1
|
Zhang C, Wang H, Aji T, Li Z, Li Y, Ainiwaer A, Rousu Z, Li J, Wang M, Deng B, Duolikun A, Kang X, Zheng X, Yu Q, Shao Y, Zhang W, Vuitton DA, Tian Z, Sun H, Wen H. Targeting myeloid-derived suppressor cells promotes antiparasitic T-cell immunity and enhances the efficacy of PD-1 blockade (15 words). Nat Commun 2024; 15:6345. [PMID: 39068159 PMCID: PMC11283557 DOI: 10.1038/s41467-024-50754-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 07/18/2024] [Indexed: 07/30/2024] Open
Abstract
Immune exhaustion corresponds to a loss of effector function of T cells that associates with cancer or chronic infection. Here, our objective was to decipher the mechanisms involved in the immune suppression of myeloid-derived suppressor cells (MDSCs) and to explore the potential to target these cells for immunotherapy to enhance checkpoint blockade efficacy in a chronic parasite infection. We demonstrated that programmed cell-death-1 (PD-1) expression was significantly upregulated and associated with T-cell dysfunction in advanced alveolar echinococcosis (AE) patients and in Echinococcus multilocularis-infected mice. PD-1 blockade ex vivo failed to reverse AE patients' peripheral blood T-cell dysfunction. PD-1/PD-L1 blockade or PD-1 deficiency had no significant effects on metacestode in mouse model. This was due to the inhibitory capacities of immunosuppressive granulocytic MDSCs (G-MDSCs), especially in the liver surrounding the parasite pseudotumor. MDSCs suppressed T-cell function in vitro in an indoleamine 2, 3 dioxygenase 1 (IDO1)-dependent manner. Although depleting MDSCs alone restored T-cell effector functions and led to some limitation of disease progression in E. multilocularis-infected mice, combination with PD-1 blockade was better to induce antiparasitic efficacy. Our findings provide preclinical evidence in support of targeting MDSC or combining such an approach with checkpoint blockade in patients with advanced AE. (200 words).
Collapse
Affiliation(s)
- Chuanshan Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University; Clinical Medicine Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P. R. China.
- Basic Medical College, Xinjiang Medical University, Urumqi, Xinjiang, P. R. China.
- Key Laboratory of High Incidence Disease Research in Xingjiang, Ministry of Education, Xinjiang Medical University, Urumqi, Xinjiang, P. R. China.
| | - Hui Wang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University; Clinical Medicine Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
- Key Laboratory of High Incidence Disease Research in Xingjiang, Ministry of Education, Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
- Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Research Institute, The First Affiliated Hospital of Xinjiang Medical University, and WHO Collaborating Centre on Prevention and Case Management of Echinococcosis, Urumqi, Xinjiang, P. R. China
| | - Tuerganaili Aji
- Key Laboratory of High Incidence Disease Research in Xingjiang, Ministry of Education, Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
- Department of Hepatic Hydatid and Hepatobiliary Surgery, Digestive and Vascular Surgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
| | - Zhide Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University; Clinical Medicine Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
| | - Yinshi Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University; Clinical Medicine Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
- Basic Medical College, Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
| | - Abidan Ainiwaer
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University; Clinical Medicine Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
- Basic Medical College, Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
| | - Zibigu Rousu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University; Clinical Medicine Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
- Basic Medical College, Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
| | - Jing Li
- Basic Medical College, Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
| | - Maolin Wang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University; Clinical Medicine Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
- Department of Hepatic Hydatid and Hepatobiliary Surgery, Digestive and Vascular Surgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
| | - Bingqing Deng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University; Clinical Medicine Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
- Basic Medical College, Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
| | - Adilai Duolikun
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University; Clinical Medicine Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
- Basic Medical College, Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
| | - Xuejiao Kang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University; Clinical Medicine Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
- Basic Medical College, Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
| | - Xuran Zheng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University; Clinical Medicine Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
- Basic Medical College, Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
| | - Qian Yu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University; Clinical Medicine Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
- Basic Medical College, Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
| | - Yingmei Shao
- Department of Hepatic Hydatid and Hepatobiliary Surgery, Digestive and Vascular Surgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
| | - Wenbao Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University; Clinical Medicine Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P. R. China
- Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Research Institute, The First Affiliated Hospital of Xinjiang Medical University, and WHO Collaborating Centre on Prevention and Case Management of Echinococcosis, Urumqi, Xinjiang, P. R. China
| | - Dominique A Vuitton
- WHO-Collaborating Centre for the Prevention and Treatment of Human Echinococcosis, Department of Parasitology, University Bourgogne Franche-Comté (EA 3181) and University Hospital, Besançon, France
| | - Zhigang Tian
- Hefei National Research Center for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Haoyu Sun
- Hefei National Research Center for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China.
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
- Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, P. R. China.
| | - Hao Wen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University; Clinical Medicine Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, P. R. China.
| |
Collapse
|
2
|
Ye C, Zhang L, Tang L, Duan Y, Liu J, Zhou H. Host genetic backgrounds: the key to determining parasite-host adaptation. Front Cell Infect Microbiol 2023; 13:1228206. [PMID: 37637465 PMCID: PMC10449477 DOI: 10.3389/fcimb.2023.1228206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/20/2023] [Indexed: 08/29/2023] Open
Abstract
Parasitic diseases pose a significant threat to global public health, particularly in developing countries. Host genetic factors play a crucial role in determining susceptibility and resistance to infection. Recent advances in molecular and biological technologies have enabled significant breakthroughs in understanding the impact of host genes on parasite adaptation. In this comprehensive review, we analyze the host genetic factors that influence parasite adaptation, including hormones, nitric oxide, immune cells, cytokine gene polymorphisms, parasite-specific receptors, and metabolites. We also establish an interactive network to better illustrate the complex relationship between host genetic factors and parasite-host adaptation. Additionally, we discuss future directions and collaborative research priorities in the parasite-host adaptation field, including investigating the impact of host genes on the microbiome, developing more sophisticated models, identifying and characterizing parasite-specific receptors, utilizing patient-derived sera as diagnostic and therapeutic tools, and developing novel treatments and management strategies targeting specific host genetic factors. This review highlights the need for a comprehensive and systematic approach to investigating the underlying mechanisms of parasite-host adaptation, which requires interdisciplinary collaborations among biologists, geneticists, immunologists, and clinicians. By deepening our understanding of the complex interactions between host genetics and parasite adaptation, we can develop more effective and targeted interventions to prevent and treat parasitic diseases. Overall, this review provides a valuable resource for researchers and clinicians working in the parasitology field and offers insights into the future directions of this critical research area.
Collapse
Affiliation(s)
- Caixia Ye
- Clinical Medical Research Center, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- Department of Pediatrics, Yunyang Women and Children’s Hospital (Yunyang Maternal and Child Health Hospital), Chongqing, China
| | - Lianhua Zhang
- Clinical Medical Research Center, The Second Affiliated Hospital, Army Medical University, Chongqing, China
- Department of Surgery, Yunyang Women and Children’s Hospital (Yunyang Maternal and Child Health Hospital), Chongqing, China
| | - Lili Tang
- The 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Tumor Hospital), Urumqi, China
| | - Yongjun Duan
- Department of Pediatrics, Yunyang Women and Children’s Hospital (Yunyang Maternal and Child Health Hospital), Chongqing, China
| | - Ji Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Hongli Zhou
- Clinical Medical Research Center, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
3
|
Gottstein B, Deplazes P. Alveolar echinococcosis: what triggers emergence in North America, Central Europe and Asia? Curr Opin Infect Dis 2021; 34:440-446. [PMID: 34524197 DOI: 10.1097/qco.0000000000000765] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Infection with the larval (metacestode) stage of Echinococcus multilocularis causes alveolar echinococcosis (AE), a serious hepatic disorder. The parasite has increased its infection extensity in wildlife and domestic dogs, mainly due to urbanization and spatial extension of wildlife hosts in Europe, Asia as well as North America, resulting in emerging infection risk for humans. RECENT FINDINGS In hyperendemic areas such as Kyrgyzstan and China, ecological and socioeconomic changes have been associated with the unpredictable increase of AE cases. In North America, the appearance of the European-like genotype is of concern. In Europe, the annual increase of human case numbers reached a plateau even in hyperendemic situations. Therefore, we conclude that most of the exposed individuals are resistant to parasite invasion and/or to disease development. Thus, AE develops in a few healthy individuals, but preferentially in immunosuppressed patients. SUMMARY In the future, improved diagnostic strategies will allow more precise estimations of transmission routes including the role of food, water and direct dog contact, which should yield improved public health recommendations. Finally, understanding protective innate and acquired immune mechanisms as well as parasite-driven immune-evasion processes will be essential to develop curative therapies in nonoperable patients and, futuristically, appropriate vaccines.
Collapse
Affiliation(s)
- Bruno Gottstein
- Institute of Infectious Diseases, Faculty of Medicine, University of Bern, Bern
| | - Peter Deplazes
- Institute of Parasitology, Vetsuisse Faculty and Faculty of Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
4
|
Kandil A, Keles AG, Balci H, Demirci Tansel C. The Effects of Nitric Oxide and Inhibitor, and Combination of Albendazole and Praziquantel On Liver in Mice Injected with Echinococcus granulosus Larvae. Acta Trop 2021; 219:105917. [PMID: 33839085 DOI: 10.1016/j.actatropica.2021.105917] [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: 09/09/2020] [Revised: 03/17/2021] [Accepted: 04/01/2021] [Indexed: 12/12/2022]
Abstract
In this study, the role of nitric oxide (NO) in the pathogenesis of hydatidosis and the interaction with effects of anthelmintic drugs, albendazole and praziquantel, were examined in larval infection caused by protoscolices obtained from hydatid cysts of sheep liver in Albino Balb/c mice. Animals were divided into ten groups including controls and infected groups. Larval infection was established with intraperitoneal injection of protoscolices. Eight months after infection with protoscolices, the infected animals were divided into 6 groups. The infected animals were given a selective inhibitor of inducible nitric oxide synthase (iNOS) L-N6-(1-Iminoethyl) lysine-hydrochloride (L-NIL), NO donor sodium nitroprusside (SNP), albendazole and praziquantel as anthelmintic drugs for 7 days. In addition, control groups were composed of intact group, control, anthelmintic drugs + L-NIL, and anthelmintic drugs + SNP. The liver and blood samples were taken for cytological, histological, immunohistochemical and biochemical analyses 7 days after treatments at the end of experiment. The animals injected with protoscolices showed histopathological changes including inflammation areas, infiltration and accumulation of leukocytes, dilation of sinusoids, and damage in endothelial cells and hepatocytes at light microscopy. Electron microscopy were revealed severe damage in sinusoidal endothelial cells, leukocytes especially eosinophils in sinusoid lumens and disorganization in endoplasmic reticulum and nuclear membrane. Endothelial nitric oxide synthase (eNOS) and iNOS reactions were increased in the tissue. Anthelmintic drugs decreased inflammation areas and damages; however, it did not change NOS reactions in the animals given protoscolices. L-NIL and SNP diminished both iNOS and eNOS reactions. Unlike the group administered the inhibitor, SNP treated group exhibited less inflammation areas. Combination of these substances and drugs resulted in decreased inflammation areas. eNOS and iNOS reactions decreased in the drugs and SNP administered group, while only iNOS reaction was decreased in L-NIL given infection group. In addition, the infected groups which received SNP displayed expanded sinusoids and hepatocytes with vacuoles, intriguingly. While levels of serum nitrite/nitrate elevated only in the infection group given drugs and SNP, it decreased in the L-NIL administered group. Tissue level of malondialdehyde increased in infection groups with drugs and SNP. In conclusion, the results indicated that NO plays an important role in the pathogenesis of hydatidosis.
Collapse
|
5
|
Chou WK, Vaikunthan M, Schröder HV, Link AJ, Kim H, Brynildsen MP. Synergy Screening Identifies a Compound That Selectively Enhances the Antibacterial Activity of Nitric Oxide. Front Bioeng Biotechnol 2020; 8:1001. [PMID: 32984281 PMCID: PMC7477088 DOI: 10.3389/fbioe.2020.01001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 07/31/2020] [Indexed: 01/04/2023] Open
Abstract
Antibiotic resistance poses a serious threat to global health. To reinforce the anti-infective arsenal, many novel therapeutic strategies to fight bacterial infections are being explored. Among them, anti-virulence therapies, which target pathways important for virulence, have attracted much attention. Nitric oxide (NO) defense systems have been identified as critical for the pathogenesis of various bacteria, making them an appealing therapeutic target. In this study, we performed chemical screens to identify inhibitors of NO detoxification in Escherichia coli. We found that 2-mercaptobenzothiazole (2-MBT) can potently inhibit cellular detoxification of NO, achieving a level of inhibition that resembled the effect of genetically removing Hmp, the dominant detoxification enzyme under oxygenated conditions. Further analysis revealed that in the presence of NO, 2-MBT impaired the catalysis of Hmp and synthesis of Hmp and other proteins, whereas in its absence there were minimal perturbations to growth and protein synthesis. In addition, by studying the structure-activity relationship of 2-MBT, we found that both sulfur atoms in 2-MBT were vital for its inhibition of NO detoxification. Interestingly, when 2-mercaptothiazole (2-MT), which lacked the benzene ring, was used, differing biological activities were observed, although they too were NO dependent. Specifically, 2-MT could still prohibit NO detoxification, though it did not interfere with Hmp catalysis; rather, it was a stronger inhibitor of protein synthesis and it reduced the transcript levels of hmp, which was not observed with 2-MBT. Overall, these results provide a strong foundation for further exploration of 2-MBT and 2-MT for therapeutic applications.
Collapse
Affiliation(s)
- Wen Kang Chou
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, United States
| | - Mathini Vaikunthan
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, United States
| | - Hendrik V. Schröder
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, United States
| | - A. James Link
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, United States
- Frick Chemistry Laboratory, Department of Chemistry, Princeton University, Princeton, NJ, United States
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States
| | - Hahn Kim
- Frick Chemistry Laboratory, Department of Chemistry, Princeton University, Princeton, NJ, United States
- Small Molecule Screening Center, Princeton University, Princeton, NJ, United States
| | - Mark P. Brynildsen
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, United States
| |
Collapse
|
6
|
Rodrigues RM, Gonçalves ALR, Silva NM, Cardoso CRDB, Araújo NR, Coutinho LB, Alves R, Ueta MT, Costa-Cruz JM. Inducible nitric oxide synthase controls experimental Strongyloides infection. Parasite Immunol 2018; 40:e12576. [PMID: 30035318 DOI: 10.1111/pim.12576] [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: 02/17/2018] [Revised: 06/18/2018] [Accepted: 07/19/2018] [Indexed: 12/12/2022]
Abstract
Infection with Strongyloides sp. induces a host immune response, predominantly the Th2 type, that is able to eliminate the parasite. However, little is known about the role of the nitric oxide (NO) mediator, induced by the enzyme nitric oxide synthase (NOS), in strongyloidiasis. Therefore, in this study, we investigated the immune response of mice genetically deficient in the enzyme inducible nitric oxide synthase (iNOS-/- ), infected with Strongyloides venezuelensis. C57BL/6 wild-type (WT) and iNOS-/- mice were individually inoculated by subcutaneous injection of 3000 S. venezuelensis L3 larvae. In the absence of iNOS, mice were more susceptible to the infection than WT animals, in which the parasite was completely eliminated. The overall production of cytokines and specific IgG, IgG1 or IgE antibodies against the parasite was significantly lowered in infected iNOS-/- mice. The expression of iNOS was observed in the intestine of WT hosts but mainly in the wall of the parasite, despite the presence of iNOS in mice. Altogether, we concluded that iNOS expression may play an important role in the control of S. venezuelensis infection.
Collapse
Affiliation(s)
| | - Ana Lúcia Ribeiro Gonçalves
- Laboratório de Diagnóstico de Parasitoses, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Neide Maria Silva
- Laboratório de Imunopatologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Cristina Ribeiro de Barros Cardoso
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | | | - Loyane Bertagnolli Coutinho
- Laboratório de Imunopatologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Ronaldo Alves
- Laboratório de Diagnóstico de Parasitoses, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| | - Marlene Tiduko Ueta
- Departamento de Parasitologia, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - Julia Maria Costa-Cruz
- Laboratório de Diagnóstico de Parasitoses, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brazil
| |
Collapse
|
7
|
Díaz Á, Sagasti C, Casaravilla C. Granulomatous responses in larval taeniid infections. Parasite Immunol 2018. [DOI: 10.1111/pim.12523] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Á. Díaz
- Área/Cátedra de Inmunología; Departamento de Biociencias (Facultad de Química) e Instituto de Química Biológica (Facultad de Ciencias); Universidad de la República; Montevideo Uruguay
| | - C. Sagasti
- Área/Cátedra de Inmunología; Departamento de Biociencias (Facultad de Química) e Instituto de Química Biológica (Facultad de Ciencias); Universidad de la República; Montevideo Uruguay
| | - C. Casaravilla
- Área/Cátedra de Inmunología; Departamento de Biociencias (Facultad de Química) e Instituto de Química Biológica (Facultad de Ciencias); Universidad de la República; Montevideo Uruguay
| |
Collapse
|
8
|
Wang J, Goepfert C, Mueller N, Piersigilli A, Lin R, Wen H, Vuitton DA, Vuitton L, Mueller C, Gottstein B. Larval Echinococcus multilocularis infection reduces dextran sulphate sodium-induced colitis in mice by attenuating T helper type 1/type 17-mediated immune reactions. Immunology 2017; 154:76-88. [PMID: 29121394 PMCID: PMC5904711 DOI: 10.1111/imm.12860] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 10/27/2017] [Accepted: 10/29/2017] [Indexed: 12/13/2022] Open
Abstract
The tumour‐like growth of larval Echinococcus multilocularis tissue (causing alveolar echinococcosis, AE) is directly linked to the nature/orientation of the periparasitic host immune‐mediated processes. Parasite‐mediated immune suppression is a hallmark triggering infection outcome in both chronic human and murine AE. So far, little is known about secondary systemic immune effects of this pathogen on other concomitant diseases, e.g. endogenous gut inflammation. We examined the influence of E. multilocularis infection on murine dextran sodium sulphate (DSS) ‐induced colitis. At 3 months after E. multilocularis infection (chronic stage), the mice were challenged with 3% DSS in the drinking water for 5 days plus subsequently with tap water (alone) for another 4 days. After necropsy, fixed tissues/organs were sectioned and stained with haematoxylin & eosin for assessing inflammatory reactions. Cytokine levels were measured by flow cytometry and quantitative RT‐PCR. Colitis severity was assessed (by board‐certified veterinary pathologists) regarding (i) colon length, (ii) weight loss and (iii) a semi‐quantitative score of morphological changes. The histopathological analysis of the colon showed a significant reduction of DSS‐induced gut inflammation by concomitant E. multilocularis infection, which correlated with down‐regulation of T helper type 1 (Th1)/Th17 T‐cell responses in the colon tissue. Echinococcus multilocularis infection markedly reduced the severity of DSS‐induced gut inflammation upon down‐regulation of Th1/Th17 cytokine expression and attenuation of CD11b+ cell activation. In conclusion, E. multilocularis infection remarkably reduces DSS‐induced colitis in mice by attenuating Th1/Th17‐mediated immune reactions.
Collapse
Affiliation(s)
- Junhua Wang
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, Institute of Parasitology, University of Bern, Bern, Switzerland.,State Key Laboratory Incubation Base of Xinjiang Major Diseases Research (2010DS890294) and Xinjiang Key Laboratory of Echinococcosis, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Christine Goepfert
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, Institute of Animal Pathology, COMPATH, University of Bern, Bern, Switzerland
| | - Norbert Mueller
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, Institute of Parasitology, University of Bern, Bern, Switzerland
| | - Alessandra Piersigilli
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, Institute of Animal Pathology, COMPATH, University of Bern, Bern, Switzerland
| | - Renyong Lin
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research (2010DS890294) and Xinjiang Key Laboratory of Echinococcosis, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Hao Wen
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research (2010DS890294) and Xinjiang Key Laboratory of Echinococcosis, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Dominique A Vuitton
- WHO-Collaborating Centre on Prevention and Treatment of Human Echinococcosis and French National Reference Centre on Alveolar Echinococcosis, University of Franche-Comté and University Hospital, Besançon, France
| | - Lucine Vuitton
- WHO-Collaborating Centre on Prevention and Treatment of Human Echinococcosis and French National Reference Centre on Alveolar Echinococcosis, University of Franche-Comté and University Hospital, Besançon, France.,Gastroenterology and Digestive Endoscopy, University Hospital, Besançon, France
| | - Christoph Mueller
- Institute of Pathology, Medical Faculty, University of Bern, Bern, Switzerland
| | - Bruno Gottstein
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, Institute of Parasitology, University of Bern, Bern, Switzerland
| |
Collapse
|
9
|
Peón AN, Ledesma-Soto Y, Terrazas LI. Regulation of immunity by Taeniids: lessons from animal models and in vitro studies. Parasite Immunol 2016; 38:124-35. [PMID: 26457989 DOI: 10.1111/pim.12289] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 10/01/2015] [Indexed: 02/06/2023]
Abstract
Taeniidae is the largest family of the Cyclophyllidea order of parasites despite being composed of just two genera: Taenia spp and Echinococcus spp. These parasites are flatworms with a terrestrial life cycle, having an immature or larval stage called metacestode, which develops into the mature form within the intestine of the primary host after being consumed in raw or poorly cooked meat. Consumed eggs hatch into oncospheres, penetrate the intestinal walls and are transported via the bloodstream to later develop into metacestodes within the muscles and internal organs of secondary and sometimes primary hosts, thereby initiating the cycle again. Larval stages of both Taenia spp and Echinococcus spp are well known to produce tissue-dwelling, long-lasting infections; in this stage, these parasites can reach centimetres (macroparasites) and both genera may cause life-threatening diseases in humans. Establishing such long-term infections requires an exceptional ability to modulate host immunity for long periods of time. In this review, we analyse the immunoregulatory mechanisms induced by these tapeworms and their products, mainly discussing the importance of taeniid strategies to successfully colonize their hosts, such as antigen-presenting cell phenotype manipulation and the consequent induction of T-cell anergy, among others.
Collapse
Affiliation(s)
- A N Peón
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, México
| | - Y Ledesma-Soto
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, México
| | - L I Terrazas
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, México
| |
Collapse
|
10
|
Abstract
Cystic and alveolar echinococcosis are severe chronic helminthic diseases caused by the cystic growth or the intrahepatic tumour-like growth of the metacestode of Echinococcus granulosus or Echinococcus multilocularis, respectively. Both parasites have evolved sophisticated strategies to escape host immune responses, mainly by manipulating and directing this immune response towards anergy and/or tolerance. Recent research studies have revealed a number of respective immunoregulatory mechanisms related to macrophages and dendritic cell as well as T cell activities (regulatory T cells, Tregs). A better understanding of this complex parasite-host relationship, and the elucidation of specific crucial events that lead to disease, represents targets towards the development of novel treatment strategies and options.
Collapse
|
11
|
Yang QL, Shen JQ, Xue Y, Cheng XB, Jiang ZH, Yang YC, Chen YD, Zhou XN. Pathological Lesions and Inducible Nitric Oxide Synthase Expressions in the Liver of Mice Experimentally Infected with Clonorchis sinensis. THE KOREAN JOURNAL OF PARASITOLOGY 2015; 53:777-83. [PMID: 26797449 PMCID: PMC4725231 DOI: 10.3347/kjp.2015.53.6.777] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 06/29/2015] [Accepted: 08/24/2015] [Indexed: 12/20/2022]
Abstract
The nitric oxide (NO) formation and intrinsic nitrosation may be involved in the possible mechanisms of liver fluke-associated carcinogenesis. We still do not know much about the responses of inducible NO synthase (iNOS) induced by Clonorchis sinensis infection. This study was conducted to explore the pathological lesions and iNOS expressions in the liver of mice with different infection intensity levels of C. sinensis. Extensive periductal inflammatory cell infiltration, bile duct hyperplasia, and fibrosis were commonly observed during the infection. The different pathological responses in liver tissues strongly correlated with the infection intensity of C. sinensis. Massive acute spotty necrosis occurred in the liver parenchyma after a severe infection. The iNOS activity in liver tissues increased, and iNOS-expressing cells with morphological differences were observed after a moderate or severe infection. The iNOS-expressing cells in liver tissues had multiple origins.
Collapse
Affiliation(s)
- Qing-Li Yang
- National Institute of Parasitic Disease, Chinese Center for Disease Control and Prevention; Key Laboratory of Parasite and Vector Biology, Ministry of Health; WHO Collaborating Centre for Malaria, Schistosomiasis and Filariasis, Shanghai 200025, P. R. China
- Guangxi Zhuang Autonomous Region Center for Disease Prevention and Control; Guangxi Key Laboratory for the Prevention and Control of Viral Hepatitis, Nanning 530028, P. R. China
| | - Ji-Qing Shen
- Department of Parasitology, Guangxi Medical University, Nanning 530021, P. R. China
| | - Yan Xue
- Department of Parasitology, Guangxi Medical University, Nanning 530021, P. R. China
| | - Xiao-Bing Cheng
- Department of Parasitology, Guangxi Medical University, Nanning 530021, P. R. China
| | - Zhi-Hua Jiang
- Guangxi Zhuang Autonomous Region Center for Disease Prevention and Control; Guangxi Key Laboratory for the Prevention and Control of Viral Hepatitis, Nanning 530028, P. R. China
| | - Yi-Chao Yang
- Guangxi Zhuang Autonomous Region Center for Disease Prevention and Control; Guangxi Key Laboratory for the Prevention and Control of Viral Hepatitis, Nanning 530028, P. R. China
| | - Ying-Dan Chen
- National Institute of Parasitic Disease, Chinese Center for Disease Control and Prevention; Key Laboratory of Parasite and Vector Biology, Ministry of Health; WHO Collaborating Centre for Malaria, Schistosomiasis and Filariasis, Shanghai 200025, P. R. China
| | - Xiao-Nong Zhou
- National Institute of Parasitic Disease, Chinese Center for Disease Control and Prevention; Key Laboratory of Parasite and Vector Biology, Ministry of Health; WHO Collaborating Centre for Malaria, Schistosomiasis and Filariasis, Shanghai 200025, P. R. China
| |
Collapse
|
12
|
Boubaker G, Hemphill A, Huber CO, Spiliotis M, Babba H, Gottstein B. Prevention and Immunotherapy of Secondary Murine Alveolar Echinococcosis Employing Recombinant EmP29 Antigen. PLoS Negl Trop Dis 2015; 9:e0003795. [PMID: 26053794 PMCID: PMC4460070 DOI: 10.1371/journal.pntd.0003795] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 04/28/2015] [Indexed: 12/13/2022] Open
Abstract
Alveolar echinococcosis (AE) is caused by infection with the larval stage of the tapeworm Echinococcus multilocularis. An increasing understanding of immunological events that account for the metacestode survival in human and murine AE infection prompted us to undertake explorative experiments tackling the potential of novel preventive and/or immunotherapeutic measures. In this study, the immunoprotective and immunotherapeutic ability of recombinant EmP29 antigen (rEmP29) was assessed in mice that were intraperitoneally infected with E. multilocularis metacestodes. For vaccination, three intraperitoneal injections with 20μg rEmP29 emulsified in saponin adjuvants were applied over 6 weeks. 2 weeks after the last boost, mice were infected, and at 90 days post-infection, rEmP29-vaccinated mice exhibited a median parasite weight that was reduced by 75% and 59% when compared to NaCl- or saponin–treated control mice, respectively. For immunotherapeutical application, the rEmP29 (20μg) vaccine was administered to experimentally infected mice, starting at 1 month post-infection, three times with 2 weeks intervals. Mice undergoing rEmP29 immunotherapy exhibited a median parasite load that was reduced by 53% and 49% when compared to NaCl- and saponin–treated control mice, respectively. Upon analysis of spleen cells, both, vaccination and treatment with rEmP29, resulted in low ratios of Th2/Th1 (IL-4/IFN-γ) cytokine mRNA and low levels of mRNA coding for IL-10 and IL-2. These results suggest that reduction of the immunosuppressive environment takes place in vaccinated as well as immunotreated mice, and a shift towards a Th1 type of immune response may be responsible for the observed increased restriction of parasite growth. The present study provides the first evidence that active immunotherapy may present a sustainable route for the control of AE. Current medical management of AE that relies on surgery and continuous benzimidazole administration is of limited effectiveness. Therefore, alternative preventive and therapeutic tools need to be explored. Here, we demonstrate that vaccination with recombinant antigen EmP29 (rEmP29), prior or after secondary infection of BALB/c mice, resulted in a significant reduction of the median parasite weight when compared to different control groups. We then characterized the transcription level of splenic IL-4 and IFN-γ cytokines as hallmarks for AE-anti-protective humoral immune reaction (Th2) and for AE-effective (restrictive) cellular response (Th1), respectively. Results revealed that vaccinated mice in pre- or post-infection situation exhibited the lowest IL-4/IFN-γ mRNA ratios. In addition, those groups showed also significantly low levels of IL-10-encoding mRNA coding (immunosuppressive cytokine), as well as IL-2. These findings suggest that reduction of parasite load in rEmP29-vaccinated mice (in pre- or post-infection status) might be triggered by a decline of the immunosuppressive environment and a change of the host immune reaction towards a Th1-re-oriented cell-mediated immune defense. A similar non-specific effect appears also to be yielded by the immunostimulating adjuvants. This study provides the first insight into the potential benefits of antigen-specific immunotherapy as new treatment option of AE.
Collapse
Affiliation(s)
- Ghalia Boubaker
- Institute of Parasitology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Faculty of Pharmacy, Department of Clinical Biology B, Laboratory of Medical and Molecular Parasitology–Mycology (LR12ES08), University of Monastir, Monastir, Tunisia
| | - Andrew Hemphill
- Institute of Parasitology, University of Bern, Bern, Switzerland
| | | | - Markus Spiliotis
- Institute of Parasitology, University of Bern, Bern, Switzerland
| | - Hamouda Babba
- Faculty of Pharmacy, Department of Clinical Biology B, Laboratory of Medical and Molecular Parasitology–Mycology (LR12ES08), University of Monastir, Monastir, Tunisia
| | - Bruno Gottstein
- Institute of Parasitology, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
13
|
Shen JQ, Yang QL, Xue Y, Cheng XB, Jiang ZH, Yang YC, Chen YD, Zhou XN. Inducible nitric oxide synthase response and associated cytokine gene expression in the spleen of mice infected with Clonorchis sinensis. Parasitol Res 2015; 114:1661-70. [PMID: 25687522 PMCID: PMC4412385 DOI: 10.1007/s00436-015-4347-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 01/22/2015] [Indexed: 12/26/2022]
Abstract
Clonorchis sinensis is a food-borne parasite that induces a permanent increase of nitrosation in the body upon infection. The spleen is an important secondary lymphoid organ for the regulation of immune responses locally and in the whole body. However, the functions and mechanisms of the spleen in nitric oxide (NO) responses after C. sinensis infection remain unknown. In this study, BALB/c mice were infected with 20, 40, and 80 C. sinensis metacercariae to simulate mild, moderate, and severe infections, respectively. We examined the expression of inducible nitric oxide synthase (iNOS) in the spleen and the relevant cytokine transcription in splenocytes from the mice infected with different amounts of metacercariae. The iNOS of the mice infected with 80 metacercariae was expressed in the spleen as early as 10 days post-infection (dpi) and gradually increased until 90 dpi. The iNOS expression in the mice infected with 40 metacercariae was detected only at 45 and 90 dpi, but not in the mice infected with 20 metacercariae. The level of interferon (IFN)-γ messenger RNA (mRNA) transcription in splenocytes significantly increased at 10 and 20 dpi (P < 0.05) in response to mild/moderate infection but gradually decreased to normal levels after 45 dpi. The level of IL-12p35 mRNA transcription did not change at 10 and 20 dpi but significantly decreased after 45 dpi under moderate/severe infection (P < 0.05/0.01/0.001). The level of IL-18 mRNA transcription significantly increased at 10 dpi (P < 0.05/0.01) but significantly decreased after 20 dpi (P < 0.05/0.01/0.001). These results suggest that spleen is an important organ for iNOS/NO responses, which correspond to the severity of C. sinensis infection, but cannot be attributed to the expression of the Th1 cytokines.
Collapse
Affiliation(s)
- Ji-Qing Shen
- Department of Parasitology, Guangxi Medical University, Nanning, 530021 People’s Republic of China
| | - Qing-Li Yang
- National Institute of Parasitic Disease, Chinese Center for Disease Control and Prevention; Key Laboratory of Parasite and Vector Biology, MOH, WHO Collaborating Centre for Malaria, Schistosomiasis and Filariasis, Shanghai, 200025 People’s Republic of China
- Guangxi Zhuang Autonomous Region Center for Disease Prevention and Control, Guangxi Key Laboratory for the Prevention and Control of Viral Hepatitis, Nanning, 530028 People’s Republic of China
| | - Yan Xue
- Department of Parasitology, Guangxi Medical University, Nanning, 530021 People’s Republic of China
| | - Xiao-Bing Cheng
- Department of Parasitology, Guangxi Medical University, Nanning, 530021 People’s Republic of China
| | - Zhi-Hua Jiang
- Guangxi Zhuang Autonomous Region Center for Disease Prevention and Control, Guangxi Key Laboratory for the Prevention and Control of Viral Hepatitis, Nanning, 530028 People’s Republic of China
| | - Yi-Chao Yang
- Guangxi Zhuang Autonomous Region Center for Disease Prevention and Control, Guangxi Key Laboratory for the Prevention and Control of Viral Hepatitis, Nanning, 530028 People’s Republic of China
| | - Ying-Dan Chen
- National Institute of Parasitic Disease, Chinese Center for Disease Control and Prevention; Key Laboratory of Parasite and Vector Biology, MOH, WHO Collaborating Centre for Malaria, Schistosomiasis and Filariasis, Shanghai, 200025 People’s Republic of China
| | - Xiao-Nong Zhou
- National Institute of Parasitic Disease, Chinese Center for Disease Control and Prevention; Key Laboratory of Parasite and Vector Biology, MOH, WHO Collaborating Centre for Malaria, Schistosomiasis and Filariasis, Shanghai, 200025 People’s Republic of China
| |
Collapse
|
14
|
MHC-DRB1/DQB1 Gene Polymorphism and Its Association with Resistance/Susceptibility to Cystic Echinococcosis in Chinese Merino Sheep. J Parasitol Res 2014; 2014:272601. [PMID: 24782918 PMCID: PMC3982463 DOI: 10.1155/2014/272601] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 10/23/2013] [Accepted: 10/23/2013] [Indexed: 11/17/2022] Open
Abstract
The aim of this study was to analyze the relationship between polymorphism of the MHC-DRB1/DQB1 gene and its resistance to Cystic Echinococcosis (C.E), as well as to screen out the molecular genetic marker of antiechinococcosis in Chinese Merino sheep. The MHCII-DRB1/DQB1 exon 2 was amplified by polymerase chain reaction (PCR) from DNA samples of healthy and hydatidosis sheep. PCR products were characterized by restriction fragment length polymorphism (RFLP) technique. Five restriction enzymes (Mval, HaeIII, SacI, SacII, and Hin1I) were employed to cut DRB1, while seven restriction enzymes (MroxI, ScaI, SacII, NciI, TaqI, Mval, and HaeIII) were employed to cut DQB1.Results showed that frequencies of patterns Mvalbb (P < 0.01), SacIab in DRB1 exon 2 (P < 0.05), and TaqIaa, HaeIIInn (P < 0.01) in DQB1 exon 2 were significantly higher in the healthy group compared with the C.E individuals, which implied that there was a strong association between these genotypes and hydatidosis resistance or susceptibility. Chi-square test showed that individuals with the genic haplotype DRB1-SacIab/DRB1-Mvalbb/DQB1-TaqIaa/DQB1-HaeIIInn (P < 0.01) were relatively resistant to C.E, while individuals with the genic haplotypes DRB1-Mvalbc/DQB1-Mvalyy/DQB1-TaqIab/DQB1-HaeIIImn (P < 0.01) and DRB1-Mvalbb/DQB1-Mvalcc/DQB1-TaqIab/DQB1-HaeIIImn (P < 0.01) were more susceptible to C.E. In addition, to confirm these results, a fielding experiment was performed with Chinese Merino sheep which were artificially infected with E.g. The result was in accordance with the results of the first study. In conclusion, MHC-DRB1/DQB1 exon 2 plays an important role as resistant to C.E in Chinese Merino sheep. In addition, the molecular genetic marker of antiechinococcosis (DRB1-SacIab/DRB1-Mvalbb/DQB1-TaqIaa/DQB1-HaeIIInn) was screened out in Chinese Merino sheep.
Collapse
|
15
|
Mejri N, Müller N, Hemphill A, Gottstein B. Intraperitoneal Echinococcus multilocularis infection in mice modulates peritoneal CD4+ and CD8+ regulatory T cell development. Parasitol Int 2010; 60:45-53. [PMID: 20965274 DOI: 10.1016/j.parint.2010.10.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2010] [Revised: 09/30/2010] [Accepted: 10/04/2010] [Indexed: 12/16/2022]
Abstract
Intraperitoneal proliferation of the metacestode stage of Echinococcus multilocularis in experimentally infected mice is followed by an impaired host immune response favoring parasite survival. We here demonstrate that infection in chronically infected mice was associated with a 3-fold increase of the percentages of CD4+ and CD8+ peritoneal T (pT) cells compared to uninfected controls. pT cells of infected mice expressed high levels of IL-4 mRNA, while only low amounts of IFN-γ mRNA were detected, suggesting that a Th2-biased immune response predominated the late stage of disease. Peritoneal dendritic cells from infected mice (AE-pDCs) expressed high levels of TGF-β mRNA and very low levels of IL-10 and IL-12 (p40) mRNA, and the expression of surface markers for DC-maturation such as MHC class II (Ia) molecules, CD80, CD86 and CD40 was down-regulated. In contrast to pDCs from non-infected mice, AE-pDCs did not enhance Concanavalin A (ConA)-induced proliferation when added to CD4+ pT and CD8+ pT cells of infected and non-infected mice, respectively. In addition, in the presence of a constant number of pDCs from non-infected mice, the proliferation of CD4+ pT cells obtained from infected animals to stimulation with ConA was lower when compared to the responses of CD4+ pT cells obtained from non-infected mice. This indicated that regulatory T cells (Treg) may interfere in the complex immunological host response to infection. Indeed, a subpopulation of regulatory CD4+ CD25+ pT cells isolated from E. multilocularis-infected mice reduced ConA-driven proliferation of CD4+ pT cells. The high expression levels of Foxp3 mRNA by CD4+ and CD8+ pT cells suggested that subpopulations of regulatory CD4+ Foxp3+ and CD8+ Foxp3+ T cells were involved in modulating the immune responses within the peritoneal cavity of E. multilocularis-infected mice.
Collapse
Affiliation(s)
- Naceur Mejri
- Institute of Parasitology, University of Bern, Bern, Switzerland
| | | | | | | |
Collapse
|
16
|
|
17
|
Gottstein B, Wittwer M, Schild M, Merli M, Leib SL, Müller N, Müller J, Jaggi R. Hepatic gene expression profile in mice perorally infected with Echinococcus multilocularis eggs. PLoS One 2010; 5:e9779. [PMID: 20368974 PMCID: PMC2848562 DOI: 10.1371/journal.pone.0009779] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Accepted: 02/26/2010] [Indexed: 12/17/2022] Open
Abstract
Background Alveolar echinococcosis (AE) is a severe chronic hepatic parasitic disease currently emerging in central and eastern Europe. Untreated AE presents a high mortality (>90%) due to a severe hepatic destruction as a result of parasitic metacestode proliferation which behaves like a malignant tumor. Despite this severe course and outcome of disease, the genetic program that regulates the host response leading to organ damage as a consequence of hepatic alveolar echinococcosis is largely unknown. Methodology/Principal Findings We used a mouse model of AE to assess gene expression profiles in the liver after establishment of a chronic disease status as a result of a primary peroral infection with eggs of the fox tapeworm Echinococcus multilocularis. Among 38 genes differentially regulated (false discovery rate adjusted p≤0.05), 35 genes were assigned to the functional gene ontology group <immune response>, while 3 associated with the functional group <intermediary metabolism>. Upregulated genes associated with <immune response> could be clustered into functional subgroups including <macrophages>, <APCs>, <lymphocytes, chemokines and regulation>, <B-cells> and <eosinophils>. Two downregulated genes related to <lymphocytes, chemokines and regulation> and <intermediary metabolism>, respectively. The <immune response> genes either associated with an <immunosupression> or an <immunostimulation> pathway. From the overexpressed genes, 18 genes were subsequently processed with a Custom Array microfluidic card system in order to assess respective expression status at the mRNA level relative to 5 reference genes (Gapdh, Est1, Rlp3, Mdh-1, Rpl37) selected upon a constitutive and stable expression level. The results generated by the two independent tools used for the assessment of gene expression, i.e., microarray and microfluidic card system, exhibited a high level of congruency (Spearman correlation rho = 0.81, p = 7.87e-5) and thus validated the applied methods. Conclusions/Significance Based on this set of biomarkers, new diagnostic targets have been made available to predict disease status and progression. These biomarkers may also offer new targets for immuno-therapeutic intervention.
Collapse
Affiliation(s)
- Bruno Gottstein
- Vetsuisse Faculty, Institute of Parasitology, University of Bern, Bern, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Echinococcus multilocularis and its intermediate host: a model of parasite-host interplay. J Biomed Biotechnol 2010; 2010:923193. [PMID: 20339517 PMCID: PMC2842905 DOI: 10.1155/2010/923193] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Accepted: 01/07/2010] [Indexed: 12/12/2022] Open
Abstract
Host-parasite interactions in the E. multilocularis-intermediate host model depend on a subtle balance between cellular immunity, which is responsible for host's resistance towards the metacestode, the larval stage of the parasite, and tolerance induction and maintenance. The pathological features of alveolar echinococcosis. the disease caused by E. multilocularis, are related both to parasitic growth and to host's immune response, leading to fibrosis and necrosis, The disease spectrum is clearly dependent on the genetic background of the host as well as on acquired disturbances of Th1-related immunity. The laminated layer of the metacestode, and especially its carbohydrate components, plays a major role in tolerance induction. Th2-type and anti-inflammatory cytokines, IL-10 and TGF-β, as well as nitric oxide, are involved in the maintenance of tolerance and partial inhibition of cytotoxic mechanisms. Results of studies in the experimental mouse model and in patients suggest that immune modulation with cytokines, such as interferon-α, or with specific antigens could be used in the future to treat patients with alveolar echinococcosis and/or to prevent this very severe parasitic disease.
Collapse
|
19
|
Nitric oxide and respiratory helminthic diseases. J Biomed Biotechnol 2010; 2010:958108. [PMID: 20169170 PMCID: PMC2821625 DOI: 10.1155/2010/958108] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Accepted: 10/12/2009] [Indexed: 11/17/2022] Open
Abstract
Nitric oxide (NO) is a very simple molecule that displays very important functions both in helminths (mainly those involved in respiratory pathology) and in mammalian hosts. In this paper we review four issues related to interaction of NO and lung helminthic diseases. Firstly, we evaluated data available on the NO synthesis and release by helminths and their biological role. Next, we summarized the effect of antigens obtained from different phases of the biological cycle on NO production by host mammalian cells (mainly from human sources). Thirdly, we revised the evaluation of NO on the biological activities and/or the viability of respiratory helminths. Lastly, the deleterious consequences of increased production of NO during helminthic human infection are detailed.
Collapse
|
20
|
Similarity and diversity in macrophage activation by nematodes, trematodes, and cestodes. J Biomed Biotechnol 2010; 2010:262609. [PMID: 20145705 PMCID: PMC2817371 DOI: 10.1155/2010/262609] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Accepted: 10/07/2009] [Indexed: 12/25/2022] Open
Abstract
This review summarizes current knowledge of macrophages in helminth infections, with a focus not only on delineating the striking similarities in macrophage phenotype between diverse infections but also on highlighting the differences. Findings from many different labs illustrate that macrophages in helminth infection can act as anti-parasite effectors but can also act as powerful immune suppressors. The specific role for their alternative (Th2-mediated) activation in helminth killing or expulsion versus immune regulation remains to be determined. Meanwhile, the rapid growth in knowledge of alternatively activated macrophages will require an even more expansive view of their potential functions to include repair of host tissue and regulation of host metabolism.
Collapse
|
21
|
Triggering and modulation of the host-parasite interplay byEchinococcus multilocularis: a review. Parasitology 2009; 137:557-68. [DOI: 10.1017/s0031182009991533] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
SUMMARYAs more facts emerge regarding the ways in whichE. multilocularis-derived molecules trigger the host immune response and modulate the host-parasite interplay, it becomes possible to envisage how the parasite can survive and proliferate in its intermediate host, while in other hosts it dies out. Through effects on cells of both the innate and adaptive arms of the immune response,E. multiloculariscan orchestrate a range of outcomes that are beneficial not only to the parasite, in terms of facilitating its intrahepatic proliferation and maturation, and thus life cycle over all, but also to its intermediate host, in limiting pathology. The present review deals with the role of metacestode surface molecules as well as excretory/secretory (E/S) metabolic products of the parasite in the modulation of the host responses such as to optimize its own survival.
Collapse
|
22
|
Echinococcus multilocularis metacestode metabolites contain a cysteine protease that digests eotaxin, a CC pro-inflammatory chemokine. Parasitol Res 2009; 105:1253-60. [PMID: 19572150 DOI: 10.1007/s00436-009-1549-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Accepted: 06/19/2009] [Indexed: 01/21/2023]
Abstract
In many helminthic infections, eotaxin, a CC-chemokine, triggers the mobilization of eosinophils, thus, contributing to an elevated blood and periparasitic eosinophil level. Following an experimental intraperitoneal infection of C57BL6 mice with Echinococcus multilocularis metacestodes, however, we observed the absence of eosinophils in the peritoneal cavity and a low number of such cells in the blood of infected animals. Therefore, we carried out an explorative study to address the question why eosinophilia did not occur especially in the peritoneal cavity of such secondarily AE-infected mice. In an in vitro assay, we showed that metacestode antigens (in vitro generated vesicle fluid and E/S products) were able to proteolytically digest eotaxin. This effect was confirmed with semiquantitative Western blotting, which demonstrated a decreasing intensity of remaining eotaxin signals. Proteolysis of eotaxin was, thus, dose-dependent and proportional to the time of incubation with the metacestode antigens. Using appropriate inhibitors, the respective protease was identified as a cysteine protease, which required the presence of Ca(++) as co-enzyme. A chromatographic fractionation procedure by successive separation of VF molecules using a superpose column and subsequently a MonoQ column mounted on an FPLC system allowed to yield a fraction, referred to us as fraction 6; containing the enriched cysteine protease, this fraction will be used for further molecular studies. Eotaxin inactivation by VF and E/S products may contribute to explain the absence of eosinophils within the peritoneal cavity of AE-secondary infected mice. Absent eosinophils, thus, may be a part of a series of events that maintain a low level of inflammation displayed within the peritoneal cavity of experimentally infected mice.
Collapse
|
23
|
Zhang W, Ross AG, McManus DP. Mechanisms of Immunity in Hydatid Disease: Implications for Vaccine Development. THE JOURNAL OF IMMUNOLOGY 2008; 181:6679-85. [DOI: 10.4049/jimmunol.181.10.6679] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
24
|
Gottstein B, Hemphill A. Echinococcus multilocularis: the parasite-host interplay. Exp Parasitol 2008; 119:447-452. [PMID: 18410929 DOI: 10.1016/j.exppara.2008.03.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2007] [Revised: 02/27/2008] [Accepted: 03/06/2008] [Indexed: 11/16/2022]
Abstract
Alveolar echinococcosis (AE) is a severe chronic helminthic disease caused by the intrahepatic tumor-like growth of the metacestode of Echinococcus multilocularis. Metacestodes are fluid-filled, asexually proliferating vesicles, which are entirely covered by the laminated layer, an acellular carbohydrate-rich surface structure that protects the parasite from immunological and physiological reactions on part of the host. The E. multilocularis metacestode has acquired specific means of manipulating and using the immunological host response to its own advantage. These include the expression of distinct immunoregulatory parasite molecules that manipulate and interfere in the functional activity of macrophages and T cells. Recent research findings have led to a better understanding of the protein- and glycoprotein composition of the laminated layer and the E/S fraction of the metacestode, including Em2- and Em492-antigens, two metacestode antigen fractions that exhibit immunosuppressive or -modulatory properties. Understanding of the events taking place at the host-parasite interface is the key for development of novel immuno-therapeutical and/or chemotherapeutical tools.
Collapse
Affiliation(s)
- Bruno Gottstein
- Institute of Parasitology, Vetsuisse Faculty and Faculty of Medicine, University of Bern, Laenggassstrasse 122, CH-3001 Bern, Switzerland.
| | | |
Collapse
|
25
|
Siles-Lucas M, Merli M, Gottstein B. 14-3-3 proteins in Echinococcus: their role and potential as protective antigens. Exp Parasitol 2008; 119:516-523. [PMID: 18316081 DOI: 10.1016/j.exppara.2008.01.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Revised: 01/22/2008] [Accepted: 01/28/2008] [Indexed: 10/22/2022]
Abstract
14-3-3 Proteins are a family of highly conserved proteins among all eukaryotic organisms studied so far. As basically intracellular proteins, they play a key role in basic cellular events related to cellular proliferation, including signal transduction, cell-cycle control, cell differentiation and cell survival. The 14-3-3 proteins have been described and characterized in several parasites, and mostly studied in Echinococcus granulosus and Echinococcus multilocularis. Here, we review the discoveries regarding this protein family in the genus Echinococcus, describing new data about specific aspects related with their implication in the parasite biology and immunology in the frame of the host-parasite relationship.
Collapse
Affiliation(s)
- Mar Siles-Lucas
- Unidad de Patología Animal, Instituto de Recursos Naturales y Agrobiología de Salamanca (IRNASA), Consejo Superior de Investigaciones Científicas (CSIC), Cordel de Merinas 40-52, 37008 Salamanca, Spain.
| | | | | |
Collapse
|
26
|
Abstract
Echinococcosis is a cosmopolitan zoonosis caused by adult or larval stages of cestodes belonging to the genus Echinococcus (family Taeniidae). The two major species of medical and public health importance are Echinococcus granulosus and Echinococcus multilocularis, which cause cystic echinococcosis and alveolar echinococcosis, respectively. Both cystic echinococcosis and alveolar echinococcosis are serious diseases, the latter especially so, with a high fatality rate and poor prognosis if managed inappropriately. This review highlights recent advances in immunity to infection and vaccination against both parasites in their intermediate and definitive hosts and procedures for diagnosis of cystic echinococcosis and alveolar echinococcosis, including the value of immunodiagnostic and DNA approaches. There is discussion also of progress in genomics and related technologies that is providing valuable insights on the functional biology of the Echinococcus organisms. These studies will underpin future research that will reveal a better understanding of the Echinococcus-host interplay, and suggest new avenues for the identification of additional targets for diagnosis, vaccination and chemotherapy.
Collapse
Affiliation(s)
- Wenbao Zhang
- Molecular Parasitology Laboratory, Australian Centre for International and Tropical Health and Nutrition, The Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | | |
Collapse
|
27
|
Gottstein B, Haag K, Walker M, Matsumoto J, Mejri N, Hemphill A. Molecular survival strategies of Echinococcus multilocularis in the murine host. Parasitol Int 2005; 55 Suppl:S45-9. [PMID: 16352460 DOI: 10.1016/j.parint.2005.11.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Larval infection with Echinococcus multilocularis starts with the intrahepatic postoncospheral development of a metacestode that-at its mature stage-consists of an inner germinal and an outer laminated layer (GL & LL). In certain cases, an appropriate host immune response may inhibit parasite proliferation. Several lines of evidence obtained in vivo and in vitro indicate the important bio-protective role of the LL. For instance, the LL has been proposed to protect the GL from nitric oxide produced by periparasitic macrophages and dendritic cells, and also to prevent immune recognition by surrounding T cells. On the other hand, the high periparasitic NO production by peritoneal exsudate cells contributes to periparasitic immunosuppression, explaining why iNOS deficienct mice exhibit a significantly lower susceptibility towards experimental infection. The intense periparasitic granulomatous infiltration indicates a strong host-parasite interaction, and the involvement of cellular immunity in control of the metacestode growth kinetics is strongly suggested by experiments carried out in T cell deficient mouse strains. Carbohydrate components of the LL, such as Em2(G11) and Em492, as well as other parasite metabolites yield immunomodulatory effects that allow the parasite to survive in the host. I.e., the IgG response to the Em2(G11)-antigen takes place independently of alpha-beta+CD4+T cells, and in the absence of interactions between CD40 and CD40 ligand. Such parasite molecules also interfere with antigen presentation and cell activation, leading to a mixed Th1/Th2-type response at the later stage of infection. Furthermore, Em492 and other (not yet published) purified parasite metabolites suppress ConA and antigen-stimulated splenocyte proliferation. Infected mouse macrophages (AE-MØ) as antigen presenting cells (APC) exhibited a reduced ability to present a conventional antigen (chicken ovalbumin, C-Ova) to specific responder lymph node T cells when compared to normal MØ. As AE-MØ fully maintain their capacity to appropriately process antigens, a failure in T cell receptor occupancy by antigen-Ia complex or/and altered co-stimulatory signals can be excluded. Studying the status of accessory molecules implicated in T cell stimulation by MØ, it could be shown that B7-1 (CD80) and B7-2 (CD86) remained unchanged, whereas CD40 was down-regulated and CD54 (=ICAM-1) slightly up-regulated. FACS analysis of peritoneal cells revealed a decrease in the percentage of CD4+ and CD8+T cells in AE-infected mice. Taken together the obstructed presenting-activity of AE-MØ appeared to trigger an unresponsiveness of T cells leading to the suppression of their clonal expansion during the chronic phase of AE infection. Interesting information on the parasite survival strategy and potential can be obtained upon in vitro and in vivo treatment. Hence, we provided very innovative results by showing that nitazoxanide, and now also, respectively, new modified compounds may represent a useful alternative to albendazole. In the context of chemotherapeutical repression of parasite growth, we searched also for parasite molecules, whose expression levels correlate with the viability and growth activity of E. multilocularis metacestode. Expression levels of 14-3-3 and II/3-10, relatively quantified by realtime reverse transcription-PCR using a housekeeping gene beta-actin, were studied in permissive nu/nu and in low-permissive wild type BALB/c mice. At 2 months p.i., the transcription level of 14-3-3 was significantly higher in parasites actively proliferating in nu/nu mice compared to parasites moderately growing in wild type mice. Immunoblotting experiments confirmed at the protein level that 14-3-3 was over-expressed in parasites derived from nu/nu mice at 2 months p.i. In vitro-treatment of E. multilocularis with an anti-echinococcal drug nitazoxanide for a period of 8 days resulted in a significant decrease of both 14-3-3 and II/3-10 transcription levels, which correlated with the kinetics of a housekeeping gene, beta-actin. This indicates that 14-3-3-exhibits a good potential as a molecular marker to assess viability and growth activity of the parasite.
Collapse
Affiliation(s)
- Bruno Gottstein
- Institute of Parasitology, Vetsuisse Faculty and Faculty of Medicine, University of Bern, Switzerland.
| | | | | | | | | | | |
Collapse
|
28
|
Andrade MA, Siles-Lucas M, Espinoza E, Pérez Arellano JL, Gottstein B, Muro A. Echinococcus multilocularis laminated-layer components and the E14t 14-3-3 recombinant protein decrease NO production by activated rat macrophages in vitro. Nitric Oxide 2005; 10:150-5. [PMID: 15158694 DOI: 10.1016/j.niox.2004.03.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2003] [Revised: 03/03/2004] [Indexed: 11/29/2022]
Abstract
Echinococcus multilocularis and Echinococcus granulosus cause alveolar and cystic (unilocular) echinococcosis, respectively, in humans and animals. It is known that these parasites can affect, among other molecules, nitric oxide (NO) production by periparasitic host cells. Nevertheless, detailed dissection of parasite components specifically affecting cell NO production has not been done to date. We compare the effect of E. granulosus and E. multilocularis defined metacestode structural (laminated-layer associated) and metabolic (14-3-3 protein, potentially related with E. multilocularis metacestode tumor-like growth) components on the NO production by rat alveolar macrophages in vitro. Our results showed that none of these antigens could stimulate macrophage NO production in vitro. However, a reversed effect of some Echinococcus antigens on NO in vitro production was found when cells were previously exposed to LPS stimulation. This inhibitory effect was found when E. multilocularis laminated-layer (LL) or cyst wall (CW) soluble components from both species were used. Pre-stimulation of cells with LPS also resulted in a strong, dose-dependent reduction of NO and iNOS mRNA production after incubation of cells with the E14t protein. Thus, the E. multilocularis 14-3-3 protein appears to be one of the components accounting for the suppressive effect of the CW and LL metacestode extracts.
Collapse
Affiliation(s)
- M Amparo Andrade
- Laboratorio de Parasitología, Facultad de Farmacia, Universidad de Salamanca, Avda, Campo Charro s/n 37007 Salamanca, España, Spain
| | | | | | | | | | | |
Collapse
|
29
|
Dai WJ, Waldvogel A, Siles-Lucas M, Gottstein B. Echinococcus multilocularis proliferation in mice and respective parasite 14-3-3 gene expression is mainly controlled by an alphabeta CD4 T-cell-mediated immune response. Immunology 2004; 112:481-8. [PMID: 15196217 PMCID: PMC1782501 DOI: 10.1111/j.1365-2567.2004.01885.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
The role of specific B lymphocytes and T-cell populations in the control of experimental Echinococus multilocularis infection was studied in micro MT, nude, T-cell receptor (TCR)-beta(-/-), major histocompatibility complex (MHC)-I(-/-) and MHC-II(-/-) mice. At 2 months postinfection, the parasite mass was more than 10 times higher in nude, TCR-beta(-/-) and MHC-II(-/-) mice than in infected C57BL/6 wild-type (WT) mice, and these T-cell-deficient mice started to die of the high parasite load at this time-point. In contrast, MHC-I(-/-) and micro MT mice exhibited parasite growth rates similar to those found in WT controls. These findings clearly point to the major role that CD4(+) alphabeta(+) T cells play in limiting the E. multilocularis proliferation, while CD8(+) T and B cells appeared to play a minor role in the control of parasite growth. In the absence of T cells, especially CD4(+) or alphabeta(+) T cells, the cellular immune response to infection was impaired, as documented by the lack of hepatic granuloma formation around the parasite and by a decreased splenocyte responsiveness to concanavalin A (Con A) and parasite antigen stimulation. Surprisingly, in T-cell-deficient mice, the ex vivo expression of interferon-gamma (IFN-gamma) and other inflammatory cytokines (except for interleukin-6) were increased in association with a high parasite load. Thus, the relative protection mediated by CD4(+) alphabeta(+) T cells against E. multilocularis infection seemed not be IFN-gamma dependent, but rather to rely on the effector's function of CD4(+) alphabeta(+) T cells. The local restriction of parasite germinal cell proliferation was reflected by a regulatory effect on the expression of 14-3-3 protein within the parasite tissue in T-cell-deficient mice. These results provide a strong indication that the CD4(+) alphabeta(+) T-cell-mediated immune response contributes to the control of the parasite growth and to the regulation of production of the parasite 14-3-3 protein in metacestode tissues.
Collapse
Affiliation(s)
- Wen Juan Dai
- Institute of Parasitology, University of Berne, Berne, Switzerland
| | | | | | | |
Collapse
|
30
|
Walker M, Baz A, Dematteis S, Stettler M, Gottstein B, Schaller J, Hemphill A. Isolation and characterization of a secretory component of Echinococcus multilocularis metacestodes potentially involved in modulating the host-parasite interface. Infect Immun 2004; 72:527-36. [PMID: 14688134 PMCID: PMC344003 DOI: 10.1128/iai.72.1.527-536.2004] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Echinococcus multilocularis metacestodes are fluid-filled, vesicle-like organisms, which are characterized by continuous asexual proliferation via external budding of daughter vesicles, predominantly in the livers of infected individuals. Tumor-like growth eventually leads to the disease alveolar echinococcosis (AE). We employed the monoclonal antibody (MAb) E492/G1, previously shown to be directed against a carbohydrate-rich, immunomodulatory fraction of Echinococcus granulosus, to characterize potentially related components in E. multilocularis. Immunofluorescence studies demonstrated that MAb E492/G1-reactive epitopes were found predominantly on the laminated layer and in the periphery of developing brood capsules. The respective molecules were continuously released into the exterior medium and were also found in the parasite vesicle fluid. The MAb E492/G1-reactive fraction in E. multilocularis, named Em492 antigen, was isolated by immunoaffinity chromatography. Em492 antigen had a protein/carbohydrate ratio of 0.25, reacted with a series of lectins, and is related to the laminated layer-associated Em2(G11) antigen. The epitope recognized by MAb E492/G1 was sensitive to sodium periodate but was not affected by protease treatment. Anti-Em492 immunoglobulin G1 (IgG1) and IgG2 and, at lower levels, IgG3 were found in sera of mice suffering from experimentally induced secondary, but not primary, AE. However, with regard to cellular immunity, a suppressive effect on concanavalin A- or crude parasite extract-induced splenocyte proliferation in these mice was observed upon addition of Em492 antigen, but trypan blue exclusion tests and transmission electron microscopy failed to reveal any cytotoxic effect in Em492 antigen-treated spleen cells. This indicated that Em492 antigen could be modulating the periparasitic cellular environment during E. multilocularis infection through as yet unidentified mechanisms and could be one of the factors contributing to immunosuppressive events that occur at the host-parasite interface.
Collapse
Affiliation(s)
- Mirjam Walker
- Institute of Parasitology, University of Berne, CH-3012 Berne, Switzerland.
| | | | | | | | | | | | | |
Collapse
|