1
|
Ma Q, Huang S, Li MY, Luo QH, Chen FM, Hong CL, Yan HH, Qiu J, Zhao KL, Du Y, Zhao JK, Zhou LQ, Lou DY, Efferth T, Li CY, Qiu P. Dihydromyricetin regulates the miR-155-5p/SIRT1/VDAC1 pathway to promote liver regeneration and improve alcohol-induced liver injury. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156522. [PMID: 39986231 DOI: 10.1016/j.phymed.2025.156522] [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: 05/31/2024] [Revised: 02/05/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Alcohol-related liver disease (ALD) has become an increasingly serious global health issue. In recent years, growing evidence has highlighted the restoration of liver regenerative capacity as an effective therapeutic strategy for improving ALD. Previous studies have demonstrated the protective effect of dihydromyricetin (DMY) in alcohol-induced liver injury, but its pharmacological role in ALD-related liver regeneration impairment remains poorly understood. OBJECTIVE This study aims to explore the therapeutic potential and molecular mechanisms of DMY in the context of liver regeneration impairment in ALD. METHODS The classic Lieber-DeCarli alcohol liquid diet was used to establish an ALD model in vivo. DMY (75 and 150 mg/kg/day) and silybin (200 mg/kg) were administered for 7 weeks to assess the hepatoprotective effects of DMY. First, biochemical markers and liver histopathology were used to evaluate liver inflammation and steatosis in ALD mice. Second, we explored the potential molecular mechanisms by which DMY improves ALD through serum untargeted metabolomics, hepatic transcriptomics, and single-cell sequencing data. Furthermore, in vivo and in vitro experiments, combined with Western blotting, dual-luciferase reporter assays, and immunofluorescence, were conducted to elucidate the protective mechanisms underlying DMY's effects on ALD. RESULTS In vivo studies showed that DMY significantly ameliorated ALT/AST abnormalities, liver inflammation, and steatosis in ALD mice. Multi-omics and bioinformatics analyses revealed that DMY may exert its anti-ALD effects by regulating the miR-155-5p/SIRT1/VDAC1 pathway, thereby mitigating cellular senescence. Notably, knockdown of miR-155 provided partial protection against ethanol-induced liver damage. Additionally, clinical ALD samples and in vivo and in vitro experiments further confirmed that excessive alcohol exposure induces the production of miR-155-5p in liver Kupffer cells. miR-155-5p targets and inhibits SIRT1, promoting the expression of mitochondrial VDAC1, leading to mitochondrial DNA leakage, thereby accelerating hepatocyte senescence and inflammation. However, DMY improved the disruption of the miR-155-5p/SIRT1/VDAC1 pathway and hepatocyte senescence, thereby restoring liver regenerative function and exerting anti-ALD effects. CONCLUSION In this study, we provide the first evidence that DMY improves liver inflammation and cellular senescence by regulating the miR-155-5p/SIRT1/VDAC1 positive feedback loop, promoting liver regeneration to improve ALD. In summary, our work provides important research evidence and theoretical support for DMY as a promising candidate drug for the prevention and treatment of ALD.
Collapse
Affiliation(s)
- Qing Ma
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China; School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Shuo Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Mei-Ya Li
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qi-Han Luo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Fang-Ming Chen
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chun-Lan Hong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Hong-Hao Yan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jiang Qiu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Kang-Lu Zhao
- Zhejiang Rehabilitation Medical Center, Rehabilitation Hospital Area of the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou Zhejiang, China; The Fourth Affiliated Hospital Zhejiang University, School of Medicine, Yiwu Zhejiang, China
| | - Yu Du
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Jin-Kai Zhao
- Zhuji People's Hospital of Zhejiang Province, Shaoxing 311800, China
| | - Li-Qin Zhou
- Zhuji People's Hospital of Zhejiang Province, Shaoxing 311800, China
| | - Da-Yong Lou
- Zhuji People's Hospital of Zhejiang Province, Shaoxing 311800, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany.
| | - Chang-Yu Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, China; Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Ping Qiu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, China.
| |
Collapse
|
2
|
Li L, Liu Y, Wang K, Mo J, Weng Z, Jiang H, Jin C. Stem cell exosomes: new hope and future potential for relieving liver fibrosis. Clin Mol Hepatol 2025; 31:333-349. [PMID: 39510097 PMCID: PMC12016649 DOI: 10.3350/cmh.2024.0854] [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: 09/29/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/15/2024] Open
Abstract
Liver fibrosis is a chronic liver injury resulting from factors like viral hepatitis, autoimmune hepatitis, non-alcoholic steatohepatitis, fatty liver disease, and cholestatic liver disease. Liver transplantation is currently the gold standard for treating severe liver diseases. However, it is limited by a shortage of donor organs and the necessity for lifelong immunosuppressive therapy. Mesenchymal stem cells (MSCs) can differentiate into various liver cells and enhance liver function when transplanted into patients due to their differentiation and proliferation capabilities. Therefore, it can be used as an alternative therapy for treating liver diseases, especially for liver cirrhosis, liver failure, and liver transplant complications. However, due to the potential tumorigenic effects of MSCs, researchers are exploring a new approach to treating liver fibrosis using extracellular vesicles (exosomes) secreted by stem cells. Many studies show that exosomes released by stem cells can promote liver injury repair through various pathways, contributing to the treatment of liver fibrosis. In this review, we focus on the molecular mechanisms by which stem cell exosomes affect liver fibrosis through different pathways and their potential therapeutic targets. Additionally, we discuss the advantages of exosome therapy over stem cell therapy and the possible future directions of exosome research, including the prospects for clinical applications and the challenges to be overcome.
Collapse
Affiliation(s)
- Lihua Li
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Yongjie Liu
- Department of Cell biology, School of Medicine, Taizhou University, Taizhou, Zhejiang Province, P. R. China
- Department of Pathophysiology, School of Basic Medicine, Shenyang Medical College, Shenyang, Liaoning Province, P. R. China
| | - Kunpeng Wang
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Jinggang Mo
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Zhiyong Weng
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Hao Jiang
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Chong Jin
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| |
Collapse
|
3
|
Li X, Li W, Xie X, Fang T, Yang J, Shen Y, Wang Y, Wang H, Tao L, Zhang H. ROS Regulate Rotenone-induced SH-SY5Y Dopamine Neuron Death Through Ferroptosis-mediated Autophagy and Apoptosis. Mol Neurobiol 2025:10.1007/s12035-025-04824-6. [PMID: 40097764 DOI: 10.1007/s12035-025-04824-6] [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: 09/04/2024] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
Rotenone, a plant-derived natural insecticide, is widely used to induce Parkinson's disease (PD) models. However, the mechanisms of rotenone-induced cell death remain unclear. Here, we found that rotenone (0.01, 0.1, or 1 μmol/L) suppressed SH-SY5Y dopamine neuron viability and led to PD-like pathological changes, such as reduced tyrosine hydroxylase (TH) but increased α-synuclein. Rotenone increased the levels of intracellular reactive oxygen species (ROS) and mitochondrial ROS, as well as the levels of the antioxidants nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), ultimately resulting in oxidative stress. Moreover, rotenone significantly downregulated the expression of GPX4 and xCT but upregulated the expression of COX2 and NCOA4, which are markers of ferroptosis. Furthermore, rotenone decreased phosphorylated mTOR level but increased Beclin-1, ATG5, LC3 and p62 expression, suggesting that rotenone enhances autophagy and reduces autophagy flux. Additionally, rotenone reduced Bcl-2 levels and the mitochondrial membrane potential (MMP) while promoting BAX and Caspase-3 expression, thus initiating cell apoptosis. N-acetylcysteine (NAC), a ROS scavenger, and ferrostatin-1 (Fer-1) and deferoxamine (DFO), two ferroptosis inhibitors, significantly eliminated rotenone-induced autophagy and apoptosis. Moreover, ML385, a specific inhibitor of Nrf2, suppressed rotenone-induced ferroptosis. Our results demonstrated that ROS might mediate rotenone-induced PD-like pathological changes by regulating iron death, autophagy, and apoptosis. Inhibiting ferroptosis blocked the rotenone-induced increase in autophagy and apoptosis. Thus, the ability of ROS to regulate rotenone-induced death through autophagy and apoptosis is dependent on ferroptosis. The findings require validation in multiple neuronal cell lines and in vivo.
Collapse
Affiliation(s)
- Xinying Li
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Weiran Li
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
- Department of Clinical Medicine, School of Medicine, Qinghai University, Xining, China
| | - Xinying Xie
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Ting Fang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Jingwen Yang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Yue Shen
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Yicheng Wang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Hongyan Wang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Liqing Tao
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Heng Zhang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China.
| |
Collapse
|
4
|
Zhu J, Jin Z, Wang J, Wu Z, Xu T, Tong G, Shen E, Fan J, Jiang C, Wang J, Li X, Cong W, Lin L. FGF21 ameliorates septic liver injury by restraining proinflammatory macrophages activation through the autophagy/HIF-1α axis. J Adv Res 2025; 69:477-494. [PMID: 38599281 PMCID: PMC11954821 DOI: 10.1016/j.jare.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/26/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024] Open
Abstract
INTRODUCTION Sepsis, a systemic immune syndrome caused by severe trauma or infection, poses a substantial threat to the health of patients worldwide. The progression of sepsis is heavily influenced by septic liver injury, which is triggered by infection and cytokine storms, and has a significant impact on the tolerance and prognosis of septic patients. The objective of our study is to elucidate the biological role and molecular mechanism of fibroblast growth factor 21 (FGF21) in the process of sepsis. OBJECTIVES This study was undertaken in an attempt to elucidate the function and molecular mechanism of FGF21 in therapy of sepsis. METHODS Serum concentrations of FGF21 were measured in sepsis patients and septic mice. Liver injury was compared between mice FGF21 knockout (KO) mice and wildtype (WT) mice. To assess the therapeutic potential, recombinant human FGF21 was administered to septic mice. Furthermore, the molecular mechanism of FGF21 was investigated in mice with myeloid-cell specific HIF-1α overexpression mice (LyzM-CreDIO-HIF-1α) and myeloid-cell specific Atg7 knockout mice (Atg7△mye). RESULTS Serum level of FGF21 was significantly increased in sepsis patients and septic mice. Through the use of recombinant human FGF21 (rhFGF21) and FGF21 KO mice, we found that FGF21 mitigated septic liver injury by inhibiting the initiation and propagation of inflammation. Treatment with rhFGF21 effectively suppressed the activation of proinflammatory macrophages by promoting macroautophagy/autophagy degradation of hypoxia-inducible factor-1α (HIF-1α). Importantly, the therapeutic effect of rhFGF21 against septic liver injury was nullified in LyzM-CreDIO-HIF-1α mice and Atg7△mye mice. CONCLUSIONS Our findings demonstrate that FGF21 considerably suppresses inflammation upon septic liver injury through the autophagy/ HIF-1α axis.
Collapse
Affiliation(s)
- Junjie Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Zhouxiang Jin
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Jie Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Zhaohang Wu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Tianpeng Xu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Gaozan Tong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China
| | - Enzhao Shen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China
| | - Junfu Fan
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Chunhui Jiang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Jiaqi Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Xiaokun Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China; Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Weitao Cong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China; Haihe Laboratory of Cell Ecosystem, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China
| | - Li Lin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000, PR China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, PR China.
| |
Collapse
|
5
|
Barberi L, Porcu C, Boccia C, Cosentino M, Nicoletti C, Peruzzi B, Iosi F, Forconi F, Bagnato G, Dobrowolny G, Di Cola S, Lapenna L, Cera G, Merli M, Musarò A. Circulating Extracellular Vesicles in Alcoholic Liver Disease Affect Skeletal Muscle Homeostasis and Differentiation. J Cachexia Sarcopenia Muscle 2025; 16:e13675. [PMID: 39921321 PMCID: PMC11806195 DOI: 10.1002/jcsm.13675] [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: 03/19/2024] [Revised: 09/13/2024] [Accepted: 11/16/2024] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND The mechanisms underlying muscle alteration associated to alcoholic liver disease (ALD) are not fully understood and the physiopathologic mediators of the liver-muscle interplay remains elusive. We investigated the role of circulating extracellular vesicles (EVs) in ALD as potential mediators of muscle atrophy. METHODS We established a mouse model of sarcopenia associated to ALD, by feeding mice with an alcoholic diet for 8 weeks. We investigated the effects of hepatic and circulating EVs isolated from these mice (EtOH mice; n = 7 females) on muscle cell cultures, comparing them with EVs from mice fed with a standard diet (CD mice; n = 6 females). Additionally, we examined the impact of circulating EVs from patients with alcohol-related cirrhosis (7 males and 2 females, mean age 55.4 years) on primary human muscle cells, comparing them with EVs from age-matched healthy subjects (6 males and 3 females). We analysed the miRNA profile of the EVs to identify potential mediators of ALD-associated sarcopenia. RESULTS We demonstrated that circulating EVs were internalized by muscle cells and that EVs from ALD mice and cirrhotic patients caused alteration in the myogenic program. Molecular analysis revealed that serum EVs from ALD mice reduced protein synthesis in C2C12 cells, decreasing levels of p-AKT/AKT (-54.6%; p < 0.05), p-mTOR/mTOR (-54.5%; p < 0.05) and p-GSK3(Ser9)/GSK3 (-30.63%). Similarly, hepatic EVs induced defects in muscle differentiation, with reduced levels of p-AKT/AKT (-39.1%; p < 0.05), p-mTOR/mTOR (-30.1%; p < 0.05) and p-GSK3(Ser9)/GSK3 (-40%). C2C12 cells treated with either serum or hepatic EtOH-EVs exhibited upregulated expression of muscle-specific atrophy markers Atrogin-1 (+61.2% and +189.5%, respectively; p < 0.05) and MuRF1 (+260.4% and +112.5%, respectively; p < 0.05), along with an increased LC3-II/-I ratio (+131.5% and +40.2%, respectively; p < 0.05), indicating enhanced autophagy. MiRNA analysis revealed that both circulating and hepatic EVs from ALD mice showed elevated expression of miR-21, miR-155, miR-223 and miR-122 (+230% and +292%, respectively; p < 0.01) suggesting their potential role in sarcopenia. Human muscle cells exposed to EVs from cirrhotic patients exhibited reduced protein synthesis and upregulated Atrogin-1 (+113%; p < 0.05) and MuRF1 (+86.3%; p < 0.05), indicating proteasome activation. Circulating EVs of alcoholic patients showed upregulation of the same miRNAs observed in EtOH mice, including the liver-specific miR-122 (+260%; p < 0.05) suggesting, also in human liver disease, a hepatic origin of circulating EVs. CONCLUSIONS Our study highlights the critical role of ALD-derived circulating EVs in affecting muscle homeostasis and myogenic program, suggesting potential therapeutic targets for mitigating muscle loss in ALD.
Collapse
Affiliation(s)
- Laura Barberi
- DAHFMO‐Unit of Histology and Medical EmbryologySapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia – Fondazione Cenci BolognettiRomeItaly
| | - Cristiana Porcu
- DAHFMO‐Unit of Histology and Medical EmbryologySapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia – Fondazione Cenci BolognettiRomeItaly
| | - Caterina Boccia
- DAHFMO‐Unit of Histology and Medical EmbryologySapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia – Fondazione Cenci BolognettiRomeItaly
| | - Marianna Cosentino
- DAHFMO‐Unit of Histology and Medical EmbryologySapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia – Fondazione Cenci BolognettiRomeItaly
| | - Carmine Nicoletti
- DAHFMO‐Unit of Histology and Medical EmbryologySapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia – Fondazione Cenci BolognettiRomeItaly
| | - Barbara Peruzzi
- Bone Pathophysiology Research UnitBambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Francesca Iosi
- Core Facilities, Microscopy AreaIstituto Superiore di SanitàRomeItaly
| | - Flavia Forconi
- DAHFMO‐Unit of Histology and Medical EmbryologySapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia – Fondazione Cenci BolognettiRomeItaly
| | - Giulia Bagnato
- DAHFMO‐Unit of Histology and Medical EmbryologySapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia – Fondazione Cenci BolognettiRomeItaly
- Bone Pathophysiology Research UnitBambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Gabriella Dobrowolny
- DAHFMO‐Unit of Histology and Medical EmbryologySapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia – Fondazione Cenci BolognettiRomeItaly
| | - Simone Di Cola
- Department of Translational and Precision MedicineSapienza University of RomeRomeItaly
| | - Lucia Lapenna
- Department of Translational and Precision MedicineSapienza University of RomeRomeItaly
| | - Gianluca Cera
- Department of Orthopaedics and TraumatologyPoliclinico Umberto IRomeItaly
| | - Manuela Merli
- Department of Translational and Precision MedicineSapienza University of RomeRomeItaly
| | - Antonio Musarò
- DAHFMO‐Unit of Histology and Medical EmbryologySapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia – Fondazione Cenci BolognettiRomeItaly
- Scuola Superiore di Studi Avanzati Sapienza (SSAS)Sapienza University of RomeRomeItaly
| |
Collapse
|
6
|
Liao S, Guo F, Xiao Z, Xiao H, Pan QR, Guo Y, Chen J, Wang X, Wang S, Huang H, Yang L, Liu HF, Pan Q. Autophagy activation within inflammatory microenvironment improved the therapeutic effect of MSC-Derived extracellular Vesicle in SLE. J Adv Res 2025:S2090-1232(25)00063-3. [PMID: 39880074 DOI: 10.1016/j.jare.2025.01.044] [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: 10/26/2024] [Revised: 01/06/2025] [Accepted: 01/25/2025] [Indexed: 01/31/2025] Open
Abstract
INTRODUCTION Developing strategies to improve the therapeutic efficacy of mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) in autoimmune diseases have garnered increased attention. OBJECTIVES To evaluate whether rapamycin-induced autophagy within the systemic lupus erythematosus (SLE) inflammatory microenvironment (Rapa-SLE) augments the therapeutic effects of MSC-derived EVs in SLE. METHODS The therapeutic potential of the resulting EVs (Rapa-SLE-EV) was assessed in MRL/lpr mice. Rapa-SLE-EVs were compared with EVs derived from MSCs from MSCs cultured with EV-depleted fetal bovine serum (FBS-EV), EVs from MSCs cultured with rapamycin-treated FBS (Rapa-FBS-EV), and EVs exposed to SLE serum without rapamycin (SLE-EV). The autoimmune response, renal function, and pathological damage were assessed among the mouse groups. Additionally, mechanistic investigations into the role of the anti-inflammatory protein IDO1 within the EVs. RESULTS Interaction with the SLE inflammatory microenvironment triggered autophagy in MSCs, which was further enhanced by rapamycin treatment. Rapa-SLE-EV administration significantly ameliorated the autoimmune response and renal damage in MRL/lpr mice, outperforming other MSC-EV groups. This treatment mitigated key manifestations of SLE, including reduced autoantibody levels, as well as splenomegaly, and lymphadenopathy. Furthermore, Rapa-SLE-EV demonstrated superior suppression of plasma inflammatory cytokines, preserved renal function, mitigated pathological damage, and reduced glomerular immune complex deposition. Mechanistically, Rapa-SLE-EV exhibits exceptional inhibitory effects on SLE-B cell function, benefited by the high expression of the anti-inflammatory protein IDO1, which was confirmed to enter SLE-B cells through EVs. CONCLUSIONS We developed a novel strategy to improve the therapeutic efficacy of MSC-EVs in SLE and confirmed that the immunomodulatory function of the MSC-EVs is enhanced through autophagic activation and interaction with the SLE serum microenvironment, a process likely benefited by the high expression of IDO1.
Collapse
Affiliation(s)
- Shuzhen Liao
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China; Yue Bei People's Hospital Postdoctoral Innovation Practice Base, Southern Medical University, Guangzhou 510515, China
| | - Fengbiao Guo
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Zengzhi Xiao
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Haiyan Xiao
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Quan-Ren Pan
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Yugan Guo
- Department of Radiation Oncology, Yuebei People's Hospital Affiliated to Shantou University School of Medicine, Shaoguan 512000, China
| | - Jiaxuan Chen
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Xi Wang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Shuting Wang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Haimin Huang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Lawei Yang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Hua-Feng Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China.
| | - Qingjun Pan
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Clinical Research and Experimental Center, Department of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China; Department of Clinical Laboratory, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University. Guangzhou 510120, China.
| |
Collapse
|
7
|
Zhang C, Qiao P, Xiao C, Cao Z, Chen J, Fang H, Yang J, Kang Z, Dang E, Shao S, Pang B, Li Q, Zhu Z, Shen S, Hasegawa A, Abe R, Qiao H, Wang G, Fu M. Exosomal miR-375-3p mediated lipid metabolism, ferritinophagy and CoQ-dependent pathway contributes to the ferroptosis of keratinocyte in SJS/TEN. Int J Biol Sci 2025; 21:1275-1293. [PMID: 39897035 PMCID: PMC11781181 DOI: 10.7150/ijbs.98592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 01/10/2025] [Indexed: 02/04/2025] Open
Abstract
Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) manifest life-threatening cutaneous adverse drug reactions characterized by keratinocyte death. Previous studies have indicated that apoptosis and necroptosis are implicated in SJS/TEN pathogeneses. However, other forms of cell death involved in this process remain unidentified. Ferroptosis, cell death driven by iron-dependent lipid peroxidation, has been implicated in various human diseases. In this study, the identification of ferroptosis and the potential effects of ferroptosis on SJS/TEN were investigated. We demonstrated that the skin lesions and plasma of SJS/TEN patients show increased levels of lipid peroxidation and iron. The biomarkers of ferroptosis correlated positively with the disease severity in SJS/TEN patients. Besides, plasma exosomes derived from patients with SJS/TEN exhibited elevated levels of cellular oxidized polyunsaturated fatty acids (PUFAs) and phospholipids phosphatidylethanolamine (PE), the key phospholipids that drive cells towards ferroptotic death. miR-375-3p, enriched in plasma-derived exosomes from SJS/TEN patients, was observed reduce both ferritin heavy chain 1 (FTH1) and ferroptosis suppressor protein 1 (FSP1) expression. Parallelly, exosomal miR-375-3p overexpression increased the level of lipid peroxidation but decreased the coenzyme Q10 (CoQ10), thus enhancing the ferroptosis rate of keratinocyte. Above all, we concluded that ferritinophagy-mediated ferroptosis, lipid metabolism, and the FSP1-CoQ-dependent pathway in ferroptosis are critical mechanisms contributing to SJS/TEN. Targeting ferroptosis in keratinocyte may be a therapeutic strategy for preventing SJS/TEN in the future.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Pei Qiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - ChunYing Xiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - ZiPeng Cao
- Department of Health Education and Management and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - JiaoLing Chen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Hui Fang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - JianKang Yang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - ZeHua Kang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - ErLe Dang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Shuai Shao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - BingYu Pang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - QingYang Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - ZhenLai Zhu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - ShengXian Shen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Akito Hasegawa
- Division of Dermatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - Riichiro Abe
- Division of Dermatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8510, Japan
| | - HongJiang Qiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Meng Fu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
8
|
Gripshover TC, Treves RS, Rouchka EC, Chariker JH, Zheng S, Hudson E, Smith ML, Singal AK, McClain CJ, Hardesty JE. Visium spatial transcriptomics and proteomics identifies novel hepatic cell populations and transcriptomic signatures of alcohol-associated hepatitis. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025; 49:106-116. [PMID: 39592394 PMCID: PMC11747835 DOI: 10.1111/acer.15494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Alcohol-associated hepatitis (AH) is the clinical manifestation of alcohol-associated liver disease (ALD). AH is a complex disease encompassing the dysregulation of many cells and cell subpopulations. This study used a hepatic spatial transcriptomic and proteomic approach (10X Genomics Visium) to identify hepatic cell populations and their associated transcriptomic and proteomic alterations in human AH. METHODS Formalin-fixed paraffin-embedded liver tissue from AH patients (n = 2) and non-ALD controls (donors) (n = 2) were used for Visium spatial transcriptomic and proteomic analysis. RESULTS AH cell clusters and cell markers were drastically different in regard to tissue pattern and number of cell types compared to non-ALD controls. Cholangiocytes, endothelial cells, macrophages, and stellate cells were more profuse in AH relative to non-ALD controls. Transcriptionally, proliferating cell nuclear antigen-positive (PCNA+) hepatocytes in AH more closely resembled cholangiocytes suggesting they were non-functional hepatocytes derived from cholangiocytes. Furthermore, mitochondria protein-coding genes were reduced in AH versus non-ALD control hepatocytes, suggesting reduced functionality and loss of regenerative mechanisms. Macrophages in AH exhibited elevated gene expression involved in exosomes as compared to non-ALD controls. The most upregulated macrophage genes observed in AH were those involved in exosome trafficking. Gene and protein signatures of disease-associated hepatocytes (ANXA2+/CXCL1+/CEACAM8+) were elevated in AH and could visually identify a pre-malignant lesion. CONCLUSIONS This study identified global cell type alterations in AH and distinct transcriptomic changes between AH and non-ALD controls. These findings characterize cellular plasticity and profuse transcriptomic and proteomic changes that are apparent in AH and contribute to the identification of novel therapeutics.
Collapse
Affiliation(s)
- Tyler C. Gripshover
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Rui S. Treves
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Eric C. Rouchka
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
- Kentucky INBRE Bioinformatics Core, University of Louisville School of Medicine, Louisville, KY, USA
| | - Julia H. Chariker
- Kentucky INBRE Bioinformatics Core, University of Louisville School of Medicine, Louisville, KY, USA
| | - Shirong Zheng
- Sequencing Technology Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - Elizabeth Hudson
- Sequencing Technology Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - Melissa L. Smith
- Department of Biochemistry & Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
- Sequencing Technology Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - Ashwani K. Singal
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, USA
- Robley Rex Veterans Medical Center, Louisville, KY, USA
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - Craig J. McClain
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, USA
- Robley Rex Veterans Medical Center, Louisville, KY, USA
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA
- University of Louisville Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - Josiah E. Hardesty
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, USA
| |
Collapse
|
9
|
Jin Y, Wang C, Meng Z, Zhang Y, Meng D, Liu J, Yuan M, Guan S. Proanthocyanidins alleviate acute alcohol liver injury by inhibiting pyroptosis via inhibiting the ROS-MLKL-CTSB-NLRP3 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156268. [PMID: 39612889 DOI: 10.1016/j.phymed.2024.156268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/04/2024] [Accepted: 11/15/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Alcoholic Liver Disease (ALD) is a hepatic disorder resulting from prolonged or excessive alcohol intake. The predominant manifestation of ALD is fatty liver, which progresses to alcoholic hepatitis as the disease worsens. Pyroptosis is a novel type of programmed cell death that is intricately linked to the inflammatory cascade, presenting a promising avenue for therapeutic intervention in the management of ALD. Oligomeric proanthocyanidins (OPCs) are polyphenols extracted from grape seeds that have anti-inflammatory and antioxidant properties. However, whether OPCs can treat ALD by suppressing pyroptosis is not completely clarified. PURPOSE To explore the role of OPCs in ALD to inhibit pyroptosis and its mechanism. METHODS In vitro, HepG2 cells were employed to evaluate the beneficial impact of OPCs on alcohol-induced pyroptosis. MTT colorimetric method, enzyme-linked immunosorbent assay (ELISA), western blot (WB), immunofluorescence, acridine orange (AO) staining, and reactive oxygen species (ROS) assay were performed. In vivo, C57BL mice were used and gavaged with alcohol and OPCs. Hematoxylin-eosin staining (HE) staining, alanine aminotransferase (ALT), aspartate aminotransferase (AST) level assay, and WB were performed. RESULTS The findings revealed that OPCs could reduce the alcohol-induced increase in pyroptosis-related proteins, such as pyrin domain-containing 3 protein (NLRP3), cleaved-caspase 1, gasdermin D (GSDMD-N), Interleukin-18 (IL-18), IL-1β (IL-1β). In in vitro mechanistic experiments, We discovered that OPCs ameliorate alcohol-induced pyroptosis by decreasing cathepsin B (CTSB) leakage-mediated NLRP3 activation. More significantly, we discovered that alcohol phosphorylates mixed lineage kinase domain-like protein (MLKL), enabling P-MLKL to translocate to the lysosomal membrane and induce lysosomal membrane permeabilization (LMP). OPCs might counteract the effects of alcohol by reducing the leakage of CTSB and inhibiting the phosphorylation of MLKL through the scavenging of ROS. CONCLUSIONS These results suggested that OPCs might counteract ALD by inhibiting pyroptosis through the ROS-MLKL-CTSB-NLRP3 pathway. Our study offered fresh insight into the ways in which naturally occurring chemicals shield ALD against harm.
Collapse
Affiliation(s)
- Yingli Jin
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, PR China
| | - Chunyun Wang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, PR China
| | - Zhuoqun Meng
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, PR China
| | - Yuxin Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, PR China
| | - Desen Meng
- The First Norman Bethune Clinical Medical College, Jilin University, Changchun, Jilin 130021, PR China
| | - Jiaqi Liu
- The First Norman Bethune Clinical Medical College, Jilin University, Changchun, Jilin 130021, PR China
| | - Meng Yuan
- The First Norman Bethune Clinical Medical College, Jilin University, Changchun, Jilin 130021, PR China
| | - Shuang Guan
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, PR China.
| |
Collapse
|
10
|
Washington AM, Kostallari E. Extracellular Vesicles and Micro-RNAs in Liver Disease. Semin Liver Dis 2024. [PMID: 39626790 DOI: 10.1055/a-2494-2233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
Progression of liver disease is dependent on intercellular signaling, including those mediated by extracellular vesicles (EVs). Within these EVs, microRNAs (miRNAs) are packaged to selectively silence gene expression in recipient cells for upregulating or downregulating a specific pathway. Injured hepatocytes secrete EV-associated miRNAs which can be taken up by liver sinusoidal endothelial cells, immune cells, hepatic stellate cells, and other cell types. In addition, these recipient cells will secrete their own EV-associated miRNAs to propagate a response throughout the tissue and the circulation. In this review, we comment on the implications of EV-miRNAs in the progression of alcohol-associated liver disease, metabolic dysfunction-associated steatohepatitis, viral and parasitic infections, liver fibrosis, and liver malignancies. We summarize how circulating miRNAs can be used as biomarkers and the potential of utilizing EVs and miRNAs as therapeutic methods to treat liver disease.
Collapse
Affiliation(s)
- Alexander M Washington
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota
| | - Enis Kostallari
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
11
|
Shi C, Hu S, Liu S, Jia X, Feng Y. Emerging role of exosomes during the pathogenesis of viral hepatitis, non-alcoholic steatohepatitis and alcoholic hepatitis. Hum Cell 2024; 38:26. [PMID: 39630211 DOI: 10.1007/s13577-024-01158-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 11/24/2024] [Indexed: 01/07/2025]
Abstract
Extracellular vesicles (EVs) refer to a diverse range of membranous vesicles that are secreted by various cell types, they can be categorized into two primary subgroups: exosomes and microvesicles. Specifically, exosomes constitute a nanosized subset of EVs characterized by their intact lipid bilayer and diameters ranging from 30 to 150 nm. These vesicles play a crucial role in intercellular communication by transporting a diverse array of biomolecules, which act as cargoes for this communication process. Exosomes have demonstrated significant implications in a wide range of biologic processes and pathologic conditions, including immunity, development, cancer, neurodegenerative diseases, and liver diseases. Liver diseases significantly contribute to the global burden of morbidity and mortality, yet their pathogenesis remains complex and effective therapies are relatively scarce. Emerging evidence suggests that exosomes play a modulatory role in the pathogenesis of liver diseases, including viral hepatitis, non-alcoholic steatohepatitis (NASH), and alcoholic hepatitis (AH). These findings bolster our confidence in the potential of exosomes as biomarkers and therapeutic tools for the diagnosis and treatment of liver diseases. In this comprehensive review, we offer a straightforward overview of exosomes and summarize the current understanding of their role in the pathogenesis of liver diseases. This provides a foundation for novel diagnostic and therapeutic approaches in the treatment of liver diseases.
Collapse
Affiliation(s)
- Congjian Shi
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, 350007, China
| | - Shuang Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China
| | - Shen Liu
- Department of Pharmacy, Linquan County People's Hospital, Fuyang, 236400, Anhui, China
| | - Xiaodi Jia
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, 350007, China
| | - Yubin Feng
- Department of Pharmacy, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China.
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, 230001, Anhui, China.
| |
Collapse
|
12
|
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: 0] [Impact Index Per Article: 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
|
13
|
Chen H, Hinz K, Zhang C, Rodriguez Y, Williams SN, Niu M, Ma X, Chao X, Frazier AL, McCarson KE, Wang X, Peng Z, Liu W, Ni HM, Zhang J, Swerdlow RH, Ding WX. Late-Life Alcohol Exposure Does Not Exacerbate Age-Dependent Reductions in Mouse Spatial Memory and Brain TFEB Activity. Biomolecules 2024; 14:1537. [PMID: 39766244 PMCID: PMC11673978 DOI: 10.3390/biom14121537] [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: 09/23/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Alcohol consumption is believed to affect Alzheimer's disease (AD) risk, but the contributing mechanisms are not well understood. A potential mediator of the proposed alcohol-AD connection is autophagy, a degradation pathway that maintains organelle and protein homeostasis. Autophagy is regulated through the activity of Transcription factor EB (TFEB), which promotes lysosome and autophagy-related gene expression. The purpose of this study is to explore whether chronic alcohol consumption worsens the age-related decline in TFEB-mediated lysosomal biogenesis in the brain and exacerbates cognitive decline associated with aging. To explore the effect of alcohol on brain TFEB and autophagy, we exposed young (3-month-old) and aged (23-month-old) mice to two alcohol-feeding paradigms and assessed biochemical, transcriptome, histology, and behavioral endpoints. In young mice, alcohol decreased hippocampal nuclear TFEB staining but increased SQSTM1/p62, LC3-II, ubiquitinated proteins, and phosphorylated Tau. Hippocampal TFEB activity was lower in aged mice than it was in young mice, and Gao-binge alcohol feeding did not worsen the age-related reduction in TFEB activity. Morris Water and Barnes Maze spatial memory tasks were used to characterize the effects of aging and chronic alcohol exposure (mice fed alcohol for 4 weeks). The aged mice showed worse spatial memory acquisition in both tests. Alcohol feeding slightly impaired spatial memory in the young mice, but had little effect or even slightly improved spatial memory acquisition in the aged mice. In conclusion, aging produces greater reductions in brain autophagy flux and impairment of spatial memory than alcohol consumption.
Collapse
Affiliation(s)
- Hao Chen
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Kaitlyn Hinz
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Chen Zhang
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Yssa Rodriguez
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Sha Neisha Williams
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Mengwei Niu
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Xiaowen Ma
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Xiaojuan Chao
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Alexandria L. Frazier
- R.L. Smith IDDRC Rodent Behavior Facility, Disease Model and Assessment Services, The University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Kenneth E. McCarson
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
- R.L. Smith IDDRC Rodent Behavior Facility, Disease Model and Assessment Services, The University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Xiaowan Wang
- Department of Neurology, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (X.W.); (R.H.S.)
| | - Zheyun Peng
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Detroit, MI 48201, USA; (Z.P.); (W.L.)
| | - Wanqing Liu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Detroit, MI 48201, USA; (Z.P.); (W.L.)
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
| | - Jianhua Zhang
- Department of Pathology, Division of Molecular Cellular Pathology, University of Alabama at Birmingham, 901 19th Street South, Birmingham, AL 35294, USA;
| | - Russell H. Swerdlow
- Department of Neurology, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (X.W.); (R.H.S.)
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.C.); (K.H.); (C.Z.); (Y.R.); (S.N.W.); (M.N.); (X.M.); (X.C.); (K.E.M.); (H.-M.N.)
- Department of Internal Medicine, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
14
|
Hong X, Huang S, Jiang H, Ma Q, Qiu J, Luo Q, Cao C, Xu Y, Chen F, Chen Y, Sun C, Fu H, Liu Y, Li C, Chen F, Qiu P. Alcohol-related liver disease (ALD): current perspectives on pathogenesis, therapeutic strategies, and animal models. Front Pharmacol 2024; 15:1432480. [PMID: 39669199 PMCID: PMC11635172 DOI: 10.3389/fphar.2024.1432480] [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: 05/14/2024] [Accepted: 10/22/2024] [Indexed: 12/14/2024] Open
Abstract
Alcohol-related liver disease (ALD) is a major cause of morbidity and mortality worldwide. It encompasses conditions such as fatty liver, alcoholic hepatitis, chronic hepatitis with liver fibrosis or cirrhosis, and hepatocellular carcinoma. Numerous recent studies have demonstrated the critical role of oxidative stress, abnormal lipid metabolism, endoplasmic reticulum stress, various forms of cell death (including apoptosis, necroptosis, and ferroptosis), intestinal microbiota dysbiosis, liver immune response, cell autophagy, and epigenetic abnormalities in the pathogenesis of ALD. Currently, abstinence, corticosteroids, and nutritional therapy are the traditional therapeutic interventions for ALD. Emerging therapies for ALD mainly include the blockade of inflammatory pathways, the promotion of liver regeneration, and the restoration of normal microbiota. Summarizing the advances in animal models of ALD will facilitate a more systematic investigation of the pathogenesis of ALD and the exploration of therapeutic targets. This review summarizes the latest insight into the pathogenesis and molecular mechanisms of ALD, as well as the pros and cons of ALD rodent models, providing a basis for further research on therapeutic strategies for ALD.
Collapse
Affiliation(s)
- Xiao Hong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuo Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - He Jiang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qing Ma
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiang Qiu
- Department of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Qihan Luo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chunlu Cao
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiyang Xu
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fuzhe Chen
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yufan Chen
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chunfeng Sun
- The First People’s Hospital of Xiaoshan District, Xiaoshan Affiliated Hospital of Wenzhou Medical University, Hangzhou, China
| | - Haozhe Fu
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiming Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Changyu Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangming Chen
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ping Qiu
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
15
|
Jiang M, Feng Y, Wang J, Xu X, Liu Z, Li T, Ma S, Wang Y, Guo X, Du S. Saikogenin A improves ethanol-induced liver injury by targeting SIRT1 to modulate lipid metabolism. Commun Biol 2024; 7:1547. [PMID: 39572758 PMCID: PMC11582619 DOI: 10.1038/s42003-024-07234-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
Chronic alcohol consumption can lead to alcohol live disease (ALD). Steatosis is a critical hallmark of ALD, making it an important stage for therapeutic intervention. Saikosaponin A (SSa), a compound found in Radix Bupleuri, has previously shown promising hepatoprotective, anti-inflammatory, and antioxidant properties. However, its role in ALD remains understudied. We employ cell-based screening models and a chronic-plus-binge ethanol-fed mouse model to investigate the protective mechanisms of SSa and its metabolite Saikogenin A (SGA), against ethanol-induced hepatocyte injury. Our RNA-seq analysis in mice unveils that SSa primarily acts through the mTOR and PPAR-α signaling pathways in the liver. Biophysical assays and loss of function experiments confirm SGA directly binds to and modulates the activity of SIRT1 protein, mitigating ethanol-induced cell injury via the SIRT1-mTOR-PPAR-α axis. Furthermore, SGA displays a survival prolonging advantage compared to resveratrol for treating ALD. This suggests SGA holds promise as a potential therapeutic agent for ALD.
Collapse
Affiliation(s)
- Mingzhu Jiang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Department of neurosurgery, Taihe Hospital, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Ying Feng
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Department of neurosurgery, Taihe Hospital, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, China
| | - Jingxian Wang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Department of neurosurgery, Taihe Hospital, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, China
| | - Xiang Xu
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Department of neurosurgery, Taihe Hospital, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, China
| | - Zegan Liu
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Tongfei Li
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Department of neurosurgery, Taihe Hospital, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, China
| | - Shinan Ma
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Department of neurosurgery, Taihe Hospital, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, China
| | - Yufeng Wang
- Institute for Systems Genetics, New York University, New York, NY, USA.
| | - Xingrong Guo
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Department of neurosurgery, Taihe Hospital, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, China.
| | - Shiming Du
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Department of neurosurgery, Taihe Hospital, School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, China.
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China.
| |
Collapse
|
16
|
Luo Y, Zang N, Tang Y, Chen H, Liu H, Wang L, Xu P. STAT3 Inhibitor Increases α-Synuclein in PFF-Treated Astroglia Cells by Dysregulating Autophagy and Potentially Affects Exosome Biogenesis. Neuromolecular Med 2024; 26:46. [PMID: 39537842 DOI: 10.1007/s12017-024-08812-2] [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: 08/17/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
Astrocytes are the most abundant cells in the brain and show neuroprotective function in CNS and reactive astrocytes are characteristic in neurodegenerative diseases. The JAK2-STAT3 pathway plays a crucial role in the process of astrocyte activation. However, as a hallmark of Parkinson's disease, the change in α-syn under the influence of STAT3 inhibitor and the underlying cellular mechanisms remain elusive. Here, we show that PFF can induce an increase in α-syn in SVG p12 cells, which is further enhanced after the inhibition of STAT3. The underlying mechanisms involve the downregulation of autophagy levels and a concurrent decrease in lysosomal function after inhibition of STAT3. Additionally, we observed inhibition of STAT3 resulted in reduced exosome secretion in SVG p12 cells. This is attributed to alterations in SNARE, leading to impaired fusion between MVBs and the cell membrane, subsequently causing the accumulation of intracellular MVBs. Taken together, our data demonstrates that inhibition of STAT3 decreases both the autophagy and lysosome function, which may increase MVBs production. However, we found a potentially decreased exosome production that may be implicated with SNARE complex and need further exploration.
Collapse
Affiliation(s)
- Yangfu Luo
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Nailiang Zang
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuting Tang
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hao Chen
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hanqun Liu
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lan Wang
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pingyi Xu
- Department of Neurology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
17
|
Li S, Cheng F, Zhang Z, Xu R, Shi H, Yan Y. The role of hepatocyte-derived extracellular vesicles in liver and extrahepatic diseases. Biomed Pharmacother 2024; 180:117502. [PMID: 39357327 DOI: 10.1016/j.biopha.2024.117502] [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: 07/21/2024] [Revised: 09/09/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024] Open
Abstract
Extracellular vesicles (EVs) are vesicle-like bodies with a double membrane structure that are released from the cell membrane or secreted by cells into the extracellular environment. These include exosomes, microvesicles, and apoptotic bodies. There is growing evidence indicating that the composition of liver cell contents changes following injury. The quantity of EVs and the biologically active substances they carry vary depending on the condition of the liver cells. Hepatocytes utilize EVs to modulate the functions of different liver cells and transfer them to distant organs via the systemic circulation, thereby playing a crucial role in intercellular communication. This review provides a concise overview of the research on the effects and potential mechanisms of hepatocyte-derived EVs (Hep-EVs) on liver diseases and extrahepatic diseases under different physiological and pathological conditions. Common liver diseases discussed include non-alcoholic fatty liver disease (NAFLD), viral hepatitis, alcoholic liver disease, drug-induced liver damage, and liver cancer. Given that NAFLD is the most prevalent chronic liver disease globally, this review particularly highlights the use of hepatocyte-derived EVs in NAFLD for disease progression, diagnosis, and treatment.
Collapse
Affiliation(s)
- Shihui Li
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Fang Cheng
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Zhuan Zhang
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Ruizi Xu
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Honglei Shi
- Wujin Hospital Affiliated With Jiangsu University, Changzhou Wujin People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213004, China; Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University (Wujin Clinical College of Xuzhou Medical University), Changzhou 213017, China; Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Changzhou 213017, China.
| | - Yongmin Yan
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China; Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University (Wujin Clinical College of Xuzhou Medical University), Changzhou 213017, China; Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Changzhou 213017, China.
| |
Collapse
|
18
|
Severtsev VV, Pavkina MA, Ivanets NN, Vinnikova MA, Yakovlev AA. Extracellular Vesicles as Potential Biomarkers in Addictive Disorders. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1970-1984. [PMID: 39647826 DOI: 10.1134/s0006297924110117] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/03/2024] [Accepted: 08/09/2024] [Indexed: 12/10/2024]
Abstract
Small extracellular vesicles (sEVs) and their role in mental and addictive disorders are extremely promising research areas. Because of their small size, sEVs can pass through the blood-brain barrier. The membrane of sEVs contain proteins that protect them against destruction by the organism's immune system. Due to these properties, sEVs circulating in the blood can be used as potential biomarkers of processes occurring in the brain. Exposure to psychoactive substances in vitro and in vivo affects sEV biogenesis and significantly alters the amount of sEVs and chemical composition of their cargo. Based on the published reports, sEVs carry numerous potential biomarkers of addictive pathologies, although the diagnostic significance of these markers still has to be evaluated. A large body of evidence indicates that psychoactive substances influence Rab family GTPases, Toll-like receptors, complement system components, and cytokines. In some studies, the effect of psychoactive substances on sEVs was found to be sex-dependent. It has become commonly accepted that sEVs are involved in the regulation of neuroinflammation and interaction between glial cells and neurons, as well as between peripheral cells and cells of the central nervous system. Here, we formulated a hypothesis on the existence of two mechanisms/stages involved in the effect of psychoactive substances on sEVs: the "fast" mechanism that provides neuroplasticity, and the "slow" one, resulting from the impaired biogenesis of sEVs and formation of aberrant vesicles.
Collapse
Affiliation(s)
- Vsevolod V Severtsev
- Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation, Moscow, 119048, Russia.
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical-Biological Agency of the Russian Federation, Moscow, 143007, Russia
| | - Margarita A Pavkina
- Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation, Moscow, 119048, Russia
| | - Nikolay N Ivanets
- Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation, Moscow, 119048, Russia
| | - Maria A Vinnikova
- Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation, Moscow, 119048, Russia
- Moscow Scientific and Practical Center of Narcology, Moscow Healthcare Department, Moscow, 109390, Russia
| | - Alexander A Yakovlev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
- Research and Clinical Center for Neuropsychiatry, Moscow Healthcare Department, Moscow, 115419, Russia
| |
Collapse
|
19
|
Li B, Liu T, Shen Y, Qin J, Chang X, Wu M, Guo J, Liu L, Wei C, Lyu Y, Tian F, Yin J, Wang T, Zhang W, Qiu Y. TFEB/LAMP2 contributes to PM 0.2-induced autophagy-lysosome dysfunction and alpha-synuclein dysregulation in astrocytes. J Environ Sci (China) 2024; 145:117-127. [PMID: 38844312 DOI: 10.1016/j.jes.2023.09.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 06/15/2024]
Abstract
Atmospheric particulate matter (PM) exacerbates the risk factor for Alzheimer's and Parkinson's diseases (PD) by promoting the alpha-synuclein (α-syn) pathology in the brain. However, the molecular mechanisms of astrocytes involvement in α-syn pathology underlying the process remain unclear. This study investigated PM with particle size <200 nm (PM0.2) exposure-induced α-syn pathology in ICR mice and primary astrocytes, then assessed the effects of mammalian target of rapamycin inhibitor (PP242) in vitro studies. We observed the α-syn pathology in the brains of exposed mice. Meanwhile, PM0.2-exposed mice also exhibited the activation of glial cell and the inhibition of autophagy. In vitro study, PM0.2 (3, 10 and 30 µg/mL) induced inflammatory response and the disorders of α-syn degradation in primary astrocytes, and lysosomal-associated membrane protein 2 (LAMP2)-mediated autophagy underlies α-syn pathology. The abnormal function of autophagy-lysosome was specifically manifested as the expression of microtubule-associated protein light chain 3 (LC3II), cathepsin B (CTSB) and lysosomal abundance increased first and then decreased, which might both be a compensatory mechanism to toxic α-syn accumulation induced by PM0.2. Moreover, with the transcription factor EB (TFEB) subcellular localization and the increase in LC3II, LAMP2, CTSB, and cathepsin D proteins were identified, leading to the restoration of the degradation of α-syn after the intervention of PP242. Our results identified that PM0.2 exposure could promote the α-syn pathological dysregulation in astrocytes, providing mechanistic insights into how PM0.2 increases the risk of developing PD and highlighting TFEB/LAMP2 as a promising therapeutic target for antagonizing PM0.2 toxicity.
Collapse
Affiliation(s)
- Ben Li
- School of Public Health, Shanxi Medical University, Taiyuan 030000, China.
| | - Ting Liu
- School of Public Health, Shanxi Medical University, Taiyuan 030000, China
| | - Yongmei Shen
- Hainan Provincial Center for Disease Control and Prevention, Haikou 570100, China
| | - Jiangnan Qin
- School of Public Health, Shanxi Medical University, Taiyuan 030000, China
| | - Xiaohan Chang
- School of Public Health, Shanxi Medical University, Taiyuan 030000, China
| | - Meiqiong Wu
- School of Public Health, Shanxi Medical University, Taiyuan 030000, China
| | - Jianquan Guo
- School of Public Health, Shanxi Medical University, Taiyuan 030000, China
| | - Liangpo Liu
- School of Public Health, Shanxi Medical University, Taiyuan 030000, China
| | - Cailing Wei
- School of Public Health, Shanxi Medical University, Taiyuan 030000, China
| | - Yi Lyu
- School of Public Health, Shanxi Medical University, Taiyuan 030000, China
| | - Fengjie Tian
- School of Public Health, Shanxi Medical University, Taiyuan 030000, China
| | - Jinzhu Yin
- Department of Neurosurgery, Sinopharm Tongmei General Hospital, Datong 037003, China
| | - Tong Wang
- School of Public Health, Shanxi Medical University, Taiyuan 030000, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, Taiyuan 030000, China
| | - Wenping Zhang
- School of Public Health, Shanxi Medical University, Taiyuan 030000, China
| | - Yulan Qiu
- School of Public Health, Shanxi Medical University, Taiyuan 030000, China.
| |
Collapse
|
20
|
Liu JY, Liu ZL, Yang M, Du CL, Zhu Y, Sun LJ, Lv XW, Huang C, Li J. Involvement of BRD4 in Alcoholic Liver Injury: Autophagy Modulation via Regulation of the SIRT1/Beclin1 Axis. J Transl Med 2024; 104:102134. [PMID: 39307311 DOI: 10.1016/j.labinv.2024.102134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 08/26/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024] Open
Abstract
Alcoholic liver disease (ALD) caused by chronic alcohol abuse involves complex processes from steatosis to fibrosis, cirrhosis, and hepatocellular carcinoma, posing a global health issue. Bromodomain protein 4 (BRD4) typically serves as a "reader" modulating the functions of transcription factors involved in various biological processes and disease progression. However, the specific mechanisms underlying alcoholic liver injury remain unclear. In this study, we detected aberrant BRD4 expression in the alcohol-induced ALD mouse model of chronic and binge ethanol feeding developed by the National Institute on Alcohol Abuse and Alcoholism, consistent with the in vitro results in Aml-12 mouse hepatocytes. Blocking and inhibiting BRD4 restored the impaired autophagic flux and lysosomal functions in alcohol-treated Aml-12 cells, whereas BRD4 overexpression reduced the expression levels of autophagy marker and lysosomal genes. Furthermore, mouse BRD4 knockdown, mediated by a short hairpin RNA carried by the adeno-associated virus serotype 8, significantly attenuated the alcohol-induced hepatocyte damage, including lipid deposition and inflammatory cell infiltration. Mechanistically, BRD4 overexpression in alcoholic liver injury inhibited the expression of sirtuin (SIRT)1 in Aml-12 cells. Chromatin immunoprecipitation and dual-luciferase reporter assays revealed that BRD4 functions as a transcription factor and suppressor, actively binding to the SIRT1 promoter region and inhibiting its transcription. SIRT1 activated autophagy, which was suppressed in alcoholic liver injury via Beclin1 deacetylation. In conclusion, our study revealed that BRD4 negatively regulated the SIRT1/Beclin1 axis and that its deficiency alleviated alcohol-induced liver injury in mice, thus providing a new strategy for ALD treatment.
Collapse
Affiliation(s)
- Jin-Yu Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Zhen-Long Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Ming Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Chang-Lin Du
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Yan Zhu
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Li-Jiao Sun
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xong-Wen Lv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.
| |
Collapse
|
21
|
Li J, Yuan Y, Fu Q, Chen M, Liang H, Chen X, Long X, Zhang B, Zhao J, Chen Q. Novel insights into the role of immunomodulatory extracellular vesicles in the pathogenesis of liver fibrosis. Biomark Res 2024; 12:119. [PMID: 39396032 PMCID: PMC11470730 DOI: 10.1186/s40364-024-00669-8] [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: 06/28/2024] [Accepted: 10/07/2024] [Indexed: 10/14/2024] Open
Abstract
Liver fibrosis, a chronic and long-term disease, can develop into hepatocellular carcinoma (HCC) and ultimately lead to liver failure. Early diagnosis and effective treatment still face significant challenges. Liver inflammation leads to liver fibrosis through continuous activation of hepatic stellate cells (HSCs) and the accumulation of immune cells. Intracellular communication among various immune cells is important for mediating the inflammatory response during fibrogenesis. Extracellular vesicles (EVs), which are lipid bilayer membrane-enclosed particles naturally secreted by cells, make great contributions to cell-cell communication and the transport of bioactive molecules. Nearly all the cells that participate in liver fibrosis release EVs loaded with lipids, proteins, and nucleic acids. EVs from hepatocytes, immune cells and stem cells are involved in mediating the inflammatory microenvironment of liver fibrosis. Recently, an increasing number of extracellular vesicle-based clinical applications have emerged, providing promising cell-free diagnostic and therapeutic tools for liver fibrosis because of their crucial role in immunomodulation during pathogenesis. The advantages of extracellular vesicle-based therapies include stability, biocompatibility, low cytotoxicity, and minimal immunogenicity, which highlight their great potential for drug delivery and specific treatments for liver fibrosis. In this review, we summarize the complex biological functions of EVs in the inflammatory response in the pathogenesis of liver fibrosis and evaluate the potential of EVs in the diagnosis and treatment of liver fibrosis.
Collapse
Affiliation(s)
- Jiaxuan Li
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yue Yuan
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qinggang Fu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Min Chen
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, China
| | - Xin Long
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, China
| | - Jianping Zhao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, China.
| | - Qian Chen
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
22
|
Tam S, Wear D, Morrone CD, Yu WH. The complexity of extracellular vesicles: Bridging the gap between cellular communication and neuropathology. J Neurochem 2024; 168:2391-2422. [PMID: 38650384 DOI: 10.1111/jnc.16108] [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: 01/14/2024] [Revised: 03/12/2024] [Accepted: 03/31/2024] [Indexed: 04/25/2024]
Abstract
Brain-derived extracellular vesicles (EVs) serve a prominent role in maintaining homeostasis and contributing to pathology in health and disease. This review establishes a crucial link between physiological processes leading to EV biogenesis and their impacts on disease. EVs are involved in the clearance and transport of proteins and nucleic acids, responding to changes in cellular processes associated with neurodegeneration, including autophagic disruption, organellar dysfunction, aging, and other cell stresses. In neurodegenerative disorders (e.g., Alzheimer's disease, Parkinson's disease, etc.), EVs contribute to the spread of pathological proteins like amyloid β, tau, ɑ-synuclein, prions, and TDP-43, exacerbating neurodegeneration and accelerating disease progression. Despite evidence for both neuropathological and neuroprotective effects of EVs, the mechanistic switch between their physiological and pathological functions remains elusive, warranting further research into their involvement in neurodegenerative disease. Moreover, owing to their innate ability to traverse the blood-brain barrier and their ubiquitous nature, EVs emerge as promising candidates for novel diagnostic and therapeutic strategies. The review uniquely positions itself at the intersection of EV cell biology, neurophysiology, and neuropathology, offering insights into the diverse biological roles of EVs in health and disease.
Collapse
Affiliation(s)
- Stephanie Tam
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Darcy Wear
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Christopher D Morrone
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Wai Haung Yu
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
23
|
Ma N, Tan J, Chen Y, Yang L, Li M, He Y. MicroRNAs in metabolic dysfunction-associated diseases: Pathogenesis and therapeutic opportunities. FASEB J 2024; 38:e70038. [PMID: 39250169 DOI: 10.1096/fj.202401464r] [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: 06/27/2024] [Revised: 08/13/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024]
Abstract
Metabolic dysfunction-associated diseases often refer to various diseases caused by metabolic problems such as glucose and lipid metabolism disorders. With the improvement of living standards, the increasing prevalence of metabolic diseases has become a severe public health problem, including metabolic dysfunction-associated steatotic liver disease (MASLD), alcohol-related liver disease (ALD), diabetes and obesity. These diseases are both independent and interdependent, with complex and diverse molecular mechanisms. Therefore, it is urgent to explore the molecular mechanisms and find effective therapeutic targets of these diseases. MicroRNAs (miRNAs) have emerged as key regulators of metabolic homoeostasis due to their multitargets and network regulatory properties within the past few decades. In this review, we discussed the latest progress in the roles of miRNA-mediated regulatory networks in the development and progression of MASLD, ALD, diabetes and obesity.
Collapse
Affiliation(s)
- Ningning Ma
- Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiaxin Tan
- Laboratory of Cellular Immunity, Shanghai Key Laboratory of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yingfen Chen
- Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Liu Yang
- Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Man Li
- Laboratory of Cellular Immunity, Shanghai Key Laboratory of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong He
- Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
24
|
Xu J, Gu J, Pei W, Zhang Y, Wang L, Gao J. The role of lysosomal membrane proteins in autophagy and related diseases. FEBS J 2024; 291:3762-3785. [PMID: 37221945 DOI: 10.1111/febs.16820] [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: 02/14/2023] [Revised: 04/12/2023] [Accepted: 05/15/2023] [Indexed: 05/25/2023]
Abstract
As a self-degrading and highly conserved survival mechanism, autophagy plays an important role in maintaining cell survival and recycling. The discovery of autophagy-related (ATG) genes has revolutionized our understanding of autophagy. Lysosomal membrane proteins (LMPs) are important executors of lysosomal function, and increasing evidence has demonstrated their role in the induction and regulation of autophagy. In addition, the functional dysregulation of the process mediated by LMPs at all stages of autophagy is closely related to neurodegenerative diseases and cancer. Here, we review the role of LMPs in autophagy, focusing on their roles in vesicle nucleation, vesicle elongation and completion, the fusion of autophagosomes and lysosomes, and degradation, as well as their broad association with related diseases.
Collapse
Affiliation(s)
- Jiahao Xu
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Department of Endocrinology and Genetic Metabolism, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- School of Clinical Medicine, Wannan Medical College, Wuhu, China
| | - Jing Gu
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Department of Endocrinology and Genetic Metabolism, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
| | - Wenjun Pei
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, China
| | - Yao Zhang
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, China
| | - Lizhuo Wang
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- Department of Biochemistry and Molecular Biology, Wannan Medical College, Wuhu, China
| | - Jialin Gao
- Department of Endocrinology and Genetic Metabolism, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Department of Endocrinology and Genetic Metabolism, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Anhui Province Key Laboratory of Biological Macro-Molecules Research, Wannan Medical College, Wuhu, China
- Anhui Provincial College Key Laboratory of Non-coding RNA Transformation Research on Critical Diseases, Wannan Medical College, Wuhu, China
| |
Collapse
|
25
|
Li M, Houben T, Bitorina AV, Meesters DM, Israelsen M, Kjærgaard M, Koek GH, Hendrikx T, Verbeek J, Krag A, Thiele M, Shiri-Sverdlov R. Plasma cathepsin D as an early indicator of alcohol-related liver disease. JHEP Rep 2024; 6:101117. [PMID: 39263329 PMCID: PMC11388167 DOI: 10.1016/j.jhepr.2024.101117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/26/2024] [Accepted: 05/02/2024] [Indexed: 09/13/2024] Open
Abstract
Background & Aims People who drink alcohol excessively are at increased risk of developing metabolic dysfunction and alcohol-related liver disease (MetALD) or the more severe form alcohol-related liver disease (ALD). One of the most significant challenges concerns the early detection of MetALD/ALD. Previously, we have demonstrated that the lysosomal enzyme cathepsin D (CTSD) is an early marker for metabolic dysfunction-associated steatohepatitis (MASH). Here, we hypothesized that plasma CTSD can also serve as an early indicator of MetALD/ALD. Methods We included 303 persistent heavy drinkers classified as having MetALD or ALD (n = 152) and abstinent patients with a history of excessive drinking (n = 151). Plasma CTSD levels of patients with MetALD/ALD without decompensation were compared with 40 healthy controls. Subsequently, the relationship between plasma CTSD levels and hepatic histological scores was established. Receiver-operating characteristic curves were generated to assess the precision of plasma CTSD levels in detecting MetALD/ALD. Lastly, plasma CTSD levels were compared between abstainers and drinkers. Results Plasma CTSD levels were higher in patients with MetALD/ALD compared to healthy controls. While hepatic disease parameters (AST/ALT ratio, liver stiffness measurement) were higher at advanced histopathological stages (assessed by liver biopsy), plasma CTSD levels were already elevated at early histopathological stages. Furthermore, combining plasma CTSD levels with liver stiffness measurement and AST/ALT ratio yielded enhanced diagnostic precision (AUC 0.872) in detecting MetALD/ALD in contrast to the utilization of CTSD alone (AUC 0.804). Plasma CTSD levels remained elevated in abstainers. Conclusion Elevated levels of CTSD in the circulation can serve as an early indicator of MetALD/ALD. Impact and implications Alcohol-related liver disease is the leading cause of liver disease-related morbidity and mortality worldwide. However, the currently available non-invasive methods to diagnose MetALD/ALD are only able to detect advanced stages of MetALD/ALD. Here, we demonstrate that plasma levels of the lysosomal enzyme cathepsin D are already elevated at early stages of MetALD/ALD. Moreover, cathepsin D levels outperformed the currently available non-invasive methods to detect MetALD/ALD. Plasma levels of cathepsin D could therefore be a useful non-invasive marker for detection of MetALD/ALD.
Collapse
Affiliation(s)
- Mengying Li
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism, Maastricht University, the Netherlands
| | - Tom Houben
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism, Maastricht University, the Netherlands
| | - Albert V. Bitorina
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism, Maastricht University, the Netherlands
| | - Dennis M. Meesters
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism, Maastricht University, the Netherlands
| | - Mads Israelsen
- Center for Liver Research, Odense University Hospital and University of Southern Denmark, Kloevervaenget 10, entrance 112, DK-5000 Odense, Denmark
| | - Maria Kjærgaard
- Center for Liver Research, Odense University Hospital and University of Southern Denmark, Kloevervaenget 10, entrance 112, DK-5000 Odense, Denmark
| | - Ger H. Koek
- Department of Internal Medicine Division of Gastroenterology and Hepatology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Tim Hendrikx
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Jef Verbeek
- Laboratory of Hepatology, Department of Chronic Diseases and Metabolism, KU Leuven, Belgium; Department of Gastroenterology & Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Aleksander Krag
- Center for Liver Research, Odense University Hospital and University of Southern Denmark, Kloevervaenget 10, entrance 112, DK-5000 Odense, Denmark
| | - Maja Thiele
- Center for Liver Research, Odense University Hospital and University of Southern Denmark, Kloevervaenget 10, entrance 112, DK-5000 Odense, Denmark
| | - Ronit Shiri-Sverdlov
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism, Maastricht University, the Netherlands
| |
Collapse
|
26
|
He W, Xu J, Wang X, Fan Z, Li H. Macrophage-derived exosomal miR-155 regulating hepatocyte pyroptosis in MAFLD. Heliyon 2024; 10:e35197. [PMID: 39157367 PMCID: PMC11328038 DOI: 10.1016/j.heliyon.2024.e35197] [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: 11/04/2023] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 08/20/2024] Open
Abstract
Background Previous studies have shown that pyroptosis in hepatocyte is essential for the development of MAFLD. Growing evidence has shown that exosomal miRNAs-mediated communication between inflammatory cells and hepatocyte is an important link in MAFLD. In the present study, we aim to elucidate whether macrophage-derived exosomal miRNAs contribute to the hepatocyte pyroptosis in the pathophysiological process of MAFLD. Methods The effects of hepatocyte pyroptosis were investigated in an HFD-induced MAFLD mouse model and in the liver tissues from patients with MAFLD using immunohistochemistry, real-time PCR, Western blotting, and luciferase reporter assay, among other techniques. MiR-155 inhibitor tail injections and AAV-FoxO3a-GFP were also administered to respectively inhibit or overexpress its expression in an HFD-induced MAFLD mouse model. Results Hepatocyte pyroptosis was heightened in the liver tissue of patients with MAFLD or HFD-induced MAFLD mouse. Importantly, treatment with a caspase-1 inhibitor or overexpression of FoxO3a reversed this trend. Our study also demonstrated that miR-155 expression and the number of infiltrated macrophages were increased, and knockdown of miR-155 attenuated hepotocyte pyroptosis and liver fibrosis in HFD-induced mouse. In addition, we demonstrated that macrophage-derived exosomal miR-155 was transferred to hepatocytes, leading to hepatocyte pyroptosis in MAFLD mouse. Furthermore, blockade of exosome secretion improved hepotocyte pyroptosis and liver fibrosis in HFD-induced mouse. On the contrary, macrophage-derived exosomal miR-155 worsened hepotocyte pyroptosis. Moreover, we found that miR-155 promoted hepatocyte pyroptosis in MAFLD by down-regulating FoxO3a. Conclusions Taken together, our results demonstrated that macrophage-derived exosomal miR-155 promotes hepatocyte pyroptosis and liver fibrosis in MAFLD.
Collapse
Affiliation(s)
- Wei He
- Corresponding author. Department of Gastroenterology, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Institute, Jiangsu Province Official Hospital, Nanjing, 210024, Jiangsu Province, China.
| | | | - Xiang Wang
- Department of Gastroenterology, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Institute, Jiangsu Province Official Hospital, Nanjing, 210024, Jiangsu Province, China
| | - Zhining Fan
- Department of Gastroenterology, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Institute, Jiangsu Province Official Hospital, Nanjing, 210024, Jiangsu Province, China
| | - Hai Li
- Department of Gastroenterology, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Institute, Jiangsu Province Official Hospital, Nanjing, 210024, Jiangsu Province, China
| |
Collapse
|
27
|
Yu J, Yu C, Jiang K, Yang G, Yang S, Tan S, Li T, Liang H, He Q, Wei F, Li Y, Cheng J, Wang F. Unveiling potential: urinary exosomal mRNAs as non-invasive biomarkers for early prostate cancer diagnosis. BMC Urol 2024; 24:163. [PMID: 39090720 PMCID: PMC11292860 DOI: 10.1186/s12894-024-01540-6] [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: 04/11/2024] [Accepted: 07/09/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND This study investigated the use of urinary exosomal mRNA as a potential biomarker for the early detection of prostate cancer (PCa). METHODS Next-generation sequencing was utilized to analyze exosomal RNA from 10 individuals with confirmed PCa and 10 individuals without cancer. Subsequent validation through qRT-PCR in a larger sample of 43 PCa patients and 92 healthy controls revealed distinct mRNA signatures associated with PCa. RESULTS Notably, mRNAs for RAB5B, WWP1, HIST2H2BF, ZFY, MARK2, PASK, RBM10, and NRSN2 showed promise as diagnostic markers, with AUC values between 0.799 and 0.906 and significance p values. Combining RAB5B and WWP1 in an exoRNA diagnostic model outperformed traditional PSA tests, achieving an AUC of 0.923, 81.4% sensitivity, and 89.1% specificity. CONCLUSIONS These findings highlight the potential of urinary exosomal mRNA profiling, particularly focusing on RAB5B and WWP1, as a valuable strategy for improving the early detection of PCa.
Collapse
Affiliation(s)
- Jiayin Yu
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu, Nanning, Guangxi, 530021, P.R. China
| | - Chifei Yu
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, No.71 Hedi Road, Qingxiu District, Nanning, Guangxi, 530021, P.R. China
| | - Kangxian Jiang
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, No. 34 Zhongshan North Road, Quanzhou, Fujian, 362000, P.R. China
| | - Guanglin Yang
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, No.71 Hedi Road, Qingxiu District, Nanning, Guangxi, 530021, P.R. China
| | - Shubo Yang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu, Nanning, Guangxi, 530021, P.R. China
| | - Shuting Tan
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu, Nanning, Guangxi, 530021, P.R. China
| | - Tingting Li
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, No.71 Hedi Road, Qingxiu District, Nanning, Guangxi, 530021, P.R. China
| | - Haiqi Liang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu, Nanning, Guangxi, 530021, P.R. China
| | - Qihuan He
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu, Nanning, Guangxi, 530021, P.R. China
| | - Faye Wei
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu, Nanning, Guangxi, 530021, P.R. China
| | - Yujian Li
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu, Nanning, Guangxi, 530021, P.R. China
| | - Jiwen Cheng
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu, Nanning, Guangxi, 530021, P.R. China.
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, No.22 Shuangyong Road, Qingxiu District, Nanning, Guangxi, 530021, P.R. China.
| | - Fubo Wang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, No.22 Shuangyong Road, Qingxiu District, Nanning, Guangxi, 530021, P.R. China.
| |
Collapse
|
28
|
Koizumi A, Kaji K, Nishimura N, Asada S, Matsuda T, Tanaka M, Yorioka N, Tsuji Y, Kitagawa K, Sato S, Namisaki T, Akahane T, Yoshiji H. Effects of elafibranor on liver fibrosis and gut barrier function in a mouse model of alcohol-associated liver disease. World J Gastroenterol 2024; 30:3428-3446. [PMID: 39091710 PMCID: PMC11290391 DOI: 10.3748/wjg.v30.i28.3428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/31/2024] [Accepted: 06/20/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND Alcohol-associated liver disease (ALD) is a leading cause of liver-related morbidity and mortality, but there are no therapeutic targets and modalities to prevent ALD-related liver fibrosis. Peroxisome proliferator activated receptor (PPAR) α and δ play a key role in lipid metabolism and intestinal barrier homeostasis, which are major contributors to the pathological progression of ALD. Meanwhile, elafibranor (EFN), which is a dual PPARα and PPARδ agonist, has reached a phase III clinical trial for the treatment of metabolic dysfunction-associated steatotic liver disease and primary biliary cholangitis. However, the benefits of EFN for ALD treatment is unknown. AIM To evaluate the inhibitory effects of EFN on liver fibrosis and gut-intestinal barrier dysfunction in an ALD mouse model. METHODS ALD-related liver fibrosis was induced in female C57BL/6J mice by feeding a 2.5% ethanol (EtOH)-containing Lieber-DeCarli liquid diet and intraperitoneally injecting carbon tetrachloride thrice weekly (1 mL/kg) for 8 weeks. EFN (3 and 10 mg/kg/day) was orally administered during the experimental period. Histological and molecular analyses were performed to assess the effect of EFN on steatohepatitis, fibrosis, and intestinal barrier integrity. The EFN effects on HepG2 lipotoxicity and Caco-2 barrier function were evaluated by cell-based assays. RESULTS The hepatic steatosis, apoptosis, and fibrosis in the ALD mice model were significantly attenuated by EFN treatment. EFN promoted lipolysis and β-oxidation and enhanced autophagic and antioxidant capacities in EtOH-stimulated HepG2 cells, primarily through PPARα activation. Moreover, EFN inhibited the Kupffer cell-mediated inflammatory response, with blunted hepatic exposure to lipopolysaccharide (LPS) and toll like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) signaling. EFN improved intestinal hyperpermeability by restoring tight junction proteins and autophagy and by inhibiting apoptosis and proinflammatory responses. The protective effect on intestinal barrier function in the EtOH-stimulated Caco-2 cells was predominantly mediated by PPARδ activation. CONCLUSION EFN reduced ALD-related fibrosis by inhibiting lipid accumulation and apoptosis, enhancing hepatocyte autophagic and antioxidant capacities, and suppressing LPS/TLR4/NF-κB-mediated inflammatory responses by restoring intestinal barrier function.
Collapse
Affiliation(s)
- Aritoshi Koizumi
- Department of Gastroenterology, Nara Medical University, Kashihara 634-8521, Japan
| | - Kosuke Kaji
- Department of Gastroenterology, Nara Medical University, Kashihara 634-8521, Japan
| | - Norihisa Nishimura
- Department of Gastroenterology, Nara Medical University, Kashihara 634-8521, Japan
| | - Shohei Asada
- Department of Gastroenterology, Nara Medical University, Kashihara 634-8521, Japan
| | - Takuya Matsuda
- Department of Gastroenterology, Nara Medical University, Kashihara 634-8521, Japan
| | - Misako Tanaka
- Department of Gastroenterology, Nara Medical University, Kashihara 634-8521, Japan
| | - Nobuyuki Yorioka
- Department of Gastroenterology, Nara Medical University, Kashihara 634-8521, Japan
| | - Yuki Tsuji
- Department of Gastroenterology, Nara Medical University, Kashihara 634-8521, Japan
| | - Koh Kitagawa
- Department of Gastroenterology, Nara Medical University, Kashihara 634-8521, Japan
| | - Shinya Sato
- Department of Gastroenterology, Nara Medical University, Kashihara 634-8521, Japan
| | - Tadashi Namisaki
- Department of Gastroenterology, Nara Medical University, Kashihara 634-8521, Japan
| | - Takemi Akahane
- Department of Gastroenterology, Nara Medical University, Kashihara 634-8521, Japan
| | - Hitoshi Yoshiji
- Department of Gastroenterology, Nara Medical University, Kashihara 634-8521, Japan
| |
Collapse
|
29
|
Luo S, Luo R, Deng G, Huang F, Lei Z. Programmed cell death, from liver Ischemia-Reperfusion injury perspective: An overview. Heliyon 2024; 10:e32480. [PMID: 39040334 PMCID: PMC11260932 DOI: 10.1016/j.heliyon.2024.e32480] [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: 01/22/2024] [Revised: 05/26/2024] [Accepted: 06/04/2024] [Indexed: 07/24/2024] Open
Abstract
Liver ischemia-reperfusion injury (LIRI) commonly occurs in liver resection, liver transplantation, shock, and other hemorrhagic conditions, resulting in profound local and systemic effects via associated inflammatory responses and hepatic cell death. Hepatocyte death is a significant component of LIRI and its mechanism was previously thought to be limited to apoptosis and necrosis. With the discovery of novel types of programmed cell death (PCD), necroptosis, ferroptosis, pyroptosis, autophagy, NETosis, and parthanatos have been shown to be involved in LIRI. Understanding the mechanisms underlying cell death following LIRI is indispensable to mitigating the widespread effects of LIRI. Here, we review the roles of different PCD and discuss potential therapy in LIRI.
Collapse
Affiliation(s)
- Shaobin Luo
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha , PR China
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Rongkun Luo
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha , PR China
| | - Gang Deng
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha , PR China
| | - Feizhou Huang
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha , PR China
| | - Zhao Lei
- Department of Hepatopancreatobiliary Surgery, The Third Xiangya Hospital, Central South University, Changsha , PR China
| |
Collapse
|
30
|
Cheng Z, Yang L, Chu H. The role of gut microbiota, exosomes, and their interaction in the pathogenesis of ALD. J Adv Res 2024:S2090-1232(24)00268-6. [PMID: 38969094 DOI: 10.1016/j.jare.2024.07.002] [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: 05/06/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND The liver disorders caused by alcohol abuse are termed alcoholic-related liver disease (ALD), including alcoholic steatosis, alcoholic steatohepatitis, alcoholic hepatitis, and alcoholic cirrhosis, posing a significant threat to human health. Currently, ALD pathogenesis has not been completely clarified, which is likely to be related to the direct damage caused by alcohol and its metabolic products, oxidative stress, gut dysbiosis, and exosomes. AIMS The existing studies suggest that both the gut microbiota and exosomes contribute to the development of ALD. Moreover, there exists an interaction between the gut microbiota and exosomes. We discuss whether this interaction plays a role in the pathogenesis of ALD and whether it can be a potential therapeutic target for ALD treatment. KEY SCIENTIFIC CONCEPTS OF REVIEW Chronic alcohol intake alters the diversity and composition of gut microbiota, which greatly contributes to ALD's progression. Some approaches targeting the gut microbiota, including probiotics, fecal microbiota transplantation, and phage therapy, have been confirmed to effectively ameliorate ALD in many animal experiments and/or several clinical trials. In ALD, the levels of exosomes and the expression profile of microRNA have also changed, which affects the pathogenesis of ALD. Moreover, there is an interplay between exosomes and the gut microbiota, which also putatively acts as a pathogenic factor of ALD.
Collapse
Affiliation(s)
- Zilu Cheng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China.
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China.
| |
Collapse
|
31
|
Biondi A, Vacante M, Catania R, Sangiorgio G. Extracellular Vesicles and Immune System Function: Exploring Novel Approaches to Colorectal Cancer Immunotherapy. Biomedicines 2024; 12:1473. [PMID: 39062046 PMCID: PMC11275211 DOI: 10.3390/biomedicines12071473] [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: 05/29/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
This review explores the emerging role of extracellular vesicles (EVs) in modulating immune system function and their application in novel cancer immunotherapy strategies, with a focus on colorectal cancer (CRC). EVs, as carriers of bioactive molecules, have shown potential in enhancing immune responses and overcoming the limitations of traditional therapies. We discuss the biogenesis, types, and functional roles of immune cell-derived EVs, their interactions with cancer cells, and their implications in antitumor immunity. Challenges such as tumor heterogeneity and immune evasion are addressed, alongside the promising therapeutic prospects of EV-based strategies. This comprehensive analysis underscores the transformative potential of EVs in cancer treatment paradigms.
Collapse
Affiliation(s)
- Antonio Biondi
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy; (A.B.); (R.C.)
| | - Marco Vacante
- Unit of Internal Medicine Critical Area—ARNAS Garibaldi, Piazza Santa Maria di Gesù, 5, 95124 Catania, Italy;
| | - Roberta Catania
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy; (A.B.); (R.C.)
| | - Giuseppe Sangiorgio
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy; (A.B.); (R.C.)
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| |
Collapse
|
32
|
Singal AK, Shah VH, Malhi H. Emerging targets for therapy in ALD: Lessons from NASH. Hepatology 2024; 80:223-237. [PMID: 36938877 PMCID: PMC10511666 DOI: 10.1097/hep.0000000000000381] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 03/09/2023] [Indexed: 03/21/2023]
Abstract
Alcohol-associated liver disease due to harmful alcohol use and NAFLD associated with metabolic syndrome are the 2 most common liver diseases worldwide. Control of respective risk factors is the cornerstone in the long-term management of these diseases. Furthermore, there are no effective therapies. Both diseases are characterized by metabolic derangements; thus, the focus of this review was to broaden our understanding of metabolic targets investigated in NAFLD, and how these can be applied to alcohol-associated liver disease. Conserved pathogenic pathways such as dysregulated lipid metabolism, cell death pathways including apoptosis and activation of innate immune cells, and stellate cells mediate both alcohol and NAFLDs, resulting in histological abnormalities of steatosis, inflammation, fibrosis, and cirrhosis. However, pathways such as gut microbiome changes, glucose metabolism and insulin resistance, inflammatory signaling, and microRNA abnormalities are distinct in these 2 diseases. In this review article, we describe conserved and distinct pathogenic pathways highlighting therapeutic targets that may be of potential in both diseases and those that are unique to each disease.
Collapse
Affiliation(s)
- Ashwani K. Singal
- Department of Internal Medicine, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, USA
- Division of Gastroenterology and Hepatology, Avera Transplant Institute, Sioux Falls, South Dakota, USA
- VA Medical Center, Sioux Falls, South Dakota, USA
| | - Vijay H. Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
33
|
Qian Z, Xiong W, Mao X, Li J. Macrophage Perspectives in Liver Diseases: Programmed Death, Related Biomarkers, and Targeted Therapy. Biomolecules 2024; 14:700. [PMID: 38927103 PMCID: PMC11202214 DOI: 10.3390/biom14060700] [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: 04/28/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Macrophages, as important immune cells of the organism, are involved in maintaining intrahepatic microenvironmental homeostasis and can undergo rapid phenotypic changes in the injured or recovering liver. In recent years, the crucial role of macrophage-programmed cell death in the development and regression of liver diseases has become a research hotspot. Moreover, macrophage-targeted therapeutic strategies are emerging in both preclinical and clinical studies. Given the macrophages' vital role in complex organismal environments, there is tremendous academic interest in developing novel therapeutic strategies that target these cells. This review provides an overview of the characteristics and interactions between macrophage polarization, programmed cell death, related biomarkers, and macrophage-targeted therapies. It aims to deepen the understanding of macrophage immunomodulation and molecular mechanisms and to provide a basis for the treatment of macrophage-associated liver diseases.
Collapse
Affiliation(s)
- Zibing Qian
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; (Z.Q.); (W.X.)
| | - Wanyuan Xiong
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; (Z.Q.); (W.X.)
| | - Xiaorong Mao
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; (Z.Q.); (W.X.)
- Department of Infectious Disease, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Junfeng Li
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; (Z.Q.); (W.X.)
- Institute of Infectious Diseases, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Department of Hepatology, The First Hospital of Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
34
|
Hu T, Liu CH, Zheng Y, Ji J, Zheng Y, He SK, Wu D, Jiang W, Zeng Q, Zhang N, Tang H. miRNAs in patients with alcoholic liver disease: a systematic review and meta-analysis. Expert Rev Gastroenterol Hepatol 2024; 18:283-292. [PMID: 38937981 DOI: 10.1080/17474124.2024.2374470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 06/26/2024] [Indexed: 06/29/2024]
Abstract
INTRODUCTION Alcoholic liver disease (ALD) encompasses a spectrum of liver conditions, including liver steatosis, alcoholic hepatitis (AH), fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). microRNAs (miRNAs) have garnered significant interest as potential biomarkers for ALD. METHODS We searched PubMed, Embase, Web of Science and Cochrane Central Register of Controlled Trials (CENTRAL) systemically from inception to June 2024. All extracted data was stratified according to the stages of ALD. The vote-counting strategy performed a meta-analysis on miRNA expression profiles. RESULTS We included 40 studies. In serum of individuals with alcohol-use vs. no alcohol-use, miRNA-122 and miRNA-155 were upregulated, and miRNA-146a was downregulated. In patients with ALD vs. healthy controls, miRNA-122 and miRNA-155 were also upregulated, and miRNA-146a was downregulated. However, in patients with AH vs. healthy individuals, only the serum miRNA-122 level was upregulated. Due to insufficient data on diagnostic accuracy, we failed to conclude the ability of miRNAs to distinguish between different stages of ALD-related liver fibrosis. The results for ALD-related HCC were also insufficient and controversial. CONCLUSIONS Circulating miRNA-122 was the most promising biomarker to manage individuals with ALD. More studies were needed for the diagnostic accuracy of miRNAs in ALD. REGISTRATION This protocol was registered on the International Prospective Register of Systematic Reviews (PROSPERO) (www.crd.york.ac.uk/prospero/) with registration number CRD42023391931.
Collapse
Affiliation(s)
- Tengyue Hu
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Chang Hai Liu
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Yurong Zheng
- West China School of Public Health, Sichuan University, Chengdu, China
| | - Jialin Ji
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Yantong Zheng
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Si-Ke He
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Dongbo Wu
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Wei Jiang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Qingmin Zeng
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Nannan Zhang
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Chengdu, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
35
|
Babuta M, Nagesh PT, Datta AA, Remotti V, Zhuang Y, Mehta J, Lami F, Wang Y, Szabo G. Combined Insults of a MASH Diet and Alcohol Binges Activate Intercellular Communication and Neutrophil Recruitment via the NLRP3-IL-1β Axis in the Liver. Cells 2024; 13:960. [PMID: 38891092 PMCID: PMC11171595 DOI: 10.3390/cells13110960] [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: 04/17/2024] [Revised: 05/16/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
Binge drinking in obese patients positively correlates with accelerated liver damage and liver-related death. However, the underlying mechanism and the effect of alcohol use on the progression of metabolic-dysfunction-associated steatotic liver disease (MASLD) remain unexplored. Here, we show that short-term feeding of a metabolic-dysfunction-associated steatohepatitis (MASH) diet plus daily acute alcohol binges for three days induce liver injury and activation of the NLRP3 inflammasome. We identify that a MASH diet plus acute alcohol binges promote liver inflammation via increased infiltration of monocyte-derived macrophages, neutrophil recruitment, and NET release in the liver. Our results suggest that both monocyte-derived macrophages and neutrophils are activated via NLRP3, while the administration of MCC950, an NLRP3 inhibitor, dampens these effects.In this study, we reveal important intercellular communication between hepatocytes and neutrophils. We discover that the MASH diet plus alcohol induces IL-1β via NLRP3 activation and that IL-1β acts on hepatocytes and promotes the production of CXCL1 and LCN2. In turn, the increase in these neutrophils recruits chemokines and causes further infiltration and activation of neutrophils in the liver. In vivo administration of the NLRP3 inhibitor, MCC950, improves the early phase of MetALD by preventing liver damage, steatosis, inflammation, and immune cells recruitment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Gyongyi Szabo
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; (M.B.)
| |
Collapse
|
36
|
Yang L, Liu S, He Y, Gan L, Ni Q, Dai A, Mu C, Liu Q, Chen H, Lu H, Sun R. Exosomes regulate SIRT3-related autophagy by delivering miR-421 to regulate macrophage polarization and participate in OSA-related NAFLD. J Transl Med 2024; 22:475. [PMID: 38764033 PMCID: PMC11103849 DOI: 10.1186/s12967-024-05283-8] [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: 12/21/2023] [Accepted: 05/08/2024] [Indexed: 05/21/2024] Open
Abstract
PURPOSE To analyze the role of and mechanism underlying obstructive sleep apnea (OSA)-derived exosomes in inducing non-alcoholic fatty liver (NAFLD). METHODS The role of OSA-derived exosomes was analyzed in inducing hepatocyte fat accumulation in mice models both in vivo and in vitro. RESULTS OSA-derived exosomes caused fat accumulation and macrophage activation in the liver tissue. These exosomes promoted fat accumulation; steatosis was more noticeable in the presence of macrophages. Macrophages could internalize OSA-derived exosomes, which promoted macrophage polarization to the M1 type. Moreover, it inhibited sirtuin-3 (SIRT3)/AMP-activated protein kinase (AMPK) and autophagy and promoted the activation of nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasomes. The use of 3-methyladenine (3-MA) to inhibit autophagy blocked NLRP3 inflammasome activation and inhibited the M1 polarization of macrophages. miR-421 targeting inhibited SIRT3 protein expression in the macrophages. miR-421 was significantly increased in OSA-derived exosomes. Additionally, miR-421 levels were increased in OSA + NAFLD mice- and patient-derived exosomes. In the liver tissues of OSA and OSA + NAFLD mice, miR-421 displayed similar co-localization with the macrophages. Intermittent hypoxia-induced hepatocytes deliver miR-421 to the macrophages via exosomes to inhibit SIRT3, thereby participating in macrophage M1 polarization. After OSA and NAFLD modeling in miR-421-/- mice, liver steatosis and M1 polarization were significantly reduced. Additionally, in the case of miR-421 knockout, the inhibitory effects of OSA-derived exosomes on SIRT3 and autophagy were significantly alleviated. Furthermore, their effects on liver steatosis and macrophage M1 polarization were significantly reduced. CONCLUSIONS OSA promotes the delivery of miR-421 from the hepatocytes to macrophages. Additionally, it promotes M1 polarization by regulating the SIRT3/AMPK-autophagy pathway, thereby causing NAFLD.
Collapse
Affiliation(s)
- Li Yang
- Hypertension Center, Yan 'an Hospital of Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China.
- Kunming Technical Diagnosis and Treatment Center for Refractory Hypertension, Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China.
| | - Shijie Liu
- Hypertension Center, Yan 'an Hospital of Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
- Kunming Technical Diagnosis and Treatment Center for Refractory Hypertension, Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
| | - Yan He
- Hypertension Center, Yan 'an Hospital of Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
- Kunming Technical Diagnosis and Treatment Center for Refractory Hypertension, Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
| | - Lulu Gan
- Hypertension Center, Yan 'an Hospital of Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
- Kunming Technical Diagnosis and Treatment Center for Refractory Hypertension, Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
| | - Qing Ni
- Hypertension Center, Yan 'an Hospital of Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
- Kunming Technical Diagnosis and Treatment Center for Refractory Hypertension, Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
| | - Anni Dai
- Hypertension Center, Yan 'an Hospital of Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
- Kunming Technical Diagnosis and Treatment Center for Refractory Hypertension, Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
| | - Changhuan Mu
- Hypertension Center, Yan 'an Hospital of Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
- Kunming Technical Diagnosis and Treatment Center for Refractory Hypertension, Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
| | - Qian Liu
- Hypertension Center, Yan 'an Hospital of Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
- Kunming Technical Diagnosis and Treatment Center for Refractory Hypertension, Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
| | - Hongyan Chen
- Hypertension Center, Yan 'an Hospital of Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
- Kunming Technical Diagnosis and Treatment Center for Refractory Hypertension, Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
| | - Hongying Lu
- Hypertension Center, Yan 'an Hospital of Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
- Kunming Technical Diagnosis and Treatment Center for Refractory Hypertension, Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
| | - Ruixue Sun
- Hypertension Center, Yan 'an Hospital of Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
- Kunming Technical Diagnosis and Treatment Center for Refractory Hypertension, Kunming Medical University, 245 Renmin East Road, Panlong District, Kunming City, 650000, Yunnan Province, China
| |
Collapse
|
37
|
Chen X, Lu T, Zheng Y, Lin Z, Liu C, Yuan D, Yuan C. miR-155-5p promotes hepatic steatosis via PICALM-mediated autophagy in aging hepatocytes. Arch Gerontol Geriatr 2024; 120:105327. [PMID: 38237377 DOI: 10.1016/j.archger.2024.105327] [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: 11/19/2023] [Revised: 12/31/2023] [Accepted: 01/08/2024] [Indexed: 03/10/2024]
Abstract
BACKGROUND Hepatic steatosis, a lipid disorder characterized by the accumulation of intrahepatic fat, is more prevalent in the elderly population. This study investigates the role of miR-155-5p in the autophagy dysregulation of aging hepatic steatosis. METHODS We established an aging mouse model in vivo and a hepatocellular senescence model induced by low serum and palmitic acid in vitro. The fluctuations of microRNAs were derived from RNA-seq data and confirmed by qPCR in 4- and 18-month-old mouse liver tissues. Hematoxylin-eosin (H&E) staining observed pathological changes. Markers of senescence, autophagy, and lipolysis genes were analyzed using Western blot and qPCR. Bioinformatics analysis predicted miR-155-5p's target gene PICALM, confirmed by dual luciferase reporter assay and transfection of miR-155-5p mimic/inhibitor into senescent hepatocytes. RESULTS Senescent markers (p21, p16, and p-P53) and miR-155-5p were up-regulated in aging liver tissues and senescent hepatocytes. Bioinformatics analysis identified PICALM as a target gene of miR-155-5p, a finding further supported by dual luciferase reporter assays. Inhibition of miR-155-5p reduced expression of senescent marker genes (p16, p21, p-P53), improved autophagy (evidenced by increased LC3B-II and ATG5, and decreased P62), and enhanced lipolysis (indicated by increased ATGL and p-HSL) in senescent hepatocytes. Oil red O staining confirmed that miR-155-5p inhibition significantly reduced lipid accumulation in these cells. CONCLUSIONS This study suggests a potential new therapeutic approach for age-related hepatic steatosis through the inhibition of miR-155-5p to enhance autophagy.
Collapse
Affiliation(s)
- Xiaoling Chen
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, HuBei, 443002, China
| | - Ting Lu
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, HuBei, 443002, China
| | - Ying Zheng
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, HuBei, 443002, China
| | - Zhiyong Lin
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, HuBei, 443002, China
| | - Chaoqi Liu
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, HuBei, 443002, China.
| | - Ding Yuan
- College of Medicine and Health Science, China Three Gorges University, Yichang, HuBei, 443002, China.
| | - Chengfu Yuan
- College of Basic Medical Science, China Three Gorges University, Yichang, HuBei, 443002, China; Third Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, China Three Gorges University, School of Medicine, Yichang, 443002, China.
| |
Collapse
|
38
|
Xu X, Wang J, Xia Y, Yin Y, Zhu T, Chen F, Hai C. Autophagy, a double-edged sword for oral tissue regeneration. J Adv Res 2024; 59:141-159. [PMID: 37356803 PMCID: PMC11081970 DOI: 10.1016/j.jare.2023.06.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/10/2023] [Accepted: 06/20/2023] [Indexed: 06/27/2023] Open
Abstract
BACKGROUND Oral health is of fundamental importance to maintain systemic health in humans. Stem cell-based oral tissue regeneration is a promising strategy to achieve the recovery of impaired oral tissue. As a highly conserved process of lysosomal degradation, autophagy induction regulates stem cell function physiologically and pathologically. Autophagy activation can serve as a cytoprotective mechanism in stressful environments, while insufficient or over-activation may also lead to cell function dysregulation and cell death. AIM OF REVIEW This review focuses on the effects of autophagy on stem cell function and oral tissue regeneration, with particular emphasis on diverse roles of autophagy in different oral tissues, including periodontal tissue, bone tissue, dentin pulp tissue, oral mucosa, salivary gland, maxillofacial muscle, temporomandibular joint, etc. Additionally, this review introduces the molecular mechanisms involved in autophagy during the regeneration of different parts of oral tissue, and how autophagy can be regulated by small molecule drugs, biomaterials, exosomes/RNAs or other specific treatments. Finally, this review discusses new perspectives for autophagy manipulation and oral tissue regeneration. KEY SCIENTIFIC CONCEPTS OF REVIEW Overall, this review emphasizes the contribution of autophagy to oral tissue regeneration and highlights the possible approaches for regulating autophagy to promote the regeneration of human oral tissue.
Collapse
Affiliation(s)
- Xinyue Xu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, PR China; Shaanxi Key Lab of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, PR China
| | - Jia Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
| | - Yunlong Xia
- Shaanxi Key Lab of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, PR China; Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Yuan Yin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
| | - Tianxiao Zhu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, PR China; Shaanxi Key Lab of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, PR China
| | - Faming Chen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases and Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
| | - Chunxu Hai
- Shaanxi Key Lab of Free Radical Biology and Medicine, Fourth Military Medical University, Xi'an, PR China.
| |
Collapse
|
39
|
Mao S, Wang X, Li M, Liu H, Liang H. The role and mechanism of hydrogen sulfide in liver fibrosis. Nitric Oxide 2024; 145:41-48. [PMID: 38360133 DOI: 10.1016/j.niox.2024.02.002] [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: 08/21/2023] [Revised: 01/20/2024] [Accepted: 02/12/2024] [Indexed: 02/17/2024]
Abstract
Hydrogen sulfide (H2S) is the third new gas signaling molecule in the human body after the discovery of NO and CO. Similar to NO, it has the functions of vasodilation, anti-inflammatory, antioxidant, and regulation of cell formation. Enzymes that can produce endogenous H2S, such as CSE, CSB, and 3-MST, are common in liver tissues and are important regulatory molecules in the liver. In the development of liver fibrosis, H2S concentration and expression of related enzymes change significantly, which makes it possible to use exogenous gases to treat liver diseases. This review summarizes the role of H2S in liver fibrosis and its complications induced by NAFLD and CCl4, and elaborates on the anti-liver fibrosis effect of H2S through the mechanism of reducing oxidative stress, inhibiting inflammation, regulating autophagy, regulating glucose and lipid metabolism, providing theoretical reference for further research on the treatment of liver fibrosis with H2S.
Collapse
Affiliation(s)
- Shaoyu Mao
- Department of Infectious Disease and Hepatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xuemei Wang
- Department of Infectious Disease and Hepatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Miaoqing Li
- Department of Infectious Disease and Hepatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hanshu Liu
- Department of Infectious Disease and Hepatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hongxia Liang
- Department of Infectious Disease and Hepatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
40
|
Yang X, Xu J, Xu Y, Wang C, Lin F, Yu J. Regulatory mechanism of perinatal nonylphenol exposure on cardiac mitochondrial autophagy and the PINK1/Parkin signaling pathway in male offspring rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 126:155434. [PMID: 38367424 DOI: 10.1016/j.phymed.2024.155434] [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: 09/09/2023] [Revised: 01/24/2024] [Accepted: 02/06/2024] [Indexed: 02/19/2024]
Abstract
OBJECTIVE This study investigated whether perinatal exposure to nonylphenol (NP) induces mitochondrial autophagy (i.e., mitophagy) damage in neonatal rat cardiomyocytes (NRCMs) and whether the PINK1/Parkin signaling pathway is involved in NP-induced primary cardiomyocyte injury. METHODS AND RESULTS In vivo: Perinatal NP exposure increased apoptosis and mitochondrial damage in NRCMs. Mitochondrial swelling and autophagosome-like structures with multiple concentric membranes were observed in the 100 mg/kg NP group, with an increase in the number of autophagosomes. Disorganized fiber arrangement and elevated serum myocardial enzyme levels were observed with increasing NP dosage. Additionally, NP exposure led to increased MDA levels and decreased SOD activity and ATP levels in myocardial tissue. The mRNA expression levels of autophagy-related genes, including Beclin-1, p62, and LC3B, as well as the expression of mitochondrial autophagy-related proteins (PINK1, p-Parkin, Parkin, Beclin-1, p62, LC3-I, LC3-II, and LC3-II/I) and apoptosis-related proteins (Bax and caspase-3), increased, whereas the expression levels of the mitochondrial membrane protein TOMM20 and the anti-apoptotic protein Bcl-2 decreased. In vitro: NP increased ROS levels, LDH release, and decreased ATP levels in NRCMs. CsA treatment significantly inhibited the expression of autophagy-related proteins (Beclin-1, LC3-II/I, and p62) and apoptosis-related proteins (caspase-3 and Bax), increased the expression levels of TOMM20 and Bcl-2 proteins, increased cellular ATP levels, and inhibited LDH release. The inhibition of the PINK1/Parkin signaling pathway suppressed the expression of mitochondrial autophagy-related proteins (PINK1, p-Parkin, Parkin, Beclin-1, LC3-II/I, and p62) and apoptosis-related proteins (caspase-3 and Bax), increased TOMM20 and Bcl-2 protein expression, increased ATP levels, and decreased LDH levels in NRCMs. CONCLUSIONS This study is novel in reporting that perinatal NP exposure induced myocardial injury in male neonatal rats, thereby inducing mitophagy. The PINK1/Parkin signaling pathway was involved in this injury by regulating mitophagy.
Collapse
Affiliation(s)
- Xiaolian Yang
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Jie Xu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Yuzhu Xu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Chengxing Wang
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Fangmei Lin
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Jie Yu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| |
Collapse
|
41
|
Du H, Yu H, Zhou M, Hui Q, Hou Y, Jiang Y. The effect of STAT1, miR-99b, and MAP2K1 in alcoholic liver disease (ALD) mouse model and hepatocyte. Aging (Albany NY) 2024; 16:4224-4235. [PMID: 38431286 PMCID: PMC10968706 DOI: 10.18632/aging.205579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 01/24/2024] [Indexed: 03/05/2024]
Abstract
Alcoholic liver disease (ALD) serves as the leading cause of chronic liver diseases-related morbidity and mortality, which threatens the life of millions of patients in the world. However, the molecular mechanisms underlying ALD progression remain unclear. Here, we applied microarray analysis and experimental approaches to identify miRNAs and related regulatory signaling that associated with ALD. Microarray analysis identified that the expression of miR-99b was elevated in the ALD mouse model. The AML-12 cells were treated with EtOH and the expression of miR-99b was enhanced in the cells. The expression of miR-99b was positively correlated with ALT levels in the ALD mice. The microarray analysis identified the abnormally expressed mRNAs in ALD mice and the overlap analysis was performed with based on the differently expressed mRNAs and the transcriptional factors of miR-99b, in which STAT1 was identified. The elevated expression of STAT1 was validated in ALD mice. Meanwhile, the treatment of EtOH induced the expression of STAT1 in the AML-12 cells. The expression of STAT1 was positively correlated with ALT levels in the ALD mice. The positive correlation of STAT1 and miR-99b expression was identified in bioinformatics analysis and ALD mice. The expression of miR-99b and pri-miR-99b was promoted by the overexpression of STAT1 in AML-12 cells. ChIP analysis confirmed the enrichment of STAT1 on miR-99b promoter in AML-12 cells. Next, we found that the expression of mitogen-activated protein kinase kinase 1 (MAP2K1) was negatively associated with miR-99b. The expression of MAP2K1 was downregulated in ALD mice. Consistently, the expression of MAP2K1 was reduced by the treatment of EtOH in AML-12 cells. The expression of MAP2K1 was negative correlated with ALT levels in the ALD mice. We identified the binding site of MAP2K1 and miR-99b. Meanwhile, the treatment of miR-99b mimic repressed the luciferase activity of MAP2K1 in AML-12 cells. The expression of MAP2K1 was suppressed by miR-99b in the cells. We observed that the expression of MAP2K1 was inhibited by the overexpression of STAT1 in AML-12 cells. Meanwhile, the apoptosis of AML-12 cells was induced by the treatment of EtOH, while miR-99b mimic promoted but the overexpression of MAP2K1 attenuated the effect of EtOH in the cells. In conclusion, we identified the correlation and effect of STAT1, miR-99b, and MAP2K1 in ALD mouse model and hepatocyte. STAT1, miR-99b, and MAP2K1 may serve as potential therapeutic target of ALD.
Collapse
Affiliation(s)
- Hongbo Du
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100015, China
| | - Hao Yu
- Beijing Ditan Hospital Capital Medical University, Beijing 100015, China
| | - Meiyue Zhou
- Beijing Ditan Hospital Capital Medical University, Beijing 100015, China
| | - Quan Hui
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100015, China
| | - Yixin Hou
- Beijing Ditan Hospital Capital Medical University, Beijing 100015, China
| | - Yuyong Jiang
- Beijing Ditan Hospital Capital Medical University, Beijing 100015, China
| |
Collapse
|
42
|
Chen H, Hinz K, Zhang C, Rodriguez Y, Williams SN, Niu M, Ma X, Chao X, Frazier AL, McCarson KE, Wang X, Peng Z, Liu W, Ni HM, Zhang J, Swerdlow RH, Ding WX. Late-Life Alcohol Exposure Does Not Exacerbate Age-Dependent Reductions in Mouse Spatial Memory and Brain TFEB Activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.23.581774. [PMID: 38464149 PMCID: PMC10925107 DOI: 10.1101/2024.02.23.581774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Alcohol consumption is believed to affect Alzheimer's disease (AD) risk, but the contributing mechanisms are not well understood. A potential mediator of the proposed alcohol-AD connection is autophagy, a degradation pathway that maintains organelle and protein homeostasis. Autophagy is in turn regulated through the activity of Transcription factor EB (TFEB), which promotes lysosome and autophagy-related gene expression. To explore the effect of alcohol on brain TFEB and autophagy, we exposed young (3-month old) and aged (23-month old) mice to two alcohol-feeding paradigms and assessed biochemical, transcriptome, histology, and behavioral endpoints. In young mice, alcohol decreased hippocampal nuclear TFEB staining but increased SQSTM1/p62, LC3-II, ubiquitinated proteins, and phosphorylated Tau. Hippocampal TFEB activity was lower in aged mice than it was in young mice, and Gao-binge alcohol feeding did not worsen the age-related reduction in TFEB activity. To better assess the impact of chronic alcohol exposure, we fed young and aged mice alcohol for four weeks before completing Morris Water and Barnes Maze spatial memory testing. The aged mice showed worse spatial memory on both tests. While alcohol feeding slightly impaired spatial memory in the young mice, it had little effect or even slightly improved spatial memory in the aged mice. These findings suggest that aging is a far more important driver of spatial memory impairment and reduced autophagy flux than alcohol consumption.
Collapse
|
43
|
Barletta B, Corinti S, Maranghi F, Tait S, Tassinari R, Martinelli A, Longo A, Longo V, Colombo P, Di Felice G, Butteroni C. The environmental pollutant BDE-47 modulates immune responses in invitro and in vivo murine models. CHEMOSPHERE 2024; 349:140739. [PMID: 38000557 DOI: 10.1016/j.chemosphere.2023.140739] [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: 07/10/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023]
Abstract
2,2',4,4'-tetra-bromodiphenyl ether (BDE-47) is widespread in the environment and biological samples. Its association with health risks is an increasing concern, yet information on BDE-47 immunotoxicity remains limited. This study investigated the impact of BDE-47 on innate and adaptive immune responses through in vitro and in vivo approaches. BDE-47's capacity to directly induce cell responses and modulate responses induced by known stimuli was studied in vitro using the RAW 264.7 murine macrophage cell line and spleen-derived lymphocytes, and in vivo using keyhole limpet hemocyanin (KLH)-immunized BALB/c mice orally administered (28 d) at dose levels (7.5, 15.0 and 30 mg/kg/bw/d) derived from relevant toxicokinetic data from rodent models. RAW 264.7 cells stimulated with lipopolysaccharide (LPS) and exposed to BDE-47 exhibited unchanged cell viability but decreased release of interleukin (IL)-6. Primary splenocytes from naïve mice stimulated with anti-CD3/anti-CD28 antibodies and exposed to BDE-47 showed a significant decrease of IL-17 A and IFNγ production. In vivo data showed that BDE-47 significantly reduced the KLH-specific antibody response. A generally decreasing trend of IFNγ, IL-10 and IL-5 production was observed after in vitro antigen-specific restimulation of spleen cells. Histopathological effects on liver, spleen, small intestine and thyroid were detected at the highest dose in the absence of general toxicity. In addition, the expression of Mm_mir155 and Mm_let7a was induced in livers of exposed mice. The data obtained in this study suggest that exposure to BDE-47 may perturb innate and adaptive immune responses, thus possibly decreasing resistance to bacterial and viral infections.
Collapse
Affiliation(s)
- Bianca Barletta
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy.
| | - Silvia Corinti
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy.
| | - Francesca Maranghi
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Sabrina Tait
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Roberta Tassinari
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Andrea Martinelli
- Center for Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy.
| | - Alessandra Longo
- Institute for Biomedical Research and Innovation, National Research Council (IRIB-CNR), Palermo, Italy.
| | - Valeria Longo
- Institute for Biomedical Research and Innovation, National Research Council (IRIB-CNR), Palermo, Italy.
| | - Paolo Colombo
- Institute for Biomedical Research and Innovation, National Research Council (IRIB-CNR), Palermo, Italy.
| | - Gabriella Di Felice
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy.
| | - Cinzia Butteroni
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
44
|
Ortega-Ribera M, Babuta M, Szabo G. Sinusoidal cell interactions—From soluble factors to exosomes. SINUSOIDAL CELLS IN LIVER DISEASES 2024:23-52. [DOI: 10.1016/b978-0-323-95262-0.00002-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
45
|
Le TV, Truong NH, Holterman AXL. Autophagy modulates physiologic and adaptive response in the liver. LIVER RESEARCH (BEIJING, CHINA) 2023; 7:304-320. [PMID: 39958781 PMCID: PMC11792069 DOI: 10.1016/j.livres.2023.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/20/2023] [Accepted: 11/14/2023] [Indexed: 01/03/2025]
Abstract
Autophagy is a physiological process that is ubiquitous and essential to the disposal or recycling of damaged cellular organelles and misfolded proteins to maintain organ homeostasis and survival. Its importance in the regulation of liver function in normal and pathological conditions is increasingly recognized. This review summarizes how autophagy regulates epithelial cell- and non-epithelial cell-specific function in the liver and how it differentially participates in hepatic homeostasis, hepatic injury response to stress-induced liver damage such as cholestasis, sepsis, non-alcoholic and alcohol-associated liver disease, viral hepatitis, hepatic fibrosis, hepatocellular and cholangiocellular carcinoma, and aging. Autophagy-based interventional studies for liver diseases that are currently registered in clinicatrials.gov are summarized. Given the broad and multidirectional autophagy response in the liver, a more refined understanding of the liver cell-specific autophagy activities in a context-dependent manner is necessary.
Collapse
Affiliation(s)
- Trinh Van Le
- Laboratory of Stem Cell Research and Application, University of Science-VNUHCM, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
| | - Nhung Hai Truong
- Faculty of Biology and Biotechnology, University of Science-VNUHCM, Ho Chi Minh City, Vietnam
| | - Ai Xuan L. Holterman
- Department of Pediatrics and Surgery, University of Illinois College of Medicine, Chicago and Peoria, IL, USA
| |
Collapse
|
46
|
Yang L, Gao X, Tian D, Yang W, Xue S, Cao Z, Sun T. Resolvin D2 activates anti-inflammatory microglia via restoring autophagy flux and alleviate neuropathic pain following spinal cord injury in rats. Exp Neurol 2023; 370:114573. [PMID: 37858697 DOI: 10.1016/j.expneurol.2023.114573] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/30/2023] [Accepted: 10/14/2023] [Indexed: 10/21/2023]
Abstract
Spinal cord injury (SCI) is a fatal and intractable disease accompanied by the comorbidity of chronic neuropathic pain. Here, we purposed to explore the therapeutic effect and the underlying mechanism of Resolvin D2 (RvD2) on neuropathic pain after SCI. The in vivo model of traumatic SCI rats was established. Primary microglia isolated from neonatal rats were induced by TNF-α in vitro. The locomotor ability was assessed by the Basso-Beattie-Besnahan score. Hargreaves methods and Von Frey fibrofilaments were used to evaluate the symptoms of neuropathic pain including allodynia and hyperalgesia in rats. The cytotoxicity of RvD2 was evaluated by MTT assay. ELISA kit was applied to access the levels of inflammatory factors. And the expression levels of related mRNA and proteins were determined by qRT-PCR, western blotting and immunofluorescence staining. The targeting relationship between miR-155 and PTEN was verified by dual-luciferase reporter (DLR) assay. We found that RvD2 mitigated locomotor dysfunction, allodynia and hyperalgesia of SCI rats. In addition, RvD2 treatment suppressed pro-inflammatory phenotype but promoted anti-inflammatory differentiation in microglia. Furthermore, RvD2 treatment inhibited the upregulated expression level of miR-155 which was caused by NF-κB activation and then recovered the autophagy flux via targeting PTEN, thereby relieving the inflammatory response in the TNF-α-induced primary microglia. In summary, RvD2 treatment could recover the autophagy flux via suppressing NF-κB-modulated miR-155 expression to activate anti-inflammatory microglia and then inhibit the inflammatory response and even mitigate neuropathic pain following SCI.
Collapse
Affiliation(s)
- Lei Yang
- Department of Pain Management, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021,China; Department of Pain Management, Weihai Municipal Hospital, Shandong University, Weihai, Shandong 264200,China
| | - Xiaoming Gao
- Department of Pain Management, Weihai Municipal Hospital, Shandong University, Weihai, Shandong 264200,China
| | - Demin Tian
- Department of Pain Management, Weihai Municipal Hospital, Shandong University, Weihai, Shandong 264200,China
| | - Wenjie Yang
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Song Xue
- Department of Pain Management, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021,China
| | - Zhenxin Cao
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Tao Sun
- Department of Pain Management, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021,China; Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| |
Collapse
|
47
|
Xiang SY, Deng KL, Yang DX, Yang P, Zhou YP. Function of macrophage-derived exosomes in chronic liver disease: From pathogenesis to treatment. World J Hepatol 2023; 15:1196-1209. [DOI: 10.4254/wjh.v15.i11.1196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/09/2023] [Accepted: 10/23/2023] [Indexed: 11/24/2023] Open
Abstract
Chronic liver disease (CLD) imposes a heavy burden on millions of people worldwide. Despite substantial research on the pathogenesis of CLD disorders, no optimal treatment is currently available for some diseases, such as liver cancer. Exosomes, which are extracellular vesicles, are composed of various cellular components. Exosomes have unique functions in maintaining cellular homeostasis and regulating cell communication, which are associated with the occurrence of disease. Furthermore, they have application potential in diagnosis and treatment by carrying diverse curative payloads. Hepatic macrophages, which are key innate immune cells, show extraordinary heterogeneity and polarization. Hence, macrophage-derived exosomes may play a pivotal role in the initiation and progression of various liver diseases. This review focuses on the effects of macrophage-derived exosomes on liver disease etiology and their therapeutic potential, which will provide new insights into alleviating the global pressure of CLD.
Collapse
Affiliation(s)
- Shi-Yi Xiang
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang Province, China
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo 315020, Zhejiang Province, China
| | - Kai-Li Deng
- Health Science Center, Ningbo University, Ningbo 315211, Zhejiang Province, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Dong-Xue Yang
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo 315020, Zhejiang Province, China
- Institute of Digestive Disease of Ningbo University, Ningbo University, Ningbo 315020, Zhejiang Province, China
| | - Ping Yang
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo 315020, Zhejiang Province, China
| | - Yu-Ping Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo 315020, Zhejiang Province, China
- Institute of Digestive Disease of Ningbo University, Ningbo University, Ningbo 315020, Zhejiang Province, China
- Ningbo Key Laboratory of Translational Medicine Research on Gastroenterology and Hepatology, Ningbo Key Laboratory, Ningbo 315020, Zhejiang Province, China
| |
Collapse
|
48
|
Zhang W, Zhou R, Liu X, You L, Chen C, Ye X, Liu J, Liang Y. Key role of exosomes derived from M2 macrophages in maintaining cancer cell stemness (Review). Int J Oncol 2023; 63:126. [PMID: 37711063 PMCID: PMC10609468 DOI: 10.3892/ijo.2023.5574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/16/2023] [Indexed: 09/16/2023] Open
Abstract
Cancer stem cells (CSCs) constitute a specific subset of cells found within tumors that are responsible for initiating, advancing and resisting traditional cancer treatments. M2 macrophages, also known as alternatively activated macrophages, contribute to the development and progression of cancer through their involvement in promoting angiogenesis, suppressing the immune system, supporting tumor growth and facilitating metastasis. Exosomes, tiny vesicles released by cells, play a crucial role in intercellular communications and have been shown to be associated with cancer development and progression by influencing the immune response; thus, they may serve as markers for diagnosis and prognosis. Currently, investigating the impact of exosomes derived from M2 macrophages on the maintenance of CSCs is a crucial area of research with the aim of developing novel therapeutic strategies to target this process and improve outcomes for individuals with cancer. Understanding the biological functions of exosomes derived from M2 macrophages and their involvement in cancer may lead to the formulation of novel diagnostic tools and treatments for this disease. By targeting M2 macrophages and the exosomes they secrete, promising prospects emerge for cancer treatment, given their substantial contribution to cancer development and progression. Further research is required to fully grasp the intricate interactions between CSCs, M2 macrophages and exosomes in cancer, and to identify fresh targets for cancer therapy. The present review explores the pivotal roles played by exosomes derived from M2 cells in maintaining the stem‑like properties of cancer cells.
Collapse
Affiliation(s)
- Weiqiong Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Ruiping Zhou
- Department of Stomatology, Yantian District People's Hospital, Southern University of Science and Technology, Shenzhen, Guangdong 518081, P.R. China
| | - Xin Liu
- Department of Stomatology, Yantian District People's Hospital, Southern University of Science and Technology, Shenzhen, Guangdong 518081, P.R. China
| | - Lin You
- Department of Stomatology, Yantian District People's Hospital, Southern University of Science and Technology, Shenzhen, Guangdong 518081, P.R. China
| | - Chang Chen
- Department of Stomatology, Yantian District People's Hospital, Southern University of Science and Technology, Shenzhen, Guangdong 518081, P.R. China
| | - Xiaoling Ye
- Department of Stomatology, Yantian District People's Hospital, Southern University of Science and Technology, Shenzhen, Guangdong 518081, P.R. China
| | - Jie Liu
- Department of Stomatology, Yantian District People's Hospital, Southern University of Science and Technology, Shenzhen, Guangdong 518081, P.R. China
| | - Youde Liang
- Department of Stomatology, Yantian District People's Hospital, Southern University of Science and Technology, Shenzhen, Guangdong 518081, P.R. China
- Department of Stomatology, The People's Hospital of Baoan Shenzhen, Shenzhen, Guangdong 518081, P.R. China
| |
Collapse
|
49
|
Srinivas AN, Suresh D, Kaur S, Kumar DP. The promise of small particles: extracellular vesicles as biomarkers in liver pathology. J Physiol 2023; 601:4953-4971. [PMID: 35708653 DOI: 10.1113/jp283074] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/07/2022] [Indexed: 11/09/2022] Open
Abstract
Extracellular vesicles (EVs) are nanoscopic packages that are heterogeneous and bona fide players in hepatic physiology and pathology as they are involved in intercellular communication. EVs carrying bioactive cargoes rich in lipids, proteins or nucleic acids are implicated in the onset and progression of liver diseases. Liver pathology using liver biopsy has been assessed for several intricate conditions such as viral hepatitis, alcoholic and non-alcoholic fatty liver disease, hepatic malignancies and drug-induced liver injury. The lacunae, however, lie in early diagnosis and timely treatment of the above conditions, underscoring the need for non-invasive, accurate diagnostic tools that could replace the gold standard method of tissue biopsy. In this regard, EVs have emerged as promising candidates that could serve as potential biomarkers. In the last two decades, EVs, owing to their multifaceted charm in bringing out cell-free therapeutic responses and the ability of their cargoes to be applied to novel biomarkers, have drawn the great attention of researchers with the advancement and clinical application of liquid biopsy. In this review, we recapitulate the role of EVs and provide insights into the promising role of these small packages as biomarkers in liver pathology.
Collapse
Affiliation(s)
- Akshatha N Srinivas
- Department of Biochemistry, CEMR, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Diwakar Suresh
- Department of Biochemistry, CEMR, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences (ILBS), New Delhi, India
| | - Divya P Kumar
- Department of Biochemistry, CEMR, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru, Karnataka, India
| |
Collapse
|
50
|
Zhou Z, Zhang D, Wang Y, Liu C, Wang L, Yuan Y, Xu X, Jiang Y. Urinary exosomes: a promising biomarker of drug-induced nephrotoxicity. Front Med (Lausanne) 2023; 10:1251839. [PMID: 37809338 PMCID: PMC10556478 DOI: 10.3389/fmed.2023.1251839] [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: 07/02/2023] [Accepted: 09/11/2023] [Indexed: 10/10/2023] Open
Abstract
Drug-induced nephrotoxicity (DIN) is a big concern for clinical medication, but the clinical use of certain nephrotoxic drugs is still inevitable. Current testing methods make it hard to detect early renal injury accurately. In addition to understanding the pathogenesis and risk factors of drug-induced nephrotoxicity, it is crucial to identify specific renal injury biomarkers for early detection of DIN. Urine is an ideal sample source for biomarkers related to kidney disease, and urinary exosomes have great potential as biomarkers for predicting DIN, which has attracted the attention of many scholars. In the present paper, we will first introduce the mechanism of DIN and the biogenesis of urinary exosomes. Finally, we will discuss the changes in urinary exosomes in DIN and compare them with other predictive indicators to enrich and boost the development of biomarkers of DIN.
Collapse
Affiliation(s)
- Zunzhen Zhou
- Clinical Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Dailiang Zhang
- Clinical Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Yongjing Wang
- Clinical Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Chongzhi Liu
- Clinical Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Limei Wang
- Clinical Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Yi Yuan
- Orthopedic Department, Dazhou Integrated TCM and Western Medicine Hospital, Dazhou Second People’s Hospital, Dazhou, China
| | - Xiaodan Xu
- Clinical Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Yuan Jiang
- Clinical Medical College, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|