1
|
Henry B, Phillips AJ, Sibley LD, Rosenberg A. A combination of four Toxoplasma gondii nuclear-targeted effectors protects against interferon gamma-driven human host cell death. mBio 2024; 15:e0212424. [PMID: 39292011 PMCID: PMC11481881 DOI: 10.1128/mbio.02124-24] [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: 07/19/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
In both mice and humans, Type II interferon gamma (IFNγ) is crucial for the regulation of Toxoplasma gondii (T. gondii) infection, during acute or chronic phases. To thwart this defense, T. gondii secretes protein effectors hindering the host's immune response. For example, T. gondii relies on the MYR translocon complex to deploy soluble dense granule effectors (GRAs) into the host cell cytosol or nucleus. Recent genome-wide loss-of-function screens in IFNγ-primed primary human fibroblasts identified MYR translocon components as crucial for parasite resistance against IFNγ-driven vacuole clearance. However, these screens did not pinpoint specific MYR-dependent GRA proteins responsible for IFNγ signaling blockade, suggesting potential functional redundancy. Our study reveals that T. gondii depends on the MYR translocon complex to prevent parasite premature egress and host cell death in human cells stimulated with IFNγ post-infection, a unique phenotype observed in various human cell lines but not in murine cells. Intriguingly, inhibiting parasite egress did not prevent host cell death, indicating this mechanism is distinct from those described previously. Genome-wide loss-of-function screens uncovered TgIST, GRA16, GRA24, and GRA28 as effectors necessary for a complete block of IFNγ response. GRA24 and GRA28 directly influenced IFNγ-driven transcription, GRA24's action depended on its interaction with p38 MAPK, while GRA28 disrupted histone acetyltransferase activity of CBP/p300. Given the intricate nature of the immune response to T. gondii, it appears that the parasite has evolved equally elaborate mechanisms to subvert IFNγ signaling, extending beyond direct interference with the JAK/STAT1 pathway, to encompass other signaling pathways as well.IMPORTANCEToxoplasma gondii, an intracellular parasite, affects nearly one-third of the global human population, posing significant risks for immunocompromised patients and infants infected in utero. In murine models, the core mechanisms of IFNγ-mediated immunity against T. gondii are consistently preserved, showcasing a remarkable conservation of immune defense mechanisms. In humans, the recognized restriction mechanisms vary among cell types, lacking a universally applicable mechanism. This difference underscores a significant variation in the genes employed by T. gondii to shield itself against the IFNγ response in human vs murine cells. Here, we identified a specific combination of four parasite-secreted effectors deployed into the host cell nucleus, disrupting IFNγ signaling. This disruption is crucial in preventing premature egress of the parasite and host cell death. Notably, this phenotype is exclusive to human cells, highlighting the intricate and unique mechanisms T. gondii employs to modulate host responses in the human cellular environment.
Collapse
Affiliation(s)
- Brittany Henry
- Department of Infectious Diseases, Center for Tropical and Emerging Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Aubrey J. Phillips
- Department of Infectious Diseases, Center for Tropical and Emerging Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - L. David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alex Rosenberg
- Department of Infectious Diseases, Center for Tropical and Emerging Infectious Diseases, University of Georgia, Athens, Georgia, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
2
|
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
|
3
|
Henry B, Sibley LD, Rosenberg A. A Combination of Four Nuclear Targeted Effectors Protects Toxoplasma Against Interferon Gamma Driven Human Host Cell Death During Acute Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.24.573224. [PMID: 38234811 PMCID: PMC10793417 DOI: 10.1101/2023.12.24.573224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
In both mice and humans, Type II interferon-gamma (IFNγ) is crucial for regulation of Toxoplasma gondii (T. gondii) infection, during acute or chronic phases. To thwart this defense, T. gondii secretes protein effectors hindering the hosťs immune response. For example, T. gondii relies on the MYR translocon complex to deploy soluble dense granule effectors (GRAs) into the host cell cytosol or nucleus. Recent genome-wide loss-of-function screens in IFNγ-primed primary human fibroblasts identified MYR translocon components as crucial for parasite resistance against IFNγ driven vacuole clearance. However, these screens did not pinpoint specific MYR-dependent GRA proteins responsible for IFNγ signaling blockade, suggesting potential functional redundancy. Our study reveals that T. gondii depends on the MYR translocon complex to prevent host cell death and parasite premature egress in human cells stimulated with IFNγ postinfection, a unique phenotype observed in various human cell lines but not in murine cells. Intriguingly, inhibiting parasite egress did not prevent host cell death, indicating this mechanism is distinct from those described previously. Genome-wide loss-of-function screens uncovered TgIST, GRA16, GRA24, and GRA28 as effectors necessary for a complete block of IFNγ response. GRA24 and GRA28 directly influenced IFNγ driven transcription, GRA24's action depended on its interaction with p38 MAPK, while GRA28 disrupted histone acetyltransferase activity of CBP/p300. Given the intricate nature of the immune response to T. gondii, it appears that the parasite has evolved equally elaborate mechanisms to subvert IFNγ signaling, extending beyond direct interference with the JAK/STAT1 pathway, to encompass other signaling pathways as well.
Collapse
Affiliation(s)
- Brittany Henry
- Department of Infectious Diseases, Center for Tropical and Emerging Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - L. David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Alex Rosenberg
- Department of Infectious Diseases, Center for Tropical and Emerging Infectious Diseases, University of Georgia, Athens, Georgia, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
4
|
Wu X, Chen L, Sui C, Hu Y, Jiang D, Yang F, Miller LC, Li J, Cong X, Hrabchenko N, Lee C, Du Y, Qi J. 3C pro of FMDV inhibits type II interferon-stimulated JAK-STAT signaling pathway by blocking STAT1 nuclear translocation. Virol Sin 2023; 38:387-397. [PMID: 36921803 PMCID: PMC10311264 DOI: 10.1016/j.virs.2023.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 03/08/2023] [Indexed: 03/16/2023] Open
Abstract
Foot-and-mouth disease virus (FMDV) has developed various strategies to antagonize the host innate immunity. FMDV Lpro and 3Cpro interfere with type I IFNs through different mechanisms. The structural protein VP3 of FMDV degrades Janus kinase 1 to suppress IFN-γ signaling transduction. Whether non-structural proteins of FMDV are involved in restraining type II IFN signaling pathways is unknown. In this study, it was shown that FMDV replication was resistant to IFN-γ treatment after the infection was established and FMDV inhibited type II IFN induced expression of IFN-γ-stimulated genes (ISGs). We also showed for the first time that FMDV non-structural protein 3C antagonized IFN-γ-stimulated JAK-STAT signaling pathway by blocking STAT1 nuclear translocation. 3Cpro expression significantly reduced the ISGs transcript levels and palindromic gamma-activated sequences (GAS) promoter activity, without affecting the protein level, tyrosine phosphorylation, and homodimerization of STAT1. Finally, we provided evidence that 3C protease activity played an essential role in degrading KPNA1 and thus inhibited ISGs mRNA and GAS promoter activities. Our results reveal a novel mechanism by which an FMDV non-structural protein antagonizes host type II IFN signaling.
Collapse
Affiliation(s)
- Xiangju Wu
- Shandong Key Laboratory of Animal Disease Control and Breeding/Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Institute of Crop Germplasm Resources, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Lei Chen
- College of Life Science, Shandong Normal University, Jinan, 250358, China
| | - Chao Sui
- Shandong Key Laboratory of Animal Disease Control and Breeding/Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Institute of Crop Germplasm Resources, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Yue Hu
- Shandong Key Laboratory of Animal Disease Control and Breeding/Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Institute of Crop Germplasm Resources, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Dandan Jiang
- Shandong Key Laboratory of Animal Disease Control and Breeding/Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Institute of Crop Germplasm Resources, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Fan Yang
- State Key Laboratory of Veterinary Etiological Biology/National Foot and Mouth Disease Reference Laboratory/Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, China
| | - Laura C Miller
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA
| | - Juntong Li
- Shandong Key Laboratory of Animal Disease Control and Breeding/Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Institute of Crop Germplasm Resources, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Xiaoyan Cong
- Shandong Key Laboratory of Animal Disease Control and Breeding/Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Institute of Crop Germplasm Resources, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Nataliia Hrabchenko
- Shandong Key Laboratory of Animal Disease Control and Breeding/Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Institute of Crop Germplasm Resources, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Changhee Lee
- College of Veterinary Medicine and Virus Vaccine Research Center, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Yijun Du
- Shandong Key Laboratory of Animal Disease Control and Breeding/Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Institute of Crop Germplasm Resources, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; College of Life Science, Shandong Normal University, Jinan, 250358, China.
| | - Jing Qi
- Shandong Key Laboratory of Animal Disease Control and Breeding/Key Laboratory of Livestock and Poultry Multi-omics of MARA, Institute of Animal Science and Veterinary Medicine, Institute of Crop Germplasm Resources, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; College of Life Science, Shandong Normal University, Jinan, 250358, China.
| |
Collapse
|
5
|
Brandão YDO, Molento MB. A Systematic Review of Apicomplexa Looking into Epigenetic Pathways and the Opportunity for Novel Therapies. Pathogens 2023; 12:pathogens12020299. [PMID: 36839571 PMCID: PMC9963874 DOI: 10.3390/pathogens12020299] [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: 12/26/2022] [Revised: 02/03/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
Interest in host epigenetic changes during apicomplexan infections increased in the last decade, mainly due to the emergence of new therapies directed to these alterations. This review aims to carry out a bibliometric analysis of the publications related to host epigenetic changes during apicomplexan infections and to summarize the main studied pathways in this context, pointing out those that represent putative drug targets. We used four databases for the article search. After screening, 116 studies were included. The bibliometric analysis revealed that the USA and China had the highest number of relevant publications. The evaluation of the selected studies revealed that Toxoplasma gondii was considered in most of the studies, non-coding RNA was the most frequently reported epigenetic event, and host defense was the most explored pathway. These findings were reinforced by an analysis of the co-occurrence of keywords. Even though we present putative targets for repurposing epidrugs and ncRNA-based drugs in apicomplexan infections, we understand that more detailed knowledge of the hosts' epigenetic pathways is still needed before establishing a definitive drug target.
Collapse
|
6
|
Overview of Apoptosis, Autophagy, and Inflammatory Processes in Toxoplasma gondii Infected Cells. Pathogens 2023; 12:pathogens12020253. [PMID: 36839525 PMCID: PMC9966443 DOI: 10.3390/pathogens12020253] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/08/2023] Open
Abstract
Toxoplasma gondii (T. gondii) is an obligate intracellular parasite. During the parasitic invasion, T. gondii creates a parasitophorous vacuole, which enables the modulation of cell functions, allowing its replication and host infection. It has effective strategies to escape the immune response and reach privileged immune sites and remain inactive in a controlled environment in tissue cysts. This current review presents the factors that affect host cells and the parasite, as well as changes in the immune system during host cell infection. The secretory organelles of T. gondii (dense granules, micronemes, and rhoptries) are responsible for these processes. They are involved with proteins secreted by micronemes and rhoptries (MIC, AMA, and RONs) that mediate the recognition and entry into host cells. Effector proteins (ROP and GRA) that modify the STAT signal or GTPases in immune cells determine their toxicity. Interference byhost autonomous cells during parasitic infection, gene expression, and production of microbicidal molecules such as reactive oxygen species (ROS) and nitric oxide (NO), result in the regulation of cell death. The high level of complexity in host cell mechanisms prevents cell death in its various pathways. Many of these abilities play an important role in escaping host immune responses, particularly by manipulating the expression of genes involved in apoptosis, necrosis, autophagy, and inflammation. Here we present recent works that define the mechanisms by which T. gondii interacts with these processes in infected host cells.
Collapse
|
7
|
Huang Z, Liu H, Nix J, Xu R, Knoverek CR, Bowman GR, Amarasinghe GK, Sibley LD. The intrinsically disordered protein TgIST from Toxoplasma gondii inhibits STAT1 signaling by blocking cofactor recruitment. Nat Commun 2022; 13:4047. [PMID: 35831295 PMCID: PMC9279507 DOI: 10.1038/s41467-022-31720-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 06/28/2022] [Indexed: 12/31/2022] Open
Abstract
Signal transducer and activator of transcription (STAT) proteins communicate from cell-surface receptors to drive transcription of immune response genes. The parasite Toxoplasma gondii blocks STAT1-mediated gene expression by secreting the intrinsically disordered protein TgIST that traffics to the host nucleus, binds phosphorylated STAT1 dimers, and occupies nascent transcription sites that unexpectedly remain silenced. Here we define a core region within internal repeats of TgIST that is necessary and sufficient to block STAT1-mediated gene expression. Cellular, biochemical, mutational, and structural data demonstrate that the repeat region of TgIST adopts a helical conformation upon binding to STAT1 dimers. The binding interface is defined by a groove formed from two loops in the STAT1 SH2 domains that reorient during dimerization. TgIST binding to this newly exposed site at the STAT1 dimer interface alters its conformation and prevents the recruitment of co-transcriptional activators, thus defining the mechanism of blocked transcription.
Collapse
Affiliation(s)
- Zhou Huang
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hejun Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jay Nix
- Molecular Biology Consortium, Advanced Light Source, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Rui Xu
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Catherine R Knoverek
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Gregory R Bowman
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Gaya K Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - L David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
8
|
Hakimi MA. Epigenetic Reprogramming in Host-Parasite Coevolution: The Toxoplasma Paradigm. Annu Rev Microbiol 2022; 76:135-155. [PMID: 35587934 DOI: 10.1146/annurev-micro-041320-011520] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Like many intracellular pathogens, the protozoan parasite Toxoplasma gondii has evolved sophisticated mechanisms to promote its transmission and persistence in a variety of hosts by injecting effector proteins that manipulate many processes in the cells it invades. Specifically, the parasite diverts host epigenetic modulators and modifiers from their native functions to rewire host gene expression to counteract the innate immune response and to limit its strength. The arms race between the parasite and its hosts has led to accelerated adaptive evolution of effector proteins and the unconventional secretion routes they use. This review provides an up-to-date overview of how T. gondii effectors, through the evolution of intrinsically disordered domains, the formation of supramolecular complexes, and the use of molecular mimicry, target host transcription factors that act as coordinating nodes, as well as chromatin-modifying enzymes, to control the fate of infected cells and ultimately the outcome of infection. Expected final online publication date for the Annual Review of Microbiology, Volume 76 is September 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Mohamed-Ali Hakimi
- Host-Pathogen Interactions and Immunity to Infection, Institute for Advanced Biosciences (IAB), INSERM U1209, CNRS UMR 5309, Grenoble Alpes University, Grenoble, France;
| |
Collapse
|
9
|
Brar HK, Roy G, Kanojia A, Madan E, Madhubala R, Muthuswami R. Chromatin-Remodeling Factor BRG1 Is a Negative Modulator of L. donovani in IFNγ Stimulated and Infected THP-1 Cells. Front Cell Infect Microbiol 2022; 12:860058. [PMID: 35433496 PMCID: PMC9011159 DOI: 10.3389/fcimb.2022.860058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Intracellular pathogens manipulate the host cell for their own survival by contributing to modifications of host epigenome, and thus, altering expression of genes involved in the pathogenesis. Both ATP-dependent chromatin remodeling complex and histone modifications has been shown to be involved in the activation of IFNγ responsive genes. Leishmania donovani is an intracellular pathogen that causes visceral leishmaniasis. The strategies employed by Leishmania donovani to modulate the host epigenome in order to overcome the host defense for their persistence has been worked out in this study. We show that L. donovani negatively affects BRG1, a catalytic subunit of mammalian SWI/SNF chromatin remodeling complex, to alter IFNγ induced host responses. We observed that L. donovani infection downregulates BRG1 expression both at transcript and protein levels in cells stimulated with IFNγ. We also observed a significant decrease in IFNγ responsive gene, Class II transactivator (CIITA), as well as its downstream genes, MHC-II (HLA-DR and HLA-DM). Also, the occupancy of BRG1 at CIITA promoters I and IV was disrupted. A reversal in CIITA expression and decreased parasite load was observed with BRG1 overexpression, thus, suggesting BRG1 is a potential negative regulator for the survival of intracellular parasites in an early phase of infection. We also observed a decrease in H3 acetylation at the promoters of CIITA, post parasite infection. Silencing of HDAC1, resulted in increased CIITA expression, and further decreased parasite load. Taken together, we suggest that intracellular parasites in an early phase of infection negatively regulates BRG1 by using host HDAC1 for its survival inside the host.
Collapse
Affiliation(s)
- Harsimran Kaur Brar
- Molecular Parasitology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Gargi Roy
- Molecular Parasitology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Akanksha Kanojia
- Chromatin Remodeling Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Evanka Madan
- Molecular Parasitology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Rentala Madhubala
- Molecular Parasitology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
- *Correspondence: Rentala Madhubala, ; Rohini Muthuswami,
| | - Rohini Muthuswami
- Chromatin Remodeling Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
- *Correspondence: Rentala Madhubala, ; Rohini Muthuswami,
| |
Collapse
|
10
|
Matta SK, Rinkenberger N, Dunay IR, Sibley LD. Toxoplasma gondii infection and its implications within the central nervous system. Nat Rev Microbiol 2021; 19:467-480. [PMID: 33627834 DOI: 10.1038/s41579-021-00518-7] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2021] [Indexed: 01/31/2023]
Abstract
Toxoplasma gondii is a parasite that infects a wide range of animals and causes zoonotic infections in humans. Although it normally only results in mild illness in healthy individuals, toxoplasmosis is a common opportunistic infection with high mortality in individuals who are immunocompromised, most commonly due to reactivation of infection in the central nervous system. In the acute phase of infection, interferon-dependent immune responses control rapid parasite expansion and mitigate acute disease symptoms. However, after dissemination the parasite differentiates into semi-dormant cysts that form within muscle cells and neurons, where they persist for life in the infected host. Control of infection in the central nervous system, a compartment of immune privilege, relies on modified immune responses that aim to balance infection control while limiting potential damage due to inflammation. In response to the activation of interferon-mediated pathways, the parasite deploys an array of effector proteins to escape immune clearance and ensure latent survival. Although these pathways are best studied in the laboratory mouse, emerging evidence points to unique mechanisms of control in human toxoplasmosis. In this Review, we explore some of these recent findings that extend our understanding for proliferation, establishment and control of toxoplasmosis in humans.
Collapse
Affiliation(s)
- Sumit K Matta
- Department of Molecular Microbiology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Nicholas Rinkenberger
- Department of Molecular Microbiology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Ildiko R Dunay
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - L David Sibley
- Department of Molecular Microbiology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
11
|
Wang Y, Sangaré LO, Paredes-Santos TC, Hassan MA, Krishnamurthy S, Furuta AM, Markus BM, Lourido S, Saeij JPJ. Genome-wide screens identify Toxoplasma gondii determinants of parasite fitness in IFNγ-activated murine macrophages. Nat Commun 2020; 11:5258. [PMID: 33067458 PMCID: PMC7567896 DOI: 10.1038/s41467-020-18991-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 09/22/2020] [Indexed: 12/12/2022] Open
Abstract
Macrophages play an essential role in the early immune response against Toxoplasma and are the cell type preferentially infected by the parasite in vivo. Interferon gamma (IFNγ) elicits a variety of anti-Toxoplasma activities in macrophages. Using a genome-wide CRISPR screen we identify 353 Toxoplasma genes that determine parasite fitness in naїve or IFNγ-activated murine macrophages, seven of which are further confirmed. We show that one of these genes encodes dense granule protein GRA45, which has a chaperone-like domain, is critical for correct localization of GRAs into the PVM and secretion of GRA effectors into the host cytoplasm. Parasites lacking GRA45 are more susceptible to IFNγ-mediated growth inhibition and have reduced virulence in mice. Together, we identify and characterize an important chaperone-like GRA in Toxoplasma and provide a resource for the community to further explore the function of Toxoplasma genes that determine fitness in IFNγ-activated macrophages.
Collapse
Affiliation(s)
- Yifan Wang
- grid.27860.3b0000 0004 1936 9684Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
| | - Lamba Omar Sangaré
- grid.27860.3b0000 0004 1936 9684Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
| | - Tatiana C. Paredes-Santos
- grid.27860.3b0000 0004 1936 9684Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
| | - Musa A. Hassan
- grid.4305.20000 0004 1936 7988College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK ,grid.4305.20000 0004 1936 7988The Roslin Institute, The University of Edinburgh, Edinburgh, UK ,grid.4305.20000 0004 1936 7988Center for Tropical Livestock Health and Genetics, The University of Edinburgh, Edinburgh, UK
| | - Shruthi Krishnamurthy
- grid.27860.3b0000 0004 1936 9684Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
| | - Anna M. Furuta
- grid.27860.3b0000 0004 1936 9684Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
| | - Benedikt M. Markus
- grid.270301.70000 0001 2292 6283Whitehead Institute for Biomedical Research, Cambridge, MA USA ,grid.5963.9Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Sebastian Lourido
- grid.270301.70000 0001 2292 6283Whitehead Institute for Biomedical Research, Cambridge, MA USA ,grid.116068.80000 0001 2341 2786Department of Biology, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Jeroen P. J. Saeij
- grid.27860.3b0000 0004 1936 9684Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
| |
Collapse
|
12
|
Nast R, Choepak T, Lüder CGK. Epigenetic Control of IFN-γ Host Responses During Infection With Toxoplasma gondii. Front Immunol 2020; 11:581241. [PMID: 33072127 PMCID: PMC7544956 DOI: 10.3389/fimmu.2020.581241] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/26/2020] [Indexed: 01/21/2023] Open
Abstract
Host defense against the human pathogen Toxoplasma gondii depends on secretion of interferon (IFN)-γ and subsequent activation of monocytic cells to combat intracellular parasites. Previous studies have shown that T. gondii evades IFN-γ-mediated immunity by secreting the effector TgIST into the host cell where it binds to STAT1, strengthens its DNA binding activity and recruits the Mi-2/NuRD complex to STAT1-responsive promoters. Here we investigated the impact of the host chromatin environment on parasite interference with IFN-γ-induced gene expression. Luciferase reporters under control of primary and secondary IFN-γ response promoters were only inhibited by T. gondii when they were stably integrated into the host genome but not when expressed from a plasmid vector. Absence of CpG islands upstream and/or downstream of the transcriptional start site allowed more vigorous up-regulation by IFN-γ as compared to CpG-rich promoters. Remarkably, it also favored parasite interference with IFN-γ-induced gene expression indicating that nucleosome occupancy at IFN-γ-responsive promoters is important. Promoter DNA of IFN-γ-responsive genes remained largely non-methylated in T. gondii-infected cells, and inhibition of DNA methylation did not impact parasite interference with host responses. IFN-γ up-regulated histone marks H4ac, H3K9ac, and H3K4me3 but down-regulated H3S10p at primary and secondary response promoters. Infection with T. gondii abolished histone modification, whereas total nuclear activities of histone acetyl transferases and histone deacetylases were not altered. Taken together, our study reveals a critical impact of the host chromatin landscape at IFN-γ-activated promoters on their inhibition by T. gondii with a comprehensive blockade of histone modifications at parasite-inactivated promoters.
Collapse
Affiliation(s)
- Roswitha Nast
- Institute for Medical Microbiology, University Medical Center Goettingen, Georg-August-University, Göttingen, Germany
| | - Tenzin Choepak
- Institute for Medical Microbiology, University Medical Center Goettingen, Georg-August-University, Göttingen, Germany
| | - Carsten G K Lüder
- Institute for Medical Microbiology, University Medical Center Goettingen, Georg-August-University, Göttingen, Germany
| |
Collapse
|
13
|
Sabou M, Doderer-Lang C, Leyer C, Konjic A, Kubina S, Lennon S, Rohr O, Viville S, Cianférani S, Candolfi E, Pfaff AW, Brunet J. Toxoplasma gondii ROP16 kinase silences the cyclin B1 gene promoter by hijacking host cell UHRF1-dependent epigenetic pathways. Cell Mol Life Sci 2020; 77:2141-2156. [PMID: 31492965 PMCID: PMC7256068 DOI: 10.1007/s00018-019-03267-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/12/2019] [Accepted: 08/05/2019] [Indexed: 12/23/2022]
Abstract
Toxoplasmosis, caused by the apicomplexan parasite Toxoplasma gondii, is one of the most common infections in the world due to the lifelong persistence of this parasite in a latent stage. This parasite hijacks host signaling pathways through epigenetic mechanisms which converge on key nuclear proteins. Here, we report a new parasite persistence strategy involving T. gondii rhoptry protein ROP16 secreted early during invasion, which targets the transcription factor UHRF1 (ubiquitin-like containing PHD and RING fingers domain 1), and leads to host cell cycle arrest. This is mediated by DNMT activity and chromatin remodeling at the cyclin B1 gene promoter through recruitment of phosphorylated UHRF1 associated with a repressive multienzymatic protein complex. This leads to deacetylation and methylation of histone H3 surrounding the cyclin B1 promoter to epigenetically silence its transcriptional activity. Moreover, T. gondii infection causes DNA hypermethylation in its host cell, by upregulation of DNMTs. ROP16 is already known to activate and phosphorylate protective immunity transcription factors such as STAT 3/6/5 and modulate host signaling pathways in a strain-dependent manner. Like in the case of STAT6, the strain-dependent effects of ROP16 on UHRF1 are dependent on a single amino-acid polymorphism in ROP16. This study demonstrates that Toxoplasma hijacks a new epigenetic initiator, UHRF1, through an early event initiated by the ROP16 parasite kinase.
Collapse
Affiliation(s)
- Marcela Sabou
- Institut de Parasitologie et de Pathologie Tropicale de Strasbourg, « Dynamics of Host-Pathogen Interactions » EA 7292, Fédération de Médecine Translationelle Université de Strasbourg, Strasbourg, France
- Service de Parasitologie et Mycologie Médicale, Hôpitaux Universitaires de Strasbourg, Centre National de Référence de la Toxoplasmose, Pôle Sérologie, Strasbourg, France
| | - Cécile Doderer-Lang
- Institut de Parasitologie et de Pathologie Tropicale de Strasbourg, « Dynamics of Host-Pathogen Interactions » EA 7292, Fédération de Médecine Translationelle Université de Strasbourg, Strasbourg, France
| | - Caroline Leyer
- Institut de Parasitologie et de Pathologie Tropicale de Strasbourg, « Dynamics of Host-Pathogen Interactions » EA 7292, Fédération de Médecine Translationelle Université de Strasbourg, Strasbourg, France
| | - Ana Konjic
- Institut de Parasitologie et de Pathologie Tropicale de Strasbourg, « Dynamics of Host-Pathogen Interactions » EA 7292, Fédération de Médecine Translationelle Université de Strasbourg, Strasbourg, France
| | - Sophie Kubina
- Institut de Parasitologie et de Pathologie Tropicale de Strasbourg, « Dynamics of Host-Pathogen Interactions » EA 7292, Fédération de Médecine Translationelle Université de Strasbourg, Strasbourg, France
| | - Sarah Lennon
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, IPHC, CNRS, UMR7178, Strasbourg, France
| | - Olivier Rohr
- Institut de Parasitologie et de Pathologie Tropicale de Strasbourg, « Dynamics of Host-Pathogen Interactions » EA 7292, Fédération de Médecine Translationelle Université de Strasbourg, Strasbourg, France
| | - Stéphane Viville
- Institut de Parasitologie et de Pathologie Tropicale de Strasbourg, « Dynamics of Host-Pathogen Interactions » EA 7292, Fédération de Médecine Translationelle Université de Strasbourg, Strasbourg, France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), Université de Strasbourg, IPHC, CNRS, UMR7178, Strasbourg, France
| | - Ermanno Candolfi
- Institut de Parasitologie et de Pathologie Tropicale de Strasbourg, « Dynamics of Host-Pathogen Interactions » EA 7292, Fédération de Médecine Translationelle Université de Strasbourg, Strasbourg, France
- Service de Parasitologie et Mycologie Médicale, Hôpitaux Universitaires de Strasbourg, Centre National de Référence de la Toxoplasmose, Pôle Sérologie, Strasbourg, France
| | - Alexander W Pfaff
- Institut de Parasitologie et de Pathologie Tropicale de Strasbourg, « Dynamics of Host-Pathogen Interactions » EA 7292, Fédération de Médecine Translationelle Université de Strasbourg, Strasbourg, France.
- Service de Parasitologie et Mycologie Médicale, Hôpitaux Universitaires de Strasbourg, Centre National de Référence de la Toxoplasmose, Pôle Sérologie, Strasbourg, France.
| | - Julie Brunet
- Institut de Parasitologie et de Pathologie Tropicale de Strasbourg, « Dynamics of Host-Pathogen Interactions » EA 7292, Fédération de Médecine Translationelle Université de Strasbourg, Strasbourg, France
- Service de Parasitologie et Mycologie Médicale, Hôpitaux Universitaires de Strasbourg, Centre National de Référence de la Toxoplasmose, Pôle Sérologie, Strasbourg, France
| |
Collapse
|
14
|
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: 5] [Impact Index Per Article: 1.0] [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
|
15
|
Lecoeur H, Prina E, Rosazza T, Kokou K, N’Diaye P, Aulner N, Varet H, Bussotti G, Xing Y, Milon G, Weil R, Meng G, Späth GF. Targeting Macrophage Histone H3 Modification as a Leishmania Strategy to Dampen the NF-κB/NLRP3-Mediated Inflammatory Response. Cell Rep 2020; 30:1870-1882.e4. [DOI: 10.1016/j.celrep.2020.01.030] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/08/2019] [Accepted: 01/08/2020] [Indexed: 12/21/2022] Open
|
16
|
Poncet AF, Blanchard N, Marion S. Toxoplasma and Dendritic Cells: An Intimate Relationship That Deserves Further Scrutiny. Trends Parasitol 2019; 35:870-886. [PMID: 31492624 DOI: 10.1016/j.pt.2019.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/01/2019] [Accepted: 08/04/2019] [Indexed: 02/07/2023]
Abstract
Toxoplasma gondii (Tg), an obligate intracellular parasite of the phylum Apicomplexa, infects a wide range of animals, including humans. A hallmark of Tg infection is the subversion of host responses, which is thought to favor parasite persistence and propagation to new hosts. Recently, a variety of parasite-secreted modulatory effectors have been uncovered in fibroblasts and macrophages, but the specific interplay between Tg and dendritic cells (DCs) is just beginning to emerge. In this review, we summarize the current knowledge on Tg-DC interactions, including innate recognition, cytokine production, and antigen presentation, and discuss open questions regarding how Tg-secreted effectors may shape DC functions to perturb innate and adaptive immunity.
Collapse
Affiliation(s)
- Anaïs F Poncet
- Centre d'Infection et d'Immunité de Lille, Université de Lille, Inserm U1019, CNRS UMR 8204, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Nicolas Blanchard
- Centre de Physiopathologie Toulouse Purpan (CPTP), Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France. @inserm.fr
| | - Sabrina Marion
- Centre d'Infection et d'Immunité de Lille, Université de Lille, Inserm U1019, CNRS UMR 8204, CHU Lille, Institut Pasteur de Lille, Lille, France. @pasteur-lille.fr
| |
Collapse
|
17
|
Lu S, Li D, Xi L, Calderone R. Interplay of interferon-gamma and macrophage polarization during Talaromyces marneffei infection. Microb Pathog 2019; 134:103594. [PMID: 31199985 DOI: 10.1016/j.micpath.2019.103594] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/29/2019] [Accepted: 06/10/2019] [Indexed: 12/19/2022]
Abstract
Talaromyces marneffei is an increasingly destructive dimorphic fungal pathogen in clinical settings that can cause lethal Talaromycosis. The activation of macrophages is known to be important for host defenses against T. marneffei, and these macrophages are known to be activated in two ways (polarization), known as M1 and M2. We investigated the plasticity of these polarizations, in order to understand if cross-conversion of macrophages may be possible even after they have been programmed. We conducted in vitro experiments using a murine macrophage cell line to investigate the ability of T. marneffei to activate these polarizations. The pre-polarized (M0) macrophage subsets were challenged with LPS as a control, and the sets of M1 markers (iNOS and CD86) and M2 markers (Arg-1 and CD206) were assessed for a possible cross-conversion among M1, M2 and M0 (unstimulated) populations. We found that either conidia or yeast forms of T. marneffei initiate the repression of Arg-1 in M2 cells with no change in the M1 subtype marker molecule iNOS. However, an additional IFN-γ stimulus caused the three macrophage groups to fully exhibit an LPS-induced M2 suppression and a shift to M1 from M0 and M2. We conclude that the conversion of macrophages is required for maintenance of sufficient iNOS production against this organism in the host. The cytokine environment is the key factor that manipulates the plasticity changes among macrophage subtypes. Furthermore, IFN-γ is a crucial host defense factor against pathogenic T. marneffei that has significant therapeutic potential to promote an M1 polarization phenotype.
Collapse
Affiliation(s)
- Sha Lu
- Department of Dermatology and Venereology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC, United States
| | - Dongmei Li
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC, United States
| | - Liyan Xi
- Department of Dermatology and Venereology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Richard Calderone
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC, United States.
| |
Collapse
|
18
|
Deng R, Zhang P, Liu W, Zeng X, Ma X, Shi L, Wang T, Yin Y, Chang W, Zhang P, Wang G, Tao K. HDAC is indispensable for IFN-γ-induced B7-H1 expression in gastric cancer. Clin Epigenetics 2018; 10:153. [PMID: 30537988 PMCID: PMC6288935 DOI: 10.1186/s13148-018-0589-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 11/21/2018] [Indexed: 12/13/2022] Open
Abstract
Background B7 homolog 1 (B7-H1) overexpression on tumor cells is an important mechanism of immune evasion in gastric cancer (GC). Elucidation of the regulation of B7-H1 expression is urgently required to guide B7-H1-targeted cancer therapy. Interferon gamma (IFN-γ) is thought to be the main driving force behind B7-H1 expression, and epigenetic factors including histone acetylation are recently linked to the process. Here, we investigated the potential role of histone deacetylase (HDAC) in IFN-γ-induced B7-H1 expression in GC. The effect of Vorinostat (SAHA), a small molecular inhibitor of HDAC, on tumor growth and B7-H1 expression in a mouse GC model was also evaluated. Results RNA-seq data from The Cancer Genome Atlas revealed that expression of B7-H1, HDAC1–3, 6–8, and 10 and SIRT1, 3, 5, and 6 was higher, and expression of HDAC5 and SIRT4 was lower in GC compared to that in normal gastric tissues; that HDAC3 and HDAC1 expression level significantly correlated with B7-H1 in GC with a respective r value of 0.42 (p < 0.001) and 0.21 (p < 0.001). HDAC inhibitor (Trichostatin A, SAHA, and sodium butyrate) pretreatment suppressed IFN-γ-induced B7-H1 expression on HGC-27 cells. HDAC1 and HDAC3 gene knockdown had the same effect. SAHA pretreatment or HDAC knockdown resulted in impaired IFN-γ signaling, demonstrated by the reduction of JAK2, p-JAK1, p-JAK2, and p-STAT1 expression and inefficient STAT1 nuclear translocation. Furthermore, SAHA pretreatment compromised IFN-γ-induced upregulation of histone H3 lysine 9 acetylation level in B7-H1 gene promoter. In the grafted mouse GC model, SAHA treatment suppressed tumor growth, inhibited B7-H1 expression, and elevated the percentage of tumor-infiltrating CD8+ T cells. Conclusion HDAC is indispensable for IFN-γ-induced B7-H1 in GC. The study suggests the possibility of targeting B7-H1 using small molecular HDAC inhibitors for cancer treatment. Electronic supplementary material The online version of this article (10.1186/s13148-018-0589-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rui Deng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Department of General Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Peng Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Weizhen Liu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiangyu Zeng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xianxiong Ma
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Liang Shi
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yuping Yin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Weilong Chang
- Department of General Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Pei Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guobin Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
19
|
Nast R, Staab J, Meyer T, Lüder CGK. Toxoplasma gondii stabilises tetrameric complexes of tyrosine-phosphorylated signal transducer and activator of transcription-1 and leads to its sustained and promiscuous DNA binding. Cell Microbiol 2018; 20:e12887. [PMID: 29968354 DOI: 10.1111/cmi.12887] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/07/2018] [Accepted: 06/25/2018] [Indexed: 11/29/2022]
Abstract
Toxoplasma gondii is an obligate intracellular parasite that infects up to 30% of humans worldwide. It can lead to severe diseases particularly in individuals with immature or defective immune responses. Control of T. gondii relies on the IFN-γ-induced signal transducer and activator of transcription-1 (STAT1) pathway. T. gondii, however, largely inactivates STAT1-mediated gene transcription by T. gondii inhibitor of STAT1-dependent transcription (TgIST), a parasite effector protein binding to STAT1. Here, we have analysed requirements of STAT1 to bind TgIST and characterised downstream effects on STAT1 signalling. TgIST bound to STAT1 dimers but more efficiently assembled with STAT1 tetramers, which are essential for effective IFN-γ responsiveness. Such binding was abrogated in N-terminal, but not C-terminal deletion mutants of STAT1. Furthermore, TgIST did not bind to the STAT1F77A substitution mutant that cannot form STAT1 tetramers, resulting in a complete unresponsiveness of parasite-infected STAT1F77A -expressing cells to IFN-γ. Remarkably, binding of TgIST considerably increased the affinity of the aberrant STAT1 tetramers for DNA consensus sequence binding motifs and even enabled binding to nonconsensus sequences. Consistent with the increased DNA binding, STAT1 from parasite-infected cells remained phosphorylated at Tyr701 and Ser727 and was retained within the nucleus in a DNA-bound state. The sustained and promiscuous binding activity particularly of STAT1 tetramers to unspecific DNA sites lacking a consensus STAT1-binding motif is an as yet unrecognised mechanism contributing to the defective IFN-γ-mediated signalling in T. gondii-infected cells.
Collapse
Affiliation(s)
- Roswitha Nast
- Institute for Medical Microbiology, University Medical Center Goettingen, Georg-August University, Göttingen, Germany
| | - Julia Staab
- Psychosomatic Medicine and Psychotherapy, University Medical Center Goettingen, Georg-August University, Göttingen, Germany
| | - Thomas Meyer
- Psychosomatic Medicine and Psychotherapy, University Medical Center Goettingen, Georg-August University, Göttingen, Germany
| | - Carsten G K Lüder
- Institute for Medical Microbiology, University Medical Center Goettingen, Georg-August University, Göttingen, Germany
| |
Collapse
|
20
|
Abstract
INTRODUCTION Parasitic diseases that pose a threat to human life include leishmaniasis - caused by protozoan parasite Leishmania species. Existing drugs have limitations due to deleterious side effects like teratogenicity, high cost and drug resistance. This calls for the need to have an insight into therapeutic aspects of disease. Areas covered: We have identified different drug targets via. molecular, imuunological, metabolic as well as by system biology approaches. We bring these promising drug targets into light so that they can be explored to their maximum. In an effort to bridge the gaps between existing knowledge and prospects of drug discovery, we have compiled interesting studies on drug targets, thereby paving the way for establishment of better therapeutic aspects. Expert opinion: Advancements in technology shed light on many unexplored pathways. Further probing of well established pathways led to the discovery of new drug targets. This review is a comprehensive report on current and emerging drug targets, with emphasis on several metabolic targets, organellar biochemistry, salvage pathways, epigenetics, kinome and more. Identification of new targets can contribute significantly towards strengthening the pipeline for disease elimination.
Collapse
Affiliation(s)
- Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi-221 005, UP, India
| | - Bhawana Singh
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi-221 005, UP, India
| |
Collapse
|
21
|
Syn G, Blackwell JM, Jamieson SE, Francis RW. An in silico pipeline to filter the Toxoplasma gondii proteome for proteins that could traffic to the host cell nucleus and influence host cell epigenetic regulation. Mem Inst Oswaldo Cruz 2018; 113:e170471. [PMID: 29846382 PMCID: PMC5963570 DOI: 10.1590/0074-02760170471] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 04/17/2018] [Indexed: 12/11/2022] Open
Abstract
Toxoplasma gondii uses epigenetic mechanisms to regulate both endogenous and host cell gene expression. To identify genes with putative epigenetic functions, we developed an in silico pipeline to interrogate the T. gondii proteome of 8313 proteins. Step 1 employs PredictNLS and NucPred to identify genes predicted to target eukaryotic nuclei. Step 2 uses GOLink to identify proteins of epigenetic function based on Gene Ontology terms. This resulted in 611 putative nuclear localised proteins with predicted epigenetic functions. Step 3 filtered for secretory proteins using SignalP, SecretomeP, and experimental data. This identified 57 of the 611 putative epigenetic proteins as likely to be secreted. The pipeline is freely available online, uses open access tools and software with user-friendly Perl scripts to automate and manage the results, and is readily adaptable to undertake any such in silico search for genes contributing to particular functions.
Collapse
Affiliation(s)
- Genevieve Syn
- Telethon Kids Institute, University of Western Australia, Subiaco, Western Australia, Australia
| | - Jenefer M Blackwell
- Telethon Kids Institute, University of Western Australia, Subiaco, Western Australia, Australia
| | - Sarra E Jamieson
- Telethon Kids Institute, University of Western Australia, Subiaco, Western Australia, Australia
| | - Richard W Francis
- Telethon Kids Institute, University of Western Australia, Subiaco, Western Australia, Australia
| |
Collapse
|
22
|
Brasil TR, Freire-de-Lima CG, Morrot A, Vetö Arnholdt AC. Host- Toxoplasma gondii Coadaptation Leads to Fine Tuning of the Immune Response. Front Immunol 2017; 8:1080. [PMID: 28955329 PMCID: PMC5601305 DOI: 10.3389/fimmu.2017.01080] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/21/2017] [Indexed: 12/22/2022] Open
Abstract
Toxoplasma gondii has successfully developed strategies to evade host's immune response and reach immune privileged sites, which remains in a controlled environment inside quiescent tissue cysts. In this review, we will approach several known mechanisms used by the parasite to modulate mainly the murine immune system at its favor. In what follows, we review recent findings revealing interference of host's cell autonomous immunity and cell signaling, gene expression, apoptosis, and production of microbicide molecules such as nitric oxide and oxygen reactive species during parasite infection. Modulation of host's metalloproteinases of extracellular matrix is also discussed. These immune evasion strategies are determinant to parasite dissemination throughout the host taking advantage of cells from the immune system to reach brain and retina, crossing crucial hosts' barriers.
Collapse
Affiliation(s)
- Thaís Rigueti Brasil
- Laboratório de Biologia do Reconhecer, Universidade Estadual do Norte Fluminense, Rio de Janeiro, Brazil
| | | | - Alexandre Morrot
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | | |
Collapse
|
23
|
Sumpf K, Nast R, Downie B, Salinas G, Lüder CG. Histone deacetylase inhibitor MS-275 augments expression of a subset of IFN-γ-regulated genes in Toxoplasma gondii-infected macrophages but does not improve parasite control. Exp Parasitol 2017; 180:45-54. [DOI: 10.1016/j.exppara.2017.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 01/10/2017] [Accepted: 02/07/2017] [Indexed: 01/17/2023]
|
24
|
Olias P, Etheridge RD, Zhang Y, Holtzman MJ, Sibley LD. Toxoplasma Effector Recruits the Mi-2/NuRD Complex to Repress STAT1 Transcription and Block IFN-γ-Dependent Gene Expression. Cell Host Microbe 2017; 20:72-82. [PMID: 27414498 DOI: 10.1016/j.chom.2016.06.006] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/09/2016] [Accepted: 06/09/2016] [Indexed: 12/12/2022]
Abstract
Interferon gamma (IFN-γ) is an essential mediator of host defense against intracellular pathogens, including the protozoan parasite Toxoplasma gondii. However, prior T. gondii infection blocks IFN-γ-dependent gene transcription, despite the downstream transcriptional activator STAT1 being activated and bound to cognate nuclear promoters. We identify the parasite effector that blocks STAT1-dependent transcription and show it is associated with recruitment of the Mi-2 nucleosome remodeling and deacetylase (NuRD) complex, a chromatin-modifying repressor. This secreted effector, toxoplasma inhibitor of STAT1-dependent transcription (TgIST), translocates to the host cell nucleus, where it recruits Mi-2/NuRD to STAT1-dependent promoters, resulting in altered chromatin and blocked transcription. TgIST is conserved across strains, underlying their shared ability to block IFN-γ-dependent transcription. TgIST deletion results in increased parasite clearance in IFN-γ-activated cells and reduced mouse virulence, which is restored in IFN-γ-receptor-deficient mice. These findings demonstrate the importance of both IFN-γ responses and the ability of pathogens to counteract these defenses.
Collapse
Affiliation(s)
- Philipp Olias
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ronald D Etheridge
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yong Zhang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael J Holtzman
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - L David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
25
|
Swierzy IJ, Händel U, Kaever A, Jarek M, Scharfe M, Schlüter D, Lüder CGK. Divergent co-transcriptomes of different host cells infected with Toxoplasma gondii reveal cell type-specific host-parasite interactions. Sci Rep 2017; 7:7229. [PMID: 28775382 PMCID: PMC5543063 DOI: 10.1038/s41598-017-07838-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 06/30/2017] [Indexed: 02/06/2023] Open
Abstract
The apicomplexan parasite Toxoplasma gondii infects various cell types in avian and mammalian hosts including humans. Infection of immunocompetent hosts is mostly asymptomatic or benign, but leads to development of largely dormant bradyzoites that persist predominantly within neurons and muscle cells. Here we have analyzed the impact of the host cell type on the co-transcriptomes of host and parasite using high-throughput RNA sequencing. Murine cortical neurons and astrocytes, skeletal muscle cells (SkMCs) and fibroblasts differed by more than 16,200 differentially expressed genes (DEGs) before and after infection with T. gondii. However, only a few hundred of them were regulated by infection and these largely diverged in neurons, SkMCs, astrocytes and fibroblasts indicating host cell type-specific transcriptional responses after infection. The heterogeneous transcriptomes of host cells before and during infection coincided with ~5,400 DEGs in T. gondii residing in different cell types. Finally, we identified gene clusters in both T. gondii and its host, which correlated with the predominant parasite persistence in neurons or SkMCs as compared to astrocytes or fibroblasts. Thus, heterogeneous expression profiles of different host cell types and the parasites’ ability to adapting to them may govern the parasite-host cell interaction during toxoplasmosis.
Collapse
Affiliation(s)
- Izabela J Swierzy
- Institute for Medical Microbiology, University Medical Center, Georg-August-University, 37075, Göttingen, Germany
| | - Ulrike Händel
- Institute of Medical Microbiology, Infection Control and Prevention, Otto-von-Guericke-University, 39120, Magdeburg, Germany
| | - Alexander Kaever
- Institute of Microbiology and Genetics, Department of Bioinformatics, Georg-August-University, 37077, Göttingen, Germany
| | - Michael Jarek
- Genome Analytics, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany
| | - Maren Scharfe
- Genome Analytics, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany
| | - Dirk Schlüter
- Institute of Medical Microbiology, Infection Control and Prevention, Otto-von-Guericke-University, 39120, Magdeburg, Germany.,Organ-Specific Immune Regulation, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany
| | - Carsten G K Lüder
- Institute for Medical Microbiology, University Medical Center, Georg-August-University, 37075, Göttingen, Germany.
| |
Collapse
|
26
|
Lamotte S, Späth GF, Rachidi N, Prina E. The enemy within: Targeting host-parasite interaction for antileishmanial drug discovery. PLoS Negl Trop Dis 2017; 11:e0005480. [PMID: 28594938 PMCID: PMC5464532 DOI: 10.1371/journal.pntd.0005480] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The state of antileishmanial chemotherapy is strongly compromised by the emergence of drug-resistant Leishmania. The evolution of drug-resistant phenotypes has been linked to the parasites’ intrinsic genome instability, with frequent gene and chromosome amplifications causing fitness gains that are directly selected by environmental factors, including the presence of antileishmanial drugs. Thus, even though the unique eukaryotic biology of Leishmania and its dependence on parasite-specific virulence factors provide valid opportunities for chemotherapeutical intervention, all strategies that target the parasite in a direct fashion are likely prone to select for resistance. Here, we review the current state of antileishmanial chemotherapy and discuss the limitations of ongoing drug discovery efforts. We finally propose new strategies that target Leishmania viability indirectly via mechanisms of host–parasite interaction, including parasite-released ectokinases and host epigenetic regulation, which modulate host cell signaling and transcriptional regulation, respectively, to establish permissive conditions for intracellular Leishmania survival.
Collapse
Affiliation(s)
- Suzanne Lamotte
- Institut Pasteur and INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
| | - Gerald F. Späth
- Institut Pasteur and INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
| | - Najma Rachidi
- Institut Pasteur and INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
| | - Eric Prina
- Institut Pasteur and INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
- * E-mail:
| |
Collapse
|
27
|
Abstract
Early electron microscopy studies revealed the elaborate cellular features that define the unique adaptations of apicomplexan parasites. Among these were bulbous rhoptry (ROP) organelles and small, dense granules (GRAs), both of which are secreted during invasion of host cells. These early morphological studies were followed by the exploration of the cellular contents of these secretory organelles, revealing them to be comprised of highly divergent protein families with few conserved domains or predicted functions. In parallel, studies on host-pathogen interactions identified many host signaling pathways that were mysteriously altered by infection. It was only with the advent of forward and reverse genetic strategies that the connections between individual parasite effectors and the specific host pathways that they targeted finally became clear. The current repertoire of parasite effectors includes ROP kinases and pseudokinases that are secreted during invasion and that block host immune pathways. Similarly, many secretory GRA proteins alter host gene expression by activating host transcription factors, through modification of chromatin, or by inducing small noncoding RNAs. These effectors highlight novel mechanisms by which T. gondii has learned to harness host signaling to favor intracellular survival and will guide future studies designed to uncover the additional complexity of this intricate host-pathogen interaction.
Collapse
|
28
|
STAT1 Signaling in Astrocytes Is Essential for Control of Infection in the Central Nervous System. mBio 2016; 7:mBio.01881-16. [PMID: 27834206 PMCID: PMC5101356 DOI: 10.1128/mbio.01881-16] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The local production of gamma interferon (IFN-γ) is important to control Toxoplasma gondii in the brain, but the basis for these protective effects is not fully understood. The studies presented here reveal that the ability of IFN-γ to inhibit parasite replication in astrocytes in vitro is dependent on signal transducer and activator of transcription 1 (STAT1) and that mice that specifically lack STAT1 in astrocytes are unable to limit parasite replication in the central nervous system (CNS). This susceptibility is associated with a loss of antimicrobial pathways and increased cyst formation in astrocytes. These results identify a critical role for astrocytes in limiting the replication of an important opportunistic pathogen. Astrocytes are the most numerous cell type in the brain, and they are activated in response to many types of neuroinflammation, but their function in the control of CNS-specific infection is unclear. The parasite Toxoplasma gondii is one of the few clinically relevant microorganisms that naturally infects astrocytes, and the studies presented here establish that the ability of astrocytes to inhibit parasite replication is essential for the local control of this opportunistic pathogen. Together, these studies establish a key role for astrocytes as effector cells and in the coordination of many aspects of the protective immune response that operates in the brain.
Collapse
|
29
|
Gay G, Braun L, Brenier-Pinchart MP, Vollaire J, Josserand V, Bertini RL, Varesano A, Touquet B, De Bock PJ, Coute Y, Tardieux I, Bougdour A, Hakimi MA. Toxoplasma gondii TgIST co-opts host chromatin repressors dampening STAT1-dependent gene regulation and IFN-γ-mediated host defenses. J Exp Med 2016; 213:1779-98. [PMID: 27503074 PMCID: PMC4995087 DOI: 10.1084/jem.20160340] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 07/22/2016] [Indexed: 11/04/2022] Open
Abstract
An early hallmark of Toxoplasma gondii infection is the rapid control of the parasite population by a potent multifaceted innate immune response that engages resident and homing immune cells along with pro- and counter-inflammatory cytokines. In this context, IFN-γ activates a variety of T. gondii-targeting activities in immune and nonimmune cells but can also contribute to host immune pathology. T. gondii has evolved mechanisms to timely counteract the host IFN-γ defenses by interfering with the transcription of IFN-γ-stimulated genes. We now have identified TgIST (T. gondii inhibitor of STAT1 transcriptional activity) as a critical molecular switch that is secreted by intracellular parasites and traffics to the host cell nucleus where it inhibits STAT1-dependent proinflammatory gene expression. We show that TgIST not only sequesters STAT1 on dedicated loci but also promotes shaping of a nonpermissive chromatin through its capacity to recruit the nucleosome remodeling deacetylase (NuRD) transcriptional repressor. We found that during mice acute infection, TgIST-deficient parasites are rapidly eliminated by the homing Gr1(+) inflammatory monocytes, thus highlighting the protective role of TgIST against IFN-γ-mediated killing. By uncovering TgIST functions, this study brings novel evidence on how T. gondii has devised a molecular weapon of choice to take control over a ubiquitous immune gene expression mechanism in metazoans, as a way to promote long-term parasitism.
Collapse
Affiliation(s)
- Gabrielle Gay
- Institute for Advanced Biosciences (IAB), Team Host-Pathogen Interactions and Immunity to Infection, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, F-38700 Grenoble, France
| | - Laurence Braun
- Institute for Advanced Biosciences (IAB), Team Host-Pathogen Interactions and Immunity to Infection, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, F-38700 Grenoble, France
| | - Marie-Pierre Brenier-Pinchart
- Institute for Advanced Biosciences (IAB), Team Host-Pathogen Interactions and Immunity to Infection, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, F-38700 Grenoble, France
| | - Julien Vollaire
- IAB, OPTIMAL Small Animal Imaging Facility, 38000 Grenoble, France
| | | | - Rose-Laurence Bertini
- Institute for Advanced Biosciences (IAB), Team Host-Pathogen Interactions and Immunity to Infection, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, F-38700 Grenoble, France
| | - Aurélie Varesano
- Institute for Advanced Biosciences (IAB), Team Host-Pathogen Interactions and Immunity to Infection, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, F-38700 Grenoble, France
| | - Bastien Touquet
- IAB, Team Membrane and Cell Dynamics of Host-Parasite Interactions, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, F-38700 Grenoble, France
| | - Pieter-Jan De Bock
- Institut de Biosciences et Biotechnologies de Grenoble-Laboratoire Biologie à Grande Échelle (BIG-BGE), Commissariat à l'énergie atomique et aux énergies alternatives (CEA), INSERM, 38000 Grenoble, France
| | - Yohann Coute
- Institut de Biosciences et Biotechnologies de Grenoble-Laboratoire Biologie à Grande Échelle (BIG-BGE), Commissariat à l'énergie atomique et aux énergies alternatives (CEA), INSERM, 38000 Grenoble, France
| | - Isabelle Tardieux
- IAB, Team Membrane and Cell Dynamics of Host-Parasite Interactions, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, F-38700 Grenoble, France
| | - Alexandre Bougdour
- Institute for Advanced Biosciences (IAB), Team Host-Pathogen Interactions and Immunity to Infection, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, F-38700 Grenoble, France
| | - Mohamed-Ali Hakimi
- Institute for Advanced Biosciences (IAB), Team Host-Pathogen Interactions and Immunity to Infection, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, F-38700 Grenoble, France
| |
Collapse
|
30
|
Epigenetics: A New Model for Intracellular Parasite–Host Cell Regulation. Trends Parasitol 2016; 32:515-521. [DOI: 10.1016/j.pt.2016.04.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 01/31/2016] [Accepted: 04/01/2016] [Indexed: 12/12/2022]
|
31
|
Functional Analysis of the Role of Toxoplasma gondii Nucleoside Triphosphate Hydrolases I and II in Acute Mouse Virulence and Immune Suppression. Infect Immun 2016; 84:1994-2001. [PMID: 27091930 DOI: 10.1128/iai.00077-16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/15/2016] [Indexed: 11/20/2022] Open
Abstract
Bioluminescent reporter assays have been widely used to study the effect of Toxoplasma gondii on host gene expression. In the present study, we extend these studies by engineering novel reporter cell lines containing a gamma-activated sequence (GAS) element driving firefly luciferase (FLUC). In RAW264.7 macrophages, T. gondii type I strain (GT1) infection blocked interferon gamma (IFN-γ)-induced FLUC activity to a significantly greater extent than infection by type II (ME49) and type III (CTG) strains. Quantitative trait locus (QTL) analysis of progeny from a prior genetic cross identified a genomic region on chromosome XII that correlated with the observed strain-dependent phenotype. This QTL region contains two isoforms of the T. gondii enzyme nucleoside triphosphate hydrolase (NTPase) that were the prime candidates for mediating the observed strain-specific effect. Using reverse genetic analysis we show that deletion of NTPase I from a type I strain (RH) background restored the higher luciferase levels seen in the type II (ME49) strain. Rather than an effect on IFN-γ-dependent transcription, our data suggest that NTPase I was responsible for the strain-dependent difference in FLUC activity due to hydrolysis of ATP. We further show that NTPases I and II were not essential for tachyzoite growth in vitro or virulence in mice. Our study reveals that although T. gondii NTPases are not essential for immune evasion, they can affect ATP-dependent reporters. Importantly, this limitation was overcome using an ATP-independent Gaussia luciferase, which provides a more appropriate reporter for use with T. gondii infection studies.
Collapse
|
32
|
Proteomic Profiling of Mouse Liver following Acute Toxoplasma gondii Infection. PLoS One 2016; 11:e0152022. [PMID: 27003162 PMCID: PMC4803215 DOI: 10.1371/journal.pone.0152022] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 03/08/2016] [Indexed: 02/05/2023] Open
Abstract
Toxoplasma gondii remains a global public health problem. However, its pathophysiology is still not-completely understood particularly the impact of infection on host liver metabolism. We performed iTRAQ-based proteomic analysis to evaluate early liver protein responses in BALB/c mice following infection with T. gondii PYS strain (genotype ToxoDB#9) infection. Our data revealed modification of protein expression in key metabolic pathways, as indicated by the upregulation of immune response and downregulation of mitochondrial respiratory chain, and the metabolism of fatty acids, lipids and xenobiotics. T. gondii seems to hijack host PPAR signaling pathway to downregulate the metabolism of fatty acids, lipids and energy in the liver. The metabolism of over 400 substances was affected by the downregulation of genes involved in xenobiotic metabolism. The top 10 transcription factors used by upregulated genes were Stat2, Stat1, Irf2, Irf1, Sp2, Egr1, Stat3, Klf4, Elf1 and Gabpa, while the top 10 transcription factors of downregulated genes were Hnf4A, Ewsr1, Fli1, Hnf4g, Nr2f1, Pparg, Rxra, Hnf1A, Foxa1 and Foxo1. These findings indicate global reprogramming of the metabolism of the mouse liver after acute T. gondii infection. Functional characterization of the altered proteins may enhance understanding of the host responses to T. gondii infection and lead to the identification of new therapeutic targets.
Collapse
|
33
|
Abstract
Our understanding of epigenetics in complex diseases is rapidly advancing and increasingly influencing the practice of medicine. Much is known about disruption of chromatin-modifying enzymes in malignant disease, but knowledge of irregular epigenetics in immune-driven disorders is just emerging. Epigenetic factors, such as DNA or histone modifications, are indispensable for precise gene expression in diverse immune cell types. Thus a disruption of epigenetic landscapes likely has a large impact on immune homeostasis. Moreover, the low concordance rates for most autoimmune diseases suggest that epigenetics contribute to immune tolerance disturbance. Here we review the important role of epigenetics for initiation, maintenance, tolerance, and training of immune responses. We discuss evolving evidence that DNA/histone modifications and chromatin-modifying enzymes are altered in immune-based diseases. Furthermore, we explore the potential of small molecules targeting epigenetic machinery, some of which are already used in oncology, as a way to reset the immune response in disease.
Collapse
|
34
|
Lüder CGK, Sumpf K, Nast R. Releasing the Brake on IFN-γ Signaling on Infection. Trends Parasitol 2015; 31:456-459. [PMID: 26422772 DOI: 10.1016/j.pt.2015.08.006] [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: 08/13/2015] [Accepted: 08/13/2015] [Indexed: 10/23/2022]
Abstract
Toxoplasma gondii effectively inhibits the responsiveness of its host cell to interferon gamma (IFN-γ). Using a genome-wide genetic screen, Beiting and colleagues have recently identified coactivators of the transcription factor STAT1 that can diminish this inhibitory effect. One of these coactivators, TLX, enhances type 1 helper (Th1) immune responses and restricts parasite replication during chronic toxoplasmosis.
Collapse
Affiliation(s)
- Carsten G K Lüder
- Institute for Medical Microbiology, Georg August University, Göttingen, Germany.
| | - Kristina Sumpf
- Institute for Medical Microbiology, Georg August University, Göttingen, Germany
| | - Roswitha Nast
- Institute for Medical Microbiology, Georg August University, Göttingen, Germany
| |
Collapse
|
35
|
STAT1 signaling within macrophages is required for antifungal activity against Cryptococcus neoformans. Infect Immun 2015; 83:4513-27. [PMID: 26351277 DOI: 10.1128/iai.00935-15] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 09/03/2015] [Indexed: 02/07/2023] Open
Abstract
Cryptococcus neoformans, the predominant etiological agent of cryptococcosis, is an opportunistic fungal pathogen that primarily affects AIDS patients and patients undergoing immunosuppressive therapy. In immunocompromised individuals, C. neoformans can lead to life-threatening meningoencephalitis. Studies using a virulent strain of C. neoformans engineered to produce gamma interferon (IFN-γ), denoted H99γ, demonstrated that protection against pulmonary C. neoformans infection is associated with the generation of a T helper 1 (Th1)-type immune response and signal transducer and activator of transcription 1 (STAT1)-mediated classical (M1) macrophage activation. However, the critical mechanism by which M1 macrophages mediate their anti-C. neoformans activity remains unknown. The current studies demonstrate that infection with C. neoformans strain H99γ in mice with macrophage-specific STAT1 ablation resulted in severely increased inflammation of the pulmonary tissue, a dysregulated Th1/Th2-type immune response, increased fungal burden, deficient M1 macrophage activation, and loss of protection. STAT1-deficient macrophages produced significantly less nitric oxide (NO) than STAT1-sufficient macrophages, correlating with an inability to control intracellular cryptococcal proliferation, even in the presence of reactive oxygen species (ROS). Furthermore, macrophages from inducible nitric oxide synthase knockout mice, which had intact ROS production, were deficient in anticryptococcal activity. These data indicate that STAT1 activation within macrophages is required for M1 macrophage activation and anti-C. neoformans activity via the production of NO.
Collapse
|
36
|
Abstract
Toxoplasmosis is the clinical and pathological consequence of acute infection with the obligate intracellular apicomplexan parasite Toxoplasma gondii. Symptoms result from tissue destruction that accompanies lytic parasite growth. This review updates current understanding of the host cell invasion, parasite replication, and eventual egress that constitute the lytic cycle, as well as the ways T. gondii manipulates host cells to ensure its survival. Since the publication of a previous iteration of this review 15 years ago, important advances have been made in our molecular understanding of parasite growth and mechanisms of host cell egress, and knowledge of the parasite's manipulation of the host has rapidly progressed. Here we cover molecular advances and current conceptual frameworks that include each of these topics, with an eye to what may be known 15 years from now.
Collapse
Affiliation(s)
- Ira J Blader
- Department of Microbiology and Immunology, University at Buffalo, Buffalo, New York 14127;
| | - Bradley I Coleman
- Department of Biology, Boston College, Chestnut Hill, Massachusetts 02467; , ,
| | - Chun-Ti Chen
- Department of Biology, Boston College, Chestnut Hill, Massachusetts 02467; , ,
| | - Marc-Jan Gubbels
- Department of Biology, Boston College, Chestnut Hill, Massachusetts 02467; , ,
| |
Collapse
|
37
|
Beiting DP, Hidano S, Baggs JE, Geskes JM, Fang Q, Wherry EJ, Hunter CA, Roos DS, Cherry S. The Orphan Nuclear Receptor TLX Is an Enhancer of STAT1-Mediated Transcription and Immunity to Toxoplasma gondii. PLoS Biol 2015. [PMID: 26196739 PMCID: PMC4509904 DOI: 10.1371/journal.pbio.1002200] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The protozoan parasite, Toxoplasma, like many intracellular pathogens, suppresses interferon gamma (IFN-γ)-induced signal transducer and activator of transcription 1 (STAT1) activity. We exploited this well-defined host–pathogen interaction as the basis for a high-throughput screen, identifying nine transcription factors that enhance STAT1 function in the nucleus, including the orphan nuclear hormone receptor TLX. Expression profiling revealed that upon IFN-γ treatment TLX enhances the output of a subset of IFN-γ target genes, which we found is dependent on TLX binding at those loci. Moreover, infection of TLX deficient mice with the intracellular parasite Toxoplasma results in impaired production of the STAT1-dependent cytokine interleukin-12 by dendritic cells and increased parasite burden in the brain during chronic infection. These results demonstrate a previously unrecognized role for this orphan nuclear hormone receptor in regulating STAT1 signaling and host defense and reveal that STAT1 activity can be modulated in a context-specific manner by such “modifiers.” Exploitation of the parasite Toxoplasma gondii identifies the host orphan nuclear hormone receptor TLX as a key enhancer of STAT1-dependent immune signaling and host defense. Immune responses are orchestrated by a diverse array of secreted ligands, yet the downstream transcriptional responses are coordinated by a relatively small set of key transcription factors, including nuclear factor kappa B (NF-κB) and signal transducers and activators of transcription (STATs). The molecular mechanisms that tailor the output of these immune signaling pathways to generate cell-, tissue-, or context-specific responses are poorly understood. In this study, we exploit a host–pathogen interaction, Toxoplasma gondii infection in mice, using a genetic screen to identify host factors that overcome parasite suppression of STAT1 signaling. We show that the orphan nuclear receptor TLX, a key regulator of brain development, enhances expression of a subset of STAT1-dependent genes in response to IFN-γ stimulation. Through genetic and pharmacological studies, we show that endogenous TLX function is required for triggering appropriate responses to IFN-γ in astrocytes. Moreover, we found that genetic disruption of TLX in mice impairs their ability to mount an effective immune response and control T. gondii infection in the brain. These data suggest that natural or synthetic ligands for TLX might be effective tools for modulating immune responses, particularly in the brain where TLX expression is highest.
Collapse
Affiliation(s)
- Daniel P. Beiting
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, Philadelphia, United States of America
| | - Shinya Hidano
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, Philadelphia, United States of America
| | - Julie E. Baggs
- Department of Pharmacology and the Institute for Translational Medicine and Therapeutics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jeanne M. Geskes
- Department of Pharmacology and the Institute for Translational Medicine and Therapeutics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Qun Fang
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, Philadelphia, United States of America
| | - E. John Wherry
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Christopher A. Hunter
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, Philadelphia, United States of America
| | - David S. Roos
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (DSR); (SC)
| | - Sara Cherry
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail: (DSR); (SC)
| |
Collapse
|
38
|
Parasite Manipulation of the Invariant Chain and the Peptide Editor H2-DM Affects Major Histocompatibility Complex Class II Antigen Presentation during Toxoplasma gondii Infection. Infect Immun 2015. [PMID: 26195549 DOI: 10.1128/iai.00415-15] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Toxoplasma gondii is an obligate intracellular protozoan parasite. This apicomplexan is the causative agent of toxoplasmosis, a leading cause of central nervous system disease in AIDS. It has long been known that T. gondii interferes with major histocompatibility complex class II (MHC-II) antigen presentation to attenuate CD4(+) T cell responses and establish persisting infections. Transcriptional downregulation of MHC-II genes by T. gondii was previously established, but the precise mechanisms inhibiting MHC-II function are currently unknown. Here, we show that, in addition to transcriptional regulation of MHC-II, the parasite modulates the expression of key components of the MHC-II antigen presentation pathway, namely, the MHC-II-associated invariant chain (Ii or CD74) and the peptide editor H2-DM, in professional antigen-presenting cells (pAPCs). Genetic deletion of CD74 restored the ability of infected dendritic cells to present a parasite antigen in the context of MHC-II in vitro. CD74 mRNA and protein levels were, surprisingly, elevated in infected cells, whereas MHC-II and H2-DM expression was inhibited. CD74 accumulated mainly in the endoplasmic reticulum (ER), and this phenotype required live parasites, but not active replication. Finally, we compared the impacts of genetic deletion of CD74 and H2-DM genes on parasite dissemination toward lymphoid organs in mice, as well as activation of CD4(+) T cells and interferon gamma (IFN-γ) levels during acute infection. Cyst burdens and survival during the chronic phase of infection were also evaluated in wild-type and knockout mice. These results highlight the fact that the infection is influenced by multiple levels of parasite manipulation of the MHC-II antigen presentation pathway.
Collapse
|
39
|
Bouchut A, Chawla AR, Jeffers V, Hudmon A, Sullivan WJ. Proteome-wide lysine acetylation in cortical astrocytes and alterations that occur during infection with brain parasite Toxoplasma gondii. PLoS One 2015; 10:e0117966. [PMID: 25786129 PMCID: PMC4364782 DOI: 10.1371/journal.pone.0117966] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 11/20/2014] [Indexed: 01/22/2023] Open
Abstract
Lysine acetylation is a reversible post-translational modification (PTM) that has been detected on thousands of proteins in nearly all cellular compartments. The role of this widespread PTM has yet to be fully elucidated, but can impact protein localization, interactions, activity, and stability. Here we present the first proteome-wide survey of lysine acetylation in cortical astrocytes, a subtype of glia that is a component of the blood-brain barrier and a key regulator of neuronal function and plasticity. We identified 529 lysine acetylation sites across 304 proteins found in multiple cellular compartments that largely function in RNA processing/transcription, metabolism, chromatin biology, and translation. Two hundred and seventy-seven of the acetylated lysines we identified on 186 proteins have not been reported previously in any other cell type. We also mapped an acetylome of astrocytes infected with the brain parasite, Toxoplasma gondii. It has been shown that infection with T. gondii modulates host cell gene expression, including several lysine acetyltransferase (KAT) and deacetylase (KDAC) genes, suggesting that the host acetylome may also be altered during infection. In the T. gondii-infected astrocytes, we identified 34 proteins exhibiting a level of acetylation >2-fold and 24 with a level of acetylation <2-fold relative to uninfected astrocytes. Our study documents the first acetylome map for cortical astrocytes, uncovers novel lysine acetylation sites, and demonstrates that T. gondii infection produces an altered acetylome.
Collapse
Affiliation(s)
- Anne Bouchut
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
| | - Aarti R. Chawla
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
| | - Victoria Jeffers
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
| | - Andy Hudmon
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
| | - William J. Sullivan
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
- Department of Microbiology & Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
- * E-mail:
| |
Collapse
|
40
|
Walwyn O, Skariah S, Lynch B, Kim N, Ueda Y, Vohora N, Choe J, Mordue DG. Forward genetics screens using macrophages to identify Toxoplasma gondii genes important for resistance to IFN-γ-dependent cell autonomous immunity. J Vis Exp 2015:52556. [PMID: 25867017 PMCID: PMC4401235 DOI: 10.3791/52556] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Toxoplasma gondii, the causative agent of toxoplasmosis, is an obligate intracellular protozoan pathogen. The parasite invades and replicates within virtually any warm blooded vertebrate cell type. During parasite invasion of a host cell, the parasite creates a parasitophorous vacuole (PV) that originates from the host cell membrane independent of phagocytosis within which the parasite replicates. While IFN-dependent-innate and cell mediated immunity is important for eventual control of infection, innate immune cells, including neutrophils, monocytes and dendritic cells, can also serve as vehicles for systemic dissemination of the parasite early in infection. An approach is described that utilizes the host innate immune response, in this case macrophages, in a forward genetic screen to identify parasite mutants with a fitness defect in infected macrophages following activation but normal invasion and replication in naïve macrophages. Thus, the screen isolates parasite mutants that have a specific defect in their ability to resist the effects of macrophage activation. The paper describes two broad phenotypes of mutant parasites following activation of infected macrophages: parasite stasis versus parasite degradation, often in amorphous vacuoles. The parasite mutants are then analyzed to identify the responsible parasite genes specifically important for resistance to induced mediators of cell autonomous immunity. The paper presents a general approach for the forward genetics screen that, in theory, can be modified to target parasite genes important for resistance to specific antimicrobial mediators. It also describes an approach to evaluate the specific macrophage antimicrobial mediators to which the parasite mutant is susceptible. Activation of infected macrophages can also promote parasite differentiation from the tachyzoite to bradyzoite stage that maintains chronic infection. Therefore, methodology is presented to evaluate the importance of the identified parasite gene to establishment of chronic infection.
Collapse
Affiliation(s)
- Odaelys Walwyn
- Department of Microbiology and Immunology, New York Medical College
| | - Sini Skariah
- Department of Microbiology and Immunology, New York Medical College
| | - Brian Lynch
- Department of Microbiology and Immunology, New York Medical College
| | - Nathaniel Kim
- Department of Microbiology and Immunology, New York Medical College
| | - Yukari Ueda
- Department of Microbiology and Immunology, New York Medical College
| | - Neal Vohora
- Department of Microbiology and Immunology, New York Medical College
| | - Josh Choe
- Department of Microbiology and Immunology, New York Medical College
| | - Dana G Mordue
- Department of Microbiology and Immunology, New York Medical College
| |
Collapse
|
41
|
Leroux LP, Dasanayake D, Rommereim LM, Fox BA, Bzik DJ, Jardim A, Dzierszinski FS. Secreted Toxoplasma gondii molecules interfere with expression of MHC-II in interferon gamma-activated macrophages. Int J Parasitol 2015; 45:319-32. [PMID: 25720921 DOI: 10.1016/j.ijpara.2015.01.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 01/16/2015] [Accepted: 01/18/2015] [Indexed: 11/15/2022]
Abstract
The obligate intracellular protozoan parasite Toxoplasma gondii interferes with major histocompatibility complex class II antigen presentation to dampen host CD4(+) T cell responses. While it is known that T. gondii inhibits major histocompatibility complex class II gene transcription and expression in infected host cells, the mechanism of this host manipulation is unknown. Here, we show that soluble parasite proteins inhibit IFNγ-induced expression of major histocompatibility complex class II on the surface of the infected cell in a dose-dependent response that was abolished by protease treatment. Subcellular fractionation of T. gondii tachyzoites revealed that the major histocompatibility complex class II inhibitory activity co-partitioned with rhoptries and/or dense granules. However, parasite mutants deleted for single rhoptries or dense granules genes (ROP1, 4/7, 14, 16 and 18 or GRA 2-9 and 12 knock-out strains) retained the ability to inhibit expression of major histocompatibility complex class II. In addition, excreted/secreted antigens released by extracellular tachyzoites displayed immunomodulatory activity characterized by an inhibition of major histocompatibility complex class II expression, and reduced expression and release of TNFα by macrophages. Tandem MS analysis of parasite excreted/secreted antigens generated a list of T. gondii secreted proteins that may participate in major histocompatibility complex class II inhibition and the modulation of host immune functions.
Collapse
Affiliation(s)
- Louis-Philippe Leroux
- Institute of Parasitology, McGill University, Parasitology Building, 21,111 Lakeshore Road, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; Centre for Host-Parasite Interaction, McGill University, Parasitology Building, 21,111 Lakeshore Road, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada
| | - Dayal Dasanayake
- Institute of Parasitology, McGill University, Parasitology Building, 21,111 Lakeshore Road, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; Centre for Host-Parasite Interaction, McGill University, Parasitology Building, 21,111 Lakeshore Road, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada
| | - Leah M Rommereim
- Geisel School of Medicine at Dartmouth, Borwell Research Building, 1 Medical Center Drive, Lebanon, NH 03756, USA
| | - Barbara A Fox
- Geisel School of Medicine at Dartmouth, Borwell Research Building, 1 Medical Center Drive, Lebanon, NH 03756, USA
| | - David J Bzik
- Geisel School of Medicine at Dartmouth, Borwell Research Building, 1 Medical Center Drive, Lebanon, NH 03756, USA
| | - Armando Jardim
- Institute of Parasitology, McGill University, Parasitology Building, 21,111 Lakeshore Road, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; Centre for Host-Parasite Interaction, McGill University, Parasitology Building, 21,111 Lakeshore Road, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada.
| | - Florence S Dzierszinski
- Institute of Parasitology, McGill University, Parasitology Building, 21,111 Lakeshore Road, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; Carleton University Research Office, Dunton Tower, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada
| |
Collapse
|
42
|
Beyond receptors and signaling: epigenetic factors in the regulation of innate immunity. Immunol Cell Biol 2015; 93:233-44. [PMID: 25559622 PMCID: PMC4885213 DOI: 10.1038/icb.2014.101] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 10/31/2014] [Indexed: 12/14/2022]
Abstract
The interaction of innate immune cells with pathogens leads to changes in gene expression that elicit our body's first line of defense against infection. Although signaling pathways and transcription factors have a central role, it is becoming increasingly clear that epigenetic factors, in the form of DNA or histone modifications, as well as noncoding RNAs, are critical for generating the necessary cell lineage as well as context‐specific gene expression in diverse innate immune cell types. Much of the epigenetic landscape is set during cellular differentiation; however, pathogens and other environmental triggers also induce changes in histone modifications that can either promote tolerance or ‘train’ innate immune cells for a more robust antigen‐independent secondary response. Here we review the important contribution of epigenetic factors to the initiation, maintenance and training of innate immune responses. In addition, we explore how pathogens have hijacked these mechanisms for their benefit and the potential of small molecules targeting chromatin machinery as a way to boost or subdue the innate immune response in disease. The March 2015 issue contains a Special Feature on the epigenetic mechanisms underlying health and disease. Epigenetic modifications to chromatin influence the transcriptional status of our genes. Thus, understanding the epigenetic mechanisms that regulate immune cell fate are of great importance as they will provide insight into not only how to boost immune responses but also alter harmful ones such as autoimmunity and cancer. Immunology and Cell Biology thanks the coordinators of this Special Feature ‐ Rhys Allan ‐ for his planning and input.
Collapse
|
43
|
Marr AK, MacIsaac JL, Jiang R, Airo AM, Kobor MS, McMaster WR. Leishmania donovani infection causes distinct epigenetic DNA methylation changes in host macrophages. PLoS Pathog 2014; 10:e1004419. [PMID: 25299267 PMCID: PMC4192605 DOI: 10.1371/journal.ppat.1004419] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 08/23/2014] [Indexed: 12/15/2022] Open
Abstract
Infection of macrophages by the intracellular protozoan Leishmania leads to down-regulation of a number of macrophage innate host defense mechanisms, thereby allowing parasite survival and replication. The underlying molecular mechanisms involved remain largely unknown. In this study, we assessed epigenetic changes in macrophage DNA methylation in response to infection with L. donovani as a possible mechanism for Leishmania driven deactivation of host defense. We quantified and detected genome-wide changes of cytosine methylation status in the macrophage genome resulting from L. donovani infection. A high confidence set of 443 CpG sites was identified with changes in methylation that correlated with live L. donovani infection. These epigenetic changes affected genes that play a critical role in host defense such as the JAK/STAT signaling pathway and the MAPK signaling pathway. These results provide strong support for a new paradigm in host-pathogen responses, where upon infection the pathogen induces epigenetic changes in the host cell genome resulting in downregulation of innate immunity thereby enabling pathogen survival and replication. We therefore propose a model whereby Leishmania induced epigenetic changes result in permanent down regulation of host defense mechanisms to protect intracellular replication and survival of parasitic cells. The L. donovani parasite causes visceral leishmaniasis, a tropical, neglected disease with an estimated number of 500,000 cases worldwide. Current drug treatments have toxic side effects, lead to drug resistance, and an effective vaccine is not available. The parasite has a complex life cycle residing within different host environments including the gut of a sand fly and immune cells of the mammalian host. Alteration of host cell gene expression including signaling pathways has been shown to be a major strategy to evade host cell immune response and thus enables the Leishmania parasite to survive, replicate and persist in its host cells. Recently it was demonstrated that intracellular pathogens such as viruses and bacteria are able to manipulate epigenetic processes, thereby perhaps facilitating their intracellular survival. Using an unbiased genome-wide DNA methylation approach, we demonstrate here that an intracellular parasite can alter host cell DNA methylation patterns resulting in altered gene expression possibly to establish disease. Thus DNA methylation changes in host cells upon infection might be a common strategy among intracellular pathogens for their uncontrolled replication and dissemination.
Collapse
Affiliation(s)
- Alexandra K. Marr
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Julia L. MacIsaac
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Ruiwei Jiang
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Adriana M. Airo
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Michael S. Kobor
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
- Human Early Learning Partnership, School of Population and Public Health, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
- * E-mail: (MSK); (WRM)
| | - W. Robert McMaster
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
- * E-mail: (MSK); (WRM)
| |
Collapse
|
44
|
Leopold Wager CM, Hole CR, Wozniak KL, Olszewski MA, Wormley FL. STAT1 signaling is essential for protection against Cryptococcus neoformans infection in mice. THE JOURNAL OF IMMUNOLOGY 2014; 193:4060-71. [PMID: 25200956 DOI: 10.4049/jimmunol.1400318] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nonprotective immune responses to highly virulent Cryptococcus neoformans strains, such as H99, are associated with Th2-type cytokine production, alternatively activated macrophages, and inability of the host to clear the fungus. In contrast, experimental studies show that protective immune responses against cryptococcosis are associated with Th1-type cytokine production and classical macrophage activation. The protective response induced during C. neoformans strain H99γ (C. neoformans strain H99 engineered to produce murine IFN-γ) infection correlates with enhanced phosphorylation of the transcription factor STAT1 in macrophages; however, the role of STAT1 in protective immunity to C. neoformans is unknown. The current studies examined the effect of STAT1 deletion in murine models of protective immunity to C. neoformans. Survival and fungal burden were evaluated in wild-type and STAT1 knockout (KO) mice infected with either strain H99γ or C. neoformans strain 52D (unmodified clinical isolate). Both strains H99γ and 52D were rapidly cleared from the lungs, did not disseminate to the CNS, or cause mortality in the wild-type mice. Conversely, STAT1 KO mice infected with H99γ or 52D had significantly increased pulmonary fungal burden, CNS dissemination, and 90-100% mortality. STAT1 deletion resulted in a shift from Th1 to Th2 cytokine bias, pronounced lung inflammation, and defective classical macrophage activation. Pulmonary macrophages from STAT1 KO mice exhibited defects in NO production correlating with inefficient inhibition of fungal proliferation. These studies demonstrate that STAT1 signaling is essential not only for regulation of immune polarization but also for the classical activation of macrophages that occurs during protective anticryptococcal immune responses.
Collapse
Affiliation(s)
- Chrissy M Leopold Wager
- Department of Biology, University of Texas, San Antonio, TX 78249; South Texas Center for Emerging Infectious Diseases, University of Texas, San Antonio, TX 78249
| | - Camaron R Hole
- Department of Biology, University of Texas, San Antonio, TX 78249; South Texas Center for Emerging Infectious Diseases, University of Texas, San Antonio, TX 78249
| | - Karen L Wozniak
- Department of Biology, University of Texas, San Antonio, TX 78249; South Texas Center for Emerging Infectious Diseases, University of Texas, San Antonio, TX 78249
| | - Michal A Olszewski
- Veterans Affairs Ann Arbor Health System, University of Michigan Health System, Ann Arbor, MI 48109; and Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109
| | - Floyd L Wormley
- Department of Biology, University of Texas, San Antonio, TX 78249; South Texas Center for Emerging Infectious Diseases, University of Texas, San Antonio, TX 78249;
| |
Collapse
|
45
|
Beiting DP. Protozoan parasites and type I interferons: a cold case reopened. Trends Parasitol 2014; 30:491-8. [PMID: 25153940 DOI: 10.1016/j.pt.2014.07.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 07/23/2014] [Accepted: 07/24/2014] [Indexed: 01/12/2023]
Abstract
Protozoan parasites, such as Plasmodium, Toxoplasma, Cryptosporidium, trypanosomes, and Leishmania, are a major cause of disease in both humans and other animals, highlighting the need to understand the full spectrum of strategies used by the host immune system to sense and respond to parasite infection. Although type II interferon (IFN-γ) has long been recognized as an essential antiparasite immune effector, much less is known about the role of type I interferons (IFN-α and -β) in host defense, particularly in vivo. Recent studies are reviewed which collectively highlight that type I IFN can be induced in response to parasite infection and influence the outcome of infection.
Collapse
Affiliation(s)
- Daniel P Beiting
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
46
|
Type II Toxoplasma gondii induction of CD40 on infected macrophages enhances interleukin-12 responses. Infect Immun 2014; 82:4047-55. [PMID: 25024369 DOI: 10.1128/iai.01615-14] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Toxoplasma gondii is an obligate intracellular parasite that can cause severe neurological disease in infected humans. CD40 is a receptor on macrophages that plays a critical role in controlling T. gondii infection. We examined the regulation of CD40 on the surface of T. gondii-infected bone marrow-derived macrophages (BMdMs). T. gondii induced CD40 expression both at the transcript level and on the cell surface, and interestingly, the effect was parasite strain specific: CD40 levels were dramatically increased in type II T. gondii-infected BMdMs compared to type I- or type III-infected cells. Type II induction of CD40 was specific to cells harboring intracellular parasites and detectable as early as 6 h postinfection (hpi) at the transcript level. CD40 protein expression peaked at 18 hpi. Using forward genetics with progeny from a type II × type III cross, we found that CD40 induction mapped to a region of chromosome X that included the gene encoding the dense granule protein 15 (GRA15). Using type I parasites stably expressing the type II allele of GRA15 (GRA15II), we found that type I GRA15II parasites induced the expression of CD40 on infected cells in an NF-κB-dependent manner. In addition, stable expression of hemagglutinin-tagged GRA15II in THP-1 cells resulted in CD40 upregulation in the absence of infection. Since CD40 signaling contributes to interleukin-12 (IL-12) production, we examined IL-12 from infected macrophages and found that CD40L engagement of CD40 amplified the IL-12 response in type II-infected cells. These data indicate that GRA15II induction of CD40 promotes parasite immunity through the production of IL-12.
Collapse
|
47
|
Toxoplasma gondii development of its replicative niche: in its host cell and beyond. EUKARYOTIC CELL 2014; 13:965-76. [PMID: 24951442 DOI: 10.1128/ec.00081-14] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Intracellular pathogens can replicate efficiently only after they manipulate and modify their host cells to create an environment conducive to replication. While diverse cellular pathways are targeted by different pathogens, metabolism, membrane and cytoskeletal architecture formation, and cell death are the three primary cellular processes that are modified by infections. Toxoplasma gondii is an obligate intracellular protozoan that infects ∼30% of the world's population and causes severe and life-threatening disease in developing fetuses, in immune-comprised patients, and in certain otherwise healthy individuals who are primarily found in South America. The high prevalence of Toxoplasma in humans is in large part a result of its ability to modulate these three host cell processes. Here, we highlight recent work defining the mechanisms by which Toxoplasma interacts with these processes. In addition, we hypothesize why some processes are modified not only in the infected host cell but also in neighboring uninfected cells.
Collapse
|
48
|
Jayachandran R, BoseDasgupta S, Pieters J. Surviving the macrophage: tools and tricks employed by Mycobacterium tuberculosis. Curr Top Microbiol Immunol 2014; 374:189-209. [PMID: 23154833 DOI: 10.1007/82_2012_273] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Mycobacterium tuberculosis has evolved to withstand one of the most inhospitable cells within the human body, namely the macrophage, a cell that is normally geared toward the destruction of any invading microbe. How M. tuberculosis achieves this is still incompletely understood; however, a number of mechanisms are now known that provide advantages to M. tuberculosis for its survival and proliferation inside the macrophage. While some of these mechanisms are mediated by factors released by M. tuberculosis, others rely on host components that are being hijacked to benefit survival of M. tuberculosis within the macrophage as well to avoid the generation of an effective immune response. Here, we describe several of these mechanisms, also pointing out the potential usage of this knowledge toward the development of novel strategies to treat tuberculosis. Furthermore, we attempt to put the 'macrophage niche' into context with other intracellular pathogens and discuss some of the generalities as well as specializations that M. tuberculosis employs to survive.
Collapse
Affiliation(s)
- Rajesh Jayachandran
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056, Basel, Switzerland
| | | | | |
Collapse
|
49
|
Toxoplasma gondii within skeletal muscle cells: a critical interplay for food-borne parasite transmission. Int J Parasitol 2014; 44:91-8. [DOI: 10.1016/j.ijpara.2013.10.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 09/12/2013] [Accepted: 10/03/2013] [Indexed: 01/30/2023]
|
50
|
New clinical and experimental insights into Old World and neotropical ocular toxoplasmosis. Int J Parasitol 2014; 44:99-107. [DOI: 10.1016/j.ijpara.2013.09.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 09/20/2013] [Accepted: 09/22/2013] [Indexed: 01/12/2023]
|