non-eosinophilic polyps. Notably, expression levels of PTHLH, S100A9, HMOX1, GRINA, and TYROBP showed strong positive correlations with VAS scores. CONCLUSIONS Our investigation delineates an RCD activation signature in CRSwNP pathogenesis, characterized by 8 key cell death modalities and their regulatory genes. The novel CDI exhibits promising diagnostic potential, while mechanistic insights suggest RCD pathways may drive disease progression through EMT potentiation and inflammatory cascade amplification. These findings provide a framework for developing RCD-targeted therapeutic strategies in CRSwNP.
Collapse
Affiliation(s)
- Hong-Xia Li
- Department of Otolaryngology-Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Luzhou, Sichuan 646000, China
| | - Jing Fei
- Department of Otolaryngology-Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Wei Xu
- Department of Otolaryngology-Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yi Peng
- Department of Otolaryngology-Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Pi-Jun Yan
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Luzhou, Sichuan 646000, China; Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Nephropathy, and Sichuan Clinical Research Center for Diabetes and Metabolic Disease, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yong Xu
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Luzhou, Sichuan 646000, China; Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Nephropathy, and Sichuan Clinical Research Center for Diabetes and Metabolic Disease, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Gang Qin
- Department of Otolaryngology-Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Fang-Yuan Teng
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, and Metabolic Vascular Diseases Key Laboratory of Sichuan-Chongqing Cooperation, Luzhou, Sichuan 646000, China; Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Nephropathy, and Sichuan Clinical Research Center for Diabetes and Metabolic Disease, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
41
|
Li Y, Cao Z, Lu Y, Lei C, Lyu W. Knowledge landscape of macrophage research in liver fibrosis: a bibliometric review of the literature from WoSCC. Front Pharmacol 2025; 16:1571879. [PMID: 40406489 PMCID: PMC12094998 DOI: 10.3389/fphar.2025.1571879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 04/16/2025] [Indexed: 05/26/2025] Open
Abstract
Recent insights into the immune response in fibrosis have provided valuable perspectives for the treatment of liver fibrosis. Macrophages, as the most abundant immune cells in the liver, are key drivers of liver fibrosis. They are extensively involved in tissue damage, chronic inflammation, and the progression and regression of liver fibrosis. This study aims to conduct a bibliometric analysis and literature review on the mechanisms by which macrophages contribute to liver fibrosis. Specifically, we analyzed a bibliometric dataset comprising 1,312 papers from 59 countries, 1,872 institutions, and 9,784 authors. Keyword co-occurrence analysis identified key research hotspots, including the role of macrophage subtypes in obesity-related metabolic disorders, the crosstalk between macrophages and hepatic stellate cells through mechanoimmunology, emerging strategies for immune modulation targeting macrophages to promote fibrosis regression and liver regeneration, and new discoveries regarding macrophage crosstalk with other immune cells. In conclusion, this study provides a visual analysis of the current research landscape, hotspots, and trends in the field of macrophages and liver fibrosis, and discusses future directions for further exploration in this area.
Collapse
Affiliation(s)
- Yanbo Li
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Zhengmin Cao
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Yanping Lu
- Shenzhen Bao’an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Chao Lei
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Wenliang Lyu
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
42
|
Bergentall M, Tremaroli V, Sun C, Henricsson M, Khan MT, Mannerås Holm L, Olsson L, Bergh PO, Molinaro A, Mardinoglu A, Caesar R, Nieuwdorp M, Bäckhed F. Gut microbiota mediates SREBP-1c-driven hepatic lipogenesis and steatosis in response to zero-fat high-sucrose diet. Mol Metab 2025; 97:102162. [PMID: 40345386 DOI: 10.1016/j.molmet.2025.102162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/23/2025] [Accepted: 05/02/2025] [Indexed: 05/11/2025] Open
Abstract
OBJECTIVES Sucrose-rich diets promote hepatic de novo lipogenesis (DNL) and steatosis through interactions with the gut microbiota. However, the role of sugar-microbiota dynamics in the absence of dietary fat remains unclear. This study aimed to investigate the effects of a high-sucrose, zero-fat diet (ZFD) on hepatic steatosis and host metabolism in conventionally raised (CONVR) and germ-free (GF) mice. METHODS CONVR and GF mice were fed a ZFD, and hepatic lipid accumulation, gene expression, and metabolite levels were analyzed. DNL activity was assessed by measuring malonyl-CoA levels, expression of key DNL enzymes, and activation of the transcription factor SREBP-1c. Metabolomic analyses of portal vein plasma identified microbiota-derived metabolites linked to hepatic steatosis. To further examine the role of SREBP-1c, its hepatic expression was knocked down using antisense oligonucleotides in CONVR ZFD-fed mice. RESULTS The gut microbiota was essential for sucrose-induced DNL and hepatic steatosis. In CONVR ZFD-fed mice, hepatic fat accumulation increased alongside elevated expression of genes encoding DNL enzymes, higher malonyl-CoA levels, and upregulation of SREBP-1c. Regardless of microbiota status, ZFD induced fatty acid elongase and desaturase gene expression and increased hepatic monounsaturated fatty acids. Metabolomic analyses identified microbiota-derived metabolites associated with hepatic steatosis. SREBP-1c knockdown in CONVR ZFD-fed mice reduced hepatic steatosis and suppressed fatty acid synthase expression. CONCLUSIONS Sucrose-microbiota interactions and SREBP-1c are required for DNL and hepatic steatosis in the absence of dietary fat. These findings provide new insights into the complex interplay between diet, gut microbiota, and metabolic regulation.
Collapse
Affiliation(s)
- Mattias Bergentall
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, Gothenburg, SE-413 45, Sweden
| | - Valentina Tremaroli
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, Gothenburg, SE-413 45, Sweden
| | - Chuqing Sun
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, Gothenburg, SE-413 45, Sweden
| | - Marcus Henricsson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, Gothenburg, SE-413 45, Sweden
| | - Muhammad Tanweer Khan
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, Gothenburg, SE-413 45, Sweden
| | - Louise Mannerås Holm
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, Gothenburg, SE-413 45, Sweden
| | - Lisa Olsson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, Gothenburg, SE-413 45, Sweden
| | - Per-Olof Bergh
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, Gothenburg, SE-413 45, Sweden
| | - Antonio Molinaro
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, Gothenburg, SE-413 45, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden; Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, UK
| | - Robert Caesar
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, Gothenburg, SE-413 45, Sweden.
| | - Max Nieuwdorp
- Department of (Experimental) Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Fredrik Bäckhed
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, Gothenburg, SE-413 45, Sweden; Department of Clinical Physiology Region Västra Götaland, Sahlgrenska University Hospital Gothenburg Sweden, Sweden.
| |
Collapse
|
43
|
Elangovan H, George J. Guidelines for the evolving landscape of liver disease: From viral hepatitis to MAFLD. Chin Med J (Engl) 2025; 138:1013-1015. [PMID: 40134300 PMCID: PMC12068753 DOI: 10.1097/cm9.0000000000003616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Indexed: 03/27/2025] Open
Affiliation(s)
- Harendran Elangovan
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead Hospital and The University of Sydney, NSW, Australia
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead Hospital and The University of Sydney, NSW, Australia
| |
Collapse
|
44
|
Lyu S, Yang J, Xin X, Sun Q, Cai B, Wang X, An Z, Sun J, Hu Y, Shi L, Feng Q, Gou X. Characteristics of serum bile acid profiles among individuals with metabolic dysfunction-associated steatotic liver disease. BMC Gastroenterol 2025; 25:334. [PMID: 40325371 PMCID: PMC12054156 DOI: 10.1186/s12876-025-03903-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 04/16/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) has become the predominant chronic liver condition globally. Bile acid (BA) metabolism contributes significantly to MASLD progression. In this multicenter clinical study, we aimed to characterize serum BA profiles in patients with MASLD and identify specific alterations compared to healthy controls. METHODS All MASLD cases were sourced from the gastroenterology outpatient departments of Shanghai Baoshan Hospital of Integrated Chinese and Western Medicine, Shanghai Baoshan District Songnan Community Health Service Center, and Lianyungang Oriental Hospital between June 2015 and December 2019. The data were analyzed using SPSS version 26.0, with a p-value of less than 0.05 considered significant. RESULTS A total of 215 participants (35.3% women) with MASLD and 49 controls (44.9% women), aged 18-65 years, were included. MASLD patients showed higher levels of serum total BA (TBA), cholic acid (CA), chenodeoxycholic acid (CDCA), and ursodeoxycholic acid (UDCA) (p < 0.05, p < 0.01) when compared to controls. Furthermore, women patients with MASLD demonstrated notably higher levels of lithocholic acid (LCA), glycolithocholic acid (GLCA), and taurolithocholic acid (TLCA) than men patients with MASLD (p < 0.025, p < 0.01). Compared to women, men exhibited a higher proportion of primary to secondary BAs. Additionally, in men patients with MASLD, the serum concentrations of CA, CDCA, glycocholic acid (GCA), glycochenodeoxycholic acid (GCDCA), and taurochenodeoxycholic acid (TCDCA) exhibited significant negative correlations with ALT levels, while deoxycholic acid (DCA) and TLCA showed negative correlations with BMI. CONCLUSIONS Patients with MASLD exhibited notable variations in BA profiles, including sex-specific differences. This study provides corresponding evidence on the association between BAs and MASLD. TRIAL REGISTRATION Chinese Clinical Trial Registry, NO: ChiCTR-OOC-15006157, registration date: March 25, 2015.
Collapse
Affiliation(s)
- Sheng Lyu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiani Yang
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Qingpu Traditional Chinese Medicine Hospital, Shanghai, China
| | - Xin Xin
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, China
| | - Qinmei Sun
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Beiyu Cai
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Wang
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ziming An
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian Sun
- Clinical Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiyang Hu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, China
| | - Lei Shi
- Clinical Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Qin Feng
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Central Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, China.
| | - Xiaojun Gou
- Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai, China.
| |
Collapse
|
45
|
Froom ZSCS, Callaghan NI, Davenport Huyer L. Cellular crosstalk in fibrosis: insights into macrophage and fibroblast dynamics. J Biol Chem 2025:110203. [PMID: 40334985 DOI: 10.1016/j.jbc.2025.110203] [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: 03/13/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
Pathological fibrosis, the excessive deposition of extracellular matrix and tissue stiffening that causes progressive organ dysfunction, underlies diverse chronic diseases. The fibrotic microenvironment is driven by the dynamic microenvironmental interaction between various cell types; macrophages and fibroblasts play central roles in fibrotic disease initiation, maintenance, and progression. Macrophage functional plasticity to microenvironmental stimuli modulates fibroblast functionality by releasing pro-inflammatory cytokines, growth factors, and matrix remodeling enzymes that promote fibroblast proliferation, activation, and differentiation into myofibroblasts. Activated fibroblasts and myofibroblasts serve as the fibrotic effector cells, secreting extracellular matrix components and initiating microenvironmental contracture. Fibroblasts also modulate macrophage function through the release of their own pro-inflammatory cytokines and growth factors, creating bidirectional crosstalk that reinforces the chronic fibrotic cycle. The intricate interplay between macrophages and fibroblasts, including their secretomes and signaling interactions, leads to tissue damage and pathological loss of tissue function. In this review, we examine macrophage-fibroblast reciprocal dynamic interactions in pathological fibrotic conditions. We discuss the specific lineages and functionality of macrophages and fibroblasts implicated in fibrotic progression, with focus on their signal transduction pathways and secretory signalling that enables their pro-fibrotic behaviour. We then finish with a set of recommendations for future experimentation with the goal of developing a set of potential targets for anti-fibrotic therapeutic candidates. Understanding the cellular interactions between macrophages and fibroblasts provides valuable insights into potential therapeutic strategies to mitigate fibrotic disease progression.
Collapse
Affiliation(s)
- Zachary S C S Froom
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Neal I Callaghan
- Department of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Locke Davenport Huyer
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Biomaterials & Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS B3H 4R2, Canada; Nova Scotia Health, Halifax, NS B3S 0H6, Canada.
| |
Collapse
|
46
|
Kaya E, Yilmaz Y, Alkhouri N. Clinical Insights on Resmetirom: Clinical Indications, Patient Selection, and Monitoring Response to Therapy. J Clin Gastroenterol 2025; 59:412-419. [PMID: 40193288 DOI: 10.1097/mcg.0000000000002150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The recent conditional approval by the Food and Drug Administration of resmetirom for treating metabolic dysfunction-associated steatohepatitis (MASH) with significant or advanced fibrosis represents a pivotal milestone in the history of metabolic dysfunction-associated steatotic liver disease (MASLD) treatment. As the first liver-directed pharmacological therapy option for MASLD, resmetirom offers a novel approach that specifically targets liver pathology, marking a transformative step forward in managing this widespread and challenging condition. For initiating therapy with resmetirom, a liver biopsy is not required. Consequently, accurately excluding patients with less severe liver histology or cirrhosis using noninvasive tests (NITs) is essential. In addition, monitoring the therapy response should be conducted using NITs. Given the recent approval, our current clinical understanding of resmetirom is primarily informed by phase 3 clinical trials. The long-term effects of the drug should be evaluated in further studies by encouraging the use of the drug in eligible patients. This review highlights key aspects of clinical resmetirom use, including identifying the target population, monitoring therapeutic response, determining appropriate discontinuation criteria, and strategies to prevent unnecessary treatment interruptions.
Collapse
Affiliation(s)
- Eda Kaya
- Department of Medicine, Knappschaftskrankenhaus Bochum, Ruhr University, Bochum, Germany
- The Global NASH Council, Washington, DC
| | - Yusuf Yilmaz
- The Global NASH Council, Washington, DC
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdoğan University, Rize, Türkiye
| | | |
Collapse
|
47
|
Luo J, Wang Y, Mao J, Yuan Y, Luo P, Wang G, Zhou S. Features, functions, and associated diseases of visceral and ectopic fat: a comprehensive review. Obesity (Silver Spring) 2025; 33:825-838. [PMID: 40075054 DOI: 10.1002/oby.24239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 03/14/2025]
Abstract
Obesity is a complex, chronic, and recurrent disease marked by abnormal or excessive fat accumulation that poses significant health risks. The distribution of body fat, especially ectopic fat deposition, plays a crucial role in the development of chronic metabolic diseases. Under normal conditions, fatty acids are primarily stored in subcutaneous adipose tissue; however, excessive intake can lead to fat accumulation in visceral adipose tissue and ectopic sites, including the pancreas, heart, and muscle. This redistribution is associated with disruptions in energy metabolism, inflammation, and insulin resistance, impairing organ function and raising the risk of cardiovascular disease, diabetes, and fatty liver. This review explores the roles of visceral and ectopic fat in the development of insulin resistance and related diseases such as type 2 diabetes and metabolic dysfunction-associated steatotic liver disease. Specifically, we examine the structure and characteristics of different fat types, their associations with disease, and the underlying pathogenic mechanisms. Future strategies for managing obesity-related diseases may include lifestyle modifications, surgical interventions, and emerging medications that target lipid metabolism and energy regulation, aiming to improve patient outcomes.
Collapse
Affiliation(s)
- Jiaqiang Luo
- Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Yi Wang
- Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Jinxin Mao
- Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Ying Yuan
- Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Peng Luo
- Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Guoze Wang
- Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Shi Zhou
- Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
48
|
Sun M, Sun H. Recent prevalence and trends of obesity and metabolic dysfunction-associated steatotic liver disease (MASLD) among US adolescents: 1999 to 2020. Pediatr Obes 2025; 20:e70003. [PMID: 39967492 DOI: 10.1111/ijpo.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/15/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease (NAFLD), is understudied among US adolescents despite rising obesity rates. METHODS This study analysed the prevalence and trends of obesity and MASLD among US adolescents aged 12-17 using data from the National Health and Nutrition Examination Survey (NHANES). We developed a new screening model utilizing FibroScan-measured controlled attenuation parameter (CAP) scores, body measurements and blood chemistry data from 2017 to 2020 to assist in analysing MASLD trends from 1999 to 2020. RESULTS Between 2017 and 2020, the prevalence of obesity and MASLD was approximately 20%, with about 70% of obese adolescents affected by MASLD. The condition was more common in boys, particularly among Mexican American adolescents. Additionally, 97.2% of those with NAFLD also had MASLD. Adolescents with MASLD had significantly higher body weight, waist circumference, triglyceride levels and alanine transaminase (ALT) levels, along with lower high-density lipoprotein (HDL) cholesterol and an increased risk of liver fibrosis. Insufficient physical activity and poor diet quality were key risk factors for developing MASLD. From 1999 to 2020, the prevalence of MASLD rose significantly, paralleling the increasing rates of obesity. CONCLUSIONS The study underscores the pressing need to screen at-risk adolescents for metabolic issues associated with steatotic liver diseases, given the rising obesity rates among adolescents. The high overlap between MASLD and NAFLD diagnoses indicates that the transition from NAFLD to MASLD can be effectively integrated into paediatric practice.
Collapse
Affiliation(s)
- Michael Sun
- Department of Pediatrics, Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware, USA
| | - Hongbing Sun
- Nutrition, Biostatistics and Health Study, Department of Earth and Chemical Sciences, Rider University, Lawrenceville, New Jersey, USA
| |
Collapse
|
49
|
Zhong YL, Xu CQ, Li J, Liang ZQ, Wang MM, Ma C, Jia CL, Cao YB, Chen J. Mitochondrial dynamics and metabolism in macrophages for cardiovascular disease: A review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156620. [PMID: 40068296 DOI: 10.1016/j.phymed.2025.156620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/18/2025] [Accepted: 03/05/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Mitochondria regulate macrophage function, affecting cardiovascular diseases like atherosclerosis and heart failure. Their dynamics interact with macrophage cell death mechanisms, including apoptosis and necroptosis. PURPOSE This review explores how mitochondrial dynamics and metabolism influence macrophage inflammation and cell death in CVDs, highlighting therapeutic targets for enhancing macrophage resilience and reducing CVD pathology, while examining molecular pathways and pharmacological agents involved. STUDY DESIGN This is a narrative review that integrates findings from various studies on mitochondrial dynamics and metabolism in macrophages, their interactions with the endoplasmic reticulum (ER) and Golgi apparatus, and their implications for CVDs. The review also considers the potential therapeutic effects of pharmacological agents on these pathways. METHODS The review utilizes a comprehensive literature search to identify relevant studies on mitochondrial dynamics and metabolism in macrophages, their role in CVDs, and the effects of pharmacological agents on these pathways. The selected studies are analyzed and synthesized to provide insights into the complex relationships between mitochondria, the ER, and Golgi apparatus, and their implications for macrophage function and fate. RESULTS The review reveals that mitochondrial metabolism intertwines with cellular architecture and function, particularly through its intricate interactions with the ER and Golgi apparatus. Mitochondrial-associated membranes (MAMs) facilitate Ca2+ transfer from the ER to mitochondria, maintaining mitochondrial homeostasis during ER stress. The Golgi apparatus transports proteins crucial for inflammatory signaling, contributing to immune responses. Inflammation-induced metabolic reprogramming in macrophages, characterized by a shift from oxidative phosphorylation to glycolysis, underscores the multifaceted role of mitochondrial metabolism in regulating immune cell polarization and inflammatory outcomes. Notably, mitochondrial dysfunction, marked by heightened reactive oxygen species generation, fuels inflammatory cascades and promotes cell death, exacerbating CVD pathology. However, pharmacological agents such as Metformin, Nitazoxanide, and Galanin emerge as potential therapeutic modulators of these pathways, offering avenues for mitigating CVD progression. CONCLUSION This review highlights mitochondrial dynamics and metabolism in macrophage inflammation and cell death in CVDs, suggesting therapeutic targets to improve macrophage resilience and reduce pathology, with new pharmacological agents offering treatment opportunities.
Collapse
Affiliation(s)
- Yi-Lang Zhong
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Chen-Qin Xu
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Ji Li
- Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Zhi-Qiang Liang
- Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Miao-Miao Wang
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Chao Ma
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Cheng-Lin Jia
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Yong-Bing Cao
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Jian Chen
- Institute of Vascular Anomalies, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China; Anhui Province Rural Revitalization Collaborative Technical Service Center, Huangshan University, Huangshan 245041, China; Department of Public Health, International College, Krirk University, Bangkok, Thailand.
| |
Collapse
|
50
|
Tang H, Liao C, Wang L, Fang W, Tang N, Wan L, Ren Z. Swertianolin regulates immunosuppression of myeloid suppressor cells in septic mice by inhibiting NF-κB and P38 signaling. Transpl Immunol 2025; 90:102217. [PMID: 40107628 DOI: 10.1016/j.trim.2025.102217] [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: 03/04/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Swertianolin is one of the main components of Gentianaceae Swertia plants, a traditional Chinese medicine used for the treatment of infection, fever, viral hepatitis, and pneumonia. An expansion of myeloid-derived suppressor cells (MDSCs) contributes to sepsis induced immunosuppression. We investigated the mechanism by which Swertianolin regulates MDSCs in a mouse model of sepsis. METHODS Severe sepsis was induced in mice using caecal ligation and puncture. These mice received an intraperitoneal injection of Swertianolin. MDSCs were isolated and analyzed by flow cytometry; serum concentrations of immunosuppressive factors were detected by ELISA; and mitogen-activated protein kinase and nuclear factor-κB (NFκB) were detected by Western blots. RESULTS We found that Swertianolin reduced the number of MDSCs in the marrow and the spleen while increased the number of CD4+ T cells in the spleen of mice with sepsis in comparison to controls (p < 0.05). Swertianolin reduced lung damage and improved the survival rate in mice with secondary infection of Legionella pneumophila (p < 0.05). Swertianolin inhibited the phosphorylation of p38 and nuclear translocation of p65 in MDSCs (p < 0.05), leading to decreased production of IL-10 and nitric oxide (both p < 0.05). CONCLUSION Swertianolin may improve immunosuppressive function of MDSCs and increased T cell activity by inhibiting p38 phosphorylation and NF-κB activation.
Collapse
Affiliation(s)
- Haoran Tang
- Department of Gastroenterological Surgery, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chen Liao
- Department of Gastroenterological Surgery, the Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lingling Wang
- Department of Medical Intensive Care Unit, General Hospital of Southern Theater Command of PLA, Guangzhou, China
| | - Wei Fang
- Department of Medical Intensive Care Unit, CiHui Hospital, Guangzhou, China
| | - Ning Tang
- Department of Neurology, The First People's Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, China
| | - Linjun Wan
- Department of Critical Care Medicine, the Second Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Zongfang Ren
- Department of Critical Care Medicine, the Second Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|