101
|
Host immune response is severely compromised during lethal Plasmodium vinckei infection. Parasitol Res 2015; 114:3445-57. [PMID: 26077756 DOI: 10.1007/s00436-015-4570-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 06/05/2015] [Indexed: 12/13/2022]
Abstract
Cytokines and immune effector cells play an important role in determining the outcome of infection with various intracellular pathogens, including protozoan parasites. However, their role during lethal and nonlethal malaria needs further validation. In the present study, we examined the role of cytokines and various immune effector cells during lethal and nonlethal malaria caused by Plasmodium vinckei in AKR mice. We show that lethal P. vinckei infection (PvAS) in AKR mice is characterized by increased parasite growth, decreased production of pro-inflammatory cytokines, and attenuated cell proliferation and nitric oxide (NO) synthesis resulting in increased parasitemia which ultimately leads to death of all animals by day 5 post infection. In contrast, AKR mice infected with lethal parasite (PvAR) showed elevated levels of pro-inflammatory cytokines, heightened cell proliferation, and NO synthesis leading to complete parasite clearance by day 22 post infection. Flow cytometric analysis performed on splenocytes from PvAS- and PvAR-infected mice shows that host immunity is severely compromised in PvAS-infected mice as was evident by decreased percentages of CD4(+) and CD8(+) T cells, B cells, plasma cells, dendritic cells (DCs), and macrophages (MΦs) which was in complete contrast to PvAR-infected animals which exhibited elevated numbers of all the cell types analyzed. Taken together, findings of the present study show that coordinated actions of pro-inflammatory cytokines and other immune effector cells are essential to control lethal malarial infection and their attenuation leads to increased parasite growth and, ultimately, death of animals.
Collapse
|
102
|
Matar CG, Anthony NR, O’Flaherty BM, Jacobs NT, Priyamvada L, Engwerda CR, Speck SH, Lamb TJ. Gammaherpesvirus Co-infection with Malaria Suppresses Anti-parasitic Humoral Immunity. PLoS Pathog 2015; 11:e1004858. [PMID: 25996913 PMCID: PMC4440701 DOI: 10.1371/journal.ppat.1004858] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 04/06/2015] [Indexed: 11/18/2022] Open
Abstract
Immunity to non-cerebral severe malaria is estimated to occur within 1-2 infections in areas of endemic transmission for Plasmodium falciparum. Yet, nearly 20% of infected children die annually as a result of severe malaria. Multiple risk factors are postulated to exacerbate malarial disease, one being co-infections with other pathogens. Children living in Sub-Saharan Africa are seropositive for Epstein Barr Virus (EBV) by the age of 6 months. This timing overlaps with the waning of protective maternal antibodies and susceptibility to primary Plasmodium infection. However, the impact of acute EBV infection on the generation of anti-malarial immunity is unknown. Using well established mouse models of infection, we show here that acute, but not latent murine gammaherpesvirus 68 (MHV68) infection suppresses the anti-malarial humoral response to a secondary malaria infection. Importantly, this resulted in the transformation of a non-lethal P. yoelii XNL infection into a lethal one; an outcome that is correlated with a defect in the maintenance of germinal center B cells and T follicular helper (Tfh) cells in the spleen. Furthermore, we have identified the MHV68 M2 protein as an important virus encoded protein that can: (i) suppress anti-MHV68 humoral responses during acute MHV68 infection; and (ii) plays a critical role in the observed suppression of anti-malarial humoral responses in the setting of co-infection. Notably, co-infection with an M2-null mutant MHV68 eliminates lethality of P. yoelii XNL. Collectively, our data demonstrates that an acute gammaherpesvirus infection can negatively impact the development of an anti-malarial immune response. This suggests that acute infection with EBV should be investigated as a risk factor for non-cerebral severe malaria in young children living in areas endemic for Plasmodium transmission.
Collapse
Affiliation(s)
- Caline G. Matar
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Microbiology and Molecular Genetics Graduate Program, Laney Graduate School, Emory University, Atlanta, Georgia, United States of America
| | - Neil R. Anthony
- Division of Pediatric Infectious Disease, Department of Pediatrics, Emory University School of Medicine, Emory Children’s Centre, Atlanta, Georgia, United States of America
| | - Brigid M. O’Flaherty
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Microbiology and Molecular Genetics Graduate Program, Laney Graduate School, Emory University, Atlanta, Georgia, United States of America
| | - Nathan T. Jacobs
- Division of Pediatric Infectious Disease, Department of Pediatrics, Emory University School of Medicine, Emory Children’s Centre, Atlanta, Georgia, United States of America
- Population Biology, Ecology and Evolution Graduate Program, Laney Graduate School, Emory University, Atlanta, Georgia, United States of America
| | - Lalita Priyamvada
- Microbiology and Molecular Genetics Graduate Program, Laney Graduate School, Emory University, Atlanta, Georgia, United States of America
- Division of Pediatric Infectious Disease, Department of Pediatrics, Emory University School of Medicine, Emory Children’s Centre, Atlanta, Georgia, United States of America
| | - Christian R. Engwerda
- Immunology and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Samuel H. Speck
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- Emory Vaccine Center, Emory University, Atlanta, Georgia, United States of America
- * E-mail: (SHS); (TJL)
| | - Tracey J. Lamb
- Division of Pediatric Infectious Disease, Department of Pediatrics, Emory University School of Medicine, Emory Children’s Centre, Atlanta, Georgia, United States of America
- * E-mail: (SHS); (TJL)
| |
Collapse
|
103
|
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
|
104
|
Lewis MD, Behrends J, Sá E Cunha C, Mendes AM, Lasitschka F, Sattler JM, Heiss K, Kooij TWA, Prudêncio M, Bringmann G, Frischknecht F, Mueller AK. Chemical attenuation of Plasmodium in the liver modulates severe malaria disease progression. THE JOURNAL OF IMMUNOLOGY 2015; 194:4860-70. [PMID: 25862814 DOI: 10.4049/jimmunol.1400863] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 03/11/2015] [Indexed: 11/19/2022]
Abstract
Cerebral malaria is one of the most severe complications of malaria disease, attributed to a complicated series of immune reactions in the host. The syndrome is marked by inflammatory immune responses, margination of leukocytes, and parasitized erythrocytes in cerebral vessels leading to breakdown of the blood-brain barrier. We show that chemical attenuation of the parasite at the very early, clinically silent liver stage suppresses parasite development, delays the time until parasites establish blood-stage infection, and provokes an altered host immune response, modifying immunopathogenesis and protecting from cerebral disease. The early response is proinflammatory and cell mediated, with increased T cell activation in the liver and spleen, and greater numbers of effector T cells, cytokine-secreting T cells, and proliferating, proinflammatory cytokine-producing T cells. Dendritic cell numbers, T cell activation, and infiltration of CD8(+) T cells to the brain are decreased later in infection, possibly mediated by the anti-inflammatory cytokine IL-10. Strikingly, protection can be transferred to naive animals by adoptive transfer of lymphocytes from the spleen at very early times of infection. Our data suggest that a subpopulation belonging to CD8(+) T cells as early as day 2 postinfection is responsible for protection. These data indicate that liver stage-directed early immune responses can moderate the overall downstream host immune response and modulate severe malaria outcome.
Collapse
Affiliation(s)
- Matthew D Lewis
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, D 69120 Heidelberg, Germany; German Centre for Infection Research, D 69120 Heidelberg, Germany
| | - Jochen Behrends
- Core Facility Fluorescence Cytometry, Research Center Borstel, D 23845 Borstel, Germany
| | - Cláudia Sá E Cunha
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - António M Mendes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Felix Lasitschka
- German Centre for Infection Research, D 69120 Heidelberg, Germany; Institute of Pathology, Heidelberg University Hospital, D 69120 Heidelberg, Germany
| | - Julia M Sattler
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, D 69120 Heidelberg, Germany
| | - Kirsten Heiss
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, D 69120 Heidelberg, Germany; MalVa GmbH, D 69121 Heidelberg, Germany
| | - Taco W A Kooij
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6500 HB Nijmegen, the Netherlands; Centre for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6500 HB Nijmegen, the Netherlands; and
| | - Miguel Prudêncio
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Gerhard Bringmann
- Institute for Organic Chemistry, University of Würzburg, 97074 Würzburg, Germany
| | - Friedrich Frischknecht
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, D 69120 Heidelberg, Germany
| | - Ann-Kristin Mueller
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, D 69120 Heidelberg, Germany; German Centre for Infection Research, D 69120 Heidelberg, Germany;
| |
Collapse
|
105
|
CD47 regulates the phagocytic clearance and replication of the Plasmodium yoelii malaria parasite. Proc Natl Acad Sci U S A 2015; 112:3062-7. [PMID: 25713361 DOI: 10.1073/pnas.1418144112] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Several Plasmodium species exhibit a strong age-based preference for the red blood cells (RBC) they infect, which in turn is a major determinant of disease severity and pathogenesis. The molecular basis underlying this age constraint on the use of RBC and its influence on parasite burden is poorly understood. CD47 is a marker of self on most cells, including RBC, which, in conjunction with signal regulatory protein alpha (expressed on macrophages), prevents the clearance of cells by the immune system. In this report, we have investigated the role of CD47 on the growth and survival of nonlethal Plasmodium yoelii 17XNL (PyNL) malaria in C57BL/6 mice. By using a quantitative biotin-labeling procedure and a GFP-expressing parasite, we demonstrate that PyNL parasites preferentially infect high levels of CD47 (CD47(hi))-expressing young RBC. Importantly, C57BL/6 CD47(-/-) mice were highly resistant to PyNL infection and developed a 9.3-fold lower peak parasitemia than their wild-type (WT) counterparts. The enhanced resistance to malaria observed in CD47(-/-) mice was associated with a higher percentage of splenic F4/80(+) cells, and these cells had a higher percentage of phagocytized parasitized RBC than infected WT mice during the acute phase of infection, when parasitemia was rapidly rising. Furthermore, injection of CD47-neutralizing antibody caused a significant reduction in parasite burden in WT C57BL/6 mice. Together, these results strongly suggest that CD47(hi) young RBC may provide a shield to the malaria parasite from clearance by the phagocytic cells, which may be an immune escape mechanism used by Plasmodium parasites that preferentially infect young RBC.
Collapse
|
106
|
Boyle MJ, Jagannathan P, Bowen K, McIntyre TI, Vance HM, Farrington LA, Greenhouse B, Nankya F, Rek J, Katureebe A, Arinaitwe E, Dorsey G, Kamya MR, Feeney ME. Effector Phenotype of Plasmodium falciparum-Specific CD4+ T Cells Is Influenced by Both Age and Transmission Intensity in Naturally Exposed Populations. J Infect Dis 2015; 212:416-25. [PMID: 25646355 DOI: 10.1093/infdis/jiv054] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 01/20/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Mechanisms mediating immunity to malaria remain unclear, but animal data and experimental human vaccination models suggest a critical role for CD4(+) T cells. Advances in multiparametric flow cytometry have revealed that the functional quality of pathogen-specific CD4(+) T cells determines immune protection in many infectious models. Little is known about the functional characteristics of Plasmodium-specific CD4(+) T-cell responses in immune and nonimmune individuals. METHODS We compared T-cell responses to Plasmodium falciparum among household-matched children and adults residing in settings of high or low malaria transmission in Uganda. Peripheral blood mononuclear cells were stimulated with P. falciparum antigen, and interferon γ (IFN-γ), interleukin 2, interleukin 10, and tumor necrosis factor α (TNF-α) production was analyzed via multiparametric flow cytometry. RESULTS We found that the magnitude of the CD4(+) T-cell responses was greater in areas of high transmission but similar between children and adults in each setting type. In the high-transmission setting, most P. falciparum-specific CD4(+) T-cells in children produced interleukin 10, while responses in adults were dominated by IFN-γ and TNF-α. In contrast, in the low-transmission setting, responses in both children and adults were dominated by IFN-γ and TNF-α. CONCLUSIONS These findings highlight major differences in the CD4(+) T-cell response of immune adults and nonimmune children that may be relevant for immune protection from malaria.
Collapse
Affiliation(s)
- Michelle J Boyle
- Department of Medicine Center for Biomedical Research, Burnet Institute, Melbourne, Australia
| | | | | | | | | | | | | | | | - John Rek
- Infectious Diseases Research Collaboration
| | | | | | | | - Moses R Kamya
- Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Margaret E Feeney
- Department of Medicine Department of Pediatrics, University of California-San Francisco
| |
Collapse
|
107
|
Besnard AG, Guabiraba R, Niedbala W, Palomo J, Reverchon F, Shaw TN, Couper KN, Ryffel B, Liew FY. IL-33-mediated protection against experimental cerebral malaria is linked to induction of type 2 innate lymphoid cells, M2 macrophages and regulatory T cells. PLoS Pathog 2015; 11:e1004607. [PMID: 25659095 PMCID: PMC4450060 DOI: 10.1371/journal.ppat.1004607] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 12/05/2014] [Indexed: 12/15/2022] Open
Abstract
Cerebral malaria (CM) is a complex parasitic disease caused by Plasmodium sp. Failure to establish an appropriate balance between pro- and anti-inflammatory immune responses is believed to contribute to the development of cerebral pathology. Using the blood-stage PbA (Plasmodium berghei ANKA) model of infection, we show here that administration of the pro-Th2 cytokine, IL-33, prevents the development of experimental cerebral malaria (ECM) in C57BL/6 mice and reduces the production of inflammatory mediators IFN-γ, IL-12 and TNF-α. IL-33 drives the expansion of type-2 innate lymphoid cells (ILC2) that produce Type-2 cytokines (IL-4, IL-5 and IL-13), leading to the polarization of the anti-inflammatory M2 macrophages, which in turn expand Foxp3 regulatory T cells (Tregs). PbA-infected mice adoptively transferred with ILC2 have elevated frequency of M2 and Tregs and are protected from ECM. Importantly, IL-33-treated mice deleted of Tregs (DEREG mice) are no longer able to resist ECM. Our data therefore provide evidence that IL-33 can prevent the development of ECM by orchestrating a protective immune response via ILC2, M2 macrophages and Tregs.
Collapse
Affiliation(s)
- Anne-Gaelle Besnard
- Institute of Infection, Immunity and Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Rodrigo Guabiraba
- Institute of Infection, Immunity and Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, United Kingdom
- INRA, UMR1282, Infectiologie et Santé publique, Nouzilly, France
| | - Wanda Niedbala
- Institute of Infection, Immunity and Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Jennifer Palomo
- CNRS-UMR7355, Orleans, France and Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France
| | - Flora Reverchon
- CNRS-UMR7355, Orleans, France and Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France
| | - Tovah N. Shaw
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Kevin N. Couper
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Bernhard Ryffel
- CNRS-UMR7355, Orleans, France and Experimental and Molecular Immunology and Neurogenetics, University of Orleans, Orleans, France
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondeboasch, Republic of South Africa
| | - Foo Y. Liew
- Institute of Infection, Immunity and Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, United Kingdom
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
108
|
Immunogenetic influences on acquisition of HIV-1 infection: consensus findings from two African cohorts point to an enhancer element in IL19 (1q32.2). Genes Immun 2015; 16:213-20. [PMID: 25633979 PMCID: PMC4409473 DOI: 10.1038/gene.2014.84] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/19/2014] [Accepted: 12/19/2014] [Indexed: 12/12/2022]
Abstract
Numerous reports have suggested that immunogenetic factors may influence HIV-1 acquisition, yet replicated findings that translate between study cohorts remain elusive. Our work aimed to test several hypotheses about genetic variants within the IL10-IL24 gene cluster that encodes interleukin (IL)-10, IL-19, IL-20, and IL-24. In aggregated data from 515 Rwandans and 762 Zambians with up to 12 years of follow-up, 190 single nucleotide polymorphisms (SNPs) passed quality control procedures. When HIV-1-exposed seronegative subjects (n = 486) were compared with newly seroconverted individuals (n = 313) and seroprevalent subjects (n = 478) who were already infected at enrollment, rs12407485 (G>A) in IL19 showed a robust association signal in adjusted logistic regression models (odds ratio = 0.64, P = 1.7 × 10−4, and q = 0.033). Sensitivity analyses demonstrated that (i) results from both cohorts and subgroups within each cohort were highly consistent; (ii) verification of HIV-1 infection status after enrollment was critical; and (iii) supporting evidence was readily obtained from Cox proportional hazards models. Data from public databases indicate that rs12407485 is part of an enhancer element for three transcription factors. Overall, these findings suggest that molecular features at the IL19 locus may modestly alter the establishment of HIV-1 infection.
Collapse
|
109
|
Perez-Mazliah D, Langhorne J. CD4 T-cell subsets in malaria: TH1/TH2 revisited. Front Immunol 2015; 5:671. [PMID: 25628621 PMCID: PMC4290673 DOI: 10.3389/fimmu.2014.00671] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 12/15/2014] [Indexed: 12/13/2022] Open
Abstract
CD4+ T-cells have been shown to play a central role in immune control of infection with Plasmodium parasites. At the erythrocytic stage of infection, IFN-γ production by CD4+ T-cells and CD4+ T-cell help for the B-cell response are required for control and elimination of infected red blood cells. CD4+ T-cells are also important for controlling Plasmodium pre-erythrocytic stages through the activation of parasite-specific CD8+ T-cells. However, excessive inflammatory responses triggered by the infection have been shown to drive pathology. Early classical experiments demonstrated a biphasic CD4+ T-cell response against erythrocytic stages in mice, in which T helper (Th)1 and antibody-helper CD4+ T-cells appear sequentially during a primary infection. While IFN-γ-producing Th1 cells do play a role in controlling acute infections, and they contribute to acute erythrocytic-stage pathology, it became apparent that a classical Th2 response producing IL-4 is not a critical feature of the CD4+ T-cell response during the chronic phase of infection. Rather, effective CD4+ T-cell help for B-cells, which can occur in the absence of IL-4, is required to control chronic parasitemia. IL-10, important to counterbalance inflammation and associated with protection from inflammatory-mediated severe malaria in both humans and experimental models, was originally considered be produced by CD4+ Th2 cells during infection. We review the interpretations of CD4+ T-cell responses during Plasmodium infection, proposed under the original Th1/Th2 paradigm, in light of more recent advances, including the identification of multifunctional T-cells such as Th1 cells co-expressing IFN-γ and IL-10, the identification of follicular helper T-cells (Tfh) as the predominant CD4+ T helper subset for B-cells, and the recognition of inherent plasticity in the fates of different CD4+ T-cells.
Collapse
Affiliation(s)
- Damian Perez-Mazliah
- Division of Parasitology, MRC National Institute for Medical Research , London , UK
| | - Jean Langhorne
- Division of Parasitology, MRC National Institute for Medical Research , London , UK
| |
Collapse
|
110
|
Fernandes P, Frank R, Lewis MD, Mueller AK. Plasmodium attenuation: connecting the dots between early immune responses and malaria disease severity. Front Microbiol 2014; 5:658. [PMID: 25520710 PMCID: PMC4251431 DOI: 10.3389/fmicb.2014.00658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 11/13/2014] [Indexed: 12/21/2022] Open
Abstract
Sterile attenuation of Plasmodium parasites at the liver-stage either by irradiation or genetic modification, or at the blood-stage by chemoprophylaxis, has been shown to induce immune responses that can protect against subsequent wild-type infection. However, following certain interventions, parasite attenuation can be incomplete or non-sterile. Instead parasites are rendered developmentally stunted but still capable of establishing an acute infection. In experiments involving Plasmodium berghei ANKA, a model of experimental cerebral malaria, it has been observed that several forms of attenuated parasites do not induce cerebral pathology. In this perspective we collect evidence from studies on murine malaria in particular, and attempt to “connect the dots” between early immune responses and protection from severe cerebral disease, highlighting potential parallels to human infection.
Collapse
Affiliation(s)
- Priyanka Fernandes
- Parasitology Unit, Centre of Infectious Diseases, University Hospital Heidelberg Heidelberg, Germany
| | - Roland Frank
- Parasitology Unit, Centre of Infectious Diseases, University Hospital Heidelberg Heidelberg, Germany
| | - Matthew D Lewis
- Parasitology Unit, Centre of Infectious Diseases, University Hospital Heidelberg Heidelberg, Germany
| | - Ann-Kristin Mueller
- Parasitology Unit, Centre of Infectious Diseases, University Hospital Heidelberg Heidelberg, Germany
| |
Collapse
|
111
|
Engwerda CR, Ng SS, Bunn PT. The Regulation of CD4(+) T Cell Responses during Protozoan Infections. Front Immunol 2014; 5:498. [PMID: 25352846 PMCID: PMC4195384 DOI: 10.3389/fimmu.2014.00498] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/25/2014] [Indexed: 12/20/2022] Open
Abstract
CD4(+) T cells are critical for defense against protozoan parasites. Intracellular protozoan parasite infections generally require the development of a Th1 cell response, characterized by the production of IFNγ and TNF that are critical for the generation of microbicidal molecules by phagocytes, as well as the expression of cytokines and cell surface molecules needed to generate cytolytic CD8(+) T cells that can recognize and kill infected host cells. Over the past 25 years, much has been learnt about the molecular and cellular components necessary for the generation of Th1 cell responses, and it has become clear that these responses need to be tightly controlled to prevent disease. However, our understanding of the immunoregulatory mechanisms activated during infection is still not complete. Furthermore, it is apparent that although these mechanisms are critical to prevent inflammation, they can also promote parasite persistence and development of disease. Here, we review how CD4(+) T cells are controlled during protozoan infections and how these regulatory mechanisms can influence parasite growth and disease outcome.
Collapse
Affiliation(s)
| | - Susanna S. Ng
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Natural Sciences, Griffith University, Nathan, QLD, Australia
| | - Patrick T. Bunn
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Institute of Glycomics, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
112
|
Kamangira B, Nyamugure P, Magombedze G. A theoretical mathematical assessment of the effectiveness of coartemether in the treatment of Plasmodium falciparum malaria infection. Math Biosci 2014; 256:28-41. [DOI: 10.1016/j.mbs.2014.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Revised: 07/29/2014] [Accepted: 07/31/2014] [Indexed: 10/24/2022]
|
113
|
Helminth parasites alter protection against Plasmodium infection. BIOMED RESEARCH INTERNATIONAL 2014; 2014:913696. [PMID: 25276830 PMCID: PMC4170705 DOI: 10.1155/2014/913696] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 08/06/2014] [Indexed: 12/17/2022]
Abstract
More than one-third of the world's population is infected with one or more helminthic parasites. Helminth infections are prevalent throughout tropical and subtropical regions where malaria pathogens are transmitted. Malaria is the most widespread and deadliest parasitic disease. The severity of the disease is strongly related to parasite density and the host's immune responses. Furthermore, coinfections between both parasites occur frequently. However, little is known regarding how concomitant infection with helminths and Plasmodium affects the host's immune response. Helminthic infections are frequently massive, chronic, and strong inductors of a Th2-type response. This implies that infection by such parasites could alter the host's susceptibility to subsequent infections by Plasmodium. There are a number of reports on the interactions between helminths and Plasmodium; in some, the burden of Plasmodium parasites increased, but others reported a reduction in the parasite. This review focuses on explaining many of these discrepancies regarding helminth-Plasmodium coinfections in terms of the effects that helminths have on the immune system. In particular, it focuses on helminth-induced immunosuppression and the effects of cytokines controlling polarization toward the Th1 or Th2 arms of the immune response.
Collapse
|
114
|
Changes in antigen-specific cytokine and chemokine responses to Plasmodium falciparum antigens in a highland area of Kenya after a prolonged absence of malaria exposure. Infect Immun 2014; 82:3775-82. [PMID: 24958707 DOI: 10.1128/iai.01924-14] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Individuals naturally exposed to Plasmodium falciparum lose clinical immunity after a prolonged lack of exposure. P. falciparum antigen-specific cytokine responses have been associated with protection from clinical malaria, but the longevity of P. falciparum antigen-specific cytokine responses in the absence of exposure is not well characterized. A highland area of Kenya with low and unstable malaria transmission provided an opportunity to study this question. The levels of antigen-specific cytokines and chemokines associated in previous studies with protection from clinical malaria (gamma interferon [IFN-γ], interleukin-10 [IL-10], and tumor necrosis factor alpha [TNF-α]), with increased risk of clinical malaria (IL-6), or with pathogenesis of severe disease in malaria (IL-5 and RANTES) were assessed by cytometric bead assay in April 2008, October 2008, and April 2009 in 100 children and adults. During the 1-year study period, none had an episode of clinical P. falciparum malaria. Two patterns of cytokine responses emerged, with some variation by antigen: a decrease at 6 months (IFN-γ and IL-5) or at both 6 and 12 months (IL-10 and TNF-α) or no change over time (IL-6 and RANTES). These findings document that P. falciparum antigen-specific cytokine responses associated in prior studies with protection from malaria (IFN-γ, TNF-α, and IL-10) decrease significantly in the absence of P. falciparum exposure, whereas those associated with increased risk of malaria (IL-6) do not. The study findings provide a strong rationale for future studies of antigen-specific IFN-γ, TNF-α, and IL-10 responses as biomarkers of increased population-level susceptibility to malaria after prolonged lack of P. falciparum exposure.
Collapse
|
115
|
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
|
116
|
de Souza JB. Protective immunity against malaria after vaccination. Parasite Immunol 2014; 36:131-9. [PMID: 24188045 DOI: 10.1111/pim.12086] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 10/29/2013] [Indexed: 11/28/2022]
Abstract
A good understanding of the immunological correlates of protective immunity is an important requirement for the development of effective vaccines against malaria. However, this concern has received little attention even in the face of two decades of intensive vaccine research. Here, we review the immune response to blood-stage malaria, with a particular focus on the type of vaccine most likely to induce the kind of response required to give strong protection against infection.
Collapse
Affiliation(s)
- J B de Souza
- Faculty of Infectious and Tropical Diseases, Department of Immunity and Infection, London School of Hygiene & Tropical Medicine, London, UK; Division of Infection & Immunity, University College London Medical School, London, UK
| |
Collapse
|
117
|
Wykes MN, Horne-Debets JM, Leow CY, Karunarathne DS. Malaria drives T cells to exhaustion. Front Microbiol 2014; 5:249. [PMID: 24904561 PMCID: PMC4034037 DOI: 10.3389/fmicb.2014.00249] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 05/07/2014] [Indexed: 01/08/2023] Open
Abstract
Malaria is a significant global burden but after >30 years of effort there is no vaccine on the market. While the complex life cycle of the parasite presents several challenges, many years of research have also identified several mechanisms of immune evasion by Plasmodium spp. Recent research on malaria, has investigated the programmed cell death-1 (PD-1) pathway which mediates exhaustion of T cells, characterized by poor effector functions and recall responses and in some cases loss of the cells by apoptosis. Such studies have shown exhaustion of CD4(+) T cells and an unappreciated role for CD8(+) T cells in promoting sterile immunity against blood stage malaria. This is because PD-1 mediates up to a 95% reduction in numbers and functional capacity of parasite-specific CD8(+) T cells, thus masking their role in protection. The role of T cell exhaustion during malaria provides an explanation for the absence of sterile immunity following the clearance of acute disease which will be relevant to future malaria-vaccine design and suggests the need for novel therapeutic solutions. This review will thus examine the role of PD-1-mediated T cell exhaustion in preventing lasting immunity against malaria.
Collapse
Affiliation(s)
- Michelle N Wykes
- Molecular Immunology Laboratory, QIMR Berghofer Medical Research Institute Brisbane, QLD, Australia
| | - Joshua M Horne-Debets
- Molecular Immunology Laboratory, QIMR Berghofer Medical Research Institute Brisbane, QLD, Australia ; The School of Medicine, University of Queensland Brisbane, QLD, Australia
| | - Chiuan-Yee Leow
- Molecular Immunology Laboratory, QIMR Berghofer Medical Research Institute Brisbane, QLD, Australia
| | | |
Collapse
|
118
|
Gonçalves RM, Lima NF, Ferreira MU. Parasite virulence, co-infections and cytokine balance in malaria. Pathog Glob Health 2014; 108:173-8. [PMID: 24854175 DOI: 10.1179/2047773214y.0000000139] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Strong early inflammatory responses followed by a timely production of regulatory cytokines are required to control malaria parasite multiplication without inducing major host pathology. Here, we briefly examine the homeostasis of inflammatory responses to malaria parasite species with varying virulence levels and discuss how co-infections with bacteria, viruses, and helminths can modulate inflammation, either aggravating or alleviating malaria-related morbidity.
Collapse
|
119
|
Lokken KL, Mooney JP, Butler BP, Xavier MN, Chau JY, Schaltenberg N, Begum RH, Müller W, Luckhart S, Tsolis RM. Malaria parasite infection compromises control of concurrent systemic non-typhoidal Salmonella infection via IL-10-mediated alteration of myeloid cell function. PLoS Pathog 2014; 10:e1004049. [PMID: 24787713 PMCID: PMC4006898 DOI: 10.1371/journal.ppat.1004049] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 02/18/2014] [Indexed: 12/23/2022] Open
Abstract
Non-typhoidal Salmonella serotypes (NTS) cause a self-limited gastroenteritis in immunocompetent individuals, while children with severe Plasmodium falciparum malaria can develop a life-threatening disseminated infection. This co-infection is a major source of child mortality in sub-Saharan Africa. However, the mechanisms by which malaria contributes to increased risk of NTS bacteremia are incompletely understood. Here, we report that in a mouse co-infection model, malaria parasite infection blunts inflammatory responses to NTS, leading to decreased inflammatory pathology and increased systemic bacterial colonization. Blunting of NTS-induced inflammatory responses required induction of IL-10 by the parasites. In the absence of malaria parasite infection, administration of recombinant IL-10 together with induction of anemia had an additive effect on systemic bacterial colonization. Mice that were conditionally deficient for either myeloid cell IL-10 production or myeloid cell expression of IL-10 receptor were better able to control systemic Salmonella infection, suggesting that phagocytic cells are both producers and targets of malaria parasite-induced IL-10. Thus, IL-10 produced during the immune response to malaria increases susceptibility to disseminated NTS infection by suppressing the ability of myeloid cells, most likely macrophages, to control bacterial infection.
Collapse
Affiliation(s)
- Kristen L. Lokken
- Department of Microbiology & Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
| | - Jason P. Mooney
- Department of Microbiology & Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
| | - Brian P. Butler
- Department of Microbiology & Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
| | - Mariana N. Xavier
- Department of Microbiology & Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
- Departamento de Clínica e Cirurgia Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jennifer Y. Chau
- Department of Microbiology & Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
| | - Nicola Schaltenberg
- Department of Microbiology & Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
| | - Ramie H. Begum
- Department of Microbiology & Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
- Department of Life Sciences & Bioinformatics, Assam University, Diphu Campus, Karbi Anglong, Assam, India
| | - Werner Müller
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Shirley Luckhart
- Department of Microbiology & Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
| | - Renée M. Tsolis
- Department of Microbiology & Immunology, School of Medicine, University of California at Davis, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
120
|
Keswani T, Bhattacharyya A. Differential role of T regulatory and Th17 in Swiss mice infected with Plasmodium berghei ANKA and Plasmodium yoelii. Exp Parasitol 2014; 141:82-92. [PMID: 24675415 DOI: 10.1016/j.exppara.2014.03.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 01/21/2014] [Accepted: 03/04/2014] [Indexed: 12/31/2022]
Abstract
The outcome of malaria infection is determined, in part, by the balance of pro-inflammatory and regulatory immune responses. Host immune responses in disease including malaria are finely regulated by the opposing effects of Th17 and T regulatory (Treg) cells. Here we have examined the role of Treg cells and Th17 cells during malaria infection and find that low levels of Treg cells possibly influence the outcome of infections with the lethal strain of Plasmodium berghei ANKA (PbA). In contrast, high level of Treg cells may influence the outcome of nonlethal Plasmodium yoelii NXL (P. yoelii) infections. We observed decreased expressions of key regulators of Treg inductions-TGF-β, CD4IL-2 and IL-10 during PbA infection, whereas their expression remains high during P. yoelii infection. On the other hand TNF-α, IL-6, IFN-γ and IL-23 expression is high during PbA infection and lower during P. yoelii infection. Thus, results from this study suggest that the differential expression of Treg and Th17 might have a key role on host pathogenesis during malaria infection. The high level of IL-6 and low level of TGF-β may composite of the advantaged local microenvironment for the production of Th17 cells in the spleen of the PBA infected mice and vice verse during nonlethal P. yoelii.
Collapse
Affiliation(s)
- Tarun Keswani
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India.
| | - Arindam Bhattacharyya
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India.
| |
Collapse
|
121
|
Torres KJ, Villasis E, Bendezú J, Chauca J, Vinetz JM, Gamboa D. Relationship of regulatory T cells to Plasmodium falciparum malaria symptomatology in a hypoendemic region. Malar J 2014; 13:108. [PMID: 24642188 PMCID: PMC3976150 DOI: 10.1186/1475-2875-13-108] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 03/14/2014] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Previous data have suggested that regulatory T cells (Tregs) balance protective immune responses with immune mediated pathology in malaria. This study aimed to determine to test the hypothesis that Treg proportions or absolute levels are associated with parasitaemia and malaria symptoms. METHODS Treg cells were quantified by flow cytometry as CD4+ CD25+, Foxp3+, CD127(low) T cells. Three patient groups were assessed: patients with symptomatic Plasmodium falciparum malaria (S), subjects with asymptomatic P. falciparum parasitaemia (AS) and uninfected control individuals (C). RESULTS S, AS and C groups had similar absolute numbers and percentage of Tregs (3.9%, 3.5% and 3.5% respectively). Levels of parasitaemia were not associated with Treg percentage (p = 0.47). CONCLUSION Neither relative nor absolute regulatory T cell numbers were found to be associated with malaria-related symptomatology in this study. Immune mechanisms other than Tregs are likely to be responsible for the state of asymptomatic P. falciparum parasitaemia in the Peruvian Amazon; but further study to explore these mechanisms is needed.
Collapse
Affiliation(s)
| | | | | | | | - Joseph M Vinetz
- Instituto de Medicina Tropical "Alexander Von Humboldt", Universidad Peruana, Cayetano Heredia, AP 4314, Lima 100, Peru.
| | | |
Collapse
|
122
|
Cheng Q, Zhang Q, Xu X, Yin L, Sun L, Lin X, Dong C, Pan W. MAPK phosphotase 5 deficiency contributes to protection against blood-stage Plasmodium yoelii 17XL infection in mice. THE JOURNAL OF IMMUNOLOGY 2014; 192:3686-96. [PMID: 24634491 DOI: 10.4049/jimmunol.1301863] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cell-mediated immunity plays a crucial role in the development of host resistance to asexual blood-stage malaria infection. However, little is known of the regulatory factors involved in this process. In this study, we investigated the impact of MAPK phosphotase 5 (MKP5) on protective immunity against a lethal Plasmodium yoelii 17XL blood-stage infection using MKP5 knockout C57BL/6 mice. Compared with wild-type control mice, MKP5 knockout mice developed significantly lower parasite burdens with prolonged survival times. We found that this phenomenon correlated with a rapid and strong IFN-γ-dependent cellular immune response during the acute phase of infection. Inactivation of IFN-γ by the administration of a neutralizing Ab significantly reduced the protective effects in MKP5 knockout mice. By analyzing IFN-γ production in innate and adaptive lymphocyte subsets, we observed that MKP5 deficiency specifically enhanced the IFN-γ response mediated by CD4+ T cells, which was attributable to the increased stimulatory capacity of splenic CD11c+ dendritic cells. Furthermore, following vaccination with whole blood-stage soluble plasmodial Ag, MKP5 knockout mice acquired strongly enhanced Ag-specific immune responses and a higher level of protection against subsequent P. yoelii 17XL challenge. Finally, we found the enhanced response mediated by MKP5 deficiency resulted in a lethal consequence in mice when infected with nonlethal P. yoelii 17XNL. Thus, our data indicate that MKP5 is a potential regulator of immune resistance against Plasmodium infection in mice, and that an understanding of the role of MKP5 in manipulating anti-malaria immunity may provide valuable information on the development of better control strategies for human malaria.
Collapse
Affiliation(s)
- Qianqian Cheng
- Institute of Infectious Disease and Vaccine Development, Tongji University School of Medicine, Shanghai 200092, China
| | | | | | | | | | | | | | | |
Collapse
|
123
|
Chattopadhyay G, Shevach EM. Antigen-specific induced T regulatory cells impair dendritic cell function via an IL-10/MARCH1-dependent mechanism. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 191:5875-84. [PMID: 24218453 PMCID: PMC3858537 DOI: 10.4049/jimmunol.1301693] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Foxp3(+) T regulatory cells (Tregs) are critically important for the maintenance of immunological tolerance, immune homeostasis, and prevention of autoimmunity. Dendritic cells (DCs) are one of the major targets of Treg-mediated suppression. Some studies have suggested that Treg-mediated suppression of DC function is mediated by the interaction of CTLA-4 on Tregs with CD80/CD86 on the DCs resulting in downregulation of CD80/CD86 expression and a decrease in costimulation. We have re-examined the effects of Tregs on mouse DC function in a model in which Ag-specific, induced Tregs (iTregs) are cocultured with DCs in the absence of T effector cells. iTreg-treated DCs are markedly defective in their capacity to activate naive T cells. iTregs from CTLA-4-deficient mice failed to induce downregulation of CD80/CD86, but DCs treated with CTLA-4-deficient iTregs still exhibited impaired capacity to activate naive T cells. The iTreg-induced defect in DC function could be completely reversed by anti-IL-10, and IL-10-deficient iTregs failed to downregulate DC function. iTreg-treated DCs expressed high levels of MARCH1, an E3 ubiquitin ligase, recently found to degrade CD86 and MHC class II on the DCs and expressed lower levels of CD83, a molecule involved in neutralizing the function of MARCH1. Both the enhanced expression of MARCH1 and the decreased expression of CD83 were mediated by IL-10 produced by the iTregs. Taken together, these studies demonstrate that a major suppressive mechanism of DC function by iTregs is secondary to the effects of IL-10 on MARCH1 and CD83 expression.
Collapse
MESH Headings
- Animals
- Antigen Presentation
- Antigens, CD/biosynthesis
- Antigens, CD/genetics
- Antigens, CD/physiology
- B7-1 Antigen/biosynthesis
- B7-1 Antigen/genetics
- B7-2 Antigen/biosynthesis
- B7-2 Antigen/genetics
- CD4-Positive T-Lymphocytes/immunology
- CTLA-4 Antigen/deficiency
- CTLA-4 Antigen/physiology
- Cell Separation
- Cells, Cultured
- Coculture Techniques
- DNA-Binding Proteins/deficiency
- Dendritic Cells/immunology
- Epitopes, T-Lymphocyte/immunology
- Flow Cytometry
- Gene Expression Regulation/immunology
- Histocompatibility Antigens Class II/immunology
- Immune Tolerance/immunology
- Immunoglobulins/biosynthesis
- Immunoglobulins/genetics
- Immunoglobulins/physiology
- Interleukin-10/antagonists & inhibitors
- Interleukin-10/deficiency
- Interleukin-10/metabolism
- Interleukin-10/physiology
- Lymphocyte Activation
- Membrane Glycoproteins/biosynthesis
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- RNA, Messenger/biosynthesis
- T-Cell Antigen Receptor Specificity
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Ubiquitin-Protein Ligases/biosynthesis
- Ubiquitin-Protein Ligases/genetics
- Ubiquitin-Protein Ligases/physiology
- CD83 Antigen
Collapse
Affiliation(s)
- Gouri Chattopadhyay
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | | |
Collapse
|
124
|
Liver accumulation of Plasmodium chabaudi-infected red blood cells and modulation of regulatory T cell and dendritic cell responses. PLoS One 2013; 8:e81409. [PMID: 24312297 PMCID: PMC3842419 DOI: 10.1371/journal.pone.0081409] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 10/12/2013] [Indexed: 12/11/2022] Open
Abstract
It is postulated that accumulation of malaria-infected Red Blood Cells (iRBCs) in the liver could be a parasitic escape mechanism against full destruction by the host immune system. Therefore, we evaluated the in vivo mechanism of this accumulation and its potential immunological consequences. A massive liver accumulation of P. c. chabaudi AS-iRBCs (Pc-iRBCs) was observed by intravital microscopy along with an over expression of ICAM-1 on day 7 of the infection, as measured by qRT-PCR. Phenotypic changes were also observed in regulatory T cells (Tregs) and dendritic cells (DCs) that were isolated from infected livers, which indicate a functional role for Tregs in the regulation of the liver inflammatory immune response. In fact, the suppressive function of liver-Tregs was in vitro tested, which demonstrated the capacity of these cells to suppress naive T cell activation to the same extent as that observed for spleen-Tregs. On the other hand, it is already known that CD4+ T cells isolated from spleens of protozoan parasite-infected mice are refractory to proliferate in vivo. In our experiments, we observed a similar lack of in vitro proliferative capacity in liver CD4+ T cells that were isolated on day 7 of infection. It is also known that nitric oxide and IL-10 are partially involved in acute phase immunosuppression; we found high expression levels of IL-10 and iNOS mRNA in day 7-infected livers, which indicates a possible role for these molecules in the observed immune suppression. Taken together, these results indicate that malaria parasite accumulation within the liver could be an escape mechanism to avoid sterile immunity sponsored by a tolerogenic environment.
Collapse
|
125
|
Butler NS, Harris TH, Blader IJ. Regulation of immunopathogenesis during Plasmodium and Toxoplasma infections: more parallels than distinctions? Trends Parasitol 2013; 29:593-602. [PMID: 24184186 DOI: 10.1016/j.pt.2013.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Revised: 10/03/2013] [Accepted: 10/04/2013] [Indexed: 01/08/2023]
Abstract
Toxoplasma and Plasmodium parasites exact a significant toll on public health. Host immunity required for efficient control of infection by these Apicomplexans involves the induction of potent T cell responses, which sometimes results in immunopathological damage. Thus, protective immune responses must be balanced by regulatory networks that limit immunopathology. We review several key cellular and molecular immunoregulatory networks operational during Toxoplasma and Plasmodium infections. Accumulating data show that despite differences in how the immune response controls these parasites, many host immunoregulatory pathways and cellular networks are common to both. Thus, understanding the cellular and molecular circuits that prevent or regulate immunopathological responses against one parasite is likely to inform our understanding of the host response to the other parasite.
Collapse
Affiliation(s)
- Noah S Butler
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | | | | |
Collapse
|
126
|
HLA class II (DR0401) molecules induce Foxp3+ regulatory T cell suppression of B cells in Plasmodium yoelii strain 17XNL malaria. Infect Immun 2013; 82:286-97. [PMID: 24166949 DOI: 10.1128/iai.00272-13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Unlike human malaria parasites that induce persistent infection, some rodent malaria parasites, like Plasmodium yoelii strain 17XNL (Py17XNL), induce a transient (self-curing) malaria infection. Cooperation between CD4 T cells and B cells to produce antibodies is thought to be critical for clearance of Py17XNL parasites from the blood, with major histocompatibility complex (MHC) class II molecules being required for activation of CD4 T cells. In order to better understand the correspondence between murine malaria models and human malaria, and in particular the role of MHC (HLA) class II molecules, we studied the ability of humanized mice expressing human HLA class II molecules to clear Py17XNL infection. We showed that humanized mice expressing HLA-DR4 (DR0401) molecules and lacking mouse MHC class II molecules (EA(0)) have impaired production of specific antibodies to Py17XNL and cannot cure the infection. In contrast, mice expressing HLA-DR4 (DR0402), HLA-DQ6 (DQ0601), HLA-DQ8 (DQ0302), or HLA-DR3 (DR0301) molecules in an EA(0) background were able to elicit specific antibodies and self-cure the infection. In a series of experiments, we determined that the inability of humanized DR0401.EA(0) mice to elicit specific antibodies was due to expansion and activation of regulatory CD4(+) Foxp3(+) T cells (Tregs) that suppressed B cells to secrete antibodies through cell-cell interactions. Treg depletion allowed the DR0401.EA(0) mice to elicit specific antibodies and self-cure the infection. Our results demonstrated a differential role of MHC (HLA) class II molecules in supporting antibody responses to Py17XNL malaria and revealed a new mechanism by which malaria parasites stimulate B cell-suppressogenic Tregs that prevent clearance of infection.
Collapse
|
127
|
Fevre C, Almeida AS, Taront S, Pedron T, Huerre M, Prevost MC, Kieusseian A, Cumano A, Brisse S, Sansonetti PJ, Tournebize R. A novel murine model of rhinoscleroma identifies Mikulicz cells, the disease signature, as IL-10 dependent derivatives of inflammatory monocytes. EMBO Mol Med 2013; 5:516-30. [PMID: 23554169 PMCID: PMC3628109 DOI: 10.1002/emmm.201202023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 01/31/2013] [Accepted: 02/14/2013] [Indexed: 12/21/2022] Open
Abstract
Rhinoscleroma is a human specific chronic disease characterized by the formation of granuloma in the airways, caused by the bacterium Klebsiella pneumoniae subspecies rhinoscleromatis, a species very closely related to K. pneumoniae subspecies pneumoniae. It is characterized by the appearance of specific foamy macrophages called Mikulicz cells. However, very little is known about the pathophysiological processes underlying rhinoscleroma. Herein, we characterized a murine model recapitulating the formation of Mikulicz cells in lungs and identified them as atypical inflammatory monocytes specifically recruited from the bone marrow upon K. rhinoscleromatis infection in a CCR2-independent manner. While K. pneumoniae and K. rhinoscleromatis infections induced a classical inflammatory reaction, K. rhinoscleromatis infection was characterized by a strong production of IL-10 concomitant to the appearance of Mikulicz cells. Strikingly, in the absence of IL-10, very few Mikulicz cells were observed, confirming a crucial role of IL-10 in the establishment of a proper environment leading to the maturation of these atypical monocytes. This is the first characterization of the environment leading to Mikulicz cells maturation and their identification as inflammatory monocytes.
Collapse
Affiliation(s)
- Cindy Fevre
- Institut Pasteur, Génotypage des Pathogènes et Santé Publique, Paris Cedex, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Immune activation and regulation in simian immunodeficiency virus-Plasmodium fragile-coinfected rhesus macaques. J Virol 2013; 87:9523-37. [PMID: 23785209 DOI: 10.1128/jvi.00861-13] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human immunodeficiency virus (HIV) is characterized by immune activation, while chronic malaria is associated with elevated interleukin-10 (IL-10) levels. How these apparently antagonizing forces interact in the coinfected host is poorly understood. Using a rhesus macaque model of simian immunodeficiency virus (SIV)-Plasmodium fragile coinfection, we evaluated how innate immune effector cells affect the balance between immune activation and regulation. In vitro Toll-like receptor (TLR) responses of peripheral blood myeloid dendritic cells (mDC) and monocytes were temporarily associated with acute parasitemic episodes and elevated plasma IL-10 levels. Prolonged infection resulted in a decline of mDC function. Monocytes maintained TLR responsiveness but, in addition to IL-12 and tumor necrosis factor alpha, also produced IL-10. Consistent with the role of spleen in the clearance of parasite-infected red blood cells, coinfected animals also had increased splenic IL-10 mRNA levels. The main cellular source of IL-10 in the spleens of coinfected animals, however, was not splenic macrophages but T cells, suggesting an impairment of adaptive immunity. In contrast to those in spleen, IL-10-positive cells in axillary lymph nodes of coinfected animals were predominantly mDC, reminiscent of the immunosuppressive phenotype of peripheral blood mDC. Concurrent with IL-10 induction, however, SIV infection promoted elevated systemic IL-12 levels. The continuously increasing ratio of plasma IL-12 to IL-10 suggested that the overall host response in SIV-P. fragile-coinfected animals was shifted toward immune activation versus immune regulation. Therefore, SIV-P. fragile coinfection might be characterized by earlier manifestation of immune dysfunction and exhaustion than that of single-pathogen infections. This could translate into increased morbidity in HIV-malaria-coinfected individuals.
Collapse
|
129
|
Bao LQ, Huy NT, Kikuchi M, Yanagi T, Senba M, Shuaibu MN, Honma K, Yui K, Hirayama K. CD19(+) B cells confer protection against experimental cerebral malaria in semi-immune rodent model. PLoS One 2013; 8:e64836. [PMID: 23724100 PMCID: PMC3665539 DOI: 10.1371/journal.pone.0064836] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 04/19/2013] [Indexed: 12/31/2022] Open
Abstract
In African endemic area, adults are less vulnerable to cerebral malaria than children probably because of acquired partial immunity or semi-immune status. Here, we developed an experimental cerebral malaria (ECM) model for semi-immune mice. C57BL/6 (B6) mice underwent one, two and three cycles of infection and radical treatment (1-cure, 2-cure and 3-cure, respectively) before being finally challenged with 104Plasmodium berghei ANKA without treatment. Our results showed that 100% of naïve (0-cure), 67% of 1-cure, 37% of 2-cure and none of 3-cure mice succumbed to ECM within 10 days post challenge infection. In the protected 3-cure mice, significantly higher levels of plasma IL-10 and lower levels of IFN-γ than the others on day 7 post challenge infection were observed. Major increased lymphocyte subset of IL-10 positive cells in 3-cure mice was CD5(−)CD19(+) B cells. Passive transfer of splenic CD19(+) cells from 3-cure mice protected naïve mice from ECM. Additionally, aged 3-cure mice were also protected from ECM 12 and 20 months after the last challenge infection. In conclusion, mice became completely resistant to ECM after three exposures to malaria. CD19(+) B cells are determinants in protective mechanism of semi-immune mice against ECM possibly via modulatory IL-10 for pathogenic IFN-γ production.
Collapse
Affiliation(s)
- Lam Quoc Bao
- Department of Immunogenetics, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
- Global Center of Excellence (GCOE), Nagasaki University, Nagasaki, Japan
| | - Nguyen Tien Huy
- Department of Immunogenetics, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
- * E-mail: (KH); (NTH)
| | - Mihoko Kikuchi
- Center for International Collaborative Research, Nagasaki University, Nagasaki, Japan
| | - Tetsuo Yanagi
- Animal Research Center for Tropical Infections, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Masachika Senba
- Department of Pathology, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Mohammed Nasir Shuaibu
- Department of Immunogenetics, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
- Global Center of Excellence (GCOE), Nagasaki University, Nagasaki, Japan
| | - Kiri Honma
- Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Katsuyuki Yui
- Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Global Center of Excellence (GCOE), Nagasaki University, Nagasaki, Japan
| | - Kenji Hirayama
- Department of Immunogenetics, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
- Global Center of Excellence (GCOE), Nagasaki University, Nagasaki, Japan
- * E-mail: (KH); (NTH)
| |
Collapse
|
130
|
McCarthy Ú, Casadei E, Wang T, Secombes CJ. Red mark syndrome in rainbow trout Oncorhynchus mykiss: investigation of immune responses in lesions using histology, immunohistochemistry and analysis of immune gene expression. FISH & SHELLFISH IMMUNOLOGY 2013; 34:1119-1130. [PMID: 23403161 DOI: 10.1016/j.fsi.2013.01.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 11/20/2012] [Accepted: 01/22/2013] [Indexed: 06/01/2023]
Abstract
Red mark syndrome (RMS) is an economically significant disease which affects farmed rainbow trout in the United Kingdom, in the US and in mainland Europe. From the pattern of incidence, it appears to be transmissable, although no causative agent has yet been identified. RMS presents as a severe lymphocytic infiltration centred on the dermis and an alternative, host-focused approach was taken to understand the disease through investigating immune responses occurring in the lesion. Lesion and non-lesion skin at different stages of lesion development were examined using histochemistry and immunohistochemistry on paraffin sections. Expression of immune-related genes was compared between lesion and non-lesion skin. Investigation of early stage lesions suggested that the initial immune response is targeted at the region of the scale pocket, with lymphocyte infiltration and anti-tumour necrosis factor (TNF)-α staining of the stratum spongiosum, and increased numbers of major histocompatibility complex (MHC) II-positive cells immediately adjacent to the scale pocket. Gene expression analysis suggested a counterbalancing T helper (Th)1 and T regulatory (Treg) - type response is occurring in the lesion, with repression of Th2 and Th17-type responses.
Collapse
Affiliation(s)
- Ú McCarthy
- Ellis Building, Marine Scotland Science, Marine Laboratory, 375 Victoria Road, PO Box 101, Aberdeen AB11 9DB, Scotland, UK.
| | | | | | | |
Collapse
|
131
|
Villegas-Mendez A, de Souza JB, Lavelle SW, Gwyer Findlay E, Shaw TN, van Rooijen N, Saris CJ, Hunter CA, Riley EM, Couper KN. IL-27 receptor signalling restricts the formation of pathogenic, terminally differentiated Th1 cells during malaria infection by repressing IL-12 dependent signals. PLoS Pathog 2013; 9:e1003293. [PMID: 23593003 PMCID: PMC3623720 DOI: 10.1371/journal.ppat.1003293] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 02/19/2013] [Indexed: 11/18/2022] Open
Abstract
The IL-27R, WSX-1, is required to limit IFN-γ production by effector CD4+ T cells in a number of different inflammatory conditions but the molecular basis of WSX-1-mediated regulation of Th1 responses in vivo during infection has not been investigated in detail. In this study we demonstrate that WSX-1 signalling suppresses the development of pathogenic, terminally differentiated (KLRG-1+) Th1 cells during malaria infection and establishes a restrictive threshold to constrain the emergent Th1 response. Importantly, we show that WSX-1 regulates cell-intrinsic responsiveness to IL-12 and IL-2, but the fate of the effector CD4+ T cell pool during malaria infection is controlled primarily through IL-12 dependent signals. Finally, we show that WSX-1 regulates Th1 cell terminal differentiation during malaria infection through IL-10 and Foxp3 independent mechanisms; the kinetics and magnitude of the Th1 response, and the degree of Th1 cell terminal differentiation, were comparable in WT, IL-10R1−/− and IL-10−/− mice and the numbers and phenotype of Foxp3+ cells were largely unaltered in WSX-1−/− mice during infection. As expected, depletion of Foxp3+ cells did not enhance Th1 cell polarisation or terminal differentiation during malaria infection. Our results significantly expand our understanding of how IL-27 regulates Th1 responses in vivo during inflammatory conditions and establishes WSX-1 as a critical and non-redundant regulator of the emergent Th1 effector response during malaria infection. The cytokine interleukin 27 (IL-27), a member of the IL-12 family, is produced by cells of the innate immune system and has been shown to exert mainly suppressive effects during a wide range of inflammatory conditions, including malaria infection, where it suppresses the development of CD4+ T cell-dependent immunopathology. In this study we show that IL-27 suppresses the production of IFN-gamma by CD4+ T cells during blood stage malaria infection by preventing the development of terminally differentiated Th1 cells. We investigated the molecular mechanisms by which IL-27 inhibits the formation of terminally differentiated Th1 cells and found that it does so specifically by restricting IL-12 signals. Importantly, we demonstrate that IL-27 mediates its regulatory effects on the Th1 response through IL-10 and Foxp3+ regulatory T cell independent mechanisms. Thus, we have identified a new pathway though which IL-27 signalling regulates the size and quality of the Th1 response during malaria infection, which we believe will have relevance to many other pro-inflammatory conditions. Manipulation of the IL-27 pathway may therefore represent an amenable therapeutic approach during chronic inflammatory disorders.
Collapse
Affiliation(s)
- Ana Villegas-Mendez
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - J. Brian de Souza
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Department of Immunology and Molecular Pathology, University College London Medical School, London, United Kingdom
| | - Seen-Wai Lavelle
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Emily Gwyer Findlay
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Tovah N. Shaw
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Nico van Rooijen
- Department of Molecular Cell Biology, VU Medical Center, Amsterdam, The Netherlands
| | - Christiaan J. Saris
- Department of Inflammation Research, Amgen, Inc., Thousand Oaks, California, United States of America
| | - Christopher A. Hunter
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Eleanor M. Riley
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Kevin N. Couper
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- * E-mail:
| |
Collapse
|
132
|
Abeles SR, Chuquiyauri R, Tong C, Vinetz JM. Human host-derived cytokines associated with Plasmodium vivax transmission from acute malaria patients to Anopheles darlingi mosquitoes in the Peruvian Amazon. Am J Trop Med Hyg 2013; 88:1130-7. [PMID: 23478585 DOI: 10.4269/ajtmh.12-0752] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Infection of mosquitoes by humans is not always successful in the setting of patent gametocytemia. This study tested the hypothesis that pro- or anti-inflammatory cytokines are associated with transmission of Plasmodium vivax to Anopheles darlingi mosquitoes in experimental infection. Blood from adults with acute, non-severe P. vivax malaria was fed to laboratory-reared F1 An. darlingi mosquitoes. A panel of cytokines at the time of mosquito infection was assessed in patient sera and levels compared among subjects who did and did not infect mosquitoes. Overall, blood from 43 of 99 (43%) subjects led to mosquito infection as shown by oocyst counts. Levels of IL-10, IL-6, TNF-α, and IFN-γ were significantly elevated in vivax infection and normalized 3 weeks later. The anti-inflammatory cytokine IL-10 was significantly higher in nontransmitters compared with top transmitters but was not in TNF-α and IFN-γ. The IL-10 elevation during acute malaria was associated with P. vivax transmission blocking.
Collapse
|
133
|
Kling JC, Körner H. Different regulatory mechanisms in protozoan parasitic infections. Int J Parasitol 2013; 43:417-25. [PMID: 23470812 DOI: 10.1016/j.ijpara.2013.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 02/01/2013] [Accepted: 02/04/2013] [Indexed: 02/07/2023]
Abstract
The immune response to the protozoan pathogens, Leishmania spp., Trypanosoma spp. and Plasmodium spp., has been studied extensively with particular focus on regulation of the immune response by immunological mechanisms. More specifically, in diseases caused by parasites, immunosuppression frequently prevents immunopathology that can injure the host. However, this allows a small number of parasites to evade the immune response and remain in the host after a clinical cure. The consequences can be chronic infections, which establish a zoonotic or anthroponotic reservoir. This review will highlight some of the identified regulatory mechanisms of the immune system that govern immune responses to parasitic diseases, in particular leishmaniasis, trypanosomiasis and malaria, and discuss implications for the development of efficient vaccines against these diseases.
Collapse
Affiliation(s)
- Jessica C Kling
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania 7000, Australia
| | | |
Collapse
|
134
|
Pagán AJ, Peters NC, Debrabant A, Ribeiro-Gomes F, Pepper M, Karp CL, Jenkins MK, Sacks DL. Tracking antigen-specific CD4+ T cells throughout the course of chronic Leishmania major infection in resistant mice. Eur J Immunol 2013; 43:427-38. [PMID: 23109292 PMCID: PMC4086308 DOI: 10.1002/eji.201242715] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 09/20/2012] [Accepted: 10/19/2012] [Indexed: 12/12/2022]
Abstract
Primary Leishmania major infection typically produces cutaneous lesions that not only heal but also harbor persistent parasites. While the opposing roles of CD4(+) T-cell-derived IFN-γ and IL-10 in promoting parasite killing and persistence have been well established, how these responses develop from naïve precursors has not been directly monitored throughout the course of infection. We used peptide:Major Histocompatibility Complex class II (pMHCII) tetramers to investigate the endogenous, parasite-specific primary CD4(+) T-cell response to L. major in mice resistant to infection. Maximal frequencies of IFN-γ(+) CD4(+) T cells were observed in the spleen and infected ears within a month after infection and were maintained into the chronic phase. In contrast, peak frequencies of IL-10(+) CD4(+) T cells emerged within 2 weeks of infection, persisted into the chronic phase, and accumulated in the infected ears but not the spleen, via a process that depended on local antigen presentation. T helper type-1 (Th1) cells, not Foxp3(+) regulatory T cells, were the chief producers of IL-10 and were not exhausted. Therefore, tracking antigen-specific CD4(+) T cells revealed that IL-10 production by Th1 cells is not due to persistent T-cell antigen receptor stimulation, but rather driven by early antigen encounter at the site of infection.
Collapse
Affiliation(s)
- Antonio J Pagán
- Department of Microbiology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, USA
| | | | | | | | | | | | | | | |
Collapse
|
135
|
Abanyie FA, McCracken C, Kirwan P, Molloy SF, Asaolu SO, Holland CV, Gutman J, Lamb TJ. Ascaris co-infection does not alter malaria-induced anaemia in a cohort of Nigerian preschool children. Malar J 2013; 12:1. [PMID: 23282136 PMCID: PMC3544581 DOI: 10.1186/1475-2875-12-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 12/26/2012] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Co-infection with malaria and intestinal parasites such as Ascaris lumbricoides is common. Malaria parasites induce a pro-inflammatory immune response that contributes to the pathogenic sequelae, such as malarial anaemia, that occur in malaria infection. Ascaris is known to create an anti-inflammatory immune environment which could, in theory, counteract the anti-malarial inflammatory immune response, minimizing the severity of malarial anaemia. This study examined whether Ascaris co-infection can minimize the severity of malarial anaemia. METHODS Data from a randomized controlled trial on the effect of antihelminthic treatment in Nigerian preschool-aged (6-59 months) children conducted in 2006-2007 were analysed to examine the effect of malaria and Ascaris co-infection on anaemia severity. Children were enrolled and tested for malaria, helminths and anaemia at baseline, four, and eight months. Six hundred and ninety subjects were analysed in this study. Generalized linear mixed models were used to assess the relationship between infection status and Ascaris and Plasmodium parasite intensity on severity of anaemia, defined as a haemoglobin less than 11 g/dL. RESULTS Malaria prevalence ranged from 35-78% over the course of this study. Of the malaria-infected children, 55% were co-infected with Ascaris at baseline, 60% were co-infected four months later and 48% were co-infected eight months later, underlining the persistent prevalence of malaria-nematode co-infections in this population. Over the course of the study the percentage of anaemic subjects in the population ranged between 84% at baseline and 77% at the eight-month time point. The odds of being anaemic were four to five times higher in children infected with malaria compared to those without malaria. Ascaris infection alone did not increase the odds of being anaemic, indicating that malaria was the main cause of anaemia in this population. There was no significant difference in the severity of anaemia between children singly infected with malaria and co-infected with malaria and Ascaris. CONCLUSION In this cohort of Nigerian preschool children, malaria infection was the major contributor to anaemia status. Ascaris co-infection neither exacerbated nor ameliorated the severity of malarial anaemia.
Collapse
|
136
|
Luckey U, Schmidt T, Pfender N, Romer M, Lorenz N, Martin SF, Bopp T, Schmitt E, Nikolaev A, Yogev N, Waisman A, Jakob T, Steinbrink K. Crosstalk of regulatory T cells and tolerogenic dendritic cells prevents contact allergy in subjects with low zone tolerance. J Allergy Clin Immunol 2012; 130:781-797.e11. [PMID: 22935591 DOI: 10.1016/j.jaci.2012.06.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 06/16/2012] [Accepted: 06/19/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND Allergic contact dermatitis is one of the most common occupational diseases. A main protective mechanism in those who do not develop allergic contact dermatitis is tolerance induction by repeated exposure to low doses of contact allergen, which is termed low zone tolerance (LZT). The mechanisms that determine the tolerance induction in subjects with LZT are still elusive. OBJECTIVE We performed analysis of the role of CD4(+)CD25(+) forkhead box protein 3 (FOXP3)-positive regulatory T (Treg) cells and dendritic cells (DCs) in mice with LZT. METHODS Mechanisms of tolerance induction were analyzed in a murine model of LZT by using FOXP3 and IL-10 reporter mice, as well as mice that allow the selective depletion of Treg cells or DCs. RESULTS Depletion of CD4(+)CD25(+)FOXP3(+) Treg cells during tolerance induction completely abolishes the development of LZT, resulting in a pronounced contact hypersensitivity response. Adoptive transfer experiments, depletion studies, and use of cell type-specific deficient mice revealed that IL-10 production is critical for the suppressor function of Treg cells in mice with LZT and that tolerogenic CD8(+)CD11c(+) DCs located in the skin-draining lymph nodes are essential for LZT. In the absence of Treg cells, DCs did not develop tolerogenic functions, indicating that activated IL-10(+) Treg cells might imprint the tolerogenic DC phenotype. Cell communication analysis revealed that the education of tolerogenic DCs might involve a direct interaction with Treg cells mediated by gap junctions. Subsequently, induction of tolerogenic CD11c(+) DCs leads to the generation of hapten-specific CD8(+) Treg cells, which protect against contact hypersensitivity. CONCLUSIONS Our data demonstrate critical interactions between CD4(+)CD25(+)FOXP3(+) Treg cells and tolerogenic CD8(+)CD11c(+) DCs during the induction of LZT.
Collapse
Affiliation(s)
- Ulrike Luckey
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University of Mainz, Mainz, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Manjurano A, Clark TG, Nadjm B, Mtove G, Wangai H, Sepulveda N, Campino SG, Maxwell C, Olomi R, Rockett KR, Jeffreys A, MalariaGen Consortium, Riley EM, Reyburn H, Drakeley C. Candidate human genetic polymorphisms and severe malaria in a Tanzanian population. PLoS One 2012; 7:e47463. [PMID: 23144702 PMCID: PMC3483265 DOI: 10.1371/journal.pone.0047463] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 09/12/2012] [Indexed: 11/18/2022] Open
Abstract
Human genetic background strongly influences susceptibility to malaria infection and progression to severe disease and death. Classical genetic studies identified haemoglobinopathies and erythrocyte-associated polymorphisms, as protective against severe disease. High throughput genotyping by mass spectrometry allows multiple single nucleotide polymorphisms (SNPs) to be examined simultaneously. We compared the prevalence of 65 human SNP's, previously associated with altered risk of malaria, between Tanzanian children with and without severe malaria. Five hundred children, aged 1–10 years, with severe malaria were recruited from those admitted to hospital in Muheza, Tanzania and compared with matched controls. Genotyping was performed by Sequenom MassArray, and conventional PCR was used to detect deletions in the alpha-thalassaemia gene. SNPs in two X-linked genes were associated with altered risk of severe malaria in females but not in males: heterozygosity for one or other of two SNPs in the G6PD gene was associated with protection from all forms of severe disease whilst two SNPs in the gene encoding CD40L were associated with respiratory distress. A SNP in the adenyl cyclase 9 (ADCY9) gene was associated with protection from acidosis whilst a polymorphism in the IL-1α gene (IL1A) was associated with an increased risk of acidosis. SNPs in the genes encoding IL-13 and reticulon-3 (RTN3) were associated with increased risk of cerebral malaria. This study confirms previously known genetic associations with protection from severe malaria (HbS, G6PD). It identifies two X-linked genes associated with altered risk of severe malaria in females, identifies mutations in ADCY9, IL1A and CD40L as being associated with altered risk of severe respiratory distress and acidosis, both of which are characterised by high serum lactate levels, and also identifies novel genetic associations with severe malaria (TRIM5) and cerebral malaria(IL-13 and RTN3). Further studies are required to test the generality of these associations and to understand their functional consequences.
Collapse
Affiliation(s)
- Alphaxard Manjurano
- Joint Malaria Programme, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Taane G. Clark
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Behzad Nadjm
- Joint Malaria Programme, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - George Mtove
- Joint Malaria Programme, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Hannah Wangai
- Joint Malaria Programme, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Nuno Sepulveda
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | - Caroline Maxwell
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Raimos Olomi
- Joint Malaria Programme, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Kirk R. Rockett
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Anna Jeffreys
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | | | - Eleanor M. Riley
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Hugh Reyburn
- Joint Malaria Programme, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Christopher Drakeley
- Joint Malaria Programme, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- * E-mail:
| |
Collapse
|
138
|
Natural transmission of Plasmodium berghei exacerbates chronic tuberculosis in an experimental co-infection model. PLoS One 2012; 7:e48110. [PMID: 23110184 PMCID: PMC3482195 DOI: 10.1371/journal.pone.0048110] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 09/19/2012] [Indexed: 01/23/2023] Open
Abstract
Human populations are rarely exposed to one pathogen alone. Particularly in high incidence regions such as sub-Saharan Africa, concurrent infections with more than one pathogen represent a widely underappreciated public health problem. Two of the world’s most notorious killers, malaria and tuberculosis, are co-endemic in impoverished populations in the tropics. However, interactions between both infections in a co-infected individual have not been studied in detail. Both pathogens have a major impact on the lung as the prime target organ for aerogenic Mycobacterium tuberculosis and the site for one of the main complications in severe malaria, malaria-associated acute respiratory distress syndrome (MA-ARDS). In order to study the ramifications caused by both infections within the same host we established an experimental mouse model of co-infection between Mycobacterium tuberculosis and Plasmodium berghei NK65, a recently described model for MA-ARDS. Our study provides evidence that malaria-induced immune responses impair host resistance to Mycobacterium tuberculosis. Using the natural routes of infection, we observed that co-infection exacerbated chronic tuberculosis while rendering mice less refractory to Plasmodium. Co-infected animals presented with enhanced inflammatory immune responses as reflected by exacerbated leukocyte infiltrates, tissue pathology and hypercytokinemia accompanied by altered T-cell responses. Our results - demonstrating striking changes in the immune regulation by co-infection with Plasmodium and Mycobacterium - are highly relevant for the medical management of both infections in humans.
Collapse
|
139
|
Das A, Ellis G, Pallant C, Lopes AR, Khanna P, Peppa D, Chen A, Blair P, Dusheiko G, Gill U, Kennedy PT, Brunetto M, Lampertico P, Mauri C, Maini MK. IL-10-producing regulatory B cells in the pathogenesis of chronic hepatitis B virus infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2012; 189:3925-35. [PMID: 22972930 PMCID: PMC3480715 DOI: 10.4049/jimmunol.1103139] [Citation(s) in RCA: 280] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A regulatory subset of B cells has been found to modulate immune responses in autoimmunity, infection, and cancer, but it has not been investigated in the setting of human persistent viral infection. IL-10 is elevated in patients with chronic hepatitis B virus infection (CHB), but its cellular sources and impact on antiviral T cells have not been addressed. We investigated the role of IL-10 and regulatory B cells in the pathogenesis of CHB. Serum IL-10 levels were studied longitudinally in patients with CHB undergoing spontaneous disease flares. There was a close temporal correlation between IL-10 levels and fluctuations in viral load or liver inflammation. Blockade of IL-10 in vitro rescued polyfunctional virus-specific CD8 T cell responses. To investigate the potential contribution of regulatory B cells, their frequency was measured directly ex vivo and after exposure to stimuli relevant to hepatitis B virus (HBV) (CpG or HBV Ags). IL-10-producing B cells were enriched in patients, and their frequency correlated temporally with hepatic flares, both after stimulation and directly ex vivo. Phenotypically, these cells were predominantly immature (CD19(+)CD24(hi)CD38(hi)) ex vivo; sorted CD19(+)CD24(hi)CD38(hi) cells suppressed HBV-specific CD8 T cell responses in an IL-10-dependent manner. In summary, these data reveal a novel IL-10-producing subset of B cells able to regulate T cell immunity in CHB.
Collapse
Affiliation(s)
- Abhishek Das
- Division of Infection and Immunity, University College London, London WC1E 6JF, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Cytokine balance in human malaria: does Plasmodium vivax elicit more inflammatory responses than Plasmodium falciparum? PLoS One 2012; 7:e44394. [PMID: 22973442 PMCID: PMC3433413 DOI: 10.1371/journal.pone.0044394] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 08/06/2012] [Indexed: 02/08/2023] Open
Abstract
Background The mechanisms by which humans regulate pro- and anti-inflammatory responses on exposure to different malaria parasites remains unclear. Although Plasmodium vivax usually causes a relatively benign disease, this parasite has been suggested to elicit more host inflammation per parasitized red blood cell than P. falciparum. Methodology/Principal Findings We measured plasma concentrations of seven cytokines and two soluble tumor necrosis factor (TNF)-α receptors, and evaluated clinical and laboratory outcomes, in Brazilians with acute uncomplicated infections with P. vivax (n = 85), P. falciparum (n = 30), or both species (n = 12), and in 45 asymptomatic carriers of low-density P. vivax infection. Symptomatic vivax malaria patients, compared to those infected with P. falciparum or both species, had more intense paroxysms, but they had no clear association with a pro-inflammatory imbalance. To the contrary, these patients had higher levels of the regulatory cytokine interleukin (IL)-10, which correlated positively with parasite density, and elevated IL-10/TNF-α, IL-10/interferon (IFN)-γ, IL-10/IL-6 and sTNFRII/TNF-α ratios, compared to falciparum or mixed-species malaria patient groups. Vivax malaria patients had the highest levels of circulating soluble TNF-α receptor sTNFRII. Levels of regulatory cytokines returned to normal values 28 days after P. vivax clearance following chemotherapy. Finally, asymptomatic carriers of low P. vivax parasitemias had substantially lower levels of both inflammatory and regulatory cytokines than did patients with clinical malaria due to either species. Conclusions Controlling fast-multiplying P. falciparum blood stages requires a strong inflammatory response to prevent fulminant infections, while reducing inflammation-related tissue damage with early regulatory cytokine responses may be a more cost-effective strategy in infections with the less virulent P. vivax parasite. The early induction of regulatory cytokines may be a critical mechanism protecting vivax malaria patients from severe clinical complications.
Collapse
|
141
|
Toxoplasma gondii infection induces suppression in a mouse model of allergic airway inflammation. PLoS One 2012; 7:e43420. [PMID: 22952678 PMCID: PMC3429463 DOI: 10.1371/journal.pone.0043420] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 07/20/2012] [Indexed: 11/19/2022] Open
Abstract
Allergic asthma is an inflammatory disorder characterized by infiltration of the airway wall with inflammatory cells driven mostly by activation of Th2-lymphocytes, eosinophils and mast cells. There is a link between increased allergy and a reduction of some infections in Western countries. Epidemiological data also show that respiratory allergy is less frequent in people exposed to orofecal and foodborne microbes such as Toxoplasma gondii. We previously showed that both acute and chronic parasite T. gondii infection substantially blocked development of airway inflammation in adult BALB/c mice. Based on the high levels of IFN-γ along with the reduction of Th2 phenotype, we hypothesized that the protective effect might be related to the strong Th1 immune response elicited against the parasite. However, other mechanisms could also be implicated. The possibility that regulatory T cells inhibit allergic diseases has received growing support from both animal and human studies. Here we investigated the cellular mechanisms involved in T. gondii induced protection against allergy. Our results show for the first time that thoracic lymph node cells from mice sensitized during chronic T. gondii infection have suppressor activity. Suppression was detected both in vitro, on allergen specific T cell proliferation and in vivo, on allergic lung inflammation after adoptive transference from infected/sensitized mice to previously sensitized animals. This ability was found to be contact- independent and correlated with high levels of TGF-β and CD4+FoxP3+ cells.
Collapse
|
142
|
Boeuf PS, Loizon S, Awandare GA, Tetteh JKA, Addae MM, Adjei GO, Goka B, Kurtzhals JAL, Puijalon O, Hviid L, Akanmori BD, Behr C. Insights into deregulated TNF and IL-10 production in malaria: implications for understanding severe malarial anaemia. Malar J 2012; 11:253. [PMID: 22853732 PMCID: PMC3469355 DOI: 10.1186/1475-2875-11-253] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 07/25/2012] [Indexed: 01/09/2023] Open
Abstract
Background Severe malarial anaemia (SMA) is a major life-threatening complication of paediatric malaria. Protracted production of pro-inflammatory cytokines promoting erythrophagocytosis and depressing erythropoiesis is thought to play an important role in SMA, which is characterized by a high TNF/IL-10 ratio. Whether this TNF/IL-10 imbalance results from an intrinsic incapacity of SMA patients to produce IL-10 or from an IL-10 unresponsiveness to infection is unknown. Monocytes and T cells are recognized as the main sources of TNF and IL-10 in vivo, but little is known about the activation status of those cells in SMA patients. Methods The IL-10 and TNF production capacity and the activation phenotype of monocytes and T cells were compared in samples collected from 332 Ghanaian children with non-overlapping SMA (n = 108), cerebral malaria (CM) (n = 144) or uncomplicated malaria (UM) (n = 80) syndromes. Activation status of monocytes and T cells was ascertained by measuring HLA-DR+ and/or CD69+ surface expression by flow cytometry. The TNF and IL-10 production was assessed in a whole-blood assay after or not stimulation with lipopolysaccharide (LPS) or phytohaemaglutinin (PHA) used as surrogate of unspecific monocyte and T cell stimulant. The number of circulating pigmented monocytes was also determined. Results Monocytes and T cells from SMA and CM patients showed similar activation profiles with a comparable decreased HLA-DR expression on monocytes and increased frequency of CD69+ and HLA-DR+ T cells. In contrast, the acute-phase IL-10 production was markedly decreased in SMA compared to CM (P = .003) and UM (P = .004). Although in SMA the IL-10 response to LPS-stimulation was larger in amplitude than in CM (P = .0082), the absolute levels of IL-10 reached were lower (P = .013). Both the amplitude and levels of TNF produced in response to LPS-stimulation were larger in SMA than CM (P = .019). In response to PHA-stimulation, absolute levels of IL-10 produced in SMA were lower than in CM (P = .005) contrasting with TNF levels, which were higher (P = .001). Conclusions These data reveal that SMA patients have the potential to mount efficient IL-10 responses and that the TNF/IL-10 imbalance may reflect a specific monocyte and T cell programming/polarization pattern in response to infection.
Collapse
Affiliation(s)
- Philippe S Boeuf
- Institut Pasteur, Unité d'Immunologie Moléculaire des Parasites URA CNRS 2581, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Owens BMJ, Beattie L, Moore JWJ, Brown N, Mann JL, Dalton JE, Maroof A, Kaye PM. IL-10-producing Th1 cells and disease progression are regulated by distinct CD11c⁺ cell populations during visceral leishmaniasis. PLoS Pathog 2012; 8:e1002827. [PMID: 22911108 PMCID: PMC3406093 DOI: 10.1371/journal.ppat.1002827] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 06/13/2012] [Indexed: 12/20/2022] Open
Abstract
IL-10 is a critical regulatory cytokine involved in the pathogenesis of visceral leishmaniasis caused by Leishmania donovani and clinical and experimental data indicate that disease progression is associated with expanded numbers of CD4⁺ IFNγ⁺ T cells committed to IL-10 production. Here, combining conditional cell-specific depletion with adoptive transfer, we demonstrate that only conventional CD11c(hi) DCs that produce both IL-10 and IL-27 are capable of inducing IL-10-producing Th1 cells in vivo. In contrast, CD11c(hi) as well as CD11c(int/lo) cells isolated from infected mice were capable of reversing the host protective effect of diphtheria toxin-mediated CD11c⁺ cell depletion. This was reflected by increased splenomegaly, inhibition of NO production and increased parasite burden. Thus during chronic infection, multiple CD11c⁺ cell populations can actively suppress host resistance and enhance immunopathology, through mechanisms that do not necessarily involve IL-10-producing Th1 cells.
Collapse
Affiliation(s)
| | - Lynette Beattie
- Centre for Immunology & Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - John W. J. Moore
- Centre for Immunology & Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Najmeeyah Brown
- Centre for Immunology & Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Jason L. Mann
- Centre for Immunology & Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Jane E. Dalton
- Centre for Immunology & Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Asher Maroof
- Centre for Immunology & Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Paul M. Kaye
- Centre for Immunology & Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
- * E-mail:
| |
Collapse
|
144
|
Carrier Y, Whitters MJ, Miyashiro JS, LaBranche TP, Ramon HE, Benoit SE, Ryan MS, Keegan SP, Guay H, Douhan J, Collins M, Dunussi-Joannopoulos K, Medley QG. Enhanced GITR/GITRL interactions augment IL-27 expression and induce IL-10-producing Tr-1 like cells. Eur J Immunol 2012; 42:1393-404. [DOI: 10.1002/eji.201142162] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
| | | | | | | | | | | | - Mark S. Ryan
- Inflammation and Immunology; Pfizer; Cambridge; MA; USA
| | | | - Heath Guay
- Inflammation and Immunology; Pfizer; Cambridge; MA; USA
| | - John Douhan
- Inflammation and Immunology; Pfizer; Cambridge; MA; USA
| | - Mary Collins
- Inflammation and Immunology; Pfizer; Cambridge; MA; USA
| | | | | |
Collapse
|
145
|
Abel S, Lückheide N, Westendorf AM, Geffers R, Roers A, Müller W, Sparwasser T, Matuschewski K, Buer J, Hansen W. Strong impact of CD4+ Foxp3+ regulatory T cells and limited effect of T cell-derived IL-10 on pathogen clearance during Plasmodium yoelii infection. THE JOURNAL OF IMMUNOLOGY 2012; 188:5467-77. [PMID: 22544931 DOI: 10.4049/jimmunol.1102223] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It is well established that CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs) play a crucial role in the course of different infectious diseases. However, contradictory results have been published regarding to malaria infection. In this study, we report that specific ablation of Foxp3(+) Tregs in Plasmodium yoelii-infected DEREG-BALB/c mice leads to an increase in T cell activation accompanied by a significant decrease in parasitemia. To better understand how Foxp3(+) Tregs orchestrate this phenotype, we used microarrays to analyze CD4(+)CD25(+)Foxp3(+) Tregs and CD4(+)CD25(-)Foxp3(-) T cells in the course of P. yoelii infection. Using this approach we identified genes specifically upregulated in CD4(+)CD25(+)Foxp3(+) Tregs in the course of infection, such as G-protein-coupled receptor 83 and Socs2. This analysis also revealed that both CD4(+)CD25(+)Foxp3(+) Tregs and CD4(+)CD25(-)Foxp3(-) T cells upregulate CTLA-4, granzyme B, and, more strikingly, IL-10 during acute blood infection. Therefore, we aimed to define the function of T cell-derived IL-10 in this context by Cre/loxP-mediated selective conditional inactivation of the IL-10 gene in T cells. Unexpectedly, IL-10 ablation in T cells exerts only a minor effect on parasite clearance, even though CD8(+) T cells are more strongly activated, the production of IFN-γ and TNF-α by CD4(+)CD25(-) T cells is increased, and the suppressive activity of CD4(+)CD25(+) Tregs is reduced upon infection. In summary, these results suggest that CD4(+)Foxp3(+) Tregs modulate the course of P. yoelii infection in BALB/c mice. Moreover, CD4(+) T cell-derived IL-10 affects T effector function and Treg activity, but has only a limited direct effect on parasite clearance in this model.
Collapse
Affiliation(s)
- Simone Abel
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, 45122 Essen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
146
|
Freitas do Rosario AP, Langhorne J. T cell-derived IL-10 and its impact on the regulation of host responses during malaria. Int J Parasitol 2012; 42:549-55. [PMID: 22549022 DOI: 10.1016/j.ijpara.2012.03.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2011] [Revised: 03/02/2012] [Accepted: 03/24/2012] [Indexed: 02/04/2023]
Abstract
Despite intense research, malaria still is the one of the most devastating diseases killing more people than any other parasitic infection. In an attempt to control the infection, the host immune system produces a potent pro-inflammatory response. However, this response is also associated with complications, such as severe anaemia, hypoglycaemia and cerebral malaria. This pronounced production of pro-inflammatory cytokines response is a common feature of malaria caused by parasites infecting humans as well as rodents and primates. A balance between pro- and anti-inflammatory responses may be fundamental to the elimination of the parasite without inducing excessive host pathology. IL-10 is a key cytokine that has been shown to have an important regulatory function in establishing this balance in malaria. Here we discuss which cells can produce IL-10 during infection, and present an overview of the evidence showing that T-cell derived IL-10 plays an important role in regulating malaria pathology. Many different subsets of T cells can produce IL-10, however, evidence is accumulating that it is effector Th1 CD4(+) T cells which provide the crucial source that down-regulates inflammatory pathology during blood-stage malaria infections.
Collapse
|
147
|
Braga M, Schiavone C, Di Gioacchino G, De Angelis I, Cavallucci E, Lazzarin F, Petrarca C, Di Gioacchino M. Environment and T regulatory cells in allergy. THE SCIENCE OF THE TOTAL ENVIRONMENT 2012; 423:193-201. [PMID: 20825978 DOI: 10.1016/j.scitotenv.2010.08.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Revised: 08/09/2010] [Accepted: 08/09/2010] [Indexed: 05/29/2023]
Abstract
The central role of T regulatory cells in the responses against harmless environmental antigens has been confirmed by many studies. Impaired T regulatory cell function is implicated in many pathological conditions, particularly allergic diseases. The "hygiene hypothesis" suggests that infections and infestations may play a protective role for allergy, whereas environmental pollutants favor the development of allergic diseases. Developing countries suffer from a variety of infections and are also facing an increasing diffusion of environmental pollutants. In these countries allergies increase in relation to the spreading use of xenobiotics (pesticides, herbicides, pollution, etc.) with a rate similar to those of developed countries, overcoming the protective effects of infections. We review here the main mechanisms of non-self tolerance, with particular regard to relations between T regulatory cell activity, infections and infestations such as helminthiasis, and exposure to environmental xenobiotics with relevant diffusion in developing countries.
Collapse
Affiliation(s)
- M Braga
- Allergy Unit, Spedali Civili, Piazzale Spedali Civili, 25123 Brescia, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
148
|
CD4+ CD25+ regulatory T cells impair HIV-1-specific CD4 T cell responses by upregulating interleukin-10 production in monocytes. J Virol 2012; 86:6586-94. [PMID: 22496237 DOI: 10.1128/jvi.06251-11] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
T cell dysfunction in the presence of ongoing antigen exposure is a cardinal feature of chronic viral infections with persistent high viremia, including HIV-1. Although interleukin-10 (IL-10) has been implicated as an important mediator of this T cell dysfunction, the regulation of IL-10 production in chronic HIV-1 infection remains poorly understood. We demonstrated that IL-10 is elevated in the plasma of individuals with chronic HIV-1 infection and that blockade of IL-10 signaling results in a restoration of HIV-1-specific CD4 T cell proliferation, gamma interferon (IFN-γ) secretion, and, to a lesser extent, IL-2 production. Whereas IL-10 blockade leads to restoration of IFN-γ secretion by HIV-1-specific CD4 T cells in all categories of subjects investigated, significant enhancement of IL-2 production and improved proliferation of CD4 T helper cells are restricted to viremic individuals. In peripheral blood mononuclear cells (PBMCs), this IL-10 is produced primarily by CD14(+) monocytes, but its production is tightly controlled by regulatory T cells (Tregs), which produce little IL-10 directly. When Tregs are depleted from PBMCs of viremic individuals, the effect of the IL-10 signaling blockade is abolished and IL-10 production by monocytes decreases, while the production of proinflammatory cytokines, such as tumor necrosis factor alpha (TNF-α), increases. The regulation of IL-10 by Tregs appears to be mediated primarily by contact or paracrine-dependent mechanisms which involve IL-27. This work describes a novel mechanism by which regulatory T cells control IL-10 production and contribute to dysfunctional HIV-1-specific CD4 T cell help in chronic HIV-1 infection and provides a unique mechanistic insight into the role of regulatory T cells in immune exhaustion.
Collapse
|
149
|
Kakita N, Kanto T, Itose I, Kuroda S, Inoue M, Matsubara T, Higashitani K, Miyazaki M, Sakakibara M, Hiramatsu N, Takehara T, Kasahara A, Hayashi N. Comparative analyses of regulatory T cell subsets in patients with hepatocellular carcinoma: a crucial role of CD25(-) FOXP3(-) T cells. Int J Cancer 2012; 131:2573-83. [PMID: 22419479 DOI: 10.1002/ijc.27535] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Accepted: 02/28/2012] [Indexed: 12/31/2022]
Abstract
Regulatory T cells (Tregs) play pivotal role in cancer-induced immunoediting. Increment of CD25(high+) FOXP3+ natural Tregs has been reported in patients with hepatocellular carcinoma (HCC); however, the involvement of other type of Tregs remain elusive. We aimed to clarify whether FOXP3- Tregs are increased and functionally suppressive or not in patients with HCC. We enrolled 184 hepatitis C-infected patients with chronic liver diseases or HCC, 57 healthy subjects and 27 HCC patients with other etiology. Distinct Treg subsets were phenotypically identified by the expression of CD4, CD25, CD127 and forkhead/winged helix transcription factor (FOXP3). Their gene profiles, frequency and suppressor functions against T cell proliferation were compared among the subjects. To examine the molecules involving in Treg differentiation, we cultured naive CD4+ T cells in the presence of HCC cells and dendritic cells. We determined two types of CD4+ CD127- T cells with comparable regulatory ability; one is CD25(high+) cells expressing FOXP3 (CD25(high+) FOXP3+ Tregs) and the other is CD25- cells without FOXP3- expression (CD25- FOXP3- cells). The peripheral or intrahepatic frequency of CD25- FOXP3- Tregs in HCC patients is higher than those in other groups, of which significance is more than CD25(high+) FOXP3+ cells. Of importance, CD25- FOXP3- Tregs, but not CD25(high+) FOXP3+ cells, dynamically change in patients accompanied by the ablation or the recurrence of HCC. CD25- FOXP3- T cells with CD127- IL-10+ phenoype are inducible in vitro from naive CD4(+) T cells, in which programmed cell death 1 ligand 1, immunoglobulin-like transcript 4 and human leukocyte antigen G are involved.. In conclusion, CD25- FOXP3- Tregs with suppressive capacity are increased in patients with HCC, suggesting their distinct roles from CD25+ FOXP3+ Tregs.
Collapse
Affiliation(s)
- Naruyasu Kakita
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Suita, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Iyer SS, Cheng G. Role of interleukin 10 transcriptional regulation in inflammation and autoimmune disease. Crit Rev Immunol 2012; 32:23-63. [PMID: 22428854 DOI: 10.1615/critrevimmunol.v32.i1.30] [Citation(s) in RCA: 1033] [Impact Index Per Article: 79.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Interleukin 10 (IL-10) is a cytokine with potent anti-inflammatory properties that plays a central role in limiting host immune response to pathogens, thereby preventing damage to the host and maintaining normal tissue homeostasis. Dysregulation of IL-10 is associated with enhanced immunopathology in response to infection as well as increased risk for development of many autoimmune diseases. Thus a fundamental understanding of IL-10 gene expression is critical for our comprehension of disease progression and resolution of host inflammatory response. In this review, we discuss modes of regulation of IL-10 gene expression in immune effector cell types, including signal transduction, epigenetics, promoter architecture, and post-transcriptional regulation, and how aberrant regulation contributes to immunopathology and disease progression.
Collapse
|