1
|
Zhang Z, Wang J, Li H, Niu Q, Tao Y, Zhao X, Zeng Z, Dong H. The role of the interleukin family in liver fibrosis. Front Immunol 2025; 16:1497095. [PMID: 39995661 PMCID: PMC11847652 DOI: 10.3389/fimmu.2025.1497095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/22/2025] [Indexed: 02/26/2025] Open
Abstract
Liver fibrosis represents a wound-healing response to chronic liver injury caused by viral infections, alcohol, and chemicals agents. It is a critical step in the progression from chronic liver disease to cirrhosis and hepatocellular carcinoma. No chemical or biological drugs have been approved for the treatment of liver fibrosis. Relevant studies have demonstrated that effective inhibition of hepatitis B virus (HBV) replication by nucleoside (acid) analogs or polyethylene glycol alpha-interferon can lead to recovery in some patients with hepatitis B liver fibrosis, However, some patients with liver fibrosis do not show improvement, even after achieving a complete serologic and virologic response. A similar situation occurs in patients with hepatitis C-related liver fibrosis. The liver, with its unique anatomical and immunological structure, is the largest immune organ and produces a large number of cytokines in response to external stimuli, which are crucial for the progression of liver fibrosis. cytokines can act either by directly affecting hepatic stellate cells (HSCs) or by indirectly regulating immune target cells. Among these, the interleukin family activates a complex cascade of responses, including cytokines, chemokines, adhesion molecules, and lipid mediators, playing a key role in the initiation and regulation of inflammation, as well as innate and adaptive immunity. In this paper, we systematically summarize recent literature to elucidate the pathogenesis of interleukin-mediated liver fibrosis and explore potential therapeutic targets for liver fibrosis treatment.
Collapse
Affiliation(s)
- Zixin Zhang
- Central Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiahui Wang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui Li
- Central Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qun Niu
- Central Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yujing Tao
- Central Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin Zhao
- Central Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zijian Zeng
- Central Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Haijian Dong
- Central Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
2
|
Yuan L, Li Y, Liu D, Zhang H, Yang J, Shen H, Xia L, Yao L, Lu J. Interleukin-35 protein inhibits osteoclastogenesis and attenuates collagen-induced arthritis in mice. J Cell Physiol 2024; 239:e31231. [PMID: 38451477 DOI: 10.1002/jcp.31231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 03/08/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease. Its pathological features include synovial inflammation, bone erosion, and joint structural damage. Our previous studies have shown that interleukin (IL)-35 is involved in the pathogenesis of bone loss in RA patients. In this study, we are further evaluating the efficacy of IL-35 on collagen-induced arthritis (CIA) in the mouse model. Male DBA/1J mice (n = 10) were initially immunized, 2 μg/mouse IL-35 was injected intraperitoneally every week for 3 weeks after the establishment of the CIA model. Clinical arthritis, histopathological analysis, and three-dimensional micro‑computed tomography (3D micro‑CT) were determined after the mice were anesthetized on the 42th day. In vitro, RANKL/M-CSF induced mouse preosteoclasts (RAW264.7 cells line) was subjected to antiarthritis mechanism study in the presence of IL-35. The results of clinical arthritis, histopathological analysis, and 3D micro‑CT, the expression of RANK/RANKL/OPG axis, inflammatory cytokines, and osteoclastogenesis-related makers demonstrated decreasing severity of synovitis and bone destruction in the ankle joints after IL-35 treatment. Furthermore, IL-35 attenuated inflammatory cytokine production and the expression of osteoclastogenesis-related makers in a mouse preosteoclasts cell line RAW264.7. The osteoclastogenesis-related makers were significantly reduced in IL-35 treated RAW264.7 cells line after blockage with the JAK/STAT1 signaling pathway. These results demonstrated that IL-35 protein could inhibits osteoclastogenesis and attenuates CIA in mice. We concluded that IL-35 can exhibit anti-osteoclastogenesis effects by reducing the expression of inflammatory cytokines and osteoclastogenesis-related makers, thus alleviating bone destruction in the ankle joint and could be a potential therapeutic target for RA.
Collapse
Affiliation(s)
- Lin Yuan
- Department of Health Management, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yuxuan Li
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Dan Liu
- Department of Critical Care Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Hui Zhang
- Department of Rheumatology and Immunology, The Fifth People Hospital, Shenyang, China
| | - Jie Yang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hui Shen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Liping Xia
- Department of Rheumatology and Immunology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Lutian Yao
- Department of Orthopedic, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jing Lu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
3
|
Jiang T, Xiang X, Wang X, Han Z, Cheng C, Zhu Y, Yang Z, Liang Y. Role of regulatory T cells in pathogenesis and therapeutics of primary biliary cholangitis and primary sclerosing cholangitis. REGULATORY T CELLS AND AUTOIMMUNE DISEASES 2024:433-452. [DOI: 10.1016/b978-0-443-13947-5.00014-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Zhao L, Li J, Mo G, Cao D, Li C, Huang G, Jiang L, Chen G, Yao H, Peng X. Recombinant protein EBI3 attenuates Clonorchis sinensis-induced liver fibrosis by inhibiting hepatic stellate cell activation in mice. Parasit Vectors 2023; 16:246. [PMID: 37480105 PMCID: PMC10360228 DOI: 10.1186/s13071-023-05863-5] [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: 03/01/2023] [Accepted: 07/03/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND Chronic infection with Clonorchis sinensis can cause hepatobiliary fibrosis and even lead to hepatobiliary carcinoma. Epstein-Barr virus-induced gene 3 protein (EBI3) is a subunit of interleukin 35, which can regulate inflammatory response and the occurrence of fibrotic diseases. Previous studies have reported that the expression of EBI3 in the serum of patients with liver cirrhosis is reduced. The present study aims to investigate the biological effects of EBI3 on liver fibrosis caused by C. sinensis and the underlying molecular mechanisms. METHODS We first established a mouse model of liver fibrosis induced by C. sinensis infection and then measured the serum expression of EBI3 during the inflammatory and fibrotic phase. GO (Gene Ontology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway analyses were performed to investigate the potential role of EBI3 in liver fibrosis by regulating the extracellular matrix structural constituent and collagen catabolic process. Recombinant protein EBI3 (rEBI3) was added to hepatic stellate cells (HSCs) in vitro with C. sinensis antigen to explore its function. Finally, the therapeutic effect of rEBI3 was verified by intravenous injection into C. sinensis-infected mice. RESULTS The results showed that the serum expression of EBI3 increased in the inflammatory response phase but decreased in the fibrotic phase. The excretory-secretory products of C. sinensis (Cs.ESP) were able to stimulate HSC activation, while rEBI3 reduced the activation of HSCs induced by Cs.ESP. Also, the protein expression of gp130 and downstream protein expressions of JAK1, p-JAK1, STAT3 and p-STAT3 in HSCs were increased after rEBI3 incubation. Finally, intravenously injected rEBI3 inhibited hepatic epithelial-mesenchymal transition in C. sinensis-infected mice by inhibiting HSC activation and reducing liver injury. CONCLUSION This study confirms that rEBI3 can attenuate C. sinensis-induced liver fibrosis by inhibiting HSC activation and may be one of the potential treatments for liver fibrosis.
Collapse
Affiliation(s)
- Lei Zhao
- Guangxi University Key Laboratory of Pathogenic Biology, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Jia Li
- Guangxi University Key Laboratory of Pathogenic Biology, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Gang Mo
- Guangxi University Key Laboratory of Pathogenic Biology, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Deping Cao
- Guangxi University Key Laboratory of Pathogenic Biology, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Chun Li
- Guangxi University Key Laboratory of Pathogenic Biology, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Guoyang Huang
- Guangxi University Key Laboratory of Pathogenic Biology, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Liping Jiang
- Guangxi University Key Laboratory of Pathogenic Biology, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Gen Chen
- Guangxi University Key Laboratory of Pathogenic Biology, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Hongbing Yao
- Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Xiaohong Peng
- Guangxi University Key Laboratory of Pathogenic Biology, Guilin Medical University, Guilin, Guangxi, People's Republic of China.
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, People's Republic of China.
| |
Collapse
|
5
|
Huang X, Huang X, Huang Y, Zheng J, Lu Y, Mai Z, Zhao X, Cui L, Huang S. The oral microbiome in autoimmune diseases: friend or foe? J Transl Med 2023; 21:211. [PMID: 36949458 PMCID: PMC10031900 DOI: 10.1186/s12967-023-03995-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/15/2023] [Indexed: 03/24/2023] Open
Abstract
The human body is colonized by abundant and diverse microorganisms, collectively known as the microbiome. The oral cavity has more than 700 species of bacteria and consists of unique microbiome niches on mucosal surfaces, on tooth hard tissue, and in saliva. The homeostatic balance between the oral microbiota and the immune system plays an indispensable role in maintaining the well-being and health status of the human host. Growing evidence has demonstrated that oral microbiota dysbiosis is actively involved in regulating the initiation and progression of an array of autoimmune diseases.Oral microbiota dysbiosis is driven by multiple factors, such as host genetic factors, dietary habits, stress, smoking, administration of antibiotics, tissue injury and infection. The dysregulation in the oral microbiome plays a crucial role in triggering and promoting autoimmune diseases via several mechanisms, including microbial translocation, molecular mimicry, autoantigen overproduction, and amplification of autoimmune responses by cytokines. Good oral hygiene behaviors, low carbohydrate diets, healthy lifestyles, usage of prebiotics, probiotics or synbiotics, oral microbiota transplantation and nanomedicine-based therapeutics are promising avenues for maintaining a balanced oral microbiome and treating oral microbiota-mediated autoimmune diseases. Thus, a comprehensive understanding of the relationship between oral microbiota dysbiosis and autoimmune diseases is critical for providing novel insights into the development of oral microbiota-based therapeutic approaches for combating these refractory diseases.
Collapse
Affiliation(s)
- Xiaoyan Huang
- Department of Preventive Dentistry, Stomatological Hospital, School of Stomatology, Southern Medical University, Haizhu District, No.366 Jiangnan Da Dao Nan, Guangzhou, 510280, China
| | - Xiangyu Huang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Haizhu District, No.366 Jiangnan Da Dao Nan, Guangzhou, 510280, China
| | - Yi Huang
- Department of Preventive Dentistry, Stomatological Hospital, School of Stomatology, Southern Medical University, Haizhu District, No.366 Jiangnan Da Dao Nan, Guangzhou, 510280, China
| | - Jiarong Zheng
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Ye Lu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Haizhu District, Guangzhou, 510280, China
| | - Zizhao Mai
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Xinyuan Zhao
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Haizhu District, No.366 Jiangnan Da Dao Nan, Guangzhou, 510280, China.
| | - Li Cui
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Haizhu District, Guangzhou, 510280, China.
- Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, CA, 90095, USA.
| | - Shaohong Huang
- Department of Preventive Dentistry, Stomatological Hospital, School of Stomatology, Southern Medical University, Haizhu District, No.366 Jiangnan Da Dao Nan, Guangzhou, 510280, China.
| |
Collapse
|
6
|
Krajewska NM, Fiancette R, Oo YH. Interplay between Mast Cells and Regulatory T Cells in Immune-Mediated Cholangiopathies. Int J Mol Sci 2022; 23:5872. [PMID: 35682552 PMCID: PMC9180565 DOI: 10.3390/ijms23115872] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 01/10/2023] Open
Abstract
Immune-mediated cholangiopathies are characterised by the destruction of small and large bile ducts causing bile acid stasis, which leads to subsequent inflammation, fibrosis, and eventual cirrhosis of the liver tissue. A breakdown of peripheral hepatic immune tolerance is a key feature of these diseases. Regulatory T cells (Tregs) are a major anti-inflammatory immune cell subset, and their quantities and functional capacity are impaired in autoimmune liver diseases. Tregs can undergo phenotypic reprogramming towards pro-inflammatory Th1 and Th17 profiles. The inflamed hepatic microenvironment influences and can impede normal Treg suppressive functions. Mast cell (MC) infiltration increases during liver inflammation, and active MCs have been shown to be an important source of pro-inflammatory mediators, thus driving pathogenesis. By influencing the microenvironment, MCs can indirectly manipulate Treg functions and inhibit their suppressive and proliferative activity. In addition, direct cell-to-cell interactions have been identified between MCs and Tregs. It is critical to consider the effects of MCs on the inflammatory milieu of the liver and their influence on Treg functions. This review will focus on the roles and crosstalk of Tregs and MCs during autoimmune cholangiopathy pathogenesis progression.
Collapse
Affiliation(s)
- Natalia M. Krajewska
- Centre for Liver and Gastrointestinal Research & NIHR Birmingham Liver Biomedical Research Unit, Institute of Biomedical Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK;
- Centre for Rare Diseases, European Reference Network Rare Liver Centre, University Hospital Birmingham NHS Foundation Trust, Birmingham B15 2TH, UK
| | - Rémi Fiancette
- Centre for Liver and Gastrointestinal Research & NIHR Birmingham Liver Biomedical Research Unit, Institute of Biomedical Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK;
- Centre for Rare Diseases, European Reference Network Rare Liver Centre, University Hospital Birmingham NHS Foundation Trust, Birmingham B15 2TH, UK
| | - Ye H. Oo
- Centre for Liver and Gastrointestinal Research & NIHR Birmingham Liver Biomedical Research Unit, Institute of Biomedical Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK;
- Centre for Rare Diseases, European Reference Network Rare Liver Centre, University Hospital Birmingham NHS Foundation Trust, Birmingham B15 2TH, UK
- Advanced Cellular Therapy Facility, University of Birmingham, Birmingham B15 2TT, UK
- Liver Transplant and Hepatobiliary Unit, Queen Elizabeth Hospital, University Hospital Birmingham NHS Foundation Trust, Birmingham B15 2TH, UK
| |
Collapse
|
7
|
Yang L, Liu S, Zhang Q, Jia S, Qiu C, Jin Z. Overexpression of ascitic interleukin-35 induces CD8 + T cell exhaustion in liver cirrhotic patients with spontaneous bacterial peritonitis. Int Immunopharmacol 2022; 108:108729. [PMID: 35349961 DOI: 10.1016/j.intimp.2022.108729] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/15/2022] [Accepted: 03/20/2022] [Indexed: 11/15/2022]
Abstract
Interleukin (IL) -35 induces immunotolerance by suppression of CD8+ T cells during chronic infections and cancers. In the present study, we amined to investigate the role of IL-35-mediated regulation of CD8+ T cells in patients with liver cirrhosis. Seventy-one patients with liver cirrhosis (46 patients with untainted ascites and 25 patients with spontaneous bacterial peritonitis [SBP]) and 22 controls were enrolled. Plasma and ascitic IL-35 levels were measured using ELISA. Peripheral and ascitic CD4+ and CD8+ T cells were purified to investigate their functional phenotypes. IL-35-stimulated CD8+ T cells were cultured with HepG2 cells in direct and indirect contact systems. Lactate dehydrogenase expression and cytokine secretion were measured to determine the cytotoxicity of CD8+ T cells. Plasma IL-35 was elevated in patients with liver cirrhosis, and ascitic IL-35 levels were higher in the SBP group than in the untainted ascites group. No significant differences in transcription factor expression or cytokine production in peripheral and ascitic CD4+ T cells were observed among groups. In the SBP group, ascitic CD8+ T cells expressed decreased cytotoxic molecules, along with the reduced secretion of interferon-γ and tumor necrosis factor-α when compared with the untainted ascites group. IL-35 stimulation suppressed ascitic CD8+ T cell cytotoxicity and cytokine production in both direct and indirect contact culture systems. This process was accompanied by decreased cytotoxic molecule expression and increased immune-checkpoint molecules in ascitic CD8+ T cells. The present findings revealed that overexpression of ascitic IL-35 dampened the cytotoxicity of CD8+ T cells in liver cirrhotic patients with SBP.
Collapse
Affiliation(s)
- Lanlan Yang
- Digestive Disease Center, Department of Hepatopancreatobiliary Medicine, The Second Hospital, Jilin University, Changchun, Jilin Province 130041, People's Republic of China
| | - Siqi Liu
- Digestive Disease Center, Department of Hepatopancreatobiliary Medicine, The Second Hospital, Jilin University, Changchun, Jilin Province 130041, People's Republic of China
| | - Qian Zhang
- Digestive Disease Center, Department of Hepatopancreatobiliary Medicine, The Second Hospital, Jilin University, Changchun, Jilin Province 130041, People's Republic of China
| | - Shengnan Jia
- Digestive Disease Center, Department of Hepatopancreatobiliary Medicine, The Second Hospital, Jilin University, Changchun, Jilin Province 130041, People's Republic of China
| | - Chen Qiu
- Digestive Disease Center, Department of Hepatopancreatobiliary Medicine, The Second Hospital, Jilin University, Changchun, Jilin Province 130041, People's Republic of China
| | - Zhenjing Jin
- Digestive Disease Center, Department of Hepatopancreatobiliary Medicine, The Second Hospital, Jilin University, Changchun, Jilin Province 130041, People's Republic of China.
| |
Collapse
|
8
|
Hu S, Lian PP, Hu Y, Zhu XY, Jiang SW, Ma Q, Li LY, Yang JF, Yang L, Guo HY, Zhou H, Yang CC, Meng XM, Li J, Li HW, Xu T, Zhou H. The Role of IL-35 in the Pathophysiological Processes of Liver Disease. Front Pharmacol 2021; 11:569575. [PMID: 33584256 PMCID: PMC7873894 DOI: 10.3389/fphar.2020.569575] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/10/2020] [Indexed: 12/23/2022] Open
Abstract
It is known that liver diseases have several characteristics of massive lipid accumulation and lipid metabolic disorder, and are divided into liver inflammation, liver fibrosis, liver cirrhosis (LC), and hepatocellular carcinoma (HCC) in patients. Interleukin (IL)-35, a new-discovered cytokine, can protect the liver from the environmental attack by increasing the ratio of Tregs (T regulatory cells) which can increase the anti-inflammatory cytokines and inhibit the proliferation of immune cellular. Interestingly, two opposite mechanisms (pro-inflammatory and anti-inflammatory) have connection with the ultimate formation of liver diseases, which suggest that IL-35 may play crucial function in the process of liver diseases through immunosuppressive regulation. Besides, some obvious advantages also imply that IL-35 can be considered as a new therapeutic target to control the progression of liver diseases, while its mechanism of function still needs further research.
Collapse
Affiliation(s)
- Shuang Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Pan-Pan Lian
- School of Pharmacy, NanJing University, NanJing, China
| | - Ying Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xing-Yu Zhu
- National Drug Clinical Trial Institution, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Shao-Wei Jiang
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qiang Ma
- Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Liang-Yun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Jun-Fa Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Li Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Hai-Yue Guo
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Hong Zhou
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Chen-Chen Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Hai-Wen Li
- The Third Affiliated Hospital of Anhui Medical University, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Huan Zhou
- National Drug Clinical Trial Institution, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
9
|
Zhao N, Liu X, Guo H, Zhao X, Qiu Y, Wang W. Interleukin-35: An emerging player in the progression of liver diseases. Clin Res Hepatol Gastroenterol 2021; 45:101518. [PMID: 33387857 DOI: 10.1016/j.clinre.2020.07.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 07/31/2020] [Indexed: 02/04/2023]
Abstract
Interleukin-35(IL-35), a newly identified immunosuppressive cytokine, has recently been shown to play a significant role in the progression of various autoimmune diseases and malignant tumors. The liver is the largest organ in the body and is generally regarded as an important lymphoid organ by an increasing number of immunologists. A number of reports have demonstrated that IL-35 plays essential roles in maintaining the immune homeostasis of the liver microenvironment. This review summarizes the existing studies of IL-35 in liver diseases, including viral hepatitis, immune liver injury, liver cirrhosis and carcinoma. We aimed to provide a comprehensive overview of the vital roles of IL-35 in hepatic damage and explore new alternative therapeutic targets for these diseases.
Collapse
Affiliation(s)
- Na Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Xin Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hao Guo
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xiangnan Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yujie Qiu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wei Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
10
|
Yang L, Zhang Q, Song J, Wang W, Jin Z. Interleukin-35 Suppresses CD8 + T Cell Activity in Patients with Viral Hepatitis-Induced Acute-on-Chronic Liver Failure. Dig Dis Sci 2020; 65:3614-3623. [PMID: 31974915 DOI: 10.1007/s10620-020-06077-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/13/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Interleukin (IL)-35 is a newly indentified cytokine and induces immunotolerance via suppression of CD8+ T cell activity in chronic viral hepatitis. AIMS To investigate the modulatory function of IL-35 to CD8+ T cells in viral hepatitis-induced acute-on-chronic liver failure (ACLF). METHODS Fifty-five ACLF patients and 21 healthy controls were enrolled. Serum IL-35 concentration was measured by ELISA. Absolute accounts for T cells, immune checkpoint molecules, and cytotoxic molecules in CD8+ T cells were measured by flow cytometry and real-time PCR, respectively. Direct and indirect contact co-culture systems between CD8+ T cells and HepG2 cells were set up. The regulatory function of IL-35 to CD8+ T cells was assessed by measuring lactate dehydrogenase expression and cytokine production. RESULTS Serum IL-35 concentration was elevated in ACLF patients and positively correlated with total bilirubin, but negatively correlated with prothrombin time activity. Peripheral CD8+ T cells showed exhausted phenotype in ACLF patients, which manifested as up-regulation of programmed death-1 (PD-1), cytotoxic T-lymphocyte-associated protein-4 (CTLA-4), and lymphocyte activation gene-3 (LAG-3) but down-regulation of perforin, granzyme B, and FasL. Recombinant IL-35 stimulation dampened cytotoxicity and interferon-γ production in both direct and indirect contact co-culture systems. This process was accompanied by elevation of PD-1, CTLA-4, and LAG3, as well as reduction of perforin, granzyme B, and FasL in CD8+ T cells. CONCLUSION Elevated IL-35 suppressed both cytolytic and non-cytolytic activity of CD8+ T cells in ACLF patients.
Collapse
Affiliation(s)
- Lanlan Yang
- Department of Hepatopancreatobiliary Medicine, The Second Hospital, Jilin University, No. 218 Ziqiang St, Nanguan District, Changchun, 130041, Jilin Province, China
| | - Qian Zhang
- Department of Hepatopancreatobiliary Medicine, The Second Hospital, Jilin University, No. 218 Ziqiang St, Nanguan District, Changchun, 130041, Jilin Province, China
| | - Jie Song
- Department of Hepatopancreatobiliary Medicine, The Second Hospital, Jilin University, No. 218 Ziqiang St, Nanguan District, Changchun, 130041, Jilin Province, China
| | - Wudong Wang
- Department of Hepatopancreatobiliary Medicine, The Second Hospital, Jilin University, No. 218 Ziqiang St, Nanguan District, Changchun, 130041, Jilin Province, China
| | - Zhenjing Jin
- Department of Hepatopancreatobiliary Medicine, The Second Hospital, Jilin University, No. 218 Ziqiang St, Nanguan District, Changchun, 130041, Jilin Province, China.
| |
Collapse
|
11
|
Fu HY, Bao WM, Yang CX, Lai WJ, Xu JM, Yu HY, Yang YN, Tan X, Gupta AK, Tang YM. Kupffer Cells Regulate Natural Killer Cells Via the NK group 2, Member D (NKG2D)/Retinoic Acid Early Inducible-1 (RAE-1) Interaction and Cytokines in a Primary Biliary Cholangitis Mouse Model. Med Sci Monit 2020; 26:e923726. [PMID: 32599603 PMCID: PMC7346879 DOI: 10.12659/msm.923726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Kupffer cells and natural killer (NK) cells has been identified as contributing factors in the pathogenesis of hepatitis, but the detailed mechanism of these cell types in the pathogenesis of primary biliary cholangitis (PBC) is poorly understood. Material/Methods In this study, polyinosinic: polycytidylic acid (poly I: C), 2-octynoic acid-bovine serum albumin (2OA-BSA) and Freund’s adjuvant (FA) were injected to establish a murine PBC model, from which NK cells and Kupffer cells were extracted and isolated. The cells were then co-cultivated in a designed culture system, and then NK group 2, member D (NKG2D), retinoic acid early inducible-1 (RAE-1), F4/80, and cytokine expression levels were detected. Results The results showed close crosstalk between Kupffer cells and NK cells. PBC mice showed increased surface RAE-1 protein expression and Kupffer cell cytokine secretion, which subsequently activated NK cell-mediated target cell killing via NKG2D/RAE-1 recognition, and increased inflammation. NK cell-derived interferon-γ (IFN-γ) and Kupffer cell-derived tumor necrosis factor α (TNF-α) were found to synergistically regulate inflammation. Moreover, interleukin (IL)-12 and IL-10 improved the crosstalk between NK cells and Kupffer cells. Conclusions Our findings in mice are the first to suggest the involvement of the NKG2D/RAE-1 interaction and cytokines in the synergistic effects of NK and Kupffer cells in PBC.
Collapse
Affiliation(s)
- Hai-Yan Fu
- Department of Gastroenterology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Wei-Min Bao
- Department of Hepatobiliary Surgery, First People's Hospital of Yunnan, Kunming, Yunnan, China (mainland)
| | - Cai-Xia Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Wei-Ju Lai
- Department of Gastroenterology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Jia-Min Xu
- Department of Gastroenterology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Hai-Yan Yu
- Department of Gastroenterology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Yi-Na Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Xu Tan
- Department of Gastroenterology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Ajay Kumar Gupta
- Department of Gastroenterology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| | - Ying-Mei Tang
- Department of Gastroenterology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China (mainland)
| |
Collapse
|
12
|
Cao H, Zhu B, Qu Y, Zhang W. Abnormal Expression of ERα in Cholangiocytes of Patients With Primary Biliary Cholangitis Mediated Intrahepatic Bile Duct Inflammation. Front Immunol 2019; 10:2815. [PMID: 31867004 PMCID: PMC6907097 DOI: 10.3389/fimmu.2019.02815] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 11/15/2019] [Indexed: 01/09/2023] Open
Abstract
ERα, one of the classical receptors of estrogen, has been found to be abnormally up-regulated in patients with primary biliary cholangitis (PBC), which is an important factor leading to ductopenia. ERα-mediated signaling pathways are involved in proliferation of human intrahepatic biliary epithelial cells (HiBECs) and portal inflammation. Our previous studies have shown that the expression levels of ERα in the liver tissues of PBC patients are positively correlated with the levels of serum pro-inflammatory cytokines. The present study was designed to assess the relationship between abnormal ERα expression in small bile ducts and the progression of PBC. We examined the levels of multiple cytokines and analyzed their relationship with clinical parameters of livers functions in a cohort of 43 PBC patients and 45 healthy controls (HC). The levels of ERα expression and the relation with the levels of cytokines were further assessed. The localization of cytokines and ERα-mediated signaling pathways in liver were examined using immunohistochemistry. The possible underlying mechanisms of these alterations in PBC were explored in vitro. Our results demonstrated that the levels of IL-6, IL-8, and TNF-α were increased in PBC patients, and positively correlated with the serum AKP levels and ERα expression levels. Moreover, the expression of these cytokines were up-regulated in HiBECs that were stimulated with 17β-estradiol and PPT (an ERα agonist) and they also were positive in intrahepatic bile duct of PBC patients. The ERα-mediated expression of pro-inflammatory cytokines was induced by JNK, P38, and STAT3 phosphorylation in HiBECs. In addition, the CD54 expression was increased in HiBECs after ERα activation, which induced peripheral blood monouclear cells (PBMCs) recruitment. In conclusion, the present study highlighted a key role of abnormal ERα expression in inducing an inflammatory phenotype of HiBECs, which was critical in the development of inflammation and damage in small bile duct.
Collapse
Affiliation(s)
| | | | | | - Wei Zhang
- Department of Liver Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
13
|
Luo M, Peng H, Chen P, Zhou Y. The immunomodulatory role of interleukin-35 in fibrotic diseases. Expert Rev Clin Immunol 2019; 15:431-439. [PMID: 30590954 DOI: 10.1080/1744666x.2019.1564041] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Fibrosis makes numerous diseases in all organs more complicated and leads to severe consequences in the lung, liver, heart, kidney, and skin. In essence, fibrosis results from excessive, persistent and oftentimes nonreversible aggregation of extracellular matrix (ECM) or simply as collagen during the process of tissue injury and repair. Recent studies suggest the pathology of fibrosis, especially in pulmonary and liver fibrosis, involves various types of immune cells and soluble mediators including interleukin (IL)-35, a recently identified heterodimeric cytokine that belongs to the IL-12 cytokine family. Furthermore, IL-35 may inhibit fibrotic diseases. However, the side effects of inhibiting IL-35 also need attention and we have a long way to go to make better use of it in fibrotic diseases. Areas covered: This review focuses on recent evidence regarding the role of IL-35 in the pathogenesis of pulmonary, hepatic, cardiac, renal and skin fibrosis. It also discusses targeting of IL-35 as a promising novel strategy for treatment of fibrotic diseases. Expert commentary: Understanding as fully as possible the relationship between IL-35 and fibrotic diseases is important for the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Man Luo
- a Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital , Central South University , Changsha , China.,b Research Unit of Respiratory Disease , Central South University , Changsha , China.,c Diagnosis and Treatment Center of Respiratory Disease , Central South University , Changsha , China
| | - Hong Peng
- a Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital , Central South University , Changsha , China.,b Research Unit of Respiratory Disease , Central South University , Changsha , China.,c Diagnosis and Treatment Center of Respiratory Disease , Central South University , Changsha , China
| | - Ping Chen
- a Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital , Central South University , Changsha , China.,b Research Unit of Respiratory Disease , Central South University , Changsha , China.,c Diagnosis and Treatment Center of Respiratory Disease , Central South University , Changsha , China
| | - Yong Zhou
- d Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine , University of Alabama at Birmingham , Birmingham , AL , USA
| |
Collapse
|