51
|
Masci I, Bozal C, Lezón C, Martin M, Brites F, Bonetto J, Alvarez L, Kurtz M, Tasat D. Exposure to airborne particulate matter and undernutrition in young rats: An in-depth histopathological and biochemical study on lung and excretory organs. Food Chem Toxicol 2025; 197:115246. [PMID: 39793949 DOI: 10.1016/j.fct.2025.115246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/13/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
Environmental stressors, such as air particulate matter (PM) and nutrient deficiencies, can significantly impact crucial organs involved in detoxifying xenobiotics, including lungs, liver, and kidneys, especially in vulnerable populations like children. This study investigated the effect of 4-week exposure to Residual Oil Fly Ash (ROFA) on these organs in young rats under growth-restricted nutrition (NGR). We assessed histological, histomorphometric and biochemical parameters. ROFA exposure induced histological changes and inflammation in all three organs when compared to control (C) animals. Specifically, in lungs ROFA caused a significant reduction in alveolar airspace (C: 55.8 ± 1.8% vs. ROFA: 38.7 ± 3.0%, p < 0.01) and alveolar number along with changes in alveolar size distribution, and disruption of the smooth muscle layer which may impaired respiratory function. In the liver, ROFA increased binucleated cells, macro and microvesicles and both AST and ALT serum biomarkers (AST: C = 77.7 ± 1.3 vs. ROFA = 81.6 ± 1.3, p < 0.05; ALT: C = 44.5 ± 0.9 vs. ROFA = 49.4 ± 1.3, p < 0.05). In the kidneys, a reduced Bowman's space (C: 2.15 ± 0.2 mm2 vs. ROFA: 1.74 ± 0.2 mm2, p < 0.05) was observed, indicative of glomerular filtration failure. NGR alone reduced Bowman's space (C: 2.15 ± 0.2 mm2 vs. NGR: 1.06 ± 0.1 mm2, p < 0.001). In lung and liver NGR showed higher levels of proinflammatory cytokine IL-6 (p < 0.01 and p < 0.001, respectively) when compared to C. In conclusion, both stressors negatively affected lung and excretory organs in young rats, with nutritional status further modulating the physiological response to ROFA. These findings highlight the compounded risks posed by environmental pollutants and poor nutrition in vulnerable populations.
Collapse
Affiliation(s)
- Ivana Masci
- Laboratorio de Bio-Toxicología Ambiental, Instituto de Tecnologías Emergentes y Ciencias Aplicadas. Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín - CONICET, San Martín, Buenos Aires, Argentina
| | - Carola Bozal
- Cátedra de Histología y Embriología. Facultad de Odontología, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Christian Lezón
- Cátedra de Fisiología. Facultad de Odontología, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maximiliano Martin
- Laboratorio de Lípidos y Lipoproteínas, Departamento de Bioquímica Clínica. Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Fernando Brites
- Laboratorio de Lípidos y Lipoproteínas, Departamento de Bioquímica Clínica. Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Julián Bonetto
- Laboratorio de Bio-Toxicología Ambiental, Instituto de Tecnologías Emergentes y Ciencias Aplicadas. Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín - CONICET, San Martín, Buenos Aires, Argentina
| | - Laura Alvarez
- Laboratorio de Efectos Biológicos de Contaminantes Ambientales, Departamento de Bioquímica Humana. Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Melisa Kurtz
- Laboratorio de Bio-Toxicología Ambiental, Instituto de Tecnologías Emergentes y Ciencias Aplicadas. Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín - CONICET, San Martín, Buenos Aires, Argentina.
| | - Deborah Tasat
- Laboratorio de Bio-Toxicología Ambiental, Instituto de Tecnologías Emergentes y Ciencias Aplicadas. Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín - CONICET, San Martín, Buenos Aires, Argentina; Cátedra de Anatomía Patológica. Facultad de Odontología, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
52
|
Borgne-Sanchez A, Fromenty B. Mitochondrial dysfunction in drug-induced hepatic steatosis: Recent findings and current concept. Clin Res Hepatol Gastroenterol 2025; 49:102529. [PMID: 39798918 DOI: 10.1016/j.clinre.2025.102529] [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: 10/01/2024] [Revised: 12/19/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
Mitochondrial activity is necessary for the maintenance of many liver functions. In particular, mitochondrial fatty acid oxidation (FAO) is required for energy production and lipid homeostasis. This key metabolic pathway is finely tuned by the mitochondrial respiratory chain (MRC) activity and different transcription factors such as peroxisome proliferator-activated receptor α (PPARα). Many drugs have been shown to cause mitochondrial dysfunction, which can lead to acute and chronic liver lesions. While severe inhibition of mitochondrial FAO would eventually cause microvesicular steatosis, hypoglycemia, and liver failure, moderate impairment of this metabolic pathway can induce macrovacuolar steatosis, which can progress in the long term to steatohepatitis and cirrhosis. Drugs can impair mitochondrial FAO through several mechanisms including direct inhibition of FAO enzymes, sequestration of coenzyme A and l-carnitine, impairment of the activity of one or several MRC complexes and reduced PPARα expression. In drug-induced macrovacuolar steatosis, non-mitochondrial mechanisms can also be involved in lipid accumulation including increased de novo lipogenesis and reduced very-low-density lipoprotein secretion. Nonetheless, mitochondrial dysfunction and subsequent oxidative stress appear to be key events in the progression of steatosis to steatohepatitis. Patients suffering from metabolic dysfunction-associated steatotic liver disease (MASLD) and treated with mitochondriotoxic drugs should be closely monitored to reduce the risk of acute liver injury or a faster transition of steatosis to steatohepatitis. Therapies based on the mitochondrial cofactor l-carnitine, the antioxidant N-acetylcysteine, or thyromimetics might be useful to prevent or treat drug-induced mitochondrial dysfunction, steatosis, and steatohepatitis.
Collapse
Affiliation(s)
| | - Bernard Fromenty
- INSERM, INRAE, Univ Rennes, Institut NUMECAN, UMR_S1317, 35000 Rennes, France.
| |
Collapse
|
53
|
Sun Y, Dai X, Yang J, Chen Y, Feng J, Shi X, Li X, Liu X. Deficiency of hepatokine orosomucoid1 aggravates NAFLD progression in mice. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167654. [PMID: 39756714 DOI: 10.1016/j.bbadis.2024.167654] [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: 08/09/2024] [Revised: 12/29/2024] [Accepted: 12/29/2024] [Indexed: 01/07/2025]
Abstract
Orosomucoid (ORM) is an important hepatokine that regulates metabolism. Previous report showed that isoform ORM2 but not ORM1 could downregulate lipogenic genes and ameliorate hepatic steatosis in obese mice, thereby categorizing ORM2 as a promising candidate for therapeutic intervention in nonalcoholic fatty liver disease (NAFLD). However, our previous studies found that mice lacking ORM1 gradually developed an obese phenotype with severe hepatic steatosis at the age of 24 weeks. Consequently, it remains imperative to further investigate the precise role of ORM1 in the context of NAFLD. The current study aims to assess the function and therapeutic prospects of ORM1 in NAFLD models induced by a high-fat diet (HFD) or a methionine- and choline-deficient diet (MCD), employing a series of loss- and gain-of-function experiments. The results showed that liver ORM levels elevated in fat NAFLD models but decreased in lean NAFLD models. Orm1-deficient mice fed either on HFD or MCD had significantly higher NAFLD activity score with more severe steatosis and ballooning, showing an aggravated NAFLD progression. However, liver-specific Orm1 overexpression in mice could not alleviate NAFLD when fed on HFD or MCD. These results suggest that systemic endogenous ORM1 is indispensable in protecting against the development of NAFLD; however, it may not serve as an effective localized therapeutic target for managing the disease.
Collapse
Affiliation(s)
- Yang Sun
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433, China
| | - XianMin Dai
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433, China
| | - JinRun Yang
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433, China
| | - Yi Chen
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433, China
| | - JiaYi Feng
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433, China
| | - XiaoFei Shi
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433, China
| | - Xiang Li
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433, China.
| | - Xia Liu
- Department of Clinical Pharmacy, School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
54
|
Jefcoate CR, Larsen MC, Song YS, Maguire M, Sheibani N. Defined Diets Link Iron and α-Linolenic Acid to Cyp1b1 Regulation of Neonatal Liver Development Through Srebp Forms and LncRNA H19. Int J Mol Sci 2025; 26:2011. [PMID: 40076634 PMCID: PMC11901102 DOI: 10.3390/ijms26052011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 03/14/2025] Open
Abstract
Cyp1b1 substantially affects hepatic vascular and stellate cells (HSC) with linkage to liver fibrosis. Despite minimal hepatocyte expression, Cyp1b1 deletion substantially impacts liver gene expression at birth and weaning. The appreciable Cyp1b1 expression in surrounding embryo mesenchyme, during early organogenesis, provides a likely source for Cyp1b1. Here defined breeder diets established major interconnected effects on neonatal liver of α-linolenic acid (ALA), vitamin A deficiency (VAD) and suboptimal iron fed mice. At birth Cyp1b1 deletion and VAD each activated perinatal HSC, while suppressing iron control by hepcidin. Cyp1b1 deletion also advanced the expression of diverse genes linked to iron regulation. Postnatal stimulations of Srebp-regulated genes in the fatty acid and cholesterol biosynthesis pathways were suppressed by Cyp1b1-deficiency. LncRNA H19 and the neutrophil alarmin S100a9 expression increased due to slower postnatal decline with Cyp1b1 deficiency. VAD reversed each of Cyp1b1 effect, probably due to enhanced HSC release of Apo-Rbp4. At birth, Cyp1b1 deletion enhanced H19 participation. Notably, a suppressor (Cnot3) decreased while an activity partner (Ezh2/H3K methylation) increased H19 expression. ALA elevated hepcidin mRNA and countered the inhibitory effects of Cyp1b1 deletion on hepcidin expression. Oxylipin metabolites of ALA from highly expressed hepatic Cyps are potential mediators. Cyp expression patterns demonstrated female dimorphism for neonatal liver. Mothers followed one of three fetal growth support programs probably linked to maturity at conception.
Collapse
Affiliation(s)
- Colin R. Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (M.C.L.); (M.M.)
| | - Michele C. Larsen
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (M.C.L.); (M.M.)
| | - Yong-Seok Song
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
| | - Meghan Maguire
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (M.C.L.); (M.M.)
| | - Nader Sheibani
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (M.C.L.); (M.M.)
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
| |
Collapse
|
55
|
Ding Y, Ye J, Liu Y, Zhang S, Xu Y, Yang Z, Liu Z. Fucoxanthin Ameliorates Kidney Injury by CCl 4-Induced via Inhibiting Oxidative Stress, Suppressing Ferroptosis, and Modulating Gut Microbiota. ACS OMEGA 2025; 10:7407-7421. [PMID: 40028144 PMCID: PMC11865997 DOI: 10.1021/acsomega.4c11437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 03/05/2025]
Abstract
Chemical-induced kidney injury represents a substantial health risk, with ferroptosis, a type of cell death caused by lipid peroxidation, playing a role in numerous kidney ailments. Fucoxanthin (Fx), a natural carotenoid known for its antioxidant capabilities, has shown promise in alleviating renal injury, but its exact mechanisms are yet to be fully understood. Carbon tetrachloride (CCl4) is recognized as a powerful nephrotoxic substance, and this study explores the therapeutic effects of Fx on oxidative stress, ferroptosis and intestinal microbiota in mouse kidneys subjected to CCl4 exposure. The mice were randomly assigned to control, model, colchicine groups (0.1 mg/kg/d), and Fx (50, 100 mg/kg/d) group and underwent related treatments for 4 weeks. Then, we evaluated their renal function, histological alterations in the kidneys, colon, and jejunum, and the levels of related proteins (i.e., Nrf2, GPX4, SLC7A11, HO-1, TFR1, NQO1, GCLM, FTL). Additionally, their gut microbiota was analyzed using 16S rRNA gene sequencing. The results showed that compared to the CCl4 group, Fx treatment led to lower serum creatinine and blood urea nitrogen levels, reduced malondialdehyde activity in kidneys and intestinal tissues, and increased activity of antioxidant enzymes. Fx also reduced dysbiosis and enhanced the diversity of intestinal flora. In summary, Fx reduced oxidative stress and ferroptosis and partially restored intestinal bacteria, thus improving CCl4-induced renal damage in mice. These results suggest Fx as a potential therapeutic option for kidney injuries related to oxidative stress. Further research is needed to clarify its precise mechanisms and potential clinical implications.
Collapse
Affiliation(s)
- Yaping Ding
- Zhoushan
Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese
Medical University, Zhoushan 316000, Zhejiang Province, P.R. China
| | - Jiena Ye
- School
of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China
| | - Ying Liu
- Zhoushan
Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese
Medical University, Zhoushan 316000, Zhejiang Province, P.R. China
| | - Shaohua Zhang
- Zhoushan
Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese
Medical University, Zhoushan 316000, Zhejiang Province, P.R. China
| | - Yan Xu
- Zhoushan
Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese
Medical University, Zhoushan 316000, Zhejiang Province, P.R. China
| | - Zuisu Yang
- School
of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China
| | - Zhongliang Liu
- Zhoushan
Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese
Medical University, Zhoushan 316000, Zhejiang Province, P.R. China
| |
Collapse
|
56
|
Hui L, Chen X, Huang M, Jiang Y, Liu T. TANK-Binding Kinase 1 in the Pathogenesis and Treatment of Inflammation-Related Diseases. Int J Mol Sci 2025; 26:1941. [PMID: 40076567 PMCID: PMC11900955 DOI: 10.3390/ijms26051941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
TANK-binding kinase 1 (TBK1) is a key signaling kinase involved in innate immune and inflammatory responses. TBK1 drives immune cells to participate in the inflammatory response by activating the NF-κB and interferon regulatory factor signaling pathways in immune cells, promoting the expression of pro-inflammatory genes, and regulating immune cell function. Thus, it plays a crucial role in initiating a signaling cascade that establishes an inflammatory environment. In inflammation-related diseases, TBK1 acts as a bridge linking inflammation to immunity, metabolism, or tumorigenesis, playing an important role in the pathogenesis of immune-mediated inflammatory diseases, metabolic, inflammatory syndromes, and inflammation-associated cancers by regulating the activation of inflammatory pathways and the production of inflammatory cytokines in cells. In this review, we focused on the mechanisms of TBK1 in immune cells and inflammatory-related diseases, providing new insights for further studies targeting TBK1 as a potential treatment for inflammation-related diseases. Thus, optimizing and investigating highly selective cell-specific TBK1 inhibitors is important for their application in these diseases.
Collapse
Affiliation(s)
- Lu Hui
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu 610041, China; (L.H.); (X.C.); (M.H.)
| | - Xiaolin Chen
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu 610041, China; (L.H.); (X.C.); (M.H.)
| | - Mengke Huang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu 610041, China; (L.H.); (X.C.); (M.H.)
| | - Yongmei Jiang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu 610041, China; (L.H.); (X.C.); (M.H.)
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Ting Liu
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu 610041, China; (L.H.); (X.C.); (M.H.)
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Biotherapy and Cancer Center/National Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
57
|
Chen CC, Hsu LW, Chen KD, Chiu KW, Kung CP, Li SR, Goto S, Chen CL, Huang KT. Extracellular calreticulin regulates fibrogenic and immunogenic properties of hepatic stellate cells. Int Immunopharmacol 2025; 148:114129. [PMID: 39862632 DOI: 10.1016/j.intimp.2025.114129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
Liver fibrosis is a persistent damage repair response triggered by various etiological factors, resulting in an excessive accumulation of extracellular matrix (ECM). Activated hepatic stellate cells (HpSCs) are the primary source of ECM proteins. Therefore, specifically targeting HpSCs has become a crucial approach for treating liver fibrosis. Calreticulin (CRT) is a molecular chaperone mainly located in the endoplasmic reticulum (ER), regulating protein folding and calcium homeostasis. Recently, CRT has gained much attention for its role outside the ER, particularly at the cell surface and extracellular space, acting as an immunomodulatory protein. The current study investigates the role of extracellular CRT in hepatic injury and its effects on HpSCs. Elevated levels of circulating CRT were observed in mouse models of liver injury, suggesting that hepatic injury may trigger CRT release. Extracellular CRT was found to moderately inhibit HpSC viability and induce morphological changes. Additionally, CRT treatment led to a decrease in α-smooth muscle actin and an upregulation of matrix metalloproteinase-2 and -9, indicating a potential fibrolytic effect. Immunomodulatory activities of CRT were also noted, as it increased cytokine expression in both macrophages and HpSCs. These effects were partially mediated through low-density lipoprotein receptor-related protein 1 (LRP1), as evidenced by altered cytokine expression upon co-treatment with a known LRP1 ligand receptor-associated protein (RAP). Overall, this study elucidates the complex role of extracellular CRT in liver injury and its potential impact on HpSC behavior and immune responses.
Collapse
Affiliation(s)
- Chien-Chih Chen
- Department of Psychiatry Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan; School of Medicine Chang Gung University Taoyuan Taiwan
| | - Li-Wen Hsu
- Liver Transplantation Center Department of General Surgery Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan
| | - Kuang-Den Chen
- Liver Transplantation Center Department of General Surgery Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan; Institute for Translational Research in Biomedicine Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan
| | - King-Wah Chiu
- Liver Transplantation Center Department of General Surgery Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan; Division of Hepato-Gastroenterology Department of Internal Medicine Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan
| | - Chao-Pin Kung
- Liver Transplantation Center Department of General Surgery Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan; Institute for Translational Research in Biomedicine Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan
| | - Shu-Rong Li
- Liver Transplantation Center Department of General Surgery Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan
| | - Shigeru Goto
- Liver Transplantation Center Department of General Surgery Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan; Nobeoka Medical Check Center Fukuoka Institute of Occupational Health Nobeoka Japan
| | - Chao-Long Chen
- Liver Transplantation Center Department of General Surgery Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan
| | - Kuang-Tzu Huang
- Liver Transplantation Center Department of General Surgery Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan; Institute for Translational Research in Biomedicine Kaohsiung Chang Gung Memorial Hospital Kaohsiung Taiwan.
| |
Collapse
|
58
|
Fu H, Wang X, Yuan M, Wang N, Zhang X. Callistephus A from Callistephus chinensis Nees alleviates concanavalin A-induced immunological liver injury in mice by inhibiting the activation of JAK/STAT1 and MAPK signaling pathways. Int Immunopharmacol 2025; 148:114153. [PMID: 39864226 DOI: 10.1016/j.intimp.2025.114153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 11/07/2024] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
Callistephus chinensis Nees is an herbaceous plant in the Asteraceae family that has various traditional effects, especially in preventing liver disease. Callistephus A (CA) is a sesquiterpene compound with a rare 6/7 ring skeleton, which has been isolated only from the Callistephus chinensis Nees, but whether CA protects the liver is unknown. Immunological liver injury (ILI) is a common liver disease mediated by the immune system. Therefore, this study investigated whether CA had a protective effect on ILI and uncovered its molecular mechanisms. To study the impact, target, and signal pathway of CA in preventing ILI, we hope to find active components from plants to avoid ILI. In this study, CA regulated the differentiation balance of CD4 + T cells (Th1/Th2 and Th17/Treg balance) and the secretion of inflammatory factors (tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), interferon-gamma (IFN-γ), interleukin-4 (IL-4), interleukin-17A (IL-17A) and transforming growth factor-β (TGF-β). CA improves liver inflammation by regulating IFN-γ-induced JAK/STAT1 signaling pathways. CA reduced hepatocyte apoptosis by decreasing protein expression of BCL2-associated X (Bax), cleaved caspase-3, and cleaved Poly (ADP-ribose) polymerase 1 (PARP-1), but increased Bcl-2 protein expression, which was achieved by regulating the MAPK pathway. To investigate the role of CA in immune liver injury, we performed in vitro cell experiments using alpha mouse liver 12 (AML12) cells. The cell experiments showed that CA potently inhibited LPS-mediated AML12 cell damage. After adding CA, damaged mitochondria are cleared through mitochondrial autophagy and reduced production of intracellular reactive oxygen species (ROS). Finally, molecular docking results showed that CA had a strong affinity for five essential target proteins (JAK1, JAK2, STAT1, JNK, and p38). CA regulates the differentiation, anti-inflammatory, and anti-apoptosis of CD4 + T cells. The mechanism of CA against ILI is related to inhibiting the activation of JAK/STAT1 and MAPK signaling pathways.
Collapse
Affiliation(s)
- Haonan Fu
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Xiaojun Wang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Mingyuan Yuan
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Ning Wang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Xiaoshu Zhang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| |
Collapse
|
59
|
Yuan Y, Zhang J, Li H, Yuan F, Cui Q, Wu D, Yuan H, Piao G. Scopoletin alleviates acetaminophen-induced hepatotoxicity through modulation of NLRP3 inflammasome activation and Nrf2/HMGB1/TLR4/NF-κB signaling pathway. Int Immunopharmacol 2025; 148:114132. [PMID: 39870009 DOI: 10.1016/j.intimp.2025.114132] [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: 10/21/2024] [Revised: 01/14/2025] [Accepted: 01/19/2025] [Indexed: 01/29/2025]
Abstract
Scopoleitin (SP), a bioactive compound from many edible plants and fruits, exerts a wide range of biological activities, however the role and mechanism of SP in acetaminophen (APAP)-induced hepatotoxicity remains unclear. In this study, we verified the protective effect of SP on APAP-induced liver injury (AILI) hepatotoxicity and explore the underlying molecular mechanisms. Here, we showed that SP alleviated AILI by reducing serum alanine transaminase (ALT) and aspartate aminotransferase (AST) levels, hepatic histopathological damage, inflammation, and liver cell apoptosis. In addition, SP attenuated the accumulation of malondialdehyde (MDA) and exhaustion of glutathione (GSH) levels and increased the superoxide dismutase (SOD) levels induced by APAP. Consistently, SP significantly reduced the gene transcription of cytochrome P450 (CYP)2E1, CYP1A2, and CYP3A11 in the livers of mice induced by APAP. Moreover, SP pretreatment effectively promoted the expression of Nrf2, Keap1, and its signal downstream HO-1, NQO1, GCLc, and GCLm, suggesting the activation of the Nrf2 signaling pathway. SP inhibited APAP-induced hepatocyte apoptosis by regulating the protein levels of apoptosis-related proteins (cytochrome C, Bax, Caspase-3, Bcl2, and PARP). SP suppressed APAP-induced expression of NLRP3 and reduced the levels of proinflammatory factors, including tumor necrosis factor-alpha (TNF-α), F4/80, Caspase-1, and interleukin (IL)-1 beta (IL-1β). Moreover, SP downregulated APAP-induced high-mobility group box 1 (HMGB1) and toll-like receptor 4 (TLR4) expression, inhibited nuclear factor kappa-B (NF-κB) and MAPK activation. Taken together, our study reveals the protective roles of SP against AILI through the downregulation of NLRP3 expression, and the inhibition of the Nrf2/HMGB1/TLR4/NF-κB signaling pathways.
Collapse
Affiliation(s)
- Yilin Yuan
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin 133002, China
| | - Jianxiu Zhang
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin 133002, China
| | - Hui Li
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin 133002, China
| | - Fengxia Yuan
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin 133002, China
| | - Qinglong Cui
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin 133002, China
| | - Di Wu
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin 133002, China
| | - Haidan Yuan
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin 133002, China.
| | - Guangchun Piao
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin 133002, China.
| |
Collapse
|
60
|
Kim DH, Go HS, Jeon EJ, Nguyen TQT, Kim DY, Park H, Eom HJ, Kim SY, Park SC, Cho KA. The Impact of Toll-Like Receptor 5 on Liver Function in Age-Related Metabolic Disorders. Aging Cell 2025:e70009. [PMID: 39957532 DOI: 10.1111/acel.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/09/2024] [Accepted: 01/10/2025] [Indexed: 02/18/2025] Open
Abstract
Toll-like receptor 5 (TLR5) plays a critical role beyond its traditional function in innate immunity, significantly impacting metabolic regulation and liver health. Previously, we reported that TLR5 activation extends the healthspan and lifespan of aging mice. This study demonstrates that TLR5 deficiency leads to pronounced metabolic abnormalities with age, primarily affecting liver metabolic functions rather than intestinal inflammation. Comprehensive RNA sequencing analysis revealed that TLR5 deficiency induces gene expression changes in liver tissue similar to those caused by the methionine-choline deficient (MCD) diet, particularly affecting lipid metabolism and circadian rhythm-related genes. TLR5 knockout (TLR5 KO) mice displayed an increased propensity for liver fibrosis and lipid accumulation under the MCD diet, exacerbating liver pathology. Both hepatocytes and hepatic stellate cells in TLR5 KO mice were functionally impacted, leading to metabolic dysfunction and fibrosis. These findings suggest that TLR5 could be a significant target for addressing metabolic diseases that arise and worsen with aging. Furthermore, understanding the mechanisms by which TLR5 activation extends healthspan could provide valuable insights into therapeutic strategies for enhancing longevity and managing age-related metabolic disorders.
Collapse
Affiliation(s)
- Dong-Hyun Kim
- MediSpan, Inc, Bundang-gu, Gyeonggi-do, Republic of Korea
| | - Hye Sun Go
- MediSpan, Inc, Bundang-gu, Gyeonggi-do, Republic of Korea
| | - Eun Jae Jeon
- MediSpan, Inc, Bundang-gu, Gyeonggi-do, Republic of Korea
| | - Thi Quynh Trang Nguyen
- Department of Biochemistry, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, Republic of Korea
- Center for Creative Biomedical Scientists, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, Republic of Korea
| | - Da Yeon Kim
- MediSpan, Inc, Bundang-gu, Gyeonggi-do, Republic of Korea
| | - Hansung Park
- Department of Biochemistry, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, Republic of Korea
| | - Hyo-Ji Eom
- MediSpan, Inc, Bundang-gu, Gyeonggi-do, Republic of Korea
| | - Sung Young Kim
- Department of Biochemistry, Konkuk University School of Medicine, Seoul, South Korea
| | - Sang Chul Park
- Future Life and Society Research Center, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, Republic of Korea
| | - Kyung A Cho
- MediSpan, Inc, Bundang-gu, Gyeonggi-do, Republic of Korea
- Department of Biochemistry, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, Republic of Korea
- Center for Creative Biomedical Scientists, Chonnam National University Medical School, Hwasun-gun, Jeonnam-do, Republic of Korea
| |
Collapse
|
61
|
Huang T, Zhou MY, Zou GL, Hu RH, Han L, Zhang QX, Zhao XK. Focal adhesion kinase promotes aerobic glycolysis in hepatic stellate cells via the cyclin D1/c-Myc/MCT-1 pathway to induce liver fibrosis. Sci Rep 2025; 15:4552. [PMID: 39915293 PMCID: PMC11802747 DOI: 10.1038/s41598-025-88538-8] [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: 09/11/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
Hepatic stellate cells (HSCs) transdifferentiate into myofibroblasts during liver fibrosis and exhibit increased glycolysis. Phosphorylated focal adhesion kinase (FAK) (pY397-FAK) promotes monocarboxylate transporter 1 (MCT-1) expression in HSCs to increase aerobic glycolysis and cause liver fibrosis. A combined multiomics analysis of C57BL/6 mice with tetrachloromethane (CCl4)-induced liver fibrosis was performed to identify the downstream FAK signaling pathway. The effect of the FAK inhibitor PF562271 on CCl4-induced liver fibrosis was explored by immunofluorescence of liver tissues. The migration, proliferation and aerobic glycolysis of LX-2 cells after stimulation and activation by transforming growth factor beta-1 (TGF-β1) or suppression by PF562271 was assessed in vitro. Multiomics analysis of a successfully generated CCl4-induced liver fibrosis mouse model was performed. FAK and cyclin D1 were significantly enriched in mice with CCl4-induced liver fibrosis. In vivo, the MCT-1 and alpha smooth muscle actin (α-SMA) levels were increased in mice with CCl4-induced liver fibrosis, and MCT-1 and α-SMA expression decreased after PF562271 treatment. In vitro, PF562271 alleviated TGF-β1-induced LX-2 activation. LX-2 cells showed diminished migration, proliferation, and aerobic glycolysis after PF562271 intervention. FAK promotes aerobic glycolysis in LX-2 cells through the cyclin D1/c-Myc/MCT-1 pathway, thereby increasing liver fibrosis.
Collapse
Affiliation(s)
- Tao Huang
- Department of Infectious Disease, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China
| | - Ming-Yu Zhou
- Department of Infectious Disease, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China
| | - Gao-Liang Zou
- Department of Infectious Disease, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China
| | - Rui-Han Hu
- Department of Cardiology, Guiqian International General Hospital, Guiyang, Guizhou Province, China
| | - Lu Han
- Department of Comprehensive Ward, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China
| | - Qing-Xiu Zhang
- Department of Infectious Disease, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China
| | - Xue-Ke Zhao
- Department of Infectious Disease, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China.
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, No. 9 Beijing Road, Guiyang, 550004, Guizhou Province, China.
| |
Collapse
|
62
|
Zhao G, Li X, Zhang Y, Wang X, Deng L, Xu J, Jin S, Zuo Z, Xun L, Luo M, Yang F, Qi J, Fu P. Intricating connections: the role of ferroptosis in systemic lupus erythematosus. Front Immunol 2025; 16:1534926. [PMID: 39967676 PMCID: PMC11832682 DOI: 10.3389/fimmu.2025.1534926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 01/17/2025] [Indexed: 02/20/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic inflammatory and autoimmune disease with multiple tissue damage. However, the pathology remains elusive, and effective treatments are lacking. Multiple types of programmed cell death (PCD) implicated in SLE progression have recently been identified. Although ferroptosis, an iron-dependent form of cell death, has numerous pathophysiological features similar to those of SLE, such as intracellular iron accumulation, mitochondrial dysfunction, lipid metabolism disorders and concentration of damage associated-molecular patterns (DAMPs), only a few reports have demonstrated that ferroptosis is involved in SLE progression and that the role of ferroptosis in SLE pathogenesis continues to be neglected. Therefore, this review elucidates the potential intricate relationship between SLE and ferroptosis to provide a reliable theoretical basis for further research on ferroptosis in the pathogenesis of SLE.
Collapse
Affiliation(s)
- Guowang Zhao
- Department of Rheumatology and Clinical Immunology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xinghai Li
- Department of Minimal Invasive Intervention Radiology, Ganzhou People’s Hospital, Ganzhou, Jiangxi, China
| | - Ying Zhang
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, The First People’s Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xingzi Wang
- Department of Nephrology, Yueyang Central Hospital, Yueyang, Hunan, China
| | - Li Deng
- Department of Internal Medicine, Community Health Service Station of Dian Mian Avenue, Kunming, Yunnan, China
| | - Juan Xu
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Shumei Jin
- Yunnan Institute of Food and Drug Supervision and Control, Medical Products Administration of Yunnan Province, Kunming, Yunnan, China
| | - Zan Zuo
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, The First People’s Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Linting Xun
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, The First People’s Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Mei Luo
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, The First People’s Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Fan Yang
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jialong Qi
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, The First People’s Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Clinical Virology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Birth Defects and Genetic Diseases, First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Ping Fu
- Department of Rheumatology and Clinical Immunology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
63
|
Xu J, Li Y, Feng Z, Chen H. Cigarette Smoke Contributes to the Progression of MASLD: From the Molecular Mechanisms to Therapy. Cells 2025; 14:221. [PMID: 39937012 PMCID: PMC11816580 DOI: 10.3390/cells14030221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/22/2025] [Accepted: 01/31/2025] [Indexed: 02/13/2025] Open
Abstract
Cigarette smoke (CS), an intricate blend comprising over 4000 compounds, induces abnormal cellular reactions that harm multiple tissues. Non-alcoholic fatty liver disease (NAFLD) is a prevalent chronic liver disease (CLD), encompassing non-alcoholic fatty liver (NAFL), non-alcoholic steatohepatitis (NASH), cirrhosis, and hepatocellular carcinoma (HCC). Recently, the term NAFLD has been changed to metabolic dysfunction-associated steatotic liver disease (MASLD), and NASH has been renamed metabolic dysfunction-associated steatohepatitis (MASH). A multitude of experiments have confirmed the association between CS and the incidence and progression of MASLD. However, the specific signaling pathways involved need to be updated with new scientific discoveries. CS exposure can disrupt lipid metabolism, induce inflammation and apoptosis, and stimulate liver fibrosis through multiple signaling pathways that promote the progression of MASLD. Currently, there is no officially approved efficacious pharmaceutical intervention in clinical practice. Therefore, lifestyle modifications have emerged as the primary therapeutic approach for managing MASLD. Smoking cessation and the application of a series of natural ingredients have been shown to ameliorate pathological changes in the liver induced by CS, potentially serving as an effective approach to decelerating MASLD development. This article aims to elucidate the specific signaling pathways through which smoking promotes MASLD, while summarizing the reversal factors identified in recent studies, thereby offering novel insights for future research on and the treatment of MASLD.
Collapse
Affiliation(s)
- Jiatong Xu
- Queen Mary School, Medical College, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (Z.F.)
| | - Yifan Li
- Queen Mary School, Medical College, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (Z.F.)
| | - Zixuan Feng
- Queen Mary School, Medical College, Nanchang University, Nanchang 330006, China; (J.X.); (Y.L.); (Z.F.)
| | - Hongping Chen
- Department of Histology and Embryology, Jiangxi Medical College, Nanchang University, Nanchang 330019, China
| |
Collapse
|
64
|
Wu X, Tian Y, Wang H, Chen H, Hou H, Hu Q. Dual Regulation of Nicotine on NLRP3 Inflammasome in Macrophages with the Involvement of Lysosomal Destabilization, ROS and α7nAChR. Inflammation 2025; 48:61-74. [PMID: 38717634 DOI: 10.1007/s10753-024-02036-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/23/2024] [Accepted: 04/23/2024] [Indexed: 02/09/2025]
Abstract
Nicotine, the primary alkaloid in tobacco products, has been shown to have immunoregulatory function in at least 20 diseases. The biological mechanism of action of nicotine immunoregulation is complex, resulting in an improvement of some disease states and exacerbation of others. Given the central role of the NLRP3 inflammasome in macrophages among multiple inflammatory diseases, this study examined how nicotine alters NLRP3 inflammasome activation in macrophages. NLRP3 inflammasome activation was examined mechanistically in the context of different nicotine dosages. We show NLRP3 inflammasome activation, apoptosis-associated speck-like protein (ASC) expression, caspase-1 activity and subsequent IL-1β secretion were positively correlated with nicotine in a dose-dependent relationship, and destabilization of lysosomes and ROS production were also involved. At high concentrations of nicotine surpassing 0.25 mM, NLRP3 inflammasome activity declined, along with increased expression of the anti-inflammatory Alpha7 nicotinic acetylcholine receptor (α7nAChR) and the inhibition of TLR4/NF-κB signaling. Consequently, high doses of nicotine also reduced ASC expression, caspase-1 activity and IL-1β secretion in macrophages. Collectively, these results suggest a dual regulatory function of nicotine on NLRP3 inflammasome activation in macrophages, that is involved with the pro-inflammatory effects of lysosomal destabilization and ROS production. We also show nicotine mediates anti-inflammatory effects by activating α7nAChR at high doses.
Collapse
Affiliation(s)
- Xiaqing Wu
- China National Tobacco Quality Supervision & Test Center, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China
- Key Laboratory of Tobacco Biological Effects, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China
- Beijing Life Science Academy, Lutuan East Road, Beijing, 102200, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Lutuan East Road, Beijing, 102200, China
| | - Yushan Tian
- China National Tobacco Quality Supervision & Test Center, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China
- Key Laboratory of Tobacco Biological Effects, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China
- Beijing Life Science Academy, Lutuan East Road, Beijing, 102200, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Lutuan East Road, Beijing, 102200, China
| | - Hongjuan Wang
- China National Tobacco Quality Supervision & Test Center, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China
- Key Laboratory of Tobacco Biological Effects, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China
- Beijing Life Science Academy, Lutuan East Road, Beijing, 102200, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Lutuan East Road, Beijing, 102200, China
| | - Huan Chen
- China National Tobacco Quality Supervision & Test Center, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China
- Key Laboratory of Tobacco Biological Effects, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China
- Beijing Life Science Academy, Lutuan East Road, Beijing, 102200, China
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Lutuan East Road, Beijing, 102200, China
| | - Hongwei Hou
- China National Tobacco Quality Supervision & Test Center, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China.
- Key Laboratory of Tobacco Biological Effects, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China.
- Beijing Life Science Academy, Lutuan East Road, Beijing, 102200, China.
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Lutuan East Road, Beijing, 102200, China.
| | - Qingyuan Hu
- China National Tobacco Quality Supervision & Test Center, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China.
- Key Laboratory of Tobacco Biological Effects, No. 2 Fengyang Street, Zhengzhou, 450001, Henan, China.
- Beijing Life Science Academy, Lutuan East Road, Beijing, 102200, China.
- Key Laboratory of Tobacco Biological Effects and Biosynthesis, Lutuan East Road, Beijing, 102200, China.
| |
Collapse
|
65
|
Ulrich H, Glaser T, Thomas AP. Purinergic signaling in liver disease: calcium signaling and induction of inflammation. Purinergic Signal 2025; 21:69-81. [PMID: 39320433 PMCID: PMC11958897 DOI: 10.1007/s11302-024-10044-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/15/2024] [Indexed: 09/26/2024] Open
Abstract
Purinergic signaling regulates many metabolic functions and is implicated in liver physiology and pathophysiology. Liver functionality is modulated by ionotropic P2X and metabotropic P2Y receptors, specifically P2Y1, P2Y2, and P2Y6 subtypes, which physiologically exert their influence through calcium signaling, a key second messenger controlling glucose and fat metabolism in hepatocytes. Purinergic receptors, acting through calcium signaling, play an important role in a range of liver diseases. Ionotropic P2X receptors, such as the P2X7 subtype, and certain metabotropic P2Y receptors can induce aberrant intracellular calcium transients that impact normal hepatocyte function and initiate the activation of other liver cell types, including Kupffer and stellate cells. These P2Y- and P2X-dependent intracellular calcium increases are particularly relevant in hepatic disease states, where stellate and Kupffer cells respond with innate immune reactions to challenges, such as excess fat accumulation, chronic alcohol abuse, or infections, and can eventually lead to liver fibrosis. This review explores the consequences of excessive extracellular ATP accumulation, triggering calcium influx through P2X4 and P2X7 receptors, inflammasome activation, and programmed cell death. In addition, P2Y2 receptors contribute to hepatic steatosis and insulin resistance, while inhibiting the expression of P2Y6 receptors can alleviate alcoholic liver steatosis. Adenosine receptors may also contribute to fibrosis through extracellular matrix production by fibroblasts. Thus, pharmacological modulation of P1 and P2 receptors and downstream calcium signaling may open novel therapeutic avenues.
Collapse
Affiliation(s)
- Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, 05508-000, Brazil.
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA.
| | - Talita Glaser
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, 05508-000, Brazil.
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA.
| | - Andrew P Thomas
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| |
Collapse
|
66
|
Matboli M, Hamady S, Saad M, Khaled R, Khaled A, Barakat EMF, Sayed SA, Agwa S, Youssef I. Innovative approaches to metabolic dysfunction-associated steatohepatitis diagnosis and stratification. Noncoding RNA Res 2025; 10:206-222. [PMID: 40248839 PMCID: PMC12004009 DOI: 10.1016/j.ncrna.2024.10.002] [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: 04/20/2024] [Revised: 08/08/2024] [Accepted: 10/10/2024] [Indexed: 01/03/2025] Open
Abstract
The global rise in Metabolic dysfunction-associated steatotic liver disease (MASLD)/Metabolic dysfunction-associated steatohepatitis (MASH) highlights the urgent necessity for noninvasive biomarkers to detect these conditions early. To address this, we endeavored to construct a diagnostic model for MASLD/MASH using a combination of bioinformatics, molecular/biochemical data, and machine learning techniques. Initially, bioinformatics analysis was employed to identify RNA molecules associated with MASLD/MASH pathogenesis and enriched in ferroptosis and exophagy. This analysis unveiled specific networks related to ferroptosis (GPX4, LPCAT3, ACSL4, miR-4266, and LINC00442) and exophagy (TSG101, HGS, SNF8, miR-4498, miR-5189-5p, and CTBP1-AS2). Subsequently, serum samples from 400 participants (151 healthy, 150 MASH, and 99 MASLD) underwent biochemical and molecular analysis, revealing significant dyslipidemia, impaired liver function, and disrupted glycemic indicators in MASLD/MASH patients compared to healthy controls. Molecular analysis indicated increased expression of LPCAT3, ACSL4, TSG101, HGS, and SNF8, alongside decreased GPX4 levels in MASH and MASLD patients compared to controls. The expression of epigenetic regulators from both networks (miR-4498, miR-5189-5p, miR-4266, LINC00442, and CTBP1-AS2) significantly differed among the studied groups. Finally, supervised machine learning models, including Neural Networks and Random Forest, were applied to molecular signatures and clinical/biochemical data. The Random Forest model exhibited superior performance, and molecular features effectively distinguished between the three studied groups. Clinical features, particularly BMI, consistently served as discriminatory factors, while biochemical features exhibited varying discriminant behavior across MASH, MASLD, and control groups. Our study underscores the significant potential of integrating diverse data types to enable early detection of MASLD/MASH, offering a promising approach for non-invasive diagnostic strategies.
Collapse
Affiliation(s)
- Marwa Matboli
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo, 11566, Egypt
- Faculty of Oral & Dental Medicine, Misr International University, Qalyubiyya Governorate, Egypt
| | - Shaimaa Hamady
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Maha Saad
- Basic Sciences Department, Faculty of Medicine, Modern University for Technology and Information, Cairo, Egypt
| | - Radwa Khaled
- Basic Sciences Department, Faculty of Medicine, Modern University for Technology and Information, Cairo, Egypt
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University & Faculty of Medicine, Modern University for Technology and Information, Cairo, Egypt
| | - Abdelrahman Khaled
- Bioinformatics Group, Center of Informatics Sciences (CIS), School of Information Technology and Computer Sciences, Nile University, Giza, Egypt
| | - Eman MF. Barakat
- Tropical Medicine Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sayed Ahmed Sayed
- Tropical Medicine Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - SaraH.A. Agwa
- Clinical Pathology and Molecular Genomics Unit, Medical Ain Shams Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo, 11382, Egypt
| | - Ibrahim Youssef
- Systems and Biomedical Engineering Department, Faculty of Engineering, Cairo University, Egypt
| |
Collapse
|
67
|
Fang Z, Liu C, Cheng Y, Ji Y, Liu C. Combined analysis of bulk, single-cell RNA sequencing, and spatial transcriptomics reveals the expression patterns of lipid metabolism and ferroptosis in the immune microenvironment of metabolic-associated fatty liver disease. Life Sci 2025; 362:123377. [PMID: 39793853 DOI: 10.1016/j.lfs.2025.123377] [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: 09/19/2024] [Revised: 12/20/2024] [Accepted: 01/04/2025] [Indexed: 01/13/2025]
Abstract
AIMS This study aims to identify key biomarkers associated with ferroptosis and lipid metabolism and investigate their roles in the progression of metabolic dysfunction-associated fatty liver disease (MAFLD). It further explores interactions between these biomarkers and the immune-infiltration environment, shedding light on how ferroptosis and lipid metabolism influence immune dynamics in MAFLD. MAIN METHODS Single-cell RNA sequencing data from liver samples were analyzed to evaluate expression variations related to ferroptosis and lipid metabolism in MAFLD patients. Gene scores were assessed to explore their impact on the immune microenvironment, particularly hepatocyte-macrophage communication. Weighted Gene Co-expression Network Analysis (WGCNA) was applied to Bulk-RNA-Seq data to identify gene clusters associated with ferroptosis and lipid metabolism. The analyses were integrated into a risk assessment system and predictive model, with validation conducted through in vivo experiments. KEY FINDINGS Integration of single-cell and WGCNA data identified 11 key genes linked to ferroptosis and lipid metabolism (e.g., IER5L, SOCS2, KLF9), significantly influencing the liver's immune microenvironment. The risk assessment system and predictive model achieved an AUC of 0.92 and revealed distinct immune and biological characteristics in MAFLD patients across risk levels. The expression patterns and biological roles of these genes were confirmed in in vivo studies. SIGNIFICANCE This study establishes a strong link between ferroptosis- and lipid metabolism-related gene expression and MAFLD's complexity. It provides novel insights into disease mechanisms, supporting personalized prognosis and targeted therapeutic strategies for MAFLD patients.
Collapse
Affiliation(s)
- Zhihao Fang
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Changxu Liu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yue Cheng
- Cardiovascular Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanchao Ji
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chang Liu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
68
|
Li Y, Zhu B, Shi K, Lu Y, Zeng X, Li Y, Zhang Q, Feng Y, Wang X. Advances in intrahepatic and extrahepatic vascular dysregulations in cirrhotic portal hypertension. Front Med (Lausanne) 2025; 12:1515400. [PMID: 39958826 PMCID: PMC11825794 DOI: 10.3389/fmed.2025.1515400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/20/2025] [Indexed: 02/18/2025] Open
Abstract
Cirrhotic portal hypertension, the most prevalent and clinically significant complication of liver cirrhosis, manifests as elevated portal venous pressure and is associated with severe complications. Although much research on the mechanisms of portal hypertension has focused on liver fibrosis, less attention has been given to the role of intrahepatic and extrahepatic vascular dysfunction, particularly with respect to extrahepatic vasculature. While the role of hepatic fibrosis in cirrhotic portal hypertension is undeniable, the underlying mechanisms involving intrahepatic and extrahepatic vasculature are highly complex. Sinusoidal capillarization and endothelial dysfunction contribute to increased intrahepatic vascular resistance. Hemodynamic changes in the extrahepatic circulation, including splanchnic vasodilation and hyperdynamic circulation, play a significant role in the development of portal hypertension. Additionally, therapeutic strategies targeting these vascular mechanisms are diverse, including improvement of sinusoidal microcirculation, therapies targeting hepatic stellate cells activation, and pharmacological modulation of systemic vascular tone. Therefore, in this review, we will discuss the vascular-related mechanisms and treatment progress of portal hypertension in cirrhosis to provide a new theoretical basis and practical guidance for clinical treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ying Feng
- Center for Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xianbo Wang
- Center for Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
69
|
Yang S, Zou Y, Zhong C, Zhou Z, Peng X, Tang C. Dual role of pyroptosis in liver diseases: mechanisms, implications, and therapeutic perspectives. Front Cell Dev Biol 2025; 13:1522206. [PMID: 39917567 PMCID: PMC11798966 DOI: 10.3389/fcell.2025.1522206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/10/2025] [Indexed: 02/09/2025] Open
Abstract
Pyroptosis, a form of programmed cell death induced by inflammasome with a mechanism distinct from that of apoptosis, occurs via one of the three pathway types: classical, non-classical, and granzyme A/B-dependent pyroptosis pathways. Pyroptosis is implicated in various diseases, notably exhibiting a dual role in liver diseases. It facilitates the clearance of damaged hepatocytes, preventing secondary injury, and triggers immune responses to eliminate pathogens and damaged cells. Conversely, excessive pyroptosis intensifies inflammatory responses, exacerbates hepatocyte damage and promotes the activation and proliferation of hepatic stellate cells, accelerating liver fibrosis. Furthermore, by sustaining an inflammatory state, impacts the survival and proliferation of cancer cells. This review comprehensively summarizes the dual role of pyroptosis in liver diseases and its therapeutic strategies, offering new theoretical foundations and practical guidance for preventing and treating of liver diseases.
Collapse
Affiliation(s)
| | | | | | - Zuoqiong Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Xiyang Peng
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Changfa Tang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| |
Collapse
|
70
|
Xuan W, Song D, Hou J, Meng X. Regulation of Hippo-YAP1/TAZ pathway in metabolic dysfunction-associated steatotic liver disease. Front Pharmacol 2025; 16:1505117. [PMID: 39917623 PMCID: PMC11798981 DOI: 10.3389/fphar.2025.1505117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/07/2025] [Indexed: 02/09/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has become the most prevalent chronic liver disease worldwide, but effective treatments are still lacking. Metabolic disorders such as iron overload, glycolysis, insulin resistance, lipid dysregulation, and glutaminolysis are found to induce liver senescence and ferroptosis, which are hot topics in the research of MASLD. Recent studies have shown that Hippo-YAP1/TAZ pathway is involved in the regulations of metabolism disorders, senescence, ferroptosis, inflammation, and fibrosis in MASLD, but their complex connections and contrast roles are also reported. In addition, therapeutics based on the Hippo-YAP1/TAZ pathway hold promising for MASLD treatment. In this review, we highlight the regulation and molecular mechanism of the Hippo-YAP1/TAZ pathway in MASLD and summarize potential therapeutic strategies for MASLD by regulating Hippo-YAP1/TAZ pathway.
Collapse
Affiliation(s)
- Wei Xuan
- Department of Hepatopancreaticobiliary Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Dandan Song
- Department of Clinical Laboratory, Second Hospital of Jilin University, Changchun, China
| | - Jianghua Hou
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xiuping Meng
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
71
|
Fan Z, Wei X, Zhu X, Du Y. Sirtuins in kidney homeostasis and disease: where are we now? Front Endocrinol (Lausanne) 2025; 15:1524674. [PMID: 39911234 PMCID: PMC11794115 DOI: 10.3389/fendo.2024.1524674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 12/31/2024] [Indexed: 02/07/2025] Open
Abstract
Sirtuins, identified as (NAD+)- dependent class III histone deacetylases, engage in a spectrum of biological functions, encompassing DNA damage repair, oxidative stress, immune modulation, mitochondrial homeostasis, apoptosis and autophagy. Sirtuins play an apoptosis role in regulating cellular operations and overall organism health. Mounting data indicate that dysregulated sirtuin expression is linked to the onset of renal diseases. Effective modulation of sirtuins expression and activity has been shown to improve renal function and attenuate the advancement of kidney diseases. In this review, we present a comprehensive overview of the biological impacts of sirtuins and their molecular targets in regulating renal diseases. Additionally, we detail advancements in elucidating sirtuin roles in the pathophysiology of both chronic and acute renal disorders. We review compounds that modulate sirtuin activity through activation or inhibition, potentially improving outcomes in renal disease. In summary, strategic manipulation of sirtuin activity represents a prospective therapeutic approach for renal diseases.
Collapse
Affiliation(s)
| | | | | | - Yujun Du
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
72
|
Zhu J, Wu X, Mu M, Zhang Q, Zhao X. TEC-mediated tRF-31R9J regulates histone lactylation and acetylation by HDAC1 to suppress hepatocyte ferroptosis and improve non-alcoholic steatohepatitis. Clin Epigenetics 2025; 17:9. [PMID: 39838504 PMCID: PMC11748747 DOI: 10.1186/s13148-025-01813-3] [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: 08/05/2024] [Accepted: 01/06/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Tectorigenin (TEC) is a monomer of anthocyanin, which we found exhibits hepatoprotective effects. tRNA-derived fragments (tRFs) and ferroptosis play important roles in the pathogenesis of non-alcoholic steatohepatitis (NASH). Recent discoveries have revealed that histone lactylation and acetylation play a crucial role in connecting cellular metabolism and epigenetic regulation through post-translational modification of histones. However, it is unclear whether TEC improves NASH by regulating histone lactylation, acetylation and hepatocyte ferroptosis through tRFs. RESULTS In this study, we demonstrated that TEC significantly inhibits free fatty acids-induced hepatocyte ferroptosis both in vitro and in vivo. We identified tRF-31R9J (tRF-31-R9JP9P9NH5HYD) involved in TEC regulation of ferroptosis in steatosis hepatocytes. Overexpression of tRF-31R9J suppressed hepatocyte ferroptosis and enhanced cell viability in steatosis HepG2 cells. Knockdown of tRF-31R9J partially counteracted the inhibitory effect of TEC on ferroptosis in hepatocytes. Mechanistically, tRF-31R9J recruited HDAC1 to reduce the levels of histone lactylation and acetylation modifications of the pro-ferroptosis genes ATF3, ATF4, and CHAC1, thereby inhibiting their gene expression. CONCLUSIONS This study demonstrates that TEC-mediated tRF-31R9J inhibits hepatocyte ferroptosis through HDAC1-regulated histone delactylation and deacetylation, thereby improving NASH. These discoveries offer a theoretical foundation and new strategies for the medical management of NASH.
Collapse
Affiliation(s)
- Juanjuan Zhu
- Department of Infection, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Guiyang, 550001, Guizhou, China
| | - Xian Wu
- Department of Infection, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Guiyang, 550001, Guizhou, China
| | - Mao Mu
- Department of Infection, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Guiyang, 550001, Guizhou, China
| | - Quan Zhang
- Department of Infection, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Guiyang, 550001, Guizhou, China
| | - Xueke Zhao
- Department of Infection, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Guiyang, 550001, Guizhou, China.
| |
Collapse
|
73
|
Zhao H, Zheng X, Lin G, Wang X, Lu H, Xie P, Jia S, Shang Y, Wang Y, Bai P, Zhang X, Tang N, Qi X. Effects of air pollution on the development and progression of digestive diseases: an umbrella review of systematic reviews and meta-analyses. BMC Public Health 2025; 25:183. [PMID: 39819486 PMCID: PMC11740668 DOI: 10.1186/s12889-024-21257-3] [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: 08/15/2024] [Accepted: 12/30/2024] [Indexed: 01/19/2025] Open
Abstract
Air pollution, especially particulate matter (PM), is one of the most common risk factors for global burden of disease. However, its effect on the risk of digestive diseases is unclear. Herein, we attempt to explore this issue by reviewing the existing evidence from published meta-analyses. We conducted a systematic literature search to identify all relevant meta-analyses regarding the association of air pollution with digestive diseases, and summarize their major findings. We assessed the methodological quality and evidence quality of the included meta-analyses using the AMSTAR-2 and GRADE tools, respectively, and the overlap of primary studies was assessed by the GROOVE tool. Nine meta-analyses were included in our analysis, containing 43 primary studies with high overlap. In the included meta-analyses, the methodological quality was from critically low to moderate, and the evidence quality was from very low to moderate. The exposure was primarily PM2.5. Seven, four, and one meta-analysis investigated the effect of air pollution on liver diseases, gastrointestinal diseases, and pancreatic diseases, respectively. PM2.5 exposure was significantly associated with liver dysfunction, chronic liver diseases, liver cancer, and colorectal cancer, but not oesophagus cancer, gastric cancer, or pancreatic cancer. Based on very low to moderate quality evidence from meta-analyses, PM2.5 exposure may contribute to the development of some digestive diseases, especially liver diseases.
Collapse
Affiliation(s)
- Haonan Zhao
- Department of Gastroenterology, General Hospital of Northern Theater Command (Teaching Hospital of Shenyang Pharmaceutical University), Shenyang, 110840, China
- Department of Life Sciences and Biopharmaceutis, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaojie Zheng
- Department of Gastroenterology, General Hospital of Northern Theater Command (Teaching Hospital of Shenyang Pharmaceutical University), Shenyang, 110840, China
- Second Affiliated Hospital of Shenyang Medical College, Shenyang, China
| | - Guo Lin
- Department of Gastroenterology, General Hospital of Northern Theater Command (Teaching Hospital of Shenyang Pharmaceutical University), Shenyang, 110840, China
- Postgraduate College, Dalian Medical University, Dalian, China
| | - Xiaomin Wang
- Department of Gastroenterology, General Hospital of Northern Theater Command (Teaching Hospital of Shenyang Pharmaceutical University), Shenyang, 110840, China
- Postgraduate College, Dalian Medical University, Dalian, China
| | - Huiyuan Lu
- Department of Gastroenterology, General Hospital of Northern Theater Command (Teaching Hospital of Shenyang Pharmaceutical University), Shenyang, 110840, China
- Department of Life Sciences and Biopharmaceutis, Shenyang Pharmaceutical University, Shenyang, China
| | - Pengpeng Xie
- Department of Gastroenterology, General Hospital of Northern Theater Command (Teaching Hospital of Shenyang Pharmaceutical University), Shenyang, 110840, China
- Department of Life Sciences and Biopharmaceutis, Shenyang Pharmaceutical University, Shenyang, China
| | - Siqi Jia
- Department of Gastroenterology, General Hospital of Northern Theater Command (Teaching Hospital of Shenyang Pharmaceutical University), Shenyang, 110840, China
- Department of Life Sciences and Biopharmaceutis, Shenyang Pharmaceutical University, Shenyang, China
| | - Yiyang Shang
- Department of Gastroenterology, General Hospital of Northern Theater Command (Teaching Hospital of Shenyang Pharmaceutical University), Shenyang, 110840, China
- Department of Life Sciences and Biopharmaceutis, Shenyang Pharmaceutical University, Shenyang, China
| | - Yan Wang
- Graduate School of Medical Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Pengchu Bai
- Graduate School of Medical Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Xuan Zhang
- National Institute of Occupational Safety and Health, Kanagawa, 214-8585, Japan
- Institute of Nature and Environmental Technology, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Ning Tang
- Institute of Nature and Environmental Technology, Kanazawa University, Kanazawa, 920-1192, Japan.
- Institute of Medical, Pharmaceutical and Health Science, Kanazawa University, Kanazawa, 920-1192, Japan.
- College of Energy and Power, Shenyang Institute of Engineering, Shenyang, 110136, China.
| | - Xingshun Qi
- Department of Gastroenterology, General Hospital of Northern Theater Command (Teaching Hospital of Shenyang Pharmaceutical University), Shenyang, 110840, China.
- Department of Life Sciences and Biopharmaceutis, Shenyang Pharmaceutical University, Shenyang, China.
- Postgraduate College, Dalian Medical University, Dalian, China.
- Institute of Nature and Environmental Technology, Kanazawa University, Kanazawa, 920-1192, Japan.
| |
Collapse
|
74
|
Na EJ, Jeong CG, Chae SB, Oem JK. Investigating the reassortment potential and pathogenicity of the S segment in Akabane virus using a reverse genetics system. BMC Vet Res 2025; 21:20. [PMID: 39815297 PMCID: PMC11734451 DOI: 10.1186/s12917-024-04459-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 12/23/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Akabane virus (AKAV) is an arthropod-borne virus that causes congenital malformations and neuropathology in cattle and sheep. In South Korea, AKAVs are classified into two main genogroups: K0505 and AKAV-7 strains. The K0505 strain infects pregnant cattle, leading to fetal abnormalities, while the AKAV-7 strain induces encephalomyelitis in post-natal cattle. The pathogenicities of K0505 and AKAV-7 strains differ significantly; however, the specific gene in the AKAV-7 strain that drives its pathogenicity remains unidentified. In this study, changes in viral replication and pathogenicity were investigated, particularly when the S segment of AKAV-7 was mutated using a T7 RNA polymerase-based reverse genetics (RG) system. RESULTS The rAKAV-7ΔNSs virus, with a deletion in the NSs protein of the wild-type AKAV-7 virus (wtAKAV-7), and the rAKAV-7(S-K0505) virus, where the S segment of wtAKAV-7 was reassorted with that from the wild type K0505 strain (wtK0505), were successfully rescued. The rAKAV-7ΔNSs virus demonstrated impaired replication in Vero cells and exhibited reduced mortality and RNA viral load in the organs of suckling mice compared to the wtAKAV-7. The rAKAV-7(S-K0505) virus displayed similar growth kinetics in Vero cells and showed no significant reduction in mortality rate in suckling mice compared to wtAKAV-7. CONCLUSIONS These observations suggest that the S segment, especially the NS protein, is associated with the pathogenicity of AKAV-7. Also, the results imply that the L and M segments might explain the differences in pathogenicity between the AKAV-7 and K0505 strains. Moreover, our findings indicate the potential for reassortment between distinct genogroups of AKAVs.
Collapse
Affiliation(s)
- Eun-Jee Na
- Laboratory of Veterinary Infectious Disease, College of Veterinary of Medicine, Jeonbuk National University, Iksan, Jeonbuk, 54596, Republic of Korea
| | - Chang-Gi Jeong
- Laboratory of Veterinary Infectious Disease, College of Veterinary of Medicine, Jeonbuk National University, Iksan, Jeonbuk, 54596, Republic of Korea
| | - Su-Beom Chae
- Laboratory of Veterinary Infectious Disease, College of Veterinary of Medicine, Jeonbuk National University, Iksan, Jeonbuk, 54596, Republic of Korea
| | - Jae-Ku Oem
- Laboratory of Veterinary Infectious Disease, College of Veterinary of Medicine, Jeonbuk National University, Iksan, Jeonbuk, 54596, Republic of Korea.
| |
Collapse
|
75
|
Hu Y, Zhang Z, Adiham A, Li H, Gu J, Gong P. In Vivo and In Vitro Models of Hepatic Fibrosis for Pharmacodynamic Evaluation and Pathology Exploration. Int J Mol Sci 2025; 26:696. [PMID: 39859410 PMCID: PMC11766297 DOI: 10.3390/ijms26020696] [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: 10/22/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Hepatic fibrosis (HF) is an important pathological state in the progression of chronic liver disease to end-stage liver disease and is usually triggered by alcohol, nonalcoholic fatty liver, chronic hepatitis viruses, autoimmune hepatitis (AIH), or cholestatic liver disease. Research on novel therapies has become a hot topic due to the reversibility of HF. Research into the molecular mechanisms of the pathology of HF and potential drug screening relies on reliable and rational biological models, mainly including animals and cells. Hence, a number of modeling approaches have been attempted based on human dietary, pathological, and physiological factors in the development of HF. In this review, classical and novel methods of modeling HF in the last 10 years were collected from electronic databases, including Web of Science, PubMed, ScienceDirect, ResearchGate, Baidu Scholar, and CNKI. Animal models of HF are usually induced by chemical toxicants, special diets, pathogenic microorganisms, surgical operations, and gene editing. The advantages and limitations of hepatic stellate cells (HSCs), organoids, and 3D coculture-based HF modeling methods established in vitro were also proposed and summarized. This information provides a scientific basis for the discovery of the pathological mechanism and treatment of HF.
Collapse
Affiliation(s)
| | | | | | | | - Jian Gu
- College of Pharmacy and Food, Southwest Minzu University, Chengdu 610093, China; (Y.H.); (Z.Z.); (A.A.); (H.L.)
| | - Puyang Gong
- College of Pharmacy and Food, Southwest Minzu University, Chengdu 610093, China; (Y.H.); (Z.Z.); (A.A.); (H.L.)
| |
Collapse
|
76
|
Zhang J, Hou L, Lei S, Li Y, Xu G. The causal relationship of cigarette smoking to metabolic disease risk and the possible mediating role of gut microbiota. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117522. [PMID: 39709709 DOI: 10.1016/j.ecoenv.2024.117522] [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: 07/31/2024] [Revised: 11/14/2024] [Accepted: 12/08/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Cigarette smoking is a leading cause of preventable death worldwide, with its associated diseases and conditions. Emerging evidence suggests that cigarette smoking contributes to a range of pathological metabolic injuries, including diabetes and nonalcoholic fatty liver disease (NAFLD). The impact of gut microbiota on metabolic health and diseases has been observed, but the causality remains uncertain. OBJECTIVE To confirm the causal relationship between cigarette smoking and metabolic diseases, and to investigate the possible mediating effect of gut microbiota on these connections. METHODS The relationships among cigarette smoking, metabolic diseases, and the gut microbiome were analyzed by Univariate Mendelian randomization (UVMR). Furthermore, to mitigate the impact of confounding factors, adjusted models were conducted via the multivariate Mendelian randomization (MVMR) method, aiming to improve the accuracy of prediction. Ultimately, the study evaluated the effect of the intermediary factor, gut microbiome, on the relationship between cigarette smoke and metabolic diseases. RESULTS The phenomenon that a causal relationship between cigarette smoke (249752 individuals) and gut microbiota (7738 individuals), diabetes (406831 individuals), NAFLD (377998 individuals), hypercholesterolaemia (463010 individuals), and obesity (463010 individuals) was observed using UVMR. In the MVMR model, the genetic connection between cigarette smoking, gut microbiota, and type 2 diabetes remained significant. Of note, paraprevotella_clara served an important mediating role in the type 2 diabetes associated with cigarette smoke. CONCLUSION This work offered genetic evidence linking cigarette smoke to metabolic diseases, suggesting that the gut microbiota, particularly paraprevotella_clara, might be a crucial mediator in the development of type 2 diabetes caused by cigarette smoke. Our future studies should consider conducting other ethnic groups MR analyses, particularly with larger sample sizes. Still, more in vivo and in vitro work should be carried out to validate the precise effect and molecular mechanisms of the gut microbiome.
Collapse
Affiliation(s)
- Jingda Zhang
- The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Lin Hou
- Department of Physiology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100730, China
| | - Shanxiang Lei
- The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Yan Li
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Guogang Xu
- The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, China; State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
77
|
Shang S, Yang H, Qu L, Fan D, Deng J. Ginsenoside, a potential natural product against liver diseases: a comprehensive review from molecular mechanisms to application. Crit Rev Food Sci Nutr 2025:1-25. [PMID: 39810734 DOI: 10.1080/10408398.2025.2451761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Liver disease constitutes a significant cause of global mortality, with its pathogenesis being multifaceted. Identifying effective pharmacological and preventive strategies is imperative for liver protection. Ginsenosides, the major bioactive compounds found in ginseng, exhibit multiple pharmacological activities including protection against liver-related diseases by mitigating liver fat accumulation and inflammation, preventing hepatic fibrosis, and exerting anti-hepatocarcinogenic effects. However, a comprehensive overview elucidating the regulatory pathways associated with ginsenosides in liver disease remains elusive. This review aims to consolidate the molecular mechanisms through which different ginsenosides ameliorate distinct liver diseases, alongside the pathogenic factors underlying liver ailments. Notably, ginsenosides Rb1 and Rg1 demonstrate significantly effective in treating fatty liver, hepatitis, and liver fibrosis, and ginsenosides CK and Rh2 exhibit potent anti-hepatocellular carcinogenic effects. Their molecular mechanisms underlying these effects primarily involve the modulation of AMPK, NF-κB, TGF-β, NFR2, JNK, and other pathways, thereby attenuating hepatic fat accumulation, inflammation, inhibition of hepatic stellate cell activation, and promoting apoptosis in hepatocellular carcinoma cells. Furthermore, it provides insights into the safety profile and current applications of ginsenosides, thereby facilitating their clinical development. Consequently, ginsenosides present promising prospects for liver disease management, underscoring their potential as valuable therapeutic agents in this context.
Collapse
Affiliation(s)
- Shiyan Shang
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, Biotech & Biomed Research Institute, School of Chemical Engineering, Northwest University, Xi'an, China
| | - Haixia Yang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Linlin Qu
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, Biotech & Biomed Research Institute, School of Chemical Engineering, Northwest University, Xi'an, China
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, Biotech & Biomed Research Institute, School of Chemical Engineering, Northwest University, Xi'an, China
| | - Jianjun Deng
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials and Fermentation Engineering, Biotech & Biomed Research Institute, School of Chemical Engineering, Northwest University, Xi'an, China
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
78
|
Fang H, Rodrigues e-Lacerda R, Barra NG, Kukje Zada D, Robin N, Mehra A, Schertzer JD. Postbiotic Impact on Host Metabolism and Immunity Provides Therapeutic Potential in Metabolic Disease. Endocr Rev 2025; 46:60-79. [PMID: 39235984 PMCID: PMC11720174 DOI: 10.1210/endrev/bnae025] [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: 04/22/2024] [Revised: 07/18/2024] [Accepted: 09/04/2024] [Indexed: 09/07/2024]
Abstract
The gut microbiota influences aspects of metabolic disease, including tissue inflammation, adiposity, blood glucose, insulin, and endocrine control of metabolism. Prebiotics or probiotics are often sought to combat metabolic disease. However, prebiotics lack specificity and can have deleterious bacterial community effects. Probiotics require live bacteria to find a colonization niche sufficient to influence host immunity or metabolism. Postbiotics encompass bacterial-derived components and molecules, which are well-positioned to alter host immunometabolism without relying on colonization efficiency or causing widespread effects on the existing microbiota. Here, we summarize the potential for beneficial and detrimental effects of specific postbiotics related to metabolic disease and the underlying mechanisms of action. Bacterial cell wall components, such as lipopolysaccharides, muropeptides, lipoteichoic acids and flagellin, have context-dependent effects on host metabolism by engaging specific immune responses. Specific types of postbiotics within broad classes of compounds, such as lipopolysaccharides and muropeptides, can have opposing effects on endocrine control of host metabolism, where certain postbiotics are insulin sensitizers and others promote insulin resistance. Bacterial metabolites, such as short-chain fatty acids, bile acids, lactate, glycerol, succinate, ethanolamine, and ethanol, can be substrates for host metabolism. Postbiotics can fuel host metabolic pathways directly or influence endocrine control of metabolism through immunomodulation or mimicking host-derived hormones. The interaction of postbiotics in the host-microbe relationship should be considered during metabolic inflammation and metabolic disease.
Collapse
Affiliation(s)
- Han Fang
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Rodrigo Rodrigues e-Lacerda
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Nicole G Barra
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Dana Kukje Zada
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Nazli Robin
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Alina Mehra
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| |
Collapse
|
79
|
Fan Y, Ren Y, Deng L, Lv D, Chen J, Ling Y, Tu J, Xu X, Wang D, Cai Z. Testosterone deficiency aggravates diet-induced non-alcoholic fatty liver disease by inducing hepatocyte ferroptosis via targeting BMAL1 in mice. Int Immunopharmacol 2025; 144:113641. [PMID: 39579542 DOI: 10.1016/j.intimp.2024.113641] [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: 08/23/2024] [Revised: 11/01/2024] [Accepted: 11/13/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND Testosterone deficiency is linked to an increased prevalence of non-alcoholic fatty liver disease (NAFLD), although the mechanisms underlying this association are not fully understood. Ferroptosis, a regulated cell death pathway driven by iron-dependent lipid peroxidation, has been suggested to play a role in NAFLD pathogenesis. Since testosterone deficiency is associated with lipid disorders and iron deposition, we hepothesize that ferroptosis may be involved in the pathogenesis of diet-induced NAFLD exacerbated by testosterone deficiency. METHODS Apolipoprotein E (APOE-/-) mice were subjected to sham surgery or bilateral castration and subsequently fed a high-fat diet for 16 weeks. Liver gene expression was analyzed using RNA sequencing. Additional assessments included blood analysis, histological staining, measurement of iron and antioxidant enzyme levels, quantitative real-time PCR, Western blotting, and electron microscopy. The effects of testosterone on ferroptosis induced by free fatty acids (FFAs) and Erastin were further investigated in HepG2 cells in vitro. RESULTS Testosterone deficiency resulted in increased hepatic lipid accumulation and macrovesicular steatosis in high-fat diet-fed APOE-/- mice, accompanied by hepatic inflammation, fibrosis, and elevated liver enzyme levels. Transcriptomic analysis revealed that testosterone deficiency affects ferroptosis and circadian rhythm-related signaling pathways. Castrated APOE-/- mice exhibited significantly higher hepatic iron deposition, lipid peroxidation, and expression of key ferroptosis-related proteins, along with decreased Brain and muscle ARNT-like gene 1 (BMAL1) protein expression. In vitro, testosterone treatment reduced lipid and iron accumulation and lipid peroxidation in HepG2 cells subjected to FFAs and Erastin. Moreover, BMAL1 knockdown negated the protective effects of testosterone against ferroptosis in hepatocytes. CONCLUSION Our study demonstrated that testosterone deficiency exacerbates NAFLD induced by a high-fat diet by promoting hepatocyte ferroptosis through modulation of the circadian protein BMAL1.
Collapse
Affiliation(s)
- Yingying Fan
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yujie Ren
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Liqun Deng
- Laboratory Animal Resources Center, Westlake University, Hangzhou, China, 310053
| | - Dongying Lv
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiayan Chen
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yun Ling
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jue Tu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaoping Xu
- Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Dejun Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China; Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhaowei Cai
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310053, China; Laboratory Animal Research Center, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
80
|
Choi H, Choi J, Go Y, Chung J. Coenzyme Q and Selenium Co-Supplementation Alleviate Methionine Choline-Deficient Diet-Induced Metabolic Dysfunction-Associated Steatohepatitis in Mice. Nutrients 2025; 17:229. [PMID: 39861360 PMCID: PMC11767553 DOI: 10.3390/nu17020229] [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: 12/23/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES The pathogenesis of metabolic dysfunction-associated steatohepatitis (MASH) is closely associated with increased oxidative stress and lipid peroxidation. Coenzyme Q (CoQ) and selenium (Se) are well-established antioxidants with protective effects against oxidative damage. This study aimed to investigate the effects of CoQ and Se in ameliorating MASH induced by a methionine choline-deficient (MCD) diet in mice. METHODS C57BL/6J male mice were fed either a methionine choline-sufficient (MCS) or MCD diet and treated with vehicle, CoQ (100 mg/kg), Se (158 μg/kg), or their combination (CoQ + Se) for 4 weeks. RESULTS The MCD diet significantly increased hepatic steatosis, inflammation, and fibrosis compared to MCS controls. Treatment with CoQ and Se, particularly in combination, markedly reduced the MAFLD activity score, hepatic inflammation, and fibrosis. Combined supplementation of CoQ and Se significantly decreased serum alanine aminotransferase and aspartate aminotransferase levels and hepatic TG and cholesterol concentrations. CoQ and Se effectively mitigated hepatic oxidative stress by enhancing catalase and superoxide dismutase activities, increasing glutathione peroxidase (GPX) activity, and restoring the GSH/GSSG ratio. Lipid peroxidation markers, such as malondialdehyde and 4-hydroxynonenal, were significantly reduced. Furthermore, the expression of ferroptosis-related markers, including acyl-CoA synthetase long-chain family member 4, arachidonate 12-lipoxygenase, and hepatic non-heme iron content, was significantly downregulated, while GPX4 expression was upregulated by combined CoQ and Se treatment. CONCLUSIONS CoQ and Se synergistically alleviate MASH progression by reducing oxidative stress and lipid peroxidation, which may contribute to the suppression of ferroptosis. Combined CoQ and Se supplementation demonstrates therapeutic potential for managing MASH and related liver injury.
Collapse
Affiliation(s)
| | | | | | - Jayong Chung
- Department of Food & Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
81
|
Hao L, Li S, Li C, Zhang Z, Hu X, Yan H. A Review of the Therapeutic Potential of Ginseng and Its Bioactive Components in Nonalcoholic Fatty Liver Disease. Drug Des Devel Ther 2025; 19:83-96. [PMID: 39803604 PMCID: PMC11725245 DOI: 10.2147/dddt.s500719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the major cause of chronic liver disease worldwide, with no universally recognized effective treatments currently available. In recent years, ginseng and its principal active components, such as ginsenosides, have shown potential protective effects in the treatment of these liver diseases. In NAFLD, studies have demonstrated that ginseng can improve hepatic lipid metabolism, reduce inflammatory responses, and inhibit oxidative stress and fibrosis, thereby attenuating the progression of NAFLD. Additionally, ginseng inhibits oxidative stress by scavenging free radicals and enhancing antioxidant enzyme activities, and it can impede fibrosis by interfering with the fibrotic signaling pathways. These combined effects contribute to attenuating the progression of NAFLD. These findings highlight the promise of ginseng as a potential therapeutic candidate for the treatment of NAFLD. However, despite the significant efficacy of ginseng in human NAFLD treatment, the number and quality of clinical studies remain limited, with a lack of large-scale, multicenter clinical trials to confirm these effects. Moreover, the pharmacokinetic properties of different ginsenosides, optimal therapeutic dosages, and the safety of long-term use require further investigation. This review summarizes the existing evidence on the mechanisms of action of ginseng and its active components in human NAFLD, assesses their potential as therapeutic options, and proposes future research directions to provide stronger scientific support for clinical application. Additionally, we performed a network pharmacology analysis of ginseng in relation to NAFLD to identify and investigate potential targets of ginseng in the treatment of NAFLD. This analysis aims to provide a theoretical foundation for the development of ginseng -based drugs for combating NAFLD.
Collapse
Affiliation(s)
- Liyuan Hao
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Shenghao Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
- Clinical Research Center, Shijiazhuang Fifth Hospital, Shijiazhuang, Hebei, People’s Republic of China
| | - Caige Li
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People’s Republic of China
| | - Zhiqin Zhang
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Traditional Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China
| | - Xiaoyu Hu
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Huimin Yan
- Clinical Research Center, Shijiazhuang Fifth Hospital, Shijiazhuang, Hebei, People’s Republic of China
| |
Collapse
|
82
|
Chen XY, Zhi LJ, Chen J, Li R, Long KL. Research hotspots and future trends in sepsis-associated acute kidney injury: a bibliometric and visualization analysis. Front Med (Lausanne) 2025; 11:1456535. [PMID: 39839617 PMCID: PMC11747655 DOI: 10.3389/fmed.2024.1456535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 11/29/2024] [Indexed: 01/23/2025] Open
Abstract
Objectives Sepsis-associated acute kidney injury (SA-AKI) commonly occurs in critically ill patients and is closely associated with adverse outcomes. A comprehensive analysis of the current research landscape in SA-AKI can help uncover trends and key issues in this field. This study aims to provide a scientific basis for research directions and critical issues through bibliometric analysis. Methods We searched all articles on SA-AKI indexed in the SCI-Expanded of WoSCC up to May 7, 2024, and conducted bibliometric and visual analyses using bibliometric software CiteSpace and VOSviewer. Results Over the past 20 years, there has been a steady increase in literature related to renal repair following AKI. China and the United States contribute over 60% of the publications, driving research in this field. The University of Pittsburgh is the most active academic institution, producing the highest number of publications. J. A. Kellum is both the most prolific and the most cited author in this area. "Shock" and "American Journal of Physiology-Renal Physiology" are the most popular journals, publishing the highest number of articles. Recent high-frequency keywords in this field include "septic AKI," "mitochondrial dysfunction," "inflammasome," "ferroptosis," and "macrophage." The terms "mitochondrial dysfunction," "inflammasome," "ferroptosis," and "macrophage" represent current research hotspots and potential targets in this area. Conclusion This is the first comprehensive bibliometric study to summarize the trends and advancements in SA-AKI research in recent years. These findings identify current research frontiers and hot topics, providing valuable insights for scholars studying SA-AKI.
Collapse
Affiliation(s)
- Xing-Yue Chen
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li-Jia Zhi
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Chen
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rong Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kun-Lan Long
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
83
|
Zhang M, Li K, Huang X, Xu D, Zong R, Hu Q, Dong X, Zhang Q, Jiang C, Ge Y, Li C, Ping J. Macrophage Notch1 signaling modulates regulatory T cells via the TGFB axis in early MASLD. JHEP Rep 2025; 7:101242. [PMID: 39717502 PMCID: PMC11664078 DOI: 10.1016/j.jhepr.2024.101242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 12/25/2024] Open
Abstract
Background & Aims Hepatic immune imbalance is crucial for driving metabolic dysfunction-associated steatotic liver disease (MASLD) progression. However, the role of hepatic regulatory T cells (Tregs) in MASLD initiation and the mechanisms responsible for their change are not completely understood. Methods A mouse model subjected to a short-term high-fat diet (HFD) to mimic early steatosis, along with liver biopsy samples from patients with simple steatosis, and macrophage-specific Notch1-knockout mice (Notch1M-KO), were used to investigate the role of Tregs in early MASLD and the effect of hepatic macrophage Notch1 signaling on Treg frequency. The miRNAs correlated with Treg differentiation were analyzed using exosomal miRNA sequencing. Results A decrease in Tregs contributed to HFD-induced hepatic steatosis and insulin resistance (five/group/time point, p <0.001). Remarkably, the frequency of Tregs was negatively correlated with Notch1 activation in hepatic macrophages during hepatic steatosis (38/group, r = -0.735, p <0.001). Furthermore, Notch1 deficiency attenuated hepatic lipid deposition and reversed Treg levels (five/group, p <0.01 and <0.05, respectively). Moreover, Treg depletion in Notch1M-KO mice greatly diminished the ameliorative effect of macrophagic Notch1 deletion on hepatic steatosis. Mechanistically, macrophage Notch1 activation increased the level of exosomal miR-142a-3p (by one- to two- fold), impairing Treg differentiation by targeting transforming growth factor beta receptor 1 (TGFBR1) on T cells. Consistently, HFD-fed Notch1M-KO mice exhibited reduced miR-142a-3p levels, elevated TGFBR1 expression on T cells, and increased Treg frequency in the liver. Conclusions These findings highlight the crucial role of hepatic Tregs during the early stage of MASLD and add a novel, non-negligible pathway for macrophage involvement in hepatic steatosis. We identify a previously unrecognized molecular mechanism involving the macrophage Notch1/exosomal miR-142a-3p/TGFBR1 pathway in regulating Treg differentiation, providing a rationale for refined therapeutic strategies for MASLD. Impact and implications The immune mechanisms driving MASLD progression, particularly during the early stages of disease, are not fully understood, which limits the development of effective interventions. This study elucidated a novel mechanism by which hepatic macrophage Notch1 signaling modulated Tregs through the exosomal miR-142a-3p/TGFBR1 axis, contributing to the progression of MASLD. These findings provide a rationale for a potential immunological approach to treat MASLD in the future.
Collapse
Affiliation(s)
- Mengya Zhang
- Department of Pharmacology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China
| | - Kun Li
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Medicine Research Center for Minimally Invasive Diagnosis and Treatment of Hepatobiliary and Pancreatic Diseases, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xiaoxing Huang
- Department of Blood Transfusion, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Dongqin Xu
- Department of Pharmacology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China
| | - Ruobin Zong
- Department of Physiology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China
| | - Qintong Hu
- Department of Pharmacology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China
| | - Xiaoyu Dong
- Department of Pharmacology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China
| | - Qinyong Zhang
- Department of Pharmacology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China
| | - Chaochen Jiang
- Department of Pharmacology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China
| | - Yue Ge
- Department of Pharmacology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China
| | - Changyong Li
- Department of Physiology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China
| | - Jie Ping
- Department of Pharmacology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan 430071, China
| |
Collapse
|
84
|
Udaipuria N, Bhattacharya S. Novel Carbohydrate Polymer-Based Systems for Precise Drug Delivery in Colon Cancer: Improving Treatment Effectiveness With Intelligent Biodegradable Materials. Biopolymers 2025; 116:e23632. [PMID: 39340194 DOI: 10.1002/bip.23632] [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: 07/31/2024] [Revised: 09/07/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024]
Abstract
Due to their biocompatibility, biodegradability, and controlled release, carbohydrates polymers are crucial to targeted drug delivery systems, notably for colon cancer treatment. This article examines how carbohydrate polymers like chitosan, pectin, guar gum, alginate, hyaluronic acid, dextran, and chondroitin sulfate are used in improved drug delivery. Modifying these polymers improves drug loading, stability, and release patterns, enhancing chemotherapeutic drugs' therapeutic index. Chitosan nanoparticles are pH-responsive, making them perfect for cancer treatment. Pectin's resistance to gastric enzymes and colonic bacteria makes it a promising colon-specific medication delivery agent. The combination of these polymers with nanotechnology, 3D printing, and AI allows the creation of stimuli-responsive systems that release drugs precisely in response to environmental signals like pH, redox potential, or colon enzymatic activity. The review highlights intelligent delivery system design advances that reduce systemic toxicity, improve treatment efficacy, and improve patient adherence. Carbohydrate polymers will revolutionize colon cancer treatment with personalized and accurate alternatives.
Collapse
Affiliation(s)
- Nikita Udaipuria
- School of Pharmacy and Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, India
| | - Sankha Bhattacharya
- School of Pharmacy and Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, India
| |
Collapse
|
85
|
Zhu A, Yan X, Chen M, Lin Y, Li L, Wang Y, Huang J, He J, Yang M, Hua W, Chen K, Qi J, Zhou Z. Sappanone A alleviates metabolic dysfunction-associated steatohepatitis by decreasing hepatocyte lipotoxicity via targeting Mup3 in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156341. [PMID: 39733550 DOI: 10.1016/j.phymed.2024.156341] [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: 08/02/2024] [Revised: 12/01/2024] [Accepted: 12/19/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND AND PURPOSE Metabolic dysfunction-associated steatohepatitis (MASH) is an inflammatory lipotoxic disorder marked by hepatic steatosis, hepatocyte damage, inflammation, and varying stages of fibrosis. Sappanone A (SA), a flavonoid, exhibits anti-inflammatory and hepatoprotection activities. Nevertheless, the effects of SA on MASH remain ambiguous. We evaluated the effects of SA on hepatocyte lipotoxicity, inflammation, and fibrosis conditions in MASH mice, as well as the underlying mechanisms. METHODS A conventional murine MASH model fed a methionine-choline-deficient (MCD) diet was utilized to assess the role of SA on MASH in vivo. Drug target prediction and liver transcriptomics were employed to elucidate the potential actions of SA. AML12 cells were applied to further explore the effects and mechanisms of SA in vitro. RESULTS The in silico prediction indicated that SA could modulate inflammation, insulin resistance, lipid metabolism, and collagen catabolic process. Treating with SA dose-dependently lessened the elevated levels of serum ALT and AST in mice with diet-triggered MASH, and high-dose SA treatment exhibited a similar effect to silymarin. Additionally, SA treatment significantly reduced lipid deposition, inflammation, and fibrosis subjected to metabolic stress in a dose-dependent manner. Besides, SA mitigated palmitate-triggered lipotoxicity in hepatocytes. Liver transcriptomics further confirmed the aforementioned findings. Of note, mRNA-sequencing analysis and molecular biology experiments demonstrated that SA statistically up-regulated the hepatic expression of major urinary protein 3 (Mup3), thereby facilitating lipid transportation and inhibiting lipotoxicity. Furthermore, Mup3 knockdown in hepatocytes significantly abolished the hepatoprotection provided by SA. CONCLUSION SA alleviates MASH by decreasing lipid accumulation and lipotoxicity in hepatocytes, at least partially by targeting Mup3, and subsequently blocks MASH process. Therefore, SA could be a promising hepatoprotective agent in the context of MASH.
Collapse
Affiliation(s)
- An Zhu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, 1 Xue Fu North Road, Fuzhou 350122, China
| | - Xueqing Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, No.1, Xuefu North Road, University Town, Fuzhou, Fujian 350122, China
| | - Mengting Chen
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, 1 Xue Fu North Road, Fuzhou 350122, China
| | - Yifan Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, 1 Xue Fu North Road, Fuzhou 350122, China
| | - Lanqian Li
- Department of Pathology & Diagnosis Pathological Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Yufei Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, No.1, Xuefu North Road, University Town, Fuzhou, Fujian 350122, China
| | - Jiabin Huang
- Department of Pathology & Diagnosis Pathological Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Jiale He
- Department of Pathology & Diagnosis Pathological Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Mengchen Yang
- Department of Pathology & Diagnosis Pathological Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Wenxi Hua
- Department of Pathology & Diagnosis Pathological Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Kunqi Chen
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, 1 Xue Fu North Road, Fuzhou 350122, China.
| | - Jing Qi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, No.1, Xuefu North Road, University Town, Fuzhou, Fujian 350122, China.
| | - Zixiong Zhou
- Department of Pathology & Diagnosis Pathological Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China.
| |
Collapse
|
86
|
Wang Y, Yang L, Xu Q, Liu T, He H, Liu L, Yin L. Tenascin C-Guided Nanosystem for Precision Delivery of Obeticholic Acid in Liver Fibrosis Therapy. Pharmaceutics 2024; 17:32. [PMID: 39861681 PMCID: PMC11768695 DOI: 10.3390/pharmaceutics17010032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 01/27/2025] Open
Abstract
Objective: Liver fibrosis, a hallmark of chronic liver diseases, is characterized by excessive extracellular matrix (ECM) deposition and scar tissue formation. Current antifibrotic nanomedicines face significant limitations, including poor penetration into fibrotic tissue, rapid clearance, and suboptimal therapeutic efficacy. The dense fibrotic ECM acts as a major physiological barrier, necessitating the development of a targeted delivery strategy to achieve effective therapeutic outcomes. Methods: We designed a liposomal delivery system functionalized with the GBI-10 aptamer and encapsulating obeticholic acid (OCA lips@Apt) to enhance selective delivery to fibrotic liver tissue while minimizing systemic toxicity. Results: Both in vitro and in vivo studies demonstrated that the aptamer-modified OCA liposomes effectively treated hepatic fibrosis through dual mechanisms: modulation of abnormal bile acid metabolism and attenuation of inflammation. The targeted delivery system leveraged the overexpression of Tenascin-C (TnC), a key ECM component in fibrotic tissues, for precise localization and enhanced endocytosis via the exposed cationic liposome surface. Conclusions: The OCA lips@Apt nanodrug demonstrated superior therapeutic efficacy with minimal off-target effects, offering a promising strategy to overcome critical barriers in liver fibrosis treatment. By precisely targeting the fibrotic ECM and modulating key pathological pathways, this TnC-guided liposomal delivery system provides a significant advancement in antifibrotic nanomedicine.
Collapse
Affiliation(s)
- Yawen Wang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Yang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Qing Xu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Taiyu Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Hongliang He
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210009, China
| | - Lisha Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China
| | - Lifang Yin
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
87
|
Gu K, Wu A, Liu C, Yu B, He J, Lai X, Chen J, Luo Y, Yan H, Zheng P, Luo J, Pu J, Wang Q, Wang H, Chen D. Absence of gut microbiota alleviates iron overload-induced colitis by modulating ferroptosis in mice. J Adv Res 2024:S2090-1232(24)00608-8. [PMID: 39710300 DOI: 10.1016/j.jare.2024.12.030] [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: 09/18/2024] [Revised: 12/02/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024] Open
Abstract
INTRODUCTION Iron overload disrupts gut microbiota and induces ferroptosis, contributing to colitis. However, whether gut microbiota directly drives iron overload-induced colitis and its underlying mechanism remain unclear. OBJECTIVES The study aimed to explore whether gut microbiota can directly regulate iron overload-induced colitis and its underling mechanism. METHODS Male C57BL/6N mice were fed with ferrous sulfate to establish an iron overload model. Antibiotics and dextran sulfate sodium salt (DSS) were used to create germ-free and colitis models, respectively. RESULTS Results showed that iron overload caused disruption of systemic iron homeostasis via activating pro-inflammation response, which caused induction of ferroptosis and eventually resulted in colitis in mice. Notably, iron overload inhibited System Xc- and activated the nuclear factor E2-related factor 2/heme oxygenase-1 pathway, driving ferroptosis and colitis progression. Similar results were observed in mouse colon epithelial cells, which were treated with high doses ferric ammonium citrate. Additionally, iron overload exacerbated DSS-induced colitis by activating the ferroptosis and increasing harmful bacteria (e.g., Mucispirillum) abundance. Interestingly, eliminating gut microbiota attenuated iron overload-induced colitis, without affecting systemic inflammation through inhibiting ferroptosis of mice. Depletion of the gut microbiota partially mitigated the exacerbating effect of iron overload on DSS-induced colitis through inhibiting ferroptosis of mice. CONCLUSION Iron overload activates ferroptosis in colonic cells, increases the relative abundance of harmful bacteria, and exacerbates DSS-induced colitis in mice. Iron overload exacerbates DSS-induced ferroptosis and colitis in a microbiota-dependent manner. Targeting gut microbiota may offer new strategies for managing iron overload-induced colitis.
Collapse
Affiliation(s)
- Ke Gu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Aimin Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Chen Liu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China; Tea Refining and Innovation Key Laboratory of Sichuan Province, College of Horticulture, Sichuan Agricultural University, Chengdu 611130, China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Xin Lai
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Junzhou Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuheng Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Junning Pu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Quyuan Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Huifen Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
88
|
Vahid ZF, Eskandani M, Dadashi H, Vandghanooni S, Rashidi MR. Recent advances in potential enzymes and their therapeutic inhibitors for the treatment of Alzheimer's disease. Heliyon 2024; 10:e40756. [PMID: 39717593 PMCID: PMC11664286 DOI: 10.1016/j.heliyon.2024.e40756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/25/2024] Open
Abstract
Alzheimer's disease (AD), a chronic neurodegenerative disease, is clinically characterized by loss of memory and learning ability among other neurological deficits. Amyloid plaques, hyperphosphorylated tau protein, and neurofibrillary tangles involve in AD etiology. Meanwhile, enzymes and their inhibitors have become the focus of research in AD treatment. In this review, the molecular mechanisms involved in the pathogenesis of AD were overviewed and various enzymes such as acetylcholinesterase (AChE), butyrylcholinesterase (BuChE), β-secretase, γ-secretase, monoamine oxidase (MAO), and receptor of advanced glycation end products (RAGE) were highlighted as potential targets for AD treatment. Several hybrid molecules with essential substructures derived from various chemotypes have demonstrated desired pharmacological activity. It is envisioned that the development of new drugs that inhibit enzymes involved in AD is a future trend in the management of the disease.
Collapse
Affiliation(s)
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Dadashi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad-Reza Rashidi
- Medicinal Chemistry Department, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
89
|
Nong WJ, Tong XY, Ouyang JM. Comparison of Endoplasmic Reticulum Stress and Pyroptosis Induced by Pathogenic Calcium Oxalate Monohydrate and Physiologic Calcium Oxalate Dihydrate Crystals in HK-2 Cells: Insights into Kidney Stone Formation. Cells 2024; 13:2070. [PMID: 39768161 PMCID: PMC11674083 DOI: 10.3390/cells13242070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Endoplasmic reticulum stress (ERS) can activate pyroptosis through CHOP and TXNIP; however, the correlation between this process and the formation of kidney stones has not been reported. The purpose is to investigate the effects of calcium oxalate monohydrate (COM) and calcium oxalate dihydrate (COD) on ERS and pyroptosis in HK-2 cells and to explore the formation mechanism of calcium oxalate stones. HK-2 cells were injured by 3 μm COM and COD. COM and COD significantly upregulated the expression levels of GRP78, CHOP, TXNIP, and pyroptosis-related proteins (NLRP3, caspase-1, GSDMD-N, and IL-1β). Fluorescence colocalization revealed that COM induced pyroptosis by inducing the interaction between TXNIP and NLRP3. Both COM and COD crystals can induce ERS and pyroptosis in HK-2 cells. COM induces the interaction with NLRP3 by the upregulation of CHOP and TXNIP and then promotes pyroptosis, while COD only promotes pyroptosis by the upregulation of CHOP. The cytotoxicity and the ability of COM to promote crystal adhesion and aggregation are higher than COD, suggesting that COM is more dangerous for calcium oxalate kidney stone formation.
Collapse
Affiliation(s)
| | | | - Jian-Ming Ouyang
- Institute of Biomineralization and Lithiasis Research, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| |
Collapse
|
90
|
Wang S, Yin J, Liu Z, Liu X, Tian G, Xin X, Qin Y, Feng X. Metabolic disorders, inter-organ crosstalk, and inflammation in the progression of metabolic dysfunction-associated steatotic liver disease. Life Sci 2024; 359:123211. [PMID: 39491769 DOI: 10.1016/j.lfs.2024.123211] [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: 07/01/2024] [Revised: 08/20/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) represents a global health concern, affecting over 30 % of adults. It is a principal driver in the development of cirrhosis and hepatocellular carcinoma. The complex pathogenesis of MASLD involves an excessive accumulation of lipids, subsequently disrupting lipid metabolism and prompting inflammation within the liver. This review synthesizes the recent research progress in understanding the mechanisms contributing to MASLD progression, with particular emphasis on metabolic disorders and interorgan crosstalk. We highlight the molecular mechanisms linked to these factors and explore their potential as novel targets for pharmacological intervention. The insights gleaned from this article have important implications for both the prevention and therapeutic management of MASLD.
Collapse
Affiliation(s)
- Shendong Wang
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Junhao Yin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Zhaojun Liu
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Xin Liu
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Ge Tian
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271000, China
| | - Xijian Xin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Yiming Qin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Xiujing Feng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China.
| |
Collapse
|
91
|
Li Y, Wang L, Yi Q, Luo L, Xiong Y. Regulation of bile acids and their receptor FXR in metabolic diseases. Front Nutr 2024; 11:1447878. [PMID: 39726876 PMCID: PMC11669848 DOI: 10.3389/fnut.2024.1447878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 11/13/2024] [Indexed: 12/28/2024] Open
Abstract
High sugar, high-fat diets and unhealthy lifestyles have led to an epidemic of obesity and obesity-related metabolic diseases, seriously placing a huge burden on socio-economic development. A deeper understanding and elucidation of the specific molecular biological mechanisms underlying the onset and development of obesity has become a key to the treatment of metabolic diseases. Recent studies have shown that the changes of bile acid composition are closely linked to the development of metabolic diseases. Bile acids can not only emulsify lipids in the intestine and promote lipid absorption, but also act as signaling molecules that play an indispensable role in regulating bile acid homeostasis, energy expenditure, glucose and lipid metabolism, immunity. Disorders of bile acid metabolism are therefore important risk factors for metabolic diseases. The farnesol X receptor, a member of the nuclear receptor family, is abundantly expressed in liver and intestinal tissues. Bile acids act as endogenous ligands for the farnesol X receptor, and erroneous FXR signaling triggered by bile acid dysregulation contributes to metabolic diseases, including obesity, non-alcoholic fatty liver disease and diabetes. Activation of FXR signaling can reduce lipogenesis and inhibit gluconeogenesis to alleviate metabolic diseases. It has been found that intestinal FXR can regulate hepatic FXR in an organ-wide manner. The crosstalk between intestinal FXR and hepatic FXR provides a new idea for the treatment of metabolic diseases. This review focuses on the relationship between bile acids and metabolic diseases and the current research progress to provide a theoretical basis for further research and clinical applications.
Collapse
Affiliation(s)
| | | | | | | | - Yuxia Xiong
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
92
|
Zha Z, Xiao D, Liu Z, Peng F, Shang X, Sun Z, Liu Y, Chen W. Endoplasmic Reticulum Stress Induces ROS Production and Activates NLRP3 Inflammasome Via the PERK-CHOP Signaling Pathway in Dry Eye Disease. Invest Ophthalmol Vis Sci 2024; 65:34. [PMID: 39699913 DOI: 10.1167/iovs.65.14.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024] Open
Abstract
Purpose The purpose of this study was to investigate the potential roles of endoplasmic reticulum (ER) stress in the development of dry eye disease (DED). Methods Single-cell RNA sequencing (scRNA-seq) data from the Gene Expression Omnibus (GEO) database, derived from corneal tissues of a dry eye mouse model, was processed using the Seurat R program. The results were validated using a scopolamine-induced dry eye mouse model and a hyperosmotic-induced cell model involving primary human corneal epithelial cells (HCECs) and immortalized human corneal epithelial (HCE-2) cells. The HCE-2 cells were treated with 4-phenylbutyric acid (4-PBA) or tunicamycin (TM) to modulate ER stress. TXNIP and PERK knockdown were performed by siRNA transfection. Immunofluorescence, Western blotting, and real-time PCR were used to assess oxidative stress, ER stress, unfolded protein response (UPR) marker proteins, and TXNIP/NLRP3 axis activation. Results The analysis of scRNAseq data shows an increase in the ER stress marker GRP78, and the activation of the PERK-CHOP of UPR in DED mouse. These findings were confirmed both in vivo and in vitro. Additionally, HCE-2 cells treated with 4-PBA or TM showed significant effects on the production of reactive oxygen species (ROS) and the activation of the TXNIP/NLRP3-IL1β signaling pathway. Furthermore, siRNA knockdown of PERK or TXNIP, which alleviated the TXNIP/NLRP3-IL1β signaling axis, showed protective effects on HCECs. Conclusions This study explores the role of ER stress-induced oxidative stress and NLRP3-IL-1β mediated inflammation in DED, and highlights the therapeutic potential of PERK-CHOP axis and TXNIP in the treatment of DED.
Collapse
Affiliation(s)
- Zhiwei Zha
- The Affiliated Ningbo Eye Hospital of Wenzhou Medical University, Ningbo, Zhejiang, China
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Decheng Xiao
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zihao Liu
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fangli Peng
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xunjie Shang
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhenzhen Sun
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yang Liu
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wei Chen
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Ningbo Eye Institute, Ningbo Eye Hospital, Wenzhou Medical University, Ningbo, China
| |
Collapse
|
93
|
El-Mahdy NA, El-Masry TA, El-Tarahony AM, Alherz FA, Osman EY. Hepatoprotective Effect of Camel Thorn Polyphenols in Concanavalin A-Induced Hepatitis in Mice. Chin J Integr Med 2024; 30:1090-1100. [PMID: 39298072 DOI: 10.1007/s11655-024-3808-3] [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] [Accepted: 03/04/2024] [Indexed: 09/21/2024]
Abstract
OBJECTIVES To explore the prophylactic and therapeutic effects of Alhagi maurorum ethanolic extract (AME) in concanavalin A (Con A)-induced hepatitis (CIH) as well as possible underlying mechanisms. METHODS Polyphenols in AME were characterized using high performance liquid chromatography (HPLC). Swiss albino mice were divided into 4 groups. Normal group received intravenous phosphate-buffered saline (PBS); Con A group received 40 mg/kg intravenous Con A. Prophylaxis group administered 300 mg/(kg·d) AME orally for 5 days before Con A intervention. Treatment group received intravenous Con A then administered 300 mg/kg AME at 30 min and 3 h after Con A intervention. After 24 h of Con A injection, hepatic injury, oxidative stress, and inflammatory mediators were assessed. Histopathological examination and markers of apoptosis, inflammation, and CD4+ cell infiltration were also investigated. RESULTS HPLC analysis revealed that AME contains abundant polyphenols with pharmacological constituents, such as ellagic acid, gallic acid, ferulic acid, methylgallate, and naringenin. AME alleviated Con A-induced hepatic injury, as manifested by a significant reduction in alanine aminotransferase, aspartate aminotransferase and alkaline phosphatase (P<0.01). Additionally, the antioxidant effect of AME was revealed by a significant reduction in oxidative stress markers (nitric oxide and malondialdehyde) and restored glutathione (P<0.01). The levels of proinflammatory cytokines (tumor necrosis factor-α, interferon-γ, and interleukin-6) and c-Jun N-terminal kinase (JNK) activity were reduced (P<0.01). Histopathological examination of liver tissue showed that AME significantly ameliorated necrotic and inflammatory lesions induced by Con A (P<0.01). Moreover, AME reduced the expression of nuclear factor kappa B, pro-apoptotic protein (Bax), caspase-3, and CD4+ T cell hepatic infiltration (P<0.01). The expression of anti-apoptotic protein Bcl-2 was increased (P<0.01). CONCLUSION AME has hepatoprotective and ameliorative effects in CIH mice. These beneficial effects are likely due to the anti-inflammatory, antioxidant, and anti-apoptotic effects of the clinically important polyphenolic content. AME could be a novel and promising hepatoprotective agent for managing immune-mediated hepatitis.
Collapse
Affiliation(s)
- Nageh Ahmed El-Mahdy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Thanaa Ahmed El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Ahmed Mahmoud El-Tarahony
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Fatemah A Alherz
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia
| | - Enass Youssef Osman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
94
|
Zhou Q, Meng Y, Le J, Sun Y, Dian Y, Yao L, Xiong Y, Zeng F, Chen X, Deng G. Ferroptosis: mechanisms and therapeutic targets. MedComm (Beijing) 2024; 5:e70010. [PMID: 39568772 PMCID: PMC11577302 DOI: 10.1002/mco2.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
Ferroptosis is a nonapoptotic form of cell death characterized by iron-dependent lipid peroxidation in membrane phospholipids. Since its identification in 2012, extensive research has unveiled its involvement in the pathophysiology of numerous diseases, including cancers, neurodegenerative disorders, organ injuries, infectious diseases, autoimmune conditions, metabolic disorders, and skin diseases. Oxidizable lipids, overload iron, and compromised antioxidant systems are known as critical prerequisites for driving overwhelming lipid peroxidation, ultimately leading to plasma membrane rupture and ferroptotic cell death. However, the precise regulatory networks governing ferroptosis and ferroptosis-targeted therapy in these diseases remain largely undefined, hindering the development of pharmacological agonists and antagonists. In this review, we first elucidate core mechanisms of ferroptosis and summarize its epigenetic modifications (e.g., histone modifications, DNA methylation, noncoding RNAs, and N6-methyladenosine modification) and nonepigenetic modifications (e.g., genetic mutations, transcriptional regulation, and posttranslational modifications). We then discuss the association between ferroptosis and disease pathogenesis and explore therapeutic approaches for targeting ferroptosis. We also introduce potential clinical monitoring strategies for ferroptosis. Finally, we put forward several unresolved issues in which progress is needed to better understand ferroptosis. We hope this review will offer promise for the clinical application of ferroptosis-targeted therapies in the context of human health and disease.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yu Meng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Jiayuan Le
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yating Dian
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Lei Yao
- Department of General Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yixiao Xiong
- Department of Dermatology Tongji Hospital Huazhong University of Science and Technology Wuhan Hubei China
| | - Furong Zeng
- Department of Oncology Xiangya Hospital Central South University Changsha Hunan Province China
| | - Xiang Chen
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Guangtong Deng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| |
Collapse
|
95
|
Zhang K, Tian L, Sun Q, Lv J, Ding R, Yu Y, Li Y, Duan J. Constructing an adverse outcome pathway framework for the impact of maternal exposure to PM 2.5 on liver development and injury in offspring. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 112:104585. [PMID: 39489199 DOI: 10.1016/j.etap.2024.104585] [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: 08/06/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
Ambient fine particulate matter (PM2.5) is a significant contributor to air pollution. PM2.5 exposure poses a substantial hazard to public health. In recent years, the adverse effects of maternal PM2.5 exposure on fetal health have gradually gained public attention. As the largest organ in the body, the liver has many metabolic and secretory functions. Liver development, as well as factors that interfere with its growth and function, are of concern. This review utilized the adverse outcome pathway (AOP) framework as the analytical approach to demonstrate the link between maternal PM2.5 exposure and potential neonatal liver injury from the molecular to the population level. The excessive generation of reactive oxygen species (ROS), subsequent endoplasmic reticulum (ER) stress, and oxidative stress were regarded as the essential components in this framework, as they could trigger adverse developmental outcomes in the offspring through DNA damage, autophagy dysfunction, mitochondrial injury, and other pathways. To the best of our knowledge, this is the first article based on an AOP framework that elaborates on the influence of maternal exposure to PM2.5 on liver injury occurrence and adverse effects on liver development in offspring. Therefore, this review offered mechanistic insights into the developmental toxicity of PM2.5 in the liver, which provided a valuable basis for future studies and prevention strategies.
Collapse
Affiliation(s)
- Kexin Zhang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Li Tian
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Qinglin Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Jianong Lv
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Ruiyang Ding
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Yang Yu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Yang Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China.
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
96
|
Chae JB, Rim JM, Han SW, Cho YK, Kang JG, Chae JS. Prevalence, Isolation, and Molecular Characterization of Severe Fever with Thrombocytopenia Syndrome Virus in Cattle from the Republic of Korea. Vector Borne Zoonotic Dis 2024; 24:826-834. [PMID: 39029504 DOI: 10.1089/vbz.2024.0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2024] Open
Abstract
Background: Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne disease caused by Bandavirus dabieense. Initially identified in China, this disease has spread throughout Asian countries via tick bites and animal-to-human transmission. However, reports of the prevalence of SFTS virus (SFTSV) in cattle in Korea are lacking. This study aimed to investigate SFTSV infections in grazing cattle in the Republic of Korea (ROK). Materials and Methods: In total, 845 grazing cattle serum samples were collected over 2 years (2019 and 2020) in the ROK, and viral RNA was extracted using a kit. One-step RT-nested PCR was performed to amplify the S-segment of SFTSV. Positive serum samples were used to isolate SFTSV in Vero E6 cells, and the full sequences were analyzed. A phylogenetic tree was constructed using the maximum-likelihood method with MEGA X. In addition, immunoglobulin G antibodies against SFTSV were investigated using an enzyme-linked immunosorbent assay. Results: Here, 4.0% of serum samples (34/845) were positive for SFTSV S-segments, and one virus isolate was cultured in Vero E6 cells. Phylogenetic analysis based on the partial S-segment classified 4 SFTSV isolates as the B-2 genotype, 9 as the B-3 genotype, 18 as an unclassified B genotype, and 3 as the D genotype. One cultured virus was classified as the B-2 genotype based on SFTSV L-, M-, and S-segments. Antibody detection results showed that 21.1% of serum samples (161/763) were positive for SFTSV. Conclusion: To the best of our knowledge, this is the first study performed to identify the prevalence of SFTSV in grazing cattle in the ROK. Our findings indicate the necessity for more intensive and continuous SFTSV monitoring, not only in cattle but also in other animals, to comprehend the genetic diversity of the virus and its potential eco-epidemiological impact on human health.
Collapse
Affiliation(s)
- Jeong-Byoung Chae
- Laboratory of Veterinary Internal Medicine, BK21 FOUR Future Veterinary Medicine Leading Education and Research Centre, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Ji-Min Rim
- Laboratory of Veterinary Internal Medicine, BK21 FOUR Future Veterinary Medicine Leading Education and Research Centre, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Sun-Woo Han
- Laboratory of Veterinary Internal Medicine, BK21 FOUR Future Veterinary Medicine Leading Education and Research Centre, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yoon-Kyoung Cho
- Laboratory of Veterinary Internal Medicine, BK21 FOUR Future Veterinary Medicine Leading Education and Research Centre, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jun-Gu Kang
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, Republic of Korea
| | - Joon-Seok Chae
- Laboratory of Veterinary Internal Medicine, BK21 FOUR Future Veterinary Medicine Leading Education and Research Centre, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
97
|
Vijayakumar S, González-Sánchez ZI, Divya M, Amanullah M, Durán-Lara EF, Li M. Efficacy of chondroitin sulfate as an emerging biomaterial for cancer-targeted drug delivery: A short review. Int J Biol Macromol 2024; 283:137704. [PMID: 39549800 DOI: 10.1016/j.ijbiomac.2024.137704] [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: 08/25/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
The global increase in cancer incidence over the past decade highlights the urgent need for more effective therapeutic strategies. Conventional cancer treatments face challenges such as drug resistance and off-target toxicity, which affect healthy tissues. Chondroitin sulfate (CHDS), a naturally occurring bioactive macromolecule, has gained attention because of its biocompatibility, biodegradability, and low toxicity, positioning it as an ideal candidate for cancer-targeted drug delivery systems. This review highlights the potential of CHDS as an emerging biomaterial in cancer therapy, focusing on its unique biological properties and applications in drug delivery platforms. Furthermore, we discuss the advantages of CHDS-based biomaterials in enhancing cancer treatment efficacy and minimizing side effects, in order to provide a comprehensive reference for future research on CHDS-based cancer therapeutics.
Collapse
Affiliation(s)
- Sekar Vijayakumar
- College of Material Science and Engineering, Huaqiao University, Engineering Research Center of Environment-Friendly Functional Materials, Ministry of Education, Xiamen 361021, PR China.
| | - Zaira I González-Sánchez
- Nanobiology Laboratory, Department of Natural and Exact Sciences, Pontificia Universidad Católica Madre y Maestra, PUCMM, Autopista Duarte Km 1 ½, Santiago de los Caballeros, Dominican Republic; Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Mani Divya
- Advanced Laboratory of Bio-nanomaterials, BioMe Live Analytical Centre, Kannappa Tower, College Road, Karaikudi - 630 003, Tamilnadu, India
| | - Mohammed Amanullah
- Department of clinical Biochemistry, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Esteban F Durán-Lara
- Bio&NanoMaterialsLab Drug Delivery and Controlled Release, Universidad de Talca, Talca 3460000, Maule, Chile; Departamento de Microbiología, Facultad de Ciencias de la Salud, Universidad de Talca, Talca 3460000, Maule, Chile
| | - Mingchun Li
- College of Material Science and Engineering, Huaqiao University, Engineering Research Center of Environment-Friendly Functional Materials, Ministry of Education, Xiamen 361021, PR China.
| |
Collapse
|
98
|
Jin H, Ma J, Xu B, Xu S, Hu T, Jin X, Wang J, Wang G, Zhen L. Thio-ProTide strategy: A novel H 2S donor-drug conjugate (DDC) alleviates hepatic injury via innate lysosomal targeting. Acta Pharm Sin B 2024; 14:5341-5356. [PMID: 39807337 PMCID: PMC11725097 DOI: 10.1016/j.apsb.2024.10.017] [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: 07/19/2024] [Revised: 10/17/2024] [Accepted: 10/25/2024] [Indexed: 01/16/2025] Open
Abstract
Hydrogen sulfide (H2S) is a gas signaling molecule with versatile bioactivities; however, its exploitation for disease treatment appears challenging. This study describes the design and characterization of a novel type of H2S donor-drug conjugate (DDC) based on the thio-ProTide scaffold, an evolution of the ProTide strategy successfully used in drug discovery. The new H2S DDCs achieved hepatic co-delivery of H2S and an anti-fibrotic drug candidate named hydronidone, which synergistically attenuated liver injury and resulted in more sufficient intracellular drug exposure. The potent hepatoprotective effects were also attributed to the H2S-mediated multipronged intervention in lipid peroxidation both at the whole cellular and lysosomal levels. Lysosomal H2S accumulation and H2S DDC activation were facilitated by the hydrolysis through the specific lysosomal hydrolase, representing a distinct mechanism for lysosomal targeting independent of the classical basic moieties. These findings provided a novel pattern for the design of optimally therapeutic H2S DDC and organelle-targeting functional molecules.
Collapse
Affiliation(s)
- Haowen Jin
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Jie Ma
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Bixin Xu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Sitao Xu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Tianyu Hu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Xin Jin
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Jiankun Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Le Zhen
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
99
|
Yuan Y, Li J, Chen M, Zhao Y, Zhang B, Chen X, Zhao J, Liang H, Chen Q. Nano-encapsulation of drugs to target hepatic stellate cells: Toward precision treatments of liver fibrosis. J Control Release 2024; 376:318-336. [PMID: 39413846 DOI: 10.1016/j.jconrel.2024.10.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024]
Abstract
Liver fibrosis is characterized by excessive extracellular matrix (ECM) deposition triggered by hepatic stellate cells (HSCs). As central players in fibrosis progression, HSCs are the most important therapeutic targets for antifibrotic therapy. However, owing to the limitations of systemic drug administration, there is still no suitable and effective clinical treatment. In recent years, nanosystems have demonstrated expansive therapeutic potential and evolved into a clinical modality. In liver fibrosis, nanosystems have undergone a paradigm shift from targeting the whole liver to locally targeted modifying processes. Nanomedicine delivered to HSCs has significant potential in managing liver fibrosis, where optimal management would benefit from targeted delivery, personalized therapy based on the specific site of interest, and minor side effects. In this review, we present a brief overview of the role of HSCs in the pathogenesis of liver fibrosis, summarize the different types of nanocarriers and their specific delivery applications in liver fibrosis, and highlight the biological barriers associated with the use of nanosystems to target HSCs and approaches available to solve this issue. We further discuss in-depth all the molecular target receptors overexpressed during HSC activation in liver fibrosis and their corresponding ligands that have been used for drug or gene delivery targeting HSCs.
Collapse
Affiliation(s)
- Yue Yuan
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Jiaxuan Li
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Min Chen
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Ying Zhao
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
| | - Jianping Zhao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China.
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China.
| | - Qian Chen
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China.
| |
Collapse
|
100
|
Guo Q, Dong D, Qiao X, Huang S, Zhao Y. Hub genes, diagnostic model, and predicted drugs related to ferroptosis in chronic rhinosinusitis with nasal polyps. Medicine (Baltimore) 2024; 103:e40624. [PMID: 39612457 PMCID: PMC11608670 DOI: 10.1097/md.0000000000040624] [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: 06/07/2024] [Accepted: 11/04/2024] [Indexed: 12/01/2024] Open
Abstract
Significant progress has been made in the pathogenesis of chronic rhinosinusitis (CRS). However, the relationship between chronic rhinosinusitis with nasal polyps (CRSwNP) and ferroptosis, as well as its underlying molecular mechanism, remains unclear. This study aimed to investigate the correlation between CRSwNP and ferroptosis and identify key gene associated with ferroptosis that could impact the diagnosis and treatment of CRS. To achieve this, gene expression profiles containing CRSwNP and CRSsNP samples were obtained from the GEO database. In addition, from the FerrDb V2 database, we acquired 2 sets of genes that are connected with ferroptosis, giving us a combined number of 260 genes associated with this particular biological process. Differential analysis and weighted gene co-expression network analysis (WGCNA) were performed on nasal tissue samples from GSE36830, leading to the identification of 1 key gene related to ferroptosis and CRS. Using stepwise regression and logistic regression analysis, we constructed a diagnostic model for CRS using ALOX15. The AUC value demonstrates that the model exhibits a strong diagnostic performance. Furthermore, the connection between immune cell infiltration in the samples and hub gene was explored, suggesting the potential significance of the hub gene in the immune response to CRS. Finally, Five drugs targeting a central gene were identified from the DrugBank database, and a few of them have exhibited efficacy in the treatment of CRS or associated ailments. In conclusion, this model holds potential for supporting the diagnosis of CRS patients, while the central gene identified may contribute to a better understanding of CRS development and drug treatment.
Collapse
Affiliation(s)
- Qian Guo
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Otolaryngology Diseases, Henan Provincial Health Commission, Changsha, China
| | - Dong Dong
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Otolaryngology Diseases, Henan Provincial Health Commission, Changsha, China
| | - Xinjie Qiao
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Otolaryngology Diseases, Henan Provincial Health Commission, Changsha, China
| | - Shuman Huang
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Otolaryngology Diseases, Henan Provincial Health Commission, Changsha, China
| | - Yulin Zhao
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Key Laboratory of Otolaryngology Diseases, Henan Provincial Health Commission, Changsha, China
| |
Collapse
|