1
|
Xing Z, Liu S, He X. Critical and diverse role of alarmin cytokines in parasitic infections. Front Cell Infect Microbiol 2024; 14:1418500. [PMID: 39559705 PMCID: PMC11570582 DOI: 10.3389/fcimb.2024.1418500] [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: 04/16/2024] [Accepted: 10/17/2024] [Indexed: 11/20/2024] Open
Abstract
Alarmin cytokines including IL-25, IL-33, and thymic stromal lymphopoietin (TSLP) function as danger signals to trigger host immunity in response to tissue injury caused by pathogenic factors such as parasitic infections. Parasitic diseases also provide an excellent context to study their functions and mechanisms. Numerous studies have indicated that alarmin cytokine released by non-immune cells such as epithelial and stromal cells induce the hosts to initiate a type 2 immunity that drives parasite expulsion but also host pathology such as tissue injury and fibrosis. By contrast, alarmin cytokines especially IL-33 derived from immune cells such as dendritic cells may elicit an immuno-suppressive milieu that promotes host tolerance to parasites. Additionally, the role of alarmin cytokines in parasite infections is reported to depend on species of parasites, cellular source of alarmin cytokines, and immune microenvironment, all of which is relevant to the parasitic sites or organs. This narrative review aims to provide information on the crucial and diverse role of alarmin cytokines in parasitic infections involved in different organs including intestine, lung, liver and brain.
Collapse
Affiliation(s)
- Zhou Xing
- Department of Tropical Diseases, Naval Medical University, Shanghai, China
| | - Suiyi Liu
- Department of Medical Engineering, Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Xing He
- Department of Tropical Diseases, Naval Medical University, Shanghai, China
| |
Collapse
|
2
|
He PY, Wu MY, Zheng LY, Duan Y, Fan Q, Zhu XM, Yao YM. Interleukin-33/serum stimulation-2 pathway: Regulatory mechanisms and emerging implications in immune and inflammatory diseases. Cytokine Growth Factor Rev 2024; 76:112-126. [PMID: 38155038 DOI: 10.1016/j.cytogfr.2023.12.001] [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: 11/30/2023] [Accepted: 12/11/2023] [Indexed: 12/30/2023]
Abstract
Interleukin (IL)- 33, a nuclear factor and pleiotropic cytokine of the IL-1 family, is gaining attention owing to its important role in chronic inflammatory and autoimmune diseases. This review extends our knowledge of the effects exerted by IL-33 on target cells by binding to its specific receptor serum stimulation-2 (ST2). Depending on the tissue context, IL-33 performs multiple functions encompassing host defence, immune response, initiation and amplification of inflammation, tissue repair, and homeostasis. The levels and activity of IL-33 in the body are controlled by complex IL-33-targeting regulatory pathways. The unique temporal and spatial expression patterns of IL-33 are associated with host homeostasis and the development of immune and inflammatory disorders. Therefore, understanding the origin, function, and processes of IL-33 under various conditions is crucial. This review summarises the regulatory mechanisms underlying the IL-33/ST2 signalling axis and its potential role and clinical significance in immune and inflammatory diseases, and discusses the current complex and conflicting findings related to IL-33 in host responses.
Collapse
Affiliation(s)
- Peng-Yi He
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; School of Medicine, Nankai University, Tianjin 300071, China
| | - Meng-Yao Wu
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Li-Yu Zheng
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Yu Duan
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Qi Fan
- Emergency Medicine Center, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Xiao-Mei Zhu
- Tissue Repair and Regeneration Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100048, China.
| | - Yong-Ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; School of Medicine, Nankai University, Tianjin 300071, China.
| |
Collapse
|
3
|
Li QF, Li YX, Yang YY, Dong PP, Mei CJ, Lu JL, Zhang JF, Hua HY, Xiong CR, Yu CX, Song LJ, Yang K. The egg ribonuclease SjCP1412 accelerates liver fibrosis caused by Schistosoma japonicum infection involving damage-associated molecular patterns (DAMPs). Parasitology 2024; 151:260-270. [PMID: 38105713 PMCID: PMC11007278 DOI: 10.1017/s0031182023001361] [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: 09/21/2023] [Revised: 11/16/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Schistosomiasis, a parasite infectious disease caused by Schistosoma japonicum, often leads to egg granuloma and fibrosis due to the inflammatory reaction triggered by egg antigens released in the host liver. This study focuses on the role of the egg antigens CP1412 protein of S. japonicum (SjCP1412) with RNase activity in promoting liver fibrosis. In this study, the recombinant egg ribonuclease SjCP1412, which had RNase activity, was successfully prepared. By analysing the serum of the population, it has been proven that the anti-SjCP1412 IgG in the serum of patients with advanced schistosomiasis was moderately correlated with liver fibrosis, and SjCP1412 may be an important antigen associated with liver fibrosis in schistosomiasis. In vitro, the rSjCP1412 protein induced the human liver cancer cell line Hep G2 and liver sinusoidal endothelial cells apoptosis and necrosis and the release of proinflammatory damage-associated molecular patterns (DAMPs). In mice infected with schistosomes, rSjCP1412 immunization or antibody neutralization of SjCP1412 activity significantly reduced cell apoptosis and necroptosis in liver tissue, thereby reducing inflammation and liver fibrosis. In summary, the SjCP1412 protein plays a crucial role in promoting liver fibrosis during schistosomiasis through mediating the liver cells apoptosis and necroptosis to release DAMPs inducing an inflammatory reaction. Blocking SjCP1412 activity could inhibit its proapoptotic and necrotic effects and alleviate hepatic fibrosis. These findings suggest that SjCP1412 may be served as a promising drug target for managing liver fibrosis in schistosomiasis japonica.
Collapse
Affiliation(s)
- Qi-Feng Li
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu 214064, China
| | - Yi-Xin Li
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu 214064, China
| | - Ying-Ying Yang
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu 214064, China
| | - Pan-Pan Dong
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu 214064, China
| | - Cong-Jin Mei
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu 214064, China
| | - Ju-Lu Lu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu 214064, China
| | - Jian-Feng Zhang
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu 214064, China
| | - Hai-Yong Hua
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu 214064, China
| | - Chun-Rong Xiong
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu 214064, China
| | - Chuan-Xin Yu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu 214064, China
| | - Li-Jun Song
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu 214064, China
| | - Kun Yang
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu 214064, China
| |
Collapse
|
4
|
Kimura M, Kothari S, Gohir W, Camargo JF, Husain S. MicroRNAs in infectious diseases: potential diagnostic biomarkers and therapeutic targets. Clin Microbiol Rev 2023; 36:e0001523. [PMID: 37909789 PMCID: PMC10732047 DOI: 10.1128/cmr.00015-23] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023] Open
Abstract
MicroRNAs (miRNAs) are conserved, short, non-coding RNAs that play a crucial role in the post-transcriptional regulation of gene expression. They have been implicated in the pathogenesis of cancer and neurological, cardiovascular, and autoimmune diseases. Several recent studies have suggested that miRNAs are key players in regulating the differentiation, maturation, and activation of immune cells, thereby influencing the host immune response to infection. The resultant upregulation or downregulation of miRNAs from infection influences the protein expression of genes responsible for the immune response and can determine the risk of disease progression. Recently, miRNAs have been explored as diagnostic biomarkers and therapeutic targets in various infectious diseases. This review summarizes our current understanding of the role of miRNAs during viral, fungal, bacterial, and parasitic infections from a clinical perspective, including critical functional mechanisms and implications for their potential use as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Muneyoshi Kimura
- Transplant Infectious Diseases, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Sagar Kothari
- Transplant Infectious Diseases, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Wajiha Gohir
- Transplant Infectious Diseases, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Jose F. Camargo
- Department of Medicine, Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Shahid Husain
- Transplant Infectious Diseases, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Maggi L, Camelo GMA, Rocha IC, Pereira Alves W, Moreira JMP, Almeida Pereira T, Tafuri WL, Rabelo ÉML, Correa A, Ecco R, Negrão-Corrêa DA. Role of the IL-33/ST2 Activation Pathway in the Development of the Hepatic Fibrosis Induced by Schistosoma mansoni Granulomas in Mice. Int J Mol Sci 2023; 24:10237. [PMID: 37373379 PMCID: PMC10299179 DOI: 10.3390/ijms241210237] [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: 05/26/2023] [Revised: 06/06/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Schistosoma mansoni eggs retained in host tissues induce innate cytokine release, contributing to the induction of Type-2 immune responses and granuloma formation, important to restrain cytotoxic antigens, but leading to fibrosis. Interleukin(IL)-33 participates in experimental models of inflammation and chemically induced fibrosis, but its role in S. mansoni-induced fibrosis is still unknown. To explore the role of the IL-33/suppressor of the tumorigenicity 2 (ST2) pathway, serum and liver cytokine levels, liver histopathology, and collagen deposition were comparatively evaluated in S. mansoni-infected wild-type (WT) and IL-33-receptor knockout (ST2-/-) BALB/c mice. Our data show similar egg counts and hydroxyproline in the livers of infected WT and ST2-/- mice; however, the extracellular matrix in ST2-/- granulomas was loose and disorganised. Pro-fibrotic cytokines, such as IL-13 and IL-17, and the tissue-repairing IL-22 were significantly lower in ST2-/- mice, especially in chronic schistosomiasis. ST2-/- mice also showed decreased α-smooth muscle actin (α-SMA) expression in granuloma cells, in addition to reduced Col III and Col VI mRNA levels and reticular fibres. Therefore, IL-33/ST2 signalling is essential for tissue repairing and myofibroblast activation during S. mansoni infection. Its disruption results in inappropriate granuloma organisation, partly due to the reduced type III and VI collagen and reticular fibre formation.
Collapse
Affiliation(s)
- Laura Maggi
- Laboratório de Esquistossomose e Imunohelmintologia, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (L.M.); (G.M.A.C.); (I.C.R.); (J.M.P.M.)
| | - Genil Mororó Araújo Camelo
- Laboratório de Esquistossomose e Imunohelmintologia, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (L.M.); (G.M.A.C.); (I.C.R.); (J.M.P.M.)
| | - Izabella Chrystina Rocha
- Laboratório de Esquistossomose e Imunohelmintologia, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (L.M.); (G.M.A.C.); (I.C.R.); (J.M.P.M.)
- Curso de Enfermagem, Instituto de Ciências Biológicas e Saúde, Universidade Federal de Mato Grosso, Barra do Garça 78698-000, MG, Brazil
| | - William Pereira Alves
- Laboratório de Parasitologia Molecular, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (W.P.A.); (É.M.L.R.)
| | - João Marcelo Peixoto Moreira
- Laboratório de Esquistossomose e Imunohelmintologia, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (L.M.); (G.M.A.C.); (I.C.R.); (J.M.P.M.)
| | - Thiago Almeida Pereira
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Wagner Luiz Tafuri
- Laboratório de Patologia das Leishmanioses, Departamento de Patologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil;
| | - Élida Mara Leite Rabelo
- Laboratório de Parasitologia Molecular, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (W.P.A.); (É.M.L.R.)
| | - Ary Correa
- Laboratório de Micologia, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil;
| | - Roselene Ecco
- Setor de Patologia, Escola Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil;
| | - Deborah Aparecida Negrão-Corrêa
- Laboratório de Esquistossomose e Imunohelmintologia, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (L.M.); (G.M.A.C.); (I.C.R.); (J.M.P.M.)
| |
Collapse
|
6
|
Kong H, Song Q, Hu W, Guo S, Xiang D, Huang S, Xu X, He J, Pan L, Tao R, Yu H, Huang J. MicroRNA-29a-3p prevents Schistosoma japonicum-induced liver fibrosis by targeting Roundabout homolog 1 in hepatic stellate cells. Parasit Vectors 2023; 16:184. [PMID: 37280619 DOI: 10.1186/s13071-023-05791-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/27/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND Schistosomiasis is a serious but neglected parasitic disease in humans that may lead to liver fibrosis and death. Activated hepatic stellate cells (HSCs) are the principal effectors that promote the accumulation of extracellular matrix (ECM) proteins during hepatic fibrosis. Aberrant microRNA-29 expression is involved in the development of fibrotic diseases. However, less is known about the role of miR-29 in Schistosoma japonicum (S. japonicum)-induced hepatic fibrosis. METHODS The levels of microRNA-29a-3p (miR-29a-3p) and Roundabout homolog 1 (Robo1) were examined in liver tissues during S. japonicum infection. The possible involvement of the miR-29a-3p-Robo1 signaling pathway was determined. We used MIR29A conditional knock-in mice and mice injected with an miR-29a-3p agomir to investigate the role of miR-29a-3p in schistosomiasis-induced hepatic fibrosis. The functional contributions of miR-29a-3p-Robo1 signaling in liver fibrosis and HSC activation were investigated using primary mouse HSCs and the human HSC cell line LX-2. RESULTS MiR-29a-3p was downregulated in humans and mice with schistosome-induced fibrosis, and Robo1 was upregulated in liver tissues. The miR-29a-3p targeted Robo1 and negatively regulated its expression. Additionally, the expression level of miR-29a-3p in schistosomiasis patients was highly correlated with the portal vein and spleen thickness diameter, which represent the severity of fibrosis. Furthermore, we demonstrated that efficient and sustained elevation of miR-29a-3p reversed schistosome-induced hepatic fibrosis. Notably, we showed that miR-29a-3p targeted Robo1 in HSCs to prevent the activation of HSCs during infection. CONCLUSIONS Our results provide experimental and clinical evidence that the miR-29a-3p-Robo1 signaling pathway in HSCs plays an important role in the development of hepatic fibrosis. Therefore, our study highlights the potential of miR-29a-3p as a therapeutic intervention for schistosomiasis and other fibrotic diseases.
Collapse
Affiliation(s)
- Hongyan Kong
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiqin Song
- Cancer Institute, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Peking University Shenzhen Hospital, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, China
| | - Wenjiang Hu
- Department of Gastroenterology, The People's Hospital of Jianshi, Enshi, China
| | - Shusen Guo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dandan Xiang
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuaiwen Huang
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Xu
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinan He
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lanyue Pan
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ran Tao
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haijing Yu
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaquan Huang
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
7
|
Xie L, Long X, Mo M, Jiang J, Zhang Q, Long M, Li M. Bone marrow mesenchymal stem cell-derived exosomes alleviate skin fibrosis in systemic sclerosis by inhibiting the IL-33/ST2 axis via the delivery of microRNA-214. Mol Immunol 2023; 157:146-157. [PMID: 37028129 DOI: 10.1016/j.molimm.2023.03.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/24/2023] [Accepted: 03/23/2023] [Indexed: 04/09/2023]
Abstract
Interleukin (IL)- 33 is a tissue-derive proinflammatory cytokine that promotes fibrosis in systemic sclerosis (SSc). microRNA (miR)- 214 expression has been elaborated to be downregulated in SSc patients and exert anti-fibrotic and anti-inflammatory effects. This study elucidates the role of bone marrow mesenchymal stem cell-derived exosome (BMSC-Exos)-delivered miR-214 in SSc and the relationship between this miR and IL-33/ST2 axis. SSc clinical samples were obtained to evaluate levels of miR-214, IL-33, and ST2. Primary fibroblasts and BMSC-Exos were extracted, followed by the co-culture of PKH6-labeled BMSC-Exos and fibroblasts. Subsequently, Exos extracted from miR-214 inhibitor-transfected BMSCs were co-cultured with TGF-β1-stimulated fibroblasts, after which the expression of fibrotic markers, miR-214, IL-33, and ST2, as well as fibroblast proliferation and migration, was determined. A skin fibrosis mouse model was induced with bleomycin (BLM) and treated with BMSC-Exos. Collagen fiber accumulation, collagen content, α-SMA expression, and IL-33 and ST2 levels were examined in BLM-treated or IL-33-knockout mice. IL-33 and ST2 were upregulated and miR-214 was downregulated in SSc patients. Mechanistically, miR-214 targeted IL-33 and blocked the IL-33/ST2 axis. BMSC-Exos delivering miR-214 inhibitor augmented proliferation, migration, and fibrotic gene expression in TGF-β1-stimulated fibroblasts. Similarly, IL-33 induced migration, proliferation, and fibrotic gene expression in fibroblasts via ST2. In BLM-treated mice, IL-33 knockout suppressed skin fibrosis, and BMSC-Exos delivered miR-214 to suppress the IL-33/ST2 axis, thus mitigating skin fibrosis. Conclusively, BMSC-Exos alleviate skin fibrosis through the blockade of the IL-33/ST2 axis by delivering miR-214.
Collapse
Affiliation(s)
- Lihu Xie
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Xiaoping Long
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Meili Mo
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Jinmei Jiang
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Qingxiu Zhang
- Department of Rehabilitation, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Mei Long
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China
| | - Mei Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
8
|
microRNAs: Critical Players during Helminth Infections. Microorganisms 2022; 11:microorganisms11010061. [PMID: 36677353 PMCID: PMC9861972 DOI: 10.3390/microorganisms11010061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
microRNAs (miRNAs) are a group of small non-coding RNAs that regulate gene expression post-transcriptionally through their interaction with the 3' untranslated regions (3' UTR) of target mRNAs, affecting their stability and/or translation. Therefore, miRNAs regulate biological processes such as signal transduction, cell death, autophagy, metabolism, development, cellular proliferation, and differentiation. Dysregulated expression of microRNAs is associated with infectious diseases, where miRNAs modulate important aspects of the parasite-host interaction. Helminths are parasitic worms that cause various neglected tropical diseases affecting millions worldwide. These parasites have sophisticated mechanisms that give them a surprising immunomodulatory capacity favoring parasite persistence and establishment of infection. In this review, we analyze miRNAs in infections caused by helminths, emphasizing their role in immune regulation and its implication in diagnosis, prognosis, and the development of therapeutic strategies.
Collapse
|
9
|
Xu L, Wei C, Chen Y, Wu Y, Shou X, Chen W, Lu D, Sun H, Li W, Yu B, Wang X, Zhang X, Yu Y, Lei Z, Tang R, Zhu J, Li Y, Lu L, Zhou H, Zhou S, Su C, Chen X. IL-33 induces thymic involution-associated naive T cell aging and impairs host control of severe infection. Nat Commun 2022; 13:6881. [PMID: 36371464 PMCID: PMC9653498 DOI: 10.1038/s41467-022-34660-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 11/01/2022] [Indexed: 11/13/2022] Open
Abstract
Severe infection commonly results in immunosuppression, which leads to impaired pathogen clearance or increased secondary infection in both humans and animals. However, the exact mechanisms remain poorly understood. Here, we demonstrate that IL-33 results in immunosuppression by inducing thymic involution-associated naive T cell dysfunction with aberrant expression of aging-associated genes and impairs host control of infection in mouse disease models of schistosomiasis or sepsis. Furthermore, we illustrate that IL-33 triggers the excessive generation of medullary thymic epithelial cell (mTEC) IV (thymic tuft cells) in a Pou2f3-dependent manner, as a consequence, disturbs mTEC/cortical TEC (cTEC) compartment and causes thymic involution during severe infection. More importantly, IL-33 deficiency, the anti-IL-33 neutralizing antibody treatment, or IL-33 receptor ST2 deficient thymus transplantation rescues T cell immunity to better control infection in mice. Our findings not only uncover a link between severe infection-induced IL-33 and thymic involution-mediated naive T cell aging, but also suggest that targeting IL-33 or ST2 is a promising strategy to rejuvenate T cell immunity to better control severe infection.
Collapse
Affiliation(s)
- Lei Xu
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Respiratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Clinical Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006 P. R. China
| | - Chuan Wei
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Ying Chen
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Yue Wu
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Xiaoli Shou
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Wenjie Chen
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Di Lu
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Haoran Sun
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Wei Li
- grid.89957.3a0000 0000 9255 8984Department of Clinical Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006 P. R. China
| | - Beibei Yu
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Xiaowei Wang
- grid.452511.6Department of Blood Transfusion, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008 P. R. China
| | - Xiaojun Zhang
- grid.452511.6Imaging Center, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu 210008 P. R. China
| | - Yanxiong Yu
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Zhigang Lei
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Rui Tang
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Jifeng Zhu
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Yalin Li
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Linrong Lu
- grid.13402.340000 0004 1759 700XInstitute of Immunology, School of Medicine, Zhejiang University, Hangzhou, 310058 P. R. China
| | - Hong Zhou
- grid.186775.a0000 0000 9490 772XDepartment of Cell Biology, School of Life Sciences, Anhui Medical University, Hefei, 230032 P. R. China
| | - Sha Zhou
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Chuan Su
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| | - Xiaojun Chen
- grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China ,grid.89957.3a0000 0000 9255 8984Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu 211166 P. R. China
| |
Collapse
|
10
|
Liu Z, Zhang L, Liang Y, Lu L. Pathology and molecular mechanisms of Schistosoma japonicum-associated liver fibrosis. Front Cell Infect Microbiol 2022; 12:1035765. [PMID: 36389166 PMCID: PMC9650140 DOI: 10.3389/fcimb.2022.1035765] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/13/2022] [Indexed: 11/23/2022] Open
Abstract
Schistosomiasis has been widely disseminated around the world, and poses a significant threat to human health. Schistosoma eggs and soluble egg antigen (SEA) mediated inflammatory responses promote the formation of egg granulomas and liver fibrosis. With continuous liver injuries and inflammatory stimulation, liver fibrosis can develop into liver cirrhosis and liver cancer. Therefore, anti-fibrotic therapy is crucial to increase the survival rate of patients. However, current research on antifibrotic treatments for schistosomiasis requires further exploration. In the complicated microenvironment of schistosome infections, it is important to understand the mechanism and pathology of schistosomiasis-associated liver fibrosis(SSLF). In this review, we discuss the role of SEA in inhibiting liver fibrosis, describe its mechanism, and comprehensively explore the role of host-derived and schistosome-derived microRNAs (miRNAs) in SSLF. Inflammasomes and cytokines are significant factors in promoting SSLF, and we discuss the mechanisms of some critical inflammatory signals and pro-fibrotic cytokines. Natural killer(NK) cells and Natural killer T(NKT) cells can inhibit SSLF but are rarely described, therefore, we highlight their significance. This summarizes and provides insights into the mechanisms of key molecules involved in SSLF development.
Collapse
Affiliation(s)
- Zhilong Liu
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang, China
| | - Lichen Zhang
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang, China
| | - Yinming Liang
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
- *Correspondence: Yinming Liang, ; Liaoxun Lu,
| | - Liaoxun Lu
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
- *Correspondence: Yinming Liang, ; Liaoxun Lu,
| |
Collapse
|
11
|
miR-182-5p attenuates Schistosoma japonicum-induced hepatic fibrosis by targeting tristetraprolin. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1421-1430. [PMID: 36148947 PMCID: PMC9828319 DOI: 10.3724/abbs.2022130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Egg granuloma formation in the liver is the main pathological lesion caused by Schistosoma japonicum infection, which generally results in liver fibrosis and may lead to death in advanced patients. MicroRNAs (miRNAs) regulate the process of liver fibrosis, but the putative function of miRNAs in liver fibrosis induced by S. japonicum infection is largely unclear. Here, we detect a new miRNA, miR-182-5p, which shows significantly decreased expression in mouse livers after stimulation by soluble egg antigen (SEA) of S. japonicum or S. japonicum infection. Knockdown or overexpression of miR-182-5p in vitro causes the increased or decreased expression of tristetraprolin (TTP), an important immunosuppressive protein in the process of liver fibrosis. Furthermore, knockdown of miR-182-5p in vivo upregulates TTP expression and significantly alleviates S. japonicum-induced hepatic fibrosis. Our data demonstrate that downregulation of miR-182-5p increases the expression of TTP in mouse livers following schistosome infection, which leads to destabilization of inflammatory factor mRNAs and attenuates liver fibrosis. Our results uncover fine-tuning of liver inflammatory reactions related to liver fibrosis caused by S. japonicum infection and provide new insights into the regulation of schistosomiasis-induced hepatic fibrosis.
Collapse
|
12
|
Abdel Aziz N, Musaigwa F, Mosala P, Berkiks I, Brombacher F. Type 2 immunity: a two-edged sword in schistosomiasis immunopathology. Trends Immunol 2022; 43:657-673. [PMID: 35835714 DOI: 10.1016/j.it.2022.06.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/13/2022] [Accepted: 06/13/2022] [Indexed: 12/14/2022]
Abstract
Schistosomiasis is the second most debilitating neglected tropical disease globally after malaria, with no available therapy to control disease-driven immunopathology. Although schistosomiasis induces a markedly heterogenous immune response, type 2 immunity is the dominating immune response following oviposition. While type 2 immunity has a crucial role in granuloma formation and host survival during the acute stage of disease, its chronic activation can result in tissue scarring, fibrosis, and organ impairment. Here, we discuss recent advances in schistosomiasis, demonstrating how different immune and non-immune cells and signaling pathways are involved in the induction, maintenance, and regulation of type 2 immunity. A better understanding of these immune responses during schistosomiasis is essential to inform the potential development of candidate therapeutic strategies that fine-tune type 2 immunity to ideally modulate schistosomiasis immunopathology.
Collapse
Affiliation(s)
- Nada Abdel Aziz
- Cytokines and Diseases Group, International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Biotechnology/Biomolecular Chemistry Program, Biotechnology Department, Faculty of Science, Cairo University, Cairo, Egypt; Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa.
| | - Fungai Musaigwa
- Cytokines and Diseases Group, International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Paballo Mosala
- Cytokines and Diseases Group, International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Inssaf Berkiks
- Cytokines and Diseases Group, International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Frank Brombacher
- Cytokines and Diseases Group, International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa.
| |
Collapse
|
13
|
Zhong H, Jin Y. Multifunctional Roles of MicroRNAs in Schistosomiasis. Front Microbiol 2022; 13:925386. [PMID: 35756064 PMCID: PMC9218868 DOI: 10.3389/fmicb.2022.925386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/20/2022] [Indexed: 11/22/2022] Open
Abstract
Schistosomiasis is a parasitic disease that is caused by helminths of the genus Schistosoma. The dioecious schistosomes mate and lay eggs after undergoing a complex life cycle. Schistosome eggs are mostly responsible for the transmission of schistosomiasis and chronic fibrotic disease induced by egg antigens is the main cause of the high mortality rate. Currently, chemotherapy with praziquantel (PZQ) is the only effective treatment against schistosomiasis, although the potential of drug resistance remains a concern. Hence, there is an urgent demand for new and effective strategies to combat schistosomiasis, which is the second most prevalent parasitic disease after malaria. MicroRNAs (miRNAs) are small non-coding RNAs that play pivotal regulatory roles in many organisms, including the development and sexual maturation of schistosomes. Thus, miRNAs are potential targets for treatment of schistosomiasis. Moreover, miRNAs can serve as multifunctional “nano-tools” for cross-species delivery in order to regulate host-parasite interactions. In this review, the multifunctional roles of miRNAs in the growth and development of schistosomes are discussed. The various regulatory functions of host-derived and worm-derived miRNAs on the progression of schistosomiasis are also thoroughly addressed, especially the promotional and inhibitory effects on schistosome-induced liver fibrosis. Additionally, the potential of miRNAs as biomarkers for the diagnosis and treatment of schistosomiasis is considered.
Collapse
Affiliation(s)
- Haoran Zhong
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China.,Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yamei Jin
- National Reference Laboratory for Animal Schistosomiasis, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China.,Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| |
Collapse
|
14
|
Hu Y, Wang X, Ding F, Liu C, Wang S, Feng T, Meng S. Periostin renders cardiomyocytes vulnerable to acute myocardial infarction via pro-apoptosis. ESC Heart Fail 2022; 9:977-987. [PMID: 35104050 PMCID: PMC8934967 DOI: 10.1002/ehf2.13675] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/07/2021] [Accepted: 10/05/2021] [Indexed: 11/08/2022] Open
Abstract
AIMS As a severe cardiovascular disease, acute myocardial infarction (AMI) could trigger congestive heart failure. Periostin (Postn) has been elucidated to be dramatically up-regulated in myocardial infarction. Abundant expression of Postn was also observed in the infarct border of human and mouse hearts with AMI. This work is dedicated to explore the mechanism through which Postn exerts its functions on AMI. METHODS AND RESULTS The expression of Postn in AMI mice and hypoxia-treated neonatal mouse cardiomyocytes (NMCMs) was quantified by qRT-PCR. The biological functions of Postn in AMI were explored by trypan blue, TUNEL, flow cytometry analysis, and JC-1 assays. Luciferase activity or MS2-RIP or RNA pull-down assay was performed to study the interaction between genes. Postn exhibited up-regulated expression in AMI mice and hypoxia-treated NMCMs. Functional assays indicated that cell apoptosis in NMCMs was promoted via the treatment of hypoxia. And Postn shortage could alleviate cell apoptosis in hypoxia-induced NMCMs. Postn was verified to bind to mmu-miR-203-3p and be down-regulated by miR-203-3p overexpression. Postn and miR-203-3p were spotted to coexist with small nucleolar RNA host gene 8 (Snhg8) in RNA-induced silencing complex. The affinity between Snhg8 and miR-203-3p was confirmed. Afterwards, Snhg8 was validated to promote cell apoptosis in hypoxia-induced NMCMs partially dependent on Postn. Furthermore, vascular endothelial growth factor A (Vegfa) was revealed to bind to miR-203-3p and be implicated in the Snhg8-mediated AML cell apoptosis and angiogenesis. CONCLUSIONS miR-203-3p availability is antagonized by Snhg8 for Postn and Vegfa-induced AMI progression.
Collapse
Affiliation(s)
- Yanlei Hu
- Department of Cardiovascular Surgery ICUHeart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou UniversityNo. 1 Fuwai Avenue, Zhengdong New DistrictZhengzhouHenan451464China
| | - Xiaohang Wang
- Department of Cardiovascular Surgery ICUHeart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou UniversityNo. 1 Fuwai Avenue, Zhengdong New DistrictZhengzhouHenan451464China
| | - Fuyan Ding
- Department of Cardiovascular Surgery ICUHeart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou UniversityNo. 1 Fuwai Avenue, Zhengdong New DistrictZhengzhouHenan451464China
| | - Chao Liu
- Department of Cardiovascular Surgery ICUHeart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou UniversityNo. 1 Fuwai Avenue, Zhengdong New DistrictZhengzhouHenan451464China
| | - Shupeng Wang
- Department of Cardiovascular Surgery ICUHeart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou UniversityNo. 1 Fuwai Avenue, Zhengdong New DistrictZhengzhouHenan451464China
| | - Tao Feng
- Department of Cardiovascular Surgery ICUHeart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou UniversityNo. 1 Fuwai Avenue, Zhengdong New DistrictZhengzhouHenan451464China
| | - Shuping Meng
- Department of Cardiovascular Surgery ICUHeart Center of Henan Provincial People’s Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou UniversityNo. 1 Fuwai Avenue, Zhengdong New DistrictZhengzhouHenan451464China
| |
Collapse
|
15
|
Comparative characterization of microRNAs of Schistosoma japonicum from SCID mice and BALB/c mice: Clues to the regulation of parasite growth and development. Acta Trop 2022; 225:106200. [PMID: 34740636 DOI: 10.1016/j.actatropica.2021.106200] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 10/07/2021] [Accepted: 10/13/2021] [Indexed: 12/17/2022]
Abstract
Schistosomiasis, caused by a parasite with a wide range of mammalian hosts, remains one of the most prevailing parasitic diseases in the world. While numerous studies have reported that the growth and reproduction of schistosomes in immunodeficient mice was significantly retarded, the underlying molecular mechanisms have yet to be revealed. In this study, we comparatively analyzed the microRNA expression of Schistosoma japonicum derived from SCID and BALB/c mice on the 35th day post-infection by high-throughput RNA sequencing as prominent morphological abnormalities had been observed in schistosomes from SCID mice when compared with those from BALB/c mice. The results revealed that more than 72% and 61% of clean reads in the small RNA libraries of female and male schistosomes, respectively, could be mapped to the selected miRs in the miRBase or the sequences of species-specific genomes. Further analysis identified 122 miRNAs using TPM >0.01 as the threshold value, including 75 known and 47 novel miRNAs, 96 of which were commonly expressed across all the four tested schistosome libraries. Comparative analysis of the libraries of schistosomes from SCID and BALB/c mice identified 15 differentially expressed miRNAs (5 up-regulated and 10 down-regulated) among females and 16 among males (9 up-regulated and 7 down-regulated). Integrated analysis of the two sets of differentially expressed miRNAs of female and male worms identified 2 miRNAs (sja-miR-3488 and sja-miR-novel_29) that overlapped between female and male datasets. Prediction of miRNA targets and Gene Ontology (GO) term enrichment analysis of the predicted target genes revealed that these genes were involved in some important biological processes, such as nucleic acid metabolic process, macromolecule modification, and cellular aromatic compound metabolic process. The predicted target genes were further matched to the differentially expressed genes in male and female schistosomes from the above two hosts, obtaining 7 genes that may be responsible for regulating the growth, development and sex maturation of schistosomes. Taken together, this study provides the first identification of differentially expressed miRNAs in schistosomes from SCID and BALB/c mice. These miRNAs and their predicted target mRNAs are probably involved in the regulation of development, growth, and maturation of schistosomes. Therefore, this study expands our understanding of schistosome development regulation and host-parasite relationship, and also provides a valuable set of potential anti-schistosomal targets for prevention and control of schistosomiasis.
Collapse
|
16
|
Cai M, Ding J, Li Y, He G, Yang J, Liu T, Guo X, Yang X, Wang X, Cho WC, Fasihi Harandi M, Zheng Y. Echinococcus multilocularis infection induces UBE2N suppression via exosomal emu-miR-4989. Acta Trop 2021; 223:106087. [PMID: 34389329 DOI: 10.1016/j.actatropica.2021.106087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/23/2021] [Accepted: 08/04/2021] [Indexed: 11/22/2022]
Abstract
Echinococcus multilocularis metacestodes mainly reside in liver in humans and animals, and cause serious damages. UBE2N was herein shown to be downregulated in response to the infection. UBE2N was further shown to be predominantly expressed in the hepatocytes, which was also significantly downregulated during the infection. UBE2N was a target of emu-miR-4989, which was loaded into the exosomes secreted by parasites. These emu-miR-4989-encapsulating exosomes were internalized by hepatocytes, and induced a significant decrease of relative luciferase activity in the cells transfected with the construct containing a wild type of UBE2N 3'-UTR compared to the control (p < 0.05). These results demonstrate that emu-miR-4989 is involved in the UBE2N inhibition in the hepatocytes during E. multilocularis through exosomes.
Collapse
Affiliation(s)
- Mengting Cai
- College of Life Science and Technology, Xinjiang University, Urumqi, China; State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China
| | - Juntao Ding
- College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Yating Li
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China
| | - Guitian He
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China
| | - Jing Yang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China
| | - Tingli Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China
| | - Xiaola Guo
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China
| | - Xing Yang
- Department of Medical Microbiology and Immunology, School of Basic Medicine, Dali University, Dali 671000, Yunnan, China
| | - Xiaoqiang Wang
- School of Chemical and Biological Engineering, Lanzhou Jiaotong University, Lanzhou 730070, China
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Majid Fasihi Harandi
- Research Center for Hydatid Disease in Iran, Department of Parasitology, Kerman University of Medical Sciences, Kerman, Iran
| | - Yadong Zheng
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, CAAS, Lanzhou 730046, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| |
Collapse
|
17
|
Zhang B, Wu X, Li J, Ning A, Zhang B, Liu J, Song L, Yan C, Sun X, Zheng K, Wu Z. Hepatic progenitor cells promote the repair of schistosomiasis liver injury by inhibiting IL-33 secretion in mice. Stem Cell Res Ther 2021; 12:546. [PMID: 34674752 PMCID: PMC8529826 DOI: 10.1186/s13287-021-02589-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 09/04/2021] [Indexed: 01/20/2023] Open
Abstract
Background Hepatic schistosomiasis, a chronic liver injury induced by long-term Schistosoma japonicum (S. japonicum) infection, is characterized by egg granulomas and fibrotic pathology. Hepatic progenitor cells (HPCs), which are nearly absent or quiescent in normal liver, play vital roles in chronic and severe liver injury. But their role in the progression of liver injury during infection remains unknown. Methods In this study, the hepatic egg granulomas, fibrosis and proliferation of HPCs were analyzed in the mice model of S. japonicum infection at different infectious stages. For validating the role of HPCs in hepatic injury, tumor necrosis factor-like-weak inducer of apoptosis (TWEAK) and TWEAK blocking antibody were used to manipulate the proliferation of HPCs in wild-type and IL-33−/− mice infected with S. japonicum. Results We found that the proliferation of HPCs was accompanied by inflammatory granulomas and fibrosis formation. HPCs expansion promoted liver regeneration and inhibited inflammatory egg granulomas, as well as the deposition of fibrotic collagen. Interestingly, the expression of IL-33 was negatively associated with HPCs’ expansion. There were no obvious differences of liver injury caused by infection between wild-type and IL-33−/− mice with HPCs’ expansion. However, liver injury was more attenuated in IL-33−/− mice than wild-type mice when the proliferation of HPCs was inhibited by anti-TWEAK. Conclusions Our data uncovered a protective role of HPCs in hepatic schistosomiasis in an IL-33-dependent manner, which might provide a promising progenitor cell therapy for hepatic schistosomiasis. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02589-y.
Collapse
Affiliation(s)
- Beibei Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Key Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaoying Wu
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jing Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Key Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - An Ning
- Jiangxi Provincial Institute of Parasitic Diseases, Nanchang, Jiangxi, China
| | - Bo Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Key Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiahua Liu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Langui Song
- The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Chao Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Key Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xi Sun
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Key Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Zhongdao Wu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China. .,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
18
|
Lei Z, Tang R, Qi Q, Gu P, Wang J, Xu L, Wei C, Pu Y, Qi X, Chen Y, Yu B, Yu Y, Chen X, Zhu J, Li Y, Zhou S, Su C. Hepatocyte CD1d protects against liver immunopathology in mice with schistosomiasis japonica. Immunology 2020; 162:328-338. [PMID: 33283278 DOI: 10.1111/imm.13288] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/17/2020] [Accepted: 11/21/2020] [Indexed: 12/16/2022] Open
Abstract
Schistosomiasis is a neglected tropical disease with over 250 million people infected worldwide. The main clinically important species Schistosoma mansoni (S. mansoni) and Schistosoma japonicum (S. japonicum) cause inflammatory responses against tissue-trapped eggs, resulting in formation of granulomas mainly in host liver. Persistent granulomatous response results in severe fibrosis in the liver, leading to irreversible impairment of the liver and even death of the host. CD1d, a highly conserved MHC class I-like molecule, is expressed by both haematopoietic and non-haematopoietic cells. CD1d on antigen-presenting cells (APCs) of haematopoietic origin presents pathogen-derived lipid antigens to natural killer T (NKT) cells, which enables them to rapidly produce large amounts of various cytokines and facilitate CD4+ T helper (Th) cell differentiation upon invading pathogens. Noteworthy, hepatocytes of non-haematopoietic origin have recently been shown to be involved in maintaining liver NKT cell homeostasis through a CD1d-dependent manner. However, whether hepatocyte CD1d-dependent regulation of NKT cell homeostasis also modulates CD4+ Th cell responses and liver immunopathology in murine schistosomiasis remains to be addressed. Here, we show in mice that CD1d expression on hepatocytes was decreased dramatically upon S. japonicum infection, accompanied by increased NKT cells, as well as upregulated Th1 and Th2 responses. Overexpression of CD1d in hepatocytes significantly decreased local NKT numbers and cytokines (IFN-γ, IL-4, IL-13), concomitantly with downregulation of both Th1 and Th2 responses and alleviation in pathological damage in livers of S. japonicum-infected mice. These findings highlight the potential of hepatocyte CD1d-targeted therapies for liver immunopathology control in schistosomiasis.
Collapse
Affiliation(s)
- Zhigang Lei
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rui Tang
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qianqian Qi
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Pan Gu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junling Wang
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Xu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chuan Wei
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yanan Pu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xin Qi
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ying Chen
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Beibei Yu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yanxiong Yu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaojun Chen
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jifeng Zhu
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yalin Li
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Sha Zhou
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chuan Su
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Center for Global Health, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
19
|
Chen J, Xu Q, Zhang W, Zhen Y, Cheng F, Hua G, Lan J, Tu C. MiR-203-3p inhibits the oxidative stress, inflammatory responses and apoptosis of mice podocytes induced by high glucose through regulating Sema3A expression. Open Life Sci 2020; 15:939-950. [PMID: 33817280 PMCID: PMC7874591 DOI: 10.1515/biol-2020-0088] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/15/2020] [Accepted: 06/15/2020] [Indexed: 01/03/2023] Open
Abstract
Diabetic nephropathy (DN) is the most serious long-term microvascular complication of diabetes, which mainly causes podocyte injury. Many studies have shown that microRNAs play a vital role in the development of DN. Studies have shown that miR-203-3p is involved in mesangial cell proliferation and apoptosis of DN mice. Therefore, we speculated that miR-203-3p might be related to the development of DN, but our study does not provide any evidence. In animal experiments, diabetic mice (db/db) were transfected with iR-203-3p overexpression lentiviral vectors (LV-miR-203-3p) and their control (LV-miR-con), with normal mice (db/m) being used as the control. High glucose (HG)-induced podocytes were used to construct a DN cell model in vitro. The expression levels of miR-203-3p, Semaphorin 3A (Sema3A) and inflammatory cytokines were detected by quantitative real-time polymerase chain reaction. Also, serum creatinine and blood urea nitrogen levels were used to evaluate the degree of renal injury in DN mice. Sema3A and apoptosis-related protein levels were assessed by the western blot analysis. Enzyme-linked immunosorbent assay was used to determine the different oxidative stress-related indicators and inflammatory cytokines. Flow cytometry and caspase-3 activity detection were used to analyze the degree of podocyte apoptosis. Our results suggested that the expression of miR-203-3p was lower in DN mice and in HG-induced podocytes. Overexpression of miR-203-3p reduced the body weight, blood glucose and renal injury of DN mice in vivo, as well as relieve the oxidative stress, inflammatory response and apoptosis of HG-induced podocytes in vitro. Functionally, Sema3A was a target of miR-203-3p, and Sema3A overexpression reversed the inhibitory effect of miR-203-3p on HG-induced podocyte injury. Our findings revealed that miR-203-3p alleviated the podocyte injury induced by HG via regulating Sema3A expression, suggesting that miR-203-3p might be a new therapeutic target to improve the progression of DN.
Collapse
Affiliation(s)
- Jingfu Chen
- Department of Cardiovascular Medicine and Dongguan Cardiovascular Institute, The Third People's Hospital of Dongguan City, No. 1, Xianglong Road, Shi Long Town, Dongguan, China
| | - Qing Xu
- Department of Cardiology, Huangpu Division of The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Zhang
- Department of Cardiology, Huangpu Division of The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - YuLan Zhen
- Department of Oncology, The Third People's Hospital of Dongguan City, Dongguan, China
| | - Fei Cheng
- Department of Cardiovascular Medicine and Dongguan Cardiovascular Institute, The Third People's Hospital of Dongguan City, No. 1, Xianglong Road, Shi Long Town, Dongguan, China
| | - Guo Hua
- Department of Cardiovascular Medicine and Dongguan Cardiovascular Institute, The Third People's Hospital of Dongguan City, No. 1, Xianglong Road, Shi Long Town, Dongguan, China
| | - Jun Lan
- Department of Cardiovascular Medicine and Dongguan Cardiovascular Institute, The Third People's Hospital of Dongguan City, No. 1, Xianglong Road, Shi Long Town, Dongguan, China
| | - Chang Tu
- Department of Cardiovascular Medicine and Dongguan Cardiovascular Institute, The Third People's Hospital of Dongguan City, No. 1, Xianglong Road, Shi Long Town, Dongguan, China
| |
Collapse
|
20
|
Maggi L, Rocha IC, Camelo GMA, Fernandes VR, Negrão-Corrêa D. The IL-33/ST2 pathway is not essential to Th2 stimulation but is key for modulation and survival during chronic infection with Schistosoma mansoni in mice. Cytokine 2020; 138:155390. [PMID: 33341001 DOI: 10.1016/j.cyto.2020.155390] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 11/11/2020] [Accepted: 11/25/2020] [Indexed: 12/30/2022]
Abstract
Morbidity during chronic schistosomiasis has been associated with the induction and modulation of type-2 granulomatous inflammatory response induced by antigens secreted by the eggs, which become trapped in capillary venules of the host tissues, especially in the liver and intestines. IL-33, an alarmin released after cell damage, binds to its ST2 (suppressor of tumorigenicity 2) receptor, expressed in an variety of immune cells, including ILC2 and macrophages, and stimulates the early production of IL-5 and IL-13, which leads to eosinophil infiltration and activation of a Th2 response. However, the role of IL-33/ST2 activation on Schistosoma-induced granuloma formation and modulation is mostly unknown. In the current work, we comparatively evaluated the immune response and granuloma formation in wild-type BALB/c (WT) and BALB/c mice genetically deficient in the IL-33 receptor (ST2-/-) experimentally infected with Schistosoma mansoni. Mice were infected with 25 or 50 S. mansoni cercariae and followed for up to 14 weeks to assess mortality. Mice from each experimental group were comparatively evaluated for parasite burden, liver immune response, and granuloma appearance during acute and chronic schistosomiasis. Our data showed that the number of circulating worms and eggs retained in the liver and eliminated in the feces was similar in WT and ST2-/- infected mice, but infected ST2-/- mice presented an enhanced rate of mortality. Interestingly, the production of type-2 cytokines by soluble egg antigens (SEA)-stimulated spleen cells, the serum concentrations of IL-5 and Immunoglobulin (Ig)-E, and the level of parasite-reactive IgG1 were similar in infected mice of both experimental groups. The concentrations of IL-4, IL-5, IL-13, and IFN-γ in liver homogenate of infected mice also did not differ between the strains at acute schistosomiasis, but there was a significant increase in IL-17 levels in ST2-/- infected mice at this phase. On the other hand, IL-4, IL-13, IL-10, IL-17, and IFN-γ concentrations were reduced and the ratios of IL-4/IFN-γ and IL-17/IFN-γ were higher in liver homogenate of chronically infected ST2-/- mice, suggesting unbalanced Th2 and Th17 responses. Moreover, liver granulomas of ST2-/- mice were larger and disorganized, showing an intense cellular infiltrate, rich in eosinophils and neutrophils. Our results suggest that the absence of the IL-33/ST2 pathway is not essential for the Schistosoma-induced Th2 response, but is necessary to prevent host mortality by modulating granuloma-mediated pathology.
Collapse
Affiliation(s)
- Laura Maggi
- Laboratório de Esquistossomose e Imunohelmintologia, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Izabella Chrystina Rocha
- Laboratório de Esquistossomose e Imunohelmintologia, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Curso de Enfermagem, Instituto de Ciências Biológicas e Saúde, Universidade Federal de Mato Grosso, Barra do Garça, Brazil
| | - Genil Mororó Araújo Camelo
- Laboratório de Esquistossomose e Imunohelmintologia, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vanessa Rodrigues Fernandes
- Laboratório de Esquistossomose e Imunohelmintologia, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Deborah Negrão-Corrêa
- Laboratório de Esquistossomose e Imunohelmintologia, Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
21
|
IL-17A-producing γδ T cells promote liver pathology in acute murine schistosomiasis. Parasit Vectors 2020; 13:334. [PMID: 32611373 PMCID: PMC7329544 DOI: 10.1186/s13071-020-04200-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 06/20/2020] [Indexed: 02/08/2023] Open
Abstract
Background The main symptoms of schistosomiasis are granuloma and fibrosis, caused by Schistosoma eggs. Numerous types of cells and cytokines are involved in the progression of Schistosoma infection. As a class of innate immune cells, γδ T cells play critical roles in the early immune response. However, their role in modulating granuloma and fibrosis remains to be clarified. Methods Liver fibrosis in wild-type (WT) mice and T cell receptor (TCR) δ knockout (KO) mice infected with Schistosoma japonicum was examined via Masson’s trichrome staining of collagen deposition and quantitative reverse transcriptase-PCR (RT-PCR) of fibrosis-related genes. Granuloma was detected by hematoxylin-eosin (H&E) staining and quantified. Flow cytometry was used for immune cell profiling and for detecting cytokine secretion. The abundance of the related cytokines was measured using quantitative RT-PCR. Results The livers of S. japonicum-infected mice had significantly increased proportions of interleukin (IL)-17A producing γδ T cells and secreted IL-17A. Compared with the WT mice, TCR δ deficiency resulted in reduced pathological impairment and fibrosis in the liver and increased survival in infected mice. In addition, the profibrogenic effects of γδ T cells in infected mice were associated with enhanced CD11b+Gr-1+ cells, concurrent with increased expression of transforming growth factor (TGF)-β in the liver. Conclusions In this mouse model of Schistosoma infection, γδ T cells may promote liver fibrosis by recruiting CD11b+Gr-1+ cells. These findings shed new light on the pathogenesis of liver pathology in murine schistosomiasis.![]()
Collapse
|
22
|
Zheng B, Zhang J, Chen H, Nie H, Miller H, Gong Q, Liu C. T Lymphocyte-Mediated Liver Immunopathology of Schistosomiasis. Front Immunol 2020; 11:61. [PMID: 32132991 PMCID: PMC7040032 DOI: 10.3389/fimmu.2020.00061] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/10/2020] [Indexed: 12/16/2022] Open
Abstract
The parasitic worms, Schistosoma mansoni and Schistosoma japonicum, reside in the mesenteric veins, where they release eggs that induce a dramatic granulomatous response in the liver and intestines. Subsequently, infection may further develop into significant fibrosis and portal hypertension. Over the past several years, uncovering the mechanism of immunopathology in schistosomiasis has become a major research objective. It is known that T lymphocytes, especially CD4+ T cells, are essential for immune responses against Schistosoma species. However, obtaining a clear understanding of how T lymphocytes regulate the pathological process is proving to be a daunting challenge. To date, CD4+ T cell subsets have been classified into several distinct T helper (Th) phenotypes including Th1, Th2, Th17, T follicular helper cells (Tfh), Th9, and regulatory T cells (Tregs). In the case of schistosomiasis, the granulomatous inflammation and the chronic liver pathology are critically regulated by the Th1/Th2 responses. Animal studies suggest that there is a moderate Th1 response to parasite antigens during the acute stage, but then, egg-derived antigens induce a sustained and dominant Th2 response that mediates granuloma formation and liver fibrosis. In addition, the newly discovered Th17 cells also play a critical role in the hepatic immunopathology of schistosomiasis. Within the liver, Tregs are recruited to hepatic granulomas and exert an immunosuppressive role to limit the granulomatous inflammation and fibrosis. Moreover, recent studies have shown that Tfh and Th9 cells might also promote liver granulomas and fibrogenesis in the murine schistosomiasis. Thus, during infection, T-cell subsets undergo complicated cross-talk with antigen presenting cells that then defines their various roles in the local microenvironment for regulating the pathological progression of schistosomiasis. This current review summarizes a vast body of literature to elucidate the contribution of T lymphocytes and their associated cytokines in the immunopathology of schistosomiasis.
Collapse
Affiliation(s)
- Bing Zheng
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China.,Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Jianqiang Zhang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Hui Chen
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Hao Nie
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China.,Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Heather Miller
- Department of Intracellular Pathogens, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China.,Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| |
Collapse
|
23
|
Wang S, Li M, Zhao X, Wang H, Zhu J, Wang C, Zhou M, Dong H, Zhou R. Upregulation of KSRP by miR-27b attenuates schistosomiasis-induced hepatic fibrosis by targeting TGF-β1. FASEB J 2020; 34:4120-4133. [PMID: 31953889 DOI: 10.1096/fj.201902438r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/25/2019] [Accepted: 12/31/2019] [Indexed: 12/13/2022]
Abstract
Hepatic stellate cells (HSCs) are the main effectors for various types of hepatic fibrosis, including Schistosome-induced hepatic fibrosis. Multiple inflammatory cytokines/chemokines, such as transforming growth factor-β1 (TGF-β1), activate HSCs, and contribute to the development of hepatic fibrosis. MicroRNAs regulate gene expression at the posttranscriptional level and are involved in regulation of inflammatory cytokine/chemokine synthesis. In this study, we showed that soluble egg antigen (SEA) stimulation and Schistosoma japonicum infection downregulate miR-27b expression and increase KH-type splicing regulatory protein (KSRP) mRNA and protein levels in vitro and in vivo. miR-27b regulates the stabilization of TGF-β1 mRNA through targeting KSRP by interacting with their AU-rich elements in hepatocytes and non-parenchymal cells, which has an effect on the activation of HSCs. Importantly, our results have shown that either knockdown miR-27b or overexpression of KSRP attenuates S. japonicum-induced hepatic fibrosis in vivo. Therefore, our study highlights the crucial role of miR-27b and KSRP in the negative regulation of immune reactions in hepatocyte and non-parenchymal cells in response to SEA stimulation and S. japonicum infection. It reveals that manipulation of miR-27b or KSRP might be a useful strategy not only for treating Schistosome-induced hepatic fibrosis but also for curing hepatic fibrosis in general.
Collapse
Affiliation(s)
- Shuhong Wang
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Hubei, China.,Department of Medical Parasitology, School of Basic Medical Sciences, Wuhan University, Hubei, China
| | - Mingxuan Li
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Hubei, China.,Department of Medical Parasitology, School of Basic Medical Sciences, Wuhan University, Hubei, China
| | - Xuejun Zhao
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Hubei, China.,Department of Medical Parasitology, School of Basic Medical Sciences, Wuhan University, Hubei, China
| | - Huan Wang
- Department of Medical Parasitology, School of Basic Medical Sciences, Wuhan University, Hubei, China
| | - Jie Zhu
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Hubei, China.,Department of Medical Parasitology, School of Basic Medical Sciences, Wuhan University, Hubei, China
| | - Cheng Wang
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Hubei, China.,Department of Medical Parasitology, School of Basic Medical Sciences, Wuhan University, Hubei, China
| | - Mengsi Zhou
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Hubei, China.,Department of Medical Parasitology, School of Basic Medical Sciences, Wuhan University, Hubei, China
| | - Huifen Dong
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Hubei, China.,Department of Medical Parasitology, School of Basic Medical Sciences, Wuhan University, Hubei, China
| | - Rui Zhou
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Hubei, China.,Department of Medical Parasitology, School of Basic Medical Sciences, Wuhan University, Hubei, China
| |
Collapse
|
24
|
The role of microRNAs in the pathogenesis, grading and treatment of hepatic fibrosis in schistosomiasis. Parasit Vectors 2019; 12:611. [PMID: 31888743 PMCID: PMC6937654 DOI: 10.1186/s13071-019-3866-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/19/2019] [Indexed: 12/16/2022] Open
Abstract
Schistosomiasis is a prevalent parasitic disease worldwide. The main pathological changes of hepatosplenic schistosomiasis are hepatic granuloma and fibrosis due to worm eggs. Portal hypertension and ascites induced by hepatic fibrosis are usually the main causes of death in patients with chronic hepatosplenic schistosomiasis. Currently, no effective vaccine exists for preventing schistosome infections. For quite a long time, praziquantel (PZQ) was widely used for the treatment of schistosomiasis and has shown benefit in treating liver fibrosis. However, drug resistance and chemical toxicity from PZQ are being increasingly reported in recent years; therefore, new and effective strategies for treating schistosomiasis-induced hepatic fibrosis are urgently needed. MicroRNA (miRNA), a non-coding RNA, has been proved to be associated with the development of many human diseases, including schistosomiasis. In this review, we present a balanced and comprehensive view of the role of miRNAs in the pathogenesis, grading, and treatment of schistosomiasis-associated hepatic fibrosis. The multiple regulatory roles of miRNAs, such as promoting or inhibiting the development of liver pathology in murine schistosomiasis are also discussed in depth. Additionally, miRNAs may serve as candidate biomarkers for diagnosing liver pathology of schistosomiasis and as novel therapeutic targets for treating schistosomiasis-associated hepatic fibrosis.![]()
Collapse
|
25
|
Yu WQ, Ji NF, Gu CJ, Sun ZX, Wang ZX, Chen ZQ, Ma Y, Wu ZZ, Wang YL, Wu CJ, Ding MD, Dai GH, Yao J, Jin RR, Huang M, Zhang MS. Downregulation of miR-4772-3p promotes enhanced regulatory T cell capacity in malignant pleural effusion by elevating Helios levels. Chin Med J (Engl) 2019; 132:2705-2715. [PMID: 31725455 PMCID: PMC6940098 DOI: 10.1097/cm9.0000000000000517] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Malignant pleural effusion (MPE) is a complicated condition of patients with advanced tumors. Further dissecting the microenvironment of infiltrated immune cells and malignant cells are warranted to understand the immune-evasion mechanisms of tumor development and progression. METHODS The possible involvement of microRNAs (miRNAs) in malignant pleural fluid was investigated using small RNA sequencing. Regulatory T cell (Treg) markers (CD4, CD25, forkhead box P3), and Helios (also known as IKAROS Family Zinc Finger 2 [IKZF2]) were detected using flow cytometry. The expression levels of IKZF2 and miR-4772-3p were measured using quantitative real-time reverse transcription polymerase chain reaction. The interaction between miR-4772-3p and Helios was determined using dual-luciferase reporter assays. The effects of miR-4772-3p on Helios expression were evaluated using an in vitro system. Correlation assays between miR-4772-3p and functional molecules of Tregs were performed. RESULTS Compared with non-malignant controls, patients with non-small cell lung cancer had an increased Tregs frequency with Helios expression in the MPE and peripheral blood mononuclear cells. The verified downregulation of miR-4772-3p was inversely related to the Helios Tregs frequency and Helios expression in the MPE. Overexpression of miR-4772-3p could inhibit Helios expression in in vitro experiments. However, ectopic expression of Helios in induced Tregs reversed the effects induced by miR-4772-3p overexpression. Additionally, miR-4772-3p could regulate Helios expression by directly targeting IKZF2 mRNA. CONCLUSION Downregulation of miR-4772-3p, by targeting Helios, contributes to enhanced Tregs activities in the MPE microenvironment.
Collapse
Affiliation(s)
- Wen-Qing Yu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Department of Infectious Diseases, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, Jiangsu 225300, China
| | - Ning-Fei Ji
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Cheng-Jing Gu
- Department of Pharmacy, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, Jiangsu 225300, China
| | - Zhi-Xiao Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Zheng-Xia Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Zhong-Qi Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yuan Ma
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Zhen-Zhen Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yan-Li Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Chao-Jie Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ming-Dong Ding
- Department of Infectious Diseases, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, Jiangsu 225300, China
| | - Gui-Hong Dai
- Department of Pathology, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, Jiangsu 225300, China
| | - Juan Yao
- Department of Oncology, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, Jiangsu 225300, China
| | - Rong-Rong Jin
- Department of Pathology, Taizhou People's Hospital Affiliated to Nantong University, Taizhou, Jiangsu 225300, China
| | - Mao Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ming-Shun Zhang
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
26
|
Nelwan ML. Schistosomiasis: Life Cycle, Diagnosis, and Control. Curr Ther Res Clin Exp 2019; 91:5-9. [PMID: 31372189 PMCID: PMC6658823 DOI: 10.1016/j.curtheres.2019.06.001] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 06/06/2019] [Indexed: 12/20/2022] Open
Abstract
Three main schistosomiasis species can infect humans; S. haematobium, S. japonicum, and S. mansoni. The parasites life cycle includes two kind of reproduction; asexual reproduction in snails and sexual reproduction in mammals. Multiple diagnostic techniques are used. Currently praziquantel is the only drug therapy approved for control of schistosomiasis but other promising candidate drugs (e.g. SpAE, and ruthenium compounds) are being tested. A number of vaccine candidates exist including SmCB1, SjAChE, and SmCB. Genetic manipulations are being investigated.
Background Human schistosomiasis is a parasitic disease caused by blood-worms that infect multiple organs, including the liver, intestine, bladder, and urethra. This disease may be eliminated with Praziquantel, vaccines, and gene therapy. Aims In this review, the author describes the progress in a study of schistosomiasis that focused on the life cycle, diagnosis, and control. Methodology The author searched the PubMed Database at NCBI for articles on schistosomiasis published between 2014 and 2018. All articles were open access and in English. Results The life cycle of this parasites involve two hosts: snails and mammals. Manifestations of schistosomiasis can be acute or chronic. Clinical manifestations of acute schistosomiasis can include fever and headache. Symptoms of chronic infections can include dysuria and hyperplasia. Infection can occur in several sites including the bile ducts, intestine, and bladder. The different sites of infection and symptoms seen are related to which of the species involved. Five species can infect humans. The three most commons are S. haematobium, S. japonicum, and S. mansoni. Detection tools for people with schistosomiasis can include the Kato-Katz and PCR. Praziquantel is at present the only effective treatment of this disease. In the future, vaccination or gene therapy may be used. Conclusion Kato-Katz and PCR are tools for detecting schistosomiasis on humans. Praziquantel, diagnosis, vaccines, and gene therapy are useful methods for eliminating schistosomiasis.
Collapse
Affiliation(s)
- Martin L. Nelwan
- Address correspondence to: Department of Animal Science, Nelwan Institution for Human Resource Development, Jl A Yani No. 24, Palu, Indonesia.
| |
Collapse
|
27
|
Abstract
Schistosomiasis is a serious but neglected tropical infectious disease, afflicting more than 240 million people in 78 countries. Lack of an effective vaccine and obscuring disease mechanism could be the main hurdles to effectively control and eradicate this disease. A better understanding of the host-schistosome interaction is the key to clearing these hurdles. Recently, accumulating evidence shows that alarmin cytokines and microRNAs (miRNAs) are crucial regulators in the host-schistosome interaction. Alarmin cytokines are proven to be potent mechanisms driving type 2 immunity, which is the central disease mechanism of schistosomiasis. MiRNA deregulation is a hallmark of a variety of human diseases, including schistosomiasis. In this review, we summarize the research advances on the role of alarmin cytokines and miRNAs in the host-schistosome interaction.
Collapse
Affiliation(s)
- Xing He
- Department of Tropical diseases, Second Military Medical University, Shanghai, 200433, China
| | - Weiqing Pan
- Department of Tropical diseases, Second Military Medical University, Shanghai, 200433, China
| |
Collapse
|