1
|
Xu X, Yang T, An J, Li B, Dou Z. Liver injury in sepsis: manifestations, mechanisms and emerging therapeutic strategies. Front Immunol 2025; 16:1575554. [PMID: 40226624 PMCID: PMC11985447 DOI: 10.3389/fimmu.2025.1575554] [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: 02/12/2025] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
Sepsis is defined as a condition related to infection that manifests with multiorgan dysfunction, representing a life-threatening state. Consequently, severe complications frequently occur, with liver injury being one of the most prevalent serious complications of sepsis. Liver dysfunction during sepsis serves as an independent predictor of mortality. This review provides a comprehensive overview of current research on sepsis-induced liver injury (SILI), encompassing the clinical manifestations, diagnostic criteria, pathogenesis and therapeutic strategies associated with this condition. SILI may manifest as hypoxic hepatitis due to ischemia and shock, cholestasis resulting from abnormal bile metabolism, or bile duct sclerosis. The pathophysiology of sepsis involves intricate interactions among the inflammatory response, oxidative stress, and cell death. All of these factors complicate treatment and represent potential targets for therapeutic intervention. Furthermore, this review addresses the limitations inherent in conventional therapies currently employed for managing SILI and emphasizes the potential of novel targeted strategies aimed at addressing the fundamental mechanisms underlying this condition.
Collapse
Affiliation(s)
- Xinqi Xu
- The First Hospital of Lanzhou University & The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Tingyu Yang
- The First Hospital of Lanzhou University & The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Jiapan An
- The First Hospital of Lanzhou University & The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Bin Li
- The First Hospital of Lanzhou University & The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Department of Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zhimin Dou
- The First Hospital of Lanzhou University & The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Department of Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
2
|
Li H, Chen C, Huang W, Shi L, Zhang Q, Zhou L, Huang H, Zhou S. Long-term expanded hepatic progenitor cells ameliorate D-GalN/LPS-induced acute liver failure through repolarizing M1 macrophage to M2-Like phenotype via activation of the IL-10/JAK2/STAT3 signaling pathway. Int Immunopharmacol 2024; 142:113127. [PMID: 39276457 DOI: 10.1016/j.intimp.2024.113127] [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: 05/02/2024] [Revised: 08/25/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024]
Abstract
Acute liver failure (ALF) is a devastating liver disease characterized by the rapid deterioration of hepatocytes, which causes a series of clinical complications, including hepatic dysfunction, coagulopathy, encephalopathy, and multiorgan failure. Cell-based therapy is a promising alternative as it can bridge patients until their livers regenerate, releasing immunomodulatory molecules to suppress inflammation. This study reports an iPSCs-derived long-term expanded hepatic progenitor cell (LTHepPCs), which can differentiate into hepatocyte-like cells (HLCs) in vivo. When introduced into drug-induced ALF models, LTHepPCs mitigate liver damage by modulating the local immune microenvironment. This is achieved by shifting macrophages/Kupffer cells towards an anti-inflammatory state, resulting in a decrease in the expression of inflammatory cytokines such as TNF-a, IL-1β, and IL-8, and an increase in the expression of anti-inflammatory cytokines such as IL-10 and ARG-1. In vitro co-culturing of THP-1 or mBMDMs with LTHepPCs suggested that LTHepPCs could activate the anti-inflammatory state of macrophages/Kupffer cells via the IL-10/JAK2/STAT3 signaling pathway. Therefore, LTHepPC transplantation is a promising therapy for ALF patients.
Collapse
Affiliation(s)
- Hongsheng Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Chen Chen
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Weijian Huang
- Celliver Biotechnology Inc., Shanghai, PR China; Department of Anesthesiology and Critical Care Medicine, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, PR China
| | - Lei Shi
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Qin Zhang
- Celliver Biotechnology Inc., Shanghai, PR China
| | - Li Zhou
- Celliver Biotechnology Inc., Shanghai, PR China
| | - Hai Huang
- Department of Urinary Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Road No.2, Shanghai, PR China.
| | - Shen'ao Zhou
- Celliver Biotechnology Inc., Shanghai, PR China; State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, CAS. Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, PR China.
| |
Collapse
|
3
|
Qian YY, Huang FF, Chen SY, Zhang WX, Wang Y, Du PF, Li G, Ding WB, Qian L, Zhan B, Chu L, Jiang DH, Yang XD, Zhou R. Therapeutic effect of recombinant Echinococcus granulosus antigen B subunit 2 protein on sepsis in a mouse model. Parasit Vectors 2024; 17:467. [PMID: 39548530 PMCID: PMC11566433 DOI: 10.1186/s13071-024-06540-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: 06/08/2024] [Accepted: 10/17/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Sepsis is a potentially fatal systemic inflammatory response syndrome (SIRS) that threatens millions of lives worldwide. Echinococcus granulosus antigen B (EgAgB) is a protein released by the larvae of the tapeworm. This protein has been shown to play an important role in modulating host immune response. In this study we expressed EgAgB as soluble recombinant protein in E. coli (rEgAgB) and explored its protective effect on sepsis. METHODS The sepsis model was established by cecal ligation and puncture (CLP) procedure in BALB/c mice. The therapeutic effect of rEgAgB on sepsis was performed by interperitoneally injecting 5 µg rEgAgB in mice with CLP-induced sepsis and observing the 72 h survival rate after onset of sepsis. The proinflammatory cytokines [tumor necrosis factor (TNF)-α, interleukin (IL)-6] and regulatory cytokines [IL-10, transforming growth factor beta (TGF-β)] were measured in sera, and the histopathological change was observed in livers, kidneys, and lungs of septic mice treated with rEgAgB compared with untreated mice. The effect of rEgAgB on the macrophage polarization was performed in vitro by incubating rEgAgB with peritoneal macrophages. The levels of TLR2 and MyD88 were measured in these tissues to determine the involvement of TLR-2/MyD88 in the sepsis-induced inflammatory signaling pathway. RESULTS In vivo, we observed that treatment with rEgAgB significantly increased the survival rate of mice with CLP-induced sepsis up to 72 h while all mice without treatment died within the same period. The increased survival was associated with reduced pathological damage in key organs such as liver, lung, and kidneys. It was supported by the reduced proinflammatory cytokine levels and increased regulatory cytokine expression in peripheral blood and key organ tissues. Further study identified that treatment with rEgAgB promoted macrophage polarization from classically activated macrophage (M1) to regulatory M2-like macrophage via inhibiting TLR2/MyD88 signal pathway. CONCLUSIONS The therapeutic effects of rEgAgB on mice with sepsis was observed in a mice model that was associated with reduced inflammatory responses and increased regulatory responses, possibly through inducing polarization of macrophages from proinflammatory M1 to regulatory M2 phenotype through inhibiting TLR2/MyD88 inflammatory pathway.
Collapse
Affiliation(s)
- Ya-Yun Qian
- First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, China
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu, 233000, China
- First People's Hospital of Changzhou, Changzhou, 213000, China
| | - Fei-Fei Huang
- First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, China
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu, 233000, China
| | - Si-Yu Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214028, China
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Wei-Xiao Zhang
- First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, China
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu, 233000, China
| | - Yin Wang
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Peng-Fei Du
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Gen Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu, 233000, China
| | - Wen-Bo Ding
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu, 233000, China
| | - Lei Qian
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu, 233000, China
| | - Bin Zhan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Liang Chu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu, 233000, China
- Second Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, China
| | - Dong-Hui Jiang
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China.
- Department of Critical Care Medicine, First People's Hospital of Haidong, Haidong, 810600, China.
| | - Xiao-Di Yang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu, 233000, China.
| | - Rui Zhou
- First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, China.
| |
Collapse
|
4
|
Jiang H, Guo Y, Wang Q, Wang Y, Peng D, Fang Y, Yan L, Ruan Z, Zhang S, Zhao Y, Zhang W, Shang W, Feng Z. The dysfunction of complement and coagulation in diseases: the implications for the therapeutic interventions. MedComm (Beijing) 2024; 5:e785. [PMID: 39445002 PMCID: PMC11496570 DOI: 10.1002/mco2.785] [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: 03/31/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
The complement system, comprising over 30 proteins, is integral to the immune system, and the coagulation system is critical for vascular homeostasis. The activation of the complement and coagulation systems involves an organized proteolytic cascade, and the overactivation of these systems is a central pathogenic mechanism in several diseases. This review describes the role of complement and coagulation system activation in critical illness, particularly sepsis. The complexities of sepsis reveal significant knowledge gaps that can be compared to a profound abyss, highlighting the urgent need for further investigation and exploration. It is well recognized that the inflammatory network, coagulation, and complement systems are integral mechanisms through which multiple factors contribute to increased susceptibility to infection and may result in a disordered immune response during septic events in patients. Given the overlapping pathogenic mechanisms in sepsis, immunomodulatory therapies currently under development may be particularly beneficial for patients with sepsis who have concurrent infections. Herein, we present recent findings regarding the molecular relationships between the coagulation and complement pathways in the advancement of sepsis, and propose potential intervention targets related to the crosstalk between coagulation and complement, aiming to provide more valuable treatment of sepsis.
Collapse
Affiliation(s)
- Honghong Jiang
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| | - Yiming Guo
- Department of Biological Science, The Dietrich School of Arts and SciencesUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Qihang Wang
- Department of Obstetrics and GynecologyThe Seventh Medical Center of Chinese PLA General HospitalBeijingChina
| | - Yiran Wang
- Department of Obstetrics and GynecologyThe sixth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Dingchuan Peng
- School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Yigong Fang
- Institute of Acupuncture and MoxibustionChina Academy of Chinese Medical SciencesBeijingChina
| | - Lei Yan
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| | - Zhuolin Ruan
- Department of Obstetrics and Gynecology,Chinese PLA General HospitalBeijingChina
| | - Sheng Zhang
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| | - Yong Zhao
- Department of Obstetrics and GynecologyThe Seventh Medical Center of Chinese PLA General HospitalBeijingChina
| | - Wendan Zhang
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| | - Wei Shang
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
- Department of Obstetrics and GynecologyThe Seventh Medical Center of Chinese PLA General HospitalBeijingChina
| | - Zhichun Feng
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| |
Collapse
|
5
|
Wen F, Yang G, Yu S, Liu H, Liao N, Liu Z. Mesenchymal stem cell therapy for liver transplantation: clinical progress and immunomodulatory properties. Stem Cell Res Ther 2024; 15:320. [PMID: 39334441 PMCID: PMC11438256 DOI: 10.1186/s13287-024-03943-6] [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: 07/05/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Although liver transplantation (LT) is an effective strategy for end-stage liver diseases, the shortage of donor organs and the immune rejection hinder its widespread implementation in clinical practice. Mesenchymal stem cells (MSCs) transplantation offers a promising approach for patients undergoing liver transplantation due to their immune regulatory capabilities, hepatic protection properties, and multidirectional differentiation potential. In this review, we summarize the potential applications of MSCs transplantation in various LT scenarios. MSCs transplantation has demonstrated effectiveness in alleviating hepatic ischemia-reperfusion injury, enhancing the viability of liver grafts, preventing acute graft-versus-host disease, and promoting liver regeneration in split LT therapy. We also discuss the clinical progress, and explore the immunomodulatory functions of MSCs in response to both adaptive and innate immune responses. Furthermore, we emphasize the interactions between MSCs and different immune cells, including T cells, B cells, plasma cells, natural killer cells, dendritic cells, Kupffer cells, and neutrophils, to provide new insights into the immunomodulatory properties of MSCs in adoptive cell therapy.
Collapse
Affiliation(s)
- Fuli Wen
- Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Guokai Yang
- Department of Nephrology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, P. R. China
| | - Saihua Yu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350028, P. R. China
| | - Haiyan Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350028, P. R. China
| | - Naishun Liao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350028, P. R. China.
| | - Zhengfang Liu
- Department of Traditional Chinese Medicine, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350028, P. R. China.
| |
Collapse
|
6
|
Tao Y, Wang Y, Wang M, Tang H, Chen E. Mesenchymal Stem Cells Alleviate Acute Liver Failure through Regulating Hepatocyte Apoptosis and Macrophage Polarization. J Clin Transl Hepatol 2024; 12:571-580. [PMID: 38974955 PMCID: PMC11224903 DOI: 10.14218/jcth.2023.00557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/06/2024] [Accepted: 03/18/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND AND AIMS Acute liver failure (ALF) is a life-threatening clinical problem with limited treatment options. Administration of human umbilical cord mesenchymal stem cells (hUC-MSCs) may be a promising approach for ALF. This study aimed to explore the role of hUC-MSCs in the treatment of ALF and the underlying mechanisms. METHODS A mouse model of ALF was induced by lipopolysaccharide and d-galactosamine administration. The therapeutic effects of hUC-MSCs were evaluated by assessing serum enzyme activity, histological appearance, and cell apoptosis in liver tissues. The apoptosis rate was analyzed in AML12 cells. The levels of inflammatory cytokines and the phenotype of RAW264.7 cells co-cultured with hUC-MSCs were detected. The C-Jun N-terminal kinase/nuclear factor-kappa B signaling pathway was studied. RESULTS The hUC-MSCs treatment decreased the levels of serum alanine aminotransferase and aspartate aminotransferase, reduced pathological damage, alleviated hepatocyte apoptosis, and reduced mortality in vivo. The hUC-MSCs co-culture reduced the apoptosis rate of AML12 cells in vitro. Moreover, lipopolysaccharide-stimulated RAW264.7 cells had higher levels of tumor necrosis factor-α, interleukin-6, and interleukin-1β and showed more CD86-positive cells, whereas the hUC-MSCs co-culture reduced the levels of the three inflammatory cytokines and increased the ratio of CD206-positive cells. The hUC-MSCs treatment inhibited the activation of phosphorylated (p)-C-Jun N-terminal kinase and p-nuclear factor-kappa B not only in liver tissues but also in AML12 and RAW264.7 cells co-cultured with hUC-MSCs. CONCLUSIONS hUC-MSCs could alleviate ALF by regulating hepatocyte apoptosis and macrophage polarization, thus hUC-MSC-based cell therapy may be an alternative option for patients with ALF.
Collapse
Affiliation(s)
- Yachao Tao
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Yonghong Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Menglan Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Enqiang Chen
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Soufihasanabad S, Mahmoudi M, Taghavi-Farahabadi M, Mirsanei Z, Mahmoudi Lamouki R, Mirza Abdalla JK, Babaei E, Hashemi SM. In vivo polarization of M2 macrophages by mesenchymal stem cell-derived extracellular vesicles: A novel approach to macrophage polarization and its potential in treating inflammatory diseases. Med Hypotheses 2024; 187:111353. [DOI: 10.1016/j.mehy.2024.111353] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
8
|
Shimizu J, Murao A, Lee Y, Aziz M, Wang P. Extracellular CIRP promotes Kupffer cell inflammatory polarization in sepsis. Front Immunol 2024; 15:1411930. [PMID: 38881891 PMCID: PMC11177612 DOI: 10.3389/fimmu.2024.1411930] [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: 04/03/2024] [Accepted: 05/16/2024] [Indexed: 06/18/2024] Open
Abstract
Introduction Sepsis is a life-threatening inflammatory condition caused by dysregulated host responses to infection. Extracellular cold-inducible RNA-binding protein (eCIRP) is a recently discovered damage-associated molecular pattern that causes inflammation and organ injury in sepsis. Kupffer cells can be activated and polarized to the inflammatory M1 phenotype, contributing to tissue damage by producing proinflammatory mediators. We hypothesized that eCIRP promotes Kupffer cell M1 polarization in sepsis. Methods We stimulated Kupffer cells isolated from wild-type (WT) and TLR4-/- mice with recombinant mouse (rm) CIRP (i.e., eCIRP) and assessed supernatant IL-6 and TNFα levels by ELISA. The mRNA expression of iNOS and CD206 for M1 and M2 markers, respectively, was assessed by qPCR. We induced sepsis in WT and CIRP-/- mice by cecal ligation and puncture (CLP) and assessed iNOS and CD206 expression in Kupffer cells by flow cytometry. Results eCIRP dose- and time-dependently increased IL-6 and TNFα release from WT Kupffer cells. In TLR4-/- Kupffer cells, their increase after eCIRP stimulation was prevented. eCIRP significantly increased iNOS gene expression, while it did not alter CD206 expression in WT Kupffer cells. In TLR4-/- Kupffer cells, however, iNOS expression was significantly decreased compared with WT Kupffer cells after eCIRP stimulation. iNOS expression in Kupffer cells was significantly increased at 20 h after CLP in WT mice. In contrast, Kupffer cell iNOS expression in CIRP-/- mice was significantly decreased compared with WT mice after CLP. CD206 expression in Kupffer cells was not different across all groups. Kupffer cell M1/M2 ratio was significantly increased in WT septic mice, while it was significantly decreased in CIRP-/- mice compared to WT mice after CLP. Conclusion Our data have clearly shown that eCIRP induces Kupffer cell M1 polarization via TLR4 pathway in sepsis, resulting in overproduction of inflammatory cytokines. eCIRP could be a promising therapeutic target to attenuate inflammation by preventing Kupffer cell M1 polarization in sepsis.
Collapse
Affiliation(s)
- Junji Shimizu
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Atsushi Murao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Yongchan Lee
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| |
Collapse
|
9
|
Liu S, Xie J, Duan C, Zhao X, Feng Z, Dai Z, Luo X, Li Y, Yang M, Zhuang R, Li J, Yin W. ADAR1 Inhibits Macrophage Apoptosis and Alleviates Sepsis-induced Liver Injury Through miR-122/BCL2A1 Signaling. J Clin Transl Hepatol 2024; 12:134-150. [PMID: 38343614 PMCID: PMC10851074 DOI: 10.14218/jcth.2023.00171] [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: 04/14/2023] [Revised: 08/18/2023] [Accepted: 10/09/2023] [Indexed: 01/05/2025] Open
Abstract
BACKGROUND AND AIMS As sepsis progresses, immune cell apoptosis plays regulatory roles in the pathogenesis of immunosuppression and organ failure. We previously reported that adenosine deaminases acting on RNA-1 (ADAR1) reduced intestinal and splenic inflammatory damage during sepsis. However, the roles and mechanism of ADAR1 in sepsis-induced liver injury remain unclear. METHODS We performed transcriptome and single-cell RNA sequencing of peripheral blood mononuclear cells (PBMCs) from patients with sepsis to investigate the effects of ADAR1 on immune cell activities. We also employed a cecal ligation and puncture (CLP) sepsis mouse model to evaluate the roles of ADAR1 in sepsis-induced liver injury. Finally, we treated murine RAW 264.7 macrophages with lipopolysaccharide to explore the underlying ADAR1-mediated mechanisms in sepsis. RESULTS PBMCs from patients with sepsis had obvious apoptotic morphological features. Single-cell RNA sequencing indicated that apoptosis-related pathways were enriched in monocytes, with significantly elevated ADAR1 and BCL2A1 expression in severe sepsis. CLP-induced septic mice had aggravated liver injury and Kupffer cell apoptosis that were largely alleviated by ADAR1 overexpression. ADAR1 directly bound to pre-miR-122 to modulate miR-122 biosynthesis. miR-122 was an upstream regulator of BCL2A1. Furthermore, ADAR1 also reduced macrophage apoptosis in mice with CLP-induced sepsis through the miR-122/BCL2A1 signaling pathway and protected against sepsis-induced liver injury. CONCLUSIONS The findings show that ADAR1 alleviates macrophage apoptosis and sepsis-induced liver damage through the miR-122/BCL2A1 signaling pathway. The study provides novel insights into the development of therapeutic interventions in sepsis.
Collapse
Affiliation(s)
- Shanshou Liu
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Jiangang Xie
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Chujun Duan
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Xiaojun Zhao
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Zhusheng Feng
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Zheng Dai
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Xu Luo
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yu Li
- Emergency Department, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Minghe Yang
- Third Student Brigade, School of Basic Medical Science, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Ran Zhuang
- Department of Immunology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Junjie Li
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Wen Yin
- Emergency Department, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
10
|
Pei L, Li R, Wang X, Xu D, Gong F, Chen W, Zheng X, Liu W, Zhao S, Wang Q, Mao E, Chen E, Chen Y, Yang Z. MSCs-derived extracellular vesicles alleviate sepsis-associated liver dysfunction by inhibiting macrophage glycolysis-mediated inflammatory response. Int Immunopharmacol 2024; 128:111575. [PMID: 38280334 DOI: 10.1016/j.intimp.2024.111575] [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: 11/29/2023] [Revised: 01/10/2024] [Accepted: 01/18/2024] [Indexed: 01/29/2024]
Abstract
Sepsis-associated liver dysfunction (SALD) aggravates the disease progression and prognosis of patients. Macrophages in the liver play a crucial role in the occurrence and development of SALD. Human umbilical cord mesenchymal stem cells (MSCs), by secreting extracellular vesicles (EVs), show beneficial effects in various inflammatory diseases. However, whether MSC-derived EVs (MSC-EVs) could ameliorate the inflammatory response in liver macrophages and the underlying mechanisms remain unclear. In this study, a mouse model of sepsis induced by lipopolysaccharide (LPS) challenge was used to investigate the immunomodulatory functions of MSC-EVs in SALD. LPS-stimulated primary Kupffer cells (KCs) and Raw264.7 were used to further explore the potential mechanisms of MSC-EVs in regulating the inflammatory response of macrophages. The results showed that MSC-EVs alleviated liver tissue injury and facilitated the polarization of M1 to M2 macrophages. Further in vitro studies confirmed that MSC-EVs treatment significantly downregulated the expression of several enzymes related to glycolysis and reduced the glycolytic flux by inhibiting hypoxia-inducible factor 1α (HIF-1α) expression, thus effectively inhibiting the inflammatory responses of macrophages. These findings reveal that the application of MSC-EVs might be a potential therapeutic strategy for treating SALD.
Collapse
Affiliation(s)
- Lei Pei
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ranran Li
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaofeng Wang
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Xu
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangchen Gong
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiwei Chen
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangtao Zheng
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenbin Liu
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shanzhi Zhao
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiuyun Wang
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Enqiang Mao
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Erzhen Chen
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ying Chen
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhitao Yang
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
11
|
Wang S, Jiang D, Huang F, Qian Y, Qi M, Li H, Wang X, Wang Z, Wang K, Wang Y, Du P, Zhan B, Zhou R, Chu L, Yang X. Therapeutic effect of Echinococcus granulosus cyst fluid on bacterial sepsis in mice. Parasit Vectors 2023; 16:450. [PMID: 38066526 PMCID: PMC10709918 DOI: 10.1186/s13071-023-06021-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/18/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND The primary pathophysiological process of sepsis is to stimulate a massive release of inflammatory mediators to trigger systemic inflammatory response syndrome (SIRS), the major cause of multi-organ dysfunction and death. Like other helminths, Echinococcus granulosus induces host immunomodulation. We sought to determine whether E. granulosus cyst fluid (EgCF) displays a therapeutic effect on sepsis-induced inflammation and tissue damage in a mouse model. METHODS The anti-inflammatory effects of EgCF were determined by in vitro culture with bone marrow-derived macrophages (BMDMs) and in vivo treatment of BALB/C mice with cecal ligation and puncture (CLP)-induced sepsis. The macrophage phenotypes were determined by flow cytometry, and the levels of cytokines in cell supernatants or in sera of mice were measured (ELISA). The therapeutic effect of EgCF on sepsis was evaluated by observing the survival rates of mice for 72 h after CLP, and the pathological injury to the liver, kidney, and lung was measured under a microscope. The expression of TLR-2/MyD88 in tissues was measured by western blot to determine whether TLR-2/MyD88 is involved in the sepsis-induced inflammatory signaling pathway. RESULTS In vitro culture with BMDMs showed that EgCF promoted macrophage polarization to M2 type and inhibited lipopolysaccharide (LPS)-induced M1 macrophages. EgCF treatment provided significant therapeutic effects on CLP-induced sepsis in mice, with increased survival rates and alleviation of tissue injury. The EgCF conferred therapeutic efficacy was associated with upregulated anti-inflammatory cytokines (IL-10 and TGF-β) and reduced pro-inflammatory cytokines (TNF-α and INF-γ). Treatment with EgCF induced Arg-1-expressed M2, and inhibited iNOS-expressed M1 macrophages. The expression of TLR-2 and MyD88 in EgCF-treated mice was reduced. CONCLUSIONS The results demonstrated that EgCF confers a therapeutic effect on sepsis by inhibiting the production of pro-inflammatory cytokines and inducing regulatory cytokines. The anti-inflammatory effect of EgCF is carried out possibly through inducing macrophage polarization from pro-inflammatory M1 to regulatory M2 phenotype to reduce excessive inflammation of sepsis and subsequent multi-organ damage. The role of EgCF in regulating macrophage polarization may be achieved by inhibiting the TLR2/MyD88 signaling pathway.
Collapse
Affiliation(s)
- Shuying Wang
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- Department of Pediatrics, Anqing First People's Hospital of Anhui Medical University, Anqing, 246000, China
| | - Donghui Jiang
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Feifei Huang
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Yayun Qian
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Meitao Qi
- Department of Pediatrics, Anqing First People's Hospital of Anhui Medical University, Anqing, 246000, China
| | - Huihui Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Xiaoli Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Zhi Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Kaigui Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Yin Wang
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Pengfei Du
- Department of Critical Care Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Bin Zhan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Rui Zhou
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Liang Chu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China.
- Second Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China.
| | - Xiaodi Yang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China.
| |
Collapse
|
12
|
Zhang W, Jiang H, Wu G, Huang P, Wang H, An H, Liu S, Zhang W. The pathogenesis and potential therapeutic targets in sepsis. MedComm (Beijing) 2023; 4:e418. [PMID: 38020710 PMCID: PMC10661353 DOI: 10.1002/mco2.418] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 10/01/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Sepsis is defined as "a life-threatening organ dysfunction caused by dysregulated host systemic inflammatory and immune response to infection." At present, sepsis continues to pose a grave healthcare concern worldwide. Despite the use of supportive measures in treating traditional sepsis, such as intravenous fluids, vasoactive substances, and oxygen plus antibiotics to eradicate harmful pathogens, there is an ongoing increase in both the morbidity and mortality associated with sepsis during clinical interventions. Therefore, it is urgent to design specific pharmacologic agents for the treatment of sepsis and convert them into a novel targeted treatment strategy. Herein, we provide an overview of the molecular mechanisms that may be involved in sepsis, such as the inflammatory response, immune dysfunction, complement deactivation, mitochondrial damage, and endoplasmic reticulum stress. Additionally, we highlight important targets involved in sepsis-related regulatory mechanisms, including GSDMD, HMGB1, STING, and SQSTM1, among others. We summarize the latest advancements in potential therapeutic drugs that specifically target these signaling pathways and paramount targets, covering both preclinical studies and clinical trials. In addition, this review provides a detailed description of the crosstalk and function between signaling pathways and vital targets, which provides more opportunities for the clinical development of new treatments for sepsis.
Collapse
Affiliation(s)
- Wendan Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Faculty of PediatricsNational Engineering Laboratory for Birth defects prevention and control of key technologyBeijing Key Laboratory of Pediatric Organ Failurethe Chinese PLA General HospitalBeijingChina
| | - Honghong Jiang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Faculty of PediatricsNational Engineering Laboratory for Birth defects prevention and control of key technologyBeijing Key Laboratory of Pediatric Organ Failurethe Chinese PLA General HospitalBeijingChina
| | - Gaosong Wu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Pengli Huang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Haonan Wang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Huazhasng An
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational MedicineThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanShandongChina
| | - Sanhong Liu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Weidong Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghaiChina
- The Research Center for Traditional Chinese MedicineShanghai Institute of Infectious Diseases and BiosecurityShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
13
|
Kholodenko IV, Yarygin KN. Hepatic Macrophages as Targets for the MSC-Based Cell Therapy in Non-Alcoholic Steatohepatitis. Biomedicines 2023; 11:3056. [PMID: 38002056 PMCID: PMC10669188 DOI: 10.3390/biomedicines11113056] [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: 10/15/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a serious public health issue associated with the obesity pandemic. Obesity is the main risk factor for the non-alcoholic fatty liver disease (NAFLD), which progresses to NASH and then to end-stage liver disease. Currently, there are no specific pharmacotherapies of NAFLD/NASH approved by the FDA or other national regulatory bodies and the treatment includes lifestyle adjustment and medicines for improving lipid metabolism, enhancing sensitivity to insulin, balancing oxidation, and counteracting fibrosis. Accordingly, further basic research and development of new therapeutic approaches are greatly needed. Mesenchymal stem cells (MSCs) and MSC-derived extracellular vesicles prevent induced hepatocyte death in vitro and attenuate NASH symptoms in animal models of the disease. They interact with hepatocytes directly, but also target other liver cells, including Kupffer cells and macrophages recruited from the blood flow. This review provides an update on the pathogenesis of NAFLD/NASH and the key role of macrophages in the development of the disease. We examine in detail the mechanisms of the cross-talk between the MSCs and the macrophages, which are likely to be among the key targets of MSCs and their derivatives in the course of NAFLD/NASH cell therapy.
Collapse
Affiliation(s)
- Irina V. Kholodenko
- Laboratory of Cell Biology, Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia;
| | | |
Collapse
|
14
|
Chen Y, Yang L, Li X. Advances in Mesenchymal stem cells regulating macrophage polarization and treatment of sepsis-induced liver injury. Front Immunol 2023; 14:1238972. [PMID: 37954578 PMCID: PMC10634316 DOI: 10.3389/fimmu.2023.1238972] [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: 07/23/2023] [Accepted: 10/17/2023] [Indexed: 11/14/2023] Open
Abstract
Sepsis is a syndrome of dysregulated host response caused by infection, which leads to life-threatening organ dysfunction. It is a familiar reason of death in critically ill patients. Liver injury frequently occurs in septic patients, yet the development of targeted and effective treatment strategies remains a pressing challenge. Macrophages are essential parts of immunity system. M1 macrophages drive inflammation, whereas M2 macrophages possess anti-inflammatory properties and contribute to tissue repair processes. Mesenchymal stem cells (MSCs), known for their remarkable attributes including homing capabilities, immunomodulation, anti-inflammatory effects, and tissue regeneration potential, hold promise in enhancing the prognosis of sepsis-induced liver injury by harmonizing the delicate balance of M1/M2 macrophage polarization. This review discusses the mechanisms by which MSCs regulate macrophage polarization, alongside the signaling pathways involved, providing an idea for innovative directions in the treatment of sepsis-induced liver injury.
Collapse
Affiliation(s)
- Yuhao Chen
- Department of Emergency Medicine, West China Second Hospital of Sichuan University, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Sichuan, China
| | - Lihong Yang
- Department of Emergency Medicine, West China Second Hospital of Sichuan University, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Sichuan, China
| | - Xihong Li
- Department of Emergency Medicine, West China Second Hospital of Sichuan University, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Sichuan, China
| |
Collapse
|
15
|
Chantree P, Tarasuk M, Prathaphan P, Ruangtong J, Jamklang M, Chumkiew S, Martviset P. Type I Cystatin Derived from Fasciola gigantica Suppresses Macrophage-Mediated Inflammatory Responses. Pathogens 2023; 12:pathogens12030395. [PMID: 36986318 PMCID: PMC10051455 DOI: 10.3390/pathogens12030395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 03/05/2023] Open
Abstract
There is an inverse relationship between the high incidence of helminth infection and the low incidence of inflammatory disease. Hence, it may be that helminth molecules have anti-inflammatory effects. Helminth cystatins are being extensively studied for anti-inflammatory potential. Therefore, in this study, the recombinant type I cystatin (stefin-1) of Fasciola gigantica (rFgCyst) was verified to have LPS-activated anti-inflammatory potential, including in human THP-1-derived macrophages and RAW 264.7 murine macrophages. The results from the MTT assay suggest that rFgCyst did not alter cell viability; moreover, it exerted anti-inflammatory activity by decreasing the production of proinflammatory cytokines and mediators, including IL-1β, IL-6, IL-8, TNF-α, iNOS, and COX-2 at the gene transcription and protein expression levels, as determined by qRT-PCR and Western blot analysis, respectively. Further, the secretion levels of IL-1β, IL-6, and TNF-α determined by ELISA and the NO production level determined by the Griess test were decreased. Furthermore, in Western blot analysis, the anti-inflammatory effects involved the downregulation of pIKKα/β, pIκBα, and pNF-κB in the NF-κB signaling pathway, hence reducing the translocation from the cytosol into the nucleus of pNF-κB, which subsequently turned on the gene of proinflammatory molecules. Therefore, cystatin type 1 of F. gigantica is a potential candidate for inflammatory disease treatment.
Collapse
Affiliation(s)
- Pathanin Chantree
- Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Thammasat University Research Unit in Nutraceuticals and Food Safety, Thammasat University, Pathumthani 12120, Thailand
- Research Group in Medical Biomolecules, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Mayuri Tarasuk
- Graduate Program in Bioclinical Sciences, Chulabhorn International College of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Parisa Prathaphan
- Thammasat University Research Unit in Nutraceuticals and Food Safety, Thammasat University, Pathumthani 12120, Thailand
| | - Jittiporn Ruangtong
- Thammasat University Research Unit in Nutraceuticals and Food Safety, Thammasat University, Pathumthani 12120, Thailand
| | - Mantana Jamklang
- Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Sirilak Chumkiew
- Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Pongsakorn Martviset
- Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Thammasat University Research Unit in Nutraceuticals and Food Safety, Thammasat University, Pathumthani 12120, Thailand
- Research Group in Medical Biomolecules, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Correspondence: ; Tel.: +66-863590511
| |
Collapse
|
16
|
Zhang L, Zhang X, Liu Y, Zhang W, Wu CT, Wang L. CD146+ Umbilical Cord Mesenchymal Stem Cells Exhibit High Immunomodulatory Activity and Therapeutic Efficacy in Septic Mice. J Inflamm Res 2023; 16:579-594. [PMID: 36818194 PMCID: PMC9930589 DOI: 10.2147/jir.s396088] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/18/2023] [Indexed: 02/12/2023] Open
Abstract
Background Several studies have shown that MSCs can significantly improve the survival of sepsis animals. CD146+ mesenchymal stem cells (MSCs) correlate with high therapeutic potency. However, their therapeutic effect on sepsis and detail mechanisms have not been explored. Methods The effect of CD146±MSCs on differentiation of Treg, Th1, Th17 subsets was evaluated by flow cytometry. The effects of CD146±MSCs on RAW264.7 phagocytosis and LPS-stimulated polarization were studied using a co-culture protocol. Luminex bead array and RNA sequencing were employed to determine the mechanisms of MSCs on LPS-stimulated RAW264.7. The Arg1 protein was detected by Western blot. CD146±MSCs were injected into LPS-induced sepsis mice by tail vein. The therapeutic effect was assessed by organ HE staining, T-cell subsets, cytokine in plasma, peritoneal macrophages, infiltrating monocytes subpopulations. Results In vitro, CD146+MSCs could significantly increase the proportion of Treg cells. Co-culture with CD146+MSCs increase the phagocytic rate of RAW264.7. CD146+MSCs regulate M2-type macrophages production more rapidly. The transcript profile differences between the CD146+MSCs and CD146-MSCs groups were clustered in arginine metabolism pathways. CD146+MSCs decreased NO production and increased ARG1 expression. CD146+MSCs secreted higher level of IL15,IFNγ, VEGF and lower level of IL1β, IL8 under LPS stimuli. In vivo, The level of IL10 at 24h and CXCL1, IFNγ at 12h in CD146+MSCs group was the highest. CD146+MSCs treatment enhances the phagocytic capacity of peritoneal macrophages. CD146+MSCs also increases the ratios of CD11b+Ly6Clo reparative monocytes and CD11b+Ly6Chi inflammatory monocytes until 24h. Conclusion Compared with CD146-MSCs, CD146+MSCs can accelerate the end of the inflammatory response and have robust anti-inflammatory effects, by increasing the Treg cells, promoting macrophage phagocytosis, enhancing the reparative macrophage, secreting more VEGF, etc.
Collapse
Affiliation(s)
- Lin Zhang
- Laboratory of Molecular Diagnosis and Regenerative Medicine, the Affiliate Hospital of Qingdao University, Qingdao, People’s Republic of China,Beijing Institute of Radiation Medicine, Beijing, People’s Republic of China
| | - Xiaoxu Zhang
- Laboratory of Molecular Diagnosis and Regenerative Medicine, the Affiliate Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Yubin Liu
- Beijing Institute of Radiation Medicine, Beijing, People’s Republic of China
| | - Weiyuan Zhang
- Laboratory of Molecular Diagnosis and Regenerative Medicine, the Affiliate Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Chu-Tse Wu
- Beijing Institute of Radiation Medicine, Beijing, People’s Republic of China
| | - Lisheng Wang
- Laboratory of Molecular Diagnosis and Regenerative Medicine, the Affiliate Hospital of Qingdao University, Qingdao, People’s Republic of China,Department of Rehabilitation Sciences, School of Nursing, Jilin University, Changchun, People’s Republic of China,Correspondence: Lisheng Wang, Laboratory of Molecular Diagnosis and Regenerative Medicine, the Affiliate Hospital of Qingdao University, Qingdao, 266000, People’s Republic of China, Email
| |
Collapse
|
17
|
Zhao X, Xue X, Cui Z, Kwame Amevor F, Wan Y, Fu K, Wang C, Peng C, Li Y. microRNAs-based diagnostic and therapeutic applications in liver fibrosis. WILEY INTERDISCIPLINARY REVIEWS. RNA 2022:e1773. [PMID: 36585388 DOI: 10.1002/wrna.1773] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 01/01/2023]
Abstract
Liver fibrosis is a process of over-extracellular matrix (ECM) aggregation and angiogenesis, which develops into cirrhosis and hepatocellular carcinoma (HCC). With the increasing pressure of liver fibrosis, new therapeutics to cure this disease requires much attention. Exosome-cargoed microRNAs (miRNAs) are emerging approaches in the precision of the liver fibrotic paradigm. In this review, we outlined the different types of hepatic cells derived miRNAs that drive intra-/extra-cellular interactive communication in liver fibrosis with different physiological and pathological processes. Specifically, we highlighted the possible mechanism of liver fibrosis pathogenesis associated with immune response and angiogenesis. In addition, potential clinical biomarkers and different stem cell transplant-derived miRNAs-based therapeutic strategies in liver fibrosis were summarized in this review. miRNAs-based approaches might help researchers devise new candidates for the cell-free treatment of liver fibrosis. This article is categorized under: RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Xingtao Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinyan Xue
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhifu Cui
- College Science and Technology, Southwest University, Chongqing, China
| | - Felix Kwame Amevor
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, China
| | - Yan Wan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ke Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
18
|
Ozkul O, Ozkul B, Erdogan MA, Erbas O. Ameliorating Effect of Propofol on Cisplatin-Induced Liver and Kidney Damage in Rats. INT J PHARMACOL 2022. [DOI: 10.3923/ijp.2022.1623.1635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
19
|
Fang H, Chen J, Luo J, Hu J, Wang D, Lv L, Zhang W. Abietic acid attenuates sepsis-induced lung injury by inhibiting nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway to inhibit M1 macrophage polarization. Exp Anim 2022; 71:481-490. [PMID: 35644586 PMCID: PMC9671762 DOI: 10.1538/expanim.22-0018] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/03/2022] [Indexed: 08/17/2024] Open
Abstract
Lung injury is one of the leading causes of death in sepsis. Abietic acid (AA) has demonstrated anti-inflammatory and bacteriostatic properties. Herein, we established a mouse model of sepsis by cecal ligation and puncture, and intraperitoneally injected AA to treat. Lung injury was assessed by H&E staining and the inflammation in bronchoalveolar lavage fluid (BALF) were assessed by counting the number of inflammatory cells and detecting the content of inflammatory factors. Meanwhile, we also designed to study the effect of AA on lipopolysaccharide (LPS)-induced inflammatory response and macrophage marker gene expression in RAW264.7 cells in vitro. In this study, we found that AA inhibited LPS-induced secretion of inflammatory mediators (IL-1β, TNF-α, IL-6 and MIP-2), and decreased the expression of M1 macrophage e markers (CD16 and iNOS) and p-p65 protein, while increased the expression of M2 macrophage markers (CD206 and Arg-1) in RAW264.7 cells in vitro. In vivo, the therapy of AA not only rescued septic animals, but also attenuated lung injury in sepsis mice. Moreover, AA decreased the number of total cells, neutrophils and macrophages, the conceration of total protein, and the levels of inflammatory mediators in BALF of sepsis mice. Further, we found that AA inhibited M1 macrophage polarization and blocked nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway in BALF of sepsis mice. In conclusion, Abietic acid attenuates sepsis-induced lung injury, and its mechanism may be related to reducing inflammation by inhibiting NF-κB signaling to inhibit M1 macrophage polarization.
Collapse
Affiliation(s)
- Honglong Fang
- Department of Critical Care Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou Zhejiang, 324000, P.R. China
| | - Juan Chen
- Department of Laboratory Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou Zhejiang, 324000, P.R. China
| | - Jian Luo
- Department of Critical Care Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou Zhejiang, 324000, P.R. China
| | - Jianhua Hu
- Department of Critical Care Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou Zhejiang, 324000, P.R. China
| | - Danqiong Wang
- Department of Critical Care Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou Zhejiang, 324000, P.R. China
| | - Liang Lv
- Department of Critical Care Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou Zhejiang, 324000, P.R. China
| | - Weiwen Zhang
- Department of Critical Care Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou Zhejiang, 324000, P.R. China
| |
Collapse
|
20
|
Li H, Qiu D, Yuan Y, Wang X, Wu F, Yang H, Wang S, Ma M, Qian Y, Zhan B, Yang X. Trichinella spiralis cystatin alleviates polymicrobial sepsis through activating regulatory macrophages. Int Immunopharmacol 2022; 109:108907. [PMID: 35691271 DOI: 10.1016/j.intimp.2022.108907] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/19/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Sepsis is a life-threateningorgandysfunction caused by the cytokine storm induced by the severe bacterial infection. Excessive inflammatory responses are responsible for the lethal organ damage during the early stage of sepsis. Helminth infection and helminth-derived proteins have been identified to have the ability to immunomodulate the host immune system by reducing inflammation against inflammatory diseases. Trichinella spiralis cystatin (Ts-Cys) is a cysteine protease inhibitor with strong immunomodulatory functions on host immune system. Our previous studies have shown that excretory-secretory proteins of T. spiralis reduced sepsis-induced inflammation and Ts-Cys was able to inhibit macrophages to produce inflammatory cytokines. Whether Ts-Cys has a therapeutic effect on polymicrobial sepsis and related immunological mechanism are not yet known. METHODS Sepsis was induced in BALB/c mice using cecal ligation and puncture (CLP), followed by intraperitoneal injection of 15 µg recombinant Ts-Cys (rTs-Cys). The therapeutic effect of rTs-Cys on sepsis was evaluated by observing the 72-hour survival rates of CLP-induced septic mice and the acute injury of lung and kidney through measuring the wet/dry weight ratio of lung, the levels of blood urea nitrogen (BUN) and creatinine (Cr) in sera and the tissue section pathology. The potential underlying mechanism was investigated using mouse bone marrow-derived macrophages (BMDMs) by observing the effect of rTs-Cys on LPS-stimulated macrophage polarization. The expression of genes associated with macrophage polarization in BMDMs and tissues of septic mice was measured by Western Blotting and qPCR. RESULTS In this study, we demonstrated the treatment with rTs-Cys alleviated CLP-induced sepsis in mice with significantly reduced pathological injury in vital organs of lung and kidney and reduced mortality of septic mice. The further study identified that treatment with rTs-Cys promoted macrophage polarization from classically activated macrophage (M1) to alternatively activated macrophage (M2) phenotype via inhibiting TLR2/MyD88 signal pathway and increasing expression of mannose receptor (MR), inhibited pro-inflammatory cytokines (TNF-α, IL-6 and IL-1β) and increased regulatory anti-inflammatory cytokines (IL-10 and TGF-β) in sera and tissues (lung and kidney) of mice with polymicrobial sepsis. CONCLUSIONS Our results demonstrated that rTs-Cys had a therapeutic effect on sepsis through activating regulatory macrophages possibly via suppressing TLR2/MyD88 signal pathway. We also identified that rTs-Cys-induced M2 macrophage differentiation was associated with increased expression of MR on the surface of macrophages. Our results underscored the importance of MR in regulating macrophages during the treatment with rTs-Cys, providing another immunological mechanism in which helminths and their derived proteins modulate the host immune system. The findings in this study suggest that rTs-Cys is a potential therapeutic agent for the prevention and treatment of sepsis and other inflammatory diseases.
Collapse
Affiliation(s)
- Huihui Li
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Dapeng Qiu
- Department of Orthopedics, Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yuan Yuan
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Xiaoli Wang
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Fengjiao Wu
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Huijuan Yang
- Department of Nephrology, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Shuying Wang
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Mengxi Ma
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Yayun Qian
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Bin Zhan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Xiaodi Yang
- Department of Basic Medical College, Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.
| |
Collapse
|
21
|
Extraembryonic Mesenchymal Stromal/Stem Cells in Liver Diseases: A Critical Revision of Promising Advanced Therapy Medicinal Products. Cells 2022; 11:cells11071074. [PMID: 35406638 PMCID: PMC8997603 DOI: 10.3390/cells11071074] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 02/04/2023] Open
Abstract
Liver disorders have been increasing globally in recent years. These diseases are associated with high morbidity and mortality rates and impose high care costs on the health system. Acute liver failure, chronic and congenital liver diseases, as well as hepatocellular carcinoma have been limitedly treated by whole organ transplantation so far. But novel treatments for liver disorders using cell-based approaches have emerged in recent years. Extra-embryonic tissues, including umbilical cord, amnion membrane, and chorion plate, contain multipotent stem cells. The pre-sent manuscript discusses potential application of extraembryonic mesenchymal stromal/stem cells, focusing on the management of liver diseases. Extra-embryonic MSC are characterized by robust and constitutive anti-inflammatory and anti-fibrotic properties, indicating as therapeutic agents for inflammatory conditions such as liver fibrosis or advanced cirrhosis, as well as chronic inflammatory settings or deranged immune responses.
Collapse
|
22
|
Wang L, Deng Z, Sun Y, Zhao Y, Li Y, Yang M, Yuan R, Liu Y, Qian Z, Zhou F, Kang H. The Study on the Regulation of Th Cells by Mesenchymal Stem Cells Through the JAK-STAT Signaling Pathway to Protect Naturally Aged Sepsis Model Rats. Front Immunol 2022; 13:820685. [PMID: 35197984 PMCID: PMC8858840 DOI: 10.3389/fimmu.2022.820685] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/17/2022] [Indexed: 12/16/2022] Open
Abstract
Sepsis is the leading cause of death among patients, especially elderly patients, in intensive care units worldwide. In this study, we established a sepsis model using naturally aged rats and injected 5×106 umbilical cord-derived MSCs via the tail vein. Each group of rats was analyzed for survival, examined for biochemical parameters, stained for organ histology, and analyzed for the Th cell subpopulation ratio and inflammatory cytokine levels by flow cytometry. Western blotting was performed to detect the activity of the JAK-STAT signaling pathway. We designed the vitro experiments to confirm the regulatory role of MSCs, and verified the possible mechanism using JAK/STAT inhibitors. It was revealed from the experiments that the 72 h survival rate of sepsis rats treated with MSCs was significantly increased, organ damage and inflammatory infiltration were reduced, the levels of organ damage indicators were decreased, the ratios of Th1/Th2 and Th17/Treg in peripheral blood and spleen were significantly decreased, the levels of pro-inflammatory cytokines such as IL-6 were decreased, the levels of anti-inflammatory cytokines such as IL-10 were increased, and the levels of STAT1 and STAT3 phosphorylation were reduced. These results were validated in in vitro experiments. Therefore, this study confirms that MSCs can control the inflammatory response induced by sepsis by regulating Th cells and inflammatory factors, and that this leads to the reduction of tissue damage, protection of organ functions and ultimately the improvement of survival in aged sepsis model rats. Inhibition of the JAK-STAT signaling pathway was surmised that it may be an important mechanism for their action.
Collapse
Affiliation(s)
- Lu Wang
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
- Department of Critical Care Medicine, the First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zihui Deng
- Department of Basic Medicine, Graduate School, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yan Sun
- School of Public Health, Capital Medical University, Beijing, China
| | - Yan Zhao
- Department of Critical Care Medicine, the First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yun Li
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
- Department of Critical Care Medicine, the First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Mengmeng Yang
- Department of Critical Care Medicine, the First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Rui Yuan
- Department of Critical Care Medicine, the First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yuyan Liu
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
- Department of Critical Care Medicine, the First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhirong Qian
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Feihu Zhou
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
- Department of Critical Care Medicine, the First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Hongjun Kang
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
- Department of Critical Care Medicine, the First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- *Correspondence: Hongjun Kang,
| |
Collapse
|
23
|
Chen W, Liu Y, Chen J, Ma Y, Song Y, Cen Y, You M, Yang G. The Notch signaling pathway regulates macrophage polarization in liver diseases. Int Immunopharmacol 2021; 99:107938. [PMID: 34371331 DOI: 10.1016/j.intimp.2021.107938] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/16/2022]
Abstract
The liver is not only the main metabolic site of exogenous compounds and drugs, but also an important immune organ in the human body. When a large number of nonself substances (such as drugs, alcohol, pathogens, microorganisms and their metabolites) enter the liver, they will cause serious liver diseases, including liver fibrosis, liver cirrhosis, liver failure, and hepatocellular carcinoma (HCC). Macrophages are the first line of defense against the invasion of exogenous pathogens and significant cellular components of the innate immune system. Macrophages have strong heterogeneity and plasticity. When different pathogens invade the body, they cause different types of polarization of macrophages through different molecular mechanisms. Notch signaling is considered to be the key regulator of the biological function of macrophages. Activating Notch signaling can regulate the differentiation of macrophages into M1 and play a role in promoting inflammation and antitumor activity, while blocking Notch signaling can polarize macrophages to M2, suppressing inflammation and promoting tumor growth. However, there are few studies on regulation of macrophage polarization by the Notch signaling pathway in liver diseases. Therefore, in this review, we will introduce the role of the Notch signaling pathway in regulating macrophage polarization in liver diseases.
Collapse
Affiliation(s)
- Wenyan Chen
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Yining Liu
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Jing Chen
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Yemei Ma
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Yawen Song
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Yanli Cen
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Mingdan You
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Guanghong Yang
- Guizhou Provincial Center for Disease Control and Prevention, Guiyang, Guizhou 550004, China.
| |
Collapse
|
24
|
Hoseinnia S, Ghane M, Norouzi J, Hosseini F. Mesenchymal stem cell and endothelial progenitor cells coinjection improves LPS-induced lung injury via Tie2 activation and downregulation of the TLR4/MyD88 pathway. J Cell Biochem 2021; 122:1791-1804. [PMID: 34397115 DOI: 10.1002/jcb.30133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/15/2022]
Abstract
Sepsis is one of the most important complications of infection with a high mortality rate. Recently, cell therapy has been widely used to reduce the symptoms of sepsis. It has been previously reported that mesenchymal stem cell (MSC) and endothelial progenitor cells (EPC) therapy have beneficial effects in experimental models of sepsis. The effects of coculture of MSC and EPC have not yet been used to treat sepsis. Therefore, the aim of this study was to investigate the therapeutic potential of EPC + MSC coculture on the residual effects of sepsis in a lipopolysaccharide (LPS)-induced mice model. Coinjections of EPC + MSC significantly enhanced the survival rate of LPS-induced mice, decreased concentrations of pro-inflammatory cytokines, and increased the level of anti-inflammatory cytokine. The LPS-induced mice that were treated with EPC + MSC showed a notable reduction in pulmonary edema, hepatic enzymes, and C-reactive protein level compared with the control group. Our results showed that coinjection of EPC + MSC up and downregulates Tie2 and TLR4/MyD88 signaling pathways in LPS-induced mice, respectively. Also, in vitro study showed that viability, adhesion, and migration in coculture cells is significantly decreased after being induced with 10 μg/ml LPS. Our results showed that LPS impaired the functional activity of the cocultured EPC + MSC via upregulation of the TLR4/MyD88 signaling pathway, which may be associated with decreased pTie2/Tie2 expression. In conclusion, coinjection of EPC and MSC modulated the TLR4/MyD88 signaling pathway that leads to reduce the inflammatory response. This study may provide promising results for the introduction of cocultured cells to manage infectious diseases and balance the immune response through immune regulatory function.
Collapse
Affiliation(s)
- Sadaf Hoseinnia
- Department of Microbiology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Ghane
- Department of Biology, Islamshahr Branch, Islamic Azad University, Islamshahr, Iran
| | - Jamile Norouzi
- Department of Microbiology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Farzaneh Hosseini
- Department of Microbiology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
25
|
Pérez-Hernández EG, Delgado-Coello B, Luna-Reyes I, Mas-Oliva J. New insights into lipopolysaccharide inactivation mechanisms in sepsis. Biomed Pharmacother 2021; 141:111890. [PMID: 34229252 DOI: 10.1016/j.biopha.2021.111890] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/03/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
The complex pathophysiology of sepsis makes it a syndrome with limited therapeutic options and a high mortality rate. Gram-negative bacteria containing lipopolysaccharides (LPS) in their outer membrane correspond to the most common cause of sepsis. Since the gut is considered an important source of LPS, intestinal damage has been considered a cause and a consequence of sepsis. Although important in the maintenance of the intestinal epithelial cell homeostasis, the microbiota has been considered a source of LPS. Recent studies have started to shed light on how sepsis is triggered by dysbiosis, and an increased inflammatory state of the intestinal epithelial cells, expanding the understanding of the gut-liver axis in sepsis. Here, we review the gut-liver interaction in Gram-negative sepsis, exploring the mechanisms of LPS inactivation, including the recently described contribution of an isoform of the cholesteryl-ester transfer protein (CETPI). Although several key questions remain to be answered when the pathophysiology of sepsis is reviewed, new contributions coming to light exploring the way LPS might be inactivated in vivo, suggest that new applications might soon reach the clinical setting.
Collapse
Affiliation(s)
| | - Blanca Delgado-Coello
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico
| | - Ismael Luna-Reyes
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico
| | - Jaime Mas-Oliva
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico.
| |
Collapse
|
26
|
LIU L, YANG F. Application of Modified Mesenchymal Stem Cells Transplantation in the Treatment of Liver Injury. Physiol Res 2021. [DOI: 10.33549/physiolres.934623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Acute and chronic hepatitis, cirrhosis, and other liver diseases pose a serious threat to human health; however, liver transplantation is the only reliable treatment for the terminal stage of liver diseases. Previous researchers have shown that mesenchymal stem cells (MSCs) are characterized by differentiation and paracrine effects, as well as anti-oxidative stress and immune regulation functions. When MSCs are transplanted into animals, they migrate to the injured liver tissue along with the circulation, to protect the liver and alleviate the injury through the paracrine, immune regulation and other characteristics, making mesenchymal stem cell transplantation a promising alternative therapy for liver diseases. Although the efficacy of MSCs transplantation has been confirmed in various animal models of liver injury, many researchers have also proposed various pretreatment methods to improve the efficacy of mesenchymal stem cell transplantation, but there is still lack a set of scientific methods system aimed at improving the efficacy of transplantation therapy in scientific research and clinical practice. In this review, we summarize the possible mechanisms of MSCs therapy and compare the existing methods of MSCs modification corresponding to the treatment mechanism, hoping to provide as a reference to help future researchers explore a safe and simple transplantation strategy.
Collapse
Affiliation(s)
- L LIU
- School of Basic Medicine, Yangtze University Health Science Center, Jingzhou, China
| | - F YANG
- School of Basic Medicine, Yangtze University Health Science Center, Jingzhou, China
| |
Collapse
|
27
|
Lu D, Xu Y, Liu Q, Zhang Q. Mesenchymal Stem Cell-Macrophage Crosstalk and Maintenance of Inflammatory Microenvironment Homeostasis. Front Cell Dev Biol 2021; 9:681171. [PMID: 34249933 PMCID: PMC8267370 DOI: 10.3389/fcell.2021.681171] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/28/2021] [Indexed: 12/13/2022] Open
Abstract
Macrophages are involved in almost every aspect of biological systems and include development, homeostasis and repair. Mesenchymal stem cells (MSCs) have good clinical application prospects due to their ability to regulate adaptive and innate immune cells, particularly macrophages, and they have been used successfully for many immune disorders, including inflammatory bowel disease (IBD), acute lung injury, and wound healing, which have been reported as macrophage-mediated disorders. In the present review, we focus on the interaction between MSCs and macrophages and summarize their methods of interaction and communication, such as cell-to-cell contact, soluble factor secretion, and organelle transfer. In addition, we discuss the roles of MSC-macrophage crosstalk in the development of disease and maintenance of homeostasis of inflammatory microenvironments. Finally, we provide optimal strategies for applications in immune-related disease treatments.
Collapse
Affiliation(s)
- Di Lu
- The Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yan Xu
- The Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qiuli Liu
- The Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qi Zhang
- The Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
28
|
Zhou JH, Lu X, Yan CL, Sheng XY, Cao HC. Mesenchymal stromal cell-dependent immunoregulation in chemically-induced acute liver failure. World J Stem Cells 2021; 13:208-220. [PMID: 33815670 PMCID: PMC8006015 DOI: 10.4252/wjsc.v13.i3.208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 01/08/2021] [Accepted: 02/15/2021] [Indexed: 02/06/2023] Open
Abstract
Drug-induced liver injury (DILI), which refers to liver damage caused by a drug or its metabolites, has emerged as an important cause of acute liver failure (ALF) in recent years. Chemically-induced ALF in animal models mimics the pathology of DILI in humans; thus, these models are used to study the mechanism of potentially effective treatment strategies. Mesenchymal stromal cells (MSCs) possess immunomodulatory properties, and they alleviate acute liver injury and decrease the mortality of animals with chemically-induced ALF. Here, we summarize some of the existing research on the interaction between MSCs and immune cells, and discuss the possible mechanisms underlying the immuno-modulatory activity of MSCs in chemically-induced ALF. We conclude that MSCs can impact the phenotype and function of macrophages, as well as the differentiation and maturation of dendritic cells, and inhibit the proliferation and activation of T lymphocytes or B lymphocytes. MSCs also have immuno-modulatory effects on the production of cytokines, such as prostaglandin E2 and tumor necrosis factor-alpha-stimulated gene 6, in animal models. Thus, MSCs have significant benefits in the treatment of chemically-induced ALF by interacting with immune cells and they may be applied to DILI in humans in the near future.
Collapse
Affiliation(s)
- Jia-Hang Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Xuan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Cui-Lin Yan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Xin-Yu Sheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Hong-Cui Cao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory for Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China.
| |
Collapse
|
29
|
Xie H, Wu L, Chen X, Gao S, Li H, Yuan Y, Liang J, Wang X, Wang S, Xu C, Chu L, Zhan B, Zhou R, Yang X. Schistosoma japonicum Cystatin Alleviates Sepsis Through Activating Regulatory Macrophages. Front Cell Infect Microbiol 2021; 11:617461. [PMID: 33718268 PMCID: PMC7943722 DOI: 10.3389/fcimb.2021.617461] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/14/2021] [Indexed: 12/13/2022] Open
Abstract
Multi-organ failure caused by the inflammatory cytokine storm induced by severe infection is the major cause of death for sepsis. Sj-Cys is a cysteine protease inhibitor secreted by Schistosoma japonicum with strong immunomodulatory functions on host immune system. Our previous studies have shown that treatment with Sj-Cys recombinant protein (rSj-Cys) attenuated inflammation caused by sepsis. However, the immunological mechanism underlying the immunomodulation of Sj-Cys for regulating inflammatory diseases is not yet known. In this study, we investigated the effect of Sj-Cys on the macrophage M2 polarization and subsequent therapeutic effect on sepsis. The rSj-Cys was expressed in yeast Pichia pastoris. Incubation of mouse bone marrow-derived macrophages (BMDMs) with yeast-expressed rSj-Cys significantly activated the polarization of macrophages to M2 subtype characterized by the expression of F4/80+ CD206+ with the elated secretion of IL-10 and TGF-β. Adoptive transfer of rSj-Cys treated BMDMs to mice with sepsis induced by cecal ligation and puncture (CLP) significantly improved their survival rates and the systemic clinical manifestations of sepsis compared with mice receiving non-treated normal BMDMs. The therapeutic effect of Sj-Cys-induced M2 macrophages on sepsis was also reflected by the reduced pathological damages in organs of heart, lung, liver and kidney and reduced serological levels of tissue damage-related ALT, AST, BUN and Cr, associated with downregulated pro-inflammatory cytokines (IFN-gamma and IL-6) and upregulated regulatory anti-inflammatory cytokines (IL-10 and TGF-β). Our results demonstrated that Sj-Cys is a strong immunomodulatory protein with anti-inflammatory features through activating M2 macrophage polarization. The findings of this study suggested that Sj-Cys itself or Sj-Cys-induced M2 macrophages could be used as therapeutic agents in the treatment of sepsis or other inflammatory diseases.
Collapse
Affiliation(s)
- Hong Xie
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Department of Basic Medical College, Bengbu Medical College, Bengbu, China
| | - Lingqin Wu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Department of Pediatric, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xingzhi Chen
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Department of Basic Medical College, Bengbu Medical College, Bengbu, China
| | - Shifang Gao
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Huihui Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Department of Basic Medical College, Bengbu Medical College, Bengbu, China
| | - Yuan Yuan
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Department of Basic Medical College, Bengbu Medical College, Bengbu, China
| | - Jinbao Liang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China
| | - Xiaoli Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Department of Basic Medical College, Bengbu Medical College, Bengbu, China
| | - Shuying Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Department of Pediatric, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Changyan Xu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Department of Basic Medical College, Bengbu Medical College, Bengbu, China
| | - Liang Chu
- Department of General Surgery, Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Bin Zhan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Rui Zhou
- Department of Pediatric, First Affiliated Hospital of Bengbu Medical College, Bengbu, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases of Bengbu Medical College, Bengbu, China
| | - Xiaodi Yang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, China.,Department of Basic Medical College, Bengbu Medical College, Bengbu, China
| |
Collapse
|
30
|
Liu F, Xie J, Zhang X, Wu Z, Zhang S, Xue M, Chen J, Yang Y, Qiu H. Overexpressing TGF-β1 in mesenchymal stem cells attenuates organ dysfunction during CLP-induced septic mice by reducing macrophage-driven inflammation. Stem Cell Res Ther 2020; 11:378. [PMID: 32883356 PMCID: PMC7469348 DOI: 10.1186/s13287-020-01894-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/05/2020] [Accepted: 08/21/2020] [Indexed: 12/13/2022] Open
Abstract
Background Sepsis remains a leading cause of death in critically ill patients. It is well known that mesenchymal stem cells (MSCs) are a promising therapy partly due to their paracrine-mediated immunoregulatory function. Previous study demonstrated that transforming growth factor-beta1 (TGF-β1) is an important cytokine secreted by MSCs and that it participates in MSC-mediated macrophage phenotype switch from pro-inflammatory to pro-resolution. In addition, the transformation of macrophage phenotype may be a potential treatment for sepsis. However, the therapeutic effect of overexpressing TGF-β1 in MSCs (MSC-TGF-β1) on sepsis is not well understood. Therefore, this study aimed to evaluate the effects of TGF-β1 overexpressing MSCs on organ injury in cecal ligation and puncture (CLP)-induced septic mice and to detect the changes in macrophage phenotype during this process. Methods Mouse MSCs stably transfected with TGF-β1 were constructed and injected into CLP-induced septic mice via tail vein. After 24 h, the mice were sacrificed; then, the histopathology of the organ was evaluated by hematoxylin-eosin (H&E) staining. Inflammatory cytokines were detected by ELISA. Macrophage infiltration and phenotype transformation in the tissues were determined by immunohistochemistry and flow cytometry. In addition, we performed adoptive transfer of mouse peritoneal macrophage pretreated with TGF-β1 overexpressing MSCs in septic mice. Results We found that infusion of TGF-β1 overexpressing MSCs attenuated the histopathological impairment of the organ, decreased the pro-inflammatory cytokine levels and inhibited macrophage infiltration in tissues. TGF-β1 overexpressing MSCs induced macrophage phenotypes changed from pro-inflammatory to pro-resolution in inflammatory environment. The adoptive transfer of mouse peritoneal macrophages pretreated with TGF-β1 overexpressing MSCs also relieved organ damage in CLP-induced septic mice. Conclusion Under septic conditions, TGF-β1 overexpressing MSCs can enhance the therapeutic effects of MSCs on organ injury and inflammation as a result of reduced macrophage infiltration and induced macrophages transformation, the adoptive transfer of macrophages treated with TGF-β1 overexpressing MSCs also relieved organ damage. This will provide new hope for the treatment of sepsis.
Collapse
Affiliation(s)
- Feng Liu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jianfeng Xie
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.
| | - Xiwen Zhang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Zongsheng Wu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Shi Zhang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Ming Xue
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jianxiao Chen
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yi Yang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Haibo Qiu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| |
Collapse
|
31
|
Immunomodulatory and Therapeutic Effects of Mesenchymal Stem Cells on Organ Dysfunction in Sepsis. Shock 2020; 55:423-440. [PMID: 32826813 DOI: 10.1097/shk.0000000000001644] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
ABSTRACT Sepsis is a life-threatening disorder that is caused by a dysregulated inflammatory response during an infection. The disease mostly affects pregnant women, newborns, and patients in intensive care units. Sepsis treatment is a significant part of a country's health budgets. Delay in the therapy causes irreversible failure of various organs due to the lack of blood supply and reduction of oxygen in the tissues and eventually increased mortality. The involvement of four or five organs by sepsis has been attributed to an increased risk of death to over 90%. Although antibiotics are at the first line of sepsis treatment, they do not possess enough potency to control the disease and prevent subsequent organ failure. The immunomodulatory, anti-inflammatory, anti-apoptotic, and anti-microbial properties of mesenchymal stem cells (MSCs) have been reported in various studies. Therefore, the application of MSCs has been considered a potentially promising therapeutic strategy. In preclinical studies, the administration of MSCs has been associated with reduced bacterial load and decreased levels of pro-inflammatory factors as well as the improved function of the different vital organs, including heart, kidney, liver, and lungs. The current study provides a brief review of sepsis and its pathophysiology, and then highlights recent findings in the therapeutic effects of MSCs and MSC-derived secretome in improving sepsis-induced organ dysfunction. Besides, eligible sepsis candidates for MSC-therapy and the latest clinical findings in these areas have been reviewed.
Collapse
|
32
|
LncRNA XIST silencing protects against sepsis-induced acute liver injury via inhibition of BRD4 expression. Inflammation 2020; 44:194-205. [PMID: 32812145 DOI: 10.1007/s10753-020-01321-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Sepsis is recognized as the acute systemic inflammatory response to severe infection. It is main cause of multiple organ system dysfunction and even organ failure. Long non-coding RNA X inactivate-specific transcript (XIST) is implicated in multiple inflammatory diseases. The aim of present work was to investigate the precise mechanism of XIST underlying sepsis-induced acute liver injury. Rats were underwent cecal ligation and puncture (CLP) to establish sepsis-induced the animal models of acute liver injury. Hematoxylin and eosin (H&E) staining was performed to observe pathological alterations. Corresponding commercial assay kits were employed to analyze the levels of inflammatory cytokines and oxidative stress. Western blot and reverse transcriptional quantitative PCR (RT-qPCR) were performed to determine the expression of proteins and target genes. Finally, TUNEL and CCK-8 assays were performed to test apoptosis rate and cell viability, respectively. In our study, XIST and BRD4 were highly expressed in serum of patients with sepsis-induced acute liver injury. XIST knockdown ameliorated sepsis-induced acute liver injury and inhibited inflammation, oxidative stress, and cell apoptosis in sepsis-induced acute liver injury rats. Interestingly, XIST knockdown downregulated the expression of BRD4, and BRD4 overexpression abolished the impacts of XIST knockdown on inflammation, oxidative stress, and apoptosis of that LPS-induced Kupffer cells. We conclude that lncRNA XIST silencing protects against sepsis-induced acute liver injury via inhibition of the BRD4 expression. Therefore, XIST may be a biomarker for sepsis diagnosis and treatment.
Collapse
|
33
|
Sun XY, Ding XF, Liang HY, Zhang XJ, Liu SH, Bing-Han, Duan XG, Sun TW. Efficacy of mesenchymal stem cell therapy for sepsis: a meta-analysis of preclinical studies. Stem Cell Res Ther 2020; 11:214. [PMID: 32493435 PMCID: PMC7268531 DOI: 10.1186/s13287-020-01730-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/28/2020] [Accepted: 05/13/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Multiple studies have reported that mesenchymal stem cell (MSC) therapy has beneficial effects in experimental models of sepsis. However, this finding remains inconclusive. This study was performed to systematically determine the connection between MSC therapy and mortality in sepsis animal models by pooling and analyzing data from newly published studies. METHODS A detailed search of related studies from 2009 to 2019 was conducted in four databases, including MEDLINE, EMBASE, Cochrane Library, and Web of Science. After browsing and filtering out articles that met the inclusion criteria for statistical analysis, the inverse variance method of the fixed effects model was used to calculate the pooled odds ratios (ORs) and their 95% confidence intervals (CIs). RESULTS Twenty-nine animal studies, including 1266 animals, were identified. None of the studies was judged to have a low risk of bias. The meta-analysis demonstrated that MSC therapy was related to a significantly lower mortality rate (OR 0.29, 95% CI 0.22-0.38, P < 0.001). Subgroup analyses performed based on the MSC injection dose (< 1.0 × 106 cells, OR = 0.33, 95% CI 0.20-0.56, P < 0.001; 1.0 × 106 cells, OR = 0.24, 95% CI 0.16-0.35, P < 0.001) and injection time (< 1 h, OR = 0.24, 95% CI 0.13-0.45, P < 0.001; 1 h, OR = 0.28, 95% CI 0.17-0.46, P < 0.001) demonstrated that treatment with MSCs significantly reduced the mortality rate of animals with sepsis. CONCLUSION This up-to-date meta-analysis showed a connection between MSC therapy and lower mortality in sepsis animal models, supporting the potential therapeutic effect of MSC treatment in future clinical trials. The results in this study contradict a previous meta-analysis with regards to the ideal dose of MSC therapy. Thus, further research is required to support these findings.
Collapse
Affiliation(s)
- Xue-Yi Sun
- General ICU, Henan Key Laboratory of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou Key Laboratory of Sepsis, Zhengzhou, 450052 China
| | - Xian-Fei Ding
- General ICU, Henan Key Laboratory of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou Key Laboratory of Sepsis, Zhengzhou, 450052 China
| | - Huo-Yan Liang
- General ICU, Henan Key Laboratory of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou Key Laboratory of Sepsis, Zhengzhou, 450052 China
| | - Xiao-Juan Zhang
- General ICU, Henan Key Laboratory of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou Key Laboratory of Sepsis, Zhengzhou, 450052 China
| | - Shao-Hua Liu
- General ICU, Henan Key Laboratory of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou Key Laboratory of Sepsis, Zhengzhou, 450052 China
| | - Bing-Han
- General ICU, Henan Key Laboratory of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou Key Laboratory of Sepsis, Zhengzhou, 450052 China
| | - Xiao-Guang Duan
- General ICU, Henan Key Laboratory of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou Key Laboratory of Sepsis, Zhengzhou, 450052 China
| | - Tong-Wen Sun
- General ICU, Henan Key Laboratory of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou Key Laboratory of Sepsis, Zhengzhou, 450052 China
| |
Collapse
|
34
|
Topcu Sarica L, Zibandeh N, Genç D, Gül F, Akkoç T, Kombak EF, Cinel L, Akkoç T, Cinel I. Immunomodulatory and Tissue-preserving Effects of Human Dental Follicle Stem Cells in a Rat Cecal Ligation and Perforation Sepsis Model. Arch Med Res 2020; 51:397-405. [PMID: 32334851 DOI: 10.1016/j.arcmed.2020.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 03/19/2020] [Accepted: 04/10/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Mesenchymal stem cells may be used for the treatment of sepsis. Dental follicle stem cells (DFSCs) are easily accessible but have not been studied in vivo or in clinical trials in sepsis models. AIM OF THE STUDY We aim to elucidate DFSC effects on host immunological functions in a rat cecal ligation and perforation (CLP) sepsis model. METHODS Adult male rats were categorized into group 1 (sham procedure SP), group 2 (SP + 1 × 106 DFSCs administered 0 h after SP), group 3 (CLP + saline), group 4 (CLP + 1 × 106 DFSCs administered 0 h after CLP), and group 5 (CLP + 1 × 106 DFSCs administered 4 h after CLP). Green fluorescent protein-labeled cells were used for imaging. Histopathological examination of ileal tissues was performed. RESULTS A significant increase in the percentage of CD4+/CD25+/Foxp3+ Treg cells in groups 4 and 5 occurred compared with that in group 3. No significant changes in CD3+/CD4+ helper T-cells and CD3+/CD8+ cytotoxic T-cells were observed. Treatment with DFSCs at 4 h significantly decreased the level of TNF-α compared with that in group 3. No significant changes in IL-10 levels and lymphocyte proliferation suppression were observed. During histopathological examination, no high scoring (Chiu scores: 3 or 4) rats were observed in the curative treatment group (group 5). CONCLUSIONS Treatment with DFSC after 4 h of sepsis induction downregulates tissue inflammatory responses by decreasing TNF-α levels and increasing Treg cell ratio. This also has a protective effect on intestinal tissues during sepsis.
Collapse
Affiliation(s)
- Leyla Topcu Sarica
- Department of Anesthesiology and Reanimation, School of Medicine, Marmara University, Istanbul, Turkey
| | - Noushin Zibandeh
- Department of Allergy and Immunology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Deniz Genç
- Department of Allergy and Immunology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Fethi Gül
- Department of Anesthesiology and Reanimation, School of Medicine, Marmara University, Istanbul, Turkey
| | - Tolga Akkoç
- TUBITAK MRC Genetic Engineering and Biotechnology Institute, Gebze, Turkey
| | - Erdem Faruk Kombak
- Department of Pathology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Leyla Cinel
- Department of Pathology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Tunç Akkoç
- Department of Allergy and Immunology, School of Medicine, Marmara University, Istanbul, Turkey
| | - Ismail Cinel
- Department of Anesthesiology and Reanimation, School of Medicine, Marmara University, Istanbul, Turkey.
| |
Collapse
|
35
|
Wu GJ, Lin YW, Tsai HC, Lee YW, Chen JT, Chen RM. Sepsis-induced liver dysfunction was ameliorated by propofol via suppressing hepatic lipid peroxidation, inflammation, and drug interactions. Life Sci 2018; 213:279-286. [PMID: 30352244 DOI: 10.1016/j.lfs.2018.10.038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/13/2022]
Abstract
AIMS Our previous study showed that propofol can protect against sepsis-induced insults through suppressing liver nitrosation and inflammation. This study further evaluated the mechanisms of propofol-caused protection from sepsis-induced liver dysfunction. MAIN METHODS Male Wistar rats were subjected to cecal ligation and puncture (CLP) and then exposed to propofol. Levels of hepatic oxidative stress and lipid peroxidation were consecutively measured. Expressions of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-4 messenger (m)RNA or proteins were quantified. Effects of propofol on microsomal pentoxyresorufin O-dealkelase (PROD) and ethoxycoumarin O-deethylase (ECOD) activities were determined. KEY FINDINGS Administration of propofol to CLP-treated rats significantly attenuated sepsis-induced insults. CLP caused augmented serum aspartate aminotransferase and alanine aminotransferase activities and concurrently triggered liver damage. In contrast, treatment with propofol protected against CLP-induced liver dysfunction. As to the mechanisms, the CLP-induced increases in oxidative stress and lipid peroxidation levels and TNF-α and IL-1β mRNA and protein expressions were subsequently attenuated by propofol. Furthermore, administration of CLP-treated rats with propofol augmented levels of IL-4 in the liver. Phenobarbital treatment of liver microsomes in CLP-treated rats produced less amplification of PROD and ECOD activities, and a smaller amount of 4-hydroxypropofol was metabolized from propofol by liver microsomes. In contrast, more drug interactions occurred with propofol, which decreased PROD and ECOD activities in liver microsomes of CLP-treated rats. SIGNIFICANCE Taken together, the present study showed that propofol can protect against sepsis-induced liver dysfunction through suppressing hepatic oxidative stress, lipid peroxidation, inflammation, and drug biotransformation and interactions in the liver.
Collapse
Affiliation(s)
- Gong-Jhe Wu
- Department of Anesthesiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yung-Wei Lin
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chien Tsai
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yuan-Wen Lee
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jui-Tai Chen
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ruei-Ming Chen
- Anesthesiology and Health Policy Research Center, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|