1
|
Geng X, Li R, Du J, Zhang M, Jiang W, Sun Q, Mi R, Qin S, Wang Q. Toxoplasma gondii Type I TR and ROP16 Synergistically Downregulate IL-12 to Inhibit Host Reactive Oxygen Species Production. Pathogens 2025; 14:171. [PMID: 40005546 PMCID: PMC11858468 DOI: 10.3390/pathogens14020171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/29/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Toxoplasma gondii is an obligate intracellular opportunistic protozoan parasite. T. gondii invasion disturbs the balance between reactive oxygen species (ROS) production and antioxidant capacity in the host, triggering the oxidative stress response. Thioredoxin reductase (TR) of T. gondii helps to escape ROS-induced damage in the host, whereas T. gondii rhoptry protein 16 (ROP16) downregulates host innate immunity to suppress excessive inflammation and inhibit ROS production. However, whether TR and ROP16 synergistically promote resistance to ROS-induced damage remains unclear. Here, we used the CRISPR/Cas9 technology to successfully obtain a double TR and ROP16 gene knockout T. gondii strain. The double deletion of TR and ROP16 in T. gondii weakened its growth ability in vitro and decreased its virulence in vivo. Moreover, the double deletion of TR and ROP16 resulted in a lower antioxidant capacity, higher degree of lipid oxidation, and elevated ROS levels in the parasite and host cells. Interestingly, the deletion of the TR and ROP16 genes in T. gondii synergistically increased IL-12 levels, which triggered host cells to produce more ROS to resist T. gondii infection. These results show that TR and ROP16 in T. gondii play a synergistic role, facilitating resistance to ROS damage incurred by host immune cells through different pathways.
Collapse
Affiliation(s)
- Xiaoling Geng
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (X.G.); (R.L.); (J.D.); (M.Z.); (W.J.); (Q.S.); (R.M.); (S.Q.)
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Ruifang Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (X.G.); (R.L.); (J.D.); (M.Z.); (W.J.); (Q.S.); (R.M.); (S.Q.)
| | - Jingying Du
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (X.G.); (R.L.); (J.D.); (M.Z.); (W.J.); (Q.S.); (R.M.); (S.Q.)
| | - Manyu Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (X.G.); (R.L.); (J.D.); (M.Z.); (W.J.); (Q.S.); (R.M.); (S.Q.)
| | - Wei Jiang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (X.G.); (R.L.); (J.D.); (M.Z.); (W.J.); (Q.S.); (R.M.); (S.Q.)
| | - Qing Sun
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (X.G.); (R.L.); (J.D.); (M.Z.); (W.J.); (Q.S.); (R.M.); (S.Q.)
| | - Rongsheng Mi
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (X.G.); (R.L.); (J.D.); (M.Z.); (W.J.); (Q.S.); (R.M.); (S.Q.)
| | - Shuang Qin
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (X.G.); (R.L.); (J.D.); (M.Z.); (W.J.); (Q.S.); (R.M.); (S.Q.)
| | - Quan Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai 200241, China; (X.G.); (R.L.); (J.D.); (M.Z.); (W.J.); (Q.S.); (R.M.); (S.Q.)
| |
Collapse
|
2
|
Gbedande K, Ibitokou SA, Endrino MJD, Yap GS, Brown MG, Stephens R. Heightened innate immune state induced by viral vector leads to enhanced response to challenge and prolongs malaria vaccine protection. iScience 2024; 27:111468. [PMID: 39758993 PMCID: PMC11697717 DOI: 10.1016/j.isci.2024.111468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/01/2024] [Accepted: 11/21/2024] [Indexed: 01/07/2025] Open
Abstract
Cytomegalovirus is a promising vaccine vector; however, mechanisms promoting CD4 T cell responses to challenge, by CMV as a vector, are unknown. The ability of MCMV to prolong immunity generated by short-lived malaria vaccine was tested. MCMV provided non-specific protection to challenge with Plasmodium and increased interleukin-12 (IL-12) and CD8α+ dendritic cell (DC) numbers through prolonged MCMV-dependent interferon gamma (IFN-γ) production. This late innate response to MCMV increased IL-12 upon challenge and increased the polyclonal CD4 effector T cell response to Plasmodium, protecting in an IL-12-dependent manner. Although Plasmodium-vaccine-induced protection decayed by d200, MCMV restored protection through IFN-γ. Mechanistically, protection depended on MCMV-induced-IFN-γ increasing CD8α+ DCs and IL-12p40. MCMV expressing a Plasmodium epitope increased parasite-specific CD4 effector and effector memory T cells persisting after malaria vaccination, both phenotypes reported to protect. Overall, enhanced innate cell status, a mechanism of heterologous protection by MCMV, led to a stronger T cell response to challenge.
Collapse
Affiliation(s)
- Komi Gbedande
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555-0435, USA
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Cancer Center, 205 S. Orange Avenue, Newark, NJ 07103, USA
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Cancer Center, 205 S. Orange Avenue, Newark, NJ 07103, USA
| | - Samad A. Ibitokou
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555-0435, USA
| | - Mark Joseph D. Endrino
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Cancer Center, 205 S. Orange Avenue, Newark, NJ 07103, USA
| | - George S. Yap
- Center for Immunity and Inflammation, and Department of Medicine, Rutgers New Jersey Medical School, Cancer Center, 205 S. Orange Avenue, Newark, NJ 07103, USA
| | - Michael G. Brown
- Department of Medicine, Division of Nephrology, and the Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA
| | - Robin Stephens
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555-0435, USA
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Cancer Center, 205 S. Orange Avenue, Newark, NJ 07103, USA
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Cancer Center, 205 S. Orange Avenue, Newark, NJ 07103, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
3
|
Doherty CM, Patterson PR, Emeanuwa JA, Belmares Ortega J, Fox BA, Bzik DJ, Denkers EY. T lymphocyte-dependent IL-10 down-regulates a cytokine storm driven by Toxoplasma gondii GRA24. mBio 2024; 15:e0145524. [PMID: 39440975 PMCID: PMC11559025 DOI: 10.1128/mbio.01455-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
As a model organism in the study of immunity to infection, Toxoplasma gondii has been instrumental in establishing key principles of host anti-microbial defense and its regulation. Here, we employed an attenuated uracil auxotroph strain of Type I Toxoplasma designated OMP to further untangle the early immune response to this parasitic pathogen. Experiments using αβ T cell-deficient Tcrb-/- mice unexpectedly revealed that an intact αβ T lymphocyte compartment was essential to survive infection with OMP. Subsequent antibody depletion and knockout mouse experiments demonstrated contributions from CD4+ T cells and most predominantly CD8+ T cells in resistance. Using transgenic knockout mice, we found only a partial requirement for IFN-γ and a lack of requirement for Toll-like receptor (TLR) adaptor MyD88 in resistance. In contrast to other studies on Toxoplasma, the ability to survive OMP infection did not require IL-12p40. Surprisingly, T cell-dependent IL-10 was found to be critical for survival, and deficiency of this cytokine triggered an abnormally high systemic inflammatory response. We also found that parasite molecule GRA24, a dense granule protein that triggers TLR-independent IL-12 production, acts as a virulence factor contributing to death of OMP-infected Tcrb-/- and IL-10-/- mice. Furthermore, resistance against OMP was restored in Tcrb-/- mice via monoclonal depletion of IL-12p40, suggesting that GRA24-induced IL-12 underlies the fatal immunopathology observed. Collectively, our studies provide insight into a novel and rapidly arising T lymphocyte-dependent anti-inflammatory response to T. gondii which operates independently of MyD88 and IL-12 and that depends on the function of parasite-dense granule protein GRA24.IMPORTANCEAs a model infectious microbe and an important human pathogen, the apicomplexan Toxoplasma gondii has provided many important insights into innate and adaptive immunity to infection. We show here that a low virulence uracil auxotrophic Toxoplasma strain emerges as a virulent parasite in the absence of an intact T cell compartment. Both CD4+ and CD8+ T lymphocytes are required for optimal protection, in line with previous findings in other models of Toxoplasma infection. Nevertheless, several novel aspects of the response were identified in our study. Protection occurs independently of IL-12 and MyD88 and only partially requires IFN-γ. This is noteworthy particularly because the cytokines IL-12 and IFN-γ have previously been regarded as essential for protective immunity to T. gondii. Instead, we identified the anti-inflammatory effects of T cell-dependent IL-10 as the critical factor enabling host survival. The parasite dense granule protein GRA24, a host-directed mitogen-activated protein kinase activator, was identified as a major virulence factor in T cell-deficient hosts. Collectively, our results provide new and unexpected insights into host resistance to Toxoplasma.
Collapse
Affiliation(s)
- Claire M. Doherty
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Paige R. Patterson
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Julie A. Emeanuwa
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Jessica Belmares Ortega
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Barbara A. Fox
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - David J. Bzik
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Eric Y. Denkers
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, USA
| |
Collapse
|
4
|
Weng S, Tian E, Gao M, Zhang S, Yang G, Zhou B. Eimeria: Navigating complex intestinal ecosystems. PLoS Pathog 2024; 20:e1012689. [PMID: 39576763 PMCID: PMC11584145 DOI: 10.1371/journal.ppat.1012689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2024] Open
Abstract
Eimeria is an intracellular obligate apicomplexan parasite that parasitizes the intestinal epithelial cells of livestock and poultry, exhibiting strong host and tissue tropism. Parasite-host interactions involve complex networks and vary as the parasites develop in the host. However, understanding the underlying mechanisms remains a challenge. Acknowledging the lack of studies on Eimeria invasion mechanism, we described the possible invasion process through comparative analysis with other apicomplexan parasites and explored the fact that parasite-host interactions serve as a prerequisite for successful recognition, penetration of the intestinal mechanical barrier, and completion of the invasion. Although it is recognized that microbiota can enhance the host immune capacity to resist Eimeria invasion, changes in the microenvironment can, in turn, contribute to Eimeria invasion and may be associated with reduced immune capacity. We also discuss the immune evasion strategies of Eimeria, emphasizing that the host employs sophisticated immune regulatory mechanisms to suppress immune evasion by parasites, thereby sustaining a balanced immune response. This review aims to deepen our understanding of Eimeria-host interactions, providing a theoretical basis for the study of the pathogenicity of Eimeria and the development of novel anticoccidial drugs.
Collapse
Affiliation(s)
- Shengjie Weng
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, People’s Republic of China
| | - Erjie Tian
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, People’s Republic of China
| | - Meng Gao
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, People’s Republic of China
| | - Siyu Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, People’s Republic of China
| | - Guodong Yang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, People’s Republic of China
| | - Bianhua Zhou
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, People’s Republic of China
| |
Collapse
|
5
|
Reis TSA, Siqueira VDS, Ferreira SRR, Domann N, Rodrigues BA, Fleury ACC, de Souza IMFNB, Cardoso LPV, Siqueira CS, Rezende HHA. Evaluation of nitazoxanide in the treatment of experimental murine neurotoxoplasmosis. Rev Inst Med Trop Sao Paulo 2024; 66:e61. [PMID: 39417497 PMCID: PMC11469426 DOI: 10.1590/s1678-9946202466061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 08/26/2024] [Indexed: 10/19/2024] Open
Abstract
Toxoplasmosis is a widespread zoonotic disease that poses significant public health concern globally, with neurotoxoplasmosis being a severe complication associated with high mortality rates. The standard therapy for neurotoxoplasmosis involves a combination of sulfadiazine and pyrimethamine, which, despite its efficacy, is often limited by adverse effects leading to treatment discontinuation. This study aimed to evaluate the in vivo efficacy of nitazoxanide in treating neurotoxoplasmosis in mice infected with the Me49 strain. The study comprised two groups: Group I, including subgroups of uninfected, infected and treated with saline, and infected and untreated mice; and Group II, comprising infected mice treated with nitazoxanide at 100 mg/kg/day, nitazoxanide at 150 mg/kg/day, and pyrimethamine combined with sulfadiazine. After 14 days of treatment, the mice were euthanized for organ collection. Histopathological examination of the brains revealed that the highest dose of nitazoxanide reduced parasitic load and cerebral hemorrhages. Biochemical and histopathological analyses of liver and kidney tissues demonstrated toxicological profiles comparable to pyrimethamine and sulfadiazine. However, despite showing efficacy and similar toxicity levels, nitazoxanide treatment was less effective regimen in controlling neurotoxoplasmosis in this experimental model compared to the pyrimethamine and sulfadiazine. Thus, while nitazoxanide presents potential in neurotoxoplasmosis treatment, pyrimethamine combined with sulfadiazine remains the preferred therapeutic choice based on better efficacy observed in this study.
Collapse
Affiliation(s)
- Thaís Santos Anjo Reis
- Universidade Federal de Jataí, Instituto de Ciências da Saúde, Programa de Pós-Graduação em Ciências Aplicadas à Saúde, Jataí, Goiás, Brazil
| | - Victor da Silva Siqueira
- Universidade Federal de Jataí, Instituto de Ciências da Saúde, Programa de Pós-Graduação em Ciências Aplicadas à Saúde, Jataí, Goiás, Brazil
| | - Stéfanne Rodrigues Rezende Ferreira
- Universidade Federal de Jataí, Instituto de Ciências da Saúde, Programa de Pós-Graduação em Ciências Aplicadas à Saúde, Jataí, Goiás, Brazil
| | - Natália Domann
- Universidade Federal de Jataí, Instituto de Ciências da Saúde, Programa de Pós-Graduação em Ciências Aplicadas à Saúde, Jataí, Goiás, Brazil
| | - Benílton Alves Rodrigues
- Universidade Federal de Jataí, Instituto de Ciências da Saúde, Curso de Biomedicina, Jataí, Goiás, Brazil
| | | | | | - Ludimila Paula Vaz Cardoso
- Universidade Federal de Jataí, Instituto de Ciências da Saúde, Programa de Pós-Graduação em Ciências Aplicadas à Saúde, Jataí, Goiás, Brazil
| | - Carla Silva Siqueira
- Universidade Federal de Jataí, Instituto de Ciências da Saúde, Programa de Pós-Graduação em Ciências Aplicadas à Saúde, Jataí, Goiás, Brazil
| | - Hanstter Hallison Alves Rezende
- Universidade Federal de Jataí, Instituto de Ciências da Saúde, Programa de Pós-Graduação em Ciências Aplicadas à Saúde, Jataí, Goiás, Brazil
| |
Collapse
|
6
|
Pinheiro AAS, Torrecilhas AC, Souza BSDF, Cruz FF, Guedes HLDM, Ramos TD, Lopes‐Pacheco M, Caruso‐Neves C, Rocco PRM. Potential of extracellular vesicles in the pathogenesis, diagnosis and therapy for parasitic diseases. J Extracell Vesicles 2024; 13:e12496. [PMID: 39113589 PMCID: PMC11306921 DOI: 10.1002/jev2.12496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 07/11/2024] [Indexed: 08/11/2024] Open
Abstract
Parasitic diseases have a significant impact on human and animal health, representing a major hazard to the public and causing economic and health damage worldwide. Extracellular vesicles (EVs) have long been recognized as diagnostic and therapeutic tools but are now also known to be implicated in the natural history of parasitic diseases and host immune response modulation. Studies have shown that EVs play a role in parasitic disease development by interacting with parasites and communicating with other types of cells. This review highlights the most recent research on EVs and their role in several aspects of parasite-host interactions in five key parasitic diseases: Chagas disease, malaria, toxoplasmosis, leishmaniasis and helminthiases. We also discuss the potential use of EVs as diagnostic tools or treatment options for these infectious diseases.
Collapse
Affiliation(s)
- Ana Acacia Sá Pinheiro
- Instituto de Biofísica Carlos Chagas FilhoUniversidade Federal do Rio de Janeiro (UFRJ)Rio de JaneiroBrazil
- Rio de Janeiro Innovation Network in Nanosystems for Health‐NanoSAÚDE/Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ)Rio de JaneiroBrazil
| | - Ana Claudia Torrecilhas
- Departamento de Ciências FarmacêuticasDiadema Campus, Instituto de Ciências Ambientais, Químicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP)DiademaSão PauloBrazil
| | - Bruno Solano de Freitas Souza
- Center for Biotechnology and Cell TherapySão Rafael HospitalSalvadorBrazil
- D'Or Institute for Research and Education (IDOR)SalvadorBrazil
| | - Fernanda Ferreira Cruz
- Instituto de Biofísica Carlos Chagas FilhoUniversidade Federal do Rio de Janeiro (UFRJ)Rio de JaneiroBrazil
- Rio de Janeiro Innovation Network in Nanosystems for Health‐NanoSAÚDE/Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ)Rio de JaneiroBrazil
| | - Herbert Leonel de Matos Guedes
- Instituto de Microbiologia Paulo de Goés (IMPG)Universidade Federal do Rio de Janeiro (UFRJ)Rio de JaneiroBrazil
- Fundação Oswaldo Cruz (FIOCRUZ)Instituto Oswaldo Cruz (IOC)Rio de JaneiroBrazil
| | - Tadeu Diniz Ramos
- Instituto de Microbiologia Paulo de Goés (IMPG)Universidade Federal do Rio de Janeiro (UFRJ)Rio de JaneiroBrazil
- Fundação Oswaldo Cruz (FIOCRUZ)Instituto Oswaldo Cruz (IOC)Rio de JaneiroBrazil
| | - Miqueias Lopes‐Pacheco
- Instituto de Biofísica Carlos Chagas FilhoUniversidade Federal do Rio de Janeiro (UFRJ)Rio de JaneiroBrazil
- Deparment of PediatricsCenter for Cystic Fibrosis and Airway Disease ResearchEmory University School of MedicineAtlantaGeorgiaUSA
| | - Celso Caruso‐Neves
- Instituto de Biofísica Carlos Chagas FilhoUniversidade Federal do Rio de Janeiro (UFRJ)Rio de JaneiroBrazil
- Rio de Janeiro Innovation Network in Nanosystems for Health‐NanoSAÚDE/Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ)Rio de JaneiroBrazil
- National Institute of Science and Technology for Regenerative MedicineINCT‐REGENERARio de JaneiroBrazil
| | - Patricia R. M. Rocco
- Instituto de Biofísica Carlos Chagas FilhoUniversidade Federal do Rio de Janeiro (UFRJ)Rio de JaneiroBrazil
- Rio de Janeiro Innovation Network in Nanosystems for Health‐NanoSAÚDE/Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ)Rio de JaneiroBrazil
- National Institute of Science and Technology for Regenerative MedicineINCT‐REGENERARio de JaneiroBrazil
| |
Collapse
|
7
|
Na J, Engwerda C. The role of CD4 + T cells in visceral leishmaniasis; new and emerging roles for NKG7 and TGFβ. Front Cell Infect Microbiol 2024; 14:1414493. [PMID: 38881737 PMCID: PMC11176485 DOI: 10.3389/fcimb.2024.1414493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/21/2024] [Indexed: 06/18/2024] Open
Abstract
Visceral leishmaniasis is a potentially devastating neglected tropical disease caused by the protozoan parasites Leishmania donovani and L. infantum (chagasi). These parasites reside in tissue macrophages and survive by deploying a number of mechanisms aimed at subverting the host immune response. CD4+ T cells play an important role in controlling Leishmania parasites by providing help in the form of pro-inflammatory cytokines to activate microbiocidal pathways in infected macrophages. However, because these cytokines can also cause tissue damage if over-produced, regulatory immune responses develop, and the balance between pro-inflammatory and regulatory CD4+ T cells responses determines the outcomes of infection. Past studies have identified important roles for pro-inflammatory cytokines such as IFNγ and TNF, as well as regulatory co-inhibitory receptors and the potent anti-inflammatory cytokine IL-10. More recently, other immunoregulatory molecules have been identified that play important roles in CD4+ T cell responses during VL. In this review, we will discuss recent findings about two of these molecules; the NK cell granule protein Nkg7 and the anti-inflammatory cytokine TGFβ, and describe how they impact CD4+ T cell functions and immune responses during visceral leishmaniasis.
Collapse
Affiliation(s)
- Jinrui Na
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Medicine, University of Queensland, Brisbane, QLD, Australia
| | | |
Collapse
|
8
|
Zhao XY, Lempke SL, Urbán Arroyo JC, Brown IG, Yin B, Magaj MM, Holness NK, Smiley J, Redemann S, Ewald SE. iNOS is necessary for GBP-mediated T. gondii clearance in murine macrophages via vacuole nitration and intravacuolar network collapse. Nat Commun 2024; 15:2698. [PMID: 38538595 PMCID: PMC10973475 DOI: 10.1038/s41467-024-46790-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 03/04/2024] [Indexed: 04/02/2024] Open
Abstract
Toxoplasma gondii is an obligate intracellular parasite of rodents and humans. Interferon-inducible guanylate binding proteins (GBPs) are mediators of T. gondii clearance, however, this mechanism is incomplete. Here, using automated spatially targeted optical micro proteomics we demonstrate that inducible nitric oxide synthetase (iNOS) is highly enriched at GBP2+ parasitophorous vacuoles (PV) in murine macrophages. iNOS expression in macrophages is necessary to limit T. gondii load in vivo and in vitro. Although iNOS activity is dispensable for GBP2 recruitment and PV membrane ruffling; parasites can replicate, egress and shed GBP2 when iNOS is inhibited. T. gondii clearance by iNOS requires nitric oxide, leading to nitration of the PV and collapse of the intravacuolar network of membranes in a chromosome 3 GBP-dependent manner. We conclude that reactive nitrogen species generated by iNOS cooperate with GBPs to target distinct structures in the PV that are necessary for optimal parasite clearance in macrophages.
Collapse
Affiliation(s)
- Xiao-Yu Zhao
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Samantha L Lempke
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jan C Urbán Arroyo
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Isabel G Brown
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Bocheng Yin
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Magdalena M Magaj
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Nadia K Holness
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jamison Smiley
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Stefanie Redemann
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Sarah E Ewald
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
9
|
Zhao XY, Lempke SL, Urbán Arroyo JC, Yin B, Holness NK, Smiley J, Ewald SE. Inducible nitric oxide synthase (iNOS) is necessary for GBP-mediated T. gondii restriction in murine macrophages via vacuole nitration and intravacuolar network collapse. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.24.549965. [PMID: 37546987 PMCID: PMC10402109 DOI: 10.1101/2023.07.24.549965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Toxoplasma gondii is an obligate intracellular, protozoan pathogen of rodents and humans. T. gondii's ability to grow within cells and evade cell-autonomous immunity depends on the integrity of the parasitophorous vacuole (PV). Interferon-inducible guanylate binding proteins (GBPs) are central mediators of T. gondii clearance, however, the precise mechanism linking GBP recruitment to the PV and T. gondii restriction is not clear. This knowledge gap is linked to heterogenous GBP-targeting across a population of vacuoles and the lack of tools to selectively purify the intact PV. To identify mediators of parasite clearance associated with GBP2-positive vacuoles, we employed a novel protein discovery tool automated spatially targeted optical micro proteomics (autoSTOMP). This approach identified inducible nitric oxide synthetase (iNOS) enriched at levels similar to the GBPs in infected bone marrow-derived myeloid cells. iNOS expression on myeloid cells was necessary for mice to control T. gondii growth in vivo and survive acute infection. T. gondii infection of IFNγ-primed macrophage was sufficient to robustly induce iNOS expression. iNOS restricted T. gondii infection through nitric oxide synthesis rather than arginine depletion, leading to robust and selective nitration of the PV. Optimal parasite restriction by iNOS and vacuole nitration depended on the chromosome 3 GBPs. Notably, GBP2 recruitment and ruffling of the PV membrane occurred in iNOS knockouts, however, these vacuoles contained dividing parasites. iNOS activity was necessary for the collapse of the intravacuolar network of nanotubular membranes which connects parasites to each other and the host cytosol. Based on these data we conclude reactive nitrogen species generated by iNOS cooperate with the chromosome 3 GBPs to target distinct biology of the PV that are necessary for optimal parasite clearance in murine myeloid cells.
Collapse
Affiliation(s)
- Xiao-Yu Zhao
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- The Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Samantha L. Lempke
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- The Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Jan C. Urbán Arroyo
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- The Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Bocheng Yin
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- The Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Nadia K. Holness
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- The Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Jamison Smiley
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- The Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Sarah E. Ewald
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- The Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
10
|
Chen J, Wang X, Li J, Sun L, Chen X, Chu Z, Zhang Z, Wu H, Zhao X, Li H, Zhang X. Influenza A Virus Weakens the Immune Response of Mice to Toxoplasma gondii, Thereby Aggravating T. gondii Infection. Vet Sci 2023; 10:vetsci10050354. [PMID: 37235437 DOI: 10.3390/vetsci10050354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
This study aimed to investigate the relationship between the T. gondii type II strain (Pru) and respiratory viral infections, specifically focusing on the co-infection with PR8 (influenza A/Puerto Rico/8/34). In this study, we found that the number of T. gondii (Pru) in the lungs of co-infected mice was significantly higher and lesions were more severe than those in the group infected with T. gondii (Pru) alone, whereas IAV (influenza A virus) copy numbers of co-infected and PR8 alone infected groups were negligible, suggesting that infection with IAV increased the pathogenicity of T. gondii (Pru) in mice. The invasion and proliferation assays demonstrated no significant effect of co-infection on T. gondii (Pru) infection or replication in vitro. To further explore the factors causing the altered pathogenicity of T. gondii (Pru) caused by co-infection, we found that decreased expression levels of IL-1β, IL-6, and IL-12 in the co-infected group were associated with the early immune responses against T. gondii (Pru), which affected the division of T. gondii (Pru). Moreover, the significant decrease in the CD4+/CD8+ ratio indicated a weakened long-term immune killing ability of the host against T. gondii (Pru) following IAV infection. In conclusion, a T. gondii type II strain (Pru) could not be properly cleared by the host immune system after IAV infection, resulting in toxoplasmosis and even death in mice.
Collapse
Affiliation(s)
- Junpeng Chen
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an 271002, China
| | - Xiaoli Wang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an 271002, China
| | - Jinxuan Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an 271002, China
| | - Lingyu Sun
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an 271002, China
| | - Xiao Chen
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an 271002, China
| | - Ziyu Chu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an 271002, China
| | - Zhenzhao Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an 271002, China
| | - Hongxia Wu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an 271002, China
| | - Xiaomin Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an 271002, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an 271002, China
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an 271002, China
| | - Hongmei Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an 271002, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an 271002, China
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an 271002, China
| | - Xiao Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an 271002, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an 271002, China
- Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an 271002, China
| |
Collapse
|
11
|
Ibitokou SA, Gbedande K, Opata MM, Carpio VH, Marshall KM, Stephens R. Effects of Low-Level Persistent Infection on Maintenance of Immunity by CD4 T Cell Subsets and Th1 Cytokines. Infect Immun 2023; 91:e0053122. [PMID: 36920200 PMCID: PMC10016079 DOI: 10.1128/iai.00531-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
CD4 T cells are required, along with antibodies, for complete protection from blood-stage infection with Plasmodium spp., which cause malaria. Without continuous exposure, as on emigration of people from endemic areas, protection from malaria decays. As in other persistent infections, low-level Plasmodium chabaudi infection protects the host from reinfection at 2 months postinfection, a phenomenon termed premunition. Premunition is correlated with T cell responses, rather than antibody levels. We previously showed that while both effector T cells (Teff) and memory T cells (Tmem) are present after infection, Teff protect better than Tmem. Here, we studied T cell kinetics post-infection by labeling dividing Ifng+ T cells with 5-bromo-2'-deoxyuridine (BrdU) in infected Ifng reporter mice. Large drops in specific T cell numbers and Ifng+ cells upon clearance of parasites suggest a mechanism for decay of protection. Although protection decays, CD4 Tmem persist, including a highly differentiated CD27- effector memory (Tem) subset that maintains some Ifng expression. In addition, pretreatment of chronically infected animals with neutralizing antibody to interferon gamma (IFN-γ) or with clodronate liposomes before reinfection decreases premunition, supporting a role for Th1-type immunity to reinfection. A pulse-chase experiment comparing chronically infected to treated animals showed that recently divided Ifng+ T cells, particularly IFN-γ+ TNF+ IL-2- T cells, are promoted by persistent infection. These data suggest that low-level persistent infection reduces CD4+ Tmem and multifunctional Teff survival, but promotes IFN-γ+ TNF+ IL-2- T cells and Ifng+ terminally differentiated effector T cells, and prolongs immunity.
Collapse
Affiliation(s)
- Samad A. Ibitokou
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
| | - Komi Gbedande
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
| | - Michael M. Opata
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
| | - Victor H. Carpio
- Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Karis M. Marshall
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
| | - Robin Stephens
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
- Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
12
|
Moretto MM, Chen J, Meador M, Phan J, Khan IA. A Lower Dose of Infection Generates a Better Long-Term Immune Response against Toxoplasma gondii. Immunohorizons 2023; 7:177-190. [PMID: 36883950 PMCID: PMC10563383 DOI: 10.4049/immunohorizons.2300006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 02/11/2023] [Indexed: 03/09/2023] Open
Abstract
Toxoplasma gondii, an obligate intracellular pathogen, induces a strong immune response in the infected host. In the encephalitis model of infection, long-term protective immunity is mediated by CD8 T cells, with the CD4 T cell population providing important help. Most of the immune studies have used a 10- to 20-cyst dose of T. gondii, which leads to T cell dysfunctionality during the late phase of chronic infection and increases the chances of reactivation. In the current study, we compared the immune response of mice orally infected with either 2 or 10 cysts of T. gondii. During the acute phase, we demonstrate that the lower dose of infection generates a reduced number of CD4 and CD8 T cells, but the frequency of functional CD4 or CD8 T cells is similar in animals infected with two different doses. However, Ag-experienced T cells (both CD4 and CD8) are better maintained in lower dose-infected mice at 8 wk postinfection, with an increase number functional cells that exhibit lower multiple inhibitory receptor expression. In addition to better long-term T cell immunity, animals infected with a lower dose display reduced inflammation manifested by lesser Ag-specific T cell and cytokine responses during the very early stage of the acute infection. Our studies suggest a previously unappreciated role of dose-dependent early programming/imprinting of the long-term CD4/CD8 T cell response during T. gondii infection. These observations point to the need for an in-depth analysis of how early events shape long-term immunity against this pathogen.
Collapse
Affiliation(s)
- Magali M. Moretto
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC
| | - Jie Chen
- Department of Medicine, The George Washington University, Washington, DC
| | - Morgan Meador
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC
| | - Jasmine Phan
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC
| | - Imtiaz A. Khan
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC
| |
Collapse
|
13
|
Mittereder LR, Swoboda J, De Pascalis R, Elkins KL. IL-12p40 is essential but not sufficient for Francisella tularensis LVS clearance in chronically infected mice. PLoS One 2023; 18:e0283161. [PMID: 36972230 PMCID: PMC10042368 DOI: 10.1371/journal.pone.0283161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 03/02/2023] [Indexed: 03/29/2023] Open
Abstract
IL-12p40 plays an important role in F. tularensis Live Vaccine Strain (LVS) clearance that is independent of its functions as a part of the heterodimeric cytokines IL-12p70 or IL-23. In contrast to WT, p35, or p19 knockout (KO) mice, p40 KO mice infected with LVS develop a chronic infection that does not resolve. Here, we further evaluated the role of IL-12p40 in F. tularensis clearance. Despite reduced IFN-γ production, primed splenocytes from p40 KO and p35 KO mice appeared functionally similar to those from WT mice during in vitro co-culture assays of intramacrophage bacterial growth control. Gene expression analysis revealed a subset of genes that were upregulated in re-stimulated WT and p35 KO splenocytes, but not p40 KO splenocytes, and thus are candidates for involvement in F. tularensis clearance. To directly evaluate a potential mechanism for p40 in F. tularensis clearance, we reconstituted protein levels in LVS-infected p40 KO mice using either intermittent injections of p40 homodimer (p80) or treatment with a p40-producing lentivirus construct. Although both delivery strategies yielded readily detectable levels of p40 in sera and spleens, neither treatment had a measurable impact on LVS clearance by p40 KO mice. Taken together, these studies demonstrate that clearance of F. tularensis infection depends on p40, but p40 monomers and/or dimers alone are not sufficient.
Collapse
Affiliation(s)
- Lara R Mittereder
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Jonathan Swoboda
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Roberto De Pascalis
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Karen L Elkins
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| |
Collapse
|
14
|
A. PORTES JULIANA, C. VOMMARO ROSSIANE, AYRES CALDAS LUCIO, S. MARTINS-DUARTE ERICA. Intracellular life of protozoan Toxoplasma gondii: Parasitophorous vacuole establishment and survival strategies. BIOCELL 2023. [DOI: 10.32604/biocell.2023.026629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
15
|
Elmasry A, Aboukamar WA, Hany H, Elmehankar MS. The immunomodulatory effects of roflumilast on tachyzoite-bradyzoite transition in a murine model of Toxoplasma gondii. Int Immunopharmacol 2022; 113:109348. [DOI: 10.1016/j.intimp.2022.109348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/09/2022] [Accepted: 10/10/2022] [Indexed: 11/05/2022]
|
16
|
Seth A, Kar S. Host-directed antileishmanial interventions: Harvesting unripe fruits to reach fruition. Int Rev Immunol 2022; 42:217-236. [PMID: 35275772 DOI: 10.1080/08830185.2022.2047670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Leishmaniasis is an exemplary paradigm of immune evasion, fraught with the perils of limited clinical assistance, escalating costs of treatment and made worse with the lack of suitable vaccine. While drugs remain central to large-scale disease control, the growing emergence of parasite resistance necessitates the need for combination therapy involving host-directed immunological agents. Also, since prolonged disease progression is associated with strong immune suppression of the host, augmentation of host immunity via restoration of the immunoregulatory circuit involving antigen-presenting cells and T-cells, activation of macrophage function and/or CD4+ T helper 1 cell differentiation may serve as an ideal approach to resolve severe cases of leishmaniasis. As such, therapies that embody a synergistic approach that involve direct killing of the parasite in addition to elevating host immunity are likely to pave the way for widespread elimination of leishmaniasis in the future. With this review, we aim to recapitulate the various immunotherapeutic agents found to hold promise in antileishmanial treatment both in vitro and in vivo. These include parasite-specific antigens, dendritic cell-targeted therapy, recombinant inhibitors of various components intrinsic to immune cell signaling and agonists or antagonists to immune cells and cytokines. We also summarize their abilities to direct therapeutic skewing of the host cell-immune response and review their potential to combat the disease either alone, or as adjunct modalities.
Collapse
Affiliation(s)
- Anuradha Seth
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Susanta Kar
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
17
|
Bąska P, Norbury LJ. The Role of Nuclear Factor Kappa B (NF-κB) in the Immune Response against Parasites. Pathogens 2022; 11:pathogens11030310. [PMID: 35335634 PMCID: PMC8950322 DOI: 10.3390/pathogens11030310] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/17/2022] [Accepted: 02/25/2022] [Indexed: 12/28/2022] Open
Abstract
The immune system consists of various cells, organs, and processes that interact in a sophisticated manner to defend against pathogens. Upon initial exposure to an invader, nonspecific mechanisms are raised through the activation of macrophages, monocytes, basophils, mast cells, eosinophils, innate lymphoid cells, or natural killer cells. During the course of an infection, more specific responses develop (adaptive immune responses) whose hallmarks include the expansion of B and T cells that specifically recognize foreign antigens. Cell to cell communication takes place through physical interactions as well as through the release of mediators (cytokines, chemokines) that modify cell activity and control and regulate the immune response. One regulator of cell states is the transcription factor Nuclear Factor kappa B (NF-κB) which mediates responses to various stimuli and is involved in a variety of processes (cell cycle, development, apoptosis, carcinogenesis, innate and adaptive immune responses). It consists of two protein classes with NF-κB1 (p105/50) and NF-κB2 (p100/52) belonging to class I, and RelA (p65), RelB and c-Rel belonging to class II. The active transcription factor consists of a dimer, usually comprised of both class I and class II proteins conjugated to Inhibitor of κB (IκB). Through various stimuli, IκB is phosphorylated and detached, allowing dimer migration to the nucleus and binding of DNA. NF-κB is crucial in regulating the immune response and maintaining a balance between suppression, effective response, and immunopathologies. Parasites are a diverse group of organisms comprised of three major groups: protozoa, helminths, and ectoparasites. Each group induces distinct effector immune mechanisms and is susceptible to different types of immune responses (Th1, Th2, Th17). This review describes the role of NF-κB and its activity during parasite infections and its contribution to inducing protective responses or immunopathologies.
Collapse
Affiliation(s)
- Piotr Bąska
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, 02-786 Warsaw, Poland
- Correspondence:
| | - Luke J. Norbury
- Department of Biosciences and Food Technology, School of Science, STEM College, RMIT University, Bundoora, VIC 3083, Australia;
| |
Collapse
|
18
|
Chen M, Yao L, Zhou L, Yang P, Zou W, Xu L, Li S, Peng H. Toxoplasma gondii
ROP18
I
inhibits host innate immunity through cGAS‐STING signaling. FASEB J 2022; 36:e22171. [DOI: 10.1096/fj.202101347r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/19/2021] [Accepted: 01/10/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Min Chen
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health Southern Medical University Guangzhou P. R. China
| | - Lijie Yao
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health Southern Medical University Guangzhou P. R. China
| | - Lijuan Zhou
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health Southern Medical University Guangzhou P. R. China
| | - Pei Yang
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health Southern Medical University Guangzhou P. R. China
| | - Weihao Zou
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health Southern Medical University Guangzhou P. R. China
| | - Liqing Xu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health Southern Medical University Guangzhou P. R. China
| | - Shengmin Li
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health Southern Medical University Guangzhou P. R. China
| | - Hongjuan Peng
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health Southern Medical University Guangzhou P. R. China
| |
Collapse
|
19
|
Nayeri T, Sarvi S, Daryani A. Toxoplasmosis: Targeting neurotransmitter systems in psychiatric disorders. Metab Brain Dis 2022; 37:123-146. [PMID: 34476718 DOI: 10.1007/s11011-021-00824-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/14/2021] [Indexed: 12/30/2022]
Abstract
The most common form of the disease caused by Toxoplasma gondii (T. gondii) is latent toxoplasmosis due to the formation of tissue cysts in various organs, such as the brain. Latent toxoplasmosis is probably a risk factor in the development of some neuropsychiatric disorders. Behavioral changes after infection are caused by the host immune response, manipulation by the parasite, central nervous system (CNS) inflammation, as well as changes in hormonal and neuromodulator relationships. The present review focused on the exact mechanisms of T. gondii effect on the alteration of behavior and neurotransmitter levels, their catabolites and metabolites, as well as the interaction between immune responses and this parasite in the etiopathogenesis of psychiatric disorders. The dysfunction of neurotransmitters in the neural transmission is associated with several neuropsychiatric disorders. However, further intensive studies are required to determine the effect of this parasite on altering the level of neurotransmitters and the role of neurotransmitters in the etiology of host behavioral changes.
Collapse
Affiliation(s)
- Tooran Nayeri
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahabeddin Sarvi
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ahmad Daryani
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
20
|
Sana M, Rashid M, Rashid I, Akbar H, Gomez-Marin JE, Dimier-Poisson I. Immune response against toxoplasmosis-some recent updates RH: Toxoplasma gondii immune response. Int J Immunopathol Pharmacol 2022; 36:3946320221078436. [PMID: 35227108 PMCID: PMC8891885 DOI: 10.1177/03946320221078436] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
AIMS Cytokines, soluble mediators of immunity, are key factors of the innate and adaptive immune system. They are secreted from and interact with various types of immune cells to manipulate host body's immune cell physiology for a counter-attack on the foreign body. A study was designed to explore the mechanism of Toxoplasma gondii (T. gondii) resistance from host immune response. METHODS AND RESULTS The published data on aspect of host (murine and human) immune response against T. gondii was taken from Google scholar and PubMed. Most relevant literature was included in this study. The basic mechanism of immune response starts from the interactions of antigens with host immune cells to trigger the production of cytokines (pro-inflammatory and anti-inflammatory) which then act by forming a cytokinome (network of cytokine). Their secretory equilibrium is essential for endowing resistance to the host against infectious diseases, particularly toxoplasmosis. A narrow balance lying between Th1, Th2, and Th17 cytokines (as demonstrated until now) is essential for the development of resistance against T. gondii as well as for the survival of host. Excessive production of pro-inflammatory cytokines leads to tissue damage resulting in the production of anti-inflammatory cytokines which enhances the proliferation of Toxoplasma. Stress and other infectious diseases (human immunodeficiency virus (HIV)) that weaken the host immunity particularly the cellular component, make the host susceptible to toxoplasmosis especially in pregnant women. CONCLUSION The current review findings state that in vitro harvesting of IL12 from DCs, Np and MΦ upon exposure with T. gondii might be a source for therapeutic use in toxoplasmosis. Current review also suggests that therapeutic interventions leading to up-regulation/supplementation of SOCS-3, IL12, and IFNγ to the infected host could be a solution to sterile immunity against T. gondii infection. This would be of interest particularly in patients passing through immunosuppression owing to any reason like the ones receiving anti-cancer therapy, the ones undergoing immunosuppressive therapy for graft/transplantation, the ones suffering from immunodeficiency virus (HIV) or having AIDS. Another imortant suggestion is to launch the efforts for a vaccine based on GRA6Nt or other similar antigens of T. gondii as a probable tool to destroy tissue cysts.
Collapse
Affiliation(s)
- Madiha Sana
- Department of Parasitology, 66920University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Muhammad Rashid
- Department of Parasitology, Faculty of Veterinary and Animal Sciences, 66920The Islamia University of Bahawalpur, Pakistan
| | - Imran Rashid
- Department of Parasitology, 66920University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Haroon Akbar
- Department of Parasitology, 66920University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Jorge E Gomez-Marin
- Grupo Gepamol, Centro de Investigaciones Biomedicas, Universidad del Quindio, Armenia, CO, South America
| | - Isabelle Dimier-Poisson
- Université de Tours, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Unité mixte de recherche 1282 (UMR1282), Infectiologie et santé publique (ISP), Tours, France
| |
Collapse
|
21
|
Transcriptomic Analysis of the Effects of Chemokine Receptor CXCR3 Deficiency on Immune Responses in the Mouse Brain during Toxoplasma gondii Infection. Microorganisms 2021; 9:microorganisms9112340. [PMID: 34835465 PMCID: PMC8620038 DOI: 10.3390/microorganisms9112340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/06/2021] [Indexed: 11/16/2022] Open
Abstract
The obligate intracellular parasite Toxoplasma gondii infects warm-blooded animals, including humans. We previously revealed through a whole-brain transcriptome analysis that infection with T. gondii in mice causes immune response-associated genes to be upregulated, for instance, chemokines and chemokine receptors such as CXC chemokine receptor 3 (CXCR3) and its ligand CXC chemokine ligand 10 (CXCL10). Here, we describe the effect of CXCR3 on responses against T. gondii infection in the mouse brain. In vivo assays using CXCR3-deficient mice showed that the absence of CXCR3 delayed the normal recovery of body weight and increased the brain parasite burden, suggesting that CXCR3 plays a role in the control of pathology in the brain, the site where chronic infection occurs. Therefore, to further analyze the function of CXCR3 in the brain, we profiled the gene expression patterns of primary astrocytes and microglia by RNA sequencing and subsequent analyses. CXCR3 deficiency impaired the normal upregulation of immune-related genes during T. gondii infection, in astrocytes and microglia alike. Collectively, our results suggest that the immune-related genes upregulated by CXCR3 perform a particular role in controlling pathology when the host is chronically infected with T. gondii in the brain.
Collapse
|
22
|
Snyder LM, Doherty CM, Mercer HL, Denkers EY. Induction of IL-12p40 and type 1 immunity by Toxoplasma gondii in the absence of the TLR-MyD88 signaling cascade. PLoS Pathog 2021; 17:e1009970. [PMID: 34597344 PMCID: PMC8513874 DOI: 10.1371/journal.ppat.1009970] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 10/13/2021] [Accepted: 09/25/2021] [Indexed: 12/18/2022] Open
Abstract
Toxoplasma gondii is an orally acquired pathogen that induces strong IFN-γ based immunity conferring protection but that can also be the cause of immunopathology. The response in mice is driven in part by well-characterized MyD88-dependent signaling pathways. Here we focus on induction of less well understood immune responses that do not involve this Toll-like receptor (TLR)/IL-1 family receptor adaptor molecule, in particular as they occur in the intestinal mucosa. Using eYFP-IL-12p40 reporter mice on an MyD88-/- background, we identified dendritic cells, macrophages, and neutrophils as cellular sources of MyD88-independent IL-12 after peroral T. gondii infection. Infection-induced IL-12 was lower in the absence of MyD88, but was still clearly above noninfected levels. Overall, this carried through to the IFN-γ response, which while generally decreased was still remarkably robust in the absence of MyD88. In the latter mice, IL-12 was strictly required to induce type I immunity. Type 1 and type 3 innate lymphoid cells (ILC), CD4+ T cells, and CD8+ T cells each contributed to the IFN-γ pool. We report that ILC3 were expanded in infected MyD88-/- mice relative to their MyD88+/+ counterparts, suggesting a compensatory response triggered by loss of MyD88. Furthermore, bacterial flagellin and Toxoplasma specific CD4+ T cell populations in the lamina propria expanded in response to infection in both WT and KO mice. Finally, we show that My88-independent IL-12 and T cell mediated IFN-γ production require the presence of the intestinal microbiota. Our results identify MyD88-independent intestinal immune pathways induced by T. gondii including myeloid cell derived IL-12 production, downstream type I immunity and IFN-γ production by ILC1, ILC3, and T lymphocytes. Collectively, our data reveal an underlying network of immune responses that do not involve signaling through MyD88. Toxoplasma gondii is an apicomplexan parasite estimated to infect 30–50% of humans worldwide. The parasite normally establishes latency in brain and muscle tissue marked by persistent asymptomatic infection. T. gondii masterfully strikes a balance between eliciting strong, anti-parasite immunity while also persisting in the host. Although the murine host recognizes Toxoplasma profilin via MyD88 and Toll-like receptors 11/12, humans lack these receptors and MyD88 deficient patients retain resistance to T. gondii infection. Given these observations, it is important to identify MyD88 independent pathways of immunity. Using an oral infection mouse model, we identified cellular sources of IL-12 and IFN-γ, two cytokines that are essential for host resistance to this microbial pathogen. We determined how these responses are impacted by the presence and absence of MyD88 and the intestinal microbiota. Our data demonstrate that T. gondii triggers MyD88-independent innate and adaptive immunity in the intestinal mucosa that requires the presence of intestinal microbes. These pathways may be conserved among species and understanding how they work in rodents will likely help determine how humans recognize and respond to T. gondii infection.
Collapse
Affiliation(s)
- Lindsay M Snyder
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Claire M Doherty
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Heather L Mercer
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Eric Y Denkers
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
| |
Collapse
|
23
|
Zhao Y, Reyes J, Rovira-Diaz E, Fox BA, Bzik DJ, Yap GS. Cutting Edge: CD36 Mediates Phagocyte Tropism and Avirulence of Toxoplasma gondii. THE JOURNAL OF IMMUNOLOGY 2021; 207:1507-1512. [PMID: 34400524 DOI: 10.4049/jimmunol.2100605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 07/13/2021] [Indexed: 11/19/2022]
Abstract
Resistance and tolerance are vital for survivability of the host-pathogen relationship. Virulence during Toxoplasma infection in mice is mediated by parasite kinase-dependent antagonism of IFN-γ-induced host resistance. Whether avirulence requires expression of parasite factors that induce host tolerance mechanisms or is a default status reflecting the absence of resistance-interfering factors is not known. In this study, we present evidence that avirulence in Toxoplasma requires parasite engagement of the scavenger receptor CD36. CD36 promotes macrophage tropism but is dispensable for the development of resistance mechanisms. Instead CD36 is critical for re-establishing tissue homeostasis and survival following the acute phase of infection. The CD36-binding capacity of T. gondii strains is negatively controlled by the virulence factor, ROP18. Thus, the absence of resistance-interfering virulence factors and the presence of tolerance-inducing avirulence factors are both required for long-term host-pathogen survival.
Collapse
Affiliation(s)
- Yanlin Zhao
- Department of Medicine and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ; and
| | - Jojo Reyes
- Department of Medicine and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ; and
| | - Eliezer Rovira-Diaz
- Department of Medicine and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ; and
| | - Barbara A Fox
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - David J Bzik
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - George S Yap
- Department of Medicine and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ; and
| |
Collapse
|
24
|
Bhattacharyya ND, Counoupas C, Daniel L, Zhang G, Cook SJ, Cootes TA, Stifter SA, Bowen DG, Triccas JA, Bertolino P, Britton WJ, Feng CG. TCR Affinity Controls the Dynamics but Not the Functional Specification of the Antimycobacterial CD4 + T Cell Response. THE JOURNAL OF IMMUNOLOGY 2021; 206:2875-2887. [PMID: 34049970 DOI: 10.4049/jimmunol.2001271] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/02/2021] [Indexed: 11/19/2022]
Abstract
The quality of T cell responses depends on the lymphocytes' ability to undergo clonal expansion, acquire effector functions, and traffic to the site of infection. Although TCR signal strength is thought to dominantly shape the T cell response, by using TCR transgenic CD4+ T cells with different peptide:MHC binding affinity, we reveal that TCR affinity does not control Th1 effector function acquisition or the functional output of individual effectors following mycobacterial infection in mice. Rather, TCR affinity calibrates the rate of cell division to synchronize the distinct processes of T cell proliferation, differentiation, and trafficking. By timing cell division-dependent IL-12R expression, TCR affinity controls when T cells become receptive to Th1-imprinting IL-12 signals, determining the emergence and magnitude of the Th1 effector pool. These findings reveal a distinct yet cooperative role for IL-12 and TCR binding affinity in Th1 differentiation and suggest that the temporal activation of clones with different TCR affinity is a major strategy to coordinate immune surveillance against persistent pathogens.
Collapse
Affiliation(s)
- Nayan D Bhattacharyya
- Immunology and Host Defense Group, Department of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia.,Tuberculosis Research Program, Centenary Institute, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Claudio Counoupas
- Microbial Pathogenesis and Immunity Group, Department of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia.,Tuberculosis Research Program, Centenary Institute, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Lina Daniel
- Immunology and Host Defense Group, Department of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia.,Tuberculosis Research Program, Centenary Institute, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Guoliang Zhang
- Immunology and Host Defense Group, Department of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia.,Tuberculosis Research Program, Centenary Institute, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia.,National Clinical Research Center for Infectious Diseases, Guangdong Key Laboratory of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Stuart J Cook
- Immune Imaging Program, Centenary Institute, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Taylor A Cootes
- Immunology and Host Defense Group, Department of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia.,Tuberculosis Research Program, Centenary Institute, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Sebastian A Stifter
- Immunology and Host Defense Group, Department of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia.,Tuberculosis Research Program, Centenary Institute, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - David G Bowen
- Liver Immunology Program, Centenary Institute, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia.,AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia; and
| | - James A Triccas
- Microbial Pathogenesis and Immunity Group, Department of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia.,Tuberculosis Research Program, Centenary Institute, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia.,Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney, Sydney, New South Wales, Australia
| | - Patrick Bertolino
- Liver Immunology Program, Centenary Institute, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia.,AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia; and
| | - Warwick J Britton
- Tuberculosis Research Program, Centenary Institute, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Carl G Feng
- Immunology and Host Defense Group, Department of Infectious Diseases and Immunology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia; .,Tuberculosis Research Program, Centenary Institute, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia.,Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
25
|
Clark JT, Christian DA, Gullicksrud JA, Perry JA, Park J, Jacquet M, Tarrant JC, Radaelli E, Silver J, Hunter CA. IL-33 promotes innate lymphoid cell-dependent IFN-γ production required for innate immunity to Toxoplasma gondii. eLife 2021; 10:e65614. [PMID: 33929319 PMCID: PMC8121546 DOI: 10.7554/elife.65614] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/29/2021] [Indexed: 12/29/2022] Open
Abstract
IL-33 is an alarmin required for resistance to the parasite Toxoplasma gondii, but its role in innate resistance to this organism is unclear. Infection with T. gondii promotes increased stromal cell expression of IL-33, and levels of parasite replication correlate with release of IL-33 in affected tissues. In response to infection, a subset of innate lymphoid cells (ILC) emerges composed of IL-33R+ NK cells and ILC1s. In Rag1-/-mice, where NK cells and ILC1 production of IFN-γ mediate innate resistance to T. gondii, the loss of the IL-33R resulted in reduced ILC responses and increased parasite replication. Furthermore, administration of IL-33 to Rag1-/- mice resulted in a marked decrease in parasite burden, increased production of IFN-γ, and the recruitment and expansion of inflammatory monocytes associated with parasite control. These protective effects of exogenous IL-33 were dependent on endogenous IL-12p40 and the ability of IL-33 to enhance ILC production of IFN-γ. These results highlight that IL-33 synergizes with IL-12 to promote ILC-mediated resistance to T. gondii.
Collapse
Affiliation(s)
- Joseph T Clark
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
| | - David A Christian
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
| | - Jodi A Gullicksrud
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
| | - Joseph A Perry
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
| | - Jeongho Park
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
- Kangwon National University College of Veterinary Medicine and Institute of Veterinary ScienceChuncheonRepublic of Korea
| | - Maxime Jacquet
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
- Liver Immunology, Department of Biomedicine, University Hospital of Basel and University of BaselBaselSwitzerland
| | - James C Tarrant
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
| | - Enrico Radaelli
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
| | - Jonathan Silver
- Department of Respiratory Inflammation and Autoimmunity, AstraZenecaGaithersburgUnited States
| | - Christopher A Hunter
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
| |
Collapse
|
26
|
Ihara F, Nishikawa Y. Toxoplasma gondii manipulates host cell signaling pathways via its secreted effector molecules. Parasitol Int 2021; 83:102368. [PMID: 33905814 DOI: 10.1016/j.parint.2021.102368] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 03/13/2021] [Accepted: 04/07/2021] [Indexed: 01/07/2023]
Abstract
The obligate intracellular parasite Toxoplasma gondii secretes a vast variety of effector molecules from organelles known as rhoptries (ROPs) and dense granules (GRAs). ROP proteins are released into the cytosol of the host cell where they are directed to the cell nucleus or to the parasitophorous vacuole (PV) membrane. ROPs secrete proteins that enable host cell penetration and vacuole formation by the parasites, as well as hijacking host-immune responses. After invading host cells, T. gondii multiplies within a PV that is maintained by the parasite proteins secreted from GRAs. Most GRA proteins remain within the PV, but some are known to access the host cytosol across the PV membrane, and a few are able to traffic into the host-cell nucleus. These effectors bind to host cell proteins and affect host cell signaling pathways to favor the parasite. Studies on host-pathogen interactions have identified many infection-altered host signal transductions. Notably, the relationship between individual parasite effector molecules and the specific targeting of host-signaling pathways is being elucidated through the advent of forward and reverse genetic strategies. Understanding the complex nature of the host-pathogen interactions underlying how the host-signaling pathway is manipulated by parasite effectors may lead to new molecular biological knowledge and novel therapeutic methods for toxoplasmosis. In this review, we discuss how T. gondii modulates cell signaling pathways in the host to favor its survival.
Collapse
Affiliation(s)
- Fumiaki Ihara
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.
| | - Yoshifumi Nishikawa
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan.
| |
Collapse
|
27
|
Li S, Zhang N, Liu S, Li J, Liu L, Wang X, Li X, Gong P, Zhang X. Protective Immunity Against Neospora caninum Infection Induced by 14-3-3 Protein in Mice. Front Vet Sci 2021; 8:638173. [PMID: 33748214 PMCID: PMC7965954 DOI: 10.3389/fvets.2021.638173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 02/03/2021] [Indexed: 11/16/2022] Open
Abstract
Neospora caninum is an apicomplexan parasite that infects many mammals and remains a threatening disease worldwide because of the lack of effective drugs and vaccines. Our previous studies demonstrated that N. caninum 14-3-3 protein (Nc14-3-3), which is included in N. caninum extracellular vesicles (NEVs), can induce effective immune responses and stimulate cytokine expression in mouse peritoneal macrophages. However, whether Nc14-3-3 has a protective effect and its mechanisms are poorly understood. Here, we evaluated the immune responses and protective effects of Nc14-3-3 against exposure to 2 × 107 Nc-1 tachyzoites. Antibody (IgG, IgGl, and IgG2a) levels and Th1-type (IFN-γ and IL-12) and Th2-type (IL-4 and IL-10) cytokines in mouse serum, survival rates, survival times, and parasite burdens were detected. In the present study, the immunostimulatory effect of Nc14-3-3 was confirmed, as it triggered Th1-type cytokine (IFN-γ and IL-12) production in mouse serum 2 weeks after the final immunization. Moreover, the immunization of C57BL/6 mice with Nc14-3-3 induced high IgG antibody levels and significant increases in CD8+ T lymphocytes in the spleens of mice, indicating that the cellular immune response was significantly stimulated. Mouse survival rates and times were significantly prolonged after immunization; the survival rates were 40% for Nc14-3-3 immunization and 60% for NEV immunization, while mice that received GST, PBS, or blank control all died at 13, 9, or 8 days, respectively, after intraperitoneal N. caninum challenge. In addition, qPCR analysis indicated that there was a reduced parasite burden and diminished pathological changes in the mice immunized with Nc14-3-3. Our data demonstrate that vaccination of mice with Nc14-3-3 elicits both cellular and humoral immune responses and provides partial protection against acute neosporosis. Thus, Nc14-3-3 could be an effective antigen candidate for vaccine development for neosporosis.
Collapse
Affiliation(s)
- Shan Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China.,Department of Social Medicine and Public Health, School of Basic Medicine, Jiujiang University, Jiujiang, China
| | - Nan Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shaoxiong Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jianhua Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Li Liu
- College of Basic Medicine, Jilin University, Changchun, China
| | - Xiaocen Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xin Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Pengtao Gong
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xichen Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
28
|
Snyder LM, Denkers EY. From Initiators to Effectors: Roadmap Through the Intestine During Encounter of Toxoplasma gondii With the Mucosal Immune System. Front Cell Infect Microbiol 2021; 10:614701. [PMID: 33505924 PMCID: PMC7829212 DOI: 10.3389/fcimb.2020.614701] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/24/2020] [Indexed: 12/26/2022] Open
Abstract
The gastrointestinal tract is a major portal of entry for many pathogens, including the protozoan parasite Toxoplasma gondii. Billions of people worldwide have acquired T. gondii at some point in their life, and for the vast majority this has led to latent infection in the central nervous system. The first line of host defense against Toxoplasma is located within the intestinal mucosa. Appropriate coordination of responses by the intestinal epithelium, intraepithelial lymphocytes, and lamina propria cells results in an inflammatory response that controls acute infection. Under some conditions, infection elicits bacterial dysbiosis and immune-mediated tissue damage in the intestine. Here, we discuss the complex interactions between the microbiota, the epithelium, as well as innate and adaptive immune cells in the intestinal mucosa that induce protective immunity, and that sometimes switch to inflammatory pathology as T. gondii encounters tissues of the gut.
Collapse
Affiliation(s)
- Lindsay M Snyder
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, NM, United States
| | - Eric Y Denkers
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
29
|
Mévélec MN, Lakhrif Z, Dimier-Poisson I. Key Limitations and New Insights Into the Toxoplasma gondii Parasite Stage Switching for Future Vaccine Development in Human, Livestock, and Cats. Front Cell Infect Microbiol 2020; 10:607198. [PMID: 33324583 PMCID: PMC7724089 DOI: 10.3389/fcimb.2020.607198] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/19/2020] [Indexed: 12/19/2022] Open
Abstract
Toxoplasmosis is a parasitic disease affecting human, livestock and cat. Prophylactic strategies would be ideal to prevent infection. In a One Health vaccination approach, the objectives would be the prevention of congenital disease in both women and livestock, prevention/reduction of T. gondii tissue cysts in food-producing animals; and oocyst shedding in cats. Over the last few years, an explosion of strategies for vaccine development, especially due to the development of genetic-engineering technologies has emerged. The field of vaccinology has been exploring safer vaccines by the generation of recombinant immunogenic proteins, naked DNA vaccines, and viral/bacterial recombinants vectors. These strategies based on single- or few antigens, are less efficacious than recombinant live-attenuated, mostly tachyzoite T. gondii vaccine candidates. Reflections on the development of an anti-Toxoplasma vaccine must focus not only on the appropriate route of administration, capable of inducing efficient immune response, but also on the choice of the antigen (s) of interest and the associated delivery systems. To answer these questions, the choice of the animal model is essential. If mice helped in understanding the protection mechanisms, the data obtained cannot be directly transposed to humans, livestock and cats. Moreover, effectiveness vaccines should elicit strong and protective humoral and cellular immune responses at both local and systemic levels against the different stages of the parasite. Finally, challenge protocols should use the oral route, major natural route of infection, either by feeding tissue cysts or oocysts from different T. gondii strains. Effective Toxoplasma vaccines depend on our understanding of the (1) protective host immune response during T. gondii invasion and infection in the different hosts, (2) manipulation and modulation of host immune response to ensure survival of the parasites able to evade and subvert host immunity, (3) molecular mechanisms that define specific stage development. This review presents an overview of the key limitations for the development of an effective vaccine and highlights the contributions made by recent studies on the mechanisms behind stage switching to offer interesting perspectives for vaccine development.
Collapse
Affiliation(s)
| | - Zineb Lakhrif
- Team BioMAP, Université de Tours, INRAE, ISP, Tours, France
| | | |
Collapse
|
30
|
Rodríguez-Serrato MA, Salinas-Carmona MC, Limón-Flores AY. Immune response to Leishmania mexicana: the host-parasite relationship. Pathog Dis 2020; 78:5917983. [PMID: 33016312 DOI: 10.1093/femspd/ftaa060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/01/2020] [Indexed: 11/12/2022] Open
Abstract
Leishmaniosis is currently considered a serious public health problem and it is listed as a neglected tropical disease by World Health Organization (WHO). Despite the efforts of the scientific community, it has not been possible to develop an effective vaccine. Current treatment consists of antimonials that is expensive and can cause adverse effects. It is essential to fully understand the immunopathogenesis of the disease to develop new strategies to prevent, treat and eradicate the disease. Studies on animal models have shown a new paradigm in the resolution or establishment of infection by Leishmania mexicana where a wide range of cytokines, antibodies and cells are involved. In recent years, the possibility of a new therapy with monoclonal antibodies has been considered, where isotype, specificity and concentration are critical for effective therapy. Would be better to create/generate a vaccine to induce host protection or produce passive immunization with engineering monoclonal antibodies to a defined antigen? This review provides an overview that includes the current known information on the immune response that are involved in the complex host-parasite relationship infection caused by L. mexicana.
Collapse
Affiliation(s)
- Mayra A Rodríguez-Serrato
- Universidad Autónoma de Nuevo León, Facultad de Medicina y Hospital Universitario Dr. Jose Eleuterio González, Servicio y Departamento de Inmunología, Av. Madero y Av. Gonzalitos s/n, Colonia Mitras Centro, Monterrey, Nuevo León, México
| | - Mario C Salinas-Carmona
- Universidad Autónoma de Nuevo León, Facultad de Medicina y Hospital Universitario Dr. Jose Eleuterio González, Servicio y Departamento de Inmunología, Av. Madero y Av. Gonzalitos s/n, Colonia Mitras Centro, Monterrey, Nuevo León, México
| | - Alberto Yairh Limón-Flores
- Universidad Autónoma de Nuevo León, Facultad de Medicina y Hospital Universitario Dr. Jose Eleuterio González, Servicio y Departamento de Inmunología, Av. Madero y Av. Gonzalitos s/n, Colonia Mitras Centro, Monterrey, Nuevo León, México
| |
Collapse
|
31
|
Song Y, Song L, Wan X, Shen B, Fang R, Hu M, Zhao J, Zhou Y. A Comparison of Transcriptional Diversity of Swine Macrophages Infected With TgHB1 Strain of Toxoplasma gondii Isolated in China. Front Cell Infect Microbiol 2020; 10:526876. [PMID: 33102248 PMCID: PMC7546811 DOI: 10.3389/fcimb.2020.526876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 08/25/2020] [Indexed: 01/08/2023] Open
Abstract
Toxoplasma gondii is an apicomplexan parasite infecting human and animals, causing huge public health concerns and economic losses. Swine alveolar macrophage plays an important role in controlling T. gondii infection. However, the mechanism by which macrophages infected with T. gondii function in the immunity to the infection is unclear, especially for local isolates such as TgHB1 isolated in China. RNA-seq as a valuable tool was applied to simultaneously analyze transcriptional changes of pig alveolar macrophages infected with TgRH (typeI), TgME49 (typeII) or TgHB1 at different time points post infection (6, 12, and 24 h). Paired-end clean reads were aligned to the Sscrofa10.2 pig genome and T. gondii ME49 genome. The differentially expressed genes of macrophages and T. gondii were enriched through Gene Ontology and Kyoto Encyclopedia of Genes and Genomes, respectively. Compared to the TgRH and TgME49 infection groups, 307 down-regulated macrophage genes (mainly enriched for development and metabolism) and 419 up-regulated genes (mainly enriched for immune pathways) were uniquely expressed in the TgHB1 infection group. Additionally, 557 down-regulated and 674 up-regulated T. gondii genes (mainly enriched in metabolism and biosynthesis) were uniquely expressed in the TgHB1 infection group. For validation purposes, some of the differentially expressed genes of macrophages involved in immune-related signaling pathways were used for further analysis via real time quantitative reverse-transcription polymerase-chain reaction (qRT-PCR). This work provides important insights into the temporal immune responses of swine alveolar macrophages to infection by the strain TgHB1 isolated from China, and is helpful for better understanding of the T. gondii genotype-associated activation of macrophages during early phase of the infection.
Collapse
Affiliation(s)
- Yongle Song
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Lindong Song
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Xiaoting Wan
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Bang Shen
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Rui Fang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Min Hu
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Junlong Zhao
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Yanqin Zhou
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
32
|
Foroutan M, Barati M, Ghaffarifar F. Enhancing immune responses by a novel multi-epitope ROP8 DNA vaccine plus interleukin-12 plasmid as a genetic adjuvant against acute Toxoplasma gondii infection in BALB/c mice. Microb Pathog 2020; 147:104435. [PMID: 32768514 DOI: 10.1016/j.micpath.2020.104435] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/27/2020] [Accepted: 08/02/2020] [Indexed: 01/24/2023]
Abstract
BACKGROUND Toxoplasmosis is a widespread zoonotic infection, caused by an obligate intracellular protozoan. The infection is often asymptomatic in immunocompetent individuals, although in persons with impaired immune system may lead to severe and progressive complications. Constant attempts of scientists have made valuable findings in the development of Toxoplasma gondii (T. gondii) candidate vaccines. However, an effective vaccine has not been successfully developed yet. In the current study, we tested the co-delivery of a novel multi-epitope pcROP8 DNA vaccine with a plasmid encoding IL-12 (pcIL-12) to assess the immune responses in BALB/c mice against acute T. gondii infection. METHODS BALB/c mice were immunized on days 0, 21, and 42. The immune responses of both vaccinated and control groups were evaluated using cytokine and antibody measurements, lymphocyte proliferation assay, and survival time. RESULTS The findings demonstrated that immunization with multi-epitope pcROP8 significantly enhanced the level of anti-T. gondii antibodies, TH1-type cellular immune responses, lymphocyte proliferation, and prolonged survival time, compared to control groups (P < 0.05). Furthermore, the use of pcIL-12 as a genetic adjuvant led to enhancements of the above-mentioned immune responses in BALB/c mice (P < 0.05). CONCLUSIONS The co-administration of pcIL-12 with multi-epitope pcROP8 vaccine, could successfully enhance the level of protection. Thus, this immunization regimen may represent an effective vaccine strategy against acute T. gondii infection.
Collapse
Affiliation(s)
- Masoud Foroutan
- Infectious Diseases Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Mohammad Barati
- Infectious Diseases Research Center, AJA University of Medical Sciences, Tehran, Iran.
| | - Fatemeh Ghaffarifar
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
33
|
Tussiwand R, Behnke MS, Kretzer NM, Grajales-Reyes GE, Murphy TL, Schreiber RD, Murphy KM, Sibley LD. An Important Role for CD4 + T Cells in Adaptive Immunity to Toxoplasma gondii in Mice Lacking the Transcription Factor Batf3. mSphere 2020; 5:e00634-20. [PMID: 32669460 PMCID: PMC7364223 DOI: 10.1128/msphere.00634-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 07/01/2020] [Indexed: 11/20/2022] Open
Abstract
Immunity to Toxoplasma gondii at early stages of infection in C57BL/6 mice depends on gamma interferon (IFN-γ) production by NK cells, while at later stages it is primarily mediated by CD8 T cells. We decided to explore the requirement for CD4 T cells during T. gondii infection in Batf3-/- mice, which lack CD8α+ dendritic cells (DCs) that are necessary for cross-presentation of cell-associated antigens to CD8 T cells. We show that in this immunodeficient background on a BALB/c background, CD4 T cells become important effector cells and are able to protect Batf3-/- mice from infection with the avirulent strain RHΔku80Δrop5 Independently of the initial NK cell activation, CD4 T cells in wild-type and Batf3-/- mice were the major source of IFN-γ. Importantly, memory CD4 T cells were sufficient to provide protective immunity following transfer into Batf3-/- mice and secondary challenge with the virulent RHΔku80 strain. Collectively, these results show that under situations where CD8 cell responses are impaired, CD4 T cells provide an important alternative immune response to T. gondiiIMPORTANCEToxoplasma gondii is a widespread parasite of animals that causes zoonotic infections in humans. Although healthy individuals generally control the infection with only moderate symptoms, it causes serious illness in newborns and those with compromised immune systems such as HIV-infected AIDS patients. Because rodents are natural hosts for T. gondii, laboratory mice provide an excellent model for studying immune responses. Here, we used a combination of an attenuated mutant strain of the parasite that effectively vaccinates mice, with a defect in a transcriptional factor that impairs a critical subset of dendritic cells, to studying the immune response to infection. The findings reveal that in BALB/c mice, CD4 memory T cells play a dominant role in producing IFN-γ needed to control chronic infection. Hence, BALB/c mice may provide a more appropriate model for declining immunity seen in HIV-AIDS patients where loss of CD4 cells is associated with emergence of opportunistic infections.
Collapse
Affiliation(s)
- Roxane Tussiwand
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Michael S Behnke
- Department of Molecular Microbiology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Nicole M Kretzer
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Gary E Grajales-Reyes
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Theresa L Murphy
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Robert D Schreiber
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
- Howard Hughes Medical Institute, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - L David Sibley
- Department of Molecular Microbiology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
34
|
Zhao XY, Ewald SE. The molecular biology and immune control of chronic Toxoplasma gondii infection. J Clin Invest 2020; 130:3370-3380. [PMID: 32609097 PMCID: PMC7324197 DOI: 10.1172/jci136226] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Toxoplasma gondii is an incredibly successful parasite owing in part to its ability to persist within cells for the life of the host. Remarkably, at least 350 host species of T. gondii have been described to date, and it is estimated that 30% of the global human population is chronically infected. The importance of T. gondii in human health was made clear with the first reports of congenital toxoplasmosis in the 1940s. However, the AIDS crisis in the 1980s revealed the prevalence of chronic infection, as patients presented with reactivated chronic toxoplasmosis, underscoring the importance of an intact immune system for parasite control. In the last 40 years, there has been tremendous progress toward understanding the biology of T. gondii infection using rodent models, human cell experimental systems, and clinical data. However, there are still major holes in our understanding of T. gondii biology, including the genes controlling parasite development, the mechanisms of cell-intrinsic immunity to T. gondii in the brain and muscle, and the long-term effects of infection on host homeostasis. The need to better understand the biology of chronic infection is underscored by the recent rise in ocular disease associated with emerging haplotypes of T. gondii and our lack of effective treatments to sterilize chronic infection. This Review discusses the cell types and molecular mediators, both host and parasite, that facilitate persistent T. gondii infection. We highlight the consequences of chronic infection for tissue-specific pathology and identify open questions in this area of host-Toxoplasma interactions.
Collapse
|
35
|
Cui J, Shen B. Transcriptomic analyses reveal distinct response of porcine macrophages to Toxoplasma gondii infection. Parasitol Res 2020; 119:1819-1828. [PMID: 32399721 DOI: 10.1007/s00436-020-06677-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/24/2020] [Indexed: 02/08/2023]
Abstract
Toxoplasma gondii is an obligate protozoan parasite infecting diverse hosts. Studies have demonstrated that different hosts respond differently to Toxoplasma infection. Pigs are among the most susceptible hosts of T. gondii, but the host-pathogen interactions that shape the outcome of infection in pigs are completely unknown. Here, we used dual RNA-seq to profile the transcriptomic changes of porcine alveolar macrophages (PAMs) upon Toxoplasma infection. Our results indicated that PAMs initiated different responses to Toxoplasma infection compared with mouse macrophages. First, although infected PAMs upregulated numerous pro-inflammatory factors, IL-12, which plays critical roles in IL-12~IFN-γ-mediated immunity against Toxoplasma infection in mice, was found unchanged during PAM infection. Second, the gene encoding iNOS that is responsible for nitric oxide (NO) production was also not induced in infected PAMs. Consistently, there was no NO level change in PAMs after infection. Third, it seems like Toxoplasma infection inhibited apoptosis in PAMs. On the parasite side, the most obvious change is the upregulation of genes involved in metabolism and macromolecule synthesis, such as the type II fatty acid synthesis in the apicoplast. Together, these results revealed distinct responses of PAMs to Toxoplasma infection and provide novel insights into Toxoplasma-pig interactions.
Collapse
Affiliation(s)
- Jianmin Cui
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.,Key Laboratory of Preventive Medicine in Hubei Province, Huazhong Agricultural University, Wuhan, 430070, China
| | - Bang Shen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China. .,Key Laboratory of Preventive Medicine in Hubei Province, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
36
|
Mercer HL, Snyder LM, Doherty CM, Fox BA, Bzik DJ, Denkers EY. Toxoplasma gondii dense granule protein GRA24 drives MyD88-independent p38 MAPK activation, IL-12 production and induction of protective immunity. PLoS Pathog 2020; 16:e1008572. [PMID: 32413093 PMCID: PMC7255617 DOI: 10.1371/journal.ppat.1008572] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/28/2020] [Accepted: 04/23/2020] [Indexed: 11/19/2022] Open
Abstract
The apicomplexan Toxoplasma gondii induces strong protective immunity dependent upon recognition by Toll-like receptors (TLR)11 and 12 operating in conjunction with MyD88 in the murine host. However, TLR11 and 12 proteins are not present in humans, inspiring us to investigate MyD88-independent pathways of resistance. Using bicistronic IL-12-YFP reporter mice on MyD88+/+ and MyD88-/- genetic backgrounds, we show that CD11c+MHCII+F4/80- dendritic cells, F4/80+ macrophages, and Ly6G+ neutrophils were the dominant cellular sources of IL-12 in both wild type and MyD88 deficient mice after parasite challenge. Parasite dense granule protein GRA24 induces p38 MAPK activation and subsequent IL-12 production in host macrophages. We show that Toxoplasma triggers an early and late p38 MAPK phosphorylation response in MyD88+/+ and MyD88-/- bone marrow-derived macrophages. Using the uracil auxotrophic Type I T. gondii strain cps1-1, we demonstrate that the late response does not require active parasite proliferation, but strictly depends upon GRA24. By i. p. inoculation with cps1-1 and cps1-1:Δgra24, we identified unique subsets of chemokines and cytokines that were up and downregulated by GRA24. Finally, we demonstrate that cps1-1 triggers a strong host-protective GRA24-dependent Th1 response in the absence of MyD88. Our data identify GRA24 as a major mediator of p38 MAPK activation, IL-12 induction and protective immunity that operates independently of the TLR/MyD88 cascade.
Collapse
Affiliation(s)
- Heather L. Mercer
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Lindsay M. Snyder
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Claire M. Doherty
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Barbara A. Fox
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - David J. Bzik
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Eric Y. Denkers
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
| |
Collapse
|
37
|
Wang JL, Liang QL, Li TT, He JJ, Bai MJ, Cao XZ, Elsheikha HM, Zhu XQ. Toxoplasma gondii tkl1 Deletion Mutant Is a Promising Vaccine against Acute, Chronic, and Congenital Toxoplasmosis in Mice. THE JOURNAL OF IMMUNOLOGY 2020; 204:1562-1570. [PMID: 31996457 DOI: 10.4049/jimmunol.1900410] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 01/01/2020] [Indexed: 12/11/2022]
Abstract
In this study, we generated a tkl1 deletion mutant in the Toxoplasma gondii type 1 RH (RHΔtkl1) strain and tested the protective efficacies of vaccination using RHΔtkl1 tachyzoites against acute, chronic, and congenital T. gondii infections in Kunming mice. Mice vaccinated with RHΔtkl1 mounted a strong humoral and cellular response as shown by elevated levels of anti-T. gondii-specific IgG, IL-2, IL-12, IFN-γ, and IL-10. All RHΔtkl1-vaccinated mice survived a lethal challenge with 1 × 103 tachyzoites of type 1 RH or ToxoDB#9 (PYS or TgC7) strain as well as 100 cysts or oocysts of Prugniuad strain. All mock-vaccinated plus infected mice have died. Vaccination also protected against cyst- or oocyst-caused chronic infection, reduced vertical transmission caused by oocysts, increased litter size, and maintained body weight of pups born to dams challenged with 10 oocysts on day 5 of gestation. In contrast, all mock-vaccinated plus oocysts-infected dams had aborted, and no fetus has survived. Vaccinated dams remained healthy postinfection, and their brain cyst burden was significantly reduced compared with mock-vaccinated dams infected with oocysts. In vivo depletion of CD4+ T cells, CD8+ T cells, and B cells revealed that CD8+ T cells are involved in the protection of mice against T. gondii infection. Additionally, adoptive transfer of CD8+ T cells from RHΔtkl1-vaccinated mice significantly enhanced the survival of naive mice infected with the pathogenic strain. Together, these data reaffirm the importance of CD8+ T cell responses in future vaccine design for toxoplasmosis and present T. gondii tkl1 gene as a promising vaccine candidate.
Collapse
Affiliation(s)
- Jin-Lei Wang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, People's Republic of China; and
| | - Qin-Li Liang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, People's Republic of China; and
| | - Ting-Ting Li
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, People's Republic of China; and
| | - Jun-Jun He
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, People's Republic of China; and
| | - Meng-Jie Bai
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, People's Republic of China; and
| | - Xue-Zhen Cao
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, People's Republic of China; and
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Loughborough LE12 5RD, United Kingdom
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, People's Republic of China; and
| |
Collapse
|
38
|
Park J, DeLong JH, Knox JJ, Konradt C, Wojno EDT, Hunter CA. Impact of Interleukin-27p28 on T and B Cell Responses during Toxoplasmosis. Infect Immun 2019; 87:e00455-19. [PMID: 31548322 PMCID: PMC6867838 DOI: 10.1128/iai.00455-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/18/2019] [Indexed: 11/20/2022] Open
Abstract
Interleukin-27 (IL-27) is a heterodimeric cytokine composed of the subunits IL-27p28 and EBi3, and while the IL-27 heterodimer influences T cell activities, there is evidence that IL-27p28 can have EBi3-independent activities; however, their relevance to infection is unclear. Therefore, the studies presented here compared how IL-27p28 transgenics and IL-27p28-/- mice responded to the intracellular parasite Toxoplasma gondii While the loss of IL-27p28 and its overexpression both result in increased susceptibility to T. gondii, the basis for this phenotype reveals distinct roles for IL-27p28. As a component of IL-27, IL-27p28 is critical to limit infection-induced T cell-mediated pathology, whereas the ectopic expression of IL-27p28 reduced the effector T cell population and had a major inhibitory effect on parasite-specific antibody titers and a failure to control parasite replication in the central nervous system. Indeed, transfer of immune serum to infected IL-27p28 transgenics resulted in reduced parasite burden and pathology. Thus, IL-27p28, independent of its role as a component of IL-27, can act as a negative regulator of humoral and cellular responses during toxoplasmosis.
Collapse
Affiliation(s)
- Jeongho Park
- University of Pennsylvania, School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Jonathan H DeLong
- University of Pennsylvania, School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - James J Knox
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Christoph Konradt
- University of Pennsylvania, School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Elia D Tait Wojno
- University of Washington, Department of Immunology, Seattle, Washington, USA
| | - Christopher A Hunter
- University of Pennsylvania, School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
39
|
Ivanova DL, Mundhenke TM, Gigley JP. The IL-12- and IL-23-Dependent NK Cell Response Is Essential for Protective Immunity against Secondary Toxoplasma gondii Infection. THE JOURNAL OF IMMUNOLOGY 2019; 203:2944-2958. [PMID: 31604804 DOI: 10.4049/jimmunol.1801525] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 09/17/2019] [Indexed: 12/22/2022]
Abstract
NK cells can develop cell-intrinsic memory-like characteristics. Whether they develop these characteristics during Toxoplasma gondii infection is unknown. We addressed this question and dissected the mechanisms involved in secondary NK cell responses using a vaccine-challenge mouse model of T. gondii infection. NK cells were required for control of and survival after secondary T. gondii infection. NK cells increased in number at the reinfection site and produced IFN-γ. To test if these T. gondii experienced NK cells were intrinsically different from naive NK cells, we performed NK cell adoptive transfer into RAG2/cγ-chain-/- mice, NK cell fate mapping, and RAG1-/- mice vaccine-challenge experiments. Although NK cells contributed to immunity after reinfection, they did not develop cell-intrinsic memory-like characteristics after T. gondii vaccination. The mechanisms required for generating these secondary NK cell responses were investigated. Secondary NK cell responses were CD4+ or CD8+ T cell independent. Although IL-12 alone is required for NK cell IFN-γ production during primary T. gondii infection, in the absence of IL-12 using IL-12p35-/- mice or anti-IL-12p70, secondary NK cell responses were only partially reduced after reinfection. IL-23 depletion with anti-IL-23p19 in vivo also significantly reduced the secondary NK cell response. IL-12 and IL-23 blockade with anti-IL-12p40 treatment completely eliminated secondary NK cell responses. Importantly, blockade of IL-12, IL-23, or both significantly reduced control of parasite reinfection and increased parasite burden. Our results define a previously unknown protective role for NK cells during secondary T. gondii infection that is dependent on IL-12 and IL-23.
Collapse
Affiliation(s)
- Daria L Ivanova
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071
| | | | - Jason P Gigley
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071
| |
Collapse
|
40
|
El-Sherbini MS, Abd El-Aal AA, El-Sherbiny WS, Attia SS, Abdel Aziz IZ, Nasr GM, Salama MS, Badr MS. Toxoplasmosis and abortion: pro- and anti-inflammatory cytokines gene expression of the host immune cells. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2019. [DOI: 10.1186/s43042-019-0006-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
41
|
French T, Düsedau HP, Steffen J, Biswas A, Ahmed N, Hartmann S, Schüler T, Schott BH, Dunay IR. Neuronal impairment following chronic Toxoplasma gondii infection is aggravated by intestinal nematode challenge in an IFN-γ-dependent manner. J Neuroinflammation 2019; 16:159. [PMID: 31352901 PMCID: PMC6661741 DOI: 10.1186/s12974-019-1539-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/09/2019] [Indexed: 02/06/2023] Open
Abstract
Background It has become increasingly evident that the immune and nervous systems are closely intertwined, relying on one another during regular homeostatic conditions. Prolonged states of imbalance between neural and immune homeostasis, such as chronic neuroinflammation, are associated with a higher risk for neural damage. Toxoplasma gondii is a highly successful neurotropic parasite causing persistent subclinical neuroinflammation, which is associated with psychiatric and neurodegenerative disorders. Little is known, however, by what means neuroinflammation and the associated neural impairment can be modulated by peripheral inflammatory processes. Methods Expression of immune and synapse-associated genes was assessed via quantitative real-time PCR to investigate how T. gondii infection-induced chronic neuroinflammation and associated neuronal alterations can be reshaped by a subsequent acute intestinal nematode co-infection. Immune cell subsets were characterized via flow cytometry in the brain of infected mice. Sulfadiazine and interferon-γ-neutralizing antibody were applied to subdue neuroinflammation. Results Neuroinflammation induced by T. gondii infection of mice was associated with increased microglia activation, recruitment of immune cells into the brain exhibiting Th1 effector functions, and enhanced production of Th1 and pro-inflammatory molecules (IFN-γ, iNOS, IL-12, TNF, IL-6, and IL-1β) following co-infection with Heligmosomoides polygyrus. The accelerated cerebral Th1 immune response resulted in enhanced T. gondii removal but exacerbated the inflammation-related decrease of synapse-associated gene expression. Synaptic proteins EAAT2 and GABAAα1, which are involved in the excitation/inhibition balance in the CNS, were affected in particular. These synaptic alterations were partially recovered by reducing neuroinflammation indirectly via antiparasitic treatment and especially by application of IFN-γ-neutralizing antibody. Impaired iNOS expression following IFN-γ neutralization directly affected EAAT2 and GABAAα1 signaling, thus contributing to the microglial regulation of neurons. Besides, reduced CD36, TREM2, and C1qa gene expression points toward inflammation induced synaptic pruning as a fundamental mechanism. Conclusion Our results suggest that neuroimmune responses following chronic T. gondii infection can be modulated by acute enteric nematode co-infection. While consecutive co-infection promotes parasite elimination in the CNS, it also adversely affects gene expression of synaptic proteins, via an IFN-γ-dependent manner. Electronic supplementary material The online version of this article (10.1186/s12974-019-1539-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Timothy French
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Henning Peter Düsedau
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Johannes Steffen
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Aindrila Biswas
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Norus Ahmed
- Department of Veterinary Medicine, Institute of Immunology, Free University Berlin, Berlin, Germany
| | - Susanne Hartmann
- Department of Veterinary Medicine, Institute of Immunology, Free University Berlin, Berlin, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Björn H Schott
- Leibniz Institute of Neurobiology, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany.,Department of Psychiatry and Psychotherapy, University Medicine Göttingen, Göttingen, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Medizinische Fakultät, Otto-von-Guericke-University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany. .,Center for Behavioral Brain Sciences, Magdeburg, Germany.
| |
Collapse
|
42
|
Ehmen HG, Lüder CGK. Long-Term Impact of Toxoplasma gondii Infection on Human Monocytes. Front Cell Infect Microbiol 2019; 9:235. [PMID: 31316920 PMCID: PMC6611340 DOI: 10.3389/fcimb.2019.00235] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 06/14/2019] [Indexed: 12/17/2022] Open
Abstract
Toxoplasma gondii is a prevalent parasite of mammals and birds including up to 30% of humans world-wide. Primary infection of immunocompetent hosts leads to a robust cell-mediated immune response, which controls but does not clear the infection, thus enabling long-term parasite persistence in brain and muscle tissues. Chronic toxoplasmosis in mice is associated with resistance to heterologous pathogens and this has been related to increased numbers of inflammatory monocytes. Here we have analyzed whether chronic T. gondii infection impacts the subset distribution and the phenotype of peripheral human monocytes in vivo and their responses to parasite infection in vitro. CD14+ monocytes from T. gondii-seropositive blood donors expressed significantly less FcγRIII (CD16) than those from seronegative controls, but they did not show a shift in the distribution of classical, intermediate and non-classical monocyte subpopulations. Percentages of CD62L+ and CD64+ monocytes were however decreased and increased, respectively, in chronically infected individuals as compared to naïve controls. Infection of monocyte-enriched PBMCs from both seropositive and seronegative individuals with T. gondii led to an increase of CD14+CD16− classical monocytes and a decrease of CD14+CD16+ double positive monocytes. Remarkably, after in vitro parasite infection, expression of the chemokine receptor CCR2 was severely impaired in monocytes from both, individuals with chronic toxoplasmosis and seronegative controls. In contrast, only monocytes from chronically infected humans but not those from controls dose-dependently up-regulated HLA-DR, DP, DQ expression following in vitro infection. Furthermore, monocyte-enriched PBMCs from seropositive individuals up-regulated IL-12 mRNA more vigorously after in vitro infection than cells from naïve controls. Collectively, our results establish that infection of humans with T. gondii exerts long-term effects on the phenotype and responsiveness of blood monocytes. This may have important implications for innate immune responses to T. gondii and unrelated pathogens.
Collapse
Affiliation(s)
- Hauke G Ehmen
- Institute for Medical Microbiology, University Medical Center Goettingen, Georg-August-University, Goettingen, Germany
| | - Carsten G K Lüder
- Institute for Medical Microbiology, University Medical Center Goettingen, Georg-August-University, Goettingen, Germany
| |
Collapse
|
43
|
Melchor SJ, Ewald SE. Disease Tolerance in Toxoplasma Infection. Front Cell Infect Microbiol 2019; 9:185. [PMID: 31245299 PMCID: PMC6563770 DOI: 10.3389/fcimb.2019.00185] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 05/14/2019] [Indexed: 01/12/2023] Open
Abstract
Toxoplasma gondii is a successful protozoan parasite that cycles between definitive felid hosts and a broad range of intermediate hosts, including rodents and humans. Within intermediate hosts, this obligate intracellular parasite invades the small intestine, inducing an inflammatory response. Toxoplasma infects infiltrating immune cells, using them to spread systemically and reach tissues amenable to chronic infection. An intact immune system is necessary to control life-long chronic infection. Chronic infection is characterized by formation of parasite cysts, which are necessary for survival through the gastrointestinal tract of the next host. Thus, Toxoplasma must evade sterilizing immunity, but still rely on the host's immune response for survival and transmission. To do this, Toxoplasma exploits a central cost-benefit tradeoff in immunity: the need to escalate inflammation for pathogen clearance vs. the need to limit inflammation-induced bystander damage. What are the consequences of sustained inflammation on host biology? Many studies have focused on aspects of the immune response that directly target Toxoplasma growth and survival, commonly referred to as "resistance mechanisms." However, it is becoming clear that a parallel arm of the immune response has evolved to mitigate damage caused by the parasite directly (for example, egress-induced cell death) or bystander damage due to the inflammatory response (for example, reactive nitrogen species, degranulation). These so-called "disease tolerance" mechanisms promote tissue function and host survival without directly targeting the pathogen. Here we review changes to host metabolism, tissue structure, and immune function that point to disease tolerance mechanisms during Toxoplasma infection. We explore the impact tolerance programs have on the health of the host and parasite biology.
Collapse
Affiliation(s)
| | - Sarah E. Ewald
- Department of Microbiology, Immunology and Cancer Biology and the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
44
|
Konstantinovic N, Guegan H, Stäjner T, Belaz S, Robert-Gangneux F. Treatment of toxoplasmosis: Current options and future perspectives. Food Waterborne Parasitol 2019; 15:e00036. [PMID: 32095610 PMCID: PMC7033996 DOI: 10.1016/j.fawpar.2019.e00036] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/24/2019] [Accepted: 01/27/2019] [Indexed: 02/08/2023] Open
Abstract
Toxoplasmosis is a worldwide parasitic disease infecting about one third of humans, with possible severe outcomes in neonates and immunocompromised patients. Despite continuous and successful efforts to improve diagnosis, therapeutic schemes have barely evolved since many years. This article aims at reviewing the main clinical trials and current treatment practices, and at addressing future perspectives in the light of ongoing researches.
Collapse
Affiliation(s)
- Neda Konstantinovic
- National Reference Laboratory for Toxoplasmosis, Institute for Medical Research, University of Belgrade, 11129 Belgrade, Serbia
| | - Hélène Guegan
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset - UMR_S 1085, F-35000 Rennes, France
| | - Tijana Stäjner
- National Reference Laboratory for Toxoplasmosis, Institute for Medical Research, University of Belgrade, 11129 Belgrade, Serbia
| | - Sorya Belaz
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset - UMR_S 1085, F-35000 Rennes, France
| | | |
Collapse
|
45
|
Caspase-8 promotes c-Rel-dependent inflammatory cytokine expression and resistance against Toxoplasma gondii. Proc Natl Acad Sci U S A 2019; 116:11926-11935. [PMID: 31147458 DOI: 10.1073/pnas.1820529116] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Caspase-8 is a key integrator of cell survival and cell death decisions during infection and inflammation. Following engagement of tumor necrosis factor superfamily receptors or certain Toll-like receptors (TLRs), caspase-8 initiates cell-extrinsic apoptosis while inhibiting RIPK3-dependent programmed necrosis. In addition, caspase-8 has an important, albeit less well understood, role in cell-intrinsic inflammatory gene expression. Macrophages lacking caspase-8 or the adaptor FADD have defective inflammatory cytokine expression and inflammasome priming in response to bacterial infection or TLR stimulation. How caspase-8 regulates cytokine gene expression, and whether caspase-8-mediated gene regulation has a physiological role during infection, remain poorly defined. Here we demonstrate that both caspase-8 enzymatic activity and scaffolding functions contribute to inflammatory cytokine gene expression. Caspase-8 enzymatic activity was necessary for maximal expression of Il1b and Il12b, but caspase-8 deficient cells exhibited a further decrease in expression of these genes. Furthermore, the ability of TLR stimuli to induce optimal IκB kinase phosphorylation and nuclear translocation of the nuclear factor kappa light chain enhancer of activated B cells family member c-Rel required caspase activity. Interestingly, overexpression of c-Rel was sufficient to restore expression of IL-12 and IL-1β in caspase-8-deficient cells. Moreover, Ripk3 -/- Casp8 -/- mice were unable to control infection by the intracellular parasite Toxoplasma gondii, which corresponded to defects in monocyte recruitment to the peritoneal cavity, and exogenous IL-12 restored monocyte recruitment and protection of caspase-8-deficient mice during acute toxoplasmosis. These findings provide insight into how caspase-8 controls inflammatory gene expression and identify a critical role for caspase-8 in host defense against eukaryotic pathogens.
Collapse
|
46
|
Tait Wojno ED, Hunter CA, Stumhofer JS. The Immunobiology of the Interleukin-12 Family: Room for Discovery. Immunity 2019; 50:851-870. [PMID: 30995503 PMCID: PMC6472917 DOI: 10.1016/j.immuni.2019.03.011] [Citation(s) in RCA: 332] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/08/2019] [Accepted: 03/14/2019] [Indexed: 12/12/2022]
Abstract
The discovery of interleukin (IL)-6 and its receptor subunits provided a foundation to understand the biology of a group of related cytokines: IL-12, IL-23, and IL-27. These family members utilize shared receptors and cytokine subunits and influence the outcome of cancer, infection, and inflammatory diseases. Consequently, many facets of their biology are being therapeutically targeted. Here, we review the landmark discoveries in this field, the combinatorial biology inherent to this family, and how patient datasets have underscored the critical role of these pathways in human disease. We present significant knowledge gaps, including how similar signals from these cytokines can mediate distinct outcomes, and discuss how a better understanding of the biology of the IL-12 family provides new therapeutic opportunities.
Collapse
Affiliation(s)
- Elia D Tait Wojno
- Baker Institute for Animal Health and Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, 235 Hungerford Hill Rd., Ithaca, NY 14853, USA
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 380 South University Ave., Philadelphia, PA 19104-4539, USA.
| | - Jason S Stumhofer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, 4301 West Markham St., Little Rock, AR 72205, USA.
| |
Collapse
|
47
|
Sánchez-López EF, Corigliano MG, Albarracín RM, Sander VA, Legarralde A, Bengoa-Luoni SA, Clemente M. Plant Hsp90 is a novel adjuvant that elicits a strong humoral and cellular immune response against B- and T-cell epitopes of a Toxoplasma gondii SAG1 peptide. Parasit Vectors 2019; 12:140. [PMID: 30909938 PMCID: PMC6434815 DOI: 10.1186/s13071-019-3362-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 02/26/2019] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The 90-kDa heat-shock protein (Hsp90) from Nicotiana benthamiana (NbHsp90.3) is a promising adjuvant, especially for those vaccines that require a T cell-mediated immune response. Toxoplasma gondii SAG1 is considered one of the most important antigens for the development of effective subunit vaccines. Some epitopes located in the SAG1 C-terminus region have showed a strong humoral and cellular immune response. In the present study, we aimed to assess the efficacy of NbHsp90.3 as carrier/adjuvant of SAG1-derived peptide (SAG1HC) in a T. gondii infection murine model. METHODS In the present study, C57BL/6 mice were intraperitoneal immunized with the NbHsp90.3-SAG1HC fusion protein (NbHsp90.3-SAG1HC group), mature SAG1 (SAG1m group), NbHsp90.3 (NbHsp90.3 group) or PBS buffer 1× (PBS group). The levels of IgG antibodies and the cytokine profile were determined by ELISA. Two weeks after the last immunization, all mice were orally challenged with 20 cysts of T. gondii Me49 strain and the number of brain cysts was determined. In addition, both humoral and cellular immune responses were also evaluated during the acute and chronic phase of T. gondii infection by ELISA. RESULTS The characterization of the immune response generated after vaccination with NbHsp90.3 as an adjuvant showed that NbHsp90.3-SAG1HC-immunized mice produced antibodies that were able to recognize not only rSAG1m but also the native SAG1 present in the total lysate antigen extract (SAG1TLA) from T. gondii tachyzoites, while control groups did not. Furthermore, anti-rSAG1m IgG2a/2b antibodies were significantly induced. In addition, only the spleen cell cultures from NbHsp90.3-SAG1HC-immunized mice showed a significantly increased production of IFN-γ. During the chronic phase of T. gondii infection, the antibodies generated by the infection were unable to detect the recombinant protein, but they did react with TLA extract. In addition, splenocytes from all groups showed a high production of IFN-γ when stimulated with rGRA4, but only those from NbHsp90.3-SAG1HC group stimulated with rSAG1m showed high production of IFN-γ. Finally, NbHsp90.3-SAG1HC-immunized mice exhibited a significant reduction in the cyst load (56%) against T. gondii infection. CONCLUSIONS We demonstrated that NbHsp90.3 enhances the humoral and cell-mediated immune response through a Th1 type cytokine production. Mice vaccinated with NbHsp90.3-SAG1HC exhibited a partial protection against T. gondii infection and it was correlated with the induction of memory immune response. We developed and validated a vaccine formulation which, to our knowledge, for the first time includes the NbHsp90.3 protein covalently fused to a peptide from T. gondii SAG1 protein that contains T- and B-cell epitopes.
Collapse
Affiliation(s)
- Edwin F. Sánchez-López
- Unidad de Biotecnología 6-UB6, IIB-INTECH, CONICET-UNSAM, Intendente Marino Km 8.2, B7130IWA Chascomús, Buenos Aires Province Argentina
| | - Mariana G. Corigliano
- Unidad de Biotecnología 6-UB6, IIB-INTECH, CONICET-UNSAM, Intendente Marino Km 8.2, B7130IWA Chascomús, Buenos Aires Province Argentina
| | - Romina M. Albarracín
- Unidad de Biotecnología 6-UB6, IIB-INTECH, CONICET-UNSAM, Intendente Marino Km 8.2, B7130IWA Chascomús, Buenos Aires Province Argentina
| | - Valeria A. Sander
- Unidad de Biotecnología 6-UB6, IIB-INTECH, CONICET-UNSAM, Intendente Marino Km 8.2, B7130IWA Chascomús, Buenos Aires Province Argentina
| | - Ariel Legarralde
- Unidad de Biotecnología 6-UB6, IIB-INTECH, CONICET-UNSAM, Intendente Marino Km 8.2, B7130IWA Chascomús, Buenos Aires Province Argentina
| | - Sofía A. Bengoa-Luoni
- Unidad de Biotecnología 6-UB6, IIB-INTECH, CONICET-UNSAM, Intendente Marino Km 8.2, B7130IWA Chascomús, Buenos Aires Province Argentina
| | - Marina Clemente
- Unidad de Biotecnología 6-UB6, IIB-INTECH, CONICET-UNSAM, Intendente Marino Km 8.2, B7130IWA Chascomús, Buenos Aires Province Argentina
| |
Collapse
|
48
|
Vijayamahantesh, Vijayalaxmi. Tinkering with targeting nucleotide signaling for control of intracellular Leishmania parasites. Cytokine 2019; 119:129-143. [PMID: 30909149 DOI: 10.1016/j.cyto.2019.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/23/2022]
Abstract
Nucleotides are one of the most primitive extracellular signalling molecules across all phyla and regulate a multitude of responses. The biological effects of extracellular nucleotides/sides are mediated via the specific purinergic receptors present on the cell surface. In mammalian system, adenine nucleotides are the predominant nucleotides found in the extracellular milieu and mediate a constellation of physiological functions. In the context of host-pathogen interaction, extracellular ATP is recognized as a danger signal and potentiates the release of pro-inflammatory mediators from activated immune cells, on the other hand, its breakdown product adenosine exerts potential anti-inflammatory and immunosuppressive actions. Therefore, it is increasingly apparent that the interplay between extracellular ATP/adenosine ratios has a significant role in coordinating the regulation of the immune system in health and diseases. Several pathogens express ectonucleotidases on their surface and exploit the purinergic signalling as one of the mechanisms to modulate the host immune response. Leishmania pathogens are one of the most successful intracellular pathogens which survive within host macrophages and manipulate protective Th1 response into disease promoting Th2 response. In this review, we discuss the regulation of extracellular ATP and adenosine levels, the role of ATP/adenosine counter signalling in regulating the inflammation and immune responses during infection and how Leishmania parasites exploit the purinergic signalling to manipulate host response. We also discuss the challenges and opportunities in targeting purinergic signalling and the future prospects.
Collapse
Affiliation(s)
- Vijayamahantesh
- Department of Biochemistry, Indian Institute of Science (IISc), Bengaluru, Karnataka, India.
| | - Vijayalaxmi
- Department of Zoology, Karnatak University, Dharwad, Karnataka, India
| |
Collapse
|
49
|
O'Brien CA, Batista SJ, Still KM, Harris TH. IL-10 and ICOS Differentially Regulate T Cell Responses in the Brain during Chronic Toxoplasma gondii Infection. THE JOURNAL OF IMMUNOLOGY 2019; 202:1755-1766. [PMID: 30718297 DOI: 10.4049/jimmunol.1801229] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 01/10/2019] [Indexed: 12/28/2022]
Abstract
Control of chronic CNS infection with the parasite Toxoplasma gondii requires ongoing T cell responses in the brain. Immunosuppressive cytokines are also important for preventing lethal immunopathology during chronic infection. To explore the loss of suppressive cytokines exclusively during the chronic phase of infection, we blocked IL-10R in chronically infected mice. Consistent with previous reports, IL-10R blockade led to severe, fatal tissue destruction associated with widespread changes in the inflammatory response, including increased APC activation, expansion of CD4+ T cells, and neutrophil recruitment to the brain. We then sought to identify regulatory mechanisms contributing to IL-10 production, focusing on ICOS, a molecule implicated in IL-10 production. Unexpectedly, ICOS ligand (ICOSL) blockade led to a local expansion of effector T cells in the brain without affecting IL-10 production or APC activation. Instead, we found that ICOSL blockade led to changes in T cells associated with their proliferation and survival. We observed increased expression of IL-2-associated signaling molecules CD25, STAT5 phosphorylation, Ki67, and Bcl-2 in T cells in the brain, along with decreased apoptosis. Interestingly, increases in CD25 and Bcl-2 were not observed following IL-10R blockade. Also, unlike IL-10R blockade, ICOSL blockade led to an expansion of both CD8+ and CD4+ T cells in the brain, with no expansion of peripheral T cells or neutrophil recruitment to the brain and no severe tissue destruction. Overall, these results suggest that IL-10 and ICOS differentially regulate T cell responses in the brain during chronic T. gondii infection.
Collapse
Affiliation(s)
- Carleigh A O'Brien
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA 22908
| | - Samantha J Batista
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA 22908
| | - Katherine M Still
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA 22908
| | - Tajie H Harris
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
50
|
Foroutan M, Ghaffarifar F, Sharifi Z, Dalimi A, Jorjani O. Rhoptry antigens as Toxoplasma gondii vaccine target. Clin Exp Vaccine Res 2019; 8:4-26. [PMID: 30775347 PMCID: PMC6369123 DOI: 10.7774/cevr.2019.8.1.4] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 07/10/2018] [Accepted: 01/14/2019] [Indexed: 01/14/2023] Open
Abstract
Toxoplasmosis is a cosmopolitan zoonotic infection, caused by a unicellular protozoan parasite known as Toxoplasma gondii that belongs to the phylum Apicomplexa. It is estimated that over one-third of the world's population has been exposed and are latently infected with the parasite. In humans, toxoplasmosis is predominantly asymptomatic in immunocompetent persons, while among immunocompromised individuals may be cause severe and progressive complications with poor prognosis. Moreover, seronegative pregnant mothers are other risk groups for acquiring the infection. The life cycle of T. gondii is very complex, indicating the presence of a plurality of antigenic epitopes. Despite of great advances, recognize and construct novel vaccines for prevent and control of toxoplasmosis in both humans and animals is still remains a great challenge for researchers to select potential protein sequences as the ideal antigens. Notably, in several past years, constant efforts of researchers have made considerable advances to elucidate the different aspects of the cell and molecular biology of T. gondii mainly on microneme antigens, dense granule antigens, surface antigens, and rhoptry proteins (ROP). These attempts thereby provided great impetus to the present focus on vaccine development, according to the defined subcellular components of the parasite. Although, currently there is no commercial vaccine for use in humans. Among the main identified T. gondii antigens, ROPs appear as a putative vaccine candidate that are vital for invasion procedure as well as survival within host cells. Overall, it is estimated that they occupy about 1%–30% of the total parasite cell volume. In this review, we have summarized the recent progress of ROP-based vaccine development through various strategies from DNA vaccines, epitope or multi epitope-based vaccines, recombinant protein vaccines to vaccines based on live-attenuated vectors and prime-boost strategies in different mouse models.
Collapse
Affiliation(s)
- Masoud Foroutan
- Abadan School of Medical Sciences, Abadan, Iran.,Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Ghaffarifar
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zohreh Sharifi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Abdolhosein Dalimi
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ogholniaz Jorjani
- Laboratory Science Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|