1
|
Lu KC, Tsai KW, Hu WC. Role of TGFβ-producing regulatory T cells in scleroderma and end-stage organ failure. Heliyon 2024; 10:e35590. [PMID: 39170360 PMCID: PMC11336735 DOI: 10.1016/j.heliyon.2024.e35590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024] Open
Abstract
Regulatory T cells (Tregs) are crucial immune cells that initiate a tolerable immune response. Transforming growth factor-beta (TGFβ) is a key cytokine produced by Tregs and plays a significant role in stimulating tissue fibrosis. Systemic sclerosis, an autoimmune disease characterized by organ fibrosis, is associated with an overrepresentation of regulatory T cells. This review aims to identify Treg-dominant tolerable host immune reactions and discuss their association with scleroderma and end-stage organ failure. End-stage organ failures, including heart failure, liver cirrhosis, uremia, and pulmonary fibrosis, are frequently linked to tissue fibrosis. This suggests that TGFβ-producing Tregs are involved in the pathogenesis of these conditions. However, the exact significance of TGFβ and the mechanisms through which it induces tolerable immune reactions during end-stage organ failure remain unclear. A deeper understanding of these mechanisms could lead to improved preventive and therapeutic strategies for these severe diseases.
Collapse
Affiliation(s)
- Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Kuo-Wang Tsai
- Department of Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, 231, Taiwan
| | - Wan-Chung Hu
- Department of Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, 231, Taiwan
- Department of Clinical Pathology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, 231, Taiwan
- Department of Biotechnology, Ming Chuan University, Taoyuan City, 333, Taiwan
| |
Collapse
|
2
|
Nóbrega DND, Carvalho TL, do Ó KP, Lima RED, Farias ICC, Souza RDS, Pereira JB, Maranhão ACN, Lima VFDS, Melo ALTMD, Silva TCG, Belmont TFDM, Gomes ECDS, Vasconcelos LRS. MicroRNA dysregulation in schistosomiasis-induced hepatic fibrosis: a systematic review. Expert Rev Mol Diagn 2023; 23:257-265. [PMID: 36803616 DOI: 10.1080/14737159.2023.2182190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
BACKGROUND MicroRNAs are involved in gene regulation in several common liver diseases and may play an essential role in activating hepatic stellate cells. The role of these post-transcriptional regulators in schistosomiasis needs to be further studied in populations from endemic areas for a better understanding of the disease, the development of new therapeutic approaches, and the use of biomarkers for the prognosis of schistosomiasis. AREAS COVERED We performed a systematic review to describe the main human microRNAs identified in non-experimental studies associated with aggravation of the disease in people infected with Schistosoma mansoni (S. mansoni) and Schistosoma japonicum (S. japonicum). Structured searches were carried out in PubMed, Medline, Science Direct, Directory of Open Access Journals, Scielo, Medcarib, and Global Index Medicus databases without time and language restrictions. This is a systematic review following the guidelines of the PRISMA platform. EXPERT OPINION The miR-146a-5p, miR-150-5p, let-7a-5p, let-7d-5p, miR-92a- 3p, and miR-532-5p are associated with liver fibrosis in schistosomiasis caused by S. japonicum, revealing that these miRNAs that have been shown to be associated with liver fibrosis are good targets for new studies that evaluate their potential as a biomarker or even treating liver fibrosis in schistosomiasis.
Collapse
Affiliation(s)
| | | | - Kleyton Palmeira do Ó
- Department of Parasitology, Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Recife, Brazil
| | - Raul Emídio de Lima
- Department of Parasitology, Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Recife, Brazil
| | | | | | - Julia Braga Pereira
- Department of Parasitology, Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Recife, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Muir R, Metcalf T, Fourati S, Bartsch Y, Lugemwa JK, Canderan G, Alter G, Muyanja E, Okech B, Namatovu T, Namara I, Namuniina A, Ssetaala A, Mpendo J, Nanvubya A, Kitandwe PK, Bagaya BS, Kiwanuka N, Nassuna J, Biribawa VM, Elliott AM, de Dood CJ, Senyonga W, Balungi P, Kaleebu P, Mayanja Y, Odongo M, Fast P, Price MA, Corstjens PLAM, van Dam GJ, Kamali A, Sekaly RP, Haddad EK. Schistosoma mansoni infection alters the host pre-vaccination environment resulting in blunted Hepatitis B vaccination immune responses. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.24.23284435. [PMID: 36865336 PMCID: PMC9980246 DOI: 10.1101/2023.02.24.23284435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
The impact of endemic infections on protective immunity is critical to inform vaccination strategies. In this study, we assessed the influence of Schistosoma mansoni infection on host responses in a Ugandan fishing cohort given a Hepatitis B (HepB) vaccine. Concentrations of schistosome-specific circulating anodic antigen (CAA) pre-vaccination showed a significant bimodal distribution associated with HepB titers, which were lower in individuals with high CAA. We established that participants with high CAA had significantly lower frequencies of circulating T follicular helper (cTfh) subpopulations pre- and post-vaccination and higher regulatory T cells (Tregs) post-vaccination. Polarization towards higher frequencies of Tregs: cTfh cells can be mediated by changes in the cytokine environment favoring Treg differentiation. In fact, we observed higher levels of CCL17 and soluble IL-2R pre-vaccination (important for Treg recruitment and development), in individuals with high CAA that negatively associated with HepB titers. Additionally, alterations in pre-vaccination monocyte function correlated with HepB titers, and changes in innate-related cytokines/chemokine production were associated with increasing CAA concentration. We report, that by influencing the immune landscape, schistosomiasis has the potential to modulate immune responses to HepB vaccination. These findings highlight multiple Schistosoma -related immune associations that could explain abrogated vaccine responses in communities with endemic infections. Author Summary Schistosomiasis drives host immune responses for optimal pathogen survival, potentially altering host responses to vaccine-related antigen. Chronic schistosomiasis and co-infection with hepatotropic viruses are common in countries where schistosomiasis is endemic. We explored the impact of Schistosoma mansoni ( S. mansoni ) infection on Hepatitis B (HepB) vaccination of individuals from a fishing community in Uganda. We demonstrate that high schistosome-specific antigen (circulating anodic antigen, CAA) concentration pre-vaccination, is associated with lower HepB antibody titers post-vaccination. We show higher pre-vaccination levels of cellular and soluble factors in instances of high CAA that are negatively associated with HepB antibody titers post-vaccination, which coincided with lower frequencies of circulating T follicular helper cell populations (cTfh), proliferating antibody secreting cells (ASCs), and higher frequencies of regulatory T cells (Tregs). We also show that monocyte function is important in HepB vaccine responses, and that high CAA is associated with alterations in the early innate cytokine/chemokine microenvironment. Our findings suggest that in individuals with high CAA and likely high worm burden, schistosomiasis creates and sustains an environment that is polarized against optimal host immune responses to the vaccine, which puts many endemic communities at risk for infection against HepB and other diseases that are preventable by vaccines.
Collapse
|
4
|
Ren J, Zhuo Y, He F, Lv L, Xing M, Guo Y, Zhang Y, Liu J, Li Y, Bai T, Chen Y, Li G, Qin Z, Zhou D. Longitudinal Immune Profiling Highlights CD4+ T Cell Exhaustion Correlated with Liver Fibrosis in Schistosoma japonicum Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:82-95. [PMID: 36445332 DOI: 10.4049/jimmunol.2200301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 11/01/2022] [Indexed: 12/24/2022]
Abstract
Schistosomiasis remains an important public health concern. The eggs deposited in livers invoke a Th2-dominant response, which mediates the fibrotic granulomatous response. However, the mechanisms involved in this immunopathological process are still not perfectly clear. Here, we report a single-cell transcriptional landscape of longitudinally collected BALB/c mouse splenocytes at different time points after Schistosoma japonicum infection. We found that exhausted CD4+ T cells were enriched after infection, changing from coproducing multiple cytokines to predominantly producing the Th2 cytokine IL-4. Regulatory B cells had high expression of Fcrl5, Ptpn22, and Lgals1, potentially regulating exhausted CD4+ T cells via direct PD-1-PD-L2 and PD-1-PD-L1 interactions. Within the myeloid compartment, the number of precursor and immature neutrophils sharply increased after infection. Moreover, dendritic cells, macrophages, and basophils showed inhibitory interactions with exhausted CD4+ T cells. Besides, in mouse livers, we found that exhausted CD4+ T cells were distributed around egg granuloma, promoting collagen expression in primary mouse hepatic stellate cells via IL-4 secretion, resulting in liver fibrosis. Our study provides comprehensive characterization of the composition and cellular states of immune cells with disease progression, which will facilitate better understanding of the mechanism underlying liver fibrotic granulomatous response in schistosomiasis.
Collapse
Affiliation(s)
- Jiling Ren
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yue Zhuo
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Furong He
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lihui Lv
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Man Xing
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yingying Guo
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuchao Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,School of Life Science, Fudan University, Shanghai, China
| | - Jiaojiao Liu
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ying Li
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Tinghui Bai
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yanan Chen
- School of Medicine, Nankai University, Tianjin, China
| | - Guangru Li
- School of Medicine, Nankai University, Tianjin, China
| | - Zhiqiang Qin
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Shanghai, China
| | - Dongming Zhou
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
5
|
Liu Y, Qu M, Jiang H, Schneider R, Qin G, Luo W, Yu H, Zhang B, Wang X, Zhang Y, Zhang H, Zhang Z, Wu Y, Zhang Y, Yin J, Zhang S, Venkatesh B, Roth O, Meyer A, Lin Q. Immunogenetic losses co-occurred with seahorse male pregnancy and mutation in tlx1 accompanied functional asplenia. Nat Commun 2022; 13:7610. [PMID: 36494371 PMCID: PMC9734139 DOI: 10.1038/s41467-022-35338-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
In the highly derived syngnathid fishes (pipefishes, seadragons & seahorses), the evolution of sex-role reversed brooding behavior culminated in the seahorse lineage's male pregnancy, whose males feature a specialized brood pouch into which females deposit eggs during mating. Then, eggs are intimately engulfed by a placenta-like tissue that facilitates gas and nutrient exchange. As fathers immunologically tolerate allogenic embryos, it was suggested that male pregnancy co-evolved with specific immunological adaptations. Indeed, here we show that a specific amino-acid replacement in the tlx1 transcription factor is associated with seahorses' asplenia (loss of spleen, an organ central in the immune system), as confirmed by a CRISPR-Cas9 experiment using zebrafish. Comparative genomics across the syngnathid phylogeny revealed that the complexity of the immune system gene repertoire decreases as parental care intensity increases. The synchronous evolution of immunogenetic alterations and male pregnancy supports the notion that male pregnancy co-evolved with the immunological tolerance of the embryo.
Collapse
Affiliation(s)
- Yali Liu
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.9227.e0000000119573309Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 PR China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, 100101 Beijing, China
| | - Meng Qu
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.9227.e0000000119573309Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 PR China
| | - Han Jiang
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, 100101 Beijing, China
| | - Ralf Schneider
- grid.9764.c0000 0001 2153 9986Marine Evolutionary Ecology, Zoological Institute, Kiel University, 24118 Kiel, Germany
| | - Geng Qin
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.9227.e0000000119573309Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 PR China
| | - Wei Luo
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China
| | - Haiyan Yu
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China
| | - Bo Zhang
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China
| | - Xin Wang
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.9227.e0000000119573309Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 PR China
| | - Yanhong Zhang
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.9227.e0000000119573309Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 PR China
| | - Huixian Zhang
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.9227.e0000000119573309Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 PR China
| | - Zhixin Zhang
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.412785.d0000 0001 0695 6482Graduate School of Marine Science and Technology, Tokyo University of Marine Science and Technology, Minato, Tokyo, Japan
| | - Yongli Wu
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China
| | - Yingyi Zhang
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, 100101 Beijing, China
| | - Jianping Yin
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.9227.e0000000119573309Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 PR China
| | - Si Zhang
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.9227.e0000000119573309Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 PR China
| | - Byrappa Venkatesh
- grid.418812.60000 0004 0620 9243Institute of Molecular and Cell Biology, A*STAR, 138673 Singapore, Singapore
| | - Olivia Roth
- grid.9764.c0000 0001 2153 9986Marine Evolutionary Ecology, Zoological Institute, Kiel University, 24118 Kiel, Germany
| | - Axel Meyer
- grid.9811.10000 0001 0658 7699Department of Biology, University of Konstanz, 78464 Konstanz, Germany
| | - Qiang Lin
- grid.9227.e0000000119573309CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 510301 Guangzhou, China ,grid.9227.e0000000119573309Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 PR China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, 100101 Beijing, China
| |
Collapse
|
6
|
Zhang S, Wan D, Zhu M, Wang G, Zhang X, Huang N, Zhang J, Zhang C, Shang Q, Zhang C, Liu X, Liang F, Zhang C, Kong G, Geng J, Yao L, Lu S, Chen Y, Li Z. CD11b + CD43 hi Ly6C lo splenocyte-derived macrophages exacerbate liver fibrosis via spleen-liver axis. Hepatology 2022; 77:1612-1629. [PMID: 36098707 PMCID: PMC10113005 DOI: 10.1002/hep.32782] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 09/03/2022] [Accepted: 09/09/2022] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS Monocyte-derived macrophages (MoMFs), a dominant population of hepatic macrophages under inflammation, play a crucial role in liver fibrosis progression. The spleen serves as an extra monocyte reservoir in inflammatory conditions; however, the precise mechanisms of involvement of the spleen in the pathogenesis of liver fibrosis remain unclear. APPROACH AND RESULTS By splenectomy and splenocyte transfusion, it was observed that splenic CD11b+ cells accumulated intrahepatically as Ly6Clo MoMFs to exacerbate CCl4 -induced liver fibrosis. The splenocyte migration into the fibrotic liver was further directly visualized by spleen-specific photoconversion with KikGR mice and confirmed by CD45.1+ /CD45.2+ spleen transplantation. Spleen-derived CD11b+ cells purified from fibrotic livers were then annotated by single-cell RNA sequencing, and a subtype of CD11b+ CD43hi Ly6Clo splenic monocytes (sM-1s) was identified, which was markedly expanded in both spleens and livers of mice with liver fibrosis. sM-1s exhibited mature feature with high expressions of F4/80, produced much ROS, and manifested preferential migration into livers. Once recruited, sM-1s underwent sequential transformation to sM-2s (highly expressed Mif, Msr1, Clec4d, and Cstb) and then to spleen-derived macrophages (sMφs) with macrophage features of higher expressions of CX3 CR1, F4/80, MHC class II, and CD64 in the fibrotic hepatic milieu. Furthermore, sM-2s and sMφs were demonstrated capable of activating hepatic stellate cells and thus exacerbating liver fibrosis. CONCLUSIONS CD11b+ CD43hi Ly6Clo splenic monocytes migrate into the liver and shift to macrophages, which account for the exacerbation of liver fibrosis. These findings reveal precise mechanisms of spleen-liver axis in hepatic pathogenesis and shed light on the potential of sM-1 as candidate target for controlling liver diseases.
Collapse
Affiliation(s)
- Shaoying Zhang
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi International Cooperation Base for Inflammation and Immunity, Xi'an, China
| | - Dan Wan
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi International Cooperation Base for Inflammation and Immunity, Xi'an, China
| | - Mengchen Zhu
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi International Cooperation Base for Inflammation and Immunity, Xi'an, China
| | - Guihu Wang
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi International Cooperation Base for Inflammation and Immunity, Xi'an, China
| | - Xurui Zhang
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi International Cooperation Base for Inflammation and Immunity, Xi'an, China
| | - Na Huang
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jian Zhang
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Chongyu Zhang
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi International Cooperation Base for Inflammation and Immunity, Xi'an, China
| | - Qi Shang
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi International Cooperation Base for Inflammation and Immunity, Xi'an, China
| | - Chen Zhang
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi International Cooperation Base for Inflammation and Immunity, Xi'an, China
| | - Xi Liu
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi International Cooperation Base for Inflammation and Immunity, Xi'an, China
| | - Fanfan Liang
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi International Cooperation Base for Inflammation and Immunity, Xi'an, China
| | - Chunyan Zhang
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Guangyao Kong
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jing Geng
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi International Cooperation Base for Inflammation and Immunity, Xi'an, China
| | - Libo Yao
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Shemin Lu
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi International Cooperation Base for Inflammation and Immunity, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, China
| | - Yongyan Chen
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Zongfang Li
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, CHESS-Shaanxi consortium, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.,Shaanxi International Cooperation Base for Inflammation and Immunity, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, China
| |
Collapse
|
7
|
Zhang S, Lu S, Li Z. Extrahepatic factors in hepatic immune regulation. Front Immunol 2022; 13:941721. [PMID: 36052075 PMCID: PMC9427192 DOI: 10.3389/fimmu.2022.941721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
The liver is a site of complex immune activity. The hepatic immune system tolerates harmless immunogenic loads in homeostasis status, shelters liver function, while maintaining vigilance against possible infectious agents or tissue damage and providing immune surveillance at the same time. Activation of the hepatic immunity is initiated by a diverse repertoire of hepatic resident immune cells as well as non-hematopoietic cells, which can sense "danger signals" and trigger robust immune response. Factors that mediate the regulation of hepatic immunity are elicited not only in liver, but also in other organs, given the dual blood supply of the liver via both portal vein blood and arterial blood. Emerging evidence indicates that inter-organ crosstalk between the liver and other organs such as spleen, gut, lung, adipose tissue, and brain is involved in the pathogenesis of liver diseases. In this review, we present the features of hepatic immune regulation, with particular attention to the correlation with factors from extrahepatic organ. We describe the mechanisms by which other organs establish an immune association with the liver and then modulate the hepatic immune response. We discuss their roles and distinct mechanisms in liver homeostasis and pathological conditions from the cellular and molecular perspective, highlighting their potential for liver disease intervention. Moreover, we review the available animal models and methods for revealing the regulatory mechanisms of these extrahepatic factors. With the increasing understanding of the mechanisms by which extrahepatic factors regulate liver immunity, we believe that this will provide promising targets for liver disease therapy.
Collapse
Affiliation(s)
- Shaoying Zhang
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- Shaanxi International Cooperation Base for Inflammation and Immunity, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Shemin Lu
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- Shaanxi International Cooperation Base for Inflammation and Immunity, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, China
| | - Zongfang Li
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- Shaanxi Provincial Clinical Medical Research Center for Liver and Spleen Diseases, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- Shaanxi International Cooperation Base for Inflammation and Immunity, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
8
|
Kadji Fassi JB, Boukeng Jatsa H, Membe Femoe U, Greigert V, Brunet J, Cannet C, Kenfack CM, Gipwe Feussom N, Tienga Nkondo E, Abou-Bacar A, Pfaff AW, Kamgang R, Kamtchouing P, Tchuem Tchuenté LA. Protein undernutrition reduces the efficacy of praziquantel in a murine model of Schistosoma mansoni infection. PLoS Negl Trop Dis 2022; 16:e0010249. [PMID: 35839247 PMCID: PMC9328564 DOI: 10.1371/journal.pntd.0010249] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 07/27/2022] [Accepted: 06/28/2022] [Indexed: 01/03/2023] Open
Abstract
Background Undernutrition and schistosomiasis are public health problems and often occur in low and middle-income countries. Protein undernutrition can alter the host-parasite environment system and aggravate the course of schistosomiasis. This study aimed to assess the impact of a low-protein diet on the efficacy of praziquantel. Methodology/Principal findings Thirty-day-old mice were fed with a low-protein diet, and 40 days later, they were individually infected with fifty Schistosoma mansoni cercariae. A 28-day-treatment with praziquantel at 100 mg/kg for five consecutive days followed by distilled water begins on the 36th day post-infection. Mice were sacrificed on the 64th day post-infection. We determined the parasitological burden, liver and intestine histomorphometry, liver injury, and immunomodulation parameters. Praziquantel treatment of infected mice fed with a standard diet (IN-PZQ) resulted in a significant reduction of worm and egg burdens and a normalization of iron and calcium levels. The therapy also improved schistosomiasis-induced hepatopathy and oxidative stress. The anti-inflammatory and immunomodulatory activities of praziquantel were also significant in these mice. When infected mice receiving the low-protein diet were treated with praziquantel (ILP-PZQ), the body weight loss and hepatomegaly were not alleviated, and the worm and liver egg burdens were significantly higher than those of IN-PZQ mice (P < 0.001). The treatment did not reduce the increased activities of ALT and γ-GGT, the high malondialdehyde concentration, and the liver granuloma volume. The iron and calcium levels were not ameliorated and differed from those of IN-PZQ mice (P < 0.001 and P < 0.05). Moreover, in these mice, praziquantel treatment did not reverse the high level of IL-5 and the low mRNA expression of CCL3/MIP-1α and CXCL-10/IP-10 induced by S. mansoni infection. Conclusion/Significance These results demonstrated that a low-protein diet reduced the schistosomicidal, antioxidant, anti-inflammatory, and immunomodulatory activities of praziquantel. Almost 90% of people requiring schistosomiasis preventive chemotherapy in 2018 lived in sub-Saharan Africa. Besides, 205.3 million children under five years suffer and die of undernutrition in low- and middle-income countries. The physiopathology of schistosomiasis mansoni involves liver damage, oxidative stress, and perturbation of the immune response. These disturbances are intensified by undernutrition. Praziquantel is used to treat schistosomiasis, but its efficacy on the comorbidity of S. mansoni infection and undernutrition has not been investigated. We conducted this study to assess the effectiveness of praziquantel on S. mansoni infection in mice fed with a low-protein diet. We recorded growth retardation, hepatomegaly, and high worm and egg burdens in mice fed with a low-protein diet and treated with PZQ. Moreover, the treatment did not reverse the liver function injury, oxidative stress, high iron level, and low calcium level. The proinflammatory cytokine IL-5 was still high, and the gene expression of some macrophage-associated chemokines was reduced. Therefore, this study demonstrated that in a murine model of a low-protein diet, the efficacy of praziquantel on S. mansoni infection was reduced. It also underlines the importance of targeting protein deficiency and malnutrition in populations living in schistosomiasis endemic areas for efficient disease control.
Collapse
Affiliation(s)
- Joseph Bertin Kadji Fassi
- Laboratory of Animal Physiology, Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
- Centre for Schistosomiasis and Parasitology, Yaoundé, Cameroon
| | - Hermine Boukeng Jatsa
- Laboratory of Animal Physiology, Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
- Centre for Schistosomiasis and Parasitology, Yaoundé, Cameroon
- * E-mail:
| | - Ulrich Membe Femoe
- Laboratory of Animal Physiology, Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
- Centre for Schistosomiasis and Parasitology, Yaoundé, Cameroon
- Institute of Parasitology and Tropical Diseases, Dynamic Host-Pathogen Interactions, University of Strasbourg, Strasbourg, France
| | - Valentin Greigert
- Institute of Parasitology and Tropical Diseases, Dynamic Host-Pathogen Interactions, University of Strasbourg, Strasbourg, France
| | - Julie Brunet
- Institute of Parasitology and Tropical Diseases, Dynamic Host-Pathogen Interactions, University of Strasbourg, Strasbourg, France
| | - Catherine Cannet
- Laboratory of Histomorphometry, Institute of Legal Medicine, University of Strasbourg, Strasbourg, France
| | - Christian Mérimé Kenfack
- Laboratory of Animal Physiology, Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
- Centre for Schistosomiasis and Parasitology, Yaoundé, Cameroon
| | - Nestor Gipwe Feussom
- Laboratory of Animal Physiology, Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
- Centre for Schistosomiasis and Parasitology, Yaoundé, Cameroon
| | - Emilienne Tienga Nkondo
- Laboratory of Animal Physiology, Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
- Centre for Schistosomiasis and Parasitology, Yaoundé, Cameroon
| | - Ahmed Abou-Bacar
- Institute of Parasitology and Tropical Diseases, Dynamic Host-Pathogen Interactions, University of Strasbourg, Strasbourg, France
| | - Alexander Wilhelm Pfaff
- Institute of Parasitology and Tropical Diseases, Dynamic Host-Pathogen Interactions, University of Strasbourg, Strasbourg, France
| | - René Kamgang
- Laboratory of Animal Physiology, Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Pierre Kamtchouing
- Laboratory of Animal Physiology, Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Louis-Albert Tchuem Tchuenté
- Centre for Schistosomiasis and Parasitology, Yaoundé, Cameroon
- Laboratory of Parasitology and Ecology, Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| |
Collapse
|
9
|
Lin D, Song Q, Zhang Y, Liu J, Chen F, Du S, Xiang S, Wang L, Wu X, Sun X. Bacillus subtilis Attenuates Hepatic and Intestinal Injuries and Modulates Gut Microbiota and Gene Expression Profiles in Mice Infected with Schistosoma japonicum. Front Cell Dev Biol 2021; 9:766205. [PMID: 34869360 PMCID: PMC8635066 DOI: 10.3389/fcell.2021.766205] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
Parasitic infection can induce pathological injuries and impact the gut microbiota diversity and composition of the host. Bacillus subtilis is a nonpathogenic and noninvasive probiotic bacterium for humans and other animals, playing an important role in improving the host immune system’s ability to respond to intestinal and liver diseases and modulating gut microbiota. However, whether B. subtilis can impact biological functions in Schistosoma japonicum–infected mice is unclear. This study used oral administration (OA) of B. subtilis to treat mice infected with S. japonicum. We evaluated changes in the gut microbiota of infected mice using 16 S rRNA gene sequencing and differentially expressed gene profiles using transcriptome sequencing after OA B. subtilis. We found that OA B. subtilis significantly attenuated hepatic and intestinal pathological injuries in infected mice. The gut microbiota of mice were significantly altered after S. japonicum infection, while OA B. subtilis remodel the diversity and composition of gut microbiomes of infected mice. We found that the S. japonicum–infected mice with OA B. subtilis had an overabundance of the most prevalent bacterial genera, including Bacteroides, Enterococcus, Lactobacillus, Blautia, Lachnoclostridium, Ruminiclostridium, and Enterobacter. Transcriptomic analysis of intestinal tissues revealed that OA B. subtilis shaped the intestinal microenvironment of the host responding to S. japonicum infection. Differentially expressed genes were classified into KEGG pathways between S. japonicum–infected mice and those without included cell adhesion molecules, intestinal immune network for IgA production, hematopoietic cell lineage, Fc epsilon RI signaling pathway, Th1 and Th2 cell differentiation, Th17 cell differentiation, calcium signaling pathway, Fc gamma R-mediated phagocytosis, chemokine signaling pathway, phospholipase D signaling pathway, NF-kappa B signaling pathway, B cell receptor signaling pathway, pancreatic secretion, and phagosome. In conclusion, our findings showed that OA B. subtilis alleviates pathological injuries and regulates gene expression, implying that B. subtilis supplementation may be a potential therapeutic strategy for schistosomiasis. Our study may highlight the value of probiotics as a beneficial supplementary therapy during human schistosomiasis, but further studies are needed.
Collapse
Affiliation(s)
- Datao Lin
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-vectors Control, Guangzhou, China
| | - Qiuyue Song
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-vectors Control, Guangzhou, China.,Department of Clinical Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Yishu Zhang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-vectors Control, Guangzhou, China
| | - Jiahua Liu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-vectors Control, Guangzhou, China
| | - Fang Chen
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Shuling Du
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-vectors Control, Guangzhou, China
| | - Suoyu Xiang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-vectors Control, Guangzhou, China
| | - Lifu Wang
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-vectors Control, Guangzhou, China
| | - Xiaoying Wu
- Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China.,The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xi Sun
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China.,Provincial Engineering Technology Research Center for Diseases-vectors Control, Guangzhou, China
| |
Collapse
|
10
|
Abstract
Introduction: NAFLD is often under-diagnosed, even though rates of its co-morbidities such as obesity and type2 diabetes mellitus, prominent statuses of inflammation, are significantly high. The spleen-liver axis is gaining much credit in the last years like other well-known organ axes.Areas covered: PubMed/MEDLINE was searched for relevant articles related to concomitant occurrence of NAFLD and spleen. Areas covered in this review include: (1) updated findings of spleen dimensions at ultrasonography, (2) discussion of current data on pathophysiological connections between obesity-related NAFLD and increased volume of the spleen, and (3) analysis of current immune-mediated mechanisms characterizing the so.called chronic low-grade inflammation leading to insulin resistance.Expert opinion: The advances in explaining mechanisms underlying the spleen involvement in immune regulation, coupled with research about the role of spleen in NAFLD, could impact real world outcomes through establishing better tools for a precocious diagnosis. Using both liver and spleen ultrasonography, technique largely dealt with in this review, could expand the possibility to cover an adequate diagnostic path toward NAFLD, reaching a good sensibility and specificity.
Collapse
Affiliation(s)
- Giovanni Tarantino
- Department of Clinical Medicine and Surgery, Federico II University Medical School of Naples, Naples, Italy
| | - Vincenzo Citro
- Department of General Medicine, "Umberto I" Hospital, Nocera Inferiore (SA), Nocera Inferiore, Italy
| | - Clara Balsano
- Department of Clinical Medicine, Life, Health & Environmental Sciences-MESVA, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
11
|
Mu Y, McManus DP, Hou N, Cai P. Schistosome Infection and Schistosome-Derived Products as Modulators for the Prevention and Alleviation of Immunological Disorders. Front Immunol 2021; 12:619776. [PMID: 33692793 PMCID: PMC7937812 DOI: 10.3389/fimmu.2021.619776] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/08/2021] [Indexed: 12/22/2022] Open
Abstract
Parasitic helminths, comprising the flatworms (tapeworms and flukes) and nematodes (roundworms), have plagued humans persistently over a considerable period of time. It is now known that the degree of exposure to these and other pathogens inversely correlates with the incidence of both T helper 1 (Th1)-mediated autoimmunity and Th2-mediated allergy. Accordingly, there has been recent increased interest in utilizing active helminth worm infections and helminth-derived products for the treatment of human autoimmune and inflammatory diseases and to alleviate disease severity. Indeed, there is an accumulating list of novel helminth derived molecules, including proteins, peptides, and microRNAs, that have been shown to exhibit therapeutic potential in a variety of disease models. Here we consider the blood-dwelling schistosome flukes, which have evolved subtle immune regulatory mechanisms that promote parasite survival but at the same time minimize host tissue immunopathology. We review and discuss the recent advances in using schistosome infection and schistosome-derived products as therapeutics to treat or mitigate human immune-related disorders, including allergic asthma, arthritis, colitis, diabetes, sepsis, cystitis, and cancer.
Collapse
Affiliation(s)
- Yi Mu
- Molecular Parasitology Laboratory, Infectious Diseases Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Donald P McManus
- Molecular Parasitology Laboratory, Infectious Diseases Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Nan Hou
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Pengfei Cai
- Molecular Parasitology Laboratory, Infectious Diseases Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
12
|
Santana JB, de Almeida TVVS, Lopes DM, Page B, Oliveira SC, Souza I, Ribeiro LEVS, Gutiérrez NAG, Carvalho EM, Cardoso LS. Phenotypic Characterization of CD4 + T Lymphocytes in Periportal Fibrosis Secondary to Schistosomiasis. Front Immunol 2021; 12:605235. [PMID: 33692784 PMCID: PMC7937650 DOI: 10.3389/fimmu.2021.605235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/04/2021] [Indexed: 01/13/2023] Open
Abstract
Schistosomiasis is a parasitic disease that affects about 166 million people around the world. It is estimated that 5%-10% of individuals with schistosomiasis develop severe forms of the disease, which are characterized by pulmonary hypertension, ascites, periportal fibrosis, and other significant complications. The chronic phase of the disease is associated with a Th2 type immune response, but evidence also suggests there are roles for Th1 and Th17 in the development of severe disease. The aim of this study was to evaluate the CD4+ T lymphocyte profile of patients with different degrees of periportal fibrosis secondary to schistosomiasis. These individuals had been treated for schistosomiasis, but since they live in a S. mansoni endemic area, they are at risk of reinfection. They were evaluated in relation to the degree of periportal fibrosis and classified into three groups: without fibrosis or with incipient fibrosis (WF/IFNE), n=12, possible periportal fibrosis/periportal fibrosis, n=13, and advanced periportal fibrosis/advanced periportal fibrosis with portal hypertension, n=4. We observed in the group without fibrosis a balance between the low expression of Th2 cytokines and high expression of T reg cells. As has already been described in the literature, we found an increase of the Th2 cytokines IL-4, IL-5, and IL-13 in the group with periportal fibrosis. In addition, this group showed higher expression of IL-17 and IL-10 but lower IL-10/IL-13 ratio than patients in the WF/IFNE group. Cells from individuals who present any level of fibrosis expressed more TGF-β compared to the WF/IFNE group and a positive correlation with left lobe enlargement and portal vein wall thickness. There was a negative correlation between IL-17 and the thickness of the portal vein wall, but more studies are necessary in order to explore the possible protective role of this cytokine. Despite the fibrosis group having presented a higher expression of pro-fibrotic molecules compared to WF/IFNE patients, it seems there is a regulation through IL-10 and T reg cells that is able to maintain the low morbidity of this group.
Collapse
Affiliation(s)
- Jordana Batista Santana
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | | | - Diego Mota Lopes
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Brady Page
- Massachusetts General Hospital, Boston, MA, United States
| | - Sergio Costa Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT–DT/CNPq), Salvador, Brazil
| | | | | | | | - Edgar M. Carvalho
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT–DT/CNPq), Salvador, Brazil
- Laboratório de Pesquisas Clínicas, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
| | - Luciana Santos Cardoso
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT–DT/CNPq), Salvador, Brazil
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| |
Collapse
|
13
|
Schistosoma japonicum Infection in Treg-Specific USP21 Knockout Mice. J Immunol Res 2021; 2021:6613162. [PMID: 33628844 PMCID: PMC7886505 DOI: 10.1155/2021/6613162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/11/2021] [Accepted: 01/21/2021] [Indexed: 12/18/2022] Open
Abstract
The E3 deubiquitinating enzyme ubiquitin-specific proteolytic enzyme 21 (USP21) plays vital roles in physiological activities and is required for Treg-cell-mediated immune tolerance. Using a murine model infected with Schistosoma japonicum, we observed that there were more cercariae developed into adults and more eggs deposited in the livers of the USP21fl/flFOXP3Cre (KO) mice. However, immunohistochemistry showed that the degree of egg granuloma formation and liver fibrosis was reduced. In USP21fl/flFOXP3Cre mice, levels of IFN-gamma, IL-4, anti-soluble egg antigen (SEA) IgG and anti-soluble worm antigen preparation (SWAP) IgG increased in blood, as determined using ELISAs and multiplex fluorescent microsphere immunoassays, while the levels of IL-10, lL-17A, IL-23, IL-9, and anti-SEA IgM decreased. In addition, the levels of the USP21 protein and mRNA in the liver and spleen of KO mice decreased. We further observed increased Th1 responses amplified by Tregs (regulatory T cells) and compromised Th17 responses, which alleviated the liver immunopathology. We speculated that these changes were related to polarization of Th1-like Tregs. Our results revealed the roles of USP21 in Treg-cell-mediated regulation of immune interactions between Schistosoma and its host. USP21 may have potential for regulating hepatic fibrosis in patients with schistosomiasis.
Collapse
|
14
|
Llanwarne F, Helmby H. Granuloma formation and tissue pathology in Schistosoma japonicum versus Schistosoma mansoni infections. Parasite Immunol 2021; 43:e12778. [PMID: 32692855 PMCID: PMC11478942 DOI: 10.1111/pim.12778] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/10/2020] [Accepted: 07/16/2020] [Indexed: 12/13/2022]
Abstract
Schistosomiasis is the most important helminth disease in the world from a public health perspective. S mansoni and S japonicum account for the majority of global intestinal schistosomiasis cases, and the pathogenesis is widely assumed to be fundamentally similar. However, the majority of research on schistosomiasis has been carried out on S mansoni and comparisons between the two species are rarely made. Here, we will discuss aspects of both older and recent literature where such comparisons have been made, with a particular focus on the pathological agent, the host granulomatous response to the egg. Major differences between the two species are apparent in features such as egg production patterns and cellular infiltration; however, it is also clear that even subtle differences in the cascade of various cytokines and chemokines contribute to the different levels of pathology observed between these two main species of intestinal schistosomiasis. A better understanding of such differences at species level will be vital when it comes to the development of new treatment strategies and vaccines.
Collapse
Affiliation(s)
- Felix Llanwarne
- Department of Infection BiologyFaculty of Infectious and Tropical DiseaseLondon School of Hygiene and Tropical MedicineLondonUK
| | - Helena Helmby
- Department of Infection BiologyFaculty of Infectious and Tropical DiseaseLondon School of Hygiene and Tropical MedicineLondonUK
| |
Collapse
|
15
|
Wang F, So KF, Xiao J, Wang H. Organ-organ communication: The liver's perspective. Am J Cancer Res 2021; 11:3317-3330. [PMID: 33537089 PMCID: PMC7847667 DOI: 10.7150/thno.55795] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 12/28/2020] [Indexed: 12/11/2022] Open
Abstract
Communication between organs participates in most physiological and pathological events. Owing to the importance of precise coordination among the liver and virtually all organs in the body for the maintenance of homeostasis, many hepatic disorders originate from impaired organ-organ communication, resulting in concomitant pathological phenotypes of distant organs. Hepatokines are proteins that are predominantly secreted from the liver, and many hepatokines and several signaling proteins have been linked to diseases of other organs, such as the heart, muscle, bone, and eyes. Although liver-centered interorgan communication has been proposed in both basic and clinical studies, to date, the regulatory mechanisms of hepatokine production, secretion, and reciprocation with signaling factors from other organs are obscure. Whether other hormones and cytokines are involved in such communication also warrants investigation. Herein, we summarize the current knowledge of organ-organ communication phenotypes in a variety of diseases and the possible involvement of hepatokines and/or other important signaling factors. This provides novel insight into the underlying roles and mechanisms of liver-originated signal transduction and, more importantly, the understanding of disease in an integrative view.
Collapse
|
16
|
Loss of natural resistance to schistosome in T cell deficient rat. PLoS Negl Trop Dis 2020; 14:e0008909. [PMID: 33347431 PMCID: PMC7785244 DOI: 10.1371/journal.pntd.0008909] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 01/05/2021] [Accepted: 10/21/2020] [Indexed: 01/08/2023] Open
Abstract
Schistosomiasis is among the major neglected tropical diseases and effective prevention by boosting the immune system is still not available. T cells are key cellular components governing adaptive immune response to various infections. While common laboratory mice, such as C57BL/6, are highly susceptible to schistosomiasis, the SD rats are extremely resistant. However, whether adaptive immunity is necessary for such natural resistance to schistosomiasis in rats remains to be determined. Therefore, it is necessary to establish genetic model deficient in T cells and adaptive immunity on the resistant SD background, and to characterize liver pathology during schistosomiasis. In this study we compared experimental schistosomiasis in highly susceptible C57BL/6 (B6) mice and in resistant SD rats, using cercariae of Schistosoma japonicum. We observed a marked T cell expansion in the spleen of infected B6 mice, but not resistant SD rats. Interestingly, CD3e−/− B6 mice in which T cells are completely absent, the infectious burden of adult worms was significantly higher than that in WT mice, suggesting an anti-parasitic role for T cells in B6 mice during schistosome infection. In further experiments, we established Lck deficient SD rats by using CRISPR/Cas9 in which T cell development was completely abolished. Strikingly, we found that such Lck deficiency in SD rats severely impaired their natural resistance to schistosome infection, and fostered parasite growth. Together with an additional genetic model deficient in T cells, the CD3e−/− SD rats, we confirmed the absence of T cell resulted in loss of natural resistance to schistosome infection, but also mitigated liver immunopathology. Our further experiments showed that regulatory T cell differentiation in infected SD rats was significantly decreased during schistosomiasis, in contrast to significant increase of regulatory T cells in infected B6 mice. These data suggest that T cell mediated immune tolerance facilitates persistent infection in mice but not in SD rats. The demonstration of an important role for T cells in natural resistance of SD rats to schistosomiasis provides experimental evidences supporting the rationale to boost T cell responses in humans to prevent and treat schistosomiasis. Schistosomiasis is among the major neglected tropical diseases and affects mainly the developing countries. Although the role of the immune system in driving immunopathology in schistosomiasis has been extensively studied, how adaptive immunity contributes to disease resistance during schistosome infection is still not completely understood. Most livestock species as well as humans are susceptible to schistosomiasis, while some mammals are extremely resistant. The common laboratory C57BL/6 mice are highly susceptible to schistosomiasis; however, the SD rats are extremely resistant. In this study, we first used T cell deficient CD3e−/− C57BL/6 mice and experimental Schistosoma japonicum infection and further established novel T cell deficient models in SD rats to assess anti-parasite roles of T cells. Strikingly, we found that the natural resistance of SD rat to schistosomiasis was abolished in the absence of T cells, despite the fact that the liver pathology was mitigated following infection. Therefore, our study presented experimental support for the rationale to boost T cell function for clearance of schistosome parasites.
Collapse
|
17
|
Zhang B, Wu X, Song Q, Ning A, Liang J, Song L, Liu J, Zhang Y, Yuan D, Sun X, Wu Z. Gut Microbiota Modulates Intestinal Pathological Injury in Schistosoma japonicum-Infected Mice. Front Med (Lausanne) 2020; 7:588928. [PMID: 33313045 PMCID: PMC7703745 DOI: 10.3389/fmed.2020.588928] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Trapping of Schistosoma japonicum (S. japonicum) eggs in host tissue, mainly in the intestine and liver, causes severe gastrointestinal and hepatic granulomatous immune responses and irreversible fibrosis. Although the gut microbiota plays a central role in regulating pathological responses in several diseases, the effect of the gut microbiota on the pathologenesis progression of schistosomiasis remains largely unknown. In this study, we aimed to investigate the regulatory function of the gut microbiota in schistosomiasis japonica. We found that the depletion of the gut microbiota significantly ameliorated egg granulomas formation and fibrosis in the intestine of infected mice. This role of the gut microbiota in intestinal granuloma formation and fibrosis was reinforced when normal and infected mice were housed together in one cage. Notably, changes in the gut microbiota induced by S. japonicum infection were partly reversible with microbiota transfer in the cohousing experiment. Transfer of the gut microbiota from normal to infected mice attenuated the intestinal pathological responses. Depletion of the gut microbiota by antibiotics, or transfer of the gut microbiota from normal to infected mice decreased the levels of IL-4, IL-5, and IL-13 and promoted the production of cytokines and mRNA levels of IL-10 and TGF-β in infected mice. Our findings indicated a regulatory effect of the gut microbiota on intestinal pathological injury associated with schistosomiasis japonica in mice, and thus suggested a potential strategy for schistosomiasis treatment.
Collapse
Affiliation(s)
- Beibei Zhang
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Sun Yat-sen University, Guangzhou, China.,Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, China
| | - Xiaoying Wu
- Department of Gastroenterology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qiuyue Song
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Sun Yat-sen University, Guangzhou, China
| | - An Ning
- Jiangxi Provincial Institute of Parasitic Diseases, Nanchang, China
| | - Jinyi Liang
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Sun Yat-sen University, Guangzhou, China
| | - Langui Song
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Sun Yat-sen University, Guangzhou, China
| | - Jiahua Liu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Sun Yat-sen University, Guangzhou, China
| | - Yishu Zhang
- College of Basic Medical Sciences, Guilin Medical University, Guilin, China
| | - Dongjuan Yuan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xi Sun
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Sun Yat-sen University, Guangzhou, China
| | - Zhongdao Wu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
18
|
Mechanism by which the combination of SjCL3 and SjGAPDH protects against Schistosoma japonicum infection. Parasitol Res 2020; 120:173-185. [PMID: 33079271 DOI: 10.1007/s00436-020-06916-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 10/04/2020] [Indexed: 10/23/2022]
Abstract
A vaccine is an important method to control schistosomiasis. Molecules related to lung-stage schistosomulum are considered potential vaccine candidates. We previously showed that glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and cathepsin L3 (CL3) displayed differential expression in the lung-stage schistosomula of Schistosoma japonicum cocultured with host cells. In the present study, we prepared the two proteins and detected the protective effects of SjGAPDH by immunizing mice with this protein alone and in combination with SjCL3 with or without Freund's adjuvant. Then, we investigated the possible mechanisms underlying S. japonicum infection. The results showed that vaccination of adjuvanted SjGAPDH decreased the worm burden (37.8%) and egg load (38.1%), and the combination of adjuvanted SjGAPDH and SjCL3 further decreased the worm burden (65.6%) and egg load (70.9%) during Schistosoma japonicum infection. However, the immunization of a combination of adjuvant-free SjGAPDH and SjCL3 displayed a lower protective effect (< 15%) than those of the adjuvanted SjCL3, the adjuvanted SjGAPDH, and a combination of adjuvanted SjGAPDH and SjCL3. Flow cytometric results showed that the frequency of regulatory T cells (Tregs) was lower (P < 0.05) in the group with adjuvanted SjGAPDH and SjCL3 (2.61%) than the remaining groups. The enzyme-linked immunosorbent assay (ELISA) results indicated that except for the uninfected and infected control groups, the remaining groups displayed a Th1-type shift in immune responses. These results showed the immunization of SjGAPDH resulted in partial protection (approximately 38%); inoculation with a combination of SjCL3 and SjGAPDH in Freund's adjuvant resulted in a high immunoprotective effect (> 65%) against Schistosoma japonicum infection in mice, which was possibly caused by the reduced percentage of Tregs and a Th1-type shift in immune responses; and SjCL3 has no adjuvant-like effect, dissimilar to SmCL3.
Collapse
|
19
|
Song LJ, Yin XR, Mu SS, Li JH, Gao H, Zhang Y, Dong PP, Mei CJ, Hua ZC. The Differential and Dynamic Progression of Hepatic Inflammation and Immune Responses During Liver Fibrosis Induced by Schistosoma japonicum or Carbon Tetrachloride in Mice. Front Immunol 2020; 11:570524. [PMID: 33117360 PMCID: PMC7575768 DOI: 10.3389/fimmu.2020.570524] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022] Open
Abstract
Liver fibrosis can result from various causes and could progress to cirrhosis and cancer; however, there are no effective treatments due to that its molecular mechanism is unclear. liver fibrosis model made by Schistosoma japonicum (S. japonicum) infection or Carbon tetrachloride (CCl4) intraperitoneal injection is a conventional model used in liver fibrosis-related studies for mechanism or pharmaceutical research purposes. But the differences in the pathological progression, immune responses and the underlying mechanism between the two liver fibrosis model have not been carefully compared and characterized, which hinders us from correctly understanding and making better use of the two models. In the present study, the pathological changes to the liver, and the cytokines, inflammatory factors, macrophages, and lymphocytes subsets involved were analyzed in the liver fibrosis model of S. japonicum infection or CCl4 intraperitoneal injection. Additionally, the pathological progression, immune responses and the underlying injury mechanism in these two models were compared and characterized. The results showed that the changing trend of interleukin-13 (IL-13), transforming growth factor beta (TGF-β), inflammatory factors, and M1, M2 macrophages, were consistent with the development trend of fibrosis regardless of whether liver fibrosis was caused by S. japonicum or CCl4. For lymphocyte subsets, the proportions of CD3+ T cells and CD4+ T cells decreased gradually, while proportion of CD8+ T cells peaked at 6 weeks in mice infected with S. japonicum and at 12 weeks in mice injected with CCl4. With prolonged S. japonicum infection time, Th1 (CD4+IFN-γ+) immunity converted to Th2 (CD4+IL-4+)/Th17 (CD4+IL-17+) with weaker regulatory T cell (Treg) (CD4+CD25+FOXP3+) immunity. However, in liver fibrosis caused by CCl4, Th1 cells occupied the dominant position, while proportions of Th2, Th17, and Treg cells decreased gradually. In conclusion, liver fibrosis was a complex pathological process that was regulated by a series of cytokines and immune cells. The pathological progressions and immune responses to S. japonicum or CCl4 induced liver fibrosis were different, possibly because of their different injury mechanisms. The appropriate animal model should be selected according to the needs of different experiments and the pathogenic factors of liver fibrosis in the study.
Collapse
Affiliation(s)
- Li-Jun Song
- School of Life Sciences and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.,National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China.,Public Health Research Center, Jiangnan University, Wuxi, China
| | - Xu-Ren Yin
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Sha-Sha Mu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Jia-Huang Li
- School of Life Sciences and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.,School of Biopharmacy, China Pharmaceutical University, Nanjing, China.,Jiangsu TargetPharma Laboratories Inc., Changzhou High-Tech Research Institute of Nanjing University, Changzhou, China
| | - Hong Gao
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Ying Zhang
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, 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 Institute of Parasitic Diseases, Wuxi, 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 Institute of Parasitic Diseases, Wuxi, China
| | - Zi-Chun Hua
- School of Life Sciences and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.,School of Biopharmacy, China Pharmaceutical University, Nanjing, China.,Jiangsu TargetPharma Laboratories Inc., Changzhou High-Tech Research Institute of Nanjing University, Changzhou, China
| |
Collapse
|
20
|
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: 1.5] [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
|
21
|
Ellwanger JH, Kaminski VDL, Rodrigues AG, Kulmann-Leal B, Chies JAB. CCR5 and CCR5Δ32 in bacterial and parasitic infections: Thinking chemokine receptors outside the HIV box. Int J Immunogenet 2020; 47:261-285. [PMID: 32212259 DOI: 10.1111/iji.12485] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/02/2020] [Accepted: 03/06/2020] [Indexed: 12/14/2022]
Abstract
The CCR5 molecule was reported in 1996 as the main HIV-1 co-receptor. In that same year, the CCR5Δ32 genetic variant was described as a strong protective factor against HIV-1 infection. These findings led to extensive research regarding the CCR5, culminating in critical scientific advances, such as the development of CCR5 inhibitors for the treatment of HIV infection. Recently, the research landscape surrounding CCR5 has begun to change. Different research groups have realized that, since CCR5 has such important effects in the chemokine system, it could also affect other different physiological systems. Therefore, the effect of reduced CCR5 expression due to the presence of the CCR5Δ32 variant began to be further studied. Several studies have investigated the role of CCR5 and the impacts of CCR5Δ32 on autoimmune and inflammatory diseases, various types of cancer, and viral diseases. However, the role of CCR5 in diseases caused by bacteria and parasites is still poorly understood. Therefore, the aim of this article is to review the role of CCR5 and the effects of CCR5Δ32 on bacterial (brucellosis, osteomyelitis, pneumonia, tuberculosis and infection by Chlamydia trachomatis) and parasitic infections (toxoplasmosis, leishmaniasis, Chagas disease and schistosomiasis). Basic information about each of these infections was also addressed. The neglected role of CCR5 in fungal disease and emerging studies regarding the action of CCR5 on regulatory T cells are briefly covered in this review. Considering the "renaissance of CCR5 research," this article is useful for updating researchers who develop studies involving CCR5 and CCR5Δ32 in different infectious diseases.
Collapse
Affiliation(s)
- Joel Henrique Ellwanger
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Valéria de Lima Kaminski
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Andressa Gonçalves Rodrigues
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - Bruna Kulmann-Leal
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| | - José Artur Bogo Chies
- Laboratory of Immunobiology and Immunogenetics, Department of Genetics, Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, Brazil
| |
Collapse
|
22
|
Zhan T, Ma H, Jiang S, Zhong Z, Wang X, Li C, Yu D, Liu L, Xu J, Xia C. Interleukin-9 blockage reduces early hepatic granuloma formation and fibrosis during Schistosoma japonicum infection in mice. Immunology 2019; 158:296-303. [PMID: 31436861 DOI: 10.1111/imm.13111] [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: 06/22/2019] [Revised: 08/19/2019] [Accepted: 08/19/2019] [Indexed: 02/07/2023] Open
Abstract
Hepatic fibrosis induced by schistosomes is regulated by a complex network of cytokines. T helper type 9 (Th9) cells are a new type of effector T helper cells, which mainly secrete the specific cytokine interleukin-9 (IL-9). Interleukin-9 has been shown to contribute to liver fibrosis in patients with chronic hepatitis B and in a mouse model due to carbon tetrachloride. However, the role of IL-9 in schistosomiasis fibrosis remains unknown. In this study, we investigated the roles of IL-9 in schistosomiasis through in vivo and in vitro studies. The in vivo studies found that neutralization of IL-9 reduced liver granulomatous inflammation and collagen deposition around parasite eggs. The in vitro studies found that the treatment of primary hepatic stellate cells with IL-9 induced a significant increase of collagen and α-smooth-muscle actin. Moreover, we also described the dynamics and relevance of IL-9 and IL-4 in mice infected with Schistosoma japonicum. We found that IL-9 might appear more quickly and at higher levels than IL-4. Hence, our findings indicated that IL-9 might play a role in regulating hepatic fibrosis in early-stage schistosomiasis and become a promising approach for regulating hepatic fibrosis caused by S. japonicum.
Collapse
Affiliation(s)
- Tingzheng Zhan
- Department of Parasitology, Medical College of Soochow University, Suzhou, China.,Department of Parasitology, Guangxi Medical University, Nanning, China
| | - Huihui Ma
- Department of Parasitology, Medical College of Soochow University, Suzhou, China
| | - Suqin Jiang
- Department of Parasitology, Medical College of Soochow University, Suzhou, China
| | - Zirong Zhong
- Department of Parasitology, Medical College of Soochow University, Suzhou, China
| | - Xiaoli Wang
- Department of Parasitology, Medical College of Soochow University, Suzhou, China.,Department of Parasitology, Bengbu Medical College, Bengbu, China
| | - Chunxiang Li
- Department of Parasitology, Medical College of Soochow University, Suzhou, China
| | - Dan Yu
- Department of Parasitology, Medical College of Soochow University, Suzhou, China
| | - Lei Liu
- Department of Parasitology, Medical College of Soochow University, Suzhou, China
| | - Jing Xu
- Department of Parasitology, Medical College of Soochow University, Suzhou, China
| | - Chaoming Xia
- Department of Parasitology, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
23
|
Hirakawa Y, Ogata T, Sasada T, Yamashita T, Itoh K, Tanaka H, Okuda K. Immunological consequences following splenectomy in patients with liver cirrhosis. Exp Ther Med 2019; 18:848-856. [PMID: 31281459 DOI: 10.3892/etm.2019.7640] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 05/16/2019] [Indexed: 12/16/2022] Open
Abstract
The immune status in patients with liver cirrhosis is generally impaired due to concomitant hypersplenism. As the spleen is the largest lymphoid organ, deleterious events resulting from splenectomy are of concern in these patients. However, the immunological consequences after splenectomy have not yet been fully elucidated. In the present study, the immune status after splenectomy was comprehensively examined. Splenectomy was performed in 11 patients with liver cirrhosis and hypersplenism, and the immune status in peripheral blood was examined and compared before and at 1, 3 and 6 months after splenectomy. Splenectomy significantly lowered the neutrophil-to-lymphocyte ratio, due to a surge in lymphocytes in the peripheral circulation at 3 and 6 months after splenectomy. The frequency of cluster of differentiation (CD)4+ T cells decreased after splenectomy, whereas the frequency of CD8+ T cells increased. Notably, the frequencies of the naïve and central memory subsets of CD4+ and CD8+ T cells decreased, whereas those of the effector memory subset trended upward. In addition, the frequencies of other immune cells such as γδ T cells, natural killer T cells and natural killer cells transiently increased, while inhibitory cells such as regulatory T cells and myeloid-derived suppressor cells significantly decreased. T-cell responses to viral- and tumor-associated antigens increased after splenectomy in five of eight and two of five patients, respectively. To the best of our knowledge, this is the first study to precisely examine the drastic changes of immunological phenotypes in peripheral blood after splenectomy in patients with cirrhosis. Our findings suggested that splenectomy in patients with cirrhosis may ameliorate the impaired immune status, possibly by reducing suppressive cells and enhancing the effector cell population and function, which could, at least in part, explain the mechanisms responsible for the clinical benefits of splenectomy.
Collapse
Affiliation(s)
- Yusuke Hirakawa
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Toshiro Ogata
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan.,Department of Surgery, St. Mary's Hospital, Kurume, Fukuoka 830-8543, Japan
| | - Tetsuro Sasada
- Cancer Vaccine Center, Kurume University, Kurume, Fukuoka 839-0863, Japan.,Cancer Vaccine Center, Kanagawa Cancer Center, Yokohama, Kanagawa 241-8515, Japan
| | - Takuto Yamashita
- Biostatistics Center, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University, Kurume, Fukuoka 839-0863, Japan
| | - Hiroyuki Tanaka
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Koji Okuda
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| |
Collapse
|
24
|
PPAR- γ Agonist Alleviates Liver and Spleen Pathology via Inducing Treg Cells during Schistosoma japonicum Infection. J Immunol Res 2018; 2018:6398078. [PMID: 30116754 PMCID: PMC6079474 DOI: 10.1155/2018/6398078] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/12/2018] [Accepted: 06/25/2018] [Indexed: 12/31/2022] Open
Abstract
Background Peroxisome proliferator-activated receptor- (PPAR-) γ plays critical roles in human metabolic disorders and has recently been implicated as a regulator of cellular proliferation and inflammatory responses. Regulatory T cells (Tregs), which express high levels of PPAR-γ protein, have the ability to maintain immune tolerance to self-antigens and regulate immune response to Schistosoma infection. However, mechanisms involved in the resolution of these responses are elusive. Methods Liver and spleen tissue samples in Schistosoma japonicum-infected mice after administration of pioglitazone (a PPAR-γ agonist) were collected. The hepatic and splenic pathologies were detected by H&E and Masson staining. The percentages of Th1/2 and Treg cells in the liver and spleen of each mouse were determined using flow cytometry. Levels of gene expression of PPAR-γ and Foxp3 in tissues or cells were determined using real-time PCR (RT-PCR). Macrophages were treated with pioglitazone in vitro or cocultured with normal purified CD4+ T cells for detecting Treg cells by flow cytometry. The interactions of PPAR-γ with Foxp3 in CD4+ T cells were detected by coimmunoprecipitation. Results Administration of pioglitazone resulted in the prevention of the development of hepatic and splenic pathologies. Activation of PPAR-γ by pioglitazone resulted in increased percentages of CD4+CD25+Foxp3+ Treg cells and decreased percentages of CD3+CD4+IFN-γ+ and CD3+CD4+IL-4+ cells in the liver and spleen of Schistosoma japonicum-infected mice. In addition, the PPAR-γ agonist can induce Treg cells in vitro directly or by modulating the macrophage's function indirectly. Furthermore, through interaction with Foxp3 in CD4+ T cells, the PPAR-γ agonist can promote the expression of Foxp3; however, the inhibitor of PPAR-γ weakened the expression of Foxp3 by modifying the coexpression of Foxp3 and PPAR-γ. Conclusions Our study reveals a previously unrecognized role for PPAR-γ/Foxp3 signaling in regulating the immunopathology that occurs during Schistosoma infection through induction of Treg cells.
Collapse
|
25
|
Ondigo BN, Ndombi EM, Nicholson SC, Oguso JK, Carter JM, Kittur N, Secor WE, Karanja DMS, Colley DG. Functional Studies of T Regulatory Lymphocytes in Human Schistosomiasis in Western Kenya. Am J Trop Med Hyg 2018; 98:1770-1781. [PMID: 29692308 PMCID: PMC6086154 DOI: 10.4269/ajtmh.17-0966] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Immunoregulation is considered a common feature of Schistosoma mansoni infections, and elevated levels of T regulatory (Treg) lymphocytes have been reported during chronic human schistosomiasis. We now report that the removal of Treg (CD4+/CD25hi/CD127low lymphocytes) from peripheral blood mononuclear cells (PBMCs) of S. mansoni–infected individuals leads to increased levels of phytohemagglutinin (PHA)-stimulated interferon gamma (IFNγ) production and decreased interleukin-10 (IL-10) responses. Exposure to schistosome antigens did not result in measurable IFNγ by either PBMC or Treg-depleted populations. Interleukin-10 responses to soluble egg antigens (SEA) by PBMC were unchanged by Treg depletion, but the depletion of Treg greatly decreased IL-10 production to soluble worm antigenic preparation (SWAP). Proliferative responses to PHA increased upon Treg removal, but responses to SEA or SWAP did not, unless only initially low responders were evaluated. Addition of anti-IL-10 increased PBMC proliferative responses to either SEA or SWAP, but did not alter responses by Treg-depleted cells. Blockade by anti-transforming growth factor-beta (TGF-β) increased SEA but not SWAP proliferative responses by PBMC, whereas anti-TGF-β increased both SEA- and SWAP-stimulated responses by Treg-depleted cultures. Addition of both anti-IL-10 and anti-TGF-β to PBMC or Treg-depleted populations increased proliferation of both populations to either SEA or SWAP. These studies demonstrate that Treg appear to produce much of the antigen-stimulated IL-10, but other cell types or subsets of Treg may produce much of the TGF-β. The elevated levels of Treg seen in chronic schistosomiasis appear functional and involve IL-10 and TGF-β in antigen-specific immunoregulation perhaps leading to regulation of immunopathology and/or possibly decreased immunoprotective responses.
Collapse
Affiliation(s)
- Bartholomew N Ondigo
- Department of Biochemistry and Molecular Biology, Egerton University, Nakuru, Kenya.,Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Eric M Ndombi
- Department of Pathology, Kenyatta University, Nairobi, Kenya.,Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Sarah C Nicholson
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| | - John K Oguso
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Jennifer M Carter
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| | - Nupur Kittur
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| | - W Evan Secor
- Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Diana M S Karanja
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Daniel G Colley
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia.,Department of Microbiology, University of Georgia, Athens, Georgia
| |
Collapse
|
26
|
Mass drug administration and the global control of schistosomiasis: successes, limitations and clinical outcomes. Curr Opin Infect Dis 2018; 29:595-608. [PMID: 27584590 DOI: 10.1097/qco.0000000000000312] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE OF REVIEW Preventive chemotherapy is advocated for the global control and elimination of schistosomiasis. Despite the well known short-term benefits of treating patients for schistosomiasis, the impact of mass drug administration (MDA) campaigns to control the disease in the long term remains unresolved. RECENT FINDINGS Many studies have advocated the success of MDA programs in order to attract donor funds for elimination efforts but such successes are often short-lived given the drug does not alter the life cycle of the organism or prevent reinfection. Within a matter of months to years after halting treatment, the prevalence, intensity of infection and morbidity of disease return to baseline levels. Other mitigating factors contribute to the failings of MDA campaigns namely: poverty, poor drug coverage, poor drug compliance, and, in the case of Asiatic schistosomiasis, zoonotic transmission. Genetic and innate and acquired immunologic mechanisms complicate the epidemiologic picture of schistosomiasis globally, and may contribute indirectly to MDA shortcomings. The possibility of drug resistance is an ever present concern because of the sole reliance on one drug, praziquantel. SUMMARY Preventive chemotherapy is advocated for the global control and elimination of schistosomiasis. The short-term benefits of MDA campaigns are well documented but the long-term benefits are questionable.
Collapse
|
27
|
Cabantous S, Hou X, Louis L, He H, Mariani O, Sastre X, Daujat-Chavanieu M, Li Y, Dessein A. Evidence for an important role of host microRNAs in regulating hepatic fibrosis in humans infected with Schistosoma japonicum. Int J Parasitol 2017; 47:823-830. [PMID: 28739251 DOI: 10.1016/j.ijpara.2017.05.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/12/2017] [Accepted: 05/18/2017] [Indexed: 11/17/2022]
Abstract
MicroRNAs (miRNAs) are short, non-coding RNAs that repress the translation of target gene transcripts. They have been implicated in various activities such as cell proliferation, survival, differentiation, migration and metabolism. We report here the first known miRNome and transcriptome analysis of human livers displaying advanced fibrosis due to Schistosoma japonicum infection. We present evidence that hsa-miR-150-5p, hsa-miR-10a-5p, hsa-miR-199a-3p, hsa-miR-4521, hsa-miR-222/221, hsa-miR-663b and hsa-miR-143-3p (associated without correction) play an important role in hepatic fibrosis by acting on metabolism, organization of the extracellular matrix proteins, lipid mobilization and limitation of oxidative damage stress.
Collapse
Affiliation(s)
- Sandrine Cabantous
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR906, GIMP, Labex ParaFrap, Aix-Marseille Université, 13005 Marseille, France.
| | - Xunya Hou
- Hunan Institute of Parasitic Diseases, Yueyang, China
| | - Laurence Louis
- INSERM UMR910, GMGF, Aix Marseille Université, 13005 Marseille, France
| | - Hongbin He
- Hunan Institute of Parasitic Diseases, Yueyang, China
| | | | | | | | - Yuesheng Li
- Queensland Institute of Medical Research, Brisbane, Australia
| | - Alain Dessein
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR906, GIMP, Labex ParaFrap, Aix-Marseille Université, 13005 Marseille, France.
| |
Collapse
|
28
|
Li L, Duan M, Chen W, Jiang A, Li X, Yang J, Li Z. The spleen in liver cirrhosis: revisiting an old enemy with novel targets. J Transl Med 2017; 15:111. [PMID: 28535799 PMCID: PMC5442653 DOI: 10.1186/s12967-017-1214-8] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/16/2017] [Indexed: 12/15/2022] Open
Abstract
The spleen is a secondary lymphoid organ which can influence the progression of multiple diseases, notably liver cirrhosis. In chronic liver diseases, splenomegaly and hypersplenism can manifest following the development of portal hypertension. These splenic abnormalities correlate with and have been postulated to facilitate the progression of liver fibrosis to cirrhosis, although precise mechanisms remain poorly understood. In this review, we summarize the literature to highlight the mechanistic contributions of splenomegaly and hypersplenism to the development of liver cirrhosis, focusing on three key aspects: hepatic fibrogenesis, hepatic immune microenvironment dysregulation and liver regeneration. We conclude with a discussion of the possible therapeutic strategies for modulating splenic abnormalities, including the novel potential usage of nanomedicine in non-surgically targetting splenic disorders for the treatment of liver cirrhosis.
Collapse
Affiliation(s)
- Liang Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China.,Liver and Spleen Diseases Research Center, Shaanxi Province, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China
| | - Mubing Duan
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Bundoora, VIC, Australia
| | - Weisan Chen
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Bundoora, VIC, Australia
| | - An Jiang
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China.,Liver and Spleen Diseases Research Center, Shaanxi Province, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China.,Department of General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China
| | - Xiaoming Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China
| | - Jun Yang
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China. .,Liver and Spleen Diseases Research Center, Shaanxi Province, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China. .,Department of Pathology, The Second Affiliated Hospital, Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China.
| | - Zongfang Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China. .,Liver and Spleen Diseases Research Center, Shaanxi Province, No.157, Xiwu Road, Xi'an, 710004, Shaanxi, China.
| |
Collapse
|
29
|
Romano A, Hou X, Sertorio M, Dessein H, Cabantous S, Oliveira P, Li J, Oyegue S, Arnaud V, Luo X, Daujat-Chavanieu M, Mariani O, Sastre X, Dombey AM, He H, Li Y, Dessein A. Correction: FOXP3+ Regulatory T Cells in Hepatic Fibrosis and Splenomegaly Caused by Schistosoma japonicum: The Spleen May Be a Major Source of Tregs in Subjects with Splenomegaly. PLoS Negl Trop Dis 2016; 10:e0004454. [PMID: 26849557 PMCID: PMC4743961 DOI: 10.1371/journal.pntd.0004454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|