1
|
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
|
2
|
Singh I, Hoti SL, Chauhan N, Joshi RK, Prasad TSK, Sarikhani M, Kaushik M, Unger BS, Jadhav P, Modi PK. Immunomodulation of streptozotocin induced Type 1 diabetes mellitus in mouse model by Macrophage migration inhibitory factor-2 (MIF-2) homologue of human lymphatic filarial parasite, Wuchereria bancrofti. Acta Trop 2024; 252:107142. [PMID: 38331083 DOI: 10.1016/j.actatropica.2024.107142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 02/05/2024] [Accepted: 02/05/2024] [Indexed: 02/10/2024]
Abstract
Helminth parasites modulate the host immune system to ensure a long-lasting asymptomatic form of infection generally, mediated by the secretion of immunomodulatory molecules and one such molecule is a homologue of human host cytokine, Macrophage migratory Inhibitory Factor (hMIF). In this study, we sought to understand the role of homologue of hMIF from the lymphatic filarial parasite, Wuchereria bancrofti (Wba-MIF2), in the immunomodulation of the Streptozotocin (STZ)-induced Type1 Diabetes Mellitus (T1DM) animal model. Full-length recombinant Wba-MIF2 was expressed and found to have both oxidoreductase and tautomerase activities. Wba-MIF2 recombinant protein was treated to STZ induced T1DM animals, and after 5 weeks pro-inflammatory (IL-1, IL-2, IL-6, TNF-α, IFN-γ) and anti-inflammatory (IL-4, IL-10) cytokines and gene expressions were determined in sera samples and spleen respectively. Pro-inflammatory and anti-inflammatory cytokine levels were significantly (p<0.05) up-regulated and down-regulated respectively, in the STZ-T1DM animals, as compared to treated groups. Histopathology showed macrophage infiltration and greater damage of islets of beta cells in the pancreatic tissue of STZ-T1DM animals, than Wba-MIF2 treated STZ-T1DM animals. The present study clearly showed the potential of Wba-MIF2 as an immunomodulatory molecule, which could modulate the host immune system in the STZ-T1DM mice model from a pro-inflammatory to anti-inflammatory milieu.
Collapse
Affiliation(s)
- Ishwar Singh
- ICMR-National Institute of Traditional Medicine, Belagavi 590010, India Karnataka, India; KLE Academy of Higher Education and Research, Belagavi 590010, India Karnataka, India
| | - S L Hoti
- ICMR-National Institute of Traditional Medicine, Belagavi 590010, India Karnataka, India.
| | - Nikhil Chauhan
- ICMR-National Institute of Traditional Medicine, Belagavi 590010, India Karnataka, India
| | - R K Joshi
- ICMR-National Institute of Traditional Medicine, Belagavi 590010, India Karnataka, India
| | - T S Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575007, Karnataka, India
| | | | - Meenakshi Kaushik
- ICMR-National Institute of Traditional Medicine, Belagavi 590010, India Karnataka, India
| | - Banappa S Unger
- ICMR-National Institute of Traditional Medicine, Belagavi 590010, India Karnataka, India
| | - Pankaj Jadhav
- Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Prashant Kumar Modi
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575007, Karnataka, India
| |
Collapse
|
3
|
Rosa AAD, Brandão-Bezerra L, Corrêa CL, Amaral G Da-Silva S, Rodrigues LS, Machado-Silva JR, Neves RH. Changes in splenic tissue and immune response profile of Schistosoma mansoni infected mice submitted to chronic ethanol intake. Exp Parasitol 2024; 259:108706. [PMID: 38309327 DOI: 10.1016/j.exppara.2024.108706] [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: 08/02/2023] [Revised: 12/08/2023] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
In Schistosoma mansoni infection, the spleen is one of the organs affected, causing its enlargement (splenomegaly). Intake of ethanol through alcoholic beverages can cause spleen atrophy and interfere with immune activity. To gain knowledge of this association on the spleen and on the immune response profile, male mice were used as an experimental model. These animals were divided into four groups: C. control; EC. uninfected/ethanol gavage; I. infected; and IE. infected/ethanol gavage. Groups I and IE were infected with about 100 cercariae (BH strain) of S. mansoni and in the fifth week of infection, gavage 200 μL/day/animal of 18 % ethanol was started for 28 consecutive days. At the end of the gavage (9th week of infection) all animals were euthanized. The spleen was removed and longitudinally divided in two parts. After histological processing, the sections were stained with H&E and Gomori's Reticulin for histopathological and stereological analyses, white pulp morphometry and quantification of megakaryocytes. The other fragment was macerated (in laminar flow) and the cell suspension, after adjusting the concentration (2 × 106), was plated to obtain cytokines produced by spleen cells that were measured by flow cytometry (Citometric Bead Array). Histopathological and quantitative analyzes in the spleen of the IE group showed an increase in the number of trabeculae and megakaryocytes, a decrease in reticular fibers, as well as important organizational changes in the white pulp and red pulp. Due to the decrease in the levels of cytokines measured and the result of the calculation of the ratio between the IFN-y and IL-10 cytokines (p = 0.0079) of the infected groups, we suggest that ethanol decreased the inflammatory and anti-inflammatory response generated by the infection (group IE, the production of cytokines was significantly decreased (p < 0.01). These changes demonstrate that ethanol ingestion interferes with some parameters of experimental S. mansoni infection, such as changes in splenic tissue and in the pattern of cytokine production.
Collapse
Affiliation(s)
- Aline Aparecida da Rosa
- Romero Lascasas Porto Laboratory of Helminthology, Department of Microbiology, Immunology and Parasitology, Faculty of Medical Sciences, Rio de Janeiro State University, Brazil
| | - Luciana Brandão-Bezerra
- Romero Lascasas Porto Laboratory of Helminthology, Department of Microbiology, Immunology and Parasitology, Faculty of Medical Sciences, Rio de Janeiro State University, Brazil
| | - Christiane Leal Corrêa
- Department of Pathology and Laboratories, School of Medical Sciences, Rio de Janeiro State University, Brazil; Medicine School, Estácio de Sá University, Brazil
| | - Silvia Amaral G Da-Silva
- Laboratory of Parasitic Immunopharmacology, Department of Microbiology, Immunology and Parasitology, Faculty of Medical Sciences, Rio de Janeiro State University, Brazil
| | - Luciana Silva Rodrigues
- Laboratory of Immunopathology, Department of Pathology and Laboratories, Faculty of Medical Sciences, Rio de Janeiro State University, Brazil
| | - José Roberto Machado-Silva
- Romero Lascasas Porto Laboratory of Helminthology, Department of Microbiology, Immunology and Parasitology, Faculty of Medical Sciences, Rio de Janeiro State University, Brazil
| | - Renata Heisler Neves
- Romero Lascasas Porto Laboratory of Helminthology, Department of Microbiology, Immunology and Parasitology, Faculty of Medical Sciences, Rio de Janeiro State University, Brazil.
| |
Collapse
|
4
|
Perera DJ, Koger-Pease C, Paulini K, Daoudi M, Ndao M. Beyond schistosomiasis: unraveling co-infections and altered immunity. Clin Microbiol Rev 2024; 37:e0009823. [PMID: 38319102 PMCID: PMC10938899 DOI: 10.1128/cmr.00098-23] [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] [Indexed: 02/07/2024] Open
Abstract
Schistosomiasis is a neglected tropical disease caused by the helminth Schistosoma spp. and has the second highest global impact of all parasites. Schistosoma are transmitted through contact with contaminated fresh water predominantly in Africa, Asia, the Middle East, and South America. Due to the widespread prevalence of Schistosoma, co-infection with other infectious agents is common but often poorly described. Herein, we review recent literature describing the impact of Schistosoma co-infection between species and Schistosoma co-infection with blood-borne protozoa, soil-transmitted helminths, various intestinal protozoa, Mycobacterium, Salmonella, various urinary tract infection-causing agents, and viral pathogens. In each case, disease severity and, of particular interest, the immune landscape, are altered as a consequence of co-infection. Understanding the impact of schistosomiasis co-infections will be important when considering treatment strategies and vaccine development moving forward.
Collapse
Affiliation(s)
- Dilhan J. Perera
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Cal Koger-Pease
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Kayla Paulini
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
| | - Mohamed Daoudi
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
| | - Momar Ndao
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
- National Reference Centre for Parasitology, Research Institute of the McGill University Health Centre, Montreal, Canada
| |
Collapse
|
5
|
Thorstenberg ML, Martins MDA, Oliveira NF, Monteiro MMLV, Santos GRC, Pereira HMG, Savio LEB, Coutinho-Silva R, Silva CLM. Altered purinergic P2X7 and A 2B receptors signaling limits macrophage-mediated host defense in schistosomiasis. Biomed J 2024; 47:100713. [PMID: 38442854 PMCID: PMC11550761 DOI: 10.1016/j.bj.2024.100713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 01/05/2024] [Accepted: 02/25/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND The occurrence of co-infections during schistosomiasis, a neglected tropical disease, with other parasites have been reported suggesting an impaired host immune defense. Macrophage purinergic P2X7 receptor (P2X7R) plays an important role against intracellular pathogens. Therefore, we investigated the P2X7R-mediated phagocytosis and killing capacity of Leishmania amazonensis by macrophages during schistosomiasis in vitro and in vivo. METHODS Swiss and C57BL/6 (Wild type) and P2X7R-/- were randomized in two groups: control (uninfected) and Schistosoma mansoni-infected. Alternatively, control Swiss and S. mansoni-infected mice were also infected with L. amazonensis. RESULTS The pre-treatment of control macrophages with the P2X7R antagonist (A74003) or TGF-β reduced the phagocytosis index, mimicking the phenotype of cells from S. mansoni-infected mice and P2X7R-/- mice. Apyrase also reduced the phagocytosis index in the control group corroborating the role of ATP to macrophage activation. Moreover, l-arginine-nitric oxide pathway was compromised during schistosomiasis, which could explain the reduced killing capacity in response to ATP in vitro and in vivo. We found an increased extracellular nucleotide (ATP, ADP and AMP) hydrolysis along with an increased frequency of F4/80+ CD39+ macrophages from the S. mansoni-infected group. Moreover, the content of adenosine in the cell supernatant was higher in the S. mansoni-infected group in relation to controls. Schistosomiasis also increased the expression of macrophage adenosine A2BR. In good accordance, both ADA and the selective A2BR antagonist restored the phagocytosis index of macrophages from S. mansoni-infected group. CONCLUSIONS Altogether, the altered P2X7R and A2BR signaling limits the role of macrophages to host defense against L. amazonensis during schistosomiasis, potentially contributing to the pathophysiology and clinically relevant co-infections.
Collapse
Affiliation(s)
- Maria Luiza Thorstenberg
- Laboratory of Biochemical and Molecular Pharmacology, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Monique Daiane Andrade Martins
- Laboratory of Immunophysiology, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil
| | - Nathália Ferreira Oliveira
- Laboratory of Biochemical and Molecular Pharmacology, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Matheus Macedo L V Monteiro
- Laboratory of Biochemical and Molecular Pharmacology, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil
| | - Gustavo R C Santos
- Brazilian Doping Control Laboratory (LBCD - LADETEC / IQ), Universidade Federal do Rio de Janeiro, Brazil
| | | | - Luiz Eduardo Baggio Savio
- Laboratory of Immunophysiology, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil
| | - Robson Coutinho-Silva
- Laboratory of Immunophysiology, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil
| | - Claudia Lucia Martins Silva
- Laboratory of Biochemical and Molecular Pharmacology, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Brazil.
| |
Collapse
|
6
|
Gomides TAR, de Souza MLM, de Figueiredo AB, Lima MR, Silveira AMS, de Assis GFM, Fraga LAO, Silveira-Nunes G, Martucci L, Garcia JD, Afonso LCC, Teixeira-Carvalho A, Leite PM. Expression of SmATPDases 1 and 2 in Schistosoma mansoni eggs favours IL-10 production in infected individuals. Parasite Immunol 2024; 46:e13017. [PMID: 37922505 DOI: 10.1111/pim.13017] [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: 04/20/2023] [Revised: 08/28/2023] [Accepted: 10/09/2023] [Indexed: 11/05/2023]
Abstract
A role of IL-10 is down-regulating T-cell responses to schistosome antigens. Since SmATPDases can be correlated to modulation of the immune response, we evaluated the expression of enzymes in S. mansoni eggs. Faecal samples were collected from 40 infected individuals to detect coding regions of the SmATPDases. The cytokines were measured in supernatants of PBMC. The analysis was performed by the global median determination and set up high producers (HP) of cytokines. Six individuals expressed SmATPDase1, six expressed SmATPDase2 and six expressed both enzymes. The group who expressed only SmATPDase1 showed a high frequency of IFN-γ, TNF IL-4 HP; individuals who expressed only SmATPDase2 showed a high frequency of IFN-γ, IL-6 and IL-4 HP; and individuals who expressed both enzymes showed a high frequency of IL-10 HP. The comparison of the IFN-γ/IL-10 ratio presented higher indices in the group who had SmATPDase 2 expression than those who had the expression of both enzymes. The positive correlation between infection intensity and IL-10 levels remained only in the positive SmATPDase group. The IL-10 is the only cytokine induced by the expression of both enzymes. Our data suggest that the expression of both enzymes seems to be a factor that modulates the host immune response by inducing high IL-10 production.
Collapse
Affiliation(s)
- Thalisson Artur Ribeiro Gomides
- Laboratório de Imunoparasitologia, Departamento de Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
- Laboratório de Imunologia da Universidade Vale do Rio Doce, Govenador Valadares, Brazil
| | | | - Amanda Braga de Figueiredo
- Laboratório de Imunoparasitologia, Departamento de Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | | | - Alda Maria Soares Silveira
- Universidade Federal de Juiz de Fora - Campus Avançado de Governador Valadares, Governador Valadares, Brazil
| | | | - Lúcia Alves Oliveira Fraga
- Universidade Federal de Juiz de Fora - Campus Avançado de Governador Valadares, Governador Valadares, Brazil
| | - Gabriela Silveira-Nunes
- Universidade Federal de Juiz de Fora - Campus Avançado de Governador Valadares, Governador Valadares, Brazil
| | - Letícia Martucci
- Universidade Federal de Juiz de Fora - Campus Avançado de Governador Valadares, Governador Valadares, Brazil
| | - Jennifer Delgado Garcia
- Universidade Federal de Juiz de Fora - Campus Avançado de Governador Valadares, Governador Valadares, Brazil
| | - Luís Carlos Crocco Afonso
- Laboratório de Imunoparasitologia, Departamento de Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Andréa Teixeira-Carvalho
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, FIOCRUZ, Belo Horizonte, Brazil
| | - Pauline Martins Leite
- Universidade Federal de Juiz de Fora - Campus Avançado de Governador Valadares, Governador Valadares, Brazil
| |
Collapse
|
7
|
Härle L, von Bülow V, Knedla L, Stettler F, Müller H, Zahner D, Haeberlein S, Windhorst A, Tschuschner A, Burg-Roderfeld M, Köhler K, Grevelding CG, Roeb E, Roderfeld M. Hepatocyte integrity depends on c-Jun-controlled proliferation in Schistosoma mansoni infected mice. Sci Rep 2023; 13:20390. [PMID: 37990129 PMCID: PMC10663609 DOI: 10.1038/s41598-023-47646-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 11/16/2023] [Indexed: 11/23/2023] Open
Abstract
Schistosomiasis is a parasitic disease affecting more than 250 million people worldwide. The transcription factor c-Jun, which is induced in S. mansoni infection-associated liver disease, can promote hepatocyte survival but can also trigger hepatocellular carcinogenesis. We aimed to analyze the hepatic role of c-Jun following S. mansoni infection. We adopted a hepatocyte-specific c-Jun knockout mouse model (Alb-Cre/c-Jun loxP) and analyzed liver tissue and serum samples by quantitative real-time PCR array, western blotting, immunohistochemistry, hydroxyproline quantification, and functional analyses. Hepatocyte-specific c-Jun knockout (c-JunΔli) was confirmed by immunohistochemistry and western blotting. Infection with S. mansoni induced elevated aminotransferase-serum levels in c-JunΔli mice. Of note, hepatic Cyclin D1 expression was induced in infected c-Junf/f control mice but to a lower extent in c-JunΔli mice. S. mansoni soluble egg antigen-induced proliferation in a human hepatoma cell line was diminished by inhibition of c-Jun signaling. Markers for apoptosis, oxidative stress, ER stress, inflammation, autophagy, DNA-damage, and fibrosis were not altered in S. mansoni infected c-JunΔli mice compared to infected c-Junf/f controls. Enhanced liver damage in c-JunΔli mice suggested a protective role of c-Jun. A reduced Cyclin D1 expression and reduced hepatic regeneration could be the reason. In addition, it seems likely that the trends in pathological changes in c-JunΔli mice cumulatively led to a loss of the protective potential being responsible for the increased hepatocyte damage and loss of regenerative ability.
Collapse
Affiliation(s)
- Lukas Härle
- Department of Gastroenterology, Justus Liebig University Giessen, Gaffkystr. 11c, 35392, Giessen, Germany
| | - Verena von Bülow
- Department of Gastroenterology, Justus Liebig University Giessen, Gaffkystr. 11c, 35392, Giessen, Germany
| | - Lukas Knedla
- Department of Gastroenterology, Justus Liebig University Giessen, Gaffkystr. 11c, 35392, Giessen, Germany
| | - Frederik Stettler
- Department of Gastroenterology, Justus Liebig University Giessen, Gaffkystr. 11c, 35392, Giessen, Germany
| | - Heike Müller
- Department of Gastroenterology, Justus Liebig University Giessen, Gaffkystr. 11c, 35392, Giessen, Germany
| | - Daniel Zahner
- Central Laboratory Animal Facility, Justus Liebig University Giessen, 35392, Giessen, Germany
| | - Simone Haeberlein
- Institute of Parasitology, BFS, Justus Liebig University Giessen, 35392, Giessen, Germany
| | - Anita Windhorst
- Institute of Medical Informatics, Justus Liebig University Giessen, 35392, Giessen, Germany
| | - Annette Tschuschner
- Department of Gastroenterology, Justus Liebig University Giessen, Gaffkystr. 11c, 35392, Giessen, Germany
| | | | - Kernt Köhler
- Institute of Veterinary Pathology, Justus Liebig University Giessen, Giessen, Germany
| | - Christoph G Grevelding
- Institute of Parasitology, BFS, Justus Liebig University Giessen, 35392, Giessen, Germany
| | - Elke Roeb
- Department of Gastroenterology, Justus Liebig University Giessen, Gaffkystr. 11c, 35392, Giessen, Germany
| | - Martin Roderfeld
- Department of Gastroenterology, Justus Liebig University Giessen, Gaffkystr. 11c, 35392, Giessen, Germany.
| |
Collapse
|
8
|
Ni Y, Xiong R, Zhu Y, Luan N, Yu C, Yang K, Wang H, Xu X, Yang Y, Sun S, Shi L, Padde JR, Chen L, Chen L, Hou M, Xu Z, Lai R, Ji M. A target-based discovery from a parasitic helminth as a novel therapeutic approach for autoimmune diseases. EBioMedicine 2023; 95:104751. [PMID: 37579625 PMCID: PMC10448429 DOI: 10.1016/j.ebiom.2023.104751] [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: 03/29/2023] [Revised: 07/25/2023] [Accepted: 07/25/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Regulatory T cells (Tregs) can alleviate the development of autoimmune and inflammatory diseases, thereby proposing their role as a new therapeutic strategy. Parasitic helminths have co-evolved with hosts to generate immunological privilege and immune tolerance through inducing Tregs. Thus, constructing a "Tregs-induction"-based discovery pipeline from parasitic helminth is a promising strategy to control autoimmune and inflammatory diseases. METHODS The gel filtration chromatography and reverse-phase high-performance liquid chromatography (RP-HPLC) were used to isolate immunomodulatory components from the egg extracts of Schistosoma japonicum. The extracted peptides were evaluated for their effects on Tregs suppressive functions using flow cytometry, ELISA and T cell suppression assay. Finally, we carried out colitis and psoriasis models to evaluate the function of Tregs induced by helminth-derived peptide in vivo. FINDINGS Here, based on target-driven discovery strategy, we successfully identified a small 3 kDa peptide (SjDX5-53) from egg extracts of schistosome, which promoted both human and murine Tregs production. SjDX5-53 presented immunosuppressive function by arresting dendritic cells (DCs) at an immature state and augmenting the proportion and suppressive capacity of Tregs. In mouse models, SjDX5-53 protected mice against autoimmune-related colitis and psoriasis through inducing Tregs and inhibiting inflammatory T-helper (Th) 1 and Th17 responses. INTERPRETATION SjDX5-53 exhibited the promising therapeutic effects in alleviating the phenotype of immune-related colitis and psoriasis. This study displayed a screening and validation pipeline of the inducer of Tregs from helminth eggs, highlighting the discovery of new biologics inspired by co-evolution of hosts and their parasites. FUNDING This study was supported by the Natural Science Foundation of China (82272368) and Natural Science Foundation of Jiangsu Province (BK20211586).
Collapse
Affiliation(s)
- Yangyue Ni
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Ruiyan Xiong
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yuxiao Zhu
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Ning Luan
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, China
| | - Chuanxin Yu
- Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Kun Yang
- Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Huiquan Wang
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Xuejun Xu
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yuxuan Yang
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Siyu Sun
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Liyun Shi
- Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, China
| | - Jon Rob Padde
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Lin Chen
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Lu Chen
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Min Hou
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Zhipeng Xu
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, China.
| | - Minjun Ji
- Department of Pathogen Biology, National Vaccine Innovation Platform, Jiangsu Province Engineering Research Center of Antibody Drug, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
9
|
Fonti N, Parisi F, Mancianti F, Freer G, Poli A. Cancerogenic parasites in veterinary medicine: a narrative literature review. Infect Agent Cancer 2023; 18:45. [PMID: 37496079 PMCID: PMC10373346 DOI: 10.1186/s13027-023-00522-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/17/2023] [Indexed: 07/28/2023] Open
Abstract
Parasite infection is one of the many environmental factors that can significantly contribute to carcinogenesis and is already known to be associated with a variety of malignancies in both human and veterinary medicine. However, the actual number of cancerogenic parasites and their relationship to tumor development is far from being fully understood, especially in veterinary medicine. Thus, the aim of this review is to investigate parasite-related cancers in domestic and wild animals and their burden in veterinary oncology. Spontaneous neoplasia with ascertained or putative parasite etiology in domestic and wild animals will be reviewed, and the multifarious mechanisms of protozoan and metazoan cancer induction will be discussed.
Collapse
Affiliation(s)
- Niccolò Fonti
- Dipartimento di Scienze veterinarie, Università di Pisa, Viale delle Piagge, 2, 56124, Pisa, Italy.
| | - Francesca Parisi
- Dipartimento di Scienze veterinarie, Università di Pisa, Viale delle Piagge, 2, 56124, Pisa, Italy
| | - Francesca Mancianti
- Dipartimento di Scienze veterinarie, Università di Pisa, Viale delle Piagge, 2, 56124, Pisa, Italy
| | - Giulia Freer
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, Università di Pisa, Via Savi, 10, 56126, Pisa, Italy
| | - Alessandro Poli
- Dipartimento di Scienze veterinarie, Università di Pisa, Viale delle Piagge, 2, 56124, Pisa, Italy
| |
Collapse
|
10
|
Smith CJ, Ashford JW. Apolipoprotein ɛ4-Associated Protection Against Pediatric Enteric Infections is a Survival Advantage in Pre-Industrial Populations. J Alzheimers Dis 2023:JAD221218. [PMID: 37125551 DOI: 10.3233/jad-221218] [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: 05/02/2023]
Abstract
Until 300,000 years ago, ancestors of modern humans ubiquitously carried the apolipoprotein E (APOE) ɛ4/ɛ4 genotype, when the ɛ3 allele mutated from the ancestral ɛ4, which elevates the risk of Alzheimer's disease. Modern humans living today predominantly carry the ɛ3 allele, which provides protection against heart disease and dementia in long-lived populations. The ancestral ɛ4 allele has been highly preserved in isolated populations in tropical and Arctic regions with high pathogen burdens, e.g., helminths. Early humans experienced serious enteric infections that exerted evolutionary selection pressure, and factors that mitigate infant and childhood mortality from enteric infections also exert selection pressure. Some bacteria can exploit the host's defensive inflammatory response to colonize and invade the host. Pathogen-induced inflammation associated with infant and childhood diarrhea can damage the gut wall long after the invading organisms are no longer present. Inflammation not only resides in the mucosal wall, but also induces systemic inflammation. Baseline systemic inflammation is lower in ɛ4 carriers, yet ɛ4 carriers display a stronger host inflammatory response that reduces pathogen burdens, increasing infant and early childhood survival. Evolutionary selection of the ɛ3 allele likely occurred after humans moved into temperate zones with lower pathogen burdens, unrelated to protection from Alzheimer's disease.
Collapse
Affiliation(s)
| | - J Wesson Ashford
- Stanford University and VA Palo Alto Health Care System, Palo Alto, CA, USA
| |
Collapse
|
11
|
Licá ICL, Frazão GCCG, Nogueira RA, Lira MGS, dos Santos VAF, Rodrigues JGM, Miranda GS, Carvalho RC, Silva LA, Guerra RNM, Nascimento FRF. Immunological mechanisms involved in macrophage activation and polarization in schistosomiasis. Parasitology 2023; 150:401-415. [PMID: 36601859 PMCID: PMC10089811 DOI: 10.1017/s0031182023000021] [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: 05/18/2022] [Revised: 12/21/2022] [Accepted: 12/24/2022] [Indexed: 01/06/2023]
Abstract
Human schistosomiasis is caused by helminths of the genus Schistosoma. Macrophages play a crucial role in the immune regulation of this disease. These cells acquire different phenotypes depending on the type of stimulus they receive. M1 macrophages can be ‘classically activated’ and can display a proinflammatory phenotype. M2 or ‘alternatively activated’ macrophages are considered anti-inflammatory cells. Despite the relevance of macrophages in controlling infections, the role of the functional types of these cells in schistosomiasis is unclear. This review highlights different molecules and/or macrophage activation and polarization pathways during Schistosoma mansoni and Schistosoma japonicum infection. This review is based on original and review articles obtained through searches in major databases, including Scopus, Google Scholar, ACS, PubMed, Wiley, Scielo, Web of Science, LILACS and ScienceDirect. Our findings emphasize the importance of S. mansoni and S. japonicum antigens in macrophage polarization, as they exert immunomodulatory effects in different stages of the disease and are therefore important as therapeutic targets for schistosomiasis and in vaccine development. A combination of different antigens can provide greater protection, as it possibly stimulates an adequate immune response for an M1 or M2 profile and leads to host resistance; however, this warrants in vitro and in vivo studies.
Collapse
Affiliation(s)
- Irlla Correia Lima Licá
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Gleycka Cristine Carvalho Gomes Frazão
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Ranielly Araujo Nogueira
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, 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
- Laboratory of Immunophysiology, 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
- Laboratory of Immunophysiology, 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, Minas Gerais, Brazil
| | - Guilherme Silva Miranda
- Department of Biology, Federal Institute of Education, Science and Technology of Maranhão, São Raimundo das Mangabeiras, Brazil
| | - Rafael Cardoso Carvalho
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
| | - Lucilene Amorim Silva
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, 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
| | - Rosane Nassar Meireles Guerra
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, 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
| | - Flávia Raquel Fernandes Nascimento
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
- Laboratory of Immunophysiology, 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
|
12
|
He X, Sun Y, Yang F, Zheng G, Li R, Liu M, Li W, Zhou DH, Zheng Y. Heat shock protein 60 in parasitic helminths: A role in immune responses and therapeutic applications. Mol Biochem Parasitol 2023; 253:111544. [PMID: 36641059 DOI: 10.1016/j.molbiopara.2023.111544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 01/13/2023]
Abstract
Heat shock protein 60 (HSP60) is an unique member of the heat shock protein family, being involved in parasite infections. To cope with harsh environments where parasites live, HSP60s are indispensable and involved in a variety of biological processes. HSP60s have relative low similarity among parasites, but their ATPase /Mg2+ active sites are highly conserved. The interactions of HSP60s with signaling pathway regulators in immune cells suggest a crucial role in immune responses, rendering them a potential therapeutic target. This paper reviews the current understandings of HSP60s in parasitic helminths in aspects of molecular characteristics, immunoregulatory responses and HSP60-based therapeutics.
Collapse
Affiliation(s)
- Xuedong He
- Key Laboratory of Applied Technology on Green-Eco Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology&College of Veterinary Medicine, Zhejiang A&F University, Hangzhou 311300, China
| | - Yue Sun
- Key Laboratory of Applied Technology on Green-Eco Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology&College of Veterinary Medicine, Zhejiang A&F University, Hangzhou 311300, China
| | - Fang Yang
- Zhejiang Kangjia Gene Technology Limited Liability Company, Hangzhou 310022, China
| | - Guanghui Zheng
- Key Laboratory of Applied Technology on Green-Eco Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology&College of Veterinary Medicine, Zhejiang A&F University, Hangzhou 311300, China
| | - Rui Li
- Key Laboratory of Applied Technology on Green-Eco Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology&College of Veterinary Medicine, Zhejiang A&F University, Hangzhou 311300, China
| | - Mengqi Liu
- Key Laboratory of Applied Technology on Green-Eco Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology&College of Veterinary Medicine, Zhejiang A&F University, Hangzhou 311300, China
| | - Wanjing Li
- Key Laboratory of Applied Technology on Green-Eco Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology&College of Veterinary Medicine, Zhejiang A&F University, Hangzhou 311300, China
| | - Dong-Hui Zhou
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| | - Yadong Zheng
- Key Laboratory of Applied Technology on Green-Eco Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology, College of Animal Science and Technology&College of Veterinary Medicine, Zhejiang A&F University, Hangzhou 311300, China.
| |
Collapse
|
13
|
Kasambala M, Mukaratirwa S, Vengesai A, Mduluza-Jokonya T, Jokonya L, Midzi H, Makota RB, Mutemeri A, Maziti E, Dube-Marimbe B, Chibanda D, Mutapi F, Mduluza T. The association of systemic inflammation and cognitive functions of pre-school aged children residing in a Schistosoma haematobium endemic area in Zimbabwe. Front Immunol 2023; 14:1139912. [PMID: 37143686 PMCID: PMC10151793 DOI: 10.3389/fimmu.2023.1139912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/31/2023] [Indexed: 05/06/2023] Open
Abstract
Background Cognitive function is negatively impacted by schistosomiasis and might be caused by systemic inflammation which has been hypothesized to be one of the mechanisms driving cognitive decline, This study explored the association of systemic inflammatory biomarkers; interleukin (IL)-10, IL-6, IL-17, transforming growth factor (TGF-β), tumor necrosis factor (TNF-α), C-reactive protein (CRP) and hematological parameters with cognitive performance of preschool-aged children (PSAC) from an Schistosoma haematobium endemic area. Methods The Griffith III tool was used to measure the cognitive performance of 136 PSAC. Whole blood and sera were collected and used to quantify levels of IL-10, TNF-α, IL-6, TGF-β, IL-17 A and CRP using the enzyme-linked immunosorbent assay and hematological parameters using the hematology analyzer. Spearman correlation analysis was used to determine the relationship between each inflammatory biomarker and cognitive performance. Multivariate logistic regression analysis was used to determine whether systemic inflammation due to S. haematobium infection affected cognitive performance in PSAC. Results Higher levels of TNF-α and IL-6, were correlated with lower performance in the Foundations of Learning domain (r = -0.30; p < 0.001 and r = -0.26; p < 0.001), respectively. Low cognitive performance in the Eye-Hand-Coordination Domain was observed in PSAC with high levels of the following inflammatory biomarkers that showed negative correlations to performance; TNF-α (r = -0.26; p < 0.001), IL-6 (r = -0.29; p < 0.001), IL-10 (r = -0.18; p < 0.04), WBC (r = -0.29; p < 0.001), neutrophils (r = -0.21; p = 0.01) and lymphocytes (r = -0.25; p = 0.003) The General Development Domain correlated with TNF-α (r = -0.28; p < 0.001) and IL-6 (r = -0.30; p < 0.001). TGF-β, L-17A and MXD had no significant correlations to performance in any of the cognitive domains. The overall general development of PSAC was negatively impacted by S. haematobium infections (OR = 7.6; p = 0.008) and (OR = 5.6; p = 0.03) where the PSAC had higher levels of TNF-α and IL-6 respectively. Conclusion Systemic inflammation and S. haematobium infections are negatively associated with cognitive function. We recommend the inclusion of PSAC into mass drug treatment programs.
Collapse
Affiliation(s)
- Maritha Kasambala
- School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
- Department of Biological Sciences and Ecology, University of Zimbabwe, Harare, Zimbabwe
- *Correspondence: Maritha Kasambala,
| | - Samson Mukaratirwa
- School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
- One Health Center for Zoonoses and Tropical Veterinary Medicine, Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis
| | - Arthur Vengesai
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Midlands State University, Gweru, Zimbabwe
| | - Tariro Mduluza-Jokonya
- Department of Surgery, College of Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Luxwell Jokonya
- Department of Surgery, College of Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Herald Midzi
- School of Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- Department of Biotechnology and Biochemistry, University of Zimbabwe, Harare, Zimbabwe
| | - Rutendo Birri Makota
- Department of Biological Sciences and Ecology, University of Zimbabwe, Harare, Zimbabwe
| | - Arnold Mutemeri
- Department of Psychiatry, College of Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Emmanuel Maziti
- Department of Psychiatry, College of Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Bazondlile Dube-Marimbe
- Department of Psychiatry, College of Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Dixon Chibanda
- Department of Psychiatry, College of Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Francisca Mutapi
- Ashworth Laboratories, Institute for Immunology and Infection Research and Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Takafira Mduluza
- School of Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
14
|
Agrawal M, Allin KH, Iversen AT, Mehandru S, Colombel JF, Jess T. Early-Life Mebendazole Exposure Increases the Risk of Adult-Onset Ulcerative Colitis: A Population-Based Cohort Study. Am J Gastroenterol 2022; 117:2025-2032. [PMID: 36040420 PMCID: PMC9722538 DOI: 10.14309/ajg.0000000000001933] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
INTRODUCTION According to the hygiene hypothesis, exposure to parasites may protect against inflammatory bowel disease (IBD). Our aim was to examine the risk of IBD with childhood exposure to mebendazole, a broad-spectrum antihelminthic agent. METHODS We conducted a population-based cohort study using prospectively collected historical data of all individuals born in Denmark between 1995 and 2018. We identified mebendazole exposure at age younger than 18 years and during early life (younger than 5 years). We performed adjusted Cox proportional hazards regression analysis to determine the risk of IBD, ulcerative colitis (UC), and Crohn's disease with mebendazole exposure after adjusting for potential confounders. RESULTS Of 1,520,290 individuals in the cohort, 615,794 had childhood or adolescence mebendazole exposure. One thousand five hundred fifty-five and 1,499 individuals were subsequently diagnosed with pediatric-onset and adult-onset IBD, respectively. On multivariable analysis, mebendazole exposure at age younger than 18 years did not affect pediatric-onset or adult-onset IBD risk (adjusted hazard ratio [aHR] 0.97, 95% confidence interval [CI] 0.87, 1.07, and 1.08, 95% CI 0.97, 1.19, respectively). On limiting mebendazole exposure to age younger than 5 years while there was no association with pediatric-onset IBD (aHR 0.98, 95% CI 0.87, 1.11), adult-onset IBD risk was increased (aHR 1.17, 95% CI 1.04, 1.31). This increase in risk was driven by UC (aHR 1.32, 95% CI 1.12, 1.55), but not Crohn's disease (1.03, 95% CI 0.87, 1.22). DISCUSSION Early-life mebendazole exposure is associated with an increase in the risk of adult-onset UC. These findings suggest the importance of early-life exposures in shaping the risk of IBD later in life.
Collapse
Affiliation(s)
- Manasi Agrawal
- Center for Molecular Prediction of Inflammatory Bowel Disease (PREDICT), Aalborg University, Department of Clinical Medicine, Copenhagen, Denmark
- The Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kristine H. Allin
- Center for Molecular Prediction of Inflammatory Bowel Disease (PREDICT), Aalborg University, Department of Clinical Medicine, Copenhagen, Denmark
- Department of Gastroenterology & Hepatology, Aalborg University Hospital, Aalborg, Denmark
| | - Aske T. Iversen
- Center for Molecular Prediction of Inflammatory Bowel Disease (PREDICT), Aalborg University, Department of Clinical Medicine, Copenhagen, Denmark
| | - Saurabh Mehandru
- The Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY
- Precision Institute of Immunology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jean-Frederic Colombel
- The Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Tine Jess
- Center for Molecular Prediction of Inflammatory Bowel Disease (PREDICT), Aalborg University, Department of Clinical Medicine, Copenhagen, Denmark
- Department of Gastroenterology & Hepatology, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
15
|
Zhang L, Wang L, Xiang S, Hu Y, Zhao S, Liao Y, Zhu Z, Wu X. CRISPR/Cas9-mediated gene knockout of Sj16 in Schistosoma japonicum eggs upregulates the host-to-egg immune response. FASEB J 2022; 36:e22615. [PMID: 36273308 DOI: 10.1096/fj.202200600rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 09/15/2022] [Accepted: 10/04/2022] [Indexed: 11/11/2022]
Abstract
Schistosomiasis is an important, neglected tropical disease. Schistosoma japonicum can evade host attacks by regulating the host's immunity, causing continuous infection. However, interactions between the host's immune system and S. japonicum are unclear. Our previous research found that the Sj16 protein isolated from S. japonicum has an anti-inflammatory effect in the host. However, the role of Sj16 in the regulation of host immunity in S. japonicum infection is not clear. Here, we applied the CRISPR/Cas9 technique to knockout Sj16 in S. japonicum eggs and investigated the effect of Sj16 in regulating host immunity. We found egg viability decreased after Sj16 knockout. In addition, we found granulomatous inflammation increased, the T-cell immune response enhanced and the immune microenvironment changed in mice model injected with Sj16-knockout eggs by tail vein. These findings suggested that S. japonicum could regulate host immunity through Sj16 to evade the host immune attack and cause continuous infection. In addition, we confirmed the application of CRISPR/Cas9-mediated gene reprogramming for functional genomics in S. japonicum.
Collapse
Affiliation(s)
- Lichao 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, Guangzhou, China
| | - Lifu Wang
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| | - Suoyu Xiang
- 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, Guangzhou, China
| | - Yunyi Hu
- 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, Guangzhou, China
| | - Siyu Zhao
- 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, Guangzhou, China
| | - Yao Liao
- 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, Guangzhou, China
| | - Zifeng Zhu
- 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, Guangzhou, China
| | - Xiaoying Wu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Gastroenterology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
16
|
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
|
17
|
Zhu T, Xue Q, Liu Y, Xu Y, Xiong C, Lu J, Yang H, Zhang Q, Huang Y. Analysis of Intestinal Microflora and Metabolites From Mice With DSS-Induced IBD Treated With Schistosoma Soluble Egg Antigen. Front Cell Dev Biol 2021; 9:777218. [PMID: 34858992 PMCID: PMC8630629 DOI: 10.3389/fcell.2021.777218] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Objective: This study aimed to analyze the changes in intestinal flora and metabolites in the intestinal contents of mice with inflammatory bowel disease (IBD) to preliminarily clarify the mechanism of action of Schistosoma soluble egg antigen (SEA) on IBD, thus, laying a research foundation for the subsequent treatment of IBD. Methods: A total of 40 Institute of Cancer Research (ICR) mice were divided into four groups: control, SEA 50 μg, dextran sulfate sodium salt (DSS), and SEA 50 μg + DSS. The overall state of the animals was observed continuously during modeling. The colonic length was measured after 10 days of modeling. The degree of colonic inflammation was observed by hematoxylin and eosin staining. 16srRNA and liquid chromatography-mass spectrometry sequencing techniques were used to determine the abundance of bacteria and metabolites in the intestinal contents of mice in the DSS and SEA 50 μg + DSS groups, and the differences were further analyzed. Results: After SEA intervention, the disease activity index score of mice with IBD decreased and the colon shortening was reduced. Microscopically, the lymphocyte aggregation, glandular atrophy, goblet cell disappearance, and colonic inflammation were less in the SEA 50 μg + DSS group than in the DSS group (p < 0.0001). After SEA intervention, the abundance of beneficial bacteria prevotellaceae_UCG-001 was upregulated, while the abundance of the harmful bacteria Helicobacter, Lachnoclostridium, and Enterococcus was downregulated in the intestinal tract of mice with IBD. The intestinal metabolite analysis showed that SEA intervention decreased the intestinal contents of glycerophospholipids (lysophosphatidylcholine, lysophosphatidylethanolamine, phatidylcholine, and phatidylethanolamine) and carboxylic acids (L-alloisoleucine and L-glutamate), whereas increased bile acids and their derivatives (3B,7A,12a-trihydroxy-5A-cholanoic acid and 3A,4B, 12a-trihydroxy-5b-cholanoic acid). Combined microbiota-metabolite analysis revealed a correlation between these differential microbiota and differential metabolites. At the same time, the changes in the contents of metabolites and differential metabolites in the two groups also correlated with the abundance of the gut microbiome. Conclusions: The study showed that SEA reduced DSS-induced inflammation in IBD and improved the symptoms of IBD in mice through the combined regulation of intestinal flora and intestinal metabolism. It suggested a potential possibility for the use of SEA in treating and regulating intestinal flora and metabolism in patients with IBD.
Collapse
Affiliation(s)
- Tianyu Zhu
- 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.,Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qingkai Xue
- 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.,Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yiyun Liu
- 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.,Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yongliang Xu
- 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.,Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chunrong Xiong
- 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.,Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jin Lu
- 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.,Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Haitao Yang
- 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.,Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Quan Zhang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Yuzheng Huang
- 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.,Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
18
|
Pierre N, Salée C, Vieujean S, Bequet E, Merli AM, Siegmund B, Meuwis MA, Louis E. Review article: distinctions between ileal and colonic Crohn's disease: from physiology to pathology. Aliment Pharmacol Ther 2021; 54:779-791. [PMID: 34297423 DOI: 10.1111/apt.16536] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/15/2021] [Accepted: 07/05/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Ileal and colonic Crohn's disease seem to be two separate entities. AIMS To describe the main physiological distinctions between the small and the large intestine and to analyse the differences between ileal and colonic Crohn's disease. METHODS The relevant literature was critically examined and synthesised. RESULTS The small and large intestine have fundamental distinctions (anatomy, cellular populations, immune defence, microbiota). The differences between ileal and colonic Crohn's disease are highlighted by a heterogeneous body of evidence including clinical features (natural history of the disease, efficacy of treatments, and monitoring), epidemiological data (smoking status, age, gender) and biological data (genetics, microbiota, immunity, mesenteric fat). However, the contribution of these factors to disease location remains poorly understood. CONCLUSION The classification of ileal and colonic Crohn's disease as distinct subphenotypes is well supported by the literature. Understanding of these differences could be exploited to develop more individualised patient care.
Collapse
Affiliation(s)
- Nicolas Pierre
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium
| | - Catherine Salée
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium
| | - Sophie Vieujean
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium.,Hepato-Gastroenterology and Digestive Oncology Department, Liège University Hospital, Liège, Belgium
| | - Emeline Bequet
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium.,Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Liège University Hospital, Liège, Belgium
| | - Angela-Maria Merli
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium
| | - Britta Siegmund
- Division of Gastroenterology, Infectiology and Rheumatology, Medical Department, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Marie-Alice Meuwis
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium.,Hepato-Gastroenterology and Digestive Oncology Department, Liège University Hospital, Liège, Belgium
| | - Edouard Louis
- Laboratory of Translational Gastroenterology, GIGA-Institute, University of Liège, Liège, Belgium.,Hepato-Gastroenterology and Digestive Oncology Department, Liège University Hospital, Liège, Belgium
| |
Collapse
|
19
|
The relationship between Schistosoma and glycolipid metabolism. Microb Pathog 2021; 159:105120. [PMID: 34358648 DOI: 10.1016/j.micpath.2021.105120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 02/02/2023]
Abstract
Diabetes and obesity have become the most popular metabolic diseases in the world. A large number of previous studies have shown that glucose and lipid metabolism disorder is an important risk factor and a main cause of diabetes and obesity. Schistosoma is a parasite transmitted by freshwater snails. It can induce a series of inflammatory and immune reactions after infecting the human body, causing schistosomiasis. However, in recent years, studies have found that Schistosoma infection or Schistosoma related products can improve or prevent some immune and inflammatory diseases, such as severe asthma, inflammatory bowel disease, diabetes and so on. Further experiments have also revealed that Schistosoma can promote the secretion of anti-inflammatory factors and regulate the glucose and lipid metabolism in the host body by polarizing immune cells such as T cells, B cells and dendritic cells (DCs). In this review, we summarize studies that investigated Schistosoma and Schistosoma-derived products and their relationship with glycolipid metabolism and related diseases, highlighting potential protective mechanisms.
Collapse
|
20
|
Oliveira NF, Silva CLM. Unveiling the Potential of Purinergic Signaling in Schistosomiasis Treatment. Curr Top Med Chem 2021; 21:193-204. [PMID: 32972342 DOI: 10.2174/1568026620666200924115113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/15/2020] [Accepted: 08/24/2020] [Indexed: 11/22/2022]
Abstract
Schistosomiasis is a neglected tropical disease. It is related to long-lasting granulomatous fibrosis and inflammation of target organs, and current sub-optimal pharmacological treatment creates global public health concerns. Intravascular worms and eggs release antigens and extracellular vesicles that target host endothelial cells, modulate the immune system, and stimulate the release of damageassociated molecular patterns (DAMPs). ATP, one of the most studied DAMPs, triggers a cascade of autocrine and paracrine actions through purinergic P2X and P2Y receptors, which are shaped by ectonucleotidases (CD39). Both P2 receptor families, and in particular P2Y1, P2Y2, P2Y12, and P2X7 receptors, have been attracting increasing interest in several inflammatory diseases and drug development. Current data obtained from the murine model unveiled a CD39-ADP-P2Y1/P2Y12 receptors signaling pathway linked to the liver and mesenteric exacerbations of schistosomal inflammation. Therefore, we proposed that members of this purinergic signaling could be putative pharmacological targets to reduce schistosomal morbidity.
Collapse
Affiliation(s)
- Nathália Ferreira Oliveira
- Laboratory of Molecular and Biochemical Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia Lucia Martins Silva
- Laboratory of Molecular and Biochemical Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
21
|
Benton ML, Abraham A, LaBella AL, Abbot P, Rokas A, Capra JA. The influence of evolutionary history on human health and disease. Nat Rev Genet 2021; 22:269-283. [PMID: 33408383 PMCID: PMC7787134 DOI: 10.1038/s41576-020-00305-9] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2020] [Indexed: 01/29/2023]
Abstract
Nearly all genetic variants that influence disease risk have human-specific origins; however, the systems they influence have ancient roots that often trace back to evolutionary events long before the origin of humans. Here, we review how advances in our understanding of the genetic architectures of diseases, recent human evolution and deep evolutionary history can help explain how and why humans in modern environments become ill. Human populations exhibit differences in the prevalence of many common and rare genetic diseases. These differences are largely the result of the diverse environmental, cultural, demographic and genetic histories of modern human populations. Synthesizing our growing knowledge of evolutionary history with genetic medicine, while accounting for environmental and social factors, will help to achieve the promise of personalized genomics and realize the potential hidden in an individual's DNA sequence to guide clinical decisions. In short, precision medicine is fundamentally evolutionary medicine, and integration of evolutionary perspectives into the clinic will support the realization of its full potential.
Collapse
Affiliation(s)
- Mary Lauren Benton
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Computer Science, Baylor University, Waco, TX, USA
| | - Abin Abraham
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
- Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA
| | - Abigail L LaBella
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Patrick Abbot
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Antonis Rokas
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - John A Capra
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA.
- Bakar Computational Health Sciences Institute and Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA.
| |
Collapse
|
22
|
Avni D, Avni O. Extracellular Vesicles: Schistosomal Long-Range Precise Weapon to Manipulate the Immune Response. Front Cell Infect Microbiol 2021; 11:649480. [PMID: 33869080 PMCID: PMC8044974 DOI: 10.3389/fcimb.2021.649480] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
Schistosomiasis (Bilharziasis), a neglected tropical disease that affects more than 240 million people around the world, is caused by infection with the helminth parasite Schistosoma. As part of their secretome, schistosomes release extracellular vesicles (EVs) that modulate the host immune response. The EV-harbored miRNAs upregulate the innate immune response of the M1 pathway and downregulate the differentiation toward the adaptive Th2 immunity. A schistosomal egg-derived miRNA increases the percentage of regulatory T cells. This schistosomal-inducible immunoediting process generates ultimately a parasitic friendly environment that is applied carefully as restrained Th2 response is crucial for the host survival and successful excretion of the eggs. Evidence indicates a selective targeting of schistosomal EVs, however, the underlying mechanisms are unclear yet. The effects of the schistosomes on the host immune system is in accordance with the hygiene hypothesis, attributing the dramatic increase in recent decades in allergy and other diseases associated with imbalanced immune response, to the reduced exposure to infectious agents that co-evolved with humans during evolution. Deciphering the bioactive cargo, function, and selective targeting of the parasite-secreted EVs may facilitate the development of novel tools for diagnostics and delivered therapy to schistosomiasis, as well as to immune-associated disorders.
Collapse
Affiliation(s)
- Dror Avni
- Laboratory of Molecular Cell Biology, Sheba Medical Center, Tel Hashomer, Israel.,Laboratory for the Study of Tropical Diseases, Sheba Medical Center, Tel Hashomer, Israel.,Department of Medicine C, Sheba Medical Center, Tel Hashomer, Israel
| | - Orly Avni
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| |
Collapse
|
23
|
Li W, Wei C, Xu L, Yu B, Chen Y, Lu D, Zhang L, Song X, Dong L, Zhou S, Xu Z, Zhu J, Chen X, Su C. Schistosome infection promotes osteoclast-mediated bone loss. PLoS Pathog 2021; 17:e1009462. [PMID: 33735306 PMCID: PMC8009420 DOI: 10.1371/journal.ppat.1009462] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 03/30/2021] [Accepted: 03/09/2021] [Indexed: 12/24/2022] Open
Abstract
Infection with schistosome results in immunological changes that might influence the skeletal system by inducing immunological states affecting bone metabolism. We investigated the relationships between chronic schistosome infection and bone metabolism by using a mouse model of chronic schistosomiasis, affecting millions of humans worldwide. Results showed that schistosome infection resulted in aberrant osteoclast-mediated bone loss, which was accompanied with an increased level of receptor activator of nuclear factor-κB (NF-κB) Ligand (RANKL) and decreased level of osteoprotegerin (OPG). The blockade of RANKL by the anti-RANKL antibody could prevent bone loss in the context of schistosome infection. Meanwhile, both B cells and CD4+ T cells, particularly follicular helper T (Tfh) cell subset, were the important cellular sources of RANKL during schistosome infection. These results highlight the risk of bone loss in schistosome-infected patients and the potential benefit of coupling bone therapy with anti-schistosome treatment. Schistosomiasis remains an important public health problem in many countries in tropical and subtropical regions, which affects about 200 million people worldwide, with another 700 million considered at risk of infection. Although the primary cause of pathogenesis of schistosomiasis is the granulomatous inflammatory responses, schistosomiasis patients experience long-term hidden pathologies that remain poorly investigated. Here, we found that schistosome infection resulted in RANKL-associated bone loss. Furthermore, our results indicated that both B cells and CD4+ T cells, particularly Tfh cell subset, in the peripheral lymphoid tissues are likely to be the important contributors to bone loss through releasing soluble RANKL. In addition, Tfh cells played a sufficient but not necessary role in schistosome infection-induced bone loss. Our findings highlight the risk of bone loss in schistosome-infected patients and the potential benefit of coupling bone therapy with anti-schistosome treatment.
Collapse
Affiliation(s)
- Wei Li
- Department of Clinical Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, P. R. China
- State Key Lab of Reproductive Medicine, Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, Center for Global Health, Nanjing Medical University, Nanjing, P. R. China
| | - Chuan Wei
- State Key Lab of Reproductive Medicine, Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, Center for Global Health, Nanjing Medical University, Nanjing, P. R. China
| | - Lei Xu
- State Key Lab of Reproductive Medicine, Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, Center for Global Health, Nanjing Medical University, Nanjing, P. R. China
| | - Beibei Yu
- State Key Lab of Reproductive Medicine, Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, Center for Global Health, Nanjing Medical University, Nanjing, P. R. China
| | - Ying Chen
- State Key Lab of Reproductive Medicine, Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, Center for Global Health, Nanjing Medical University, Nanjing, P. R. China
| | - Di Lu
- State Key Lab of Reproductive Medicine, Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, Center for Global Health, Nanjing Medical University, Nanjing, P. R. China
| | - Lina Zhang
- State Key Lab of Reproductive Medicine, Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, Center for Global Health, Nanjing Medical University, Nanjing, P. R. China
| | - Xian Song
- State Key Lab of Reproductive Medicine, Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, Center for Global Health, Nanjing Medical University, Nanjing, P. R. China
| | - Liyang Dong
- State Key Lab of Reproductive Medicine, Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, Center for Global Health, Nanjing Medical University, Nanjing, P. R. China
| | - Sha Zhou
- State Key Lab of Reproductive Medicine, Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, Center for Global Health, Nanjing Medical University, Nanjing, P. R. China
| | - Zhipeng Xu
- State Key Lab of Reproductive Medicine, Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, Center for Global Health, Nanjing Medical University, Nanjing, P. R. China
| | - Jifeng Zhu
- State Key Lab of Reproductive Medicine, Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, Center for Global Health, Nanjing Medical University, Nanjing, P. R. China
| | - Xiaojun Chen
- State Key Lab of Reproductive Medicine, Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, Center for Global Health, Nanjing Medical University, Nanjing, P. R. China
- * E-mail: (XC); (CS)
| | - Chuan Su
- State Key Lab of Reproductive Medicine, Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, Center for Global Health, Nanjing Medical University, Nanjing, P. R. China
- * E-mail: (XC); (CS)
| |
Collapse
|
24
|
Tootee A, Nikbin B, Ghahary A, Esfahani EN, Arjmand B, Aghayan H, Qorbani M, Larijani B. Immunopathology of Type 1 Diabetes and Immunomodulatory Effects of Stem Cells: A Narrative Review of the Literature. Endocr Metab Immune Disord Drug Targets 2021; 22:169-197. [PMID: 33538679 DOI: 10.2174/1871530321666210203212809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/11/2020] [Accepted: 10/27/2020] [Indexed: 11/22/2022]
Abstract
Type 1 Diabetes (T1D) is a complex autoimmune disorder which occurs as a result of an intricate series of pathologic interactions between pancreatic β-cells and a wide range of components of both the innate and the adaptive immune systems. Stem-cell therapy, a recently-emerged potentially therapeutic option for curative treatment of diabetes, is demonstrated to cause significant alternations to both different immune cells such as macrophages, natural killer (NK) cells, dendritic cells, T cells, and B cells and non-cellular elements including serum cytokines and different components of the complement system. Although there exists overwhelming evidence indicating that the documented therapeutic effects of stem cells on patients with T1D is primarily due to their potential for immune regulation rather than pancreatic tissue regeneration, to date, the precise underlying mechanisms remain obscure. On the other hand, immune-mediated rejection of stem cells remains one of the main obstacles to regenerative medicine. Moreover, the consequences of efferocytosis of stem-cells by the recipients' lung-resident macrophages have recently emerged as a responsible mechanism for some immune-mediated therapeutic effects of stem-cells. This review focuses on the nature of the interactions amongst different compartments of the immune systems which are involved in the pathogenesis of T1D and provides explanation as to how stem cell-based interventions can influence immune system and maintain the physiologic equilibrium.
Collapse
Affiliation(s)
- Ali Tootee
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| | - Behrouz Nikbin
- Research Center of Molecular Immunology, Tehran University of Medical Sciences, Tehran, . Iran
| | - Aziz Ghahary
- British Columbia Professional Firefighters' Burn and Wound Healing Research Laboratory, Department of Surgery, Plastic Surgery, University of British Columbia, Vancouver, . Canada
| | - Ensieh Nasli Esfahani
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| | - Babak Arjmand
- Cell therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| | - Hamidreza Aghayan
- Cell therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| | - Mostafa Qorbani
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, . Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| |
Collapse
|
25
|
Ng YQ, Gupte TP, Krause PJ. Tick hypersensitivity and human tick-borne diseases. Parasite Immunol 2021; 43:e12819. [PMID: 33428244 DOI: 10.1111/pim.12819] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/22/2020] [Accepted: 01/08/2021] [Indexed: 12/15/2022]
Abstract
Immune-mediated hypersensitivity reactions to ticks and other arthropods are well documented. Hypersensitivity to ixodid (hard bodied) ticks is especially important because they transmit infection to humans throughout the world and are responsible for most vector-borne diseases in the United States. The causative pathogens of these diseases are transmitted in tick saliva that is secreted into the host while taking a blood meal. Tick salivary proteins inhibit blood coagulation, block the local itch response and impair host anti-tick immune responses, which allows completion of the blood meal. Anti-tick host immune responses are heightened upon repeated tick exposure and have the potential to abrogate tick salivary protein function, interfere with the blood meal and prevent pathogen transmission. Although there have been relatively few tick bite hypersensitivity studies in humans compared with those in domestic animals and laboratory animal models, areas of human investigation have included local hypersensitivity reactions at the site of tick attachment and generalized hypersensitivity reactions. Progress in the development of anti-tick vaccines for humans has been slow due to the complexities of such vaccines but has recently accelerated. This approach holds great promise for future prevention of tick-borne diseases.
Collapse
Affiliation(s)
- Yu Quan Ng
- Yale School of Public Health and Yale School of Medicine, New Haven, CT, USA
| | - Trisha P Gupte
- Yale School of Public Health and Yale School of Medicine, New Haven, CT, USA
| | - Peter J Krause
- Yale School of Public Health and Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
26
|
Macháček T, Šmídová B, Pankrác J, Majer M, Bulantová J, Horák P. Nitric oxide debilitates the neuropathogenic schistosome Trichobilharzia regenti in mice, partly by inhibiting its vital peptidases. Parasit Vectors 2020; 13:426. [PMID: 32819437 PMCID: PMC7439556 DOI: 10.1186/s13071-020-04279-9] [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: 03/17/2020] [Accepted: 08/03/2020] [Indexed: 12/12/2022] Open
Abstract
Background Avian schistosomes, the causative agents of human cercarial dermatitis (or swimmer’s itch), die in mammals but the mechanisms responsible for parasite elimination are unknown. Here we examined the role of reactive nitrogen species, nitric oxide (NO) and peroxynitrite, in the immune response of mice experimentally infected with Trichobilharzia regenti, a model species of avian schistosomes remarkable for its neuropathogenicity. Methods Inducible NO synthase (iNOS) was localized by immunohistochemistry in the skin and the spinal cord of mice infected by T. regenti. The impact of iNOS inhibition by aminoguanidine on parasite burden and growth was then evaluated in vivo. The vulnerability of T. regenti schistosomula to NO and peroxynitrite was assessed in vitro by viability assays and electron microscopy. Additionally, the effect of NO on the activity of T. regenti peptidases was tested using a fluorogenic substrate. Results iNOS was detected around the parasites in the epidermis 8 h post-infection and also in the spinal cord 3 days post-infection (dpi). Inhibition of iNOS resulted in slower parasite growth 3 dpi, but the opposite effect was observed 7 dpi. At the latter time point, moderately increased parasite burden was also noticed in the spinal cord. In vitro, NO did not impair the parasites, but inhibited the activity of T. regenti cathepsins B1.1 and B2, the peptidases essential for parasite migration and digestion. Peroxynitrite severely damaged the surface tegument of the parasites and decreased their viability in vitro, but rather did not participate in parasite clearance in vivo. Conclusions Reactive nitrogen species, specifically NO, do not directly kill T. regenti in mice. NO promotes the parasite growth soon after penetration (3 dpi), but prevents it later (7 dpi) when also suspends the parasite migration in the CNS. NO-related disruption of the parasite proteolytic machinery is partly responsible for this effect. ![]()
Collapse
Affiliation(s)
- Tomáš Macháček
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia.
| | - Barbora Šmídová
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
| | - Jan Pankrác
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia.,Center for Advanced Preclinical Imaging, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Martin Majer
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
| | - Jana Bulantová
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
| | - Petr Horák
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
| |
Collapse
|
27
|
Castro Rocha FA, Duarte-Monteiro AM, Henrique da Mota LM, Matias Dinelly Pinto AC, Fonseca JE. Microbes, helminths, and rheumatic diseases. Best Pract Res Clin Rheumatol 2020; 34:101528. [PMID: 32448639 PMCID: PMC7203059 DOI: 10.1016/j.berh.2020.101528] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There has been a progressive interest on modifications of the human defense system following insults occurring in the interface between our body and the external environment, as they may provoke or worsen disease states. Studies suggest that billions of germs, which compose the gut microbiota influence one's innate and adaptive immune responses at the intestinal level, but these microorganisms may also impact rheumatic diseases. The microbiota of the skin, respiratory, and urinary tracts may also be relevant in rheumatology. Evidence indicates that changes in the gut microbiome alter the pathogenesis of immune-mediated diseases such as rheumatoid arthritis and ankylosing spondylitis but also of other disorders like atherosclerosis and osteoarthritis. Therapeutic strategies to modify the microbiota, including probiotics and fecal microbiota transplantation, have been received with skepticism, which, in turn, has drawn attention back to previously developed interventions such as antibiotics. Helminths adapted to humans over the evolution process, but their role in disease modulation, particularly immune-mediated diseases, remains to be understood. The present review focuses on data concerning modifications of the immune system induced by interactions with microbes and pluricellular organisms, namely helminths, and their impact on rheumatic diseases. Practical aspects, including specific microbiota-targeted therapies, are also discussed.
Collapse
Affiliation(s)
- Francisco Airton Castro Rocha
- Departamento de Medicina Clínica, Liga de Reumatologia e Doenças Autoimunes, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brazil.
| | - Ana Margarida Duarte-Monteiro
- Serviço de Reumatologia e Doenças Ósseas Metabólicas, Hospital de Santa Maria, CHULN and Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisboa, Portugal
| | - Licia Maria Henrique da Mota
- Hospital Universitário de Brasília, Programa de Pós-graduação em Ciências Médicas, Faculdade de Medicina, Universidade de Brasília, Brazil
| | - Ana Carolina Matias Dinelly Pinto
- Departamento de Medicina Clínica, Liga de Reumatologia e Doenças Autoimunes, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - João Eurico Fonseca
- Serviço de Reumatologia e Doenças Ósseas Metabólicas, Hospital de Santa Maria, CHULN and Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisboa, Portugal
| |
Collapse
|
28
|
Bexkens ML, van Gestel RA, van Breukelen B, Urbanus RT, Brouwers JF, Nieuwland R, Tielens AGM, van Hellemond JJ. Schistosoma mansoni infection affects the proteome and lipidome of circulating extracellular vesicles in the host. Mol Biochem Parasitol 2020; 238:111296. [PMID: 32603736 DOI: 10.1016/j.molbiopara.2020.111296] [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: 02/22/2020] [Revised: 06/19/2020] [Accepted: 06/25/2020] [Indexed: 12/19/2022]
Abstract
Eggs, schistosomula and adult Schistosoma worms are known to release extracellular vesicles (EV) during in vitro incubations and these EVs are postulated to affect the host responses. So far only those EVs released during in vitro incubations of schistosomes have been studied and it is unknown whether in blood of infected hosts the schistosomal EVs can be detected amidst all the circulating EVs of the host itself. In this study we analyzed the protein as well as the phospholipid composition of EVs circulating in blood plasma of S. mansoni infected hamsters and compared those with the EVs circulating in blood of non-infected hamsters. Although neither proteins nor lipids specific for schistosomes could be detected in the circulating EVs of the infected hamsters, the infection with schistosomes had a marked effect on the circulating EVs of the host, as the protein as well as the lipid composition of EVs circulating in infected hamsters were different from the EVs of uninfected hamsters. The observed changes in the EV lipid and protein content suggest that more EVs are released by the diseased liver, the affected erythrocytes and activated immune cells.
Collapse
Affiliation(s)
- Michiel L Bexkens
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Renske A van Gestel
- Biomolecular Mass Spectrometry & Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Bas van Breukelen
- Biomolecular Mass Spectrometry & Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Rolf T Urbanus
- Department of Clinical Chemistry and Haematology, Center for Circulatory Health, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jos F Brouwers
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Vesicle Observation Centre, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Aloysius G M Tielens
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Jaap J van Hellemond
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
29
|
Kuipers ME, Nolte-'t Hoen ENM, van der Ham AJ, Ozir-Fazalalikhan A, Nguyen DL, de Korne CM, Koning RI, Tomes JJ, Hoffmann KF, Smits HH, Hokke CH. DC-SIGN mediated internalisation of glycosylated extracellular vesicles from Schistosoma mansoni increases activation of monocyte-derived dendritic cells. J Extracell Vesicles 2020; 9:1753420. [PMID: 32489529 PMCID: PMC7241508 DOI: 10.1080/20013078.2020.1753420] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 12/13/2022] Open
Abstract
Helminths like Schistosoma mansoni release excretory/secretory (E/S) products that modulate host immunity to enable infection. Extracellular vesicles (EVs) are among these E/S products, yet molecular mechanisms and functionality of S. mansoni EV interaction with host immune cells is unknown. Here we demonstrate that EVs released by S. mansoni schistosomula are internalised by human monocyte-derived dendritic cells (moDCs). Importantly, we show that this uptake was mainly mediated via DC-SIGN (CD209). Blocking DC-SIGN almost completely abrogated EV uptake, while blocking mannose receptor (MR, CD206) or dendritic cell immunoreceptor (DCIR, CLEC4A) had no effect on EV uptake. Mass spectrometric analysis of EV glycans revealed the presence of surface N-glycans with terminal Galβ1-4(Fucα1-3)GlcNAc (LewisX) motifs, and a wide array of fucosylated lipid-linked glycans, including LewisX, a known ligand for DC-SIGN. Stimulation of moDCs with schistosomula EVs led to increased expression of costimulatory molecules CD86 and CD80 and regulatory surface marker PD-L1. Furthermore, schistosomula EVs increased expression of IL-12 and IL-10 by moDCs, which was partly dependent on the interaction with DC-SIGN. These results provide the first evidence that glycosylation of S. mansoni EVs facilitates the interaction with host immune cells and reveals a role for DC-SIGN and EV-associated glycoconjugates in parasite-induced immune modulation.
Collapse
Affiliation(s)
- Marije E Kuipers
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands.,Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Esther N M Nolte-'t Hoen
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Alwin J van der Ham
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | - D Linh Nguyen
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Clarize M de Korne
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Roman I Koning
- Department of Cell & Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - John J Tomes
- Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth University, Aberystwyth, UK
| | - Karl F Hoffmann
- Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth University, Aberystwyth, UK
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Cornelis H Hokke
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
30
|
Crane AL, Brown GE, Chivers DP, Ferrari MCO. An ecological framework of neophobia: from cells to organisms to populations. Biol Rev Camb Philos Soc 2020; 95:218-231. [PMID: 31599483 DOI: 10.1111/brv.12560] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/09/2019] [Accepted: 09/11/2019] [Indexed: 01/24/2023]
Abstract
Neophobia is the fear of novel stimuli or situations. This phenotype has recently received much ecological attention, primarily in the context of decision making. Here, we explore neophobia across biological levels of organisation, first describing types of neophobia among animals and the underlying causes of neophobia, highlighting high levels of risk and uncertainty as key drivers. We place neophobia in the framework of Error Management Theory and Signal Detection Theory, showing how increases in overall risk and uncertainty can lead to costly non-responses towards novel threats unless individuals lower their response threshold and become neophobic. We then discuss how neophobic behaviour translates into population and evolutionary consequences before introducing neophobia-like processes at the cellular level, where some phenomena such as allergy and autoimmunity can parallel neophobic behaviour. Finally, we discuss neophobia attenuation, considering how a sudden change in the environment from dangerous to safe can lead to problematic over-responses (i.e. the 'maladaptive defensive carry-over' hypothesis), and discuss treatment methods for such over-responses. We anticipate that bridging the concept of neophobia with a process-centered perspective can facilitate a transfer of insight across organisational levels.
Collapse
Affiliation(s)
- Adam L Crane
- Department of Biology, Concordia University, 7141 Sherbrooke Street West, Montreal, QC, H4B 1R6, Canada.,Department of Biology, University of Saskatchewan, 112 Science Pl., Saskatoon, SK, S7N 5E2, Canada
| | - Grant E Brown
- Department of Biology, Concordia University, 7141 Sherbrooke Street West, Montreal, QC, H4B 1R6, Canada
| | - Douglas P Chivers
- Department of Biology, University of Saskatchewan, 112 Science Pl., Saskatoon, SK, S7N 5E2, Canada
| | - Maud C O Ferrari
- Department of Biomedical Sciences, WCVM, University of Saskatchewan, 52 Campus Drive, Saskatoon, SK, S7N 5B4, Canada
| |
Collapse
|
31
|
Meningher T, Barsheshet Y, Ofir‐Birin Y, Gold D, Brant B, Dekel E, Sidi Y, Schwartz E, Regev‐Rudzki N, Avni O, Avni D. Schistosomal extracellular vesicle-enclosed miRNAs modulate host T helper cell differentiation. EMBO Rep 2020; 21:e47882. [PMID: 31825165 PMCID: PMC6944914 DOI: 10.15252/embr.201947882] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 11/08/2019] [Accepted: 11/13/2019] [Indexed: 12/22/2022] Open
Abstract
During the chronic stage of Schistosoma infection, the female lays fertile eggs, triggering a strong anti-parasitic type 2 helper T-cell (Th2) immune response. It is unclear how this Th2 response gradually declines even though the worms live for years and continue to produce eggs. Here, we show that Schistosoma mansoni downregulates Th2 differentiation in an antigen-presenting cell-independent manner, by modulating the Th2-specific transcriptional program. Adult schistosomes secrete miRNA-harboring extracellular vesicles that are internalized by Th cells in vitro. Schistosomal miRNAs are found also in T helper cells isolated from Peyer's patches and mesenteric lymph nodes of infected mice. In T helper cells, the schistosomal miR-10 targets MAP3K7 and consequently downmodulates NF-κB activity, a critical transcription factor for Th2 differentiation and function. Our results explain, at least partially, how schistosomes tune down the Th2 response, and provide further insight into the reciprocal geographic distribution between high prevalence of parasitic infections and immune disorders such as allergy. Furthermore, this worm-host crosstalk mechanism can be harnessed to develop diagnostic and therapeutic approaches for human schistosomiasis and Th2-associated diseases.
Collapse
Affiliation(s)
- Tal Meningher
- Laboratory of Molecular Cell BiologyCenter for Cancer Research and Department of Medicine CSheba Medical CenterTel HashomerIsrael
- Molecular Laboratory for the Study of Tropical DiseasesSheba Medical CenterTel HashomerIsrael
| | | | - Yifat Ofir‐Birin
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
| | - Daniel Gold
- Department of Clinical Microbiology and ImmunologyFaculty of MedicineSackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Boris Brant
- Azrieli Faculty of MedicineBar Ilan UniversitySafedIsrael
| | - Elya Dekel
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
| | - Yechezkel Sidi
- Laboratory of Molecular Cell BiologyCenter for Cancer Research and Department of Medicine CSheba Medical CenterTel HashomerIsrael
- Faculty of MedicineSackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Eli Schwartz
- Molecular Laboratory for the Study of Tropical DiseasesSheba Medical CenterTel HashomerIsrael
- Faculty of MedicineSackler School of MedicineTel Aviv UniversityTel AvivIsrael
- The Center for Geographic MedicineSheba Medical CenterTel HashomerIsrael
| | - Neta Regev‐Rudzki
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
| | - Orly Avni
- Azrieli Faculty of MedicineBar Ilan UniversitySafedIsrael
| | - Dror Avni
- Laboratory of Molecular Cell BiologyCenter for Cancer Research and Department of Medicine CSheba Medical CenterTel HashomerIsrael
- Molecular Laboratory for the Study of Tropical DiseasesSheba Medical CenterTel HashomerIsrael
| |
Collapse
|
32
|
Shen J, Wang L, Peng M, Liu Z, Zhang B, Zhou T, Sun X, Wu Z. Recombinant Sj16 protein with novel activity alleviates hepatic granulomatous inflammation and fibrosis induced by Schistosoma japonicum associated with M2 macrophages in a mouse model. Parasit Vectors 2019; 12:457. [PMID: 31547847 PMCID: PMC6755699 DOI: 10.1186/s13071-019-3697-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 09/03/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Potent granulomatous inflammation responses induced by schistosome eggs and resultant fibrosis are the primary causes of morbidity in schistosomiasis. Recombinant Sj16 (rSj16), a 16-kDa protein of Schistosoma japonicum produced in Escherichia coli, has been demonstrated to have novel immunoregulatory effects in vivo and in vitro. Thus, this study investigated the anti-inflammatory and anti-fibrotic effects of rSj16 treatment in S. japonicum-infected mice and demonstrated the immune modulation between the schistosome and the host. METHODS Schistosoma japonicum infected mice were treated with the rSj16 protein and Sj16 peptide at different time points post-infection to assess their efficacy at the optimal time point. Sj16 peptide and/or Praziquantel (PZQ) treatments were initiated at week 5 post-infection to compare the therapeutic efficacy of each regimen. Hepatic granulomatous inflammation, fibrosis and cytokine production (pro-inflammatory, Th1, Th2, Th17 and regulatory cytokines IL-10) were detected. Moreover, M2 macrophages were measured to illuminate the mechanisms of Sj16. RESULTS The rSj16 protein and Sj16 peptide had significant protective effects in S. japonicum-infected mice, as shown by decreased granuloma formation, areas of collagen deposition and inhibition of pro-inflammatory Th1, Th2 and Th17 cytokine production. These protective activities were more obvious when animals were treated with either the Sj16 protein or peptide at early stages post-infection. Interestingly, the combined treatment of PZQ and Sj16 was more effective and upregulated IL-10 production than administration of PZQ alone in infected mice. Furthermore, the Sj16 treatment alleviated the pathological effects associated with activated M2 macrophages. CONCLUSIONS This study demonstrates the anti-inflammatory and anti-fibrotic effects of rSj16 in schistosomiasis. Therefore, the combination of rSj16 with PZQ could be a viable and promising therapeutic strategy for schistosomiasis. In addition, this investigation provides additional information on schistosome-mediated immune modulation and host-parasite interactions.
Collapse
Affiliation(s)
- Jia Shen
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, 510080, Guangdong, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Lifu Wang
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, 510080, Guangdong, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Mei Peng
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, 510080, Guangdong, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Zhen Liu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, 510080, Guangdong, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Beibei Zhang
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, 510080, Guangdong, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Tao Zhou
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, 510080, Guangdong, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China
| | - Xi Sun
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China. .,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, 510080, Guangdong, China. .,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China.
| | - Zhongdao Wu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China. .,Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, 510080, Guangdong, China. .,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
33
|
Sun S, Li H, Yuan Y, Wang L, He W, Xie H, Gao S, Cheng R, Qian H, Jiang H, Wang X, Zhan B, Fang Q, Yang X. Preventive and therapeutic effects of Trichinella spiralis adult extracts on allergic inflammation in an experimental asthma mouse model. Parasit Vectors 2019; 12:326. [PMID: 31253164 PMCID: PMC6599242 DOI: 10.1186/s13071-019-3561-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 06/09/2019] [Indexed: 01/09/2023] Open
Abstract
Background Helminths immunomodulate the host immune system by secreting proteins to create an inhibitory environment as a strategy for survival in the host. As a bystander effect, this balances the host immune system to reduce hypersensitivity to allergens or autoantigens. Based on this, helminth therapy has been used to treat some allergic or autoimmune diseases. As a tissue-dwelling helminth, Trichinella spiralis infection has been identified to have strong immunomodulatory effects; the effective components in the worm have not yet been identified. Methods The soluble extracts of T. spiralis adult worms and muscle larvae were used to treat airway inflammation before and after an ovalbumin (OVA)-sensitization/challenge in an OVA-induced asthma mouse model. The therapeutic effects were observed by measuring the level of inflammation in the lungs. Results The soluble products derived from T. spiralis parasites, especially from adult worms, were able to ameliorate OVA-induced airway inflammatory responses which were associated with reduced eosinophil infiltration, OVA-specific IgE, Th2 cytokine IL-4, and increased IL-10 and TGF-β. The stimulation of the Treg response may contribute to the alleviated allergic inflammation. Conclusions Trichinella spiralis worm extracts stimulate regulatory cytokines that are associated with reduced allergic airway inflammation. The identification of effective components in the adult worm extracts will be a crucial approach for developing a novel therapeutic for allergic and autoimmune diseases.
Collapse
Affiliation(s)
- Siying Sun
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Huihui Li
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Yuan Yuan
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Liyuan Wang
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Wenxin He
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Hong Xie
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Shifang Gao
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Ruoxue Cheng
- Pharmacy College of Anhui Medical University, Hefei, 230001, China
| | - Haichun Qian
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Hui Jiang
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Xiaoli Wang
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China
| | - Bin Zhan
- Section of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Qiang Fang
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China. .,Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, 233030, China.
| | - Xiaodi Yang
- Department of Microbiology and Parasitology of Bengbu Medical College, Bengbu, 233000, China. .,Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, 233030, China.
| |
Collapse
|
34
|
Wolde M, Berhe N, Medhin G, Chala F, van Die I, Tsegaye A. Inverse Associations of Schistosoma mansoni Infection and Metabolic Syndromes in Humans: A Cross-Sectional Study in Northeast Ethiopia. Microbiol Insights 2019; 12:1178636119849934. [PMID: 31205419 PMCID: PMC6537292 DOI: 10.1177/1178636119849934] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 02/07/2019] [Indexed: 01/02/2023] Open
Abstract
Background: Recent animal and retrospective human studies have demonstrated that
Schistosoma mansoni infection may have potential to
protect against development of metabolic syndromes. Thus, the aim of this
study was to assess metabolic panel among S. mansoni egg
positives and egg negatives in stool examinations. This study was a
cross-sectional study, conducted involving 120 participants from S.
mansoni endemic town (Kemise) and 61 from non-endemic town
(Kombolcha), Northeast Ethiopia. Stool samples were collected and examined
for S. mansoni and other helminths using Kato-Katz method.
Furthermore, blood samples were collected and used for determination of
blood sugar, lipid profile tests, insulin, and C-reactive protein. Data were
analyzed using SPSS software version 20. Chi-square test, independent mean
t-test, and logistic regression models were employed on data.
P values less than .05 were considered as statistically
significant. Results: S. mansoni infected participants (n = 41; all from Kemise)
had significantly lower levels of fasting blood sugar, low prevalence of
dyslipidemia (at least one or more abnormal lipid profile tests; total
cholesterol, low-density lipoprotein cholesterol [LDL-C], high-density
lipoprotein cholesterol [HDL-C], and triglycerides) as compared with
controls (n = 79 in Kemise and 61 in Kombolcha). Moreover, logistic
regression model indicated that with the adjusted odds ratios, there was
significant inverse association between S. mansoni
infection and impaired fasting glucose (adjusted odds ratio −0.181, 95%
confidence interval: 0.042-0.774). Conclusions: Low fasting blood sugar and reduced prevalence of dyslipidemia in S.
mansoni egg positive participants might suggest inverse
association of S. mansoni infection and development of
metabolic syndromes. Furthermore, large-scale studies are recommended to
assess the role of S. mansoni egg and/or worm antigens in
modulating the host metabolic profile and reducing the risk of metabolic
syndromes, including diabetes mellitus and cardiovascular diseases.
Collapse
Affiliation(s)
- Mistire Wolde
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia.,Department of Medical Laboratory Sciences, College of Health Science, Addis Ababa University, Addis Ababa, Ethiopia
| | - Nega Berhe
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia.,Centre for Imported and Tropical Diseases, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Girmay Medhin
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Feyissa Chala
- Ethiopian Public Health Institute (EPHI), Addis Ababa, Ethiopia
| | - Irma van Die
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Aster Tsegaye
- Department of Medical Laboratory Sciences, College of Health Science, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
35
|
Øverli Ø, Johansen IB. Kindness to the Final Host and Vice Versa: A Trend for Parasites Providing Easy Prey? Front Ecol Evol 2019. [DOI: 10.3389/fevo.2019.00050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
36
|
Wang X, Fu Q, Song R, Duan B, Bergquist R, Xu J, Li S, Zhou D, Qin Z. Antinuclear antibodies and interleukin responses in patients with Schistosoma japonicum infection. Parasite Immunol 2018; 40:e12577. [PMID: 30074250 DOI: 10.1111/pim.12577] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 07/10/2018] [Accepted: 07/30/2018] [Indexed: 01/12/2023]
Abstract
Schistosomiasis poses a serious threat to public health, and the infection will develop into chronic and advanced late-stage disease if not treated. Apart from the clinical signs due to immune reactions to schistosome eggs trapped in host tissues, it also increases the risk for the development of autoimmunity reflected by dysfunctional, auto-reactive antibodies. Antinuclear antibodies (ANA) have been reported in schistosomiasis due to S. mansoni and S. haematobium. We demonstrate ANA in schistosomiasis japonica and explore the relationship between this infection and autoimmune disease by measuring ANA and interleukin (IL)-10, IL-12 and IL-17 responses in the sera of 125 Chinese patients with different stages of schistosomiasis japonica. The incidence rates of ANA in the patients with acute, chronic and late stages of schistosomiasis infection were 6.7%, 23.3% and 70.0%, respectively, with statistically significant differences between each stage (P = 0.000). IL-17 concentrations were high at the acute stage of schistosomiasis compared to the other stages of the disease (P = 0.000). This pattern was also seen for IL-10 and IL-12 concentrations (P = 0.01). IL concentrations in patients in the chronic and late stages of the disease were low and showed no difference compared to the healthy adults.
Collapse
Affiliation(s)
- Xiang Wang
- Institute of Biology and Medical Sciences, Soochow University, Suzhou, China.,Key Laboratory of Molecular Virology & Immunology, Vaccine Research Center, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Qiong Fu
- RenjiHospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Song
- RenjiHospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bowen Duan
- Key Laboratory of Molecular Virology & Immunology, Vaccine Research Center, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | | | - Jing Xu
- Key Laboratory of Parasite and Vector Biology, Ministry of Health, National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
| | - Shizhu Li
- Key Laboratory of Parasite and Vector Biology, Ministry of Health, National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
| | - Dongming Zhou
- Key Laboratory of Molecular Virology & Immunology, Vaccine Research Center, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Zhiqiang Qin
- Key Laboratory of Parasite and Vector Biology, Ministry of Health, National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
| |
Collapse
|
37
|
Abstract
In this Review, I present evidence supporting a multifactorial causation of childhood acute lymphoblastic leukaemia (ALL), a major subtype of paediatric cancer. ALL evolves in two discrete steps. First, in utero initiation by fusion gene formation or hyperdiploidy generates a covert, pre-leukaemic clone. Second, in a small fraction of these cases, the postnatal acquisition of secondary genetic changes (primarily V(D)J recombination-activating protein (RAG) and activation-induced cytidine deaminase (AID)-driven copy number alterations in the case of ETS translocation variant 6 (ETV6)-runt-related transcription factor 1 (RUNX1)+ ALL) drives conversion to overt leukaemia. Epidemiological and modelling studies endorse a dual role for common infections. Microbial exposures earlier in life are protective but, in their absence, later infections trigger the critical secondary mutations. Risk is further modified by inherited genetics, chance and, probably, diet. Childhood ALL can be viewed as a paradoxical consequence of progress in modern societies, where behavioural changes have restrained early microbial exposure. This engenders an evolutionary mismatch between historical adaptations of the immune system and contemporary lifestyles. Childhood ALL may be a preventable cancer.
Collapse
Affiliation(s)
- Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
| |
Collapse
|
38
|
Zhou S, Qi Q, Wang X, Zhang L, Xu L, Dong L, Zhu J, Li Y, Wang X, Xu Z, Liu F, Hu W, Zhou L, Chen X, Su C. SjHSP60 induces CD4 + CD25 + Foxp3 + Tregs via TLR4-Mal-drived production of TGF-β in macrophages. Immunol Cell Biol 2018; 96:958-968. [PMID: 29697865 DOI: 10.1111/imcb.12160] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 03/25/2018] [Accepted: 04/18/2018] [Indexed: 12/21/2022]
Abstract
CD4+ CD25+ Foxp3+ regulatory T cells (Tregs) play a pivotal role in limiting immunopathological damage to host organs after schistosome infection. Transforming growth factor-β (TGF-β) is an essential factor for the periphery conversion of CD4+ CD25- T cells into CD4+ CD25+ Foxp3+ Tregs by inducing the key transcription factor Foxp3. Antigen presenting cells (APCs), which highly express TGF-β, are involved in parasite antigen-induced Treg conversion in peripheral. However, the mechanisms underlying high TGF-β induction in APCs by parasite antigens remain to be clarified during schistosome infection. Here, we demonstrated that Schistosoma japonicum stress protein, heat shock protein 60 (SjHSP60), promoted TGF-β production in macrophages (Mφ). Furthermore, we showed that activation of TLR4-Mal (MyD88 adaptor-like protein) signaling by SjHSP60 is necessary for induction of TGF-β expression in Mφ, which subsequently promoted Treg induction. Our results not only demonstrate a novel mechanism of TGF-β production in Mφ for inducing Tregs in mice with schistosomiasis, but also allude to the possibility of targeting parasite stress protein for potential therapeutics.
Collapse
Affiliation(s)
- Sha Zhou
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qianqian Qi
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaofan Wang
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lina Zhang
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Xu
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liyang Dong
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jifeng Zhu
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yalin Li
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuefeng Wang
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhipeng Xu
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Feng Liu
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei Hu
- Department of Microbiology and Microbial Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Liang Zhou
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL, USA
| | - Xiaojun Chen
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chuan Su
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
39
|
Schwartz C, Hams E, Fallon PG. Helminth Modulation of Lung Inflammation. Trends Parasitol 2018; 34:388-403. [PMID: 29339033 DOI: 10.1016/j.pt.2017.12.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/11/2017] [Accepted: 12/18/2017] [Indexed: 12/21/2022]
Abstract
Parasitic helminths must establish chronic infections to complete their life cycle and therefore are potent modulators of multiple facets of host physiology. Parasitic helminths have coevolved with humans to become arguably master selectors of our immune system, whereby they have impacted on the selection of genes with beneficial mutations for both host and parasite. While helminth infections of humans are a significant health burden, studies have shown that helminths or helminth products can alter susceptibility to unrelated infectious or inflammatory diseases. This has generated interest in the use of helminth infections or molecules as therapeutics. In this review, we focus on the impact of helminth infections on pulmonary immunity, especially with regard to homeostatic lung function, pulmonary viral and bacterial (co)infections, and asthma.
Collapse
Affiliation(s)
- Christian Schwartz
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| | - Emily Hams
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Padraic G Fallon
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
40
|
Karaoglan M, Eksi F. The Coincidence of Newly Diagnosed Type 1 Diabetes Mellitus with IgM Antibody Positivity to Enteroviruses and Respiratory Tract Viruses. J Diabetes Res 2018; 2018:8475341. [PMID: 30186878 PMCID: PMC6116462 DOI: 10.1155/2018/8475341] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/01/2018] [Accepted: 07/24/2018] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Viruses trigger and promote islet cell destruction and cause type 1 diabetes mellitus (T1DM). However, the existence of a cause-and-effect relationship is under debate. The aim of this study is to investigate the sero-epidemiological and molecular evidence on enteroviruses and respiratory viruses in patients with newly diagnosed T1DM during the cold season. DESIGN Forty children newly diagnosed with T1DM and 30 healthy children who presented to the clinic over the course of a year were included in the study. The IgM antibodies against enteroviruses and respiratory viruses were studied using the indirect immunofluorescence assay (IFA) test, and no CBV4-specific RNA was detected in the children. The onset times of T1DM were classified into fall-winter and spring-summer seasons and separated into cold, moderate, or warm months in terms of temperature. RESULTS The percentages of viral IgM antibodies against most common viruses were detected in the patients as follows: influenza B (IVB) (70%), echovirus 7 (ECHO7) (45%), parainfluenza virus 4 (PIV4) (40%), coxsackievirus A7 (CAV7) (27.5%), and H3N2 (22.5%). Compared with the control group, the above viruses had a significant association with T1DM (p ≤ 0.001, p ≤ 0.001, p = 0.035, p = 0.003, and p = 0.023, resp.). CBV4-specific RNA was not detected in any serum. A total of 75% and 95% patients were diagnosed with T1DM in the fall-winter seasons and cold-moderate months, respectively. CONCLUSION Our study demonstrates the significant association between T1DM and the presence of IgM antibodies against IVB, ECHO7, PIV4, CAV7, and H3N2, and the majority of newly diagnosed T1DM appeared in the fall-winter season. It suggests that enteroviruses and respiratory viruses, in addition to seasonal variation, could play a role in the etiopathogenesis and clinical onset of T1DM.
Collapse
Affiliation(s)
- Murat Karaoglan
- Division of Pediatric Endocrinology, Gaziantep University Faculty of Medicine, Gaziantep, Turkey
| | - Fahriye Eksi
- Department of Medical Microbiology, Gaziantep University Faculty of Medicine, Gaziantep, Turkey
| |
Collapse
|
41
|
Lopes DM, de Almeida TVVS, de Souza RDP, Ribeiro LEV, Page B, Fernandes JDS, Carvalho EM, Cardoso LS. Susceptibility of dendritic cells from individuals with schistosomiasis to infection by Leishmania braziliensis. Mol Immunol 2017; 93:173-183. [PMID: 29197260 DOI: 10.1016/j.molimm.2017.11.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/09/2017] [Accepted: 11/18/2017] [Indexed: 12/18/2022]
Abstract
Coinfection with leishmaniasis and schistosomiasis has been associated with increased time to healing of cutaneous lesions of leishmaniasis. The objective of this study was to evaluate the effect of Leishmania braziliensis infection on co-cultures of monocyte-derived dendritic cells (MoDCs) with autologous lymphocytes from patients with schistosomiasis and patients with cutaneous leishmaniasis. MoDCs were differentiated from peripheral blood monocytes, isolated by magnetic beads, infected with L. braziliensis, and co-cultured with autologous lymphocytes. Expression of HLA-DR, CD1a, CD83, CD80, CD86, CD40, and the IL-10 receptor (IL-10R) on MoDCs as well as CD28, CD40L, CD25, and CTLA-4 on lymphocytes were evaluated by flow cytometry. The production of the cytokines IL-10, TNF, IL-12p40, and IFN-γ were evaluated by sandwich ELISA of the culture supernatant. The infectivity evaluation was performed by light microscopy after concentration of cells by cytospin and Giemsa staining. It was observed that the frequency of MoDCs expressing CD83, CD80, and CD86 as well as the MFI of HLA-DR were smaller in the group of patients with schistosomiasis compared to the group of patients with leishmaniasis. On the other hand, the frequency of IL-10R on MoDCs was higher in patients with schistosomiasis than in patients with leishmaniasis. CD4+ and CD8+ T lymphocytes from patients with schistosomiasis presented a lower frequency of CD28 and a higher frequency of CTLA-4 compared to lymphocytes from patients with leishmaniasis. Levels of IL-10 were higher in the supernatants of co-cultures from individuals with schistosomiasis compared to those with leishmaniasis. However, levels of TNF, IL-12p40, and IFN-γ were lower in the group of individuals with schistosomiasis. Regarding the frequency of MoDCs infected by L. braziliensis after 72h in culture, it was observed that higher frequencies of cells from patients with schistosomiasis were infected compared to cells from patients with leishmaniasis. It was concluded that MoDCs from patients with schistosomiasis are more likely to be infected by L. braziliensis, possibly due to a lower degree of activation and a regulatory profile.
Collapse
Affiliation(s)
- Diego Mota Lopes
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT- DT) -CNPQ/MCT, Brazil
| | - Tarcísio Vila Verde S de Almeida
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil; Escola Bahiana de Medicina e Saúde Pública, Salvador, Bahia, Brazil
| | - Robson da Paixão de Souza
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Luís Eduardo Viana Ribeiro
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Brady Page
- Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Edgar M Carvalho
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT- DT) -CNPQ/MCT, Brazil; Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA, Brazil
| | - Luciana Santos Cardoso
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT- DT) -CNPQ/MCT, Brazil; Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, UFBA, Salvador, Bahia, Brazil.
| |
Collapse
|
42
|
He L, Zhou S, Qi Q, Chi Y, Zhu J, Xu Z, Wang X, Hoellwarth J, Liu F, Chen X, Su C. The regulation of regulation: interleukin-10 increases CD4 + CD25 + regulatory T cells but impairs their immunosuppressive activity in murine models with schistosomiasis japonica or asthma. Immunology 2017; 153:84-96. [PMID: 28799262 DOI: 10.1111/imm.12813] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 08/02/2017] [Accepted: 08/04/2017] [Indexed: 12/13/2022] Open
Abstract
CD4+ CD25+ Foxp3+ regulatory T (Treg) cells play an important role in maintaining immune homeostasis. Interleukin-10 (IL-10), a cytokine with anti-inflammatory capacities, also has a critical role in controlling immune responses. In addition, it is well known that production of IL-10 is one of the suppression mechanisms of Treg cells. However, the action of IL-10 on Treg cells themselves remains insufficiently understood. In this study, by using a Schistosoma japonicum-infected murine model, we show that the elevated IL-10 contributed to Treg cell induction but impaired their immunosuppressive function. Our investigations further suggest that this may relate to the up-regulation of serum transforming growth factor (TGF-β) level but the decrease in membrane-bound TGF-β of Treg cells by IL-10 during S. japonicum infection. In addition, similar IL-10-mediated regulation on Treg cells was also confirmed in the murine model of asthma. In general, our findings identify a previously unrecognized opposing regulation of IL-10 on Treg cells and provide a deep insight into the precise regulation in immune responses.
Collapse
Affiliation(s)
- Lei He
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Province Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Sha Zhou
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Province Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qianqian Qi
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Province Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ying Chi
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Province Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jifeng Zhu
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Province Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhipeng Xu
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Province Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuefeng Wang
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Province Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jason Hoellwarth
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Feng Liu
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Province Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaojun Chen
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Province Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chuan Su
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Province Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
43
|
Amezcua L. MS in self-identified Hispanic/Latino individuals living in the US. Mult Scler J Exp Transl Clin 2017; 3:2055217317725103. [PMID: 28979795 PMCID: PMC5617095 DOI: 10.1177/2055217317725103] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 07/07/2017] [Indexed: 12/26/2022] Open
Abstract
Self-identified Hispanic/Latino individuals living with multiple sclerosis (MS) in the continental United States (US) are a diverse group that represents different cultural and ancestral backgrounds. A marked variability in the way MS affects various subgroups of Hispanics in the US has been observed. We reviewed and synthesized available data about MS in Hispanics in the US. There are likely a host of multifactorial elements contributing to these observations that could be explained by genetic, environmental, and social underpinnings. Barriers to adequate MS care in Hispanics are likely to include delivery of culturally competent care and social and economic disadvantages. Considerable efforts, including the formation of a national consortium known as the Alliance for Research in Hispanic Multiple Sclerosis (ARHMS), are underway to help further explore these various factors.
Collapse
Affiliation(s)
- Lilyana Amezcua
- Department of Neurology, University of Southern California, Keck School of Medicine, USA
| |
Collapse
|
44
|
Chen L, Zhang D, Zhang W, Zhu Y, Hou M, Yang B, Xu Z, Ji M, Wu G. Absence of Batf3 results in reduced liver pathology in mice infected with Schistosoma japonicum. Parasit Vectors 2017. [PMID: 28646891 PMCID: PMC5483257 DOI: 10.1186/s13071-017-2250-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background The involvement of CD8+T cells in schistosomiasis is being increasingly appreciated, but the underlying mechanism is not well defined. Results In this study, we showed that the absence of Batf3 alleviated liver damage in Batf3−/− mice infected with S. japonicum. We found alleviated liver granulomatous inflammation in Batf3−/− mice with schistosomiasis japonica could not be attributed to the difference in schistosome egg or worm burden. The stronger Tc1 cell responses observed in Batf3−/− mice suggested that the deletion of Batf3 resulted in more activation of CD8+T cells unexpectedly during the natural infection of schistosomes. We detected a small amount of CD8α+ DCs in the spleen of Batf3−/− mice at 9w post-infection. This small amount of newly generated CD8α+ DCs might contribute to enhanced activation of CD8+T cells via cross-presentation and activation which then attenuate hepatic pathological damage found in Batf3−/− mice. Conclusions Our study provides evidence that Batf3 is associated with the immunoregulation of the liver granuloma formation, which may confer a new options for schistosomiasis treatment.
Collapse
Affiliation(s)
- Lin Chen
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China. .,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China.
| | - Donghui Zhang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China.,School of International Education, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenyue Zhang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Yuxiao Zhu
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| | - Min Hou
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| | - Bingya Yang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| | - Zhipeng Xu
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| | - Minjun Ji
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| | - Guanling Wu
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| |
Collapse
|
45
|
Zhou CL, Kong DL, Liu JF, Lu ZK, Guo HF, Wang W, Qiu JF, Liu XJ, Wang Y. MHC II -, but not MHC II +, hepatic Stellate cells contribute to liver fibrosis of mice in infection with Schistosoma japonicum. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1848-1857. [PMID: 28483578 DOI: 10.1016/j.bbadis.2017.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 04/12/2017] [Accepted: 05/04/2017] [Indexed: 01/18/2023]
Abstract
Hepatic stellate cells (HSCs) are considered as the main effector cells in vitamin A metabolism and liver fibrosis, as well as in hepatic immune regulation. Recently, researches have revealed that HSCs have plasticity and heterogeneity, which depend on their lobular location and whether liver is normal or injured. This research aimed to explore the biological characteristics and heterogeneity of HSCs in mice with Schistosoma japonicum (S. japonicum) infection, and determine the subpopulation of HSCs in pathogenesis of hepatic fibrosis caused by S. japonicum infection. Results revealed that HSCs significantly increased the expressions of MHC II and fibrogenic genes after S. japonicum infection, and could be classified into MHC II+ HSCs and MHC II- HSCs subsets. Both two HSCs populations suppressed the proliferation of activated CD4+T cells, whereas only MHC II- HSCs displayed a myofibroblast-like phenotype. In response to IFN-γ, HSCs up-regulated the expressions of MHC II and CIITA, while down-regulated the expression of fibrogenic gene Col1. In addition, praziquantel treatment decreased the expressions of fibrogenic genes in MHC II- HSCs. These results confirmed that HSCs from S. japonicum-infected mice have heterogeneity. The MHC II- α-SMA+ HSCs were major subsets of HSCs contributing to liver fibrosis and could be considered as a potential target of praziquantel anti-fibrosis treatment.
Collapse
Affiliation(s)
- Chun-Lei Zhou
- Department of Pathogen Biology, Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - De-Long Kong
- Department of Pathogen Biology, Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - Jin-Feng Liu
- Department of Pathogen Biology, Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - Zhong-Kui Lu
- Department of Pathogen Biology, Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - Hong-Fei Guo
- Department of Pathogen Biology, Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - Wei Wang
- Department of Pathogen Biology, Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - Jing-Fan Qiu
- Department of Pathogen Biology, Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - Xin-Jian Liu
- Department of Pathogen Biology, Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China
| | - Yong Wang
- Department of Pathogen Biology, Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, Jiangsu 211166, PR China.
| |
Collapse
|
46
|
Yu HS, Park MK, Kang SA, Cho KS, Mun SJ, Roh HJ. Culture supernatant of adipose stem cells can ameliorate allergic airway inflammation via recruitment of CD4 +CD25 +Foxp3 T cells. Stem Cell Res Ther 2017; 8:8. [PMID: 28114970 PMCID: PMC5259897 DOI: 10.1186/s13287-016-0462-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 10/19/2016] [Accepted: 12/17/2016] [Indexed: 12/16/2022] Open
Abstract
Background In a previous study, we demonstrated that intravenous administration of adipose tissue stem cells (ASCs) could significantly reduce allergic symptoms and suppress eosinophilic inflammation. Methods To evaluate the secretome of ASCs, we administrated culture supernatant of ASCs (ASC sup, which contains the ASC secretome) and uncultured fresh medium (con sup) into a mouse model of allergic airway inflammation. Subsequently we observed the mice for signs of inflammation and investigated Th1-, Th2-, and Treg-related cytokine levels as well as recruitment of Treg cells into the airway. Results We found that ASC sup could ameliorate allergic airway inflammation in this model; the value of airway hyperresponsiveness, and the occurrence of inflammatory cell infiltration in the lung, as well as the number of eosinophils, and goblet cells in the lung epithelium were all significantly decreased by ASC sup treatment. In addition, ASC sup treatment significantly decreased the levels of IL-4, IL-5, and IL-13 in the bronchial alveolar lavage fluid and in culture medium of lung-draining lymph node cells of the animal model of acute asthma. We detected numerous CTLA-4 and Foxp3-expressing cells in the lung after ASC sup treatment. ASC sup was found to have a higher concentration of IL-10 and TGF-β compared to con sup. Conclusions Stem cells have powerful potential for therapeutic functions in various diseases, but they also have many drawbacks. In this study, we found strong immunosuppressive ability of ASC sup in an allergic airway mouse model. It may be possible to use ASC sup for treatment of many immunological diseases in the near future. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0462-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hak Sun Yu
- Department of Parasitology, Pusan National University School of Medicine, Yangsan, 626-870, Republic of Korea.,Immunoregulatory Therapeutics Group in Brain Busan 21 project, Yangsan, Republic of Korea
| | - Mi-Kyung Park
- Department of Parasitology, Pusan National University School of Medicine, Yangsan, 626-870, Republic of Korea.,Immunoregulatory Therapeutics Group in Brain Busan 21 project, Yangsan, Republic of Korea
| | - Shin Ae Kang
- Department of Parasitology, Pusan National University School of Medicine, Yangsan, 626-870, Republic of Korea.,Immunoregulatory Therapeutics Group in Brain Busan 21 project, Yangsan, Republic of Korea
| | - Kyu-Sup Cho
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University Hospital, Busan, 602-739, Republic of Korea
| | - Sue Jean Mun
- Department of Otorhinolaryngology and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Beomeo-li, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do, Yangsan, 626-770, Republic of Korea
| | - Hwan-Jung Roh
- Department of Otorhinolaryngology and Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Beomeo-li, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do, Yangsan, 626-770, Republic of Korea.
| |
Collapse
|
47
|
Kim TS, Pak JH, Kim JB, Bahk YY. Clonorchis sinensis, an oriental liver fluke, as a human biological agent of cholangiocarcinoma: a brief review. BMB Rep 2017; 49:590-597. [PMID: 27418285 PMCID: PMC5346318 DOI: 10.5483/bmbrep.2016.49.11.109] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Indexed: 01/11/2023] Open
Abstract
Parasitic diseases remain an unarguable public health problem worldwide. Liver fluke Clonorchis sinensis is a high risk pathogenic parasitic helminth which is endemic predominantly in Asian countries, including Korea, China, Taiwan, Vietnam, and the far eastern parts of Russia, and is still actively transmitted. According to the earlier 8th National Survey on the Prevalence of Intestinal Parasitic Infections in 2012, C. sinensis was revealed as the parasite with highest prevalence of 1.86% in general population among all parasite species surveyed in Korea. This fluke is now classified under one of the definite Group 1 human biological agents (carcinogens) by International Agency of Research on Cancer (IARC) along with two other parasites, Opisthorchis viverrini and Schistosoma haematobium. C. sinensis infestation is mainly linked to liver and biliary disorders, especially cholangiocarcinoma (CCA). For the purposes of this mini-review, we will only focus on C. sinensis and review pathogenesis and carcinogenesis of clonorchiasis, disease condition by C. sinensis infestation, and association between C. sinensis infestation and CCA. In this presentation, we briefly consider the current scientific status for progression of CCA by heavy C. sinensis infestation from the food-borne trematode and development of CCA.
Collapse
Affiliation(s)
- Tong-Soo Kim
- Department of Parasitology and Tropical Medicine, School of Medicine, Inha University, Incheon 22212, Korea
| | - Jhang Ho Pak
- Department of Convergence Medicine, College of Medicine, University of Ulsan, Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea
| | - Jong-Bo Kim
- Department of Biotechnology, Konkuk University, Chungju 27478, Korea
| | - Young Yil Bahk
- Department of Biotechnology, Konkuk University, Chungju 27478, Korea
| |
Collapse
|
48
|
Wang X, Li L, Wang J, Dong L, Shu Y, Liang Y, Shi L, Xu C, Zhou Y, Wang Y, Chen D, Mao C. Inhibition of cytokine response to TLR stimulation and alleviation of collagen-induced arthritis in mice by Schistosoma japonicum peptide SJMHE1. J Cell Mol Med 2016; 21:475-486. [PMID: 27677654 PMCID: PMC5323857 DOI: 10.1111/jcmm.12991] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 08/18/2016] [Indexed: 12/28/2022] Open
Abstract
Helminth‐derived products have recently been shown to prevent the development of inflammatory diseases in mouse models. However, most identified immunomodulators from helminthes are mixtures or macromolecules with potentially immunogenic side effects. We previously identified an immunomodulatory peptide called SJMHE1 from the HSP60 protein of Schistosoma japonicum. In this study, we assessed the ability of SJMHE1 to affect murine splenocytes and human peripheral blood mononuclear cells (PBMCs) stimulated by toll‐like receptor (TLR) ligands in vitro and its treatment effect on mice with collagen‐induced arthritis (CIA). We show that SJMHE1 not only modulates the cytokine production of murine macrophage (MΦ) and dendritic cell but also affects cytokine production upon coculturing with allogeneic CD4+ T cell. SJMHE1 potently inhibits the cytokine response to TLR ligands lipopolysaccharide (LPS), CpG oligodeoxynucleotides (CpG) or resiquimod (R848) from mouse splenocytes, and human PBMCs stimulated by LPS. Furthermore, SJMHE1 suppressed clinical signs of CIA in mice and blocked joint erosion progression. This effect was mediated by downregulation of key cytokines involved in the pathogenesis of CIA, such as interferon‐γ (IFN‐γ), tumour necrosis factor‐α (TNF‐α), interleukin (IL)‐6, IL‐17, and IL‐22 and up‐regulation of the inhibitory cytokine IL‐10, Tgf‐β1 mRNA, and CD4+CD25+Foxp3+ Tregs. This study provides new evidence that the peptide from S. japonicum, which is the ‘safe’ selective generation of small molecule peptide that has evolved during host–parasite interactions, is of great value in the search for novel anti‐inflammatory agents and therapeutic targets for autoimmune diseases.
Collapse
Affiliation(s)
- Xuefeng Wang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Li Li
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jun Wang
- Department of Nuclear Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Liyang Dong
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yang Shu
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yong Liang
- Clinical Laboratory, Huai'an Hospital Affiliated of Xuzhou Medical College, Huaian, Jiangsu, China
| | - Liang Shi
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Chengcheng Xu
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yuepeng Zhou
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yi Wang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Deyu Chen
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Chaoming Mao
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
49
|
Hablützel PI, Brown M, Friberg IM, Jackson JA. Changing expression of vertebrate immunity genes in an anthropogenic environment: a controlled experiment. BMC Evol Biol 2016; 16:175. [PMID: 27586387 PMCID: PMC5009682 DOI: 10.1186/s12862-016-0751-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 08/23/2016] [Indexed: 12/01/2022] Open
Abstract
Background The effect of anthropogenic environments on the function of the vertebrate immune system is a problem of general importance. For example, it relates to the increasing rates of immunologically-based disease in modern human populations and to the desirability of identifying optimal immune function in domesticated animals. Despite this importance, our present understanding is compromised by a deficit of experimental studies that make adequately matched comparisons between wild and captive vertebrates. Results We transferred post-larval fishes (three-spined sticklebacks), collected in the wild, to an anthropogenic (captive) environment. We then monitored, over 11 months, how the systemic expression of immunity genes changed in comparison to cohort-matched wild individuals in the originator population (total n = 299). We found that a range of innate (lyz, defbl2, il1r-like, tbk1) and adaptive (cd8a, igmh) immunity genes were up-regulated in captivity, accompanied by an increase in expression of the antioxidant enzyme, gpx4a. For some genes previously known to show seasonality in the wild, this appeared to be reduced in captive fishes. Captive fishes tended to express immunity genes, including igzh, foxp3b, lyz, defbl2, and il1r-like, more variably. Furthermore, although gene co-expression patterns (analyzed through gene-by-gene correlations and mutual information theory based networks) shared common structure in wild and captive fishes, there was also significant divergence. For one gene in particular, defbl2, high expression was associated with adverse health outcomes in captive fishes. Conclusion Taken together, these results demonstrate widespread regulatory changes in the immune system in captive populations, and that the expression of immunity genes is more constrained in the wild. An increase in constitutive systemic immune activity, such as we observed here, may alter the risk of immunopathology and contribute to variance in health in vertebrate populations exposed to anthropogenic environments. Electronic supplementary material The online version of this article (doi:10.1186/s12862-016-0751-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Martha Brown
- IBERS, Aberystwyth University, Aberystwyth, SY23 3DA, UK
| | - Ida M Friberg
- School of Life and Environmental Sciences, University of Salford, Salford, M5 4WT, UK
| | - Joseph A Jackson
- School of Life and Environmental Sciences, University of Salford, Salford, M5 4WT, UK.
| |
Collapse
|
50
|
Liu F, Cheng W, Pappoe F, Hu X, Wen H, Luo Q, Wang S, Deng F, Xie Y, Xu Y, Shen J. Schistosoma japonicum cystatin attenuates murine collagen-induced arthritis. Parasitol Res 2016; 115:3795-806. [PMID: 27393379 DOI: 10.1007/s00436-016-5140-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/18/2016] [Indexed: 01/09/2023]
Abstract
Recombinant SjCystatin (rSjCystatin), a recombinant protein of Schistosoma japonicum cystatin, has been reported to have an effect on immunoregulation mediated by IL-10 induction. Rheumatoid arthritis (RA) is a common autoimmune inflammatory arthropathy, and recombinant immune-modulating drugs for RA treatment are under development. We aimed to study the putative immune regulation of rSjCystatin and its prophylactic/therapeutic effects on murine collagen-induced arthritis (CIA). CIA was induced in DBA/1 mice by inoculation with bovine collagen II (CII). rSjCystatin was administered prior or post development of CIA. The severity of CIA was assessed using established clinical and histopathological scoring systems. The incidence was also determined. The CII-specific antibodies in sera and cytokines in splenocyte culture supernatants were measured by ELISA. Th1/Th2/Th17 cells and Tregs development in splenocytes were monitored by flow cytometry. The inflammatory mediators in the diseased joint were semiquantitated by qPCR. Prophylactic injection of rSjCystatin attenuated paw clinical scores, incidence, and histopathology scores of joints in CIA mice. The arthritis-alleviative effects were closely associated with the augmentation of IL-4, IL-10, and collagen-specific IgG1, and with the distinct reduction of IFN-γ, collagen-specific IgG2a, and the marked decrease of proinflammatory cytokines IL-6, IL-17, and TNF-α and RANKL. The data indicate that rSjCystatin may prevent cartilage destruction and inflammation of joints in CIA mice. The effects are related to the inhibitory modulation of Th1 and Th17 and upregulation of Tregs and Th2 via a shift of cytokines profiling from Th1 to Th2 response.
Collapse
Affiliation(s)
- Fang Liu
- Department of Pathogen Biology, Provincial Laboratory of Pathogen Biology and Key Laboratory of Zoonoses Anhui, Anhui Medical University, Hefei, 230022, China
| | - Weisheng Cheng
- Department of Pathogen Biology, Provincial Laboratory of Pathogen Biology and Key Laboratory of Zoonoses Anhui, Anhui Medical University, Hefei, 230022, China.,Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Faustina Pappoe
- Department of Pathogen Biology, Provincial Laboratory of Pathogen Biology and Key Laboratory of Zoonoses Anhui, Anhui Medical University, Hefei, 230022, China
| | - Xiaodong Hu
- Department of Pathogen Biology, Provincial Laboratory of Pathogen Biology and Key Laboratory of Zoonoses Anhui, Anhui Medical University, Hefei, 230022, China
| | - Huiqin Wen
- Department of Pathogen Biology, Provincial Laboratory of Pathogen Biology and Key Laboratory of Zoonoses Anhui, Anhui Medical University, Hefei, 230022, China.,Department of Blood Transfusion, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Qingli Luo
- Department of Pathogen Biology, Provincial Laboratory of Pathogen Biology and Key Laboratory of Zoonoses Anhui, Anhui Medical University, Hefei, 230022, China
| | - Shushu Wang
- Department of Pathogen Biology, Provincial Laboratory of Pathogen Biology and Key Laboratory of Zoonoses Anhui, Anhui Medical University, Hefei, 230022, China
| | - Fang Deng
- Department of Laboratory Diagnosis, the Provincial Tumor Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yuanyuan Xie
- Department of Pathogen Biology, Provincial Laboratory of Pathogen Biology and Key Laboratory of Zoonoses Anhui, Anhui Medical University, Hefei, 230022, China
| | - Yuanhong Xu
- Department of Pathogen Biology, Provincial Laboratory of Pathogen Biology and Key Laboratory of Zoonoses Anhui, Anhui Medical University, Hefei, 230022, China. .,Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China. .,Department of Pathogen Biology, Anhui Medical University, Hefei, 230022, China. .,Department of Clinical Laboratory Medicine, Anhui Medical University, Hefei, 230022, China.
| | - Jilong Shen
- Department of Pathogen Biology, Provincial Laboratory of Pathogen Biology and Key Laboratory of Zoonoses Anhui, Anhui Medical University, Hefei, 230022, China. .,Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China. .,Department of Pathogen Biology, Anhui Medical University, Hefei, 230022, China. .,Department of Clinical Laboratory Medicine, Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|