1
|
Liu L, Wang P, Xie SQ, Pu WJ, Xu J, Xia CM. ICOSL deficiency promotes M1 polarization to alleviate liver fibrosis in schistosomiasis mice. Acta Trop 2025; 261:107470. [PMID: 39581561 DOI: 10.1016/j.actatropica.2024.107470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/26/2024]
Abstract
The expression of inducible co-stimulator ligand (ICOSL) on macrophage (Mφ) implies their ability to interact with inducible co-stimulator (ICOS)-expressing T cells, thereby modulating immune responses within the liver microenvironment. This study aimed to elucidate the mechanism underlying ICOS/ICOSL signaling in the regulation of Mφ polarization during Schistosomiasis-induced liver fibrosis. To investigate this, ICOSL-knock out (KO) and wildtype (WT) C57BL/6 mice were infected with Schistosoma japonicum (S. japonicum) to examine the dynamic changes in Mφ phenotype and observe the pathology alterations in the liver. There was significantly decreased expression of ICOSL both in monocytes of cirrhosis patients and the liver tissue of mice infected with S. japonicum. Furthermore, ICOSL-KO mice exhibited reduced liver granuloma formation and fibrosis during S. japonicum infection. Simultaneously, Mφ in ICOSL-KO mice polarized towards M1-type and induced apoptosis of hepatic stellate cells (HSCs). Overall, the blockade of ICOSL signaling could promote M1 polarization, induce HSCs apoptosis, and ameliorate hepatic fibrosis, suggesting that ICOSL may serve as a potential biomarker for prognosis and therapeutic target for schistosomiasis-induced hepatic fibrosis.
Collapse
Affiliation(s)
- Lei Liu
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, Jiangsu, China; Department of Blood Transfusion, The First Affiliated Hospital of University of Science and Technology of China, 17 Lujiang Road, Hefei 230032, Anhui, China
| | - Peng Wang
- Department of Health Promotion and Behavioral Sciences, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China
| | - Shi-Qi Xie
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, Jiangsu, China
| | - Wen-Jie Pu
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, Jiangsu, China
| | - Jing Xu
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, Jiangsu, China
| | - Chao-Ming Xia
- Department of Parasitology, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, Jiangsu, China.
| |
Collapse
|
2
|
El-kady AM, Altwaim SA, Wakid MH, Banjar AS, Mohammed K, Alfaifi MS, Elshazly H, Al-Megrin WAI, Alshehri EA, Sayed E, Elshabrawy HA. Prior Trichinella spiralis infection protects against Schistosoma mansoni induced hepatic fibrosis. Front Vet Sci 2024; 11:1443267. [PMID: 39439825 PMCID: PMC11494294 DOI: 10.3389/fvets.2024.1443267] [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: 06/03/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Background Schistosomiasis affects approximately 250 million people worldwide, with 200,000 deaths annually. It has been documented that the granulomatous response to Schistosoma mansoni (S. mansoni) oviposition is the root cause of progressive liver fibrosis in chronic infection, in 20% of the patients, and can lead to liver cirrhosis and/or liver cancer. The influence of helminths coinfection on schistosomiasis-induced liver pathological alterations remains poorly understood. Therefore, in this study, we investigated the effect of Trichinella spiralis (T. spiralis) infection on S. mansoni-induced hepatic fibrosis. Materials and methods Thirty adult male Balb-c mice were divided into three groups. Group 1 was left uninfected; group 2 was infected with S. mansoni cercariae and group 3 was orally infected with T. spiralis larvae, then 28 days later, this group was infected with S. mansoni cercariae. All groups were sacrificed at the end of the 8th week post infection with S. mansoni to evaluate the effect of pre-infection with T. spiralis on S. mansoni induced liver fibrosis was evaluated parasitologically (worm burden and egg count in tissues), biochemically (levels of alanine aminotransferase and aspartate aminotransferase), histopathologically (H&E and MT staining, and immunohistochemical staining for the expression of α-SMA, IL-6, IL-1β, IL-17, IL-23, TNF-α, and TGF-β). Results The results in the present study demonstrated marked protective effect of T. spiralis against S. mansoni induced liver pathology. We demonstrated that pre-infection with T. spirais caused marked reduction in the number of S. mansoni adult worms (3.17 ± 0.98 vs. 18 ± 2.16, P = 0.114) and egg count in both the intestine (207.2 ± 64.3 vs. 8,619.43 ± 727.52, P = 0.009) and liver tissues (279 ± 87.2 vs. 7,916.86 ± 771.34; P = 0.014). Consistently, we found significant reductions in both number (3.4 ± 1.1 vs. 11.8.3 ± 1.22; P = 0.007) and size (84 ± 11 vs. 294.3 ± 16.22; P = 0.001) of the hepatic granulomas in mice pre-infected with T. spiralis larvae compared to those infected with only S. mansoni. Furthermore, pre- infection with T. spiralis markedly reduced S. mansoni- induced hepatic fibrosis, as evidenced by decreased collagen deposition, low expression of α-SMA, and significantly reduced levels of IL-17, IL-1B, IL-6, TGF-B, IL-23, and TNF-α compared to mice infected with S. mansoni only. Conclusions Our data show that pre-infection with T. spiralis effectively protected mice from severe schistosomiasis and liver fibrosis. We believe that our findings support the potential utility of helminths for the preventing and ameliorating severe pathological alterations induced by schistosomiasis.
Collapse
Affiliation(s)
- Asmaa M. El-kady
- Department of Medical Parasitology, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Sarah A. Altwaim
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, Jeddah, Saudi Arabia
| | - Majed H. Wakid
- Special Infectious Agents Unit, King Fahd Medical Research Center, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Alaa S. Banjar
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Khalil Mohammed
- Department of Epidemiology and Medical Statistics, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Mashael S. Alfaifi
- Department of Epidemiology and Medical Statistics, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Hayam Elshazly
- Department of Biology, Faculty of Sciences-Scientific Departments, Qassim University, Buraidah, Qassim, Saudi Arabia
- Department of Zoology, Faculty of Science, Beni-Suef University, Beni Suef, Egypt
| | - Wafa Abdullah I. Al-Megrin
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | | | - Eman Sayed
- Department of Parasitology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Hatem A. Elshabrawy
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX, United States
| |
Collapse
|
3
|
Li Q, Wang J, Lv J, Liu D, Xiao S, Mo J, Lu Z, Qiu R, Li C, Tang L, He S, Tang Z, Cheng Q, Zhan T. Total flavonoids of litchi Seed alleviates schistosomiasis liver fibrosis in mice by suppressing hepatic stellate cells activation and modulating the gut microbiomes. Biomed Pharmacother 2024; 178:117240. [PMID: 39094546 DOI: 10.1016/j.biopha.2024.117240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/20/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024] Open
Abstract
Infection with Schistosoma japonicum (S. japonicum) is an important zoonotic parasitic disease that causes liver fibrosis in both human and domestic animals. The activation of hepatic stellate cells (HSCs) is a crucial phase in the development of liver fibrosis, and inhibiting their activation can alleviate this progression. Total flavonoids of litchi seed (TFL) is a naturally extracted drug, and modern pharmacological studies have shown its anti-fibrotic and liver-protective effects. However, the role of TFL in schistosomiasis liver fibrosis is still unclear. This study investigated the therapeutic effects of TFL on liver fibrosis in S. japonicum infected mice and explored its potential mechanisms. Animal study results showed that TFL significantly reduced the levels of Interleukin-1β (IL-1β), Tumor Necrosis Factor-α (TNF-α), Interleukin-4 (IL-4), and Interleukin-6 (IL-6) in the serum of S. japonicum infected mice. TFL reduced the spleen index of mice and markedly improved the pathological changes in liver tissues induced by S. japonicum infection, decreasing the expression of alpha-smooth muscle actin (α-SMA), Collagen I and Collagen III protein in liver tissues. In vitro studies indicated that TFL also inhibited the activation of HCSs induced by Transforming Growth Factor-β1 (TGF-β1) and reduced the levels of α-SMA. Gut microbes metagenomics study revealed that the composition, abundance, and functions of the mice gut microbiomes changed significantly after S. japonicum infection, and TLF treatment reversed these changes. Therefore, our study indicated that TFL alleviated granulomatous lesions and improved S. japonicum induced liver fibrosis in mice by inhibiting the activation of HSCs and by improving the gut microbiomes.
Collapse
Affiliation(s)
- Qing Li
- Department of Cell Biology and Genetics, Guangxi Medical University, Nanning, Guangxi, China; Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China; Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Jilong Wang
- Department of Parasitology, Guangxi Medical University, Nanning, Guangxi, China
| | - Jiahui Lv
- Department of Parasitology, Guangxi Medical University, Nanning, Guangxi, China
| | - Dengyu Liu
- Department of Parasitology, Guangxi Medical University, Nanning, Guangxi, China
| | - Suyu Xiao
- Department of Parasitology, Guangxi Medical University, Nanning, Guangxi, China
| | - Jingquan Mo
- School of Pre-clinical Medicine, Guangxi Medical University, Nanning, China
| | - Zuochao Lu
- Department of Parasitology, Guangxi Medical University, Nanning, Guangxi, China
| | - Ran Qiu
- School of Pre-clinical Medicine, Guangxi Medical University, Nanning, China
| | - Caiqi Li
- School of Pre-clinical Medicine, Guangxi Medical University, Nanning, China
| | - Lili Tang
- Department of Parasitology, Guangxi Medical University, Nanning, Guangxi, China
| | - Shanshan He
- Department of Parasitology, Guangxi Medical University, Nanning, Guangxi, China
| | - Zeli Tang
- Department of Cell Biology and Genetics, Guangxi Medical University, Nanning, Guangxi, China; Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China; Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China.
| | - Qiuchen Cheng
- Department of Gastroenterology, the People's Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, Guangxi, China.
| | - Tingzheng Zhan
- Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China; Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China; Department of Parasitology, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
4
|
Mertelsmann AM, Bowers SF, Wright D, Maganga JK, Mazigo HD, Ndhlovu LC, Changalucha JM, Downs JA. Effects of Schistosoma haematobium infection and treatment on the systemic and mucosal immune phenotype, gene expression and microbiome: A systematic review. PLoS Negl Trop Dis 2024; 18:e0012456. [PMID: 39250522 PMCID: PMC11412685 DOI: 10.1371/journal.pntd.0012456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 09/19/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Urogenital schistosomiasis caused by Schistosoma haematobium affects approximately 110 million people globally, with the majority of cases in low- and middle-income countries. Schistosome infections have been shown to impact the host immune system, gene expression, and microbiome composition. Studies have demonstrated variations in pathology between schistosome subspecies. In the case of S. haematobium, infection has been associated with HIV acquisition and bladder cancer. However, the underlying pathophysiology has been understudied compared to other schistosome species. This systematic review comprehensively investigates and assimilates the effects of S. haematobium infection on systemic and local host mucosal immunity, cellular gene expression and microbiome. METHODS We conducted a systematic review assessing the reported effects of S. haematobium infections and anthelmintic treatment on the immune system, gene expression and microbiome in humans and animal models. This review followed PRISMA guidelines and was registered prospectively in PROSPERO (CRD42022372607). Randomized clinical trials, cohort, cross-sectional, case-control, experimental ex vivo, and animal studies were included. Two reviewers performed screening independently. RESULTS We screened 3,177 studies and included 94. S. haematobium was reported to lead to: (i) a mixed immune response with a predominant type 2 immune phenotype, increased T and B regulatory cells, and select pro-inflammatory cytokines; (ii) distinct molecular alterations that would compromise epithelial integrity, such as increased metalloproteinase expression, and promote immunological changes and cellular transformation, specifically upregulation of genes p53 and Bcl-2; and (iii) microbiome dysbiosis in the urinary, intestinal, and genital tracts. CONCLUSION S. haematobium induces distinct alterations in the host's immune system, molecular profile, and microbiome. This leads to a diverse range of inflammatory and anti-inflammatory responses and impaired integrity of the local mucosal epithelial barrier, elevating the risks of secondary infections. Further, S. haematobium promotes cellular transformation with oncogenic potential and disrupts the microbiome, further influencing the immune system and genetic makeup. Understanding the pathophysiology of these interactions can improve outcomes for the sequelae of this devastating parasitic infection.
Collapse
Affiliation(s)
- Anna M Mertelsmann
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, United States of America
- Center for Global Health, Weill Cornell Medicine, New York, New York, United States of America
| | - Sheridan F Bowers
- Center for Global Health, Weill Cornell Medicine, New York, New York, United States of America
| | - Drew Wright
- Samuel J. Wood Library & C.V. Starr Biomedical Information Center, Weill Cornell Medical College, New York, New York, United States of America
| | - Jane K Maganga
- Mwanza Intervention Trials Unit/National Institute for Medical Research, Mwanza, Tanzania
| | - Humphrey D Mazigo
- Department of Parasitology and Entomology, Catholic University of Health and Allied Sciences, Mwanza, Tanzania
| | - Lishomwa C Ndhlovu
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - John M Changalucha
- Mwanza Intervention Trials Unit/National Institute for Medical Research, Mwanza, Tanzania
| | - Jennifer A Downs
- Center for Global Health, Weill Cornell Medicine, New York, New York, United States of America
- Mwanza Intervention Trials Unit/National Institute for Medical Research, Mwanza, Tanzania
- Weill Bugando School of Medicine, Mwanza, Tanzania
| |
Collapse
|
5
|
Huang P, Xu J, Jiang S, Zhang Y, Wang X, Xiong C, Xia C. The impact of ICOSL/ICOS pathway-regulated long non-coding RNAs on liver fibrosis in mice infected with Schistosoma japonicum. Parasit Vectors 2024; 17:317. [PMID: 39044218 PMCID: PMC11267842 DOI: 10.1186/s13071-024-06399-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/08/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND The primary pathogenic mechanism of schistosomiasis-associated liver fibrosis involves the deposition of schistosome eggs, leading to the formation of liver egg granulomas and subsequent liver fibrosis. Hepatic stellate cells are abnormally activated, resulting in excessive collagen deposition and fibrosis development. While specific long non-coding RNAs (lncRNAs) have been associated with fibrotic processes, their roles in schistosomiasis-associated liver fibrosis remain unclear. METHODS Our previous research indicated that downregulating the ICOSL/ICOS could partially alleviate liver fibrosis. In this study, we established a schistosomiasis infection model in C57BL/6 and ICOSL knockout (KO) mice, and the liver pathology changes were observed at various weeks postinfection (wpi) using hematoxylin and eosin and Masson's trichrome staining. Within the first 4 wpi, no significant liver abnormalities were observed. However, mice exhibited evident egg granulomas and fibrosis in their livers at 7 wpi. Notably, ICOSL-KO mice had significantly smaller pathological variations compared with simultaneously infected C57BL/6 mice. To investigate the impact of lncRNAs on schistosomiasis-associated liver fibrosis, quantitative real-time polymerase chain reaction (RT-qPCR) was used to monitor the dynamic changes of lncRNAs in hepatic stellate cells of infected mice. RESULTS The results demonstrated that lncRNA-H19, -MALAT1, -PVT1, -P21 and -GAS5 all participated in liver fibrosis formation after schistosome infection. In addition, ICOSL-KO mice exhibited significantly inhibited expression of lncRNA-H19, -MALAT1 and -PVT1 after 7 wpi. In contrast, they showed enhanced expression of lncRNA-P21 and -GAS5 compared with C57BL/6 mice, influencing liver fibrosis development. Furthermore, small interfering RNA transfection (siRNA) in JS-1 cells in vitro confirmed that lncRNA-H19, -MALAT1, and -PVT1 promoted liver fibrosis, whereas lncRNA-P21 and -GAS5 had the opposite effect on key fibrotic molecules, including α- smooth muscle actin and collagen I expression. CONCLUSIONS This study uncovers that ICOSL/ICOS may play a role in activating hepatic stellate cells and promoting liver fibrosis in mice infected with Schistosoma japonicum by dynamically regulating the expression of specific lncRNAs. These findings offer potential therapeutic targets for schistosomiasis-associated liver fibrosis.
Collapse
Affiliation(s)
- Ping Huang
- Department of Parasitology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Jing Xu
- Department of Parasitology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Suqin Jiang
- Department of Parasitology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Yanan Zhang
- Department of Parasitology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Xinyi Wang
- Department of Parasitology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Chunrong Xiong
- Jiangsu Institute of Parasitic Diseases, Wuxi City, Jiangsu Province, China
| | - Chaoming Xia
- Department of Parasitology, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China.
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, School of Biology & Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China.
| |
Collapse
|
6
|
Nogueira RA, Lira MGS, Licá ICL, Frazão GCCG, Dos Santos VAF, Filho ACCM, Rodrigues JGM, Miranda GS, Carvalho RC, Nascimento FRF. Praziquantel: An update on the mechanism of its action against schistosomiasis and new therapeutic perspectives. Mol Biochem Parasitol 2022; 252:111531. [PMID: 36375598 DOI: 10.1016/j.molbiopara.2022.111531] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/03/2022] [Accepted: 11/10/2022] [Indexed: 11/13/2022]
Abstract
Praziquantel (PZQ) is the drug of choice for the treatment of all forms of schistosomiasis, although its mechanisms of action are not completely understood. PZQ acts largely on adult worms. This narrative literature review describes what is known about the mechanisms of action of PZQ against schistosomes from in vitro and in vivo studies and highlights the molecular targets in parasites and immune responses induced in definitive hosts by this drug. Moreover, new therapeutic uses of PZQ are discussed. Studies have demonstrated that in addition to impacting voltage-operated Ca2 + channels, PZQ may interact with other schistosome molecules, such as myosin regulatory light chain, glutathione S-transferase, and transient receptor potential channels. Following PZQ administration, increased T regulatory type 1 (Tr1) cell differentiation and decreased inflammation were observed, indicating that PZQ promotes immunoregulatory pathways. Although PZQ is widely used in mass drug administration schemes, the existence of resistant parasites has not been proven; however, it is a concern that should be constantly investigated in human populations. In addition, we discuss studies that evaluate health applications of PZQ (other than helminth infection), such as its effect in cancer therapy and its adjuvant action in vaccines against viruses.
Collapse
Affiliation(s)
- Ranielly Araujo Nogueira
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Maria Gabriela Sampaio Lira
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil; Department of Education, Federal Institute of Education, Science and Technology of Maranhão, Zé Doca, MA, Brazil
| | - Irlla Correia Lima Licá
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | | | - Vitor Augusto Ferreira Dos Santos
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | | | - João Gustavo Mendes Rodrigues
- Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Guilherme Silva Miranda
- Department of Education, Federal Institute of Education, Science and Technology of Maranhão, São Raimundo das Mangabeiras, MA, Brazil
| | - Rafael Cardoso Carvalho
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Flávia Raquel Fernandes Nascimento
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil; Department of Pathology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil.
| |
Collapse
|
7
|
Miranda GS, Rodrigues JGM, de Rezende MC, Resende SD, Camelo GMA, de Oliveira Silva JKA, Maggi L, Rodrigues VF, de Oliveira VG, Negrão-Corrêa DA. Experimental infection with Schistosoma mansoni isolated from the wild rodent Holochilus sciureus shows a low parasite burden but induces high schistosomiasis severity in BALB/c mice. Parasitology 2022; 149:1381-1396. [PMID: 35641335 PMCID: PMC11010505 DOI: 10.1017/s0031182022000774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 11/06/2022]
Abstract
Wild mammals, especially rodents, can participate in the life cycle of Schistosoma mansoni; however, the impact of these parasite strains on the severity of schistosomiasis remains unclear. The aim of this study was to comparatively evaluate the parasitological and immunopathological alterations induced by an S. mansoni strain isolated from the wild rodent Holochilus sciureus (HS strain) and a parasite strain isolated from a human (LE strain) in experimentally infected mice. Male BALB/c mice were subcutaneously infected with 50 cercariae/mouse of either the HS or the LE strain and were evaluated for 12 weeks. In the experimental groups, the parasite burden was estimated by worm and egg (feces and tissues) count, and immunopathological alterations were evaluated in the liver and intestines. Compared to experimental infection with the LE parasite strain, HS-infected mice showed reduced number of parasite worms but higher fecundity rate, significant reduction in IL-5, IL-10 and IL-13 concentrations, lower EPO-activity in liver homogenate and higher concentrations of TNF-α, IFN-γ, IL-12 and IL-17 in the small intestine homogenate. Moreover, HS infection resulted in higher concentrations of NO end-products in both the liver and intestine, suggesting a predominance of the Th1/Th17 immune response. HS-infected mice also showed higher plasma transaminase levels, formed larger granulomas, and had a higher mortality rate in comparison with LE-infected mice. Data indicate that BALB/c mice infected with the HS strain of S. mansoni showed reduced susceptibility to the parasite but stronger tissue inflammation and high disease severity.
Collapse
Affiliation(s)
- Guilherme Silva Miranda
- Department of Parasitology, Federal University of Minas Gerais, Institute of Biological Sciences, Belo Horizonte, Brazil
- Department of Biology, Federal Institute of Education, Science and Technology of Maranhão, São Raimundo das Mangabeiras, Brazil
| | - João Gustavo Mendes Rodrigues
- Department of Parasitology, Federal University of Minas Gerais, Institute of Biological Sciences, Belo Horizonte, Brazil
| | - Michelle Carvalho de Rezende
- Department of Parasitology, Federal University of Minas Gerais, Institute of Biological Sciences, Belo Horizonte, Brazil
| | - Samira Diniz Resende
- Department of Parasitology, Federal University of Minas Gerais, Institute of Biological Sciences, Belo Horizonte, Brazil
| | - Genil Mororó Araújo Camelo
- Department of Parasitology, Federal University of Minas Gerais, Institute of Biological Sciences, Belo Horizonte, Brazil
| | | | - Laura Maggi
- Department of Parasitology, Federal University of Minas Gerais, Institute of Biological Sciences, Belo Horizonte, Brazil
| | - Vanessa Fernandes Rodrigues
- Department of Parasitology, Federal University of Minas Gerais, Institute of Biological Sciences, Belo Horizonte, Brazil
| | - Vinícius Gustavo de Oliveira
- Department of Parasitology, Federal University of Minas Gerais, Institute of Biological Sciences, Belo Horizonte, Brazil
| | | |
Collapse
|
8
|
Chen H, Li G, Zhang J, Zheng T, Chen Q, Zhang Y, Yang F, Wang C, Nie H, Zheng B, Gong Q. Sodium butyrate ameliorates Schistosoma japonicum-induced liver fibrosis by inhibiting HMGB1 expression. Exp Parasitol 2021; 231:108171. [PMID: 34736899 DOI: 10.1016/j.exppara.2021.108171] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/16/2021] [Accepted: 10/31/2021] [Indexed: 11/25/2022]
Abstract
Schistosomiasis is a prevalent zoonotic parasitic disease caused by schistosomes. Its main threat to human health is hepatic granuloma and fibrosis due to worm eggs. Praziquantel remains the first choice for the treatment of schistosomiasis but has limited benefit in treating liver fibrosis. Therefore, the need to develop effective drugs for treating schistosomiasis-induced hepatic fibrosis is urgent. High-mobility group box 1 protein (HMGB1) is a potential immune mediator that is highly associated with the development of some fibrotic diseases and may be involved in the liver pathology of schistosomiasis. We speculated that HMGB1 inhibitors could have an anti-fibrotic effect. Sodium butyrate (SB), a potent inhibitor of HMGB1, has shown anti-inflammatory activity in some animal disease models. In this study, we evaluated the effects of SB on a murine schistosomiasis model. Mice were percutaneously infected with 20 ± 2 cercariae of Schistosoma japonicum. SB (500 mg/kg/day) was administered every 3 days for the entire experiment period. The activity of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), liver histopathology, HMGB1 expression, and the levels of interferon gamma (IFN-γ), transforming growth factor-β1 (TGF-β1), and interleukin-6 (IL-6) in serum were analyzed. SB reduced hepatic granuloma and fibrosis of schistosomiasis, reflected by the decreased levels of ALT and AST in serum and the reduced expression of pro-inflammatory and fibrogenic cytokines (IFN-γ, TGF-β1, and IL-6). The protective effect could be attributable to the inhibition of the expression of HMGB1 and release by SB.
Collapse
Affiliation(s)
- Hui Chen
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Gang Li
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Department of Gastroenterology, Jingmen Second People's Hospital, Jingmen, Hubei Province, 448000, PR China
| | - Jianqiang Zhang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Ting Zheng
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Qianglin Chen
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Yanxiang Zhang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Fei Yang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Chao Wang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Hao Nie
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Bing Zheng
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China.
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China.
| |
Collapse
|
9
|
Wan M, Han J, Ding L, Hu F, Gao P. Novel Immune Subsets and Related Cytokines: Emerging Players in the Progression of Liver Fibrosis. Front Med (Lausanne) 2021; 8:604894. [PMID: 33869241 PMCID: PMC8047058 DOI: 10.3389/fmed.2021.604894] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/05/2021] [Indexed: 12/12/2022] Open
Abstract
Liver fibrosis is a pathological process caused by persistent chronic injury of the liver. Kupffer cells, natural killer (NK) cells, NKT cells, and dendritic cells (DCs), which are in close contact with T and B cells, serve to bridge innate and adaptive immunity in the liver. Meanwhile, an imbalanced inflammatory response constitutes a challenge in liver disease. The dichotomous roles of novel immune cells, including T helper 17 (Th17), regulatory T cells (Tregs), mucosa-associated invariant T cells (MAIT), and innate lymphoid cells (ILCs) in liver fibrosis have gradually been revealed. These cells not only induce damage during liver fibrosis but also promote tissue repair. Hence, immune cells have unique, and often opposing, roles during the various stages of fibrosis. Due to this heterogeneity, the treatment, or reversal of fibrosis through the target of immune cells have attracted much attention. Moreover, activation of hepatic stellate cells (HSCs) constitutes the core of fibrosis. This activation is regulated by various immune mediators, including Th17, Th22, and Th9, MAIT, ILCs, and γδ T cells, as well as their related cytokines. Thus, liver fibrosis results from the complex interaction of these immune mediators, thereby complicating the ability to elucidate the mechanisms of action elicited by each cell type. Future developments in biotechnology will certainly aid in this feat to inform the design of novel therapeutic targets. Therefore, the aim of this review was to summarize the role of specific immune cells in liver fibrosis, as well as biomarkers and treatment methods related to these cells.
Collapse
Affiliation(s)
- Minjie Wan
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, China.,Central Laboratory, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Jiawen Han
- Central Laboratory, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Lili Ding
- Central Laboratory, The First Hospital of Jilin University, Jilin University, Changchun, China.,Intensive Care Unit, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Feng Hu
- Department of Hepatology and Gastroenterology, The Second Part of First Hospital, Jilin University, Changchun, China
| | - Pujun Gao
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
10
|
Miranda GS, Resende SD, Cardoso DT, Camelo GMA, Silva JKAO, de Castro VN, Geiger SM, Carneiro M, Negrão-Corrêa D. Previous History of American Tegumentary Leishmaniasis Alters Susceptibility and Immune Response Against Schistosoma mansoni Infection in Humans. Front Immunol 2021; 12:630934. [PMID: 33777015 PMCID: PMC7990892 DOI: 10.3389/fimmu.2021.630934] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/18/2021] [Indexed: 01/21/2023] Open
Abstract
Schistosomiasis and Leishmaniasis are chronic parasitic diseases with high prevalence in some tropical regions and, due to their wide distribution, a risk of co-infections is present in some areas. Nevertheless, the impact of this interaction on human populations is still poorly understood. Thus, the current study evaluated the effect of previous American Tegumentary Leishmaniasis (ATL) on the susceptibility and immune response to Schistosoma mansoni infection in residents from a rural community in Northern of Minas Gerais state, Brazil, an area endemic for both parasitic infections. The participants answered a socioeconomic questionnaire and provided stool and blood samples for parasitological and immunological evaluations. Stool samples were examined by a combination of parasitological techniques to identify helminth infections, especially S. mansoni eggs. Blood samples were used for hemograms and to measure the serum levels of cytokines and chemokines. Reports on previous ATL were obtained through interviews, clinical evaluation forms, and medical records. S. mansoni infection was the most prevalent parasitic infection in the study population (46%), and the majority of the infected individuals had a very low parasite burden. In the same population, 93 individuals (36.2%) reported previous ATL, and the prevalence of S. mansoni infection among these individuals was significantly higher than among individuals with no ATL history. A multiple logistic regression model revealed that S. mansoni infection was positively associated with higher levels of CCL3 and CCL17, and a higher frequency of IL-17 responders. Moreover, this model demonstrated that individuals with an ATL history had a 2-fold higher probability to be infected with S. mansoni (OR = 2.0; 95% CI 1.04–3.68). Among S. mansoni-infected individuals, the logistic regression demonstrated that a previous ATL history was negatively associated with the frequency of IL-17 responders and CXCL10 higher responders, but positively associated with higher IL-27 responders. Altogether, our data suggest that previous ATL may alter the susceptibility and the immune response in S. mansoni-infected individuals, which may likely affect the outcome of schistosomiasis and the severity of the disease in humans.
Collapse
Affiliation(s)
- Guilherme Silva Miranda
- Laboratory of Immunohelminthology and Schistosomiasis, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil.,Laboratory of Biology, Department of Biology, Institute of Education, Science and Technology of Maranhão, São Raimundo das Mangabeiras, Brazil
| | - Samira Diniz Resende
- Laboratory of Immunohelminthology and Schistosomiasis, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Diogo Tavares Cardoso
- Laboratory of Intestinal Helminthiasis, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Genil Mororó Araújo Camelo
- Laboratory of Immunohelminthology and Schistosomiasis, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Jeferson Kelvin Alves Oliveira Silva
- Laboratory of Immunohelminthology and Schistosomiasis, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Vanessa Normandio de Castro
- Laboratory of Intestinal Helminthiasis, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Stefan Michael Geiger
- Laboratory of Intestinal Helminthiasis, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Mariângela Carneiro
- Laboratory of Epidemiology of Infectious and Parasitic Diseases, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Deborah Negrão-Corrêa
- Laboratory of Immunohelminthology and Schistosomiasis, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
11
|
Zhang W, Le L, Ahmad G, Molehin AJ, Siddiqui AJ, Torben W, Karmakar S, Rojo JU, Sennoune S, Lazarus S, Khatoon S, Freeborn J, Sudduth J, Rezk AF, Carey D, Wolf RF, Papin JF, Damian R, Gray SA, Marks F, Carter D, Siddiqui AA. Fifteen Years of Sm-p80-Based Vaccine Trials in Nonhuman Primates: Antibodies From Vaccinated Baboons Confer Protection in vivo and in vitro From Schistosoma mansoni and Identification of Putative Correlative Markers of Protection. Front Immunol 2020; 11:1246. [PMID: 32636844 PMCID: PMC7318103 DOI: 10.3389/fimmu.2020.01246] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022] Open
Abstract
Recent advances in systems biology have shifted vaccine development from a largely trial-and-error approach to an approach that promote rational design through the search for immune signatures and predictive correlates of protection. These advances will doubtlessly accelerate the development of a vaccine for schistosomiasis, a neglected tropical disease that currently affects over 250 million people. For over 15 years and with contributions of over 120 people, we have endeavored to test and optimize Sm-p80-based vaccines in the non-human primate model of schistosomiasis. Using RNA-sequencing on eight different Sm-p80-based vaccine strategies, we sought to elucidate immune signatures correlated with experimental protective efficacy. Furthermore, we aimed to explore the role of antibodies through in vivo passive transfer of IgG obtained from immunized baboons and in vitro killing of schistosomula using Sm-p80-specific antibodies. We report that passive transfer of IgG from Sm-p80-immunized baboons led to significant worm burden reduction, egg reduction in liver, and reduced egg hatching percentages from tissues in mice compared to controls. In addition, we observed that sera from Sm-p80-immunized baboons were able to kill a significant percent of schistosomula and that this effect was complement-dependent. While we did not find a universal signature of immunity, the large datasets generated by this study will serve as a substantial resource for further efforts to develop vaccine or therapeutics for schistosomiasis.
Collapse
Affiliation(s)
- Weidong Zhang
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Loc Le
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Gul Ahmad
- Department of Natural Sciences, Peru State College, Peru, NE, United States
| | - Adebayo J. Molehin
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | | | - Workineh Torben
- Department of Biological Sciences, Louisiana State University of Alexandria, Alexandria, LA, United States
| | - Souvik Karmakar
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Juan U. Rojo
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH, United States
| | - Souad Sennoune
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Samara Lazarus
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Sabiha Khatoon
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Jasmin Freeborn
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Justin Sudduth
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Ashraf F. Rezk
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - David Carey
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Roman F. Wolf
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Oklahoma City VA Health Care System, Oklahoma City, OK, United States
| | - James F. Papin
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Ray Damian
- Department of Cellular Biology, University of Georgia, Athens, GA, United States
| | | | - Florian Marks
- International Vaccine Institute, SNU Research Park, Seoul, South Korea
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Darrick Carter
- PAI Life Sciences, Seattle, WA, United States
- Infectious Disease Research Institute, Seattle, WA, United States
| | - Afzal A. Siddiqui
- Center for Tropical Medicine and Infectious Diseases, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
12
|
Zheng B, Zhang J, Chen H, Nie H, Miller H, Gong Q, Liu C. T Lymphocyte-Mediated Liver Immunopathology of Schistosomiasis. Front Immunol 2020; 11:61. [PMID: 32132991 PMCID: PMC7040032 DOI: 10.3389/fimmu.2020.00061] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/10/2020] [Indexed: 12/16/2022] Open
Abstract
The parasitic worms, Schistosoma mansoni and Schistosoma japonicum, reside in the mesenteric veins, where they release eggs that induce a dramatic granulomatous response in the liver and intestines. Subsequently, infection may further develop into significant fibrosis and portal hypertension. Over the past several years, uncovering the mechanism of immunopathology in schistosomiasis has become a major research objective. It is known that T lymphocytes, especially CD4+ T cells, are essential for immune responses against Schistosoma species. However, obtaining a clear understanding of how T lymphocytes regulate the pathological process is proving to be a daunting challenge. To date, CD4+ T cell subsets have been classified into several distinct T helper (Th) phenotypes including Th1, Th2, Th17, T follicular helper cells (Tfh), Th9, and regulatory T cells (Tregs). In the case of schistosomiasis, the granulomatous inflammation and the chronic liver pathology are critically regulated by the Th1/Th2 responses. Animal studies suggest that there is a moderate Th1 response to parasite antigens during the acute stage, but then, egg-derived antigens induce a sustained and dominant Th2 response that mediates granuloma formation and liver fibrosis. In addition, the newly discovered Th17 cells also play a critical role in the hepatic immunopathology of schistosomiasis. Within the liver, Tregs are recruited to hepatic granulomas and exert an immunosuppressive role to limit the granulomatous inflammation and fibrosis. Moreover, recent studies have shown that Tfh and Th9 cells might also promote liver granulomas and fibrogenesis in the murine schistosomiasis. Thus, during infection, T-cell subsets undergo complicated cross-talk with antigen presenting cells that then defines their various roles in the local microenvironment for regulating the pathological progression of schistosomiasis. This current review summarizes a vast body of literature to elucidate the contribution of T lymphocytes and their associated cytokines in the immunopathology of schistosomiasis.
Collapse
Affiliation(s)
- Bing Zheng
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China.,Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Jianqiang Zhang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Hui Chen
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Hao Nie
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China.,Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Heather Miller
- Department of Intracellular Pathogens, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China.,Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| |
Collapse
|
13
|
Mandal S, Rigopoulou EI, Mandal M, Bogdanos DP. Immunity of Parasitic Infections of the Liver. LIVER IMMUNOLOGY 2020:197-209. [DOI: 10.1007/978-3-030-51709-0_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
14
|
Solaymani-Mohammadi S, Eckmann L, Singer SM. Interleukin (IL)-21 in Inflammation and Immunity During Parasitic Diseases. Front Cell Infect Microbiol 2019; 9:401. [PMID: 31867283 PMCID: PMC6904299 DOI: 10.3389/fcimb.2019.00401] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/11/2019] [Indexed: 12/30/2022] Open
Abstract
Parasitic diseases cause significant morbidity and mortality in the developing and underdeveloped countries. No efficacious vaccines are available against most parasitic diseases and there is a critical need for developing novel vaccine strategies for care. IL-21 is a pleiotropic cytokine whose functions in protection and immunopathology during parasitic diseases have been explored in limited ways. IL-21 and its cognate receptor, IL-21R, are highly expressed in parasitized organs of infected humans as well in murine models of the human parasitic diseases. Prior studies have indicated the ability of the IL-21/IL-21R signaling axis to regulate the effector functions (e.g., cytokine production) of T cell subsets by enhancing the expression of T-bet and STAT4 in human T cells, resulting in an augmented production of IFN-γ. Mice deficient for either IL-21 (Il21−/−) or IL-21R (Il21r−/−) showed significantly reduced inflammatory responses following parasitic infections as compared with their WT counterparts. Targeting the IL-21/IL-21R signaling axis may provide a novel approach for the development of new therapeutic agents for the prevention of parasite-induced immunopathology and tissue destruction.
Collapse
Affiliation(s)
- Shahram Solaymani-Mohammadi
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Lars Eckmann
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Steven M Singer
- Department of Biology, Georgetown University, Washington, DC, United States
| |
Collapse
|
15
|
Inhibition of Rho-Kinase Downregulates Th17 Cells and Ameliorates Hepatic Fibrosis by Schistosoma japonicum Infection. Cells 2019; 8:cells8101262. [PMID: 31623153 PMCID: PMC6829618 DOI: 10.3390/cells8101262] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Schistosomiasis is an immunopathogenic disease in which Th17 cells play vital roles. Hepatic granuloma formation and subsequent fibrosis are its main pathologic manifestations and the leading causes of hepatic cirrhosis, and effective therapeutic interventions are lacking. In this study, we explored the effects of fasudil, a selective RhoA-Rho-associated kinase (ROCK) inhibitor, on Th17 cells and the pathogenesis of schistosomiasis. METHODS Mice were infected with Schistosoma japonicum and treated with fasudil. The worm burden, hepatic granuloma formation, and fibrosis were evaluated. The roles of fasudil on Th17, Treg, and hepatic stellate cells were analyzed. RESULTS Fasudil therapy markedly reduced the granuloma size and collagen deposit in livers from mice infected with S. japonicum. However, fasudil therapy did not affect the worm burden in infected mice. The underlying cellular and molecular mechanisms were investigated. Fasudil suppressed the activation and induced the apoptosis of CD4+ T cells. Fasudil inhibited the differentiation and effector cytokine secretion of Th17 cells, whereas it upregulated Treg cells in vitro. It also restrained the in vivo interleukin (IL)-4 and IL-17 levels in infected mice. Fasudil directly induced the apoptosis of hepatic stellate cells and downregulated the expressions of hepatic fibrogenic genes, such as collagen type I (Col-I), Col-III, and transforming growth factor-1 (TGF-β1). These effects may contribute to its anti-pathogenic roles in schistosomiasis. CONCLUSIONS Fasudil inhibits hepatic granuloma formation and fibrosis with downregulation of Th17 cells. Fasudil might serve as a novel therapeutic agent for hepatic fibrosis due to schistosome infections and perhaps other disorders.
Collapse
|
16
|
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: 16] [Impact Index Per Article: 2.7] [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
|
17
|
Omar HH. Impact of chronic schistosomiasis and HBV/HCV co-infection on the liver: current perspectives. Hepat Med 2019; 11:131-136. [PMID: 31565002 PMCID: PMC6731971 DOI: 10.2147/hmer.s155962] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/25/2019] [Indexed: 12/19/2022] Open
Abstract
Schistosomiasis is a public health problem in many countries. Its prevalence is increasing annually; the current infection rate is one in 30 individuals. The WHO reported that at least 206.4 million people all over the world required preventive treatments for schistosomiasis in 2016. Chronic schistosomiasis, hepatitis B virus (HBV) and hepatitis C virus (HCV) co-infection are common in countries where schistosomiasis is endemic. The effects of the hepatotropic virus co-infection may modify the Th2-dominated granulomatous phase of schistosomal infection. These viruses induce a strong-specific T cell response, with infiltration of large numbers of specific interferon-γ-producing CD8+ cells into the liver. The outcome of liver diseases depends on the underlying causes, host immune response and concomitant infections. Co-infection of schistosomiasis with HBV/HCV infection causes advanced liver disease and worsens the outcome, especially with higher viral load titers, which increase the mortality rate through an increased incidence of liver cirrhosis and hepatocellular carcinoma. The exposure risk for HBV in patients with HCV and schistosomiasis was two and half times greater than that in CHC patients without schistosomiasis. Finally, chronic schistosomiasis and HBV/HCV co-infection have serious effects on liver pathology. Co-infection accelerates the progression of liver disease and leads to advanced liver diseases and liver failure.
Collapse
Affiliation(s)
- Hanan Hassan Omar
- Clinical Pathology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
18
|
Radiation induces changes in toll-like receptors of the uterine cervix of the rat. PLoS One 2019; 14:e0215250. [PMID: 30998706 PMCID: PMC6472742 DOI: 10.1371/journal.pone.0215250] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/28/2019] [Indexed: 01/02/2023] Open
Abstract
Radiotherapy is an important therapeutic approach against cervical cancer but associated with adverse effects including vaginal fibrosis and dyspareunia. We here assessed the immunological and oxidative responses to cervical irradiation in an animal model for radiation-induced cervicitis. Rats were sedated and either exposed to 20 Gy of ionising radiation given by a linear accelerator or only sedated (controls) and euthanized 1–14 days later. The expressions of toll-like receptors (TLRs) and coupled intracellular pathways in the cervix were assessed with immunohistofluorescence and western blot. Expression of cytokines were analysed with the Bio-Plex Suspension Array System (Bio-Rad). We showed that TLRs 2–9 were expressed in the rat cervix and cervical irradiation induced up-regulation of TLR5, TRIF and NF-κB. In the irradiated cervical epithelium, TLR5 and TRIF were increased in concert with an up-regulation of oxidative stress (8-OHdG) and antioxidant enzymes (SOD-1 and catalase). G-CSF, M-CSF, IL-10, IL- 17A, IL-18 and RANTES expressions in the cervix decreased two weeks after cervical irradiation. In conclusion, the rat uterine cervix expresses the TLRs 2–9. Cervical irradiation induces immunological changes and oxidative stress, which could have importance in the development of adverse effects to radiotherapy.
Collapse
|
19
|
Kalantari P, Bunnell SC, Stadecker MJ. The C-type Lectin Receptor-Driven, Th17 Cell-Mediated Severe Pathology in Schistosomiasis: Not All Immune Responses to Helminth Parasites Are Th2 Dominated. Front Immunol 2019; 10:26. [PMID: 30761125 PMCID: PMC6363701 DOI: 10.3389/fimmu.2019.00026] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/08/2019] [Indexed: 01/09/2023] Open
Abstract
Schistosomiasis is a major helminthic disease in which damage to the affected organs is orchestrated by a pathogenic host CD4 T helper (Th) cell-mediated immune response against parasite eggs. In the case of the species Schistosoma mansoni, the resulting granulomatous inflammation and fibrosis takes place in the liver and intestines. The magnitude of disease varies greatly from individual to individual but in a minority of patients, there is severe disease and death. S. mansoni infection in a murine model similarly results in marked strain variation of immunopathology. In the most commonly examined mouse strain, C57BL/6 (BL/6), there is relatively mild hepatic pathology arising in a Th2-dominated cytokine environment. In contrast, CBA mice develop decisively more severe lesions largely driven by proinflammatory IL-17-producing Th17 cells. Dendritic cells (DCs) from CBA mice differ sharply with those from BL/6 mice in that they vastly over-express the C-type lectin receptor (CLR) CD209a (SIGNR5), a homolog of human DC-SIGN, which senses glycans such as those produced by schistosome eggs. Silencing of CD209a, and recent studies with CD209a KO CBA mice have shown that this receptor is crucial to induce the pathogenic Th17 cell response; indeed, CD209a KO mice display markedly reduced immunopathology akin to that seen in BL/6 mice. Mechanistically, CD209a synergizes with the related CLRs Dectin-2 and Mincle to stimulate increased DC production of IL-1β and IL-23, necessary for pathogenic Th17 cell development. These findings denote key molecular underpinnings of disease variability based on selection and function of contrasting Th cell subsets.
Collapse
Affiliation(s)
- Parisa Kalantari
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
| | - Stephen C Bunnell
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
| | - Miguel J Stadecker
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
20
|
Zhang H, Jiang Z, Zhang L. Dual effect of T helper cell 17 (Th17) and regulatory T cell (Treg) in liver pathological process: From occurrence to end stage of disease. Int Immunopharmacol 2019; 69:50-59. [PMID: 30669025 DOI: 10.1016/j.intimp.2019.01.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 01/02/2019] [Accepted: 01/04/2019] [Indexed: 02/06/2023]
Abstract
Liver disease is a complicated pathological status with acute or chronic progressions, causing a series of damages to liver and massive burden to public health and society. Th17 and Treg, two subsets of CD4+ T helper cells, seem to keep a subtle balance in the maintenance of organic immune homeostasis including liver. The dysfunction of Th17/Treg balance in liver has been proved associated with hepatic injury and disease. Herein, we summarized the research advance of Th17 and Treg cells in different phenotypes of liver diseases in the past decade. It is known to all that hepatic diseases start from stimulations or infections like virus, autoimmune, alcohol and so on in the early stage, which would cause inflammation. With the disease consistently existed, severe outcomes like cirrhosis and hepatocellular carcinoma appear finally. In conclusion, it is found that Th17 and Treg cells serve as an important role in the immune response imbalance of liver diseases from the beginning to the end stage. However, the effect of these two subsets of CD4+ T helper cells is not a stereotype. Pathological role which exacerbates the disease and protective character which inhibits damage to liver are co-existed in the effect of Th17 and Treg cells. Still, more studies should be carried out to enrich the understandings of liver disease and Th17/Treg immune balance in the future.
Collapse
Affiliation(s)
- Haoran Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China.
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Center for Drug Screening and Pharmacodynamics Evaluation, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
21
|
Wang Y, Wang BS, Hui X, Qiao J, Li WZ, Sun N. [Role of inducible costimulatory molecule-mediated Th17 cell polarization in renal fibrosis in spontaneously hypertensive rats]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:534-540. [PMID: 29891448 PMCID: PMC6743898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Indexed: 07/30/2024]
Abstract
OBJECTIVE To explore the role of inducible costimulatory molecule (ICOS) signaling pathway-mediated Th17 cells polarization in renal damage in essential hypertension. METHODS Four-week-old spontaneously hypertensive rats (SHR) were randomly divided into control (SHR-C) group and intervention (SHR-I) group and subjected to intraperitoneal injections of PBS and ICOS monoclonal antibody for 2 weeks, respectively. Blood pressure of the rats was monitored using noninvasive tail artery blood pressure measuring instrument. The percentage of Th17 cells in the splenocytes was analyzed using flow cytometry, and the expression levels of IL-17A mRNA in the rat's kidneys were detected using RT-PCR. The levels of IL-17A and TGF-β1 in the plasma and kidneys were dynamically detected using ELISA and immunohistochemistry, respectively. Renal pathological changes in the rats were detected using Masson staining. RESULTS At the age of 10 and 30 weeks, the rats in SHR-C group had a significantly higher blood pressure than those in SHR-I group (P<0.05 or 0.01). In rats in SHR-C group, Th17 cells percentage in the splenocytes and IL-17A mRNA level in the kidney was significantly higher than those in SHR-I group from the age of 6 weeks (P<0.05). The expressions of IL-17A and TGF-β1 in the plasma and kidney were significantly higher in SHR-C group than that in SHR-I group at 6 weeks (P<0.05). Compared with those in SHR-C group, the rats in SHR-I group showed significant alleviation of renal fibrosis from the age of 30 weeks (P<0.05). CONCLUSION The ICOS signaling pathway-mediated Th17 cells polarization plays an important role in renal fibrosis in hypertensive rats.
Collapse
Affiliation(s)
- Yu Wang
- Department of Basic Medical Sciences, Medical College, Anhui University of Science & Technology, Huainan 232001, China.E-mail:
| | | | | | | | | | | |
Collapse
|
22
|
Wang Y, Wang BS, Hui X, Qiao J, Li WZ, Sun N. [Role of inducible costimulatory molecule-mediated Th17 cell polarization in renal fibrosis in spontaneously hypertensive rats]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:534-540. [PMID: 29891448 PMCID: PMC6743898 DOI: 10.3969/j.issn.1673-4254.2018.05.05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Indexed: 06/08/2023]
Abstract
OBJECTIVE To explore the role of inducible costimulatory molecule (ICOS) signaling pathway-mediated Th17 cells polarization in renal damage in essential hypertension. METHODS Four-week-old spontaneously hypertensive rats (SHR) were randomly divided into control (SHR-C) group and intervention (SHR-I) group and subjected to intraperitoneal injections of PBS and ICOS monoclonal antibody for 2 weeks, respectively. Blood pressure of the rats was monitored using noninvasive tail artery blood pressure measuring instrument. The percentage of Th17 cells in the splenocytes was analyzed using flow cytometry, and the expression levels of IL-17A mRNA in the rat's kidneys were detected using RT-PCR. The levels of IL-17A and TGF-β1 in the plasma and kidneys were dynamically detected using ELISA and immunohistochemistry, respectively. Renal pathological changes in the rats were detected using Masson staining. RESULTS At the age of 10 and 30 weeks, the rats in SHR-C group had a significantly higher blood pressure than those in SHR-I group (P<0.05 or 0.01). In rats in SHR-C group, Th17 cells percentage in the splenocytes and IL-17A mRNA level in the kidney was significantly higher than those in SHR-I group from the age of 6 weeks (P<0.05). The expressions of IL-17A and TGF-β1 in the plasma and kidney were significantly higher in SHR-C group than that in SHR-I group at 6 weeks (P<0.05). Compared with those in SHR-C group, the rats in SHR-I group showed significant alleviation of renal fibrosis from the age of 30 weeks (P<0.05). CONCLUSION The ICOS signaling pathway-mediated Th17 cells polarization plays an important role in renal fibrosis in hypertensive rats.
Collapse
Affiliation(s)
- Yu Wang
- Department of Basic Medical Sciences, Medical College, Anhui University of Science & Technology, Huainan 232001, China.E-mail:
| | | | | | | | | | | |
Collapse
|
23
|
Mutengo MM, Mduluza T, Kelly P, Mwansa JCL, Kwenda G, Musonda P, Chipeta J. Low IL-6, IL-10, and TNF- α and High IL-13 Cytokine Levels Are Associated with Severe Hepatic Fibrosis in Schistosoma mansoni Chronically Exposed Individuals. J Parasitol Res 2018; 2018:9754060. [PMID: 29610679 PMCID: PMC5828471 DOI: 10.1155/2018/9754060] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/21/2017] [Indexed: 01/29/2023] Open
Abstract
Several studies have attributed the etiopathogenesis of chronic Schistosoma mansoni related hepatic fibrosis to unregulated immune responses against trapped parasite ova in the host. However, there is limited data on immune profiles associated with varying degrees of the disease in a population under chronic exposure to the parasite. We therefore investigated the role of selected T-helper (Th)1, Th2, and Th17 cytokines in relation to hepatic fibrosis severity among individuals resident in a hyper-Schistosoma mansoni endemic region of Western Zambia. Two hundred and forty-four S. mansoni infected individuals with and without fibrosis were analysed for cytokine profiles. Based on hepatic fibrosis stage as determined by ultrasound, participants were categorized into Group 0, Group I, Group II, and Group III. Cytokines were measured in S. mansoni egg stimulated whole blood culture supernatants using the BD Cytometric Bead Array kits. Compared to the nonfibrotic group, participants in the severe hepatic fibrotic group produced less interleukin- (IL-) 6, IL-10, and tumour necrosis factor-alpha (TNF-α). On the other hand, IL-13 was significantly elevated in this group compared to the nonfibrotic group (p < 0.001). Our results suggest that low IL-6, IL-10, and TNF-α and high IL-13 levels may influence S. mansoni disease progression.
Collapse
Affiliation(s)
- Mable M. Mutengo
- Department of Pathology and Microbiology, University Teaching Hospital, Lusaka, Zambia
- University of Zambia, Lusaka, Zambia
| | - Takafira Mduluza
- Department of Biochemistry, University of Zimbabwe, Mount Pleasant, Harare, Zimbabwe
| | - Paul Kelly
- Department of Internal Medicine, School of Medicine, University of Zambia, Lusaka, Zambia
| | - James C. L. Mwansa
- Department of Pathology and Microbiology, University Teaching Hospital, Lusaka, Zambia
- University of Zambia, Lusaka, Zambia
| | - Geoffrey Kwenda
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka, Zambia
| | - Patrick Musonda
- Department of Epidemiology and Biostatistics, School of Public Health, University of Zambia, Lusaka, Zambia
| | - James Chipeta
- Department of Pediatrics and Child Health, School of Medicine, University of Zambia, Lusaka, Zambia
| |
Collapse
|
24
|
Wang Y, Lin C, Cao Y, Duan Z, Guan Z, Xu J, Zhu XQ, Xia C. Up-regulation of Interleukin-21 Contributes to Liver Pathology of Schistosomiasis by Driving GC Immune Responses and Activating HSCs in Mice. Sci Rep 2017; 7:16682. [PMID: 29192177 PMCID: PMC5709429 DOI: 10.1038/s41598-017-16783-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 11/17/2017] [Indexed: 01/07/2023] Open
Abstract
The pathology of schistosome egg-induced liver granuloma, fibrosis and eventually liver scarring is complicated. CD4+ helper T (Th) cells play critical roles in both host humoral immunity and cellular immunity against parasitic infection and immunopathology in schistosomiasis. Follicular helper T (Tfh) cells are another specialized subset of Th cells and involved in infectious diseases. However, the immune regulatory mechanism of Tfh cells in severe liver pathology of schistosomiasis is still poorly understood. In this study, using a S. japonicum-infected mouse model, we studied the dynamics and effects of Tfh cells in vivo and demonstrated that Tfh phenotype molecules ICOS, PD-1 and functional factor IL-21 were positively correlated with disease development by flow cytometry. Meanwhile, our results also showed that Tfh cells enriched in splenic germinal center (GC) and promoted B cells producing IgM with the progress of hepatic immunopathology by B-T co-culture experiments. More importantly, our data indicated that IL-21 contributed to the formation and development of hepatic egg granuloma and subsequent fibrosis by driving GC responses and activating HSCs by immunohistochemical detection and blocking assay in vitro. Our findings contribute to the better understanding of the immunopathogenesis of schistosomiasis and have implications for therapeutic intervention of hepatic fibrotic diseases.
Collapse
Affiliation(s)
- Yanyan Wang
- Department of Parasitology, Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Cai Lin
- Department of Parasitology, Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Yun Cao
- Department of Parasitology, Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhongliang Duan
- Department of Parasitology, Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhixun Guan
- Department of Parasitology, Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Jing Xu
- Department of Parasitology, Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, China
| | - Chaoming Xia
- Department of Parasitology, Medical College of Soochow University, Suzhou, Jiangsu Province, China.
| |
Collapse
|
25
|
Zhang C, Shao Y, Yang S, Bi X, Li L, Wang H, Yang N, Li Z, Sun C, Li L, Lü G, Aji T, Vuitton DA, Lin R, Wen H. T-cell tolerance and exhaustion in the clearance of Echinococcus multilocularis: role of inoculum size in a quantitative hepatic experimental model. Sci Rep 2017; 7:11153. [PMID: 28894272 PMCID: PMC5593833 DOI: 10.1038/s41598-017-11703-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/29/2017] [Indexed: 12/31/2022] Open
Abstract
The local immune mechanisms responsible for either self-healing or sustained chronic infection are not clear, in the development of E. multilocularis larvae. Here, we developed a suitable experimental model that mimics naturally infected livers, according to the parasite load. We demonstrated that local cellular immunity and fibrogenesis are actually protective and fully able to limit metacestode growth in the liver of low or medium dose-infected mice (LDG or MDG), or even to clear it, while impairment of cellular immunity is followed by a more rapid and severe course of the disease in high dose-infected mice (HDG). And recruitment and/ or proliferation of memory T cells (including CD4 Tem, CD8 Tcm and CD8 Tem) and imbalance of T1/T2/T17/Treg-type T cells in liver were not only associated with clearance of the parasite infection in LDG, but also with increased hepatic injury in HDG; in particular the dual role of CD8 T cells depending on the parasite load and the various stages of metacestode growth. Besides, we first demonstrate the association between LAG3- or 2B4-expressing T cells exhaustion and HD inocula in late stages. Our quantitative experimental model appears fully appropriate to study immunomodulation as a therapeutic strategy for patients with Alveolar Echinococcosis.
Collapse
Affiliation(s)
- Chuanshan Zhang
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, and WHO Collaborating Centre on Prevention and Case Management of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yingmei Shao
- Department of Hepatic Hydatid and Hepatobiliary Surgery, Digestive and Vascular Surgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Shuting Yang
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, and WHO Collaborating Centre on Prevention and Case Management of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xiaojuan Bi
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, and WHO Collaborating Centre on Prevention and Case Management of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Liang Li
- Institute of Immunology, The Key Laboratory of Innate Immunity and Chronic Disease (Chinese Academy of Medical Science), School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, China
| | - Hui Wang
- Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Ning Yang
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, and WHO Collaborating Centre on Prevention and Case Management of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Zhide Li
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, and WHO Collaborating Centre on Prevention and Case Management of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Cheng Sun
- Institute of Immunology, The Key Laboratory of Innate Immunity and Chronic Disease (Chinese Academy of Medical Science), School of Life Sciences and Medical Center, University of Science & Technology of China, Hefei, Anhui, China
| | - Liang Li
- Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Guodong Lü
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, and WHO Collaborating Centre on Prevention and Case Management of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Tuerganaili Aji
- Department of Hepatic Hydatid and Hepatobiliary Surgery, Digestive and Vascular Surgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Dominique A Vuitton
- WHO-Collaborating Centre for the Prevention and Treatment of Human Echinococcosis, Department of Parasitology, University Bourgogne Franche-Comté (EA 3181) and University Hospital, Besançon, France
| | - Renyong Lin
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, and WHO Collaborating Centre on Prevention and Case Management of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China.
| | - Hao Wen
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, and WHO Collaborating Centre on Prevention and Case Management of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China. .,Department of Hepatic Hydatid and Hepatobiliary Surgery, Digestive and Vascular Surgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China.
| |
Collapse
|
26
|
Genetic Adjuvantation of a Cell-Based Therapeutic Vaccine for Amelioration of Chagasic Cardiomyopathy. Infect Immun 2017; 85:IAI.00127-17. [PMID: 28674032 DOI: 10.1128/iai.00127-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 06/21/2017] [Indexed: 12/19/2022] Open
Abstract
Chagas disease, caused by infection with the protozoan parasite Trypanosoma cruzi, is a leading cause of heart disease ("chagasic cardiomyopathy") in Latin America, disproportionately affecting people in resource-poor areas. The efficacy of currently approved pharmaceutical treatments is limited mainly to acute infection, and there are no effective treatments for the chronic phase of the disease. Preclinical models of Chagas disease have demonstrated that antigen-specific CD8+ gamma interferon (IFN-γ)-positive T-cell responses are essential for reducing parasite burdens, increasing survival, and decreasing cardiac pathology in both the acute and chronic phases of Chagas disease. In the present study, we developed a genetically adjuvanted, dendritic cell-based immunotherapeutic for acute Chagas disease in an attempt to delay or prevent the cardiac complications that eventually result from chronic T. cruzi infection. Dendritic cells transduced with the adjuvant, an adenoviral vector encoding a dominant negative isoform of Src homology region 2 domain-containing tyrosine phosphatase 1 (SHP-1) along with the T. cruzi Tc24 antigen and trans-sialidase antigen 1 (TSA1), induced significant numbers of antigen-specific CD8+ IFN-γ-positive cells following injection into BALB/c mice. A vaccine platform transduced with the adenoviral vector and loaded in tandem with the recombinant protein reduced parasite burdens by 76% to >99% in comparison to a variety of different controls and significantly reduced cardiac pathology in a BALB/c mouse model of live Chagas disease. Although no statistical differences in overall survival rates among cohorts were observed, the data suggest that immunotherapeutic strategies for the treatment of acute Chagas disease are feasible and that this approach may warrant further study.
Collapse
|
27
|
NLRP3 inflammasome activation results in liver inflammation and fibrosis in mice infected with Schistosoma japonicum in a Syk-dependent manner. Sci Rep 2017; 7:8120. [PMID: 28808303 PMCID: PMC5556086 DOI: 10.1038/s41598-017-08689-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 07/06/2017] [Indexed: 02/06/2023] Open
Abstract
Granulomatous and fibrosing inflammation in response to soluble egg antigen (SEA) from Schistosoma japonicum (S. japonicum) is the main pathological process of S. japonicum infection. Inflammasome activation has recently been implicated in the pathogenesis of liver disease. However, the role of inflammasome activation in schistosomiasis-associated liver fibrosis (SSLF) has not been extensively studied. In this study, it is demonstrated that the NLRP3 inflammasome is markedly activated in mouse HSCs both in vivo and in vitro during S. japonicum infection. Furthermore, it is demonstrated that inhibition of NLRP3 inflammasome significantly alleviates the liver inflammation and collagen deposition that are induced by infection with S. japonicum. The mechanism of SEA-induced NLRP3 inflammasome activation is studied in isolated, cultured mouse HSCs and it is shown that SEA-induced NLRP3 inflammasome activation in HSCs is dependent upon the activities of spleen tyrosine kinase (Syk), an enzyme usually associated with a pathogen recognition receptor for fungal pathogens. Moreover, it is demonstrated that Dectin-1 and JNK signaling are also involved in SEA-induced NLRP3 inflammasome activation in HSCs. These data shed new light on the mechanisms of NLRP3 inflammasome activation during an infection with S. japonicum, and further characterize its role in schistosomiasis-associated liver fibrosis (SSLF).
Collapse
|