1
|
Shendi SS, Selim SM, Sharaf SA, Gouda MA, Sallam HM, Sweed DM, Shafey DA. Anti-toxoplasmic effects of celecoxib alone and combined with spiramycin in experimental mice. Acta Trop 2024; 260:107448. [PMID: 39477047 DOI: 10.1016/j.actatropica.2024.107448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/27/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024]
Abstract
Even though toxoplasmosis is a worldwide parasitic disease caused by Toxoplasma gondii (T. gondii), the available drugs used for the treatment of symptomatic toxoplasmosis have multiple drawbacks. So, there is a considerable need to discover new potential therapeutic agents. The current study aimed to assess the effect of celecoxib (CELE) alone or combined with spiramycin against chronic toxoplasmosis in experimentally infected mice. The study documented the reduction rate of T. gondii cysts in brain tissues and ultrastructural changes through transmission electron microscopy after treatment. We also investigated pathological changes in the brain, liver, lung, and spleen, as well as the expression of TGF-β, iNOS, and pSTAT-1 in brain tissues. Other markers for kidney function and serum levels of interleukins 10 and 12 were also assessed. The study reported a reduction rate of T. gondii brain cyst count of 32.9 % after CELE treatment, 71.7 % after spiramycin treatment, and 75.7 % after combined treatment. Furthermore, the CELE and spiramycin combination improved the ultrastructure and histopathology in brain tissues while decreasing TGF-β, iNOS, and pSTAT-1 expression. The combined therapy ameliorated the inflammation of the liver, lung, and spleen, upregulated the IL-12 level, reduced the IL-10 level, and was accompanied by a reduction in creatinine and urea in serum. In conclusion, CELE increased spiramycin therapeutic efficacy, and their combination showed a better response than spiramycin alone. Thus, the CELE combination with spiramycin represents a hopeful therapy against chronic toxoplasmosis.
Collapse
Affiliation(s)
- Sawsan S Shendi
- Department of Clinical and Molecular Parasitology, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Sahar M Selim
- Department of Clinical and Molecular Parasitology, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Soraya A Sharaf
- Department of Clinical and Molecular Parasitology, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Marwa A Gouda
- Department of Clinical and Molecular Parasitology, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Hebatallah M Sallam
- Department of Clinical and Molecular Parasitology, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt.
| | - Dina M Sweed
- Department of Pathology, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Dalia A Shafey
- Department of Clinical and Molecular Parasitology, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| |
Collapse
|
2
|
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:e0145524. [PMID: 39440975 DOI: 10.1128/mbio.01455-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
3
|
Wang K, Espinosa V, Wang Y, Lemenze A, Kumamoto Y, Xue C, Rivera A. Innate cells and STAT1-dependent signals orchestrate vaccine-induced protection against invasive Cryptococcus infection. mBio 2024; 15:e0194424. [PMID: 39324785 PMCID: PMC11481872 DOI: 10.1128/mbio.01944-24] [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: 06/24/2024] [Accepted: 08/22/2024] [Indexed: 09/27/2024] Open
Abstract
Fungal pathogens are underappreciated causes of significant morbidity and mortality worldwide. In previous studies, we determined that a heat-killed, Cryptococcus neoformans fbp1-deficient strain (HK-fbp1) is a potent vaccine candidate. We determined that vaccination with HK-fbp1 confers protective immunity against lethal Cryptococcosis in an interferon γ (IFNγ)-dependent manner. In this study, we set out to uncover cellular sources and relevant targets of the protective effects of IFNγ in response to the HK-fbp1 vaccine. We found that early IFNγ production peaks at day 3 and that monocytes and neutrophils are important sources of this cytokine after vaccination. Neutralization of IFNγ at day 3 results in impaired CCR2+ monocyte recruitment and reduced differentiation into monocyte-derived dendritic cells (Mo-DC). In turn, depletion of CCR2+ cells prior to immunization results in impaired activation of IFNγ-producing CD4 and CD8 T cells. Thus, monocytes are important targets of innate IFNγ and help promote further IFNγ production by lymphocytes. We employed monocyte-fate mapper and conditional STAT1 knockout mice to uncover that STAT1 activation in CD11c+ cells, including alveolar macrophages, Mo-DCs, and monocyte-derived macrophages (Mo-Mac) is essential for HK-fbp1 vaccine-induced protection. Altogether, our aggregate findings suggest critical roles for innate cells as orchestrators of vaccine-induced protection against Cryptococcus infection.IMPORTANCEThe number of patients susceptible to invasive fungal infections across the world continues to rise at an alarming pace yet current antifungal drugs are often inadequate. Immune-based interventions and novel antifungal vaccines hold the promise of significantly improving patient outcomes. In previous studies, we identified a Cryptococcus neoformans mutant strain (Fbp1-deficient) as a potent, heat-inactivated vaccine candidate capable of inducing homologous and heterologous antifungal protection. In this study, we used a combination of methods together with a cohort of conditional knockout mouse strains to interrogate the roles of innate cells in the orchestration of vaccine-induced antifungal protection. We uncovered novel roles for neutrophils and monocytes as coordinators of a STAT1-dependent cascade of responses that mediate vaccine-induced protection against invasive cryptococcosis. This new knowledge will help guide the future development of much-needed antifungal vaccines.
Collapse
Affiliation(s)
- Keyi Wang
- Graduate School of Biomedical Sciences, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Vanessa Espinosa
- Department of Pediatrics and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Yina Wang
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Alexander Lemenze
- Department of Medicine and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Yosuke Kumamoto
- Department of Medicine and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Chaoyang Xue
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Amariliz Rivera
- Graduate School of Biomedical Sciences, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
- Department of Pediatrics and Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| |
Collapse
|
4
|
Aguiar A, Menezes de Brito ASS, Santos AGAD, Watanabe PDS, Cuman RKN, Trevizan AR, de Lima LL, Bersani-Amado CA, Rinaldi JDC, Sant Ana DDMG, Nogueira-Melo GDA. Mastocytosis and intraepithelial lymphocytosis in the ileum and colon characterize chronic Toxoplasma gondii infection in mice. Tissue Cell 2024; 91:102533. [PMID: 39213782 DOI: 10.1016/j.tice.2024.102533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Toxoplasma gondii is the causative agent of toxoplasmosis, a common zoonotic disease affecting vertebrates with high global incidence. For the parasite to disseminate throughout the body, it crosses the intestinal barrier, triggering inflammatory reactions. This study aimed to assess the tissue response in the ileum and colon of mice following chronic infection with T. gondii. Fourteen mice were divided into two groups: the infected group received 1000 T. gondii oocysts via gavage, and after 60 days, the mice were euthanized. The ileum and colon were collected and processed for histological analysis, inflammatory marker measurement and myenteric neuron analysis. Chronic infection resulted in a significant increase in intraepithelial lymphocytes and mast cells, as well as morphometric changes such as increased total intestinal wall thickness of the ileum, crypt depth, collagen fiber area, and a decrease in myeloperoxidase activity, without altering nitric oxide levels. While the number of myenteric neurons remained unchanged, there was an increase in vasoactive intestinal peptide expression. These results suggest persistence intestinal inflammatory stimuli in chronic T. gondii infection.
Collapse
Affiliation(s)
- Aline Aguiar
- Graduate Program in Biosciences and Pathophysiology, State University of Maringá, Maringá, Paraná, Brazil
| | | | | | - Paulo da Silva Watanabe
- Graduate Program in Biosciences and Pathophysiology, State University of Maringá, Maringá, Paraná, Brazil
| | | | - Aline Rosa Trevizan
- Graduate Program in Biosciences and Pathophysiology, State University of Maringá, Maringá, Paraná, Brazil
| | - Lainy Leiny de Lima
- Graduate Program in Biosciences and Pathophysiology, State University of Maringá, Maringá, Paraná, Brazil
| | | | | | | | | |
Collapse
|
5
|
Nasuhidehnavi A, Yap GS. To kill a tachyzoite: assault and battery. Trends Parasitol 2024; 40:449-451. [PMID: 38762372 PMCID: PMC11156527 DOI: 10.1016/j.pt.2024.05.002] [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: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/20/2024]
Abstract
Polymeric guanylate-binding proteins (GBPs) physically dismember the vacuole membrane formed by Toxoplasma gondii while nitric oxide (NO) poisons and inhibits parasite replication within interferon (IFN)-γ activated macrophages. Zhao et al. report a novel mechanism for synergy between these classical microbicidal and microbistatic effectors in cell-autonomous immunity to the intracellular parasites.
Collapse
Affiliation(s)
- Azadeh Nasuhidehnavi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - George S Yap
- Center for Immunity and Inflammation, Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ, USA.
| |
Collapse
|
6
|
Kreimendahl S, Pernas L. Metabolic immunity against microbes. Trends Cell Biol 2024; 34:496-508. [PMID: 38030541 DOI: 10.1016/j.tcb.2023.10.013] [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: 06/09/2023] [Revised: 10/11/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023]
Abstract
Pathogens, including viruses, bacteria, fungi, and parasites, remodel the metabolism of their host to acquire the nutrients they need to proliferate. Thus, host cells are often perceived as mere exploitable nutrient pools during infection. Mounting reports challenge this perception and instead suggest that host cells can actively reprogram their metabolism to the detriment of the microbial invader. In this review, we present metabolic mechanisms that host cells use to defend against pathogens. We highlight the contribution of domesticated microbes to host defenses and discuss examples of host-pathogen arms races that are derived from metabolic conflict.
Collapse
Affiliation(s)
| | - Lena Pernas
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA, USA; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
| |
Collapse
|
7
|
Nayeri T, Sarvi S, Daryani A. Effective factors in the pathogenesis of Toxoplasmagondii. Heliyon 2024; 10:e31558. [PMID: 38818168 PMCID: PMC11137575 DOI: 10.1016/j.heliyon.2024.e31558] [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: 01/03/2023] [Revised: 05/11/2024] [Accepted: 05/17/2024] [Indexed: 06/01/2024] Open
Abstract
Toxoplasma gondii (T. gondii) is a cosmopolitan protozoan parasite in humans and animals. It infects about 30 % of the human population worldwide and causes potentially fatal diseases in immunocompromised hosts and neonates. For this study, five English-language databases (ScienceDirect, ProQuest, Web of Science, PubMed, and Scopus) and the internet search engine Google Scholar were searched. This review was accomplished to draw a global perspective of what is known about the pathogenesis of T. gondii and various factors affecting it. Virulence and immune responses can influence the mechanisms of parasite pathogenesis and these factors are in turn influenced by other factors. In addition to the host's genetic background, the type of Toxoplasma strain, the routes of transmission of infection, the number of passages, and different phases of parasite life affect virulence. The identification of virulence factors of the parasite could provide promising insights into the pathogenesis of this parasite. The results of this study can be an incentive to conduct more intensive research to design and develop new anti-Toxoplasma agents (drugs and vaccines) to treat or prevent this infection. In addition, further studies are needed to better understand the key agents in the pathogenesis of T. gondii.
Collapse
Affiliation(s)
- Tooran Nayeri
- Infectious and Tropical Diseases Research Center, Dezful University of Medical Sciences, Dezful, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahabeddin Sarvi
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ahmad Daryani
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
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: 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: 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
|
Ewald S, Nasuhidehnavi A, Feng TY, Lesani M, McCall LI. The intersection of host in vivo metabolism and immune responses to infection with kinetoplastid and apicomplexan parasites. Microbiol Mol Biol Rev 2024; 88:e0016422. [PMID: 38299836 PMCID: PMC10966954 DOI: 10.1128/mmbr.00164-22] [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: 02/02/2024] Open
Abstract
SUMMARYProtozoan parasite infection dramatically alters host metabolism, driven by immunological demand and parasite manipulation strategies. Immunometabolic checkpoints are often exploited by kinetoplastid and protozoan parasites to establish chronic infection, which can significantly impair host metabolic homeostasis. The recent growth of tools to analyze metabolism is expanding our understanding of these questions. Here, we review and contrast host metabolic alterations that occur in vivo during infection with Leishmania, trypanosomes, Toxoplasma, Plasmodium, and Cryptosporidium. Although genetically divergent, there are commonalities among these pathogens in terms of metabolic needs, induction of the type I immune responses required for clearance, and the potential for sustained host metabolic dysbiosis. Comparing these pathogens provides an opportunity to explore how transmission strategy, nutritional demand, and host cell and tissue tropism drive similarities and unique aspects in host response and infection outcome and to design new strategies to treat disease.
Collapse
Affiliation(s)
- Sarah Ewald
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Azadeh Nasuhidehnavi
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
| | - Tzu-Yu Feng
- Department of Microbiology, Immunology, and Cancer Biology at the Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Mahbobeh Lesani
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, USA
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, Oklahoma, USA
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, USA
| |
Collapse
|
10
|
Lüder CGK. IFNs in host defence and parasite immune evasion during Toxoplasma gondii infections. Front Immunol 2024; 15:1356216. [PMID: 38384452 PMCID: PMC10879624 DOI: 10.3389/fimmu.2024.1356216] [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: 12/15/2023] [Accepted: 01/23/2024] [Indexed: 02/23/2024] Open
Abstract
Interferons (IFNs) are a family of cytokines with diverse functions in host resistance to pathogens and in immune regulation. Type II IFN, i.e. IFN-γ, is widely recognized as a major mediator of resistance to intracellular pathogens, including the protozoan Toxoplasma gondii. More recently, IFN-α/β, i.e. type I IFNs, and IFN-λ (type III IFN) have been identified to also play important roles during T. gondii infections. This parasite is a widespread pathogen of humans and animals, and it is a model organism to study cell-mediated immune responses to intracellular infection. Its success depends, among other factors, on the ability to counteract the IFN system, both at the level of IFN-mediated gene expression and at the level of IFN-regulated effector molecules. Here, I review recent advances in our understanding of the molecular mechanisms underlying IFN-mediated host resistance and immune regulation during T. gondii infections. I also discuss those mechanisms that T. gondii has evolved to efficiently evade IFN-mediated immunity. Knowledge of these fascinating host-parasite interactions and their underlying signalling machineries is crucial for a deeper understanding of the pathogenesis of toxoplasmosis, and it might also identify potential targets of parasite-directed or host-directed supportive therapies to combat the parasite more effectively.
Collapse
Affiliation(s)
- Carsten G. K. Lüder
- Institute for Medical Microbiology and Virology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
11
|
Kovacs MA, Babcock IW, Royo Marco A, Sibley LA, Kelly AG, Harris TH. Vascular Endothelial Growth Factor-C Treatment Enhances Cerebrospinal Fluid Outflow during Toxoplasma gondii Brain Infection but Does Not Improve Cerebral Edema. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:225-237. [PMID: 38065361 PMCID: PMC10835445 DOI: 10.1016/j.ajpath.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 10/02/2023] [Accepted: 11/06/2023] [Indexed: 01/22/2024]
Abstract
Cerebral edema frequently develops in the setting of brain infection and can contribute to elevated intracranial pressure, a medical emergency. How excess fluid is cleared from the brain is not well understood. Previous studies have shown that interstitial fluid is transported out of the brain along perivascular channels that collect into the cerebrospinal fluid (CSF)-filled subarachnoid space. CSF is then removed from the central nervous system through venous and lymphatic routes. The current study tested the hypothesis that increasing lymphatic drainage of CSF would promote clearance of cerebral edema fluid during infection with the neurotropic parasite Toxoplasma gondii. Fluorescent microscopy and magnetic resonance imaging was used to show that C57BL/6 mice develop vasogenic edema 4 to 5 weeks after infection with T. gondii. Tracer experiments were used to evaluate how brain infection affects meningeal lymphatic function, which demonstrated a decreased rate in CSF outflow in T. gondii-infected mice. Next, mice were treated with a vascular endothelial growth factor (VEGF)-C-expressing viral vector, which induced meningeal lymphangiogenesis and improved CSF outflow in chronically infected mice. No difference in cerebral edema was observed between mice that received VEGF-C and those that rececived sham treatment. Therefore, although VEGF-C treatment can improve lymphatic outflow in mice infected with T. gondii, this effect does not lead to increased clearance of edema fluid from the brains of these mice.
Collapse
Affiliation(s)
- Michael A Kovacs
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia
| | - Isaac W Babcock
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia
| | - Ana Royo Marco
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia
| | - Lydia A Sibley
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia
| | - Abigail G Kelly
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia
| | - Tajie H Harris
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
12
|
Okuma H, Saijo-Hamano Y, Yamada H, Sherif AA, Hashizaki E, Sakai N, Kato T, Imasaki T, Kikkawa S, Nitta E, Sasai M, Abe T, Sugihara F, Maniwa Y, Kosako H, Takei K, Standley DM, Yamamoto M, Nitta R. Structural basis of Irgb6 inactivation by Toxoplasma gondii through the phosphorylation of switch I. Genes Cells 2024; 29:17-38. [PMID: 37984375 PMCID: PMC11448365 DOI: 10.1111/gtc.13080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/12/2023] [Accepted: 10/29/2023] [Indexed: 11/22/2023]
Abstract
Irgb6 is a priming immune-related GTPase (IRG) that counteracts Toxoplasma gondii. It is known to be recruited to the low virulent type II T. gondii parasitophorous vacuole (PV), initiating cell-autonomous immunity. However, the molecular mechanism by which immunity-related GTPases become inactivated after the parasite infection remains obscure. Here, we found that Thr95 of Irgb6 is prominently phosphorylated in response to low virulent type II T. gondii infection. We observed that a phosphomimetic T95D mutation in Irgb6 impaired its localization to the PV and exhibited reduced GTPase activity in vitro. Structural analysis unveiled an atypical conformation of nucleotide-free Irgb6-T95D, resulting from a conformational change in the G-domain that allosterically modified the PV membrane-binding interface. In silico docking corroborated the disruption of the physiological membrane binding site. These findings provide novel insights into a T. gondii-induced allosteric inactivation mechanism of Irgb6.
Collapse
Affiliation(s)
- Hiromichi Okuma
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yumiko Saijo-Hamano
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroshi Yamada
- Department of Neuroscience, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Aalaa Alrahman Sherif
- Department of Genome Informatics, Research Institute for Microbial Diseases, Osaka, Japan
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Emi Hashizaki
- Laboratory of Immunoparasitology, Osaka University, Osaka, Japan
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka, Japan
| | | | - Takaaki Kato
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tsuyoshi Imasaki
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Satoshi Kikkawa
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Eriko Nitta
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Miwa Sasai
- Laboratory of Immunoparasitology, Osaka University, Osaka, Japan
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka, Japan
| | - Tadashi Abe
- Department of Neuroscience, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Fuminori Sugihara
- Core Instrumentation Facility, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yoshimasa Maniwa
- Division of Thoracic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Kohji Takei
- Department of Neuroscience, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Daron M Standley
- Department of Genome Informatics, Research Institute for Microbial Diseases, Osaka, Japan
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Masahiro Yamamoto
- Laboratory of Immunoparasitology, Osaka University, Osaka, Japan
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka, Japan
| | - Ryo Nitta
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
13
|
Orchanian SB, Still K, Harris TH, Lodoen MB. Deficiency in astrocyte CCL2 production reduces neuroimmune control of Toxoplasma gondii infection. PLoS Pathog 2024; 20:e1011710. [PMID: 38206985 PMCID: PMC10807779 DOI: 10.1371/journal.ppat.1011710] [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: 02/17/2023] [Revised: 01/24/2024] [Accepted: 09/25/2023] [Indexed: 01/13/2024] Open
Abstract
Toxoplasma gondii is an obligate intracellular parasite that infects one-third of the world's human population and establishes infection in the brain. Cerebral immune cell infiltration is critical for controlling the parasite, but little is known about the molecular cues guiding immune cells to the brain during infection. Activated astrocytes produce CCL2, a chemokine that mediates inflammatory monocyte recruitment to tissues by binding to the CCR2 receptor. We detected elevated CCL2 production in the brains of C57BL/6J mice by 15 days after T. gondii infection. Utilizing confocal microscopy and intracellular flow cytometry, we identified microglia and brain-infiltrating myeloid cells as the main producers of CCL2 during acute infection, and CCL2 was specifically produced in regions of parasite infection in the brain. In contrast, astrocytes became the dominant CCL2 producer during chronic T. gondii infection. To determine the role of astrocyte-derived CCL2 in mobilizing immune cells to the brain and controlling T. gondii infection, we generated GFAP-Cre x CCL2fl/fl mice, in which astrocytes are deficient in CCL2 production. We observed significantly decreased immune cell recruitment and increased parasite burden in the brain during chronic, but not acute, infection of mice deficient in astrocyte CCL2 production, without an effect on peripheral immune responses. To investigate potential mechanisms explaining the reduced control of T. gondii infection, we analyzed key antimicrobial and immune players in host defense against T. gondii and detected a reduction in iNOS+ myeloid cells, and T. gondii-specific CD4+ T cells in the knockout mice. These data uncover a critical role for astrocyte-derived CCL2 in immune cell recruitment and parasite control in the brain during chronic, but not acute, T. gondii infection.
Collapse
Affiliation(s)
- Stephanie B. Orchanian
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, United States of America
- Institute for Immunology, University of California, Irvine, Irvine, California, United States of America
| | - Katherine Still
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Tajie H. Harris
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Melissa B. Lodoen
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, United States of America
- Institute for Immunology, University of California, Irvine, Irvine, California, United States of America
| |
Collapse
|
14
|
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
|
15
|
Denton SL, Mejia A, Nevarez LL, Soares MP, Fox BA, Bzik DJ, Gigley JP. Theft of Host Transferrin Receptor-1 by Toxoplasma gondii is required for infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.23.546322. [PMID: 39372795 PMCID: PMC11451604 DOI: 10.1101/2023.06.23.546322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Nutrient acquisition by apicomplexan parasites is essential to drive their intracellular replication, yet the mechanisms that underpin essential nutrient acquisition are not defined. Using the apicomplexan model Toxoplasma gondii , we show that host cell proteins including the transferrin receptor 1, transferrin, ferritin heavy and light chains, and clathrin light chain are robustly taken up by tachyzoites. Tachyzoite acquisition of host cell protein was not related to host cell type or parasite virulence phenotypes. Bradyzoites possessed little capacity to acquire host cell proteins consistent with the cyst wall representing a barrier to host cell protein cargo. Increased trafficking of host cell transferrin receptor 1 and transferrin to endolysosomes boosted tachyzoite acquisition of host proteins and growth rate. Theft of host transferrin 1 and transferrin did not significantly affect iron levels in the tachyzoite. This study provides insight into essential functions associated with parasite theft of host iron sequestration and storage proteins.
Collapse
|
16
|
Ezzatkhah F, Mahmoudvand H, Raziani Y. The role of Curcuma longa essential oil in controlling acute toxoplasmosis by improving the immune system and reducing inflammation and oxidative stress. Front Cell Infect Microbiol 2023; 13:1161133. [PMID: 37249978 PMCID: PMC10214415 DOI: 10.3389/fcimb.2023.1161133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/06/2023] [Indexed: 05/31/2023] Open
Abstract
Background Chemotherapy with synthetic drugs is the principal approach for toxoplasmosis treatment; however, recent studies reported the limitations and adverse side effects of these chemical drugs. Objective This study aimed to examine the in vitro and in vivo effects of Curcuma longa essential oil (CLE) against the Toxoplasma gondii RH strain. Methods The in vitro effect of different concentrations of CLE on T. gondii tachyzoites was assessed by cell viability assay. Flow cytometry and apoptosis analysis were performed, and nitric oxide production by CLE was also evaluated in tachyzoites. BALB/c mice were orally treated with various doses (1.25, 2.5, and 5 mg·kg-1·day-1) of CLE for 2 weeks. After the induction of acute toxoplasmosis in the mice, their survival rate and the mean number of peritoneal parasites were checked. The hepatic level of antioxidant enzymes and oxidative stress markers was evaluated by commercial kits. The mRNA expression level of proinflammatory cytokines such as interleukin 1-beta (IL-1β) and interferon-gamma (IFN-γ) was evaluated by quantitative real-time PCR. Results CLE, especially at 50 µg/ml, showed potent inhibitory effects on T. gondii tachyzoites. It increased the survival rate (ninth day) and reduced the mean number of peritoneal tachyzoites in the infected mice. CLE dependently increased (p < 0.01) the number of necrotic and apoptotic cells as well as NO production. CLE significantly (p < 0.05) reduced the hepatic level of oxidative stress markers but increased (p < 0.001) the antioxidant enzymes and proinflammatory cytokines in the infected mice, with no important toxicity for vital organs. Conclusion The findings of this survey revealed the significant in vitro inhibitory effects of CLE on T. gondii tachyzoites. The results also exhibited promising in vivo effects of CLE. CLE improved the survival rate of infected mice and reduced the parasite number in them. Although the mechanisms of action of CLE are not clear, our study demonstrated its beneficial effects on acute toxoplasmosis by strengthening the immune system and reducing inflammation and oxidative stress. Still, more studies are required to confirm these results.
Collapse
Affiliation(s)
- Fatemeh Ezzatkhah
- Department of Laboratory Sciences, Sirjan School of Medical Sciences, Sirjan, Iran
| | - Hossein Mahmoudvand
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Yosra Raziani
- Nursing Department, Al-Mustaqbal University College, Hillah, Babylon, Iraq
| |
Collapse
|
17
|
Clark JT, Weizman OE, Aldridge DL, Shallberg LA, Eberhard J, Lanzar Z, Wasche D, Huck JD, Zhou T, Ring AM, Hunter CA. IL-18BP mediates the balance between protective and pathological immune responses to Toxoplasma gondii. Cell Rep 2023; 42:112147. [PMID: 36827187 PMCID: PMC10131179 DOI: 10.1016/j.celrep.2023.112147] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 12/02/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
Interleukin-18 (IL-18) promotes natural killer (NK) and T cell production of interferon (IFN)-γ, a key factor in resistance to Toxoplasma gondii, but previous work has shown a limited role for endogenous IL-18 in control of this parasite. Although infection with T. gondii results in release of IL-18, the production of IFN-γ induces high levels of the IL-18 binding protein (IL-18BP). Antagonism of IL-18BP with a "decoy-to-the-decoy" (D2D) IL-18 construct that does not signal but rather binds IL-18BP results in enhanced innate lymphoid cell (ILC) and T cell responses and improved parasite control. In addition, the use of IL-18 resistant to IL-18BP ("decoy-resistant" IL-18 [DR-18]) is more effective than exogenous IL-18 at promoting innate resistance to infection. DR-18 enhances CD4+ T cell production of IFN-γ but results in CD4+ T cell-mediated pathology. Thus, endogenous IL-18BP restrains aberrant immune pathology, and this study highlights strategies that can be used to tune this regulatory pathway for optimal anti-pathogen responses.
Collapse
Affiliation(s)
- Joseph T Clark
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Orr-El Weizman
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Daniel L Aldridge
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Lindsey A Shallberg
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Julia Eberhard
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Zachary Lanzar
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Devon Wasche
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06519, USA
| | - John D Huck
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Ting Zhou
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Aaron M Ring
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06519, USA.
| | - Christopher A Hunter
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
18
|
Deng S, Graham ML, Chen XM. The Complexity of Interferon Signaling in Host Defense against Protozoan Parasite Infection. Pathogens 2023; 12:319. [PMID: 36839591 PMCID: PMC9962834 DOI: 10.3390/pathogens12020319] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Protozoan parasites, such as Plasmodium, Leishmania, Toxoplasma, Cryptosporidium, and Trypanosoma, are causative agents of health-threatening diseases in both humans and animals, leading to significant health risks and socioeconomic losses globally. The development of effective therapeutic and prevention strategies for protozoan-caused diseases requires a full understanding of the pathogenesis and protective events occurring in infected hosts. Interferons (IFNs) are a family of cytokines with diverse biological effects in host antimicrobial defense and disease pathogenesis, including protozoan parasite infection. Type II IFN (IFN-γ) has been widely recognized as the essential defense cytokine in intracellular protozoan parasite infection, whereas recent studies also revealed the production and distinct function of type I and III IFNs in host defense against these parasites. Decoding the complex network of the IFN family in host-parasite interaction is critical for exploring potential new therapeutic strategies against intracellular protozoan parasite infection. Here, we review the complex effects of IFNs on the host defense against intracellular protozoan parasites and the crosstalk between distinct types of IFN signaling during infections.
Collapse
Affiliation(s)
- Silu Deng
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Marion L. Graham
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
| | - Xian-Ming Chen
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
19
|
Omar M, Abdelal HO. Nitric oxide in parasitic infections: a friend or foe? J Parasit Dis 2022; 46:1147-1163. [PMID: 36457767 PMCID: PMC9606182 DOI: 10.1007/s12639-022-01518-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/20/2022] [Indexed: 11/28/2022] Open
Abstract
The complex interaction between the host and the parasite remains a puzzling question. Control of parasitic infections requires an efficient immune response that must be balanced against destructive pathological consequences. Nitric oxide is a nitrogenous free radical which has many molecular targets and serves diverse functions. Apart from being a signaling messenger, nitric oxide is critical for controlling numerous infections. There is still controversy surrounding the exact role of nitric oxide in the immune response against different parasitic species. It proved protective against intracellular protozoa, as well as extracellular helminths. At the same time, it plays a pivotal role in stimulating detrimental pathological changes in the infected hosts. Several reports have discussed the anti-parasitic and immunoregulatory functions of nitric oxide, which could directly influence the control of the infection. Nevertheless, there is scarce literature addressing the harmful cytotoxic impacts of this mediator. Thus, this review provides insights into the most updated concepts and controversies regarding the dual nature and opposing sides of nitric oxide during the course of different parasitic infections.
Collapse
Affiliation(s)
- Marwa Omar
- Department of Medical Parasitology, Faculty of Medicine, Zagazig University, Gameyet Almohafza St. 1, Menya Al-Kamh, City of Zagazig, 44511 Sharkia Governorate Egypt
| | - Heba O. Abdelal
- LIS: Cross-National Data Center, Maison des Sciences Humaines - 5e étage, 11- porte des Sciences, L-4366 Esch-Belval, Luxembourg
| |
Collapse
|
20
|
Cowan MN, Kovacs MA, Sethi I, Babcock IW, Still K, Batista SJ, O’Brien CA, Thompson JA, Sibley LA, Labuzan SA, Harris TH. Microglial STAT1-sufficiency is required for resistance to toxoplasmic encephalitis. PLoS Pathog 2022; 18:e1010637. [PMID: 36067217 PMCID: PMC9481170 DOI: 10.1371/journal.ppat.1010637] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/16/2022] [Accepted: 08/11/2022] [Indexed: 12/02/2022] Open
Abstract
Toxoplasma gondii is a ubiquitous intracellular protozoan parasite that establishes a life-long chronic infection largely restricted to the central nervous system (CNS). Constant immune pressure, notably IFN-γ-STAT1 signaling, is required for preventing fatal pathology during T. gondii infection. Here, we report that abrogation of STAT1 signaling in microglia, the resident immune cells of the CNS, is sufficient to induce a loss of parasite control in the CNS and susceptibility to toxoplasmic encephalitis during the early stages of chronic infection. Using a microglia-specific genetic labeling and targeting system that discriminates microglia from blood-derived myeloid cells that infiltrate the brain during infection, we find that, contrary to previous in vitro reports, microglia do not express inducible nitric-oxide synthase (iNOS) during T. gondii infection in vivo. Instead, transcriptomic analyses of microglia reveal that STAT1 regulates both (i) a transcriptional shift from homeostatic to “disease-associated microglia” (DAM) phenotype conserved across several neuroinflammatory models, including T. gondii infection, and (ii) the expression of anti-parasitic cytosolic molecules that are required for eliminating T. gondii in a cell-intrinsic manner. Further, genetic deletion of Stat1 from microglia during T. gondii challenge leads to fatal pathology despite largely equivalent or enhanced immune effector functions displayed by brain-infiltrating immune populations. Finally, we show that microglial STAT1-deficiency results in the overrepresentation of the highly replicative, lytic tachyzoite form of T. gondii, relative to its quiescent, semi-dormant bradyzoite form typical of chronic CNS infection. Our data suggest an overall protective role of CNS-resident microglia against T. gondii infection, illuminating (i) general mechanisms of CNS-specific immunity to infection (ii) and a clear role for IFN-STAT1 signaling in regulating a microglial activation phenotype observed across diverse neuroinflammatory disease states. The brain, an immune-privileged organ, can be invaded and colonized by pathogens such as the opportunistic parasite, Toxoplasma gondii. How microglia, the resident immune cells of the brain, provide resistance to infection is an active area of investigation. In this study, we used a genetic approach to generate and study mice with microglia that lack STAT1, a critical transcription factor that confers protection against intracellular pathogens in both humans and mice. We find that despite robust activation and recruitment of immune cells from the blood to the brain during infection, STAT1 deficiency in microglia leads to increased brain parasite burden and uniform lethality in mice when challenged with T. gondii. Our bioinformatic analyses also indicate that STAT1 in microglia regulates (i) the expression of large families of genes associated with parasite killing and (ii) a microglial activation state that has been classically seen in neurodegeneration. Our findings identify mechanisms by which microglia contribute to parasite control and contribute to a greater understanding of their cellular physiology during neuroinflammation.
Collapse
Affiliation(s)
- Maureen N. Cowan
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Michael A. Kovacs
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Ish Sethi
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Isaac W. Babcock
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Katherine Still
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Samantha J. Batista
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Carleigh A. O’Brien
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jeremy A. Thompson
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Lydia A. Sibley
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Sydney A. Labuzan
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Tajie H. Harris
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| |
Collapse
|
21
|
Activity of isoflavone biochanin A in chronic experimental toxoplasmosis: impact on inflammation. Parasitol Res 2022; 121:2405-2414. [PMID: 35710847 PMCID: PMC9279236 DOI: 10.1007/s00436-022-07571-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 06/03/2022] [Indexed: 11/07/2022]
Abstract
Toxoplasma gondii is a worldwide prevalent parasite. The infection has been linked to variable inflammatory effects including neuroinflammation. Biochanin A (BCA) is an isoflavone, known for its anti-inflammatory and anti-oxidative properties. In this study, we examined the effect of BCA on the brain and liver inflammatory lesions in a murine model with chronic toxoplasmosis. Mice were divided in to six groups: non-infected control, non-infected BCA-treated, and four infected groups with Toxoplasmagondii Me49-type II cystogenic strain: infected control, BCA (50 mg/kg/day)-treated, combined BCA/cotrimoxazole-treated and cotrimoxazole (370 mg/kg/day) alone-treated. Gene expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and inducible nitric oxide synthase (iNOS) was evaluated by quantitative real-time PCR in the brain and liver tissues. In the infected control group, an upregulation of TNF-α and IL-1β mRNA expression levels was found. However, a downregulation of iNOS expression was detected in the brain of infected control mice. In both BCA- and combined-treated groups, the brain and liver tissues showed significantly reduced inflammatory lesions compared to the infected control mice with inhibited TNF-α and IL-1β mRNA levels. The iNOS expression levels in the brain tissues of BCA group were significantly higher than the levels of the infected control group. BCA alone or combined significantly reduced T. gondii cyst count in the brain tissues. In conclusion, the anti-inflammatory activity of BCA was demonstrated in the brain tissues of mice with chronic toxoplasmosis with decreased TNF-α and IL-1β expression levels and increased iNOS expression levels.
Collapse
|
22
|
Sánchez-Arcila JC, Jensen KDC. Forward Genetics in Apicomplexa Biology: The Host Side of the Story. Front Cell Infect Microbiol 2022; 12:878475. [PMID: 35646724 PMCID: PMC9133346 DOI: 10.3389/fcimb.2022.878475] [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: 02/18/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Forward genetic approaches have been widely used in parasitology and have proven their power to reveal the complexities of host-parasite interactions in an unbiased fashion. Many aspects of the parasite's biology, including the identification of virulence factors, replication determinants, antibiotic resistance genes, and other factors required for parasitic life, have been discovered using such strategies. Forward genetic approaches have also been employed to understand host resistance mechanisms to parasitic infection. Here, we will introduce and review all forward genetic approaches that have been used to identify host factors involved with Apicomplexa infections, which include classical genetic screens and QTL mapping, GWAS, ENU mutagenesis, overexpression, RNAi and CRISPR-Cas9 library screens. Collectively, these screens have improved our understanding of host resistance mechanisms, immune regulation, vaccine and drug designs for Apicomplexa parasites. We will also discuss how recent advances in molecular genetics give present opportunities to further explore host-parasite relationships.
Collapse
Affiliation(s)
- Juan C. Sánchez-Arcila
- Department of Molecular and Cell Biology, University of California Merced, Merced, CA, United States
| | - Kirk D. C. Jensen
- Department of Molecular and Cell Biology, University of California Merced, Merced, CA, United States
- Health Science Research Institute, University of California, Merced, Merced, CA, United States
| |
Collapse
|
23
|
Sasai M, Yamamoto M. Anti-toxoplasma host defense systems and the parasitic counterdefense mechanisms. Parasitol Int 2022; 89:102593. [DOI: 10.1016/j.parint.2022.102593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 04/12/2022] [Accepted: 04/26/2022] [Indexed: 10/18/2022]
|
24
|
Hou Z, Zhang H, Xu K, Zhu S, Wang L, Su D, Liu J, Su S, Liu D, Huang S, Xu J, Pan Z, Tao J. Cluster analysis of splenocyte microRNAs in the pig reveals key signal regulators of immunomodulation in the host during acute and chronic Toxoplasma gondii infection. Parasit Vectors 2022; 15:58. [PMID: 35177094 PMCID: PMC8851844 DOI: 10.1186/s13071-022-05164-3] [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: 11/08/2021] [Accepted: 01/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Toxoplasma gondii is an obligate intracellular protozoan parasite that can cause a geographically widespread zoonosis. Our previous splenocyte microRNA profile analyses of pig infected with T. gondii revealed that the coordination of a large number of miRNAs regulates the host immune response during infection. However, the functions of other miRNAs involved in the immune regulation during T. gondii infection are not yet known. METHODS Clustering analysis was performed by K-means, self-organizing map (SOM), and hierarchical clustering to obtain miRNA groups with the similar expression patterns. Then, the target genes of the miRNA group in each subcluster were further analyzed for functional enrichment by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Reactome pathway to recognize the key signaling molecules and the regulatory signatures of the innate and adaptive immune responses of the host during T. gondii infection. RESULTS A total of 252 miRNAs were successfully divided into 22 subclusters by K-means clustering (designated as K1-K22), 29 subclusters by SOM clustering (designated as SOM1-SOM29), and six subclusters by hierarchical clustering (designated as H1-H6) based on their dynamic expression levels in the different infection stages. A total of 634, 660, and 477 GO terms, 15, 26, and 14 KEGG pathways, and 16, 15, and 7 Reactome pathways were significantly enriched by K-means, SOM, and hierarchical clustering, respectively. Of note, up to 22 miRNAs mainly showing downregulated expression at 50 days post-infection (dpi) were grouped into one subcluster (namely subcluster H3-K17-SOM1) through the three algorithms. Functional analysis revealed that a large group of immunomodulatory signaling molecules were controlled by the different miRNA groups to regulate multiple immune processes, for instance, IL-1-mediated cellular response and Th1/Th2 cell differentiation partly depending on Notch signaling transduction for subclusters K1 and K2, innate immune response involved in neutrophil degranulation and TLR4 cascade signaling for subcluster K15, B cell activation for subclusters SOM17, SOM1, and SOM25, leukocyte migration, and chemokine activity for subcluster SOM9, cytokine-cytokine receptor interaction for subcluster H2, and interleukin production, chemotaxis of immune cells, chemokine signaling pathway, and C-type lectin receptor signaling pathway for subcluster H3-K17-SOM1. CONCLUSIONS Cluster analysis of splenocyte microRNAs in the pig revealed key regulatory properties of subcluster miRNA molecules and important features in the immune regulation induced by acute and chronic T. gondii infection. These results contribute new insight into the identification of physiological immune responses and maintenance of tolerance in pig spleen tissues during T. gondii infection.
Collapse
Affiliation(s)
- Zhaofeng Hou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Hui Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Kangzhi Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Shifan Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Lele Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Dingzeyang Su
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Jiantao Liu
- YEBIO Bioengineering Co., Ltd. of QINGDAO, Qingdao, 266109, People's Republic of China
| | - Shijie Su
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Dandan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Siyang Huang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Jinjun Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Zhiming Pan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China.,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China
| | - Jianping Tao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China. .,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225009, People's Republic of China. .,Jiangsu Key Laboratory of Zoonosis, Yangzhou, 225009, People's Republic of China.
| |
Collapse
|
25
|
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: 5.0] [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
|
26
|
Lodoen MB, Smith NC, Soldati-Favre D, Ferguson DJP, van Dooren GG. Nanos gigantium humeris insidentes: old papers informing new research into Toxoplasma gondii. Int J Parasitol 2021; 51:1193-1212. [PMID: 34736901 PMCID: PMC10538201 DOI: 10.1016/j.ijpara.2021.10.004] [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: 09/14/2021] [Revised: 10/19/2021] [Accepted: 10/19/2021] [Indexed: 11/25/2022]
Abstract
Since Nicolle, Manceaux and Splendore first described Toxoplasma gondii as a parasite of rodents and rabbits in the early 20th century, a diverse and vigorous research community has been built around studying this fascinating intracellular parasite. In addition to its importance as a pathogen of humans, livestock and wildlife, modern researchers are attracted to T. gondii as a facile experimental system to study many aspects of evolutionary biology, cellular biology, host-microbe interactions, and host immunity. For new researchers entering the field, the extensive literature describing the biology of the parasite, and the interactions with its host, can be daunting. In this review, we examine four foundational studies that describe various aspects of T. gondii biology, presenting a 'journal club'-style analysis of each. We have chosen a paper that established the beguiling life cycle of the parasite (Hutchison et al., 1971), a paper that described key features of its cellular biology that the parasite shares with related organisms (Gustafson et al., 1954), a paper that characterised the origin of the unique compartment in which the parasite resides within host cells (Jones and Hirsch, 1972), and a paper that established a key mechanism in the host immune response to parasite infection (Pfefferkorn, 1984). These interesting and far-reaching studies set the stage for subsequent research into numerous facets of parasite biology. As well as providing new researchers with an entry point into the literature surrounding the parasite, revisiting these studies can remind us of the roots of our discipline, how far we have come, and the new directions in which we might head.
Collapse
Affiliation(s)
- Melissa B Lodoen
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA
| | - Nicholas C Smith
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; Research School of Biology, Australian National University, Canberra, ACT 2600, Australia
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, CMU, University of Geneva, Geneva, Switzerland
| | - David J P Ferguson
- Nuffield Department of Clinical Laboratory Science, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK; Department of Biological and Medical Sciences, Faculty of Health and Life Science, Oxford Brookes University, Gipsy Lane, Oxford OX3 0BP, UK
| | - Giel G van Dooren
- Research School of Biology, Australian National University, Canberra, ACT 2600, Australia.
| |
Collapse
|
27
|
Effect of spiramycin versus aminoguanidine and their combined use in experimental toxoplasmosis. J Parasit Dis 2021; 45:1014-1025. [PMID: 34789985 DOI: 10.1007/s12639-021-01396-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 04/17/2021] [Indexed: 10/21/2022] Open
Abstract
Toxoplasmosis is one of the widest spread parasitic infections which is caused by Toxoplasma gondii protozoon. Many experimental studies have evaluated the effect of aminoguanidine upon parasitic load and inflammatory process. However, few reports have illustrated the impact of combining aminoguanidine with spiramycin in the treatment of toxoplasmosis. Therefore, our study aimed to explore the possible effects of spiramycin used alone and combined with aminoguanidine against the avirulent (ME49) Toxoplasma gondii strain in experimental toxoplasmosis. Fifty-five Swiss albino mice were included in the study and were divided into five groups: (GI): non-infected control group; (GII): infected untreated control group; (GIII): infected- spiramycin treated group; (GIV): infected-aminoguanidine treated group; (GV): infected and received combination of spiramycin and aminoguanidine. Obtained results exhibited a significant increase in brain cysts numbers in aminoguanidine treated groups compared to infected untreated control groups. Histopathological studies denoted that combination between spiramycin and aminoguanidine improved the pathological features only in liver and heart tissues of the studied groups. Moreover, it was noticed that spiramycin administered alone had no effect on nitric oxide expression, whereas its combination with aminoguanidine had an inhibitory effect on inducible nitric oxide synthase enzyme in brain, liver and heart tissues of different study groups. In conclusion, the combination of spiramycin and aminoguanidine significantly reduced the parasitic burden, yet, it failed to resolve the pathological sequels in brain tissues of Toxoplasma gondii infected mice.
Collapse
|
28
|
Wang ZJ, Yu SM, Gao JM, Zhang P, Hide G, Yamamoto M, Lai DH, Lun ZR. High resistance to Toxoplasma gondii infection in inducible nitric oxide synthase knockout rats. iScience 2021; 24:103280. [PMID: 34765911 PMCID: PMC8571494 DOI: 10.1016/j.isci.2021.103280] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/08/2021] [Accepted: 10/13/2021] [Indexed: 11/29/2022] Open
Abstract
Nitric oxide (NO) is an important immune molecule that acts against extracellular and intracellular pathogens in most hosts. However, after the knockout of inducible nitric oxide synthase (iNOS−/−) in Sprague Dawley (SD) rats, these iNOS−/− rats were found to be completely resistant to Toxoplasma gondii infection. Once the iNOS−/− rat peritoneal macrophages (PMs) were infected with T. gondii, they produced high levels of reactive oxygen species (ROS) triggered by GRA43 secreted by T. gondii, which damaged the parasitophorous vacuole membrane and PM mitochondrial membranes within a few hours post-infection. Further evidence indicated that the high levels of ROS caused mitochondrial superoxide dismutase 2 depletion and induced PM pyroptosis and cell death. This discovery of complete resistance to T. gondii infection, in the iNOS−/−-SD rat, demonstrates a strong link between NO and ROS in immunity to T. gondii infection and showcases a potentially novel and effective backup innate immunity system. iNOS−/−-SD rats show strong resistance to Toxoplasma gondii infection iNOS−/−-SD rat PMs resist T. gondii infection through ROS upregulation The T. gondii infection results in PM pyroptosis in iNOS−/−-SD rats GRAs play a key role in the activation of resistance in iNOS−/−-SD rat PMs
Collapse
Affiliation(s)
- Zhen-Jie Wang
- Guangdong Provincial Key Laboratory of Aquatic Economic Animals, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, The People's Republic of China
| | - Shao-Meng Yu
- Guangdong Provincial Key Laboratory of Aquatic Economic Animals, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, The People's Republic of China
| | - Jiang-Mei Gao
- Department of Parasitology, Key Laboratory of Tropical Disease Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, The People's Republic of China
| | - Peng Zhang
- Guangdong Provincial Key Laboratory of Aquatic Economic Animals, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, The People's Republic of China
| | - Geoff Hide
- Biomedical Research Centre, School of Science, Engineering and Environment, University of Salford, Salford M5 4WT, UK
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - De-Hua Lai
- Guangdong Provincial Key Laboratory of Aquatic Economic Animals, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, The People's Republic of China
| | - Zhao-Rong Lun
- Guangdong Provincial Key Laboratory of Aquatic Economic Animals, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, The People's Republic of China.,Department of Parasitology, Key Laboratory of Tropical Disease Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, The People's Republic of China.,Biomedical Research Centre, School of Science, Engineering and Environment, University of Salford, Salford M5 4WT, UK
| |
Collapse
|
29
|
Saijo-Hamano Y, Sherif AA, Pradipta A, Sasai M, Sakai N, Sakihama Y, Yamamoto M, Standley DM, Nitta R. Structural basis of membrane recognition of Toxoplasma gondii vacuole by Irgb6. Life Sci Alliance 2021; 5:5/1/e202101149. [PMID: 34753804 PMCID: PMC8605323 DOI: 10.26508/lsa.202101149] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 11/24/2022] Open
Abstract
Upon Toxoplasma gondii infection, Irgb6 is recruited to the parasitophorous vacuole membrane (PVM) where Irgb6 disrupts it. We solved the atomic structures of Irgb6 in two distinct nucleotide states, revealing a unique PVM-binding interface sensitive to GTPase cycling. The p47 immunity-related GTPase (IRG) Irgb6 plays a pioneering role in host defense against Toxoplasma gondii infection. Irgb6 is recruited to the parasitophorous vacuole membrane (PVM) formed by T. gondii and disrupts it. Despite the importance of this process, the molecular mechanisms accounting for PVM recognition by Irgb6 remain elusive because of lack of structural information on Irgb6. Here we report the crystal structures of mouse Irgb6 in the GTP-bound and nucleotide-free forms. Irgb6 exhibits a similar overall architecture to other IRGs in which GTP binding induces conformational changes in both the dimerization interface and the membrane-binding interface. The membrane-binding interface of Irgb6 assumes a unique conformation, composed of N- and C-terminal helical regions forming a phospholipid binding site. In silico docking of phospholipids further revealed membrane-binding residues that were validated through mutagenesis and cell-based assays. Collectively, these data demonstrate a novel structural basis for Irgb6 to recognize T. gondii PVM in a manner distinct from other IRGs.
Collapse
Affiliation(s)
- Yumiko Saijo-Hamano
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Aalaa Alrahman Sherif
- Depertment of Genome Informatics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Ariel Pradipta
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Immunoparasitology, World Premier International Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Miwa Sasai
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Immunoparasitology, World Premier International Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Division of Microbiology and Immunology, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| | | | - Yoshiaki Sakihama
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Immunoparasitology, World Premier International Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Division of Microbiology and Immunology, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| | - Daron M Standley
- Depertment of Genome Informatics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Ryo Nitta
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
30
|
Hamed EFA, Mostafa NE, Fawzy EM, Ibrahim MN, Attia R, Salama MA. The delayed death-causing nature of Rosmarinus officinalis leaf extracts and their mixture within experimental chronic toxoplasmosis: Therapeutic and prophylactic implications. Acta Trop 2021; 221:105992. [PMID: 34089696 DOI: 10.1016/j.actatropica.2021.105992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/22/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND The restricted effect, significant toxicity, and emerging resistance of anti-toxoplasmosis synthetic agents impose the search for alternatives. The current research aimed to evaluate the prophylactic and therapeutic efficacy of Rosmarinus officinalis extracts and their mixtures against chronic murine toxoplasmosis and to clarify the phenomenon of delayed death. METHODS This research included two experimental designs, the first to test the preventive and curative efficacy of the extracts and the second to assess delayed death in mice infected with the ME49 strain of Toxoplasma gondii. The essential oils of the plant were analyzed by gas chromatography/mass spectrometry. RESULTS Treatment with a mixture of rosemary extracts displayed reduction rates of 81% for T. gondii cyst burden and 23% for cyst viability. The reinfected group with the pretreated cysts reported 93.4% reduction in cyst burden and 95.4% in cyst viability. Moreover, 90% reduction of the infectivity rate was obtained. The therapeutic efficacy of this mixture was superior to its valuable prophylactic effect. Histopathological examination of liver and brain tissue exhibited marked improvement. Both extracts possess free radical scavenging and antioxidant activities evidenced by high expression of iNOS stain. Our results were signified by low BAG-1 gene expression and massive mutilation of T. gondii cyst in the targeted group using scanning electron microscopy. Analysis of R. officinalis revealed the presence of isobornylformate as a novel ingredient. CONCLUSIONS R. officinalis displays a therapeutic rather than prophylactic potential, indicating the emergence of an effective safe alternative therapy.
Collapse
|
31
|
Hamie M, Tawil N, El Hajj R, Najm R, Moodad S, Hleihel R, Karam M, El Sayyed S, Besteiro S, El-Sabban M, Dubremetz JF, Lebrun M, El Hajj H. P18 (SRS35/TgSAG4) Plays a Role in the Invasion and Virulence of Toxoplasma gondii. Front Immunol 2021; 12:643292. [PMID: 34262559 PMCID: PMC8273438 DOI: 10.3389/fimmu.2021.643292] [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: 12/17/2020] [Accepted: 06/10/2021] [Indexed: 11/13/2022] Open
Abstract
Toxoplasmosis is a prevalent parasitic disease caused by Toxoplasma gondii (T. gondii). Under the control of the host immune system, T. gondii persists as latent bradyzoite cysts. Immunosuppression leads to their reactivation, a potentially life-threatening condition. Interferon-gamma (IFN-γ) controls the different stages of toxoplasmosis. Here, we addressed the role of the parasite surface antigen P18, belonging to the Surface-Antigen 1 (SAG-1) Related Sequence (SRS) family, in a cyst-forming strain. Deletion of P18 gene (KO P18) impaired the invasion of parasites in macrophages and IFN-γ-mediated activation of macrophages further reduced the invasion capacity of this KO, as compared to WT strain. Mice infected by KO P18, showed a marked decrease in virulence during acute toxoplasmosis. This was consequent to less parasitemia, accompanied by a substantial recruitment of dendritic cells, macrophages and natural killer cells (NK). Furthermore, KO P18 resulted in a higher number of bradyzoite cysts, and a stronger inflammatory response. A prolonged survival of mice was observed upon immunosuppression of KO P18 infected BALB/c mice or upon oral infection of Severe Combined Immunodeficiency (SCID) mice, with intact macrophages and natural killer (NK) cells. In stark contrast, oral infection of NSG (NOD/Shi-scid/IL-2Rγnull) mice, defective in macrophages and NK cells, with KO P18, was as lethal as that of the control strain showing that the conversion from bradyzoites to tachyzoites is intact and, suggesting a role of P18 in the response to host IFN-γ. Collectively, these data demonstrate a role for P18 surface antigen in the invasion of macrophages and in the virulence of the parasite, during acute and chronic toxoplasmosis.
Collapse
Affiliation(s)
- Maguy Hamie
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nadim Tawil
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Rana El Hajj
- Department of Biological Sciences, Beirut Arab University, Beirut, Lebanon
| | - Rania Najm
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Sara Moodad
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Rita Hleihel
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Martin Karam
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Sana El Sayyed
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Maryse Lebrun
- LPHI UMR5235, Univ Montpellier, CNRS, Montpellier, France
| | - Hiba El Hajj
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
32
|
Lymphotoxin β Receptor: a Crucial Role in Innate and Adaptive Immune Responses against Toxoplasma gondii. Infect Immun 2021; 89:IAI.00026-21. [PMID: 33753412 PMCID: PMC8316152 DOI: 10.1128/iai.00026-21] [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: 01/15/2021] [Accepted: 03/12/2021] [Indexed: 11/21/2022] Open
Abstract
The lymphotoxin β receptor (LTβR) plays an essential role in the initiation of immune responses to intracellular pathogens. In mice, the LTβR is crucial for surviving acute toxoplasmosis; however, until now, a functional analysis was largely incomplete. Here, we demonstrate that the LTβR is a key regulator required for the intricate balance of adaptive immune responses. Toxoplasma gondii-infected LTβR-deficient (LTβR−/−) mice show globally altered interferon-γ (IFN-γ) regulation, reduced IFN-γ-controlled host effector molecule expression, impaired T cell functionality, and an absent anti-parasite-specific IgG response, resulting in a severe loss of immune control of the parasites. Reconstitution of LTβR−/− mice with toxoplasma immune serum significantly prolongs survival following T. gondii infection. Notably, analysis of RNA-seq data clearly indicates a specific effect of T. gondii infection on the B cell response and isotype switching. This study uncovers the decisive role of the LTβR in cytokine regulation and adaptive immune responses to control T. gondii.
Collapse
|
33
|
Abdel-Hamed EF, Ibrahim MN, Mostafa NE, Moawad HSF, Elgammal NE, Darwiesh EM, El-Rafey DS, ElBadawy NE, Al-Khoufi EA, Hindawi SI. Role of interferon gamma in SARS-CoV-2-positive patients with parasitic infections. Gut Pathog 2021; 13:29. [PMID: 33947467 PMCID: PMC8096133 DOI: 10.1186/s13099-021-00427-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/29/2021] [Indexed: 12/25/2022] Open
Abstract
Background By 27 June 2020, almost half a million people had died due to COVID-19 infections. The susceptibility and severity of infection vary significantly across nations. The contribution of chronic viral and parasitic infections to immune homeostasis remains a concern. By investigating the role of interferon (IFN)-γ, we conducted this study to understand the connection between the decrease in numbers and severity of COVID-19 cases within parasitic endemic regions. Our research included 375 patients referred to hospitals for diagnosis of COVID-19 infection. Patients were subjected to full investigations, in particular severe acute respiratory syndrome coronavirus-2 nucleic acid and Toxoplasma IgM and IgG antibody detection, stool examination, and quantitative IFN-γ measurement. Results The majority of the studied cases had chest manifestation either alone (54.7%) or in association with gastrointestinal (GIT) manifestations (19.7%), whereas 25.6% had GIT symptoms. We reported parasitic infections in 72.8% of mild COVID-19 cases and 20.7% of severe cases. Toxoplasma gondii, Cryptosporidium, Blastocyst, and Giardia were the most common parasitic infections among the COVID-19 cases studied. Conclusion The remarkable adaptation of human immune response to COVID-19 infection by parasitic infections with high levels of IFN-γ was observed in moderate cases compared with low levels in extreme cases. The potential therapeutic efforts aimed at the role of parasitic infection in immune system modulation are needed if this hypothesis is confirmed.
Collapse
Affiliation(s)
- Enas Fakhry Abdel-Hamed
- Medical Parasitology Department, Faculty of Medicine, Zagazig University, El Kawmia Square, Zagazig, 44511, Sharkia Governorate, Egypt.
| | - Mohamed N Ibrahim
- Clinical Laboratories Department, College of Applied Medical Sciences, Jouf University, Al-Jouf, 77451, Saudi Arabia
| | - Nahed E Mostafa
- Medical Parasitology Department, Faculty of Medicine, Zagazig University, El Kawmia Square, Zagazig, 44511, Sharkia Governorate, Egypt
| | - Howayda S F Moawad
- Medical Parasitology Department, Faculty of Medicine, Zagazig University, El Kawmia Square, Zagazig, 44511, Sharkia Governorate, Egypt
| | - Nahla E Elgammal
- Tropical Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, 44511, Egypt
| | - Ehab M Darwiesh
- Tropical Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, 44511, Egypt
| | - Dina S El-Rafey
- Community, Environmental and Occupational Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, 44511, Egypt
| | - Nissreen E ElBadawy
- Microbiology Department, Faculty of Medicine, Zagazig University, Zagazig, 44511, Egypt
| | - Emad Ali Al-Khoufi
- Internal Medicine Department, College of Medicine, King Faisal University, 31982, Al-Ahsa, Saudi Arabia
| | - Salwa I Hindawi
- Haematology and Transfusion Medicine, King Abdulaziz University, Jeddah, 21577, Saudi Arabia
| |
Collapse
|
34
|
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: 21] [Impact Index Per Article: 7.0] [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
|
35
|
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: 17] [Impact Index Per Article: 5.7] [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
|
36
|
French T, Israel N, Düsedau HP, Tersteegen A, Steffen J, Cammann C, Topfstedt E, Dieterich D, Schüler T, Seifert U, Dunay IR. The Immunoproteasome Subunits LMP2, LMP7 and MECL-1 Are Crucial Along the Induction of Cerebral Toxoplasmosis. Front Immunol 2021; 12:619465. [PMID: 33968021 PMCID: PMC8099150 DOI: 10.3389/fimmu.2021.619465] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 03/16/2021] [Indexed: 11/28/2022] Open
Abstract
Cell survival and function critically relies on the fine-tuned balance of protein synthesis and degradation. In the steady state, the standard proteasome is sufficient to maintain this proteostasis. However, upon inflammation, the sharp increase in protein production requires additional mechanisms to limit protein-associated cellular stress. Under inflammatory conditions and the release of interferons, the immunoproteasome (IP) is induced to support protein processing and recycling. In antigen-presenting cells constitutively expressing IPs, inflammation-related mechanisms contribute to the formation of MHC class I/II-peptide complexes, which are required for the induction of T cell responses. The control of Toxoplasma gondii infection relies on Interferon-γ (IFNγ)-related T cell responses. Whether and how the IP affects the course of anti-parasitic T cell responses along the infection as well as inflammation of the central nervous system is still unknown. To answer this question we used triple knockout (TKO) mice lacking the 3 catalytic subunits of the immunoproteasome (β1i/LMP2, β2i/MECL-1 and β5i/LMP7). Here we show that the numbers of dendritic cells, monocytes and CD8+ T cells were reduced in Toxoplasma gondii-infected TKO mice. Furthermore, impaired IFNγ, TNF and iNOS production was accompanied by dysregulated chemokine expression and altered immune cell recruitment to the brain. T cell differentiation was altered, apoptosis rates of microglia and monocytes were elevated and STAT3 downstream signaling was diminished. Consequently, anti-parasitic immune responses were impaired in TKO mice leading to elevated T. gondii burden and prolonged neuroinflammation. In summary we provide evidence for a critical role of the IP subunits β1i/LMP2, β2i/MECL-1 and β5i/LMP7 for the control of cerebral Toxoplasma gondii infection and subsequent neuroinflammation.
Collapse
Affiliation(s)
- Timothy French
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Nicole Israel
- Friedrich Loeffler-Institute of Medical Microbiology-Virology, University Medicine Greifswald, Greifswald, Germany.,Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Henning Peter Düsedau
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Anne Tersteegen
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Johannes Steffen
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Clemens Cammann
- Friedrich Loeffler-Institute of Medical Microbiology-Virology, University Medicine Greifswald, Greifswald, Germany.,Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Eylin Topfstedt
- Friedrich Loeffler-Institute of Medical Microbiology-Virology, University Medicine Greifswald, Greifswald, Germany
| | - Daniela Dieterich
- Institute of Pharmacology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Ulrike Seifert
- Friedrich Loeffler-Institute of Medical Microbiology-Virology, University Medicine Greifswald, Greifswald, Germany.,Institute of Molecular and Clinical Immunology, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany
| |
Collapse
|
37
|
Gaignage M, Uyttenhove C, Jones LL, Bourdeaux C, Chéou P, Mandour MF, Coutelier JP, Vignali DAA, Van Snick J. Novel antibodies that selectively block mouse IL-12 enable the re-evaluation of the role of IL-12 in immune protection and pathology. Eur J Immunol 2021; 51:1482-1493. [PMID: 33788263 DOI: 10.1002/eji.202048936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 02/11/2021] [Accepted: 03/23/2021] [Indexed: 01/01/2023]
Abstract
The dimeric cytokine IL-12 is important in the control of various infections but also contributes to the pathology of certain diseases making it a potential target for therapy. However, its specific inhibition with antibodies is complicated by the fact that its two subunits are present in other cytokines: p40 in IL-23 and p35 in IL-35. This has led to erroneous conclusions like the alleged implication of IL-12 in experimental autoimmune encephalomyelitis (EAE). Here, we report the development of a mouse anti-mouse IL-12 vaccine and the production of monoclonal antibodies (mAbs) that do not react with p40 or p35 (in IL-35) but specifically recognize and functionally inhibit the IL-12 heterodimer. Using one of these mAbs, MM12A1.6, that strongly inhibited IFN-γ production and LPS-induced septic shock after viral infection, we demonstrate the critical role played by IL-12 in the rejection of male skin graft by female C57BL/6 syngeneic recipients and in the clearance of an immunogenic mastocytoma tumor variant by DBA/2 mice, but not in a parent to F1 immune aggression model nor in MOG-induced EAE, which was clearly prevented by anti-p40 mAb C17.8. Given this selective inhibition of IL-12, these mAbs provide new options for reassessing IL-12 function in vivo.
Collapse
Affiliation(s)
| | - Catherine Uyttenhove
- de Duve Institute, Université de Louvain, Brussels, Belgium.,Ludwig Cancer Research, Brussels, Belgium
| | - Lindsay L Jones
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Paméla Chéou
- de Duve Institute, Université de Louvain, Brussels, Belgium
| | - Mohamed F Mandour
- de Duve Institute, Université de Louvain, Brussels, Belgium.,Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | | | - Dario A A Vignali
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.,Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Jacques Van Snick
- de Duve Institute, Université de Louvain, Brussels, Belgium.,Ludwig Cancer Research, Brussels, Belgium
| |
Collapse
|
38
|
Elsheikha HM, Marra CM, Zhu XQ. Epidemiology, Pathophysiology, Diagnosis, and Management of Cerebral Toxoplasmosis. Clin Microbiol Rev 2021; 34:e00115-19. [PMID: 33239310 PMCID: PMC7690944 DOI: 10.1128/cmr.00115-19] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Toxoplasma gondii is known to infect a considerable number of mammalian and avian species and a substantial proportion of the world's human population. The parasite has an impressive ability to disseminate within the host's body and employs various tactics to overcome the highly regulatory blood-brain barrier and reside in the brain. In healthy individuals, T. gondii infection is largely tolerated without any obvious ill effects. However, primary infection in immunosuppressed patients can result in acute cerebral or systemic disease, and reactivation of latent tissue cysts can lead to a deadly outcome. It is imperative that treatment of life-threatening toxoplasmic encephalitis is timely and effective. Several therapeutic and prophylactic regimens have been used in clinical practice. Current approaches can control infection caused by the invasive and highly proliferative tachyzoites but cannot eliminate the dormant tissue cysts. Adverse events and other limitations are associated with the standard pyrimethamine-based therapy, and effective vaccines are unavailable. In this review, the epidemiology, economic impact, pathophysiology, diagnosis, and management of cerebral toxoplasmosis are discussed, and critical areas for future research are highlighted.
Collapse
Affiliation(s)
- Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Christina M Marra
- Departments of Neurology and Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - 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, People's Republic of China
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi Province, People's Republic of China
| |
Collapse
|
39
|
Sikorski PM, Commodaro AG, Grigg ME. A Protective and Pathogenic Role for Complement During Acute Toxoplasma gondii Infection. Front Cell Infect Microbiol 2021; 11:634610. [PMID: 33692968 PMCID: PMC7937796 DOI: 10.3389/fcimb.2021.634610] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/06/2021] [Indexed: 11/13/2022] Open
Abstract
The infection competence of the protozoan pathogen Toxoplasma gondii is critically dependent on the parasite’s ability to inactivate the host complement system. Toxoplasma actively resists complement-mediated killing in non-immune serum by recruiting host-derived complement regulatory proteins C4BP and Factor H (FH) to the parasite surface to inactivate surface-bound C3 and limit formation of the C5b-9 membrane attack complex (MAC). While decreased complement activation on the parasite surface certainly protects Toxoplasma from immediate lysis, the biological effector functions of C3 split products C3b and C3a are maintained, which includes opsonization of the parasite for phagocytosis and potent immunomodulatory effects that promote pro-inflammatory responses and alters mucosal defenses during infection, respectively. In this review, we discuss how complement regulation by Toxoplasma controls parasite burden systemically but drives exacerbated immune responses locally in the gut of genetically susceptible C57BL/6J mice. In effect, Toxoplasma has evolved to strike a balance with the complement system, by inactivating complement to protect the parasite from immediate serum killing, it generates sufficient C3 catabolites that signal through their cognate receptors to stimulate protective immunity. This regulation ultimately controls tachyzoite proliferation and promotes host survival, parasite persistence, and transmissibility to new hosts.
Collapse
Affiliation(s)
- Patricia M Sikorski
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.,Department of Microbiology and Immunology, Georgetown University Medical Center, Georgetown University, Washington, DC, United States
| | - Alessandra G Commodaro
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Michael E Grigg
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
40
|
Sokol-Borrelli SL, Coombs RS, Boyle JP. A Comparison of Stage Conversion in the Coccidian Apicomplexans Toxoplasma gondii, Hammondia hammondi, and Neospora caninum. Front Cell Infect Microbiol 2020; 10:608283. [PMID: 33344268 PMCID: PMC7744739 DOI: 10.3389/fcimb.2020.608283] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 11/06/2020] [Indexed: 01/31/2023] Open
Abstract
Stage conversion is a critical life cycle feature for several Apicomplexan parasites as the ability to switch between life forms is critical for replication, dissemination, pathogenesis and ultimately, transmission to a new host. In order for these developmental transitions to occur, the parasite must first sense changes in their environment, such as the presence of stressors or other environmental signals, and then respond to these signals by initiating global alterations in gene expression. As our understanding of the genetic components required for stage conversion continues to broaden, we can better understand the conserved mechanisms for this process and unique components and their contribution to pathogenesis by comparing stage conversion in multiple closely related species. In this review, we will discuss what is currently known about the mechanisms driving stage conversion in Toxoplasma gondii and its closest relatives Hammondia hammondi and Neospora caninum. Work by us and others has shown that these species have some important differences in the way that they (1) progress through their life cycle and (2) respond to stage conversion initiating stressors. To provide a specific example of species-specific complexities associated with stage conversion, we will discuss our recent published and unpublished work comparing stress responses in T. gondii and H. hammondi.
Collapse
Affiliation(s)
| | | | - Jon P. Boyle
- University of Pittsburgh, Department of Biological Sciences, Kenneth P. Dietrich School of Arts and Sciences, Pittsburgh, PA, United States
| |
Collapse
|
41
|
Salinas-Carmona MC, Longoria-Lozano O, Garza-Esquivel HR, López-Ulloa J, Reyes-Carrillo J, Vázquez-Marmolejo AV. Inducible nitric oxide synthase blockade with aminoguanidine, protects mice infected with Nocardia brasiliensis from actinomycetoma development. PLoS Negl Trop Dis 2020; 14:e0008775. [PMID: 33091049 PMCID: PMC7580934 DOI: 10.1371/journal.pntd.0008775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 09/04/2020] [Indexed: 12/14/2022] Open
Abstract
Mycetoma is a chronic infectious disease that can be caused by fungi or bacteria, Madurella mycetomatis and Nocardia brasiliensis are frequent etiologic agents of this disease. Mycetoma produced by bacteria is known as actinomycetoma. In mycetoma produced by fungi (eumycetoma) and actinomycetoma, diagnosis of the disease is based on clinical findings: severe inflammation, with deformities of affected tissues, abscesses, fistulae, sinuses and discharge of purulent material that contains micro colonies of the etiologic agent. Microscopic examination of infected tissue is similar regardless of the offending microbe; hallmark of infected tissue is severe inflammation with abundant neutrophils around micro colonies and granuloma formation with macrophages, lymphocytes, dendritic and foamy cells. Even though medical treatment is available for mycetoma patients, amputation, or surgical intervention is frequently needed. The pathogenesis of actinomycetoma is little known, most information was obtained from experimental animal models infected with bacteria. In other experimental mice infections with different microbes, it was demonstrated that nitric oxide is responsible for the intracellular killing of Mycobacterium tuberculosis by activated macrophages. Nitric oxide is a free radical with potent stimulatory and suppressive effects in innate and adaptive immunity. The unstable nitric oxide molecule is produced by action of nitric oxide synthases on L-arginine. There are three nitric oxide synthases expressed in different cells and tissues, two are constitutively expressed one in neurons, and the other in endothelial cells and one that is inducible in macrophages. Aminoguanidine is a competitive inhibitor of inducible nitric oxide synthase. Its administration in experimental animals may favor or harm them. We used aminoguanidine in mice infected with Nocardia brasiliensis, and demonstrated that all treated animals were protected from actinomycetoma development. Anti N. brasiliensis antibodies and T cell proliferation were not affected, but inflammation was reduced.
Collapse
Affiliation(s)
- Mario C. Salinas-Carmona
- Universidad Autonoma de Nuevo Leon, Facultad de Medicina y Hospital Universitario, Servicio de Inmunología, Monterrey, Nuevo Leon, Mexico
- * E-mail:
| | - Ossian Longoria-Lozano
- Universidad Autonoma de Nuevo Leon, Facultad de Medicina y Hospital Universitario, Servicio de Inmunología, Monterrey, Nuevo Leon, Mexico
| | - Humberto R. Garza-Esquivel
- Universidad Autonoma de Nuevo Leon, Facultad de Medicina y Hospital Universitario, Servicio de Inmunología, Monterrey, Nuevo Leon, Mexico
| | - Juan López-Ulloa
- Universidad Autonoma de Nuevo Leon, Facultad de Medicina y Hospital Universitario, Servicio de Inmunología, Monterrey, Nuevo Leon, Mexico
| | - Jorge Reyes-Carrillo
- Universidad Autonoma de Nuevo Leon, Facultad de Medicina y Hospital Universitario, Servicio de Inmunología, Monterrey, Nuevo Leon, Mexico
| | - Anna Velia Vázquez-Marmolejo
- Universidad Autonoma de Nuevo Leon, Facultad de Medicina y Hospital Universitario, Servicio de Inmunología, Monterrey, Nuevo Leon, Mexico
| |
Collapse
|
42
|
Gossner A, Hassan MA. Transcriptional Analyses Identify Genes That Modulate Bovine Macrophage Response to Toxoplasma Infection and Immune Stimulation. Front Cell Infect Microbiol 2020; 10:437. [PMID: 33014886 PMCID: PMC7508302 DOI: 10.3389/fcimb.2020.00437] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/16/2020] [Indexed: 11/26/2022] Open
Abstract
The obligate intracellular parasite, Toxoplasma gondii, is highly prevalent among livestock species. Although cattle are generally resistant to Toxoplasma strains circulating in Europe and North America, the underlying mechanisms are largely unknown. Here, we report that bovine bone marrow-derived macrophage (BMDM) pre-stimulated with interferon gamma (IFNγ) restricts intracellular Toxoplasma growth independently of nitric oxide. While Toxoplasma promoted the expression of genes associated with alternative macrophage activation and lipid metabolism, IFNγ abrogated parasite-induced transcriptional responses and promoted the expression of genes linked to the classical macrophage activation phenotype. Additionally, several chemokines, including CCL22, that are linked to parasite-induced activation of the Wnt/β-catenin signaling were highly expressed in Toxoplasma-exposed naïve BMDMs. A chemical Wnt/β-catenin signaling pathway antagonist (IWR-1-endo) significantly reduced intracellular parasite burden in naïve BMDMs, suggesting that Toxoplasma activates this pathway to evade bovine macrophage anti-parasitic responses. Congruently, intracellular burden of a mutant Toxoplasma strain (RHΔASP5) that does not secrete dense granule proteins into the host cell, which is an essential requirement for parasite-induced activation of the Wnt/β-catenin pathway, was significantly reduced in naïve BMDMs. However, both the Wnt/β-catenin antagonist and RHASPΔ5 did not abolish parasite burden differences in naïve and IFNγ-stimulated BMDMs. Finally, we observed that parasites infecting IFNγ-stimulated BMDMs largely express genes associated with the slow dividing bradyzoite stage. Overall, this study provides novel insights into bovine macrophage transcriptional response to Toxoplasma. It establishes a foundation for a mechanistic analysis IFNγ-induced bovine anti-Toxoplasma responses and the counteracting Toxoplasma survival strategies.
Collapse
Affiliation(s)
- Anton Gossner
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Musa A Hassan
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom.,Centre for Tropical Livestock Genetics and Health, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
43
|
Still KM, Batista SJ, O’Brien CA, Oyesola OO, Früh SP, Webb LM, Smirnov I, Kovacs MA, Cowan MN, Hayes NW, Thompson JA, Tait Wojno ED, Harris TH. Astrocytes promote a protective immune response to brain Toxoplasma gondii infection via IL-33-ST2 signaling. PLoS Pathog 2020; 16:e1009027. [PMID: 33108405 PMCID: PMC7647122 DOI: 10.1371/journal.ppat.1009027] [Citation(s) in RCA: 24] [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: 05/06/2020] [Revised: 11/06/2020] [Accepted: 09/29/2020] [Indexed: 12/27/2022] Open
Abstract
It is of great interest to understand how invading pathogens are sensed within the brain, a tissue with unique challenges to mounting an immune response. The eukaryotic parasite Toxoplasma gondii colonizes the brain of its hosts, and initiates robust immune cell recruitment, but little is known about pattern recognition of T. gondii within brain tissue. The host damage signal IL-33 is one protein that has been implicated in control of chronic T. gondii infection, but, like many other pattern recognition pathways, IL-33 can signal peripherally, and the specific impact of IL-33 signaling within the brain is unclear. Here, we show that IL-33 is expressed by oligodendrocytes and astrocytes during T. gondii infection, is released locally into the cerebrospinal fluid of T. gondii-infected animals, and is required for control of infection. IL-33 signaling promotes chemokine expression within brain tissue and is required for the recruitment and/or maintenance of blood-derived anti-parasitic immune cells, including proliferating, IFN-γ-expressing T cells and iNOS-expressing monocytes. Importantly, we find that the beneficial effects of IL-33 during chronic infection are not a result of signaling on infiltrating immune cells, but rather on radio-resistant responders, and specifically, astrocytes. Mice with IL-33 receptor-deficient astrocytes fail to mount an adequate adaptive immune response in the CNS to control parasite burden-demonstrating, genetically, that astrocytes can directly respond to IL-33 in vivo. Together, these results indicate a brain-specific mechanism by which IL-33 is released locally, and sensed locally, to engage the peripheral immune system in controlling a pathogen.
Collapse
Affiliation(s)
- Katherine M. Still
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Samantha J. Batista
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Carleigh A. O’Brien
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Oyebola O. Oyesola
- Baker Institute for Animal Health and Department of Microbiology and Immunology, Cornell University, Ithaca, New York, United States of America
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
| | - Simon P. Früh
- Baker Institute for Animal Health and Department of Microbiology and Immunology, Cornell University, Ithaca, New York, United States of America
| | - Lauren M. Webb
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
| | - Igor Smirnov
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Michael A. Kovacs
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Maureen N. Cowan
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Nikolas W. Hayes
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jeremy A. Thompson
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Elia D. Tait Wojno
- Department of Immunology, University of Washington, Seattle, Washington, United States of America
| | - Tajie H. Harris
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
44
|
Shyu LY, Chen KM, Lu CY, Lai SC. Regulation of Proinflammatory Enzymes by Peroxisome Proliferator-Activated Receptor Gamma in Astroglia Infected with Toxoplasma gondii. J Parasitol 2020; 106:564-571. [PMID: 32916705 DOI: 10.1645/18-184] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) regulates neuroinflammation, and its agonists act as neuroprotective agents. This study aims to investigate the correlation between PPARγ and proinflammatory enzyme expression in astroglia infected with Toxoplasma gondii tachyzoite in vitro. Our results showed that matrix metalloprotease (MMP)-2, MMP-9, cyclooxygenase-2 (COX-2), prostaglandin (PGE)-2, inducible nitric-oxide synthase (iNOS), and nitric oxide (NO) were significantly increased in T. gondii-infected astroglia. Furthermore, the expression levels of MMP-2, MMP-9, COX-2, PGE-2, iNOS, and NO were significantly decreased by rosiglitazone-a PPARγ agonist. By contrast, the treatment with GW9662, a PPARγ antagonist, efficiently increased the expression levels of MMP-2, MMP-9, COX-2, PGE-2, iNOS, and NO. These results suggested that the treatment with rosiglitazone offers a potential strategy for controlling the inflammatory factors in T. gondii infection.
Collapse
Affiliation(s)
- Ling-Yuh Shyu
- Department of Parasitology, Chung Shan Medical University, 110, Section 1, Chien-Kuo North Road, Taichung 402, Taiwan
| | - Ke-Min Chen
- Department of Parasitology, Chung Shan Medical University, 110, Section 1, Chien-Kuo North Road, Taichung 402, Taiwan
| | - Cheng-You Lu
- Department of Parasitology, Chung Shan Medical University, 110, Section 1, Chien-Kuo North Road, Taichung 402, Taiwan
| | - Shih-Chan Lai
- Department of Parasitology, Chung Shan Medical University, 110, Section 1, Chien-Kuo North Road, Taichung 402, Taiwan.,Clinical Laboratory, Chung Shan Medical University Hospital, 110, Section 1, Chien-Kuo North Road, Taichung 402, Taiwan
| |
Collapse
|
45
|
Kupz A, Pai S, Giacomin PR, Whan JA, Walker RA, Hammoudi PM, Smith NC, Miller CM. Treatment of mice with S4B6 IL-2 complex prevents lethal toxoplasmosis via IL-12- and IL-18-dependent interferon-gamma production by non-CD4 immune cells. Sci Rep 2020; 10:13115. [PMID: 32753607 PMCID: PMC7403597 DOI: 10.1038/s41598-020-70102-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/23/2020] [Indexed: 01/08/2023] Open
Abstract
Toxoplasmic encephalitis is an AIDS-defining condition. The decline of IFN-γ-producing CD4+ T cells in AIDS is a major contributing factor in reactivation of quiescent Toxoplasma gondii to an actively replicating stage of infection. Hence, it is important to characterize CD4-independent mechanisms that constrain acute T. gondii infection. We investigated the in vivo regulation of IFN-γ production by CD8+ T cells, DN T cells and NK cells in response to acute T. gondii infection. Our data show that processing of IFN-γ by these non-CD4 cells is dependent on both IL-12 and IL-18 and the secretion of bioactive IL-18 in response to T. gondii requires the sensing of viable parasites by multiple redundant inflammasome sensors in multiple hematopoietic cell types. Importantly, our results show that expansion of CD8+ T cells, DN T cells and NK cell by S4B6 IL-2 complex pre-treatment increases survival rates of mice infected with T. gondii and this is dependent on IL-12, IL-18 and IFN-γ. Increased survival is accompanied by reduced pathology but is independent of expansion of TReg cells or parasite burden. This provides evidence for a protective role of IL2C-mediated expansion of non-CD4 cells and may represent a promising lead to adjunct therapy for acute toxoplasmosis.
Collapse
Affiliation(s)
- Andreas Kupz
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, 4878, Australia.
| | - Saparna Pai
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, 4878, Australia
| | - Paul R Giacomin
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, 4878, Australia
| | - Jennifer A Whan
- Advanced Analytical Centre, James Cook University, Cairns, QLD, 4878, Australia
| | - Robert A Walker
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, 4878, Australia
| | - Pierre-Mehdi Hammoudi
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Nicholas C Smith
- School of Science and Health, Western Sydney University, Parramatta South Campus, Sydney, NSW, 2116, Australia.,School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Catherine M Miller
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, 4878, Australia.,Discipline of Biomedicine, College of Public Health, Medical and Veterinary Science, James Cook University, Cairns, QLD, 4878, Australia
| |
Collapse
|
46
|
Briukhovetska D, Ohm B, Mey FT, Aliberti J, Kleingarn M, Huber-Lang M, Karsten CM, Köhl J. C5aR1 Activation Drives Early IFN-γ Production to Control Experimental Toxoplasma gondii Infection. Front Immunol 2020; 11:1397. [PMID: 32733463 PMCID: PMC7362728 DOI: 10.3389/fimmu.2020.01397] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/01/2020] [Indexed: 12/11/2022] Open
Abstract
Toxoplasma gondii (T. gondii) is a parasite infecting animals and humans. In intermediate hosts, such as humans or rodents, rapidly replicating tachyzoites drive vigorous innate and adaptive immune responses resulting in bradyzoites that survive within tissue cysts. Activation of the innate immune system is critical during the early phase of infection to limit pathogen growth and to instruct parasite-specific adaptive immunity. In rodents, dendritic cells (DCs) sense T. gondii through TLR11/12, leading to IL-12 production, which activates NK cells to produce IFN-γ as an essential mechanism for early parasite control. Further, C3 can bind to T. gondii resulting in limited complement activation. Here, we determined the role of C5a/C5aR1 axis activation for the early innate immune response in a mouse model of peritoneal T. gondii infection. We found that C5ar1−/− animals suffered from significantly higher weight loss, disease severity, mortality, and parasite burden in the brain than wild type control animals. Severe infection in C5ar1−/− mice was associated with diminished serum concentrations of IL-12, IL-27, and IFN-γ. Importantly, the serum levels of pro-inflammatory cytokines, including IL-1α, IL-6, and TNF-α, as well as several CXC and CC chemokines, were decreased in comparison to wt animals, whereas anti-inflammatory IL-10 was elevated. The defect in IFN-γ production was associated with diminished Ifng mRNA expression in the spleen and the brain, reduced frequency of IFN-γ+ NK cells in the spleen, and decreased Nos2 expression in the brain of C5ar1−/− mice. Mechanistically, DCs from the spleen of C5ar1−/− mice produced significantly less IL-12 in response to soluble tachyzoite antigen (STAg) stimulation in vivo and in vitro. Our findings suggest a model in which the C5a/C5aR1 axis promotes IL-12 induction in splenic DCs that is critical for IFN-γ production from NK cells and subsequent iNOS expression in the brain as a critical mechanism to control acute T. gondii infection.
Collapse
Affiliation(s)
- Daria Briukhovetska
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Birte Ohm
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Fabian T Mey
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Julio Aliberti
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Marie Kleingarn
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital of Ulm, Ulm, Germany
| | - Christian M Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany.,Division of Immunobiology, Cincinnati Children's Hospital and College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
47
|
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: 3] [Impact Index Per Article: 0.8] [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
|
48
|
Wong ZS, Borrelli SLS, Coyne CC, Boyle JP. Cell type- and species-specific host responses to Toxoplasma gondii and its near relatives. Int J Parasitol 2020; 50:423-431. [PMID: 32407716 PMCID: PMC8281328 DOI: 10.1016/j.ijpara.2020.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/30/2020] [Accepted: 04/03/2020] [Indexed: 10/24/2022]
Abstract
Toxoplasma gondii is remarkably unique in its ability to successfully infect vertebrate hosts from multiple phyla and can successfully infect most cells within these organisms. The infection outcome in each of these species is determined by the complex interaction between parasite and host genotype. As techniques to quantify global changes in cell function become more readily available and precise, new data are coming to light about how (i) different host cell types respond to parasitic infection and (ii) different parasite species impact the host. Here we focus on recent studies comparing the response to intracellular parasitism by different cell types and insights into understanding host-parasite interactions from comparative studies on T. gondii and its close extant relatives.
Collapse
Affiliation(s)
- Zhee S Wong
- Department of Biological Sciences, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, United States
| | - Sarah L Sokol Borrelli
- Department of Biological Sciences, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, United States
| | - Carolyn C Coyne
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jon P Boyle
- Department of Biological Sciences, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
49
|
Park J, Hunter CA. The role of macrophages in protective and pathological responses to Toxoplasma gondii. Parasite Immunol 2020; 42:e12712. [PMID: 32187690 DOI: 10.1111/pim.12712] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/12/2020] [Accepted: 02/24/2020] [Indexed: 02/06/2023]
Abstract
The ability of Toxoplasma gondii to cause clinical disease in immune-competent and immune-deficient hosts coupled with its ease of use in vitro and availability of murine models has led to its use as a model organism to study how the immune system controls an intracellular infection. This article reviews the studies that established the role of the cytokine IFN-γ in the activation of macrophages to control T gondii and the events that lead to the mobilization and expansion of macrophage populations and their ability to limit parasite replication. Macrophages also have pro-inflammatory functions that promote protective NK and T-cell activities as well as regulatory properties that facilitate the resolution of inflammation. Nevertheless, while macrophages are important in determining the outcome of infection, T gondii has evolved mechanisms to subvert macrophage activation and can utilize their migratory activities to promote dissemination and these two properties underlie the ability of this parasite to persist and cause disease.
Collapse
Affiliation(s)
- Jeongho Park
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA.,Kangwon National University College of Veterinary Medicine and Institute of Veterinary Science, Chuncheon, Korea
| | | |
Collapse
|
50
|
Coombs RS, Blank ML, English ED, Adomako-Ankomah Y, Urama ICS, Martin AT, Yarovinsky F, Boyle JP. Immediate Interferon Gamma Induction Determines Murine Host Compatibility Differences between Toxoplasma gondii and Neospora caninum. Infect Immun 2020; 88:e00027-20. [PMID: 32014892 PMCID: PMC7093116 DOI: 10.1128/iai.00027-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 12/24/2022] Open
Abstract
Rodents are critical for the transmission of Toxoplasma gondii to the definitive feline host via predation, and this relationship has been extensively studied as a model for immune responses to parasites. Neospora caninum is a closely related coccidian parasite of ruminants and canines but is not naturally transmitted by rodents. We compared mouse innate immune responses to N. caninum and T. gondii and found marked differences in cytokine levels and parasite growth kinetics during the first 24 h postinfection (hpi). N. caninum-infected mice produced significantly higher levels of interleukin-12 (IL-12) and interferon gamma (IFN-γ) by as early as 4 hpi, but the level of IFN-γ was significantly lower or undetectable in T. gondii-infected mice during the first 24 hpi. "Immediate" IFN-γ and IL-12p40 production was not detected in MyD88-/- mice. However, unlike IL-12p40-/- and IFN-γ-/- mice, MyD88-/- mice survived N. caninum infections at the dose used in this study. Serial measures of parasite burden showed that MyD88-/- mice were more susceptible to N. caninum infections than wild-type (WT) mice, and control of parasite burdens correlated with a pulse of serum IFN-γ at 3 to 4 days postinfection in the absence of detectable IL-12. Immediate IFN-γ was partially dependent on the T. gondii mouse profilin receptor Toll-like receptor 11 (TLR11), but the ectopic expression of N. caninum profilin in T. gondii had no impact on early IFN-γ production or parasite proliferation. Our data indicate that T. gondii is capable of evading host detection during the first hours after infection, while N. caninum is not, and this is likely due to the early MyD88-dependent recognition of ligands other than profilin.
Collapse
Affiliation(s)
- Rachel S Coombs
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Matthew L Blank
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Elizabeth D English
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yaw Adomako-Ankomah
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Andrew T Martin
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
| | - Felix Yarovinsky
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
| | - Jon P Boyle
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|