1
|
Ding Q, Wu Y, Triglia ET, Gommerman JL, Subramanian A, Kuchroo VK, Rothstein DM. TIM-4 Identifies Effector B Cells Expressing a RORγt-Driven Proinflammatory Cytokine Module That Promotes Immune Responsiveness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.22.558524. [PMID: 37790513 PMCID: PMC10542535 DOI: 10.1101/2023.09.22.558524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
B cells can express pro-inflammatory cytokines that promote a wide variety of immune responses. Here we show that B cells expressing the phosphatidylserine receptor TIM-4, preferentially express IL-17A, as well as IL-22, IL-6, IL-1β, and GM-CSF - a collection of cytokines reminiscent of pathogenic Th17 cells. Expression of this proinflammatory module requires IL-23R signaling and selective expression of RORγt and IL-17A by TIM-4+ B cells. TIM-4+ B cell-derived-IL-17A not only enhances the severity of experimental autoimmune encephalomyelitis (EAE) and promotes allograft rejection, but also acts in an autocrine manner to prevent their conversion into IL-10-expressing B cells with regulatory function. Thus, IL-17A acts as an inflammatory mediator and also enforces the proinflammatory activity of TIM-4+ B cells. Thus, TIM-4 serves as a broad marker for RORγt+ effector B cells (Beff) and allows further study of the signals regulating Beff differentiation and effector molecule expression.
Collapse
Affiliation(s)
- Qing Ding
- Thomas E. Starzl Transplantation Institute; University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yufan Wu
- Klarman Cell Observatory, Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | - Elena Torlai Triglia
- Klarman Cell Observatory, Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | | | - Ayshwarya Subramanian
- Klarman Cell Observatory, Broad Institute of MIT and Harvard; Cambridge, MA, USA
- The Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | - Vijay K. Kuchroo
- Klarman Cell Observatory, Broad Institute of MIT and Harvard; Cambridge, MA, USA
- The Broad Institute of MIT and Harvard; Cambridge, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, USA
| | - David M. Rothstein
- Thomas E. Starzl Transplantation Institute; University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
2
|
Papadakos SP, Chatzikalil E, Vakadaris G, Reppas L, Arvanitakis K, Koufakis T, Siakavellas SI, Manolakopoulos S, Germanidis G, Theocharis S. Exploring the Role of GITR/GITRL Signaling: From Liver Disease to Hepatocellular Carcinoma. Cancers (Basel) 2024; 16:2609. [PMID: 39061246 PMCID: PMC11275207 DOI: 10.3390/cancers16142609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer and presents a continuously growing incidence and high mortality rates worldwide. Besides advances in diagnosis and promising results of pre-clinical studies, established curative therapeutic options for HCC are not currently available. Recent progress in understanding the tumor microenvironment (TME) interactions has turned the scientific interest to immunotherapy, revolutionizing the treatment of patients with advanced HCC. However, the limited number of HCC patients who benefit from current immunotherapeutic options creates the need to explore novel targets associated with improved patient response rates and potentially establish them as a part of novel combinatorial treatment options. Glucocorticoid-induced TNFR-related protein (GITR) belongs to the TNFR superfamily (TNFRSF) and promotes CD8+ and CD4+ effector T-cell function with simultaneous inhibition of Tregs function, when activated by its ligand, GITRL. GITR is currently considered a potential immunotherapy target in various kinds of neoplasms, especially with the concomitant use of programmed cell-death protein-1 (PD-1) blockade. Regarding liver disease, a high GITR expression in liver progenitor cells has been observed, associated with impaired hepatocyte differentiation, and decreased progenitor cell-mediated liver regeneration. Considering real-world data proving its anti-tumor effect and recently published evidence in pre-clinical models proving its involvement in pre-cancerous liver disease, the idea of its inclusion in HCC therapeutic options theoretically arises. In this review, we aim to summarize the current evidence supporting targeting GITR/GITRL signaling as a potential treatment strategy for advanced HCC.
Collapse
Affiliation(s)
- Stavros P. Papadakos
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.P.P.); (E.C.)
| | - Elena Chatzikalil
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.P.P.); (E.C.)
| | - Georgios Vakadaris
- First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (G.V.); (K.A.)
- Basic and Translational Research Unit (BTRU), Special Unit for Biomedical Research and Education (BRESU), Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Lampros Reppas
- 4th Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece;
| | - Konstantinos Arvanitakis
- First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (G.V.); (K.A.)
- Basic and Translational Research Unit (BTRU), Special Unit for Biomedical Research and Education (BRESU), Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Theocharis Koufakis
- 2nd Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, 54642 Thessaloniki, Greece;
| | - Spyros I. Siakavellas
- 2nd Academic Department of Internal Medicine, Liver-GI Unit, General Hospital of Athens “Hippocration”, National and Kapodistrian University of Athens, 114 Vas. Sofias str, 11527 Athens, Greece; (S.I.S.); (S.M.)
| | - Spilios Manolakopoulos
- 2nd Academic Department of Internal Medicine, Liver-GI Unit, General Hospital of Athens “Hippocration”, National and Kapodistrian University of Athens, 114 Vas. Sofias str, 11527 Athens, Greece; (S.I.S.); (S.M.)
| | - Georgios Germanidis
- First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (G.V.); (K.A.)
- Basic and Translational Research Unit (BTRU), Special Unit for Biomedical Research and Education (BRESU), Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Stamatios Theocharis
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.P.P.); (E.C.)
| |
Collapse
|
3
|
Prasanphanich NS, Leon K, Secor WE, Shoemaker CB, Heimburg-Molinaro J, Cummings RD. Anti-schistosomal immunity to core xylose/fucose in N-glycans. Front Mol Biosci 2023; 10:1142620. [PMID: 37081851 PMCID: PMC10110957 DOI: 10.3389/fmolb.2023.1142620] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/20/2023] [Indexed: 04/07/2023] Open
Abstract
Schistosomiasis is a globally prevalent, debilitating disease that is poorly controlled by chemotherapy and for which no vaccine exists. While partial resistance in people may develop over time with repeated infections and treatments, some animals, including the brown rat (Rattus norvegicus), are only semi-permissive and have natural protection. To understand the basis of this protection, we explored the nature of the immune response in the brown rat to infection by Schistosoma mansoni. Infection leads to production of IgG to Infection leads to production of IgG to parasite glycoproteins parasite glycoproteins with complex-type N-glycans that contain a non-mammalian-type modification by core α2-Xylose and core α3-Fucose (core Xyl/Fuc). These epitopes are expressed on the surfaces of schistosomula and adult worms. Importantly, IgG to these epitopes can kill schistosomula by a complement-dependent process in vitro. Additionally, sera from both infected rhesus monkey and infected brown rat were capable of killing schistosomula in a manner inhibited by glycopeptides containing core Xyl/Fuc. These results demonstrate that protective antibodies to schistosome infections in brown rats and rhesus monkeys include IgG responses to the core Xyl/Fuc epitopes in surface-expressed N-glycans, and raise the potential of novel glyco-based vaccines that might be developed to combat this disease.
Collapse
Affiliation(s)
| | - Kristoffer Leon
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, United States
| | - W. Evan Secor
- Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Charles B. Shoemaker
- Department of Infectious Disease and Global Health, Tufts University Cummings School of Veterinary Medicine, North Grafton, MA, United States
| | - Jamie Heimburg-Molinaro
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, United States
- National Center for Functional Glycomics, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Richard D. Cummings
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, United States
- National Center for Functional Glycomics, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- *Correspondence: Richard D. Cummings,
| |
Collapse
|
4
|
You H, Jones MK, Gordon CA, Arganda AE, Cai P, Al-Wassiti H, Pouton CW, McManus DP. The mRNA Vaccine Technology Era and the Future Control of Parasitic Infections. Clin Microbiol Rev 2023; 36:e0024121. [PMID: 36625671 PMCID: PMC10035331 DOI: 10.1128/cmr.00241-21] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Despite intensive long-term efforts, with very few exceptions, the development of effective vaccines against parasitic infections has presented considerable challenges, given the complexity of parasite life cycles, the interplay between parasites and their hosts, and their capacity to escape the host immune system and to regulate host immune responses. For many parasitic diseases, conventional vaccine platforms have generally proven ill suited, considering the complex manufacturing processes involved and the costs they incur, the inability to posttranslationally modify cloned target antigens, and the absence of long-lasting protective immunity induced by these antigens. An effective antiparasite vaccine platform is required to assess the effectiveness of novel vaccine candidates at high throughput. By exploiting the approach that has recently been used successfully to produce highly protective COVID mRNA vaccines, we anticipate a new wave of research to advance the use of mRNA vaccines to prevent parasitic infections in the near future. This article considers the characteristics that are required to develop a potent antiparasite vaccine and provides a conceptual foundation to promote the development of parasite mRNA-based vaccines. We review the recent advances and challenges encountered in developing antiparasite vaccines and evaluate the potential of developing mRNA vaccines against parasites, including those causing diseases such as malaria and schistosomiasis, against which vaccines are currently suboptimal or not yet available.
Collapse
Affiliation(s)
- Hong You
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Malcolm K. Jones
- School of Veterinary Science, The University of Queensland, Brisbane, Australia
| | - Catherine A. Gordon
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Alexa E. Arganda
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Pengfei Cai
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Harry Al-Wassiti
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Colin W. Pouton
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Donald P. McManus
- Department of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| |
Collapse
|
5
|
El-Derbawy MM, Salem HS, Raboo M, Baiuomy IR, Fadil SA, Fadil HA, Ibrahim SRM, El Kholy WA. In Vivo Evaluation of the Anti-Schistosomal Potential of Ginger-Loaded Chitosan Nanoparticles on Schistosoma mansoni: Histopathological, Ultrastructural, and Immunological Changes. Life (Basel) 2022; 12:life12111834. [PMID: 36362992 PMCID: PMC9696985 DOI: 10.3390/life12111834] [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: 09/29/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/12/2022] Open
Abstract
Chemotherapy is the most widely advocated method of Schistosome control. However, repeated chemotherapy leads to the emergence of drug-resistant Schistosoma strains. Therefore, efforts to find alternative drugs, especially those of natural origin, have risen globally. Nanoparticles (NPs) have received special interest as efficient drug delivery systems. This work aimed to investigate the anti-schistosomal potential of Zingiber officinale (ginger, Zingiberaceae)-loaded chitosan nanoparticles (GCsNPs) on Schistosoma mansoni experimentally infected mice that were exposed to 80 ± 10 cercariae/mouse. The study groups are: (G1) negative control; (G2) positive control; (G3) praziquantel in a dose of 500 mg/kg/day for two consecutive days; (G4) ginger in a dose of 500 mg/kg treated; (G5) chitosan nanoparticles in a dose 3 mg/kg (G6) GCsNPs in a dose 250 mg/kg; and (G7) GCsNPs in a dose 500 mg/kg. The anti-schistosome potential was assessed using histopathological scanning electron microscopically and immunological parameters. The results showed that there was a significant decrease in cellular granuloma count (p < 0.05) and granuloma diameter (p < 0.001) in all infected treated mice groups, in comparison to the infected non-treated group with the highest reduction in both G3 and G7. SEM of S. mansoni adult worm recovered from G3 showed mild edema of oral and ventral suckers with some peeling and blebs around them, while that recovered from G7 showed abnormal oedematous oral and retracted ventral sucker, edema of the tegument, rupture of many tubercles with vacuolation and complete loss of spines. All infected treated mice groups, in comparison to positive control G2, showed a significant reduction in IL-4, IL-10, and TNF-α levels (p-value < 0.001), especially groups G6 and G7 (p-value < 0.05); both G6 and G7 values were nearer to the normal that indicated recovery of the liver tissue.
Collapse
Affiliation(s)
- Mona M. El-Derbawy
- Department of Medical Parasitology, Faculty of Medicine, Al-Azhar University, Cairo 11751, Egypt
| | - Hala S. Salem
- Department of Medical Parasitology, Faculty of Medicine, Al-Azhar University, Cairo 11751, Egypt
| | - Mona Raboo
- Department of Medical Parasitology, Faculty of Medicine, Al-Azhar University, Cairo 11751, Egypt
| | - Ibrahim R. Baiuomy
- Department of Immunology and Parasitology, Theodor Bilharz Research Institute, Giza 12411, Egypt
| | - Sana A. Fadil
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Haifa A. Fadil
- Department of Clinical and Hospital Pharmacy, Faculty of Pharmacy, Taibah University, Almadinah Almunawarah 30078, Saudi Arabia
| | - Sabrin R. M. Ibrahim
- Preparatory Year Program, Department of Chemistry, Batterjee Medical College, Jeddah 21442, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
- Correspondence: ; Tel.: +966-581183034
| | - Walaa A. El Kholy
- Department of Medical Parasitology, Faculty of Medicine, Al-Azhar University, Cairo 11751, Egypt
| |
Collapse
|
6
|
Hanton AJ, Scott F, Stenzel K, Nausch N, Zdesenko G, Mduluza T, Mutapi F. Frequency distribution of cytokine and associated transcription factor single nucleotide polymorphisms in Zimbabweans: Impact on schistosome infection and cytokine levels. PLoS Negl Trop Dis 2022; 16:e0010536. [PMID: 35759449 PMCID: PMC9236240 DOI: 10.1371/journal.pntd.0010536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 05/25/2022] [Indexed: 11/18/2022] Open
Abstract
Cytokines mediate T-helper (TH) responses that are crucial for determining the course of infection and disease. The expression of cytokines is regulated by transcription factors (TFs). Here we present the frequencies of single nucleotide polymorphisms (SNPs) in cytokine and TF genes in a Zimbabwean population, and further relate SNPs to susceptibility to schistosomiasis and cytokine levels. Individuals (N = 850) were genotyped for SNPs across the cytokines IL4, IL10, IL13, IL33, and IFNG, and their TFs STAT4, STAT5A/B, STAT6, GATA3, FOXP3, and TBX21 to determine allele frequencies. Circulatory levels of systemic and parasite-specific IL-4, IL-5, IL-10, IL-13, and IFNγ were quantified via enzyme-linked immunosorbent assay. Schistosoma haematobium infection was determined by enumerating parasite eggs excreted in urine by microscopy. SNP allele frequencies were related to infection status by case-control analysis and logistic regression, and egg burdens and systemic and parasite-specific cytokine levels by analysis of variance and linear regression. Novel findings were i) IL4 rs2070874*T's association with protection from schistosomiasis, as carriage of ≥1 allele gave an odds ratio of infection of 0.597 (95% CIs, 0.421-0.848, p = 0.0021) and IFNG rs2069727*G's association with susceptibility to schistosomiasis as carriage of ≥1 allele gave an odds ratio of infection of 1.692 (1.229-2.33, p = 0.0013). Neither IL4 rs2070874*T nor IFNG rs2069727*G were significantly associated with cytokine levels. This study found TH2-upregulating SNPs were more frequent among the Zimbabwean sample compared to African and European populations, highlighting the value of immunogenetic studies of African populations in the context of infectious diseases and other conditions, including allergic and atopic disease. In addition, the identification of novel infection-associated alleles in both TH1- and TH2-associated genes highlights the role of both in regulating and controlling responses to Schistosoma.
Collapse
Affiliation(s)
- Andrew John Hanton
- Institute of Immunology & Infection Research, University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
- NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
| | - Fiona Scott
- Institute of Immunology & Infection Research, University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
- NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
| | - Katharina Stenzel
- Institute of Immunology & Infection Research, University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
| | - Norman Nausch
- Institute of Immunology & Infection Research, University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
| | - Grace Zdesenko
- Institute of Immunology & Infection Research, University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
- NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
| | - Takafira Mduluza
- Department of Biochemistry, University of Zimbabwe, Harare, Zimbabwe
| | - Francisca Mutapi
- Institute of Immunology & Infection Research, University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
- NIHR Global Health Research Unit Tackling Infections to Benefit Africa (TIBA), University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
| |
Collapse
|
7
|
Nono JK, Mpotje T, Mosala P, Aziz NA, Musaigwa F, Hlaka L, Spangenberg T, Brombacher F. Praziquantel Treatment of Schistosoma mansoni Infected Mice Renders Them Less Susceptible to Reinfection. Front Immunol 2021; 12:748387. [PMID: 34956183 PMCID: PMC8703194 DOI: 10.3389/fimmu.2021.748387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/23/2021] [Indexed: 11/13/2022] Open
Abstract
Beyond transient control of the infection, additional benefits of mass drug administration of praziquantel in endemic communities have been suggested in communities but not mechanistically investigated experimentally. The present study sought to evaluate the additional and hitherto unreported benefits of repeated mass drug administration of praziquantel. We used a tractable mouse model of Schistosoma mansoni infection to assess the effects of repeated infection-treatment cycles on the host susceptibility to reinfection. Parasitaemia was assessed by quantification of Schistosoma egg burden in liver tissues and morbidity was followed up by histological observation of liver lesions by microscopy and using biochemical measurement of liver transaminases. Immune responses were further determined by serum probing of schistosoma-specific antibodies, cytokines and quantification of liver cellular and soluble mediator responses by flow cytometry and ELISA, respectively. At similar ages and comparable gender distribution, groups of mice undergoing higher number of infections treatment cycles over a longer period, remained susceptible to reinfection by the parasite, as judged by the presence of eggs and the associated increasing pathology in the liver tissues. However, notably, there was a clear and significantly higher propensity to lower egg burden upon reinfection when compared to counterparts undergoing a lower number of infection-treatment cycles. This relative reduction of susceptibility to infection was paralleled by a more robust humoral response against parasite antigens, elevated serum IL-4 and liver cytokines. Of note, praziquantel treatment of infected mice left them at a higher baseline of serum IL-4, IgE and liver cytokines but lower CD4+ T cell -derived cytokines when compared to infected non-treated mice supporting an immunological treatment-induced advantage of previously infected mice over naïve mice and infected/not treated mice. Notably, repeated infection-treatment cycles did not preclude the infection-driven aggravation of collagen deposition in the livers over time and was corroborated by a more robust local production of inflammatory cytokines in the most exposed livers. Taken together, our data reveal that treatment of S. mansoni-infected hosts with praziquantel rewires the immune system to a conformation less permissive to subsequent reinfection in mice. Provided the data are translatable from mouse to human, our findings may provide mechanistic support to the potential benefits of more frequent MDAs in high transmission areas to allow rapid acquisition of protective immunity against reinfection.
Collapse
Affiliation(s)
- Justin Komguep Nono
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Laboratory of ImmunoBiology and Helminth Infections, Institute of Medical Research and Medicinal Plant Studies, Ministry of Scientific Research and Innovation, Yaoundé, Cameroon
| | - Thabo Mpotje
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
| | - Paballo Mosala
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
| | - Nada Abdel Aziz
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Chemistry Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Fungai Musaigwa
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
| | - Lerato Hlaka
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
| | - Thomas Spangenberg
- Global Health Institute of Merck, Ares Trading S.A., a Subsidiary of Merck KGaA Darmstadt Germany, Eysins, Switzerland
- *Correspondence: Thomas Spangenberg, ; Frank Brombacher,
| | - Frank Brombacher
- Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- *Correspondence: Thomas Spangenberg, ; Frank Brombacher,
| |
Collapse
|
8
|
Liu C, Zhang W, Wang J, Si T, Xing W. Tumor-associated macrophage-derived transforming growth factor-β promotes colorectal cancer progression through HIF1-TRIB3 signaling. Cancer Sci 2021; 112:4198-4207. [PMID: 34375482 PMCID: PMC8486199 DOI: 10.1111/cas.15101] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor-associated macrophages (TAMs), one of the most common cell components in the tumor microenvironment, have been reported as key contributors to cancer-related inflammation and enhanced metastatic progression of tumors. To explore the underlying mechanism of TAM-induced tumor progression, TAMs were isolated from colorectal cancer patients, and the functional interaction with colorectal cancer cells was analyzed. Our study found that coculture of TAMs contributed to a glycolytic state in colorectal cancer, which promoted the stem-like phenotypes and invasion of tumor cells. TAMs produced the cytokine transforming growth factor-β to support hypoxia-inducible factor 1α (HIF1α) expression, thereby upregulating Tribbles pseudokinase 3 (TRIB3) in tumor cells. Elevated expression of TRIB3 resulted in activation of the β-catenin/Wnt signaling pathway, which eventually enhanced the stem-like phenotypes and cell invasion in colorectal cancer. Our findings provided evidence that TAMs promoted colorectal cancer progression in a HIF1α/TRIB3-dependent manner, and blockade of HIF1α signals efficiently improved the outcome of chemotherapy, describing an innovative approach for colorectal cancer treatment.
Collapse
Affiliation(s)
- Changfu Liu
- Department of Interventional TreatmentNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin’s Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Weihao Zhang
- Department of Interventional TreatmentNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin’s Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Junfeng Wang
- Department of Colorectal CancerNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin’s Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Tongguo Si
- Department of Interventional TreatmentNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin’s Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Wenge Xing
- Department of Interventional TreatmentNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin’s Clinical Research Center for CancerTianjin Medical University Cancer Institute and HospitalTianjinChina
| |
Collapse
|
9
|
Marume A, Vengesai A, Mann J, Mduluza T. Interleukin-10 and tumour necrosis factor alpha promoter region polymorphisms and susceptibility to urogenital schistosomiasis in young Zimbabwean children living in Schistosoma haematobium endemic regions. S Afr J Infect Dis 2021; 35:11. [PMID: 34485462 PMCID: PMC8378000 DOI: 10.4102/sajid.v35i1.11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 06/23/2020] [Indexed: 11/11/2022] Open
Abstract
Background Host genetic factors can influence susceptibility, morbidity and mortality from schistosomiasis. The study explored the association between single nucleotide polymorphisms (SNPs) in interleukin-10 (IL-10) and tumour necrosis factor alpha (TNF-α) promoter regions and susceptibility to Schistosoma haematobium infection. Methods Urine specimens were collected from 361 primary school children aged 5–15 years from schistosomiasis endemic areas of Manicaland and Mashonaland central provinces. Schistosoma haematobium was diagnosed using the urine filtration method. Only 272 participants provided adequate blood for genotyping. Genotyping was performed using the amplification refractory mutation system-polymerase chain reaction. The association between IL-10 and TNF-α SNPs and S. haematobium infection was analysed using the chi-square test. Results Schistosoma haematobium infection was confirmed in 26.8% of the participants. No significant difference in S. haematobium prevalence between men (51.6% of those infected) and women (48.4%) (χ2 = 0.008, df = 1, p = 0.928) was observed. The total IL-10 -1082 G, IL-10 -819 C and TNF-α -308G allele distribution between S. haematobium infected and uninfected participants was 50.7% and 51.5% (χ2 = 0.025, df = 1, p = 0.87), 54.3% and 60.6% (χ2 = 1.187, df = 1, p = 0.187) and 82.1% and 80.9% (χ2 = 0.099, df = 1, p = 0.753), respectively, and the differences were not significant. Conclusion Interleukin-10 -1082 G/A, IL-10 -819 C/T and TNF-α -308 G/A SNPs were not significantly associated with susceptibility to S. haematobium infection. The prevalence of schistosomiasis is still in the moderate range and is similar in boys and girls.
Collapse
Affiliation(s)
- Amos Marume
- Department of Infection Prevention and Control, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.,Paraclinical Department, Faculty of Veterinary Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Arthur Vengesai
- Department of Infection Prevention and Control, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.,Department of Biochemistry, University of Zimbabwe, Harare, Zimbabwe
| | - Jaclyn Mann
- Department of Infection Prevention and Control, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Takafira Mduluza
- Department of Infection Prevention and Control, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.,Department of Biochemistry, University of Zimbabwe, Harare, Zimbabwe
| |
Collapse
|
10
|
Xiong D, Luo S, Wu K, Yu Y, Sun J, Wang Y, Hu J, Hu W. Transcriptional profiling of Microtus fortis responses to S. japonicum: New sight into Mf-Hsp90α resistance mechanism. Parasite Immunol 2021; 43:e12842. [PMID: 33959966 PMCID: PMC8365665 DOI: 10.1111/pim.12842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 04/23/2021] [Accepted: 04/30/2021] [Indexed: 12/22/2022]
Abstract
AIMS Schistosomiasis is a parasitic disease with a chronic debilitating character caused by parasitic flatworms of the genus Schistosoma. The main disease-causing species of Schistosoma in China is S. japonicum. M fortis has been proved to be a nonpermissive host of S. japonicum. Mf-HSP90α (Microtus fortis heat shock protein 90alpha), the homologue of HSP90α, display anti-schistosome effect in vitro and in vivo. In the current study, in order to investigate the mechanism of anti-schistosome effect of Mf-HSP90α, we conducted RNA-Seq to obtain the transcriptome profile of M. fortis liver infected with S. japonicum at different time points. METHODS AND RESULTS By mapping the differential expressed genes (DEGs) to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), we found that the JAK2/STAT1 pathway was highly enriched with an elevated level of IL-10 and HSP90α. We then checked the IL-10-JAK2/STAT1-HSP90α pathway, and found that this pathway was activated in the infected mice with S. japonicum. The expression of the molecules in this pathway was elevated on the 10th day after infection and gradually decreased on the 20th day. CONCLUSIONS The IL-10-JAK2/STAT1-HSP90α axis was associated with the anti-schistosome effect of Mf-HSP90α, and targeting IL-10-JAK2/STAT1-HSP90α axis might be a novel therapeutic strategy for developing resistance to S. japonicum infection.
Collapse
Affiliation(s)
- Dehui Xiong
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha, China
| | - Saiqun Luo
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha, China
| | - Kunlu Wu
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha, China
| | - Yuanjing Yu
- Department of Laboratory Animal, Central South University, Changsha, China
| | - Jiameng Sun
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha, China
| | - Yanpeng Wang
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha, China
| | - Jingping Hu
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha, China
| | - Weixin Hu
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha, China
| |
Collapse
|
11
|
Marume A, Chimponda T, Vengesai A, Mushayi C, Mann J, Mduluza T. Effects of TNF-α and IL-10-819 T>C single nucleotide polymorphisms on urogenital schistosomiasis in preschool children in Zimbabwe. Afr J Lab Med 2021; 10:1138. [PMID: 34007813 PMCID: PMC8111665 DOI: 10.4102/ajlm.v10i1.1138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/28/2021] [Indexed: 12/02/2022] Open
Abstract
Background Knowledge gaps exist between host genetic factors and susceptibility to schistosomiasis. Objective This study determined cytokine levels and single nucleotide polymorphisms of tumour necrosis factor (TNF)-α (rs1800629) and interleukin (IL)-10 (rs1800871) and their possible impact on susceptibility to schistosomiasis in preschool-age children in the Madziva area of Shamva district, Mashonaland Central province, Zimbabwe. Methods Urogenital schistosomiasis was diagnosed using the urine filtration method, while a sandwich enzyme-linked immunosorbent assay was used for cytokine level determination. The survey was done in August 2015 and reinfection levels post treatment were assessed at 3, 6 and 12 months. Amplification refractory mutation system polymerase chain reaction with visualisation on 2% agarose gel electrophoresis was used for genotyping. Results Schistosomiasis prevalence was found to be 10.5% (59/563). Reinfections were detected in only six children at 3 months and only one was reinfected at 12 months. There were no significant differences in TNF-α-308 G/A allele or genotype frequencies between the Schistosoma haematobium infected participants (p = 0.360) and uninfected participants (p = 0.279). However, no children with the IL-10-819 TT genotype had schistosomiasis. The TNF-α GG genotype corresponded with significantly lower TNF-α levels when compared with the GA or AA genotypes (p < 0.001), and TNF-α levels were significantly lower in infected children compared to uninfected children (p < 0.001). Conclusion Higher TNF-α levels and lower IL-10 levels are potentially protective against schistosomiasis infection. The IL-10-819 TT genotype is potentially protective against infection through its association with lower IL-10 levels.
Collapse
Affiliation(s)
- Amos Marume
- Department of Infection Prevention and Control, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.,Paraclinical Department, Faculty of Veterinary Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Theresa Chimponda
- Department of Biochemistry, Faculty of Science, University of Zimbabwe, Harare, Zimbabwe
| | - Arthur Vengesai
- Department of Biochemistry, Faculty of Science, University of Zimbabwe, Harare, Zimbabwe
| | - Caroline Mushayi
- Department of Biochemistry, Faculty of Science, University of Zimbabwe, Harare, Zimbabwe
| | - Jaclyn Mann
- Department of Infection Prevention and Control, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Takafira Mduluza
- Department of Infection Prevention and Control, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.,Department of Biochemistry, Faculty of Science, University of Zimbabwe, Harare, Zimbabwe
| |
Collapse
|
12
|
Molehin AJ. Current Understanding of Immunity Against Schistosomiasis: Impact on Vaccine and Drug Development. Res Rep Trop Med 2020; 11:119-128. [PMID: 33173371 PMCID: PMC7646453 DOI: 10.2147/rrtm.s274518] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 09/29/2020] [Indexed: 12/17/2022] Open
Abstract
Schistosomiasis is a neglected tropical disease inflicting significant morbidity in humans worldwide. The disease is caused by infections with a parasitic trematode belonging to the genus Schistosoma. Over 250 million people are currently infected globally, with an estimated disability-adjusted life-years of 1.9 million attributed to the disease. Current understanding, based on several immunological studies using experimental and human models of schistosomiasis, reveals that complex immune mechanisms play off each other in the acquisition of immune resistance to infection/reinfection. Nevertheless, the precise characteristics of these responses, the specific antigens against which they are elicited, and how these responses are intricately regulated are still being investigated. What is apparent is that immunity to schistosome infections develops slowly and over a prolonged period of time, augmented by the death of adult worms occurring naturally or by praziquantel therapy. In this review, aspects of immunity to schistosomiasis, host–parasite interactions and their impact on schistosomiasis vaccine development are discussed.
Collapse
Affiliation(s)
- Adebayo J Molehin
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.,Center for Tropical Medicine and Infectious Diseases, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
13
|
Mechanism by which the combination of SjCL3 and SjGAPDH protects against Schistosoma japonicum infection. Parasitol Res 2020; 120:173-185. [PMID: 33079271 DOI: 10.1007/s00436-020-06916-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 10/04/2020] [Indexed: 10/23/2022]
Abstract
A vaccine is an important method to control schistosomiasis. Molecules related to lung-stage schistosomulum are considered potential vaccine candidates. We previously showed that glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and cathepsin L3 (CL3) displayed differential expression in the lung-stage schistosomula of Schistosoma japonicum cocultured with host cells. In the present study, we prepared the two proteins and detected the protective effects of SjGAPDH by immunizing mice with this protein alone and in combination with SjCL3 with or without Freund's adjuvant. Then, we investigated the possible mechanisms underlying S. japonicum infection. The results showed that vaccination of adjuvanted SjGAPDH decreased the worm burden (37.8%) and egg load (38.1%), and the combination of adjuvanted SjGAPDH and SjCL3 further decreased the worm burden (65.6%) and egg load (70.9%) during Schistosoma japonicum infection. However, the immunization of a combination of adjuvant-free SjGAPDH and SjCL3 displayed a lower protective effect (< 15%) than those of the adjuvanted SjCL3, the adjuvanted SjGAPDH, and a combination of adjuvanted SjGAPDH and SjCL3. Flow cytometric results showed that the frequency of regulatory T cells (Tregs) was lower (P < 0.05) in the group with adjuvanted SjGAPDH and SjCL3 (2.61%) than the remaining groups. The enzyme-linked immunosorbent assay (ELISA) results indicated that except for the uninfected and infected control groups, the remaining groups displayed a Th1-type shift in immune responses. These results showed the immunization of SjGAPDH resulted in partial protection (approximately 38%); inoculation with a combination of SjCL3 and SjGAPDH in Freund's adjuvant resulted in a high immunoprotective effect (> 65%) against Schistosoma japonicum infection in mice, which was possibly caused by the reduced percentage of Tregs and a Th1-type shift in immune responses; and SjCL3 has no adjuvant-like effect, dissimilar to SmCL3.
Collapse
|
14
|
Ayelign B, Akalu Y, Teferi B, Molla MD, Shibabaw T. Helminth Induced Immunoregulation and Novel Therapeutic Avenue of Allergy. J Asthma Allergy 2020; 13:439-451. [PMID: 33116652 PMCID: PMC7548329 DOI: 10.2147/jaa.s273556] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/17/2020] [Indexed: 12/24/2022] Open
Abstract
Allergic diseases are increasing at an alarming rate worldwide, particularly in developed countries. In contrast, there is a decrease in the prevalence of helminthic infections and other neglected diseases. The hygiene hypothesis elaborates parasitic infection, and allergy-associated diseases have an inverse relationship. Acute helminthic infection and allergic reaction stimulate Type 2 helper cells (Th2) immune response with up-regulation of cytokines IL-4-, IL-5-, and IL-13-mediated IgE and mast cell production, as well as eosinophilia. However, people who chronically suffer from helminthic infections are demarcated through polarized Th2 resulting in alternative macrophage activation and T regulatory response. This regulatory system reduces allergy incidence in individuals that are chronically diseased through helminth. As a result, the excretory-secretory (ES) substance derived from parasites and extracellular vesicular components can be used as a novel therapeutic modality of allergy. Therefore, the aim of this review meticulously explored the link between helminth infection and allergy, and utilization of the helminth secretome for therapeutic immunomodulation.
Collapse
Affiliation(s)
- Birhanu Ayelign
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Yonas Akalu
- Department of Physiology, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Banchamlak Teferi
- Department of Clinical Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Meseret Derbew Molla
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Tewodros Shibabaw
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
15
|
Preventive CTLA-4-Ig Treatment Reduces Hepatic Egg Load and Hepatic Fibrosis in Schistosoma mansoni-Infected Mice. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1704238. [PMID: 31950032 PMCID: PMC6948272 DOI: 10.1155/2019/1704238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 10/02/2019] [Accepted: 11/12/2019] [Indexed: 11/21/2022]
Abstract
Background Hepatic fibrosis and granuloma formation as a consequence of tissue entrapped eggs produced by female schistosomes characterize the pathology of Schistosoma mansoni infection. We have previously shown that single-sex infection with female schistosomes mitigates hepatic fibrosis after secondary infection. This was associated with an increased expression of cytotoxic T-lymphocyte-associated protein-4 (CTLA-4), known as a negative regulator of T cell activation. Based on these findings, we hypothesized that administration of agonistic CTLA-4-Ig (Belatacept) is capable to prevent and/or treat hepatic fibrosis during schistosomiasis. Methods Mice were infected with 50 S. mansoni cercariae and CTLA-4-Ig, or appropriated control-Ig was administered for 4 weeks. Preventive treatment started 4 weeks after infection, before onset of egg production, and therapeutic treatment started 8 weeks after infection when hepatic fibrosis was already established. Results When given early after infection, livers of CTLA-4-Ig-treated mice showed significantly reduced collagen deposition and decreased expression of profibrotic genes in comparison to controls. In addition, administration of CTLA-4-Ig suppressed the inflammatory T cell response in infected mice. If therapy was started at a later time point when fibrogenesis was initiated, CTLA-4-Ig had no impact on hepatic fibrosis. Conclusion We could demonstrate that an early preventive administration of CTLA-4-Ig suppresses effector T cell function and therefore ameliorates liver fibrosis. CTLA-4-Ig administration after onset of egg production fails to treat hepatic fibrosis.
Collapse
|
16
|
Ndombi EM, Abudho B, Kittur N, Carter JM, Korir H, Riner DK, Ochanda H, Lee YM, Secor WE, Karanja DM, Colley DG. Effect of four rounds of annual school-wide mass praziquantel treatment for schistosoma mansoni control on schistosome-specific immune responses. Parasite Immunol 2018; 40:e12530. [PMID: 29604074 PMCID: PMC6001474 DOI: 10.1111/pim.12530] [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] [Received: 10/23/2017] [Accepted: 03/22/2018] [Indexed: 11/27/2022]
Abstract
This study evaluated potential changes in antischistosome immune responses in children from schools that received 4 rounds of annual mass drug administration (MDA) of praziquantel (PZQ). In a repeated cross‐sectional study design, 210 schistosome egg‐positive children were recruited at baseline from schools in western Kenya (baseline group). Another 251 children of the same age range were recruited from the same schools and diagnosed with schistosome infection by microscopy (post‐MDA group). In‐vitro schistosome‐specific cytokines and plasma antibody levels were measured by ELISA and compared between the 2 groups of children. Schistosome soluble egg antigen (SEA) and soluble worm antigen preparation (SWAP) stimulated higher IL‐5 production by egg‐negative children in the post‐MDA group compared to the baseline group. Similarly, anti‐SEA IgE levels were higher in egg‐negative children in the post‐MDA group compared to the baseline group. Anti‐SEA and anti‐SWAP IgG4 levels were lower in egg‐negative children in the post‐MDA group compared to baseline. This resulted in higher anti‐SEA IgE/IgG4 ratios for children in the post‐MDA group compared to baseline. These post‐MDA immunological changes are compatible with the current paradigm that treatment shifts immune responses to higher antischistosome IgE:IgG4 ratios in parallel with a potential increase in resistance to reinfection.
Collapse
Affiliation(s)
- E M Ndombi
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya.,School of Biological Sciences, University of Nairobi, Nairobi, Kenya.,Department of Pathology, Kenyatta University, Nairobi, Kenya
| | - B Abudho
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya.,Department of Biomedical Sciences, Maseno University, Maseno, Kenya
| | - N Kittur
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| | - J M Carter
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| | - H Korir
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - D K Riner
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| | - H Ochanda
- School of Biological Sciences, University of Nairobi, Nairobi, Kenya
| | - Y-M Lee
- Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - W E Secor
- Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - D M Karanja
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - D G Colley
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA.,Department of Microbiology, University of Georgia, Athens, GA, USA
| |
Collapse
|
17
|
Ondigo BN, Ndombi EM, Nicholson SC, Oguso JK, Carter JM, Kittur N, Secor WE, Karanja DMS, Colley DG. Functional Studies of T Regulatory Lymphocytes in Human Schistosomiasis in Western Kenya. Am J Trop Med Hyg 2018; 98:1770-1781. [PMID: 29692308 PMCID: PMC6086154 DOI: 10.4269/ajtmh.17-0966] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Immunoregulation is considered a common feature of Schistosoma mansoni infections, and elevated levels of T regulatory (Treg) lymphocytes have been reported during chronic human schistosomiasis. We now report that the removal of Treg (CD4+/CD25hi/CD127low lymphocytes) from peripheral blood mononuclear cells (PBMCs) of S. mansoni–infected individuals leads to increased levels of phytohemagglutinin (PHA)-stimulated interferon gamma (IFNγ) production and decreased interleukin-10 (IL-10) responses. Exposure to schistosome antigens did not result in measurable IFNγ by either PBMC or Treg-depleted populations. Interleukin-10 responses to soluble egg antigens (SEA) by PBMC were unchanged by Treg depletion, but the depletion of Treg greatly decreased IL-10 production to soluble worm antigenic preparation (SWAP). Proliferative responses to PHA increased upon Treg removal, but responses to SEA or SWAP did not, unless only initially low responders were evaluated. Addition of anti-IL-10 increased PBMC proliferative responses to either SEA or SWAP, but did not alter responses by Treg-depleted cells. Blockade by anti-transforming growth factor-beta (TGF-β) increased SEA but not SWAP proliferative responses by PBMC, whereas anti-TGF-β increased both SEA- and SWAP-stimulated responses by Treg-depleted cultures. Addition of both anti-IL-10 and anti-TGF-β to PBMC or Treg-depleted populations increased proliferation of both populations to either SEA or SWAP. These studies demonstrate that Treg appear to produce much of the antigen-stimulated IL-10, but other cell types or subsets of Treg may produce much of the TGF-β. The elevated levels of Treg seen in chronic schistosomiasis appear functional and involve IL-10 and TGF-β in antigen-specific immunoregulation perhaps leading to regulation of immunopathology and/or possibly decreased immunoprotective responses.
Collapse
Affiliation(s)
- Bartholomew N Ondigo
- Department of Biochemistry and Molecular Biology, Egerton University, Nakuru, Kenya.,Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Eric M Ndombi
- Department of Pathology, Kenyatta University, Nairobi, Kenya.,Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Sarah C Nicholson
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| | - John K Oguso
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Jennifer M Carter
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| | - Nupur Kittur
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| | - W Evan Secor
- Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Diana M S Karanja
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Daniel G Colley
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia.,Department of Microbiology, University of Georgia, Athens, Georgia
| |
Collapse
|
18
|
Uhde AK, Ciurkiewicz M, Herder V, Khan MA, Hensel N, Claus P, Beckstette M, Teich R, Floess S, Baumgärtner W, Jung K, Huehn J, Beineke A. Intact interleukin-10 receptor signaling protects from hippocampal damage elicited by experimental neurotropic virus infection of SJL mice. Sci Rep 2018; 8:6106. [PMID: 29666403 PMCID: PMC5904160 DOI: 10.1038/s41598-018-24378-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 03/27/2018] [Indexed: 12/24/2022] Open
Abstract
Theiler’s murine encephalomyelitis virus (TMEV) infection represents an experimental mouse model to study hippocampal damage induced by neurotropic viruses. IL-10 is a pleiotropic cytokine with profound anti-inflammatory properties, which critically controls immune homeostasis. In order to analyze IL-10R signaling following virus-induced polioencephalitis, SJL mice were intracerebrally infected with TMEV. RNA-based next generation sequencing revealed an up-regulation of Il10, Il10rα and further genes involved in IL-10 downstream signaling, including Jak1, Socs3 and Stat3 in the brain upon infection. Subsequent antibody-mediated blockade of IL-10R signaling led to enhanced hippocampal damage with neuronal loss and increased recruitment of CD3+ T cells, CD45R+ B cells and an up-regulation of Il1α mRNA. Increased expression of Tgfβ and Foxp3 as well as accumulation of Foxp3+ regulatory T cells and arginase-1+ macrophages/microglia was detected in the hippocampus, representing a potential compensatory mechanism following disturbed IL-10R signaling. Additionally, an increased peripheral Chi3l3 expression was found in spleens of infected mice, which may embody reactive regulatory mechanisms for prevention of excessive immunopathology. The present study highlights the importance of IL-10R signaling for immune regulation and its neuroprotective properties in the context of an acute neurotropic virus infection.
Collapse
Affiliation(s)
- Ann-Kathrin Uhde
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Malgorzata Ciurkiewicz
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Vanessa Herder
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Muhammad Akram Khan
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany.,Department of Pathobiology, Faculty of Veterinary & Animal Sciences, PMAS-Arid Agriculture University, Rawalpindi, Pakistan
| | - Niko Hensel
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
| | - Peter Claus
- Center for Systems Neuroscience, Hannover, Germany.,Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany.,Niedersachsen-Research Network on Neuroinfectiology (N-RENNT), Hannover, Germany
| | - Michael Beckstette
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - René Teich
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Stefan Floess
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany.,Niedersachsen-Research Network on Neuroinfectiology (N-RENNT), Hannover, Germany
| | - Klaus Jung
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany. .,Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
19
|
Sachdev D, Gough KC, Flynn RJ. The Chronic Stages of Bovine Fasciola hepatica Are Dominated by CD4 T-Cell Exhaustion. Front Immunol 2017; 8:1002. [PMID: 28871261 PMCID: PMC5566560 DOI: 10.3389/fimmu.2017.01002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/04/2017] [Indexed: 01/28/2023] Open
Abstract
Fasciola hepatica infection of ruminants leads to non-resolving chronic infection, as patency develops, there is switching to a TGF-β and IL-10 led response. Here, we explore the responses of CD4 T-cells within the major draining lymph nodes. We found minimal expression of Foxp3 within CD4 cells but elevated levels within the γδ (WC1+) population. There is a strong T-cell-intrinsic exhaustion phenotype within the hepatic lymph node (HLN) characterized by a lack of antigen-specific proliferation and cytokine secretion. CD4 T-cells recovered from the HLN had high levels of PD-1 expression and low levels of IL-2 secretion. Exogenous IL-2 partially rescued this defect; when combined with neutralization of IL-10 and TGF-β, full restoration of proliferation, and cytokine production was achieved. Moreover, there is a clear uncoupling of the mechanisms that facilitate this regulation with parasite-specific proliferation and cytokine secretion being governed by independent means. These data would suggest that there is a CD4 T-cell-intrinsic regulation in place early in chronic infection, potentially leading to failure in resistance to reinfection.
Collapse
Affiliation(s)
- Divya Sachdev
- School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Kevin C Gough
- School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Robin J Flynn
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
20
|
Lynch M, Walsh TA, Marszalowska I, Webb AE, Mac Aogain M, Rogers TR, Windle H, Kelleher D, O'Connell MJ, Loscher CE. Surface layer proteins from virulent Clostridium difficile ribotypes exhibit signatures of positive selection with consequences for innate immune response. BMC Evol Biol 2017; 17:90. [PMID: 28335725 PMCID: PMC5364705 DOI: 10.1186/s12862-017-0937-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/15/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Clostridium difficile is a nosocomial pathogen prevalent in hospitals worldwide and increasingly common in the community. Sequence differences have been shown to be present in the Surface Layer Proteins (SLPs) from different C. difficile ribotypes (RT) however whether these differences influence severity of infection is still not clear. RESULTS We used a molecular evolutionary approach to analyse SLPs from twenty-six C. difficile RTs representing different slpA sequences. We demonstrate that SLPs from RT 027 and 078 exhibit evidence of positive selection (PS). We compared the effect of these SLPs to those purified from RT 001 and 014, which did not exhibit PS, and demonstrate that the presence of sites under positive selection correlates with ability to activate macrophages. SLPs from RTs 027 and 078 induced a more potent response in macrophages, with increased levels of IL-6, IL-12p40, IL-10, MIP-1α, MIP-2 production relative to RT 001 and 014. Furthermore, RTs 027 and 078 induced higher expression of CD40, CD80 and MHC II on macrophages with decreased ability to phagocytose relative to LPS. CONCLUSIONS These results tightly link sequence differences in C. difficile SLPs to disease susceptibility and severity, and suggest that positively selected sites in the SLPs may play a role in driving the emergence of hyper-virulent strains.
Collapse
Affiliation(s)
- Mark Lynch
- Immunomodulation Research Group, School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Thomas A Walsh
- Bioinformatics and Molecular Evolution Group, School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Izabela Marszalowska
- Immunomodulation Research Group, School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Andrew E Webb
- Bioinformatics and Molecular Evolution Group, School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Micheál Mac Aogain
- Department of Clinical Microbiology, Trinity College Dublin, St James Hospital Dublin, Dublin, Ireland
| | - Thomas R Rogers
- Department of Clinical Microbiology, Trinity College Dublin, St James Hospital Dublin, Dublin, Ireland
| | - Henry Windle
- Institute of Molecular Medicine, Trinity College Dublin, Dublin, Ireland
| | - Dermot Kelleher
- Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Mary J O'Connell
- Bioinformatics and Molecular Evolution Group, School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland. .,Computational and Molecular Evolutionary Biology Research Group, School of Biology, Faculty of Biological Sciences, The University of Leeds, Leeds, LS2 9JT, UK.
| | - Christine E Loscher
- Immunomodulation Research Group, School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| |
Collapse
|
21
|
Jagannathan P, Bowen K, Nankya F, McIntyre TI, Auma A, Wamala S, Sikyomu E, Naluwu K, Nalubega M, Boyle MJ, Farrington LA, Bigira V, Kapisi J, Aweeka F, Greenhouse B, Kamya M, Dorsey G, Feeney ME. Effective Antimalarial Chemoprevention in Childhood Enhances the Quality of CD4+ T Cells and Limits Their Production of Immunoregulatory Interleukin 10. J Infect Dis 2016; 214:329-38. [PMID: 27067196 DOI: 10.1093/infdis/jiw147] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Accepted: 04/04/2016] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Experimental inoculation of viable Plasmodium falciparum sporozoites administered with chemoprevention targeting blood-stage parasites results in protective immunity. It is unclear whether chemoprevention similarly enhances immunity following natural exposure to malaria. METHODS We assessed P. falciparum-specific T-cell responses among Ugandan children who were randomly assigned to receive monthly dihydroartemisinin-piperaquine (DP; n = 87) or no chemoprevention (n = 90) from 6 to 24 months of age, with pharmacologic assessments for adherence, and then clinically followed for an additional year. RESULTS During the intervention, monthly DP reduced malaria episodes by 55% overall (P < .001) and by 97% among children who were highly adherent to DP (P < .001). In the year after the cessation of chemoprevention, children who were highly adherent to DP had a 55% reduction in malaria incidence as compared to children given no chemoprevention (P = .004). Children randomly assigned to receive DP had higher frequencies of blood-stage specific CD4(+) T cells coproducing interleukin-2 and tumor necrosis factor α (P = .003), which were associated with protection from subsequent clinical malaria and parasitemia, and fewer blood-stage specific CD4(+) T cells coproducing interleukin-10 and interferon γ (P = .001), which were associated with increased risk of malaria. CONCLUSIONS In this setting, effective antimalarial chemoprevention fostered the development of CD4(+) T cells that coproduced interleukin 2 and tumor necrosis factor α and were associated with prospective protection, while limiting CD4(+) T-cell production of the immunoregulatory cytokine IL-10.
Collapse
Affiliation(s)
| | | | | | | | - Ann Auma
- Infectious Diseases Research Collaboration
| | | | | | | | | | - Michelle J Boyle
- Department of Medicine, San Francisco General Hospital Center for Biomedical Research, The Burnet Institute, Melbourne, Australia
| | | | | | | | | | | | - Moses Kamya
- Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Grant Dorsey
- Department of Medicine, San Francisco General Hospital
| | - Margaret E Feeney
- Department of Medicine, San Francisco General Hospital Department of Pediatrics, University of California-San Francisco
| |
Collapse
|
22
|
Nogueira DS, Gazzinelli-Guimarães PH, Barbosa FS, Resende NM, Silva CC, de Oliveira LM, Amorim CCO, Oliveira FMS, Mattos MS, Kraemer LR, Caliari MV, Gaze S, Bueno LL, Russo RC, Fujiwara RT. Multiple Exposures to Ascaris suum Induce Tissue Injury and Mixed Th2/Th17 Immune Response in Mice. PLoS Negl Trop Dis 2016; 10:e0004382. [PMID: 26814713 PMCID: PMC4729520 DOI: 10.1371/journal.pntd.0004382] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 12/19/2015] [Indexed: 02/07/2023] Open
Abstract
Ascaris spp. infection affects 800 million people worldwide, and half of the world population is currently at risk of infection. Recurrent reinfection in humans is mostly due to the simplicity of the parasite life cycle, but the impact of multiple exposures to the biology of the infection and the consequences to the host's homeostasis are poorly understood. In this context, single and multiple exposures in mice were performed in order to characterize the parasitological, histopathological, tissue functional and immunological aspects of experimental larval ascariasis. The most important findings revealed that reinfected mice presented a significant reduction of parasite burden in the lung and an increase in the cellularity in the bronchoalveolar lavage (BAL) associated with a robust granulocytic pulmonary inflammation, leading to a severe impairment of respiratory function. Moreover, the multiple exposures to Ascaris elicited an increased number of circulating inflammatory cells as well as production of higher levels of systemic cytokines, mainly IL-4, IL-5, IL-6, IL-10, IL-17A and TNF-α when compared to single-infected animals. Taken together, our results suggest the intense pulmonary inflammation associated with a polarized systemic Th2/Th17 immune response are crucial to control larval migration after multiple exposures to Ascaris.
Collapse
Affiliation(s)
- Denise Silva Nogueira
- Laboratory of Immunology and Parasite Genomics, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Pedro Henrique Gazzinelli-Guimarães
- Laboratory of Immunology and Parasite Genomics, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernando Sérgio Barbosa
- Laboratory of Immunology and Parasite Genomics, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Nathália Maria Resende
- Laboratory of Immunology and Parasite Genomics, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Institute of Biological and Health Sciences, Universidade Federal do Mato Grosso, Cuiabá, Brazil
| | - Caroline Cavalcanti Silva
- Laboratory of Immunology and Parasite Genomics, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luciana Maria de Oliveira
- Laboratory of Immunology and Parasite Genomics, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Chiara Cássia Oliveira Amorim
- Laboratory of Immunology and Parasite Genomics, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fabrício Marcus Silva Oliveira
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Matheus Silvério Mattos
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lucas Rocha Kraemer
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marcelo Vidigal Caliari
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Soraya Gaze
- Laboratory of Cellular and Molecular Immunology, René Rachou Institute, Oswaldo Cruz Foundation–FIOCRUZ, Belo Horizonte, Brazil
| | - Lilian Lacerda Bueno
- Laboratory of Immunology and Parasite Genomics, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo Toshio Fujiwara
- Laboratory of Immunology and Parasite Genomics, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Institute of Biological and Health Sciences, Universidade Federal do Mato Grosso, Cuiabá, Brazil
| |
Collapse
|
23
|
Long X, Chen Q, Zhao J, Rafaels N, Mathias P, Liang H, Potee J, Campbell M, Zhang B, Gao L, Georas SN, Vercelli D, Beaty TH, Ruczinski I, Mathias R, Barnes KC, Chen X. An IL-13 promoter polymorphism associated with liver fibrosis in patients with Schistosoma japonicum. PLoS One 2015; 10:e0135360. [PMID: 26258681 PMCID: PMC4530950 DOI: 10.1371/journal.pone.0135360] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/21/2015] [Indexed: 12/20/2022] Open
Abstract
The aim of this study was to determine whether two polymorphisms in the gene encoding IL13 previously associated with Schistosoma hematobium (S. hematobium) and S. mansoni infection are associated with S. japonicum infection. Single nucleotide polymorphisms (SNPs) rs1800925 (IL13/-1112C>T) and rs20541 (IL13R130Q) were genotyped in 947 unrelated individuals (307 chronically infected, 339 late-stage with liver fibrosis, 301 uninfected controls) from a schistosomiasis-endemic area of Hubei province in China. Regression models were used to evaluate allelic and haplotypic associations with chronic and late-stage schistosomiasis adjusted for non-genetic covariates. Expression of IL-13 was measured in S. japonicun-infected liver fibrosis tissue and normal liver tissue from uninfected controls by immunohistochemistry (IHC). The role of rs1800925 in IL-13 transcription was further determined by Luciferase report assay using the recombinant PGL4.17-rs180092 plasmid. We found SNP rs1800925T was associated with late-stage schistosomiasis caused by S. japonicum but not chronic schistosomiasis (OR = 1.39, 95%CI = 1.02-1.91, p = 0.03) and uninfected controls (OR = 1.49, 95%CI = 1.03-2.13, p = 0.03). Moreover, the haplotype rs1800925T-rs20541C increased the risk of disease progression to late-stage schistosomiasis (OR = 1.46, p = 0.035), whereas haplotype rs1800925C-rs20541A showed a protective role against development of late-stage schistosomiasis (F = 0.188, OR = 0.61, p = 0.002). Furthermore, S. japonicum-induced fibrotic liver tissue had higher IL13 expression than normal liver tissue. Plasmid PGL4.17-rs1800925T showed a stronger relative luciferase activity than Plasmid PGL4.17-rs1800925C in 293FT, QSG-7701 and HL-7702 cell lines. In conclusion, the functional IL13 polymorphism, rs1800925T, previously associated with risk of schistosomiasis, also contributes to risk of late-stage schistosomiasis caused by S. japonicum.
Collapse
Affiliation(s)
- Xin Long
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei province, China
- Johns Hopkins Asthma & Allergy Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Qian Chen
- Division of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei province, China
| | - Jianping Zhao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei province, China
| | - Nicholas Rafaels
- Johns Hopkins Asthma & Allergy Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Priyanka Mathias
- Johns Hopkins Asthma & Allergy Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei province, China
| | - Joseph Potee
- Johns Hopkins Asthma & Allergy Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Monica Campbell
- Johns Hopkins Asthma & Allergy Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei province, China
| | - Li Gao
- Johns Hopkins Asthma & Allergy Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Steve N. Georas
- University of Rochester Medical Center, Rochester, New York, United States of America
| | - Donata Vercelli
- Arizona Respiratory Center, University of Arizona, Tucson, Arizona, United States of America
| | - Terri H. Beaty
- Bloomberg School of Public Health, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Ingo Ruczinski
- Bloomberg School of Public Health, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Rasika Mathias
- Johns Hopkins Asthma & Allergy Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Kathleen C. Barnes
- Johns Hopkins Asthma & Allergy Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei province, China
| |
Collapse
|
24
|
Sanin DE, Prendergast CT, Mountford AP. IL-10 Production in Macrophages Is Regulated by a TLR-Driven CREB-Mediated Mechanism That Is Linked to Genes Involved in Cell Metabolism. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:1218-32. [PMID: 26116503 PMCID: PMC4505959 DOI: 10.4049/jimmunol.1500146] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 05/27/2015] [Indexed: 12/14/2022]
Abstract
IL-10 is produced by macrophages in diverse immune settings and is critical in limiting immune-mediated pathology. In helminth infections, macrophages are an important source of IL-10; however, the molecular mechanism underpinning production of IL-10 by these cells is poorly characterized. In this study, bone marrow-derived macrophages exposed to excretory/secretory products released by Schistosoma mansoni cercariae rapidly produce IL-10 as a result of MyD88-mediated activation of MEK/ERK/RSK and p38. The phosphorylation of these kinases was triggered by TLR2 and TLR4 and converged on activation of the transcription factor CREB. Following phosphorylation, CREB is recruited to a novel regulatory element in the Il10 promoter and is also responsible for regulating a network of genes involved in metabolic processes, such as glycolysis, the tricarboxylic acid cycle, and oxidative phosphorylation. Moreover, skin-resident tissue macrophages, which encounter S. mansoni excretory/secretory products during infection, are the first monocytes to produce IL-10 in vivo early postinfection with S. mansoni cercariae. The early and rapid release of IL-10 by these cells has the potential to condition the dermal microenvironment encountered by immune cells recruited to this infection site, and we propose a mechanism by which CREB regulates the production of IL-10 by macrophages in the skin, but also has a major effect on their metabolic state.
Collapse
Affiliation(s)
- David E Sanin
- Department of Biology, Centre for Immunology and Infection, University of York, York YO10 5DD, United Kingdom
| | - Catriona T Prendergast
- Department of Biology, Centre for Immunology and Infection, University of York, York YO10 5DD, United Kingdom
| | - Adrian P Mountford
- Department of Biology, Centre for Immunology and Infection, University of York, York YO10 5DD, United Kingdom
| |
Collapse
|
25
|
Yepes E, Varela-M RE, López-Abán J, Rojas-Caraballo J, Muro A, Mollinedo F. Inhibition of Granulomatous Inflammation and Prophylactic Treatment of Schistosomiasis with a Combination of Edelfosine and Praziquantel. PLoS Negl Trop Dis 2015; 9:e0003893. [PMID: 26191954 PMCID: PMC4507859 DOI: 10.1371/journal.pntd.0003893] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 06/09/2015] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Schistosomiasis is the third most devastating tropical disease worldwide caused by blood flukes of the genus Schistosoma. This parasitic disease is due to immunologic reactions to Schistosoma eggs trapped in tissues. Egg-released antigens stimulate tissue-destructive inflammatory and granulomatous reactions, involving different immune cell populations, including T cells and granulocytes. Granulomas lead to collagen fibers deposition and fibrosis, resulting in organ damage. Praziquantel (PZQ) is the drug of choice for treating all species of schistosomes. However, PZQ kills only adult Schistosoma worms, not immature stages. The inability of PZQ to abort early infection or prevent re-infection, and the lack of prophylactic effect prompt the need for novel drugs and strategies for the prevention of schistosomiasis. METHODOLOGY/PRINCIPAL FINDINGS Using in vitro and in vivo approaches, we have found that the alkylphospholipid analog edelfosine kills schistosomula, and displays anti-inflammatory activity. The combined treatment of PZQ and edelfosine during a few days before and after cercariae infection in a schistosomiasis mouse model, simulating a prophylactic treatment, led to seven major effects: a) killing of Schistosoma parasites at early and late development stages; b) reduction of hepatomegaly; c) granuloma size reduction; d) down-regulation of Th1, Th2 and Th17 responses at late post-infection times, thus inhibiting granuloma formation; e) upregulation of IL-10 at early post-infection times, thus potentiating anti-inflammatory actions; f) down-regulation of IL-10 at late post-infection times, thus favoring resistance to re-infection; g) reduction in the number of blood granulocytes in late post-infection times as compared to infected untreated animals. CONCLUSIONS/SIGNIFICANCE Taken together, these data suggest that the combined treatment of PZQ and edelfosine promotes a high decrease in granuloma formation, as well as in the cellular immune response that underlies granuloma development, with changes in the cytokine patterns, and may provide a promising and effective strategy for a prophylactic treatment of schistosomiasis.
Collapse
Affiliation(s)
- Edward Yepes
- IBSAL-CIETUS (Instituto de Investigación Biomédica de Salamanca-Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca), Facultad de Farmacia, Universidad de Salamanca, Salamanca, Spain
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, CSIC-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Rubén E. Varela-M
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, CSIC-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Julio López-Abán
- IBSAL-CIETUS (Instituto de Investigación Biomédica de Salamanca-Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca), Facultad de Farmacia, Universidad de Salamanca, Salamanca, Spain
| | - Jose Rojas-Caraballo
- IBSAL-CIETUS (Instituto de Investigación Biomédica de Salamanca-Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca), Facultad de Farmacia, Universidad de Salamanca, Salamanca, Spain
| | - Antonio Muro
- IBSAL-CIETUS (Instituto de Investigación Biomédica de Salamanca-Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca), Facultad de Farmacia, Universidad de Salamanca, Salamanca, Spain
| | - Faustino Mollinedo
- Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer, CSIC-Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain
- * E-mail:
| |
Collapse
|
26
|
Schistosoma mansoni Larvae Do Not Expand or Activate Foxp3+ Regulatory T Cells during Their Migratory Phase. Infect Immun 2015. [PMID: 26195548 PMCID: PMC4567639 DOI: 10.1128/iai.00408-15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Foxp3+ regulatory T (Treg) cells play a key role in suppression of immune responses during parasitic helminth infection, both by controlling damaging immunopathology and by inhibiting protective immunity. During the patent phase of Schistosoma mansoni infection, Foxp3+ Treg cells are activated and suppress egg-elicited Th2 responses, but little is known of their induction and role during the early prepatent larval stage of infection. We quantified Foxp3+ Treg cell responses during the first 3 weeks of murine S. mansoni infection in C57BL/6 mice, a time when larval parasites migrate from the skin and transit the lungs en route to the hepatic and mesenteric vasculature. In contrast to other helminth infections, S. mansoni did not elicit a Foxp3+ Treg cell response during this early phase of infection. We found that the numbers and proportions of Foxp3+ Treg cells remained unchanged in the lungs, draining lymph nodes, and spleens of infected mice. There was no increase in the activation status of Foxp3+ Treg cells upon infection as assessed by their expression of CD25, Foxp3, and Helios. Furthermore, infection failed to induce Foxp3+ Treg cells to produce the suppressive cytokine interleukin 10 (IL-10). Instead, only CD4+ Foxp3− IL-4+ Th2 cells showed increased IL-10 production upon infection. These data indicate that Foxp3+ Treg cells do not play a prominent role in regulating immunity to S. mansoni larvae and that the character of the initial immune response invoked by S. mansoni parasites contrasts with the responses to other parasitic helminth infections that promote rapid Foxp3+ Treg cell responses.
Collapse
|
27
|
van der Ree AM, Mutapi F. The helminth parasite proteome at the host-parasite interface - Informing diagnosis and control. Exp Parasitol 2015; 157:48-58. [PMID: 26116863 DOI: 10.1016/j.exppara.2015.06.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 05/16/2015] [Accepted: 06/15/2015] [Indexed: 12/27/2022]
Abstract
Helminth parasites are a significant health burden for humans in the developing world and also cause substantial economic losses in livestock production across the world. The combined lack of vaccines for the major human and veterinary helminth parasites in addition to the development of drug resistance to anthelmintics in sheep and cattle mean that controlling helminth infection and pathology remains a challenge. However, recent high throughput technological advances mean that screening for potential drug and vaccine candidates is now easier than in previous decades. A better understanding of the host-parasite interactions occurring during infection and pathology and identifying pathways that can be therapeutically targeted for more effective and 'evolution proof' interventions is now required. This review highlights some of the advances that have been made in understanding the host-parasite interface in helminth infections using studies of the temporal expression of parasite proteins, i.e. the parasite proteome, and discuss areas for potential future research and translation.
Collapse
Affiliation(s)
- Anna M van der Ree
- Institute of Immunology & Infection Research, Centre for Immunity, Infection and Evolution, University of Edinburgh, Edinburgh, UK
| | - Francisca Mutapi
- Institute of Immunology & Infection Research, Centre for Immunity, Infection and Evolution, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
28
|
Bah GS, Tanya VN, Makepeace BL. Immunotherapy with mutated onchocystatin fails to enhance the efficacy of a sub-lethal oxytetracycline regimen against Onchocerca ochengi. Vet Parasitol 2015; 212:25-34. [PMID: 26100152 DOI: 10.1016/j.vetpar.2015.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/30/2015] [Accepted: 06/06/2015] [Indexed: 11/26/2022]
Abstract
Human onchocerciasis (river blindness), caused by the filarial nematode Onchocerca volvulus, has been successfully controlled by a single drug, ivermectin, for over 25 years. Ivermectin prevents the disease symptoms of severe itching and visual impairment by killing the microfilarial stage, but does not eliminate the adult parasites, necessitating repeated annual treatments. Mass drug administration with ivermectin does not always break transmission in forest zones and is contraindicated in individuals heavily co-infected with Loa loa, while reports of reduced drug efficacy in Ghana and Cameroon may signal the development of resistance. An alternative treatment for onchocerciasis involves targeting the essential Wolbachia symbiont with tetracycline or its derivatives, which are adulticidal. However, implementation of antibiotic therapy has not occurred on a wide scale due to the prolonged treatment regimen required (several weeks). In the bovine Onchocerca ochengi system, it has been shown previously that prolonged oxytetracycline therapy increases eosinophil counts in intradermal nodules, which kill the adult worms by degranulating on their surface. Here, in an "immunochemotherapeutic" approach, we sought to enhance the efficacy of a short, sub-lethal antibiotic regimen against O. ochengi by prior immunotherapy targeting onchocystatin, an immunomodulatory protein located in the adult female worm cuticle. A key asparagine residue in onchocystatin was mutated to ablate immunomodulatory activity, which has been demonstrated previously to markedly improve the protective efficacy of this vaccine candidate when used as an immunoprophylactic. The immunochemotherapeutic regimen was compared with sub-lethal oxytetracycline therapy alone; onchocystatin immunotherapy alone; a gold-standard prolonged, intermittent oxytetracycline regimen; and no treatment (negative control) in naturally infected Cameroonian cattle. Readouts were collected over one year and comprised adult worm viability, dermal microfilarial density, anti-onchocystatin IgG in sera, and eosinophil counts in nodules. Only the gold-standard antibiotic regimen achieved significant killing of adult worms, a profound reduction in microfilarial load, and a sustained increase in local tissue eosinophilia. A small but statistically significant elevation in anti-onchocystatin IgG was observed for several weeks after immunisation in the immunotherapy-only group, but the antibody response in the immunochemotherapy group was more variable. At 12 weeks post-treatment, only a transient and non-significant increase in eosinophil counts was apparent in the immunochemotherapy group. We conclude that the addition of onchocystatin immunotherapy to a sub-lethal antibiotic regimen is insufficient to induce adulticidal activity, although with booster immunisations or the targeting of additional filarial immunomodulatory proteins, the efficacy of this strategy could be strengthened.
Collapse
Affiliation(s)
- Germanus S Bah
- Institute of Infection & Global Health, University of Liverpool, 146 Brownlow Hill, Liverpool Science Park IC2, Liverpool L3 5RF, UK; Institut de Recherche Agricole pour le Développement, Regional Centre of Wakwa, BP 65 Ngaoundéré, Adamawa Region, Cameroon
| | - Vincent N Tanya
- Institut de Recherche Agricole pour le Développement, Regional Centre of Wakwa, BP 65 Ngaoundéré, Adamawa Region, Cameroon; Cameroon Academy of Sciences, BP 1457 Yaoundé, Centre Region, Cameroon
| | - Benjamin L Makepeace
- Institute of Infection & Global Health, University of Liverpool, 146 Brownlow Hill, Liverpool Science Park IC2, Liverpool L3 5RF, UK.
| |
Collapse
|
29
|
Carvalho-Queiroz C, Nyakundi R, Ogongo P, Rikoi H, Egilmez NK, Farah IO, Kariuki TM, LoVerde PT. Protective Potential of Antioxidant Enzymes as Vaccines for Schistosomiasis in a Non-Human Primate Model. Front Immunol 2015; 6:273. [PMID: 26082781 PMCID: PMC4451692 DOI: 10.3389/fimmu.2015.00273] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 05/16/2015] [Indexed: 12/15/2022] Open
Abstract
Schistosomiasis remains a major cause of morbidity in the world. The challenge today is not so much in the clinical management of individual patients, but rather in population-based control of transmission in endemic areas. Despite recent large-scale efforts, such as integrated control programs aimed at limiting schistosomiasis by improving education and sanitation, molluscicide treatment programs and chemotherapy with praziquantel, there has only been limited success. There is an urgent need for complementary approaches, such as vaccines. We demonstrated previously that anti-oxidant enzymes, such as Cu-Zn superoxide dismutase (SOD) and glutathione S peroxidase (GPX), when administered as DNA-based vaccines induced significant levels of protection in inbred mice, greater than the target 40% reduction in worm burden compared to controls set as a minimum by the WHO. These results led us to investigate if immunization of non-human primates with antioxidants would stimulate an immune response that could confer protection as a prelude study for human trials. Issues of vaccine toxicity and safety that were difficult to address in mice were also investigated. All baboons in the study were examined clinically throughout the study and no adverse reactions occurred to the immunization. When our outbred baboons were vaccinated with two different formulations of SOD (SmCT-SOD and SmEC-SOD) or one of GPX (SmGPX), they showed a reduction in worm number to varying degrees, when compared with the control group. More pronounced, vaccinated animals showed decreased bloody diarrhea, days of diarrhea, and egg excretion (transmission), as well as reduction of eggs in the liver tissue and in the large intestine (pathology) compared to controls. Specific IgG antibodies were present in sera after immunizations and 10 weeks after challenge infection compared to controls. Peripheral blood mononuclear cells, mesenteric, and inguinal node cells from vaccinated animals proliferated and produced high levels of cytokines and chemokines in response to crude and recombinant antigens compared with controls. All together, these data demonstrate the potential of antioxidants as a vaccine in a non-human primate model.
Collapse
Affiliation(s)
- Claudia Carvalho-Queiroz
- Department of Biochemistry, University of Texas Health Science Center , San Antonio, TX , USA ; Department of Pathology, University of Texas Health Science Center , San Antonio, TX , USA
| | - Ruth Nyakundi
- Institute of Primate Research, National Museums of Kenya , Nairobi , Kenya
| | - Paul Ogongo
- Institute of Primate Research, National Museums of Kenya , Nairobi , Kenya
| | - Hitler Rikoi
- Institute of Primate Research, National Museums of Kenya , Nairobi , Kenya
| | - Nejat K Egilmez
- Department of Microbiology and Immunology, School of Medicine and Biomedical Sciences, University at Buffalo , Buffalo, NY , USA
| | - Idle O Farah
- Institute of Primate Research, National Museums of Kenya , Nairobi , Kenya
| | - Thomas M Kariuki
- Institute of Primate Research, National Museums of Kenya , Nairobi , Kenya
| | - Philip T LoVerde
- Department of Biochemistry, University of Texas Health Science Center , San Antonio, TX , USA ; Department of Pathology, University of Texas Health Science Center , San Antonio, TX , USA
| |
Collapse
|
30
|
Sanin DE, Prendergast CT, Bourke CD, Mountford AP. Helminth Infection and Commensal Microbiota Drive Early IL-10 Production in the Skin by CD4+ T Cells That Are Functionally Suppressive. PLoS Pathog 2015; 11:e1004841. [PMID: 25974019 PMCID: PMC4431738 DOI: 10.1371/journal.ppat.1004841] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 03/30/2015] [Indexed: 12/12/2022] Open
Abstract
The skin provides an important first line of defence and immunological barrier to invasive pathogens, but immune responses must also be regulated to maintain barrier function and ensure tolerance of skin surface commensal organisms. In schistosomiasis-endemic regions, populations can experience repeated percutaneous exposure to schistosome larvae, however little is known about how repeated exposure to pathogens affects immune regulation in the skin. Here, using a murine model of repeated infection with Schistosoma mansoni larvae, we show that the skin infection site becomes rich in regulatory IL-10, whilst in its absence, inflammation, neutrophil recruitment, and local lymphocyte proliferation is increased. Whilst CD4+ T cells are the primary cellular source of regulatory IL-10, they expressed none of the markers conventionally associated with T regulatory (Treg) cells (i.e. FoxP3, Helios, Nrp1, CD223, or CD49b). Nevertheless, these IL-10+ CD4+ T cells in the skin from repeatedly infected mice are functionally suppressive as they reduced proliferation of responsive CD4+ T cells from the skin draining lymph node. Moreover, the skin of infected Rag-/- mice had impaired IL-10 production and increased neutrophil recruitment. Finally, we show that the mechanism behind IL-10 production by CD4+ T cells in the skin is due to a combination of an initial (day 1) response specific to skin commensal bacteria, and then over the following days schistosome-specific CD4+ T cell responses, which together contribute towards limiting inflammation and tissue damage following schistosome infection. We propose CD4+ T cells in the skin that do not express markers of conventional T regulatory cell populations have a significant role in immune regulation after repeated pathogen exposure and speculate that these cells may also help to maintain skin barrier function in the context of repeated percutaneous insult by other skin pathogens. The skin is a major barrier protecting the host from pathogen infection, but is also a site for immune regulation. Using a murine model of repeated percutaneous exposure to infectious Schistosoma mansoni cercariae, we show that, in the skin, CD4+ T cells that do not express markers of conventional regulatory T cells are the main early source of immunoregulatory IL-10 and are functionally suppressive of adaptive immune responses. We demonstrate that the production of regulatory IL-10 in the skin is greatly enhanced after repeated schistosome infection compared to levels present after a single infection and that it limits both neutrophil recruitment and local CD4+ T cell proliferation, thereby preventing excessive inflammation and tissue damage. Initially (day 1), IL-10 producing CD4+ T cells are reactive towards skin commensal bacteria, although over succeeding days they progressively become specific for schistosome antigens. Consequently, our findings highlight a role for early IL-10 produced by dermal CD4+ T cells to mediate immune regulation in advance of later stage chronic infection conventionally associated with the presence of IL-10. Our work provides a mechanistic insight into the triggers of early IL-10 production at barrier sites like the skin, and suggests how tolerance and pathogen clearance might be co-regulated early after exposure to infectious agents.
Collapse
Affiliation(s)
- David E. Sanin
- Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom
| | - Catriona T. Prendergast
- Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom
| | - Claire D. Bourke
- Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom
| | - Adrian P. Mountford
- Centre for Immunology and Infection, Department of Biology, University of York, York, United Kingdom
- * E-mail:
| |
Collapse
|
31
|
Ballesteros NA, Rodriguez Saint-Jean S, Perez-Prieto SI. Immune responses to oral pcDNA-VP2 vaccine in relation to infectious pancreatic necrosis virus carrier state in rainbow trout Oncorhynchus mykiss. Vet Immunol Immunopathol 2015; 165:127-37. [PMID: 25892368 DOI: 10.1016/j.vetimm.2015.04.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 03/10/2015] [Accepted: 04/01/2015] [Indexed: 10/23/2022]
Abstract
The VP2 gene of infectious pancreatic necrosis virus, encoded in an expression plasmid and encapsulated in alginate microspheres, was used for oral DNA vaccination of fish to better understand the carrier state and the action of the vaccine. The efficacy of the vaccine was evaluated by measuring the prevention of virus persistence in the vaccinated fish that survived after waterborne virus challenge. A real-time RT-qPCR analysis revealed lower levels of IPNV-VP4 transcripts in rainbow trout survivors among vaccinated and challenged fish compared with the control virus group at 45 days post-infection. The infective virus was recovered from asymptomatic virus control fish, but not from the vaccinated survivor fish, suggesting an active role of the vaccine in the control of IPNV infection. Moreover, the levels of IPNV and immune-related gene expression were quantified in fish showing clinical infection as well as in asymptomatic rainbow trout survivors. The vaccine mimicked the action of the virus, although stronger expression of immune-related genes, except for IFN-1 and IL12, was detected in survivors from the virus control (carrier) group than in those from the vaccinated group. The transcriptional levels of the examined genes also showed significant differences in the virus control fish at 10 and 45 days post-challenge.
Collapse
Affiliation(s)
- Natalia A Ballesteros
- Centro de Investigaciones Biologicas (CSIC), Dpto Microbiologia Molecular y Biología de las Infecciones, C/ Ramiro de Maeztu 9, 28040 Madrid, Spain.
| | - Sylvia Rodriguez Saint-Jean
- Centro de Investigaciones Biologicas (CSIC), Dpto Microbiologia Molecular y Biología de las Infecciones, C/ Ramiro de Maeztu 9, 28040 Madrid, Spain.
| | - Sara I Perez-Prieto
- Centro de Investigaciones Biologicas (CSIC), Dpto Microbiologia Molecular y Biología de las Infecciones, C/ Ramiro de Maeztu 9, 28040 Madrid, Spain.
| |
Collapse
|
32
|
Drug-induced exposure of Schistosoma mansoni antigens SmCD59a and SmKK7. PLoS Negl Trop Dis 2015; 9:e0003593. [PMID: 25774883 PMCID: PMC4361651 DOI: 10.1371/journal.pntd.0003593] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 02/05/2015] [Indexed: 11/22/2022] Open
Abstract
Background Schistosomiasis is a serious health problem especially in developing countries and affects more than 243 million people. Only few anthelmintic drugs are available up to now. A major obstacle for drug treatment is the different developmental stages and the varying host compartments during worm development. Anthelmintic drugs have been tested mainly on adult schistosomes or freshly transformed cercariae. Knowledge concerning the larval stages is lacking. Methodology/Principal Findings In this study, we used in vitro-grown schistosomula (aged between 2 to 14 days) to investigate drug effects of the three anthelmintics praziquantel, artemether, and oxamniquine. Further, we analyzed the antibody accessibility of two exemplary schistosome antigens SmCD59a and SmKK7, before and after drug treatment. Our results demonstrated that praziquantel applied at a concentration of 1 μM inhibited development of all life stages. Application of 10 μM praziquantel led to dramatic morphological changes of all schistosomula. Artemether at 1 and 10 μM had differential effects depending on whether it was applied to 2-day as compared to 7- and 14-day schistosomula. While 2-day schistosomula were not killed but inhibited from further development, severe morphological damage was seen in 7- and 14-day schistosomula. Oxamniquine (1 and 10 μM) led to severe morphological impairment in all life stages. Analyzing the accessibility of the antigens SmCD59a and SmKK7 before drug treatment showed no antibody binding on living intact schistosomula. However, when schistosomula were treated with anthelmintics, both antigens became exposed on the larvae. Oxamniquine turned out to be most effective in promoting antibody binding to all schistosomula stages. Conclusion This study has revealed marked differences in anthelmintic drug effects against larvae. Drug treatment increases surface antigen presentation and renders larvae accessible to antibody attack. Schistosomiasis is one of the major parasitic diseases in developing countries and still causes 200,000 deaths per year. Mass drug administration programs with praziquantel, the drug of choice against schistosomiasis, are currently undertaken in Sub-Saharan Africa. Praziquantel, although efficient against adult worms, fails to cure early infection. The complex developmental stages of schistosomes and migration through varying host compartments with different local drug concentration are a challenge for drug treatment. After infecting their mammalian host, schistosomula traverse through skin and the vasculature of lung, liver and intestines. During their migration, they develop from larvae to paired adults in approximately 4 to 5 weeks. So far, drug effects have been analyzed on adult worms or freshly transformed schistosomula only. Information about the effects on the larval stages is lacking. We were able to transfer the larval development of the first three weeks into the culture dish. This tool can be used for the analysis of drug effects against schistosomula and for investigation of the accessibility, expression and localization of antigens. Rendering the parasite’s larvae vulnerable to the host’s immune system by increasing antigen presentation is an important aspect of drug activity. We demonstrate on in vitro-cultured Schistosoma mansoni larvae, that SmCD59a and SmKK7, as examples for hidden antigens, become accessible to antibodies following drug treatment.
Collapse
|
33
|
Fonseca CT, Oliveira SC, Alves CC. Eliminating Schistosomes through Vaccination: What are the Best Immune Weapons? Front Immunol 2015; 6:95. [PMID: 25806033 PMCID: PMC4353369 DOI: 10.3389/fimmu.2015.00095] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 02/18/2015] [Indexed: 12/24/2022] Open
Abstract
The successful development of vaccines depends on the knowledge of the immunological mechanisms associated with the elimination of the pathogen. In the case of schistosomes, its complex life cycle and the mechanisms developed to evade host immune system, turns the development of a vaccine against the disease into a very difficult task. Identifying the immunological effector mechanisms involved in parasite attrition and the major targets for its response is a key step to formulate an effective vaccine. Recent studies have described some promising antigens to compose a subunit vaccine and have pointed to some immune factors that play a role in parasite elimination. Here, we review the immune components and effector mechanisms associated with the protective immunity induced by those vaccine candidates and the lessons we have learned from the studies of the acquired resistance to infection in humans. We will also discuss the immune factors that correlate with protection and therefore could help to evaluate those vaccine formulations in clinical trials.
Collapse
Affiliation(s)
- Cristina Toscano Fonseca
- Laboratório de Esquistossomose do Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz , Belo Horizonte , Brazil ; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação, Universidade Federal de Minas Gerais , Belo Horizonte , Brazil
| | - Sergio Costa Oliveira
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação, Universidade Federal de Minas Gerais , Belo Horizonte , Brazil ; Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais , Belo Horizonte , Brazil
| | - Clarice Carvalho Alves
- Laboratório de Esquistossomose do Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz , Belo Horizonte , Brazil
| |
Collapse
|
34
|
Nausch N, Appleby LJ, Sparks AM, Midzi N, Mduluza T, Mutapi F. Group 2 innate lymphoid cell proportions are diminished in young helminth infected children and restored by curative anti-helminthic treatment. PLoS Negl Trop Dis 2015; 9:e0003627. [PMID: 25799270 PMCID: PMC4370749 DOI: 10.1371/journal.pntd.0003627] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 02/19/2015] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Group 2 Innate lymphoid cells (ILC2s) are innate cells that produce the TH2 cytokines IL-5 and IL-13. The importance of these cells has recently been demonstrated in experimental models of parasitic diseases but there is a paucity of data on ILC2s in the context of human parasitic infections and in particular of the blood dwelling parasite Schistosoma haematobium. METHODOLOGY/PRINCIPAL FINDINGS In this case-control study human peripheral blood ILC2s were analysed in relation to infection with the helminth parasite Schistosoma haematobium. Peripheral blood mononuclear cells of 36 S. haematobium infected and 36 age and sex matched uninfected children were analysed for frequencies of ILC2s identified as Lin-CD45+CD127+CD294+CD161+. ILC2s were significantly lower particularly in infected children aged 6-9 years compared to healthy participants. Curative anti-helminthic treatment resulted in an increase in levels of the activating factor TSLP and restoration of ILC2 levels. CONCLUSION This study demonstrates that ILC2s are diminished in young helminth infected children and restored by removal of the parasites by treatment, indicating a previously undescribed association between a human parasitic infection and ILC2s and suggesting a role of ILC2s before the establishment of protective acquired immunity in human schistosomiasis.
Collapse
Affiliation(s)
- Norman Nausch
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
| | - Laura J. Appleby
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
| | - Alexandra M. Sparks
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
| | - Nicholas Midzi
- National Institute of Health Research, Causeway, Harare, Zimbabwe
| | - Takafira Mduluza
- University of Zimbabwe, Department of Biochemistry, University of Zimbabwe, Mount Pleasant, Harare, Zimbabwe
| | - Francisca Mutapi
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
| |
Collapse
|
35
|
Mbanefo EC, Kumagai T, Kodama Y, Kurosaki T, Furushima-Shimogawara R, Cherif MS, Mizukami S, Kikuchi M, Huy NT, Ohta N, Sasaki H, Hirayama K. Immunogenicity and anti-fecundity effect of nanoparticle coated glutathione S-transferase (SjGST) DNA vaccine against murine Schistosoma japonicum infection. Parasitol Int 2015; 64:24-31. [PMID: 25603531 DOI: 10.1016/j.parint.2015.01.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 12/10/2014] [Accepted: 01/11/2015] [Indexed: 01/10/2023]
Abstract
There is still urgent need for a vaccine against schistosomiasis, especially in Schistosoma japonicum endemic areas where even a vaccine that will interrupt zoonotic transmission will be potentially effective as an intervention tool. We had developed a novel nanoparticle gene delivery system, which has proven efficacious in gene transfection to target immune cells with complementary adjuvant effect and high protective efficacy in several diseases. Here, we applied this nanoparticle system in combination with S. japonicum glutathione S-transferase (SjGST) DNA vaccine to show the immunogenicity and anti-fecundity effect of the nanoparticle coated vaccine formulation against murine schistosomiasis. The nanoparticle-coated DNA vaccine formulation induced desired immune responses. In comparison with the nanoparticle coated empty vector, it produced significantly increased antigen-specific humoral response, T-helper 1 polarized cytokine environment, higher proportion of IFN-γ producing CD4(+) T-cells and the concomitant decrease in IL-4 producing CD4(+) T-cells. Although there was no effect on worm burden, we recorded a marked reduction in tissue egg burden. There was up to 71.3% decrease in tissue egg burden and 55% reduction in the fecundity of female adult worms. Our data showed that SjGST DNA vaccine, delivered using the nanoparticle gene delivery system, produced anti-fecundity effect on female adult schistosomes as previously described by using conventional subunit vaccine with adjuvant, proving this DNA vaccine formulation as a promising candidate for anti-pathology and transmission blocking application.
Collapse
Affiliation(s)
- Evaristus Chibunna Mbanefo
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN) and Global COE Program, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Department of Parasitology and Entomology, Faculty of Bioscience, Nnamdi Azikiwe University, P.M.B. 5025, Awka, Nigeria
| | - Takashi Kumagai
- Section of Environmental Parasitology, Tokyo Medical and Dental University Graduate School of Medical and Dental Science, 113-8519, Japan
| | - Yukinobu Kodama
- Department of Hospital Pharmacy, Nagasaki University Hospital, 1-7-1 Sakamoto, 852-8501, Japan
| | - Tomoaki Kurosaki
- Department of Hospital Pharmacy, Nagasaki University Hospital, 1-7-1 Sakamoto, 852-8501, Japan
| | - Rieko Furushima-Shimogawara
- Section of Environmental Parasitology, Tokyo Medical and Dental University Graduate School of Medical and Dental Science, 113-8519, Japan
| | - Mahamoud Sama Cherif
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN) and Global COE Program, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan
| | - Shusaku Mizukami
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN) and Global COE Program, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan
| | - Mihoko Kikuchi
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN) and Global COE Program, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan
| | - Nguyen Tien Huy
- Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Department of Clinical Product Development, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan
| | - Nobuo Ohta
- Section of Environmental Parasitology, Tokyo Medical and Dental University Graduate School of Medical and Dental Science, 113-8519, Japan
| | - Hitoshi Sasaki
- Department of Hospital Pharmacy, Nagasaki University Hospital, 1-7-1 Sakamoto, 852-8501, Japan
| | - Kenji Hirayama
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN) and Global COE Program, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan.
| |
Collapse
|
36
|
Colley DG, Secor WE. Immunology of human schistosomiasis. Parasite Immunol 2014; 36:347-57. [PMID: 25142505 PMCID: PMC4278558 DOI: 10.1111/pim.12087] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 10/30/2013] [Indexed: 12/25/2022]
Abstract
There is a wealth of immunologic studies that have been carried out in experimental and human schistosomiasis that can be classified into three main areas: immunopathogenesis, resistance to reinfection and diagnostics. It is clear that the bulk of, if not all, morbidity due to human schistosomiasis results from immune-response-based inflammation against eggs lodged in the body, either as regulated chronic inflammation or resulting in fibrotic lesions. However, the exact nature of these responses, the antigens to which they are mounted and the mechanisms of the critical regulatory responses are still being sorted out. It is also becoming apparent that protective immunity against schistosomula as they develop into adult worms develops slowly and is hastened by the dying of adult worms, either naturally or when they are killed by praziquantel. However, as with anti-egg responses, the responsible immune mechanisms and inducing antigens are not clearly established, nor are any potential regulatory responses known. Finally, a wide variety of immune markers, both cellular and humoral, can be used to demonstrate exposure to schistosomes, and immunologic measurement of schistosome antigens can be used to detect, and thus diagnose, active infections. All three areas contribute to the public health response to human schistosome infections.
Collapse
Affiliation(s)
- D G Colley
- Department of Microbiology, Center for Tropical and Emerging Global Disease, The University of Georgia, Athens, GA, USA
| | | |
Collapse
|
37
|
Abstract
Helminth parasites infect over one fourth of the human population and are highly prevalent in livestock worldwide. In model systems, parasites are strongly immunomodulatory, but the immune system can be driven to expel them by prior vaccination. However, no vaccines are currently available for human use. Recent advances in vaccination with recombinant helminth antigens have been successful against cestode infections of livestock and new vaccines are being tested against nematode parasites of animals. Numerous vaccine antigens are being defined for a wide range of helminth parasite species, but greater understanding is needed to define the mechanisms of vaccine-induced immunity, to lay a rational platform for new vaccines and their optimal design. With human trials underway for hookworm and schistosomiasis vaccines, a greater integration between veterinary and human studies will highlight the common molecular and mechanistic pathways, and accelerate progress towards reducing the global health burden of helminth infection.
Collapse
|
38
|
Carey AJ, Tan CK, Ulett GC. Infection-induced IL-10 and JAK-STAT: A review of the molecular circuitry controlling immune hyperactivity in response to pathogenic microbes. JAKSTAT 2014; 1:159-67. [PMID: 24058765 PMCID: PMC3670239 DOI: 10.4161/jkst.19918] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2012] [Revised: 03/04/2012] [Accepted: 03/06/2012] [Indexed: 12/19/2022] Open
Abstract
Generation of effective immune responses against pathogenic microbes depends on a fine balance between pro- and anti-inflammatory responses. Interleukin-10 (IL-10) is essential in regulating this balance and has garnered renewed interest recently as a modulator of the response to infection at the JAK-STAT signaling axis of host responses. Here, we examine how IL-10 functions as the “master regulator” of immune responses through JAK-STAT, and provide a perspective from recent insights on bacterial, protozoan, and viral infection model systems. Pattern recognition and subsequent molecular events that drive activation of IL-10-associated JAK-STAT circuitry are reviewed and the implications for microbial pathogenesis are discussed.
Collapse
Affiliation(s)
- Alison J Carey
- School of Medical Sciences; Centre for Medicine and Oral Health; Griffith University; Gold Coast, QLD Australia
| | | | | |
Collapse
|
39
|
Redpath SA, Fonseca NM, Perona-Wright G. Protection and pathology during parasite infection: IL-10 strikes the balance. Parasite Immunol 2014; 36:233-52. [PMID: 24666543 DOI: 10.1111/pim.12113] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 03/18/2014] [Indexed: 12/16/2022]
Abstract
The host response to infection requires an immune response to be strong enough to control the pathogen but also restrained, to minimize immune-mediated pathology. The conflicting pressures of immune activation and immune suppression are particularly apparent in parasite infections, where co-evolution of host and pathogen has selected many different compromises between protection and pathology. Cytokine signals are critical determinants of both protective immunity and immunopathology, and, in this review, we focus on the regulatory cytokine IL-10 and its role in protozoan and helminth infections. We discuss the sources and targets of IL-10 during parasite infection, the signals that initiate and reinforce its action, and its impact on the invading parasite, on the host tissue, and on coincident immune responses.
Collapse
Affiliation(s)
- S A Redpath
- Department of Microbiology & Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | | | | |
Collapse
|
40
|
Cytokine responses to the anti-schistosome vaccine candidate antigen glutathione-S-transferase vary with host age and are boosted by praziquantel treatment. PLoS Negl Trop Dis 2014; 8:e2846. [PMID: 24810615 PMCID: PMC4014416 DOI: 10.1371/journal.pntd.0002846] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 03/25/2014] [Indexed: 01/17/2023] Open
Abstract
Background Improved helminth control is required to alleviate the global burden of schistosomiasis and schistosome-associated pathologies. Current control efforts rely on the anti-helminthic drug praziquantel (PZQ), which enhances immune responses to crude schistosome antigens but does not prevent re-infection. An anti-schistosome vaccine based on Schistosoma haematobium glutathione-S-transferase (GST) is currently in Phase III clinical trials, but little is known about the immune responses directed against this antigen in humans naturally exposed to schistosomes or how these responses change following PZQ treatment. Methodology Blood samples from inhabitants of a Schistosoma haematobium-endemic area were incubated for 48 hours with or without GST before (n = 195) and six weeks after PZQ treatment (n = 107). Concentrations of cytokines associated with innate inflammatory (TNFα, IL-6, IL-8), type 1 (Th1; IFNγ, IL-2, IL-12p70), type 2 (IL-4, IL-5, IL-13), type 17 (IL-17A, IL-21, IL-23p19) and regulatory (IL-10) responses were quantified in culture supernatants via enzyme-linked immunosorbent assay (ELISA). Factor analysis and multidimensional scaling were used to analyse multiple cytokines simultaneously. Principal Findings A combination of GST-specific type 2 (IL-5 and IL-13) and regulatory (IL-10) cytokines was significantly lower in 10–12 year olds, the age group at which S. haematobium infection intensity and prevalence peak, than in 4–9 or 13+ year olds. Following PZQ treatment there was an increase in the number of participants producing detectable levels of GST-specific cytokines (TNFα, IL-6, IL-8, IFNγ, IL-12p70, IL-13 and IL-23p19) and also a shift in the GST-specific cytokine response towards a more pro-inflammatory phenotype than that observed before treatment. Participant age and pre-treatment infection status significantly influenced post-treatment cytokine profiles. Conclusions/Significance In areas where schistosomiasis is endemic host age, schistosome infection status and PZQ treatment affect the cellular cytokine response to GST. Thus the efficacy of a GST-based vaccine may also be shaped by the demographic and epidemiological characteristics of targeted populations. Schistosomiasis is caused by infection with Schistosoma spp. parasites, for which the main treatment is the drug praziquantel (PZQ). Since PZQ does not prevent reinfection, an anti-schistosome vaccine based on the Schistosoma haematobium enzyme glutathione-S-transferase (GST) is being developed. In this study we investigated the GST-specific immune responses of people naturally exposed to schistosomes and the affect that PZQ has on these responses. We cultured blood samples from a schistosome-exposed community with GST before and six weeks after PZQ treatment and measured a range of soluble proteins (cytokines) in culture supernatants as indicators of blood cell activation and phenotype. Before treatment, GST-specific cytokine responses varied with host age, particularly in children with high intensity schistosome infections. Following treatment, GST activated blood samples from more individuals to produce a broader range of cytokines and the combination of GST-specific cytokine responses reflected a more pro-inflammatory immune phenotype than that observed pre-treatment. Post-treatment responses varied according to host age and pre-treatment infection status. Taken together, our study suggests that current and future GST-based vaccine trials should take host age, schistosome infection status and PZQ treatment history into account since these factors influence GST-specific immune activation.
Collapse
|
41
|
Scheer S, Gross S, Mouahid G, Moné H, Lamers MC. A novel tool to identify the relative contribution of lymphoid cell types that contribute to IL-10 production during the infection with Schistosoma mansoni: the TIGER index. J Immunol Methods 2014; 406:66-73. [PMID: 24657590 DOI: 10.1016/j.jim.2014.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 02/18/2014] [Accepted: 03/11/2014] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Infection with the trematode helminth Schistosoma mansoni affects more than 200 million people worldwide. Infected patients are thought to show a decreased incidence of asthma and autoimmune diseases, which is, among others, considered a result of an increased production of the immunoregulatory cytokine IL-10. However, the location and the type of cell that is responsible for the highest production of IL-10 in vivo are still unknown. AIM Identification of the hierarchy of IL-10 producing cell types in the mesenteric lymph node and spleen during the course of the murine infection with S. mansoni without the need of an external standard. METHODS We describe the use of the IL-10 reporter mouse TIGER for the study of murine schistosomiasis and introduce a novel tool, which we have called the TIGER index (TI). This index combines data from flow cytometric measurements and cell count analysis and allows identifying the cell type with the highest contribution of IL-10 during the course of infection in the secondary lymphoid organs, sites of extensive immunoregulatory activity in schistosomiasis. RESULTS In this paper we have calculated the TI for the mesenteric lymph nodes and the spleen in the course of a chronic infection with S. mansoni. Using the TI, we identified CD4(pos) CD25pos and CD4(pos) CD25(neg) cell populations as the highest producers of IL-10 in the mesenteric lymph node and the spleen in chronic schistosomiasis, respectively, whereas B cells, NK cells and NKT cells showed a lower contribution to IL-10 production throughout the infection. CONCLUSION The TI is a highly useful tool to measure the relative contribution of different cell types, which are responsible for the in vivo production of IL-10 in the secondary lymphoid organs during the infection with S. mansoni. Thus, the strength of the TI ensures the possibility to analyze IL-10 production in a long term experiment without the need of an external standard between each time point of analysis.
Collapse
Affiliation(s)
- Sebastian Scheer
- Max Planck Institute of Immunobiology and Epigenetics, Stuebeweg 51, 79108 Freiburg, Germany; University of Freiburg, Germany; International Max Planck Research School of Cellular and Molecular Biology, Freiburg, Germany.
| | - Sandra Gross
- Max Planck Institute of Immunobiology and Epigenetics, Stuebeweg 51, 79108 Freiburg, Germany
| | - Gabriel Mouahid
- Ecologie et Evolution des Interactions, Université de Perpignan Via Domitia, UMR 5244, 66860 Perpignan, France
| | - Hélène Moné
- Ecologie et Evolution des Interactions, Université de Perpignan Via Domitia, UMR 5244, 66860 Perpignan, France
| | - Marinus C Lamers
- Max Planck Institute of Immunobiology and Epigenetics, Stuebeweg 51, 79108 Freiburg, Germany
| |
Collapse
|
42
|
Negrão-Corrêa D, Fittipaldi JF, Lambertucci JR, Teixeira MM, Antunes CMDF, Carneiro M. Association of Schistosoma mansoni-specific IgG and IgE antibody production and clinical schistosomiasis status in a rural area of Minas Gerais, Brazil. PLoS One 2014; 9:e88042. [PMID: 24505371 PMCID: PMC3913716 DOI: 10.1371/journal.pone.0088042] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 01/02/2014] [Indexed: 02/07/2023] Open
Abstract
Background Studies in murine models and human populations have indicated that the collagen-rich granulomatous response against parasite eggs trapped in the liver is associated with the development of severe hepatosplenic schistosomiasis, characterized by periportal fibrosis and portal hypertension. The role of the humoral response in parasite susceptibility has been well established, but its participation in disease severity remains poorly understood. In this work, we evaluated the relationship between parasite-reactive IgE and IgG levels and schistosomiasis morbidity in infected patients with similar parasite burdens. Methodology/Principal Findings Ninety-seven Schistosoma mansoni-infected individuals were subjected to clinical examination and abdominal ultrasound analysis. IgG reactivity and IgE concentration against Schistosoma mansoni soluble egg antigens (SEA) and adult worm antigen preparation (SWAP) were evaluated by ELISA assay. Multivariable linear regression models were used to evaluate the relationship between parasite-reactive antibodies and the co-variables investigated. The study population showed low parasite burden (median 30 eggs/g feces), constant re-infection, and signs of fibrosis was detected in more than 30% of individuals. Most infected individuals showed IgG reactivity, and the median concentrations of IgE anti-SEA and anti-SWAP antibodies were 1,870 and 1,375 ng/mL, respectively. There was no association between parasite burden and antibody response or any parameter of disease severity. However, IgG anti-SWAP level was positively associated with morbidity parameters, such as spleen size and thickness of portal vein at the entrance and secondary branch. In contrast, the data also revealed independent inverse correlations between concentration of parasite-reactive IgE and gallbladder wall thickness, a marker of fibrosis in schistosomiasis. Conclusions/Significance The data indicate that IgG anti-SWAP is positively associated with severe schistosomiasis, independently of parasite burden, while high production of parasite-specific IgE is associated with mild disease in the human population. Antibody profiles are good correlates for schistosomiasis severity and could be tested as biomarkers of disease severity.
Collapse
Affiliation(s)
- Deborah Negrão-Corrêa
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Programa de Pós-Graduação em Parasitologia, Belo Horizonte, MG, Brazil
- * E-mail:
| | - Juliana F. Fittipaldi
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Programa de Pós-Graduação em Parasitologia, Belo Horizonte, MG, Brazil
| | - José Roberto Lambertucci
- Faculdade de Medicina Universidade Federal de Minas Gerais, Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Belo Horizonte, MG, Brazil
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Mariângela Carneiro
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Programa de Pós-Graduação em Parasitologia, Belo Horizonte, MG, Brazil
- Faculdade de Medicina Universidade Federal de Minas Gerais, Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Belo Horizonte, MG, Brazil
| |
Collapse
|
43
|
Layland LE, Straubinger K, Ritter M, Loffredo-Verde E, Garn H, Sparwasser T, Prazeres da Costa C. Schistosoma mansoni-mediated suppression of allergic airway inflammation requires patency and Foxp3+ Treg cells. PLoS Negl Trop Dis 2013; 7:e2379. [PMID: 23967364 PMCID: PMC3744427 DOI: 10.1371/journal.pntd.0002379] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 07/05/2013] [Indexed: 12/13/2022] Open
Abstract
The continual rise of asthma in industrialised countries stands in strong contrast to the situation in developing lands. According to the modified Hygiene Hypothesis, helminths play a major role in suppressing bystander immune responses to allergens, and both epidemiological and experimental studies suggest that the tropical parasitic trematode Schistosoma mansoni elicits such effects. The focus of this study was to investigate which developmental stages of schistosome infection confer suppression of allergic airway inflammation (AAI) using ovalbumin (OVA) as a model allergen. Moreover, we assessed the functional role and localization of infection-induced CD4+Foxp3+ regulatory T cells (Treg) in mediating such suppressive effects. Therefore, AAI was elicited using OVA/adjuvant sensitizations with subsequent OVA aerosolic challenge and was induced during various stages of infection, as well as after successful anti-helminthic treatment with praziquantel. The role of Treg was determined by specifically depleting Treg in a genetically modified mouse model (DEREG) during schistosome infection. Alterations in AAI were determined by cell infiltration levels into the bronchial system, OVA-specific IgE and Th2 type responses, airway hyper-sensitivity and lung pathology. Our results demonstrate that schistosome infection leads to a suppression of OVA-induced AAI when mice are challenged during the patent phase of infection: production of eggs by fecund female worms. Moreover, this ameliorating effect does not persist after anti-helminthic treatment, and depletion of Treg reverts suppression, resulting in aggravated AAI responses. This is most likely due to a delayed reconstitution of Treg in infected-depleted animals which have strong ongoing immune responses. In summary, we conclude that schistosome-mediated suppression of AAI requires the presence of viable eggs and infection-driven Treg cells. These data provide evidence that helminth derived products could be incorporated into treatment strategies that specifically target suppression of immune responses in AAI by inducing Treg cells. Infections with schistosomes, such as S. mansoni, S. japonicum and S. haematobium, are considered a major public health concern. Morbidity arises through granulomatous responses to eggs that become trapped in infected tissues. Interestingly, schistosomes belong to the group of helminths that have been shown to reduce allergy or autoimmunity. Indeed, the evidence provided by epidemiological surveys and experimental animal models has been so overwhelming that such helminths are now included in the Hygiene Hypothesis. However, since helminths provoke immunological responses that are similar to those seen in allergy (increased eosinophilia and IgE) it is suggested that additional mechanisms dampen such allergic responses. Helminth-induced regulatory T cells (Treg) are considered a component of these modulatory networks. Using an allergic airway inflammation model, we have elucidated that schistosome-mediated protection requires patency, that is, active egg production from fecund female worms. In addition, protection was shown to be mediated by infection-induced Treg. Interestingly, in endemic countries it is usually individuals with strong patent infections that show reduced allergic prevalence. Thus, further research into the immunomodulatory capacity of schistosome-egg derived factors may elucidate novel drug candidates or enhance treatment strategies to reduce allergic responses on the cellular level.
Collapse
Affiliation(s)
- Laura E. Layland
- Institute of Medical Microbiology, Immunology and Hygiene (MIH), Technische Universität München, Munich, Germany
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Clinic Bonn, Bonn, Germany
| | - Kathrin Straubinger
- Institute of Medical Microbiology, Immunology and Hygiene (MIH), Technische Universität München, Munich, Germany
| | - Manuel Ritter
- Institute of Medical Microbiology, Immunology and Hygiene (MIH), Technische Universität München, Munich, Germany
| | - Eva Loffredo-Verde
- Institute of Medical Microbiology, Immunology and Hygiene (MIH), Technische Universität München, Munich, Germany
| | - Holger Garn
- Institute of Laboratory Medicine and Pathobiochemistry, Medical Faculty, Philipps-University Marburg, Marburg, Germany
| | - Tim Sparwasser
- Institut für Infektionsimmunologie TWINCORE - Zentrum für Experimentelle und Klinische Infektionsforschung GmbH, Hannover, Germany
| | - Clarissa Prazeres da Costa
- Institute of Medical Microbiology, Immunology and Hygiene (MIH), Technische Universität München, Munich, Germany
- * E-mail:
| |
Collapse
|
44
|
Mutapi F, Billingsley PF, Secor WE. Infection and treatment immunizations for successful parasite vaccines. Trends Parasitol 2013; 29:135-41. [PMID: 23415733 PMCID: PMC3884123 DOI: 10.1016/j.pt.2013.01.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 01/16/2013] [Accepted: 01/16/2013] [Indexed: 12/20/2022]
Abstract
Since the advent of techniques for the expression of recombinant peptide antigens, the availability of human vaccines for parasitic diseases has been ‘imminent’. Yet vaccines based on recombinant proteins are still largely aspirations, not realities. It is now apparent that vaccine development needs additional knowledge about host protective immune response(s), antigen characteristics, and the delivery required to induce those responses. The most successful immune protection against parasites has been generated by infection and treatment, the induction of protective immunity by truncating the course of an infection with drug treatment. Here, we consider the characteristics of an effective, protective anti-parasite vaccine and propose a conceptual framework to aid parasite vaccine development using malaria and schistosomiasis as examples.
Collapse
Affiliation(s)
- Francisca Mutapi
- Institute for Immunology and Infection Research, Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, EH9 3JT, UK.
| | | | | |
Collapse
|
45
|
Coomes SM, Pelly VS, Wilson MS. Plasticity within the αβ⁺CD4⁺ T-cell lineage: when, how and what for? Open Biol 2013; 3:120157. [PMID: 23345540 PMCID: PMC3603458 DOI: 10.1098/rsob.120157] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Following thymic output, αβ⁺CD4⁺ T cells become activated in the periphery when they encounter peptide-major histocompatibility complex. A combination of cytokine and co-stimulatory signals instructs the differentiation of T cells into various lineages and subsequent expansion and contraction during an appropriate and protective immune response. Our understanding of the events leading to T-cell lineage commitment has been dominated by a single fate model describing the commitment of T cells to one of several helper (T(H)), follicular helper (T(FH)) or regulatory (T(REG)) phenotypes. Although a single lineage-committed and dedicated T cell may best execute a single function, the view of a single fate for T cells has recently been challenged. A relatively new paradigm in αβ⁺CD4⁺ T-cell biology indicates that T cells are much more flexible than previously appreciated, with the ability to change between helper phenotypes, between helper and follicular helper, or, most extremely, between helper and regulatory functions. In this review, we comprehensively summarize the recent literature identifying when T(H) or T(REG) cell plasticity occurs, provide potential mechanisms of plasticity and ask if T-cell plasticity is beneficial or detrimental to immunity.
Collapse
Affiliation(s)
- Stephanie M Coomes
- Division of Molecular Immunology, National Institute for Medical Research, MRC, London NW7 1AA, UK
| | | | | |
Collapse
|
46
|
Tang H, Ming Z, Liu R, Xiong T, Grevelding CG, Dong H, Jiang M. Development of adult worms and granulomatous pathology are collectively regulated by T- and B-cells in mice infected with Schistosoma japonicum. PLoS One 2013; 8:e54432. [PMID: 23349889 PMCID: PMC3551845 DOI: 10.1371/journal.pone.0054432] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 12/11/2012] [Indexed: 12/12/2022] Open
Abstract
Schistosoma blood flukes, which cause schistosomiasis affecting 200 million people in the world, are dependent on signals from host CD4+ T cells to facilitate parasite growth and development in the mammalian host and to induce Th2-biased inflammatory granulomas. B cells, however, are reported to down-regulate granulomatous pathology in schistosomiasis, but not to affect the development of blood flukes together with CD4+ T lymphocytes. Thus it is not clear whether B cells mediate parasite development, reproduction and egg granuloma formation of schistosomes without the help of CD4+ T lymphocytes. Using mice that have severe combined immunodeficiency (scid) and mice lacking T cells (nude), we found that the absence of B cells can more seriously hamper the development and paring of adult worms, but granuloma formation of Schistosoma japonicum in scid mice was not down-regulated comparing with that in nude mice. The level of IL-10 in the sera of nude mice was significantly higher than of scid mice at 43 days post infection (p.i.). Thus multiple mechanisms of immune modulation seem to be involved in parasite development and reproduction by helminth-induced regulatory B cells. Our findings have significance for understanding the molecular connections between schistosomes and T- and B-cells, indicating that more research is needed to develop efficient vaccine-based therapies for schistosomiasis.
Collapse
Affiliation(s)
- Hongbin Tang
- Laboratory Animal Center, medicine school, Wuhan University, Wuhan, China
| | - Zhenping Ming
- Department of Medical Parasitology, School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Rong Liu
- Department of Medical Parasitology, School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Tao Xiong
- Department of Medical Parasitology, School of Basic Medical Science, Wuhan University, Wuhan, China
| | | | - Huifeng Dong
- Department of Medical Parasitology, School of Basic Medical Science, Wuhan University, Wuhan, China
- * E-mail: (HD) (HD); (MJ) (MJ)
| | - Mingsen Jiang
- Department of Medical Parasitology, School of Basic Medical Science, Wuhan University, Wuhan, China
- * E-mail: (HD) (HD); (MJ) (MJ)
| |
Collapse
|
47
|
Fairfax K, Nascimento M, Huang SCC, Everts B, Pearce EJ. Th2 responses in schistosomiasis. Semin Immunopathol 2012; 34:863-71. [PMID: 23139101 DOI: 10.1007/s00281-012-0354-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 10/08/2012] [Indexed: 01/04/2023]
Abstract
Schistosomiasis is caused by infection with parasitic flatworms of the genus Schistosoma. It is characterized by the development of strong CD4(+) T cell and B cell responses that, during primary infection, fail to eliminate the parasites, but in collaboration with cells of the innate immune system allow survival in the face of ongoing tissue damage caused by the lodging of parasite eggs in the liver and the passage of eggs across the intestinal epithelium. Mounting a tightly controlled Th2 response is key to this outcome, and while this type of response is a risk factor for the development of fibrosis, it also underpins the development of resistance to further infection; as such, understanding how Th2 responses are induced and regulated in schistosomiasis remains a critical area of research.
Collapse
Affiliation(s)
- Keke Fairfax
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | | |
Collapse
|
48
|
Yin J, Dai A, Arango T, Kasinathan RS, Greenberg RM, Boyer JD. IL-4 and IFN-γ induced by human immunodeficiency virus vaccine in a schistosome infection model. Hum Vaccin Immunother 2012; 8:1555-63. [PMID: 23151453 DOI: 10.4161/hv.22142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The co-infection of HIV and helminth parasites, such as Schistosoma spp, has increased in sub-Saharan Africa. Many HIV vaccine candidate studies have been completed or are in ongoing clinical trials, but it is not clear how HIV vaccines might affect the course of schistosome infections. In this study, we immunized S. mansoni-infected mice with an efficient DNA vaccine that included HIV gag. Using this model, we found that Th2 cytokines, such as IL-4 and IL-13, were highly induced after schistosome infection. Treatment of infected mice with the HIV DNA vaccine resulted in a significant attenuation of this rise in IL-13 expression and an increase in expression of the Th1 cytokine, TNF-α. However, vaccine administration did not significantly influence the expression of IL-4, or IFN-γ, and did not affect T cell proliferative capacity. Interestingly, the IL-4 (+) IFN-γ (+) phenotype appears in schistosome-infected mice that received HIV vaccination, and is associated with the expression of transcription factors GATA3 (+) T-bet (+) in these mice. These studies indicate that DNA vaccination can have an impact on ongoing chronic infection.
Collapse
Affiliation(s)
- Jiangmei Yin
- Department of Pathology and Laboratory Medicine; University of Pennsylvania School of Medicine; Philadelphia, PA USA
| | | | | | | | | | | |
Collapse
|
49
|
Colton CA. Immune heterogeneity in neuroinflammation: dendritic cells in the brain. J Neuroimmune Pharmacol 2012; 8:145-62. [PMID: 23114889 PMCID: PMC4279719 DOI: 10.1007/s11481-012-9414-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 10/22/2012] [Indexed: 12/20/2022]
Abstract
Dendritic cells (DC) are critical to an integrated immune response and serve as the key link between the innate and adaptive arms of the immune system. Under steady state conditions, brain DC’s act as sentinels, continually sampling their local environment. They share this function with macrophages derived from the same basic hemopoietic (bone marrow-derived) precursor and with parenchymal microglia that arise from a unique non-hemopoietic origin. While multiple cells may serve as antigen presenting cells (APCs), dendritic cells present both foreign and self-proteins to naïve T cells that, in turn, carry out effector functions that serve to protect or destroy. The resulting activation of the adaptive response is a critical step to resolution of injury or infection and is key to survival. In this review we will explore the critical roles that DCs play in the brain’s response to neuroinflammatory disease with emphasis on how the brain’s microenvironment impacts these actions.
Collapse
Affiliation(s)
- Carol A Colton
- Neurology, Duke University Medical Center, Box 2900, Durham, NC 27710, USA.
| |
Collapse
|
50
|
Bourke CD, Mutapi F, Nausch N, Photiou DMF, Poulsen LK, Kristensen B, Arnved J, Rønborg S, Roepstorff A, Thamsborg S, Kapel C, Melbye M, Bager P. Trichuris suisova therapy for allergic rhinitis does not affect allergen-specific cytokine responses despite a parasite-specific cytokine response. Clin Exp Allergy 2012; 42:1582-95. [DOI: 10.1111/j.1365-2222.2012.04063.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- C. D. Bourke
- Institute of Immunology and Infection Research; School of Biological Sciences; University of Edinburgh; Edinburgh; UK
| | - F. Mutapi
- Institute of Immunology and Infection Research; School of Biological Sciences; University of Edinburgh; Edinburgh; UK
| | - N. Nausch
- Institute of Immunology and Infection Research; School of Biological Sciences; University of Edinburgh; Edinburgh; UK
| | - D. M. F. Photiou
- Institute of Immunology and Infection Research; School of Biological Sciences; University of Edinburgh; Edinburgh; UK
| | - L. K. Poulsen
- Allergy Clinic; National University Hospital; Copenhagen; Denmark
| | | | - J. Arnved
- Pulmonology and Allergy Clinic of Copenhagen; Copenhagen; Denmark
| | - S. Rønborg
- Pulmonology and Allergy Clinic of Copenhagen; Copenhagen; Denmark
| | - A. Roepstorff
- Department of Veterinary Disease Biology; Faculty of Life Sciences; University of Copenhagen; Copenhagen; Denmark
| | - S. Thamsborg
- Department of Veterinary Disease Biology; Faculty of Life Sciences; University of Copenhagen; Copenhagen; Denmark
| | - C. Kapel
- Department of Agriculture and Ecology; Faculty of Life Sciences; University of Copenhagen; Frederiksberg; Denmark
| | - M. Melbye
- Statens Serum Institut, Department of Epidemiology Research; Copenhagen; Denmark
| | - P. Bager
- Statens Serum Institut, Department of Epidemiology Research; Copenhagen; Denmark
| |
Collapse
|