1
|
Warunek J, Jin RM, Blair SJ, Garis M, Marzullo B, Wohlfert EA. Tbet Expression by Regulatory T Cells Is Needed to Protect against Th1-Mediated Immunopathology during Toxoplasma Infection in Mice. Immunohorizons 2021; 5:931-943. [PMID: 34893511 DOI: 10.4049/immunohorizons.2100080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 11/14/2021] [Indexed: 11/19/2022] Open
Abstract
Toxoplasma gondii infection has proven to be an ideal model to understand the delicate balance between protective immunity and immune-mediated pathology during infection. Lethal infection causes a collapse of T regulatory cells (Tregs) mediated by the loss of IL-2 and conversion of Tregs to IFN-γ-producing cells. Importantly, these Tregs highly express the Th1 transcription factor Tbet. To determine the role of Tbet in Tregs, we infected Tbx21f/f -Foxp3YFPCre and control Foxp3YFPCre mice with the type II strain of T. gondii, ME49. The majority of Tbx21f/f -Foxp3YFPCre mice succumbed to a nonlethal dose. Notably, parasite burden was reduced in Tbx21f/f -Foxp3YFPCre compared with Foxp3YFPCre control mice. We found that Tbx21f/f -Foxp3YFPCre mice have significantly higher serum levels of proinflammatory cytokines IFN-γ and TNF-α, suggestive of a heightened immune response. To test if CD4+ T cells were driving immunopathology, we treated Tbx21f/f -Foxp3YFPCre mice with anti-CD4-depleting Abs and partially rescued these mice. Broad-spectrum antibiotic treatment also improved survival, demonstrating a role for commensal flora in immunopathology in Tbx21f/f -Foxp3YFPCre mice. RNA sequencing analysis reinforced that Tbet regulates several key cellular pathways, including leukocyte activation, regulation of lymphocyte activation, and cell cycle progression, that help to maintain fitness in Tregs during Th1 responses. Taken together, our data show an important role for Tbet in Tregs in preventing lethal immunopathology during T. gondii infection, further highlighting the protective role of Treg plasticity in controlling immune responses to infection and the microbiota.
Collapse
Affiliation(s)
- Jordan Warunek
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Amherst, NY; and
| | - Richard M Jin
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Amherst, NY; and
| | - Sarah J Blair
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Amherst, NY; and
| | - Matthew Garis
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Amherst, NY; and
| | - Brandon Marzullo
- Genomics and Bioinformatics Core, State University of New York at Buffalo, Amherst, NY
| | - Elizabeth A Wohlfert
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Amherst, NY; and
| |
Collapse
|
2
|
Immunobiotic and Paraprobiotic Potential Effect of Lactobacillus casei in a Systemic Toxoplasmosis Murine Model. Microorganisms 2020; 8:microorganisms8010113. [PMID: 31947510 PMCID: PMC7023318 DOI: 10.3390/microorganisms8010113] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/20/2022] Open
Abstract
One of the main characteristics of probiotics is their ability to stimulate and modulate the immune response regardless of their viability. Lactobacillus casei (Lc) can stimulate local and systemic immunity, in addition to the activation of macrophages at sites distant from the intestine. Activated macrophages limit the replication of intracellular protozoa, such as Toxoplasma gondii, through the production of nitric oxide. The present study aimed to evaluate the protection generated by treatment with viable and non-viable Lc in the murine systemic toxoplasmosis model. CD1 male mice were treated with viable Lc (immunobiotic) and non-viable Lc (paraprobiotic), infected with tachyzoites of Toxoplasma gondii RH strain. The reduction of the parasitic load, activation of peritoneal macrophages, inflammatory cytokines, and cell populations was evaluated at 7 days post-infection, in addition to the survival. The immunobiotic and paraprobiotic reduced the parasitic load, but only the immunobiotic increased the activation of peritoneal macrophages, and the production of interferon-gamma (IFN-γ), tumor necrosis factor (TNF), and interleukin-6 (IL-6) while the paraprobiotic increased the production of monocyte chemoattractant protein-1 (MCP-1) and T CD4+CD44+ lymphocytes. Viable and non-viable Lc increases survival but does not prevent the death of animals. The results provide evidence about the remote immunological stimulation of viable and non-viable Lc in an in vivo parasitic model.
Collapse
|
3
|
Pandiyan P, Bhaskaran N, Zou M, Schneider E, Jayaraman S, Huehn J. Microbiome Dependent Regulation of T regs and Th17 Cells in Mucosa. Front Immunol 2019; 10:426. [PMID: 30906299 PMCID: PMC6419713 DOI: 10.3389/fimmu.2019.00426] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 02/18/2019] [Indexed: 12/19/2022] Open
Abstract
Mammals co-exist with resident microbial ecosystem that is composed of an incredible number and diversity of bacteria, viruses and fungi. Owing to direct contact between resident microbes and mucosal surfaces, both parties are in continuous and complex interactions resulting in important functional consequences. These interactions govern immune homeostasis, host response to infection, vaccination and cancer, as well as predisposition to metabolic, inflammatory and neurological disorders. Here, we discuss recent studies on direct and indirect effects of resident microbiota on regulatory T cells (Tregs) and Th17 cells at the cellular and molecular level. We review mechanisms by which commensal microbes influence mucosa in the context of bioactive molecules derived from resident bacteria, immune senescence, chronic inflammation and cancer. Lastly, we discuss potential therapeutic applications of microbiota alterations and microbial derivatives, for improving resilience of mucosal immunity and combating immunopathology.
Collapse
Affiliation(s)
- Pushpa Pandiyan
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Natarajan Bhaskaran
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Mangge Zou
- Experimental Immunology, Helmholtz Centre for Infection Research, Hamburg, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Elizabeth Schneider
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Sangeetha Jayaraman
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, Hamburg, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
4
|
Silva-Gutierrez N, Bahsas Zaky R, Bouchard M, Teran Angel G, Amoroso A, Peterson DL, Salmen S. T-cell profiles elicited by Toxoplasma gondii in acutely/chronically infected humans. Parasite Immunol 2018; 40:e12532. [PMID: 29633283 DOI: 10.1111/pim.12532] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 03/19/2018] [Indexed: 11/30/2022]
Abstract
Toxoplasma gondii (T. gondii) is an obligate intracellular protozoan parasite that can infect almost all warm-blooded species and induce a chronic infection in human hosts. The aim of this work was to investigate Th1, Th2, Th17 and Treg polarization, induced by four important T. gondii antigens (SAG1, ROP1, GRA8 and MAG1) in acutely and chronically infected patients. For this purpose, SAG1, ROP1, GRA8 and MAG1 were expressed as recombinant proteins, purified, and used to evaluate the proinflammatory and regulatory immune response profiles in seropositive and seronegative individuals. Our results show that SAG1 and ROP1 elicited a proinflammatory profile (INF-γ, IL-12 and IL-17) in individuals in the acute phase, whereas MAG1 and GRA8 induced a regulatory pattern (Treg and TGF-β) in chronically infected patients. These results reveal fundamental differences in T-cell polarization induced by T. gondii antigens, which could have important implications in the immunopathogenesis of the disease and in future proposals of therapeutic strategies.
Collapse
Affiliation(s)
- N Silva-Gutierrez
- Facultad de Medicina, Instituto de Inmunologia Clinica, Universidad de Los Andes, Mérida, Venezuela
| | - R Bahsas Zaky
- Facultad de Medicina, Instituto de Inmunologia Clinica, Universidad de Los Andes, Mérida, Venezuela
| | - M Bouchard
- Facultad de Medicina, Instituto de Inmunologia Clinica, Universidad de Los Andes, Mérida, Venezuela
| | - G Teran Angel
- Facultad de Medicina, Instituto de Inmunologia Clinica, Universidad de Los Andes, Mérida, Venezuela
| | - A Amoroso
- Facultad de Medicina, Instituto de Inmunologia Clinica, Universidad de Los Andes, Mérida, Venezuela
| | - D L Peterson
- Department of Biochemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - S Salmen
- Facultad de Medicina, Instituto de Inmunologia Clinica, Universidad de Los Andes, Mérida, Venezuela
| |
Collapse
|
5
|
Role of CD4+ Foxp3+ Regulatory T Cells in Protection Induced by a Live Attenuated, Replicating Type I Vaccine Strain of Toxoplasma gondii. Infect Immun 2015; 83:3601-11. [PMID: 26123802 DOI: 10.1128/iai.00217-15] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 06/24/2015] [Indexed: 11/20/2022] Open
Abstract
Vaccination with the live attenuated Toxoplasma gondii Mic1.3KO strain induced long-lasting immunity against challenge with Toxoplasma gondii type I and type II strains. The involvement of regulatory T cells (Tregs) in the protection mechanism was investigated. Intraperitoneal injection of Mic1.3KO induced a weak and transient influx of CD4(+) Foxp3(+) T regulatory cells followed by recruitment/expansion of CD4(+) Foxp3(-) CD25(+) effector cells and control of the parasite at the site of infection. The local and systemic cytokine responses associated with this recruitment of Tregs were of the TH1/Treg-like type. In contrast, injection of RH, the wild-type strain from which the vaccinal strain is derived, induced a low CD4(+) Foxp3(+) cell influx and uncontrolled multiplication of the parasites at this local site, followed by death of the mice. The associated local and systemic cytokine responses were of the TH1/TH17-like type. In addition, in vivo Treg induction in RH-infected mice with interleukin-2 (IL-2)/anti-IL-2 complexes induced control of the parasite and a TH1/Treg cytokine response similar to the response after Mic1.3KO vaccination. These results suggest that Tregs may contribute to the protective response after vaccination with Mic1.3KO.
Collapse
|
6
|
Verhelst D, De Craeye S, Entrican G, Dorny P, Cox E. Parasite distribution and associated immune response during the acute phase of Toxoplasma gondii infection in sheep. BMC Vet Res 2014; 10:293. [PMID: 25511864 PMCID: PMC4279818 DOI: 10.1186/s12917-014-0293-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 12/02/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In many countries, Toxoplasma gondii (T. gondii) is a major cause of reproductive disorders and abortions in the sheep industry, and therefore responsible for important financial and economic losses. In addition, undercooked infected lamb is an important risk factor for human toxoplasmosis. In the present study, the initial phase of the infection was investigated: the parasite's entry site, the subsequent distribution of the parasite and the host-immune response. RESULTS Parasite DNA was already detected in the cranial small intestinal mucosa the first days after oral infection with T. gondii tissue cysts. Simultaneously, high IFN-gamma and IL-12 responses were induced mainly in the mesenteric lymph nodes. The emergence of IgG1 (at 8dpi), and IgG2 (at 11 dpi) was accompanied by a decrease or even disappearance of the IFN-gamma and IL-12 response in the Peyers' patches (PP), PBMC's and popliteal LN's. Meanwhile the parasite DNA could be recovered from most mucosal and systemic tissues to become undetectable in the small intestine, popliteal LN, PBMC and spleen 3 weeks pi. CONCLUSIONS Our results indicate that parasites enter the cranial small intestine the first days after infection and that after an increase the first two weeks after infection, the parasite DNA levels in the intestine drop below the detection limit three weeks after infection. This coincides with an increase in parastic-specific serum IgG1 and IgG2 and a decrease of the antigen-specific IFN-gamma response in PP, PBMC and popliteal LN. We suggest a role for IFN-gamma and IL-12 in controlling the infection.
Collapse
Affiliation(s)
- Delfien Verhelst
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.
| | - Stéphane De Craeye
- National Reference Laboratory for Toxoplasmosis, Operational Direction Communicable and Infectious Diseases, Scientific Institute of Public Health, Brussels, Belgium.
| | - Gary Entrican
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Midlothian, Penicuik, EH26 0PZ, Scotland, UK.
| | - Pierre Dorny
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium. .,Laboratory of Parasitology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.
| | - Eric Cox
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium.
| |
Collapse
|
7
|
Fenoy IM, Sanchez VR, Soto AS, Picchio MS, Maglioco A, Corigliano MG, Dran GI, Martin V, Goldman A. Regulatory cells induced by acute toxoplasmosis prevent the development of allergic lung inflammation. Immunobiology 2014; 220:641-8. [PMID: 25532793 DOI: 10.1016/j.imbio.2014.11.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 11/18/2014] [Accepted: 11/22/2014] [Indexed: 01/31/2023]
Abstract
The increased prevalence of allergies in developed countries has been attributed to a reduction of some infections. Supporting epidemiological studies, we previously showed that both acute and chronic Toxoplasma gondii infection can diminish allergic airway inflammation in BALB/c mice. The mechanisms involved when sensitization occurs during acute phase would be related to the strong Th1 response induced by the parasite. Here, we further investigated the mechanisms involved in T. gondii allergy protection in mice sensitized during acute T. gondii infection. Adoptive transference assays and ex vivo co-cultures experiments showed that not only thoracic lymph node cells from infected and sensitized mice but also from non-sensitized infected animals diminished both allergic lung inflammation and the proliferation of effector T cells from allergic mice. This ability was found to be contact-independent and correlated with high levels of CD4(+)FoxP3(+) cells. IL-10 would not be involved in allergy suppression since IL-10-deficient mice behaved similar to wild type mice. Our results extend earlier work and show that, in addition to immune deviation, acute T. gondii infection can suppress allergic airway inflammation through immune suppression.
Collapse
Affiliation(s)
- Ignacio M Fenoy
- Laboratorio de Inmunología, vacunas y alergia, CESyMA, Escuela de Ciencia y Tecnología, UNSAM, San Martín, Argentina
| | - Vanesa R Sanchez
- Laboratorio de Inmunología, vacunas y alergia, CESyMA, Escuela de Ciencia y Tecnología, UNSAM, San Martín, Argentina
| | - Ariadna S Soto
- Laboratorio de Inmunología, vacunas y alergia, CESyMA, Escuela de Ciencia y Tecnología, UNSAM, San Martín, Argentina
| | - Mariano S Picchio
- Laboratorio de Inmunología, vacunas y alergia, CESyMA, Escuela de Ciencia y Tecnología, UNSAM, San Martín, Argentina
| | - Andrea Maglioco
- Instituto de Medicina Experimental (IMEX-CONICET), Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Mariana G Corigliano
- Laboratorio de Biotecnología Vegetal, IIB-INTECH, CONICET-UNSAM, Chascomús, Provincia de Buenos Aires, Argentina
| | - Graciela I Dran
- Instituto de Medicina Experimental (IMEX-CONICET), Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Valentina Martin
- Laboratorio de Inmunología, vacunas y alergia, CESyMA, Escuela de Ciencia y Tecnología, UNSAM, San Martín, Argentina
| | - Alejandra Goldman
- Laboratorio de Inmunología, vacunas y alergia, CESyMA, Escuela de Ciencia y Tecnología, UNSAM, San Martín, Argentina.
| |
Collapse
|
8
|
Haben I, Hartmann W, Breloer M. Nematode-induced interference with vaccination efficacy targets follicular T helper cell induction and is preserved after termination of infection. PLoS Negl Trop Dis 2014; 8:e3170. [PMID: 25255463 PMCID: PMC4177885 DOI: 10.1371/journal.pntd.0003170] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 08/05/2014] [Indexed: 12/14/2022] Open
Abstract
One-third of the human population is infected with parasitic worms. To avoid being eliminated, these parasites actively dampen the immune response of their hosts. This immune modulation also suppresses immune responses to third-party antigens such as vaccines. Here, we used Litomosoides sigmodontis-infected BALB/c mice to analyse nematode-induced interference with vaccination. Chronic nematode infection led to complete suppression of the humoral response to thymus-dependent vaccination. Thereby the numbers of antigen-specific B cells as well as the serum immunoglobulin (Ig) G titres were reduced. TH2-associated IgG1 and TH1-associated IgG2 responses were both suppressed. Thus, nematode infection did not bias responses towards a TH2 response, but interfered with Ig responses in general. We provide evidence that this suppression indirectly targeted B cells via accessory T cells as number and frequency of vaccine-induced follicular B helper T cells were reduced. Moreover, vaccination using model antigens that stimulate Ig response independently of T helper cells was functional in nematode-infected mice. Using depletion experiments, we show that CD4+Foxp3+ regulatory T cells did not mediate the suppression of Ig response during chronic nematode infection. Suppression was induced by fourth stage larvae, immature adults and mature adults, and increased with the duration of the infection. By contrast, isolated microfilariae increased IgG2a responses to vaccination. This pro-inflammatory effect of microfilariae was overruled by the simultaneous presence of adults. Strikingly, a reduced humoral response was still observed if vaccination was performed more than 16 weeks after termination of L. sigmodontis infection. In summary, our results suggest that vaccination may not only fail in helminth-infected individuals, but also in individuals with a history of previous helminth infections. Parasitic worms, called helminths, infect one-third of the world population. Despite exposure to their host's immune system many helminths establish chronic infections and survive several years within their host. They avoid elimination by dampening the immune response of their hosts. This immune suppression also affects immune responses to third-party antigens such as vaccines. Indeed, accumulating evidence suggests that helminth-infected humans display impaired responses to vaccination. Thus, anthelminthic treatment before vaccination is discussed. Here, we use helminth-infected mice to analyse kinetics and mechanism of helminth-induced interference with vaccination efficacy more precisely. We show that chronic helminth infection completely suppressed antibody responses to a model vaccine. Thereby helminths suppressed the antibody-producing B cells indirectly via suppression of accessory T helper cells. The suppression was more pronounced at later time points of infection and still observed in mice that had terminated the helminth infection for more than 16 weeks. In summary, our results suggest that vaccination may not only fail in helminth-infected individuals, but also in individuals with a history of previous helminth infections. Thus, our report highlights the importance to develop vaccination strategies that are functional despite concurrent helminth infection rather than deworming humans before vaccination.
Collapse
Affiliation(s)
- Irma Haben
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Wiebke Hartmann
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Minka Breloer
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- * E-mail:
| |
Collapse
|
9
|
Blankenhaus B, Reitz M, Brenz Y, Eschbach ML, Hartmann W, Haben I, Sparwasser T, Huehn J, Kühl A, Feyerabend TB, Rodewald HR, Breloer M. Foxp3⁺ regulatory T cells delay expulsion of intestinal nematodes by suppression of IL-9-driven mast cell activation in BALB/c but not in C57BL/6 mice. PLoS Pathog 2014; 10:e1003913. [PMID: 24516385 PMCID: PMC3916398 DOI: 10.1371/journal.ppat.1003913] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 12/19/2013] [Indexed: 12/11/2022] Open
Abstract
Accumulating evidence suggests that IL-9-mediated immunity plays a fundamental role in control of intestinal nematode infection. Here we report a different impact of Foxp3⁺ regulatory T cells (Treg) in nematode-induced evasion of IL-9-mediated immunity in BALB/c and C57BL/6 mice. Infection with Strongyloides ratti induced Treg expansion with similar kinetics and phenotype in both strains. Strikingly, Treg depletion reduced parasite burden selectively in BALB/c but not in C57BL/6 mice. Treg function was apparent in both strains as Treg depletion increased nematode-specific humoral and cellular Th2 response in BALB/c and C57BL/6 mice to the same extent. Improved resistance in Treg-depleted BALB/c mice was accompanied by increased production of IL-9 and accelerated degranulation of mast cells. In contrast, IL-9 production was not significantly elevated and kinetics of mast cell degranulation were unaffected by Treg depletion in C57BL/6 mice. By in vivo neutralization, we demonstrate that increased IL-9 production during the first days of infection caused accelerated mast cell degranulation and rapid expulsion of S. ratti adults from the small intestine of Treg-depleted BALB/c mice. In genetically mast cell-deficient (Cpa3-Cre) BALB/c mice, Treg depletion still resulted in increased IL-9 production but resistance to S. ratti infection was lost, suggesting that IL-9-driven mast cell activation mediated accelerated expulsion of S. ratti in Treg-depleted BALB/c mice. This IL-9-driven mast cell degranulation is a central mechanism of S. ratti expulsion in both, BALB/c and C57BL/6 mice, because IL-9 injection reduced and IL-9 neutralization increased parasite burden in the presence of Treg in both strains. Therefore our results suggest that Foxp3⁺ Treg suppress sufficient IL-9 production for subsequent mast cell degranulation during S. ratti infection in a non-redundant manner in BALB/c mice, whereas additional regulatory pathways are functional in Treg-depleted C57BL/6 mice.
Collapse
Affiliation(s)
| | - Martina Reitz
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Yannick Brenz
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | - Wiebke Hartmann
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Irma Haben
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Tim Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Helmholtz Centre for Infection Research Braunschweig and the Hanover Medical School, Hanover, Germany
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Anja Kühl
- Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie, Charité-Campus Benjamin Franklin, Berlin, Germany
| | | | - Hans-Reimer Rodewald
- Division for Cellular Immunology, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Minka Breloer
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| |
Collapse
|
10
|
Araujo ECB, Barbosa BF, Coutinho LB, Barenco PVC, Sousa LA, Milanezi CM, Bonfá G, Pavanelli WR, Silva JS, Ferro EAV, Silva DAO, Cunha-Junior JP, Silva NM. Heme oxygenase-1 activity is involved in the control of Toxoplasma gondii infection in the lung of BALB/c and C57BL/6 and in the small intestine of C57BL/6 mice. Vet Res 2013; 44:89. [PMID: 24088531 PMCID: PMC3851451 DOI: 10.1186/1297-9716-44-89] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 06/13/2013] [Indexed: 01/12/2023] Open
Abstract
Heme oxygenase-1 (HO-1) is an enzyme that catabolizes free heme, which induces an intense inflammatory response. The expression of HO-1 is induced by different stimuli, triggering an anti-inflammatory response during biological stress. It was previously verified that HO-1 is able to induce indoleamine 2,3-dioxygenase (IDO), an enzyme that is induced by IFN-γ in Toxoplasma gondii infection. To verify the role of HO-1 during in vivo T. gondii infection, BALB/c and C57BL/6 mice were infected with the ME49 strain and treated with zinc protoporphyrin IX (ZnPPIX) or hemin, which inhibit or induce HO-1 activity, respectively. The results show that T. gondii infection induced high levels of HO-1 expression in the lung of BALB/c and C57BL6 mice. The animals treated with ZnPPIX presented higher parasitism in the lungs of both lineages of mice, whereas hemin treatment decreased the parasite replication in this organ and in the small intestine of infected C57BL/6 mice. Furthermore, C57BL/6 mice infected with T. gondii and treated with hemin showed higher levels of IDO expression in the lungs and small intestine than uninfected mice. In conclusion, our data suggest that HO-1 activity is involved in the control of T. gondii in the lungs of both mouse lineages, whereas the hemin, a HO-1 inducer, seems to be involved in the control of parasitism in the small intestine of C57BL/6 mice.
Collapse
Affiliation(s)
- Ester C B Araujo
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, MG, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Costa DL, Guimarães LH, Cardoso TM, Queiroz A, Lago E, Roselino AM, Bacellar O, Carvalho EM, Silva JS. Characterization of regulatory T cell (Treg) function in patients infected with Leishmania braziliensis. Hum Immunol 2013; 74:1491-500. [PMID: 23993989 DOI: 10.1016/j.humimm.2013.08.269] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 07/30/2013] [Accepted: 08/10/2013] [Indexed: 11/27/2022]
Abstract
Th1 immune responses are crucial for eliminating Leishmania parasites. However, despite strong Th1 responses, cutaneous leishmaniasis (CL) patients infected with Leishmania braziliensis develop the disease, while milder Th1 responses are found in sub-clinical (SC) infections. Therefore, CL patients may experience impaired regulatory T cell (Treg) function, causing excessive Th1 responses and tissue damage. To address this hypothesis, we characterized the function of circulating Tregs in L. braziliensis infected CL patients and compared them to Tregs from uninfected controls (UC) and SC subjects. The frequency of circulating Tregs was similar in CL patients, UC and SC subjects. Moreover, CL patients Tregs suppressed lymphocyte proliferation and PBMC pro-inflammatory cytokine production more efficiently than UC Tregs, and also produced higher levels of IL-10 than UC and SC Tregs. Furthermore, PBMC and mononuclear cells from lesions of CL patients responded normally to Treg-induced suppression. Therefore, the lesion development in CL patients infected with L. braziliensis is not associated with impairment in Treg function or failure of cells to respond to immunomodulation. Rather, the increased Treg activation in CL patients may impair parasite elimination, resulting in establishment of chronic infection. Thus, immunological strategies that interfere with this response may improve leishmaniasis treatment.
Collapse
Affiliation(s)
- Diego L Costa
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes, 3900, Ribeirão Preto, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
MAP kinase phosphatase-2 plays a key role in the control of infection with Toxoplasma gondii by modulating iNOS and arginase-1 activities in mice. PLoS Pathog 2013; 9:e1003535. [PMID: 23966857 PMCID: PMC3744406 DOI: 10.1371/journal.ppat.1003535] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 06/18/2013] [Indexed: 11/19/2022] Open
Abstract
The dual specific phosphatase, MAP kinase phosphatase-2 (MKP-2) has recently been demonstrated to negatively regulate macrophage arginase-1 expression, while at the same time to positively regulate iNOS expression. Consequently, MKP-2 is likely to play a significant role in the host interplay with intracellular pathogens. Here we demonstrate that MKP-2(-/-) mice on the C57BL/6 background have enhanced susceptibility compared with wild-type counterparts following infection with type-2 strains of Toxoplasma gondii as measured by increased parasite multiplication during acute infection, increased mortality from day 12 post-infection onwards and increased parasite burdens in the brain, day 30 post-infection. MKP-2(-/-) mice did not, however, demonstrate defective type-1 responses compared with MKP-2(+/+) mice following infection although they did display significantly reduced serum nitrite levels and enhanced tissue arginase-1 expression. Early resistance to T. gondii in MKP-2(+/+), but not MKP-2(-/-), mice was nitric oxide (NO) dependent as infected MKP-2(+/+), but not MKP-2(-/-) mice succumbed within 10 days post-infection with increased parasite burdens following treatment with the iNOS inhibitor L-NAME. Conversely, treatment of infected MKP-2(-/-) but not MKP-2(+/+) mice with nor-NOHA increased parasite burdens indicating a protective role for arginase-1 in MKP-2(-/-) mice. In vitro studies using tachyzoite-infected bone marrow derived macrophages and selective inhibition of arginase-1 and iNOS activities confirmed that both iNOS and arginase-1 contributed to inhibiting parasite replication. However, the effects of arginase-1 were transient and ultimately the role of iNOS was paramount in facilitating long-term inhibition of parasite multiplication within macrophages.
Collapse
|
13
|
Akbar H, Germon S, Berthon P, Dimier-Poisson I, Moiré N. Depletion of CD25⁺ cells during acute toxoplasmosis does not significantly increase mortality in Swiss OF1 mice. Mem Inst Oswaldo Cruz 2013; 107:155-62. [PMID: 22415252 DOI: 10.1590/s0074-02762012000200002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 12/11/2011] [Indexed: 11/21/2022] Open
Abstract
The interleukin (IL)-2R alpha chain (CD25) is expressed on regulatory T cells (Treg), which constitute more than 85% of the CD25+ T cell population in a naïve mouse. CD25 is also expressed on effector T cells in mice suffering from an acute infection by the obligate intracellular protozoan parasite, Toxoplasma gondii. Lethal toxoplasmosis is accompanied by a significant loss of Treg in mice naturally susceptible to toxoplasmosis. The present study was done to explore the role of Treg cells using an anti-CD25 antibody-mediated depletion in mice naturally resistant to toxoplasmosis. Although a significant decrease in the percentage of Treg cells was observed following anti-CD25 monoclonal antibody injections, the depletion of CD25+ cells during acute toxoplasmosis did not significantly increase the mortality of Swiss OF1 mice and no significant difference was observed in the brain parasitic load between the mice in the depleted-infected and isotype-infected groups. We found no significant difference between the titres of total IgG in the sera of the mice from the two groups in the chronic phase. However, CD25+ cells depletion was followed by significantly higher levels of IL-12 in the serum of depleted mice than in that of mice injected with the isotype control antibody.
Collapse
Affiliation(s)
- Haroon Akbar
- Université François Rabelais de Tours, UMR1282 Infectiologie et Santé Publique, F-37000 Tours, France.
| | | | | | | | | |
Collapse
|
14
|
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
|
15
|
Neill DR, Fernandes VE, Wisby L, Haynes AR, Ferreira DM, Laher A, Strickland N, Gordon SB, Denny P, Kadioglu A, Andrew PW. T regulatory cells control susceptibility to invasive pneumococcal pneumonia in mice. PLoS Pathog 2012; 8:e1002660. [PMID: 22563306 PMCID: PMC3334885 DOI: 10.1371/journal.ppat.1002660] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 03/05/2012] [Indexed: 11/18/2022] Open
Abstract
Streptococcus pneumoniae is an important human pathogen responsible for a spectrum of diseases including pneumonia. Immunological and pro-inflammatory processes induced in the lung during pneumococcal infection are well documented, but little is known about the role played by immunoregulatory cells and cytokines in the control of such responses. We demonstrate considerable differences in the immunomodulatory cytokine transforming growth factor (TGF)-β between the pneumococcal pneumonia resistant BALB/c and susceptible CBA/Ca mouse strains. Immunohistochemistry and flow cytometry reveal higher levels of TGF-β protein in BALB/c lungs during pneumococcal pneumonia that correlates with a rapid rise in lung Foxp3+Helios+ T regulatory cells. These cells have protective functions during pneumococcal pneumonia, because blocking their induction with an inhibitor of TGF-β impairs BALB/c resistance to infection and aids bacterial dissemination from lungs. Conversely, adoptive transfer of T regulatory cells to CBA/Ca mice, prior to infection, prolongs survival and decreases bacterial dissemination from lungs to blood. Importantly, strong T regulatory cell responses also correlate with disease-resistance in outbred MF1 mice, confirming the importance of immunoregulatory cells in controlling protective responses to the pneumococcus. This study provides exciting new evidence for the importance of immunomodulation during pulmonary pneumococcal infection and suggests that TGF-β signalling is a potential target for immunotherapy or drug design. Streptococcus pneumoniae is a major human bacterial pathogen that causes a wide range of diseases including pneumonia, meningitis, sepsis and ear infections. The bacterium is responsible for around 1.2 million deaths per year, mostly in high-risk groups such as children, the elderly and those with a weakened immune system. Infection with the pneumococcus can induce a wide-variety of immune responses and disease symptoms and it is not known why some people are more resistant to infection than others. Here, we identify an important role in natural resistance against pneumococcal pneumonia for a group of cells – known as T regulatory cells – that control the immune response to pneumococcal infection. In mice, strong T regulatory cell responses correlate with resistance to invasive pneumococcal pneumonia. Disease-resistance can be boosted by administering T regulatory cells to highly susceptible mice or inhibited by blocking the activity of these cells in resistant mice. These results advance our understanding of the host immunity differences that underpin resistance to pneumococcal pneumonia and offer hope that in the future we might boost resistance in susceptible individuals through modulation of their immune system.
Collapse
Affiliation(s)
- Daniel R. Neill
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Vitor E. Fernandes
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Laura Wisby
- MRC Harwell, Mammalian Genetics Unit, Harwell Science and Innovation Campus, Oxfordshire, United Kingdom
| | - Andrew R. Haynes
- MRC Harwell, Mammalian Genetics Unit, Harwell Science and Innovation Campus, Oxfordshire, United Kingdom
| | - Daniela M. Ferreira
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Ameera Laher
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Natalie Strickland
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Stephen B. Gordon
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Paul Denny
- MRC Harwell, Mammalian Genetics Unit, Harwell Science and Innovation Campus, Oxfordshire, United Kingdom
| | - Aras Kadioglu
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
- * E-mail: (AK); (PWA)
| | - Peter W. Andrew
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
- * E-mail: (AK); (PWA)
| |
Collapse
|