51
|
de Faria Junior GM, Murata FHA, Lorenzi HA, Castro BBP, Assoni LCP, Ayo CM, Brandão CC, de Mattos LC. The Role of microRNAs in the Infection by T. gondii in Humans. Front Cell Infect Microbiol 2021; 11:670548. [PMID: 34055667 PMCID: PMC8160463 DOI: 10.3389/fcimb.2021.670548] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 04/26/2021] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs are molecules belonging to an evolutionarily conserved family of small non-coding RNAs, which act on post-transcriptional gene regulation, causing messenger RNA (mRNA) degradation or inhibiting mRNA translation into proteins. These molecules represent potential biomarkers for diagnosis, non-invasive prognosis, and monitoring the development of the disease. Moreover, they may provide additional information on the pathophysiology of parasitic infections and guide strategies for treatment. The Apicomplexan parasite Toxoplasma gondii modifies the levels of microRNAs and mRNAs in infected host cells by modulating the innate and adaptive immune responses, facilitating its survival within the host. Some studies have shown that microRNAs are promising molecular markers for developing diagnostic tools for human toxoplasmosis. MicroRNAs can be detected in human specimens collected using non-invasive procedures. changes in the circulating host microRNAs have been associated with T. gondii infection in mice and ocular toxoplasmosis in humans. Besides, microRNAs can be amplified from samples using sensitive and molecular-specific approaches such as real-time PCR. This review presents recent findings of the role that microRNAs play during T. gondii infection and discuss their potential use of these small nuclei acid molecules to different approaches such as laboratory diagnosis, modulation of cell and tissue infected as other potential applications in human toxoplasmosis.
Collapse
Affiliation(s)
- Geraldo Magela de Faria Junior
- Immunogenetics Laboratory, Molecular Biology Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil
| | - Fernando Henrique Antunes Murata
- Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, United States Department of Agriculture, Agricultural Research Service, Beltsville, MD, United States
| | | | - Bruno Bello Pede Castro
- Department of Preventive Veterinary Medicine and Animal Health, Faculty of Veterinary Medicine, University of São Paulo, São Paulo, Brazil
| | - Letícia Carolina Paraboli Assoni
- Immunogenetics Laboratory, Molecular Biology Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil
| | - Christiane Maria Ayo
- Immunogenetics Laboratory, Molecular Biology Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil
| | - Cinara Cássia Brandão
- Immunogenetics Laboratory, Molecular Biology Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil
| | - Luiz Carlos de Mattos
- Immunogenetics Laboratory, Molecular Biology Department, Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil
| |
Collapse
|
52
|
Kochanowsky JA, Thomas KK, Koshy AA. ROP16-Mediated Activation of STAT6 Suppresses Host Cell Reactive Oxygen Species Production, Facilitating Type III Toxoplasma gondii Growth and Survival. mBio 2021; 12:e03305-20. [PMID: 33653884 PMCID: PMC8092286 DOI: 10.1128/mbio.03305-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/22/2021] [Indexed: 12/21/2022] Open
Abstract
Polymorphic effector proteins determine the susceptibility of Toxoplasma gondii strains to IFN-γ-mediated clearance mechanisms deployed by murine host cells. However, less is known about the influence of these polymorphic effector proteins on IFN-γ-independent clearance mechanisms. Here, we show that deletion of one such polymorphic effector protein, ROP16, from a type III background leads to a defect in parasite growth and survival in unstimulated human fibroblasts and murine macrophages. Rescue of these defects requires a ROP16 with a functional kinase domain and the ability to activate a specific family of host cell transcription factors (STAT3, 5a, and 6). The growth and survival defects correlate with an accumulation of host cell reactive oxygen species (ROS) and are prevented by treatment with an ROS inhibitor. Exogenous activation of STAT3 and 6 suppresses host cell ROS production during infection with ROP16-deficient parasites and depletion of STAT6, but not STAT3 or 5a, causes an accumulation of ROS in cells infected with wild-type parasites. Pharmacological inhibition of NOX2 and mitochondrially derived ROS also rescues growth and survival of ROP16-deficient parasites. Collectively, these findings reveal an IFN-γ-independent mechanism of parasite restriction in human cells that is subverted by injection of ROP16 by type III parasites.IMPORTANCEToxoplasma gondii is an obligate intracellular parasite that infects up to one-third of the world's population. Control of the parasite is largely accomplished by IFN-γ-dependent mechanisms that stimulate innate and adaptive immune responses. Parasite suppression of IFN-γ-stimulated responses has been linked to proteins that the parasite secretes into its host cell. These secreted proteins vary by T. gondii strain and determine strain-specific lethality in mice. How these strain-specific polymorphic effector proteins affect IFN-γ-independent parasite control mechanisms in human and murine cells is not well known. This study shows that one such secreted protein, ROP16, enables efficient parasite growth and survival by suppressing IFN-γ-independent production of ROS by human and mouse cells.
Collapse
Affiliation(s)
| | | | - Anita A Koshy
- Department of Immunobiology, University of Arizona, Tucson, Arizona, USA
- BIO5 Institute, University of Arizona, Tucson, Arizona, USA
- Department of Neurology, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
53
|
Li T, Cui L, Xu X, Zhang H, Jiang Y, Ren L, Yang C, Liu X, Hu X. The Role of Tim-3 on dNK Cells Dysfunction During Abnormal Pregnancy With Toxoplasma gondii Infection. Front Cell Infect Microbiol 2021; 11:587150. [PMID: 33718261 PMCID: PMC7953497 DOI: 10.3389/fcimb.2021.587150] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 01/15/2021] [Indexed: 11/18/2022] Open
Abstract
Vertical transmission of Toxoplasma gondii (T. gondii) infection during gestation can result in severe complications such as abortion, congenital malformation, fetal teratogenesis, etc. Immune inhibitory molecule Tim-3 was discovered to be expressed on some decidual immune cells and participates in the maintenance of maternal-fetal tolerance. Dysregulation of Tim-3 expression on decidual NK (dNK) cells was observed in several cases of pregnancy complications, whereas the role of Tim-3 on dNK cells during T. gondii infection remains unclear. In the present study, T. gondii infected Tim-3-/- pregnant mice, and anti-Tim-3 neutralizing antibody treated and infected human dNK cells were successfully established to explore the role of Tim-3 in dysfunction of dNK cells during abnormal pregnancy. Our results illustrated that Tim-3-/- pregnant mice displayed more worse pregnancy outcomes with T. gondii infection compared to infected WT pregnant mice. Also, it demonstrated that Tim-3 expression on dNK cells was significantly down-regulated following T. gondii infection. Data suggested a remarkable activation of dNK cells in Tim-3-/- mice and anti-Tim-3 neutralizing antibody treated and infected groups, with higher ratios of activating receptor NKG2D to inhibitory receptor NKG2A or KIR2DL4, IFN-γ/IL-10, and increased granule production compared with that of the infected group. Mechanism analysis proved that T. gondii-induced Tim-3 down-regulation significantly activated the phosphatidylinositol-3-kinase (PI3K)-AKT and JAK-STAT signaling pathway, by which the GranzymeB, Perforin, IFN-γ, and IL-10 production were further up-regulated. Our research demonstrated that the decrease of Tim-3 on dNK cells caused by T. gondii infection further led to dNK cells function disorder, which finally contributed to the development of abnormal pregnancy outcomes.
Collapse
Affiliation(s)
- Teng Li
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Lijun Cui
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Xiaoyan Xu
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Haixia Zhang
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Yuzhu Jiang
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Liqin Ren
- Medicine & Pharmacy Research Center, Binzhou Medical University, Yantai, China
| | - Chunyan Yang
- School of Stomatology, Binzhou Medical University, Yantai, China
| | - Xianbing Liu
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Xuemei Hu
- Department of Immunology, Binzhou Medical University, Yantai, China
| |
Collapse
|
54
|
Menard KL, Bu L, Denkers EY. Transcriptomics analysis of Toxoplasma gondii-infected mouse macrophages reveals coding and noncoding signatures in the presence and absence of MyD88. BMC Genomics 2021; 22:130. [PMID: 33622246 PMCID: PMC7903719 DOI: 10.1186/s12864-021-07437-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 02/11/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Toxoplasma gondii is a globally distributed protozoan parasite that establishes life-long asymptomatic infection in humans, often emerging as a life-threatening opportunistic pathogen during immunodeficiency. As an intracellular microbe, Toxoplasma establishes an intimate relationship with its host cell from the outset of infection. Macrophages are targets of infection and they are important in early innate immunity and possibly parasite dissemination throughout the host. Here, we employ an RNA-sequencing approach to identify host and parasite transcriptional responses during infection of mouse bone marrow-derived macrophages (BMDM). We incorporated into our analysis infection with the high virulence Type I RH strain and the low virulence Type II strain PTG. Because the well-known TLR-MyD88 signaling axis is likely of less importance in humans, we examined transcriptional responses in both MyD88+/+ and MyD88-/- BMDM. Long noncoding (lnc) RNA molecules are emerging as key regulators in infection and immunity, and were, therefore, included in our analysis. RESULTS We found significantly more host genes were differentially expressed in response to the highly virulent RH strain rather than with the less virulent PTG strain (335 versus 74 protein coding genes for RH and PTG, respectively). Enriched in these protein coding genes were subsets associated with the immune response as well as cell adhesion and migration. We identified 249 and 83 non-coding RNAs as differentially expressed during infection with RH and PTG strains, respectively. Although the majority of these are of unknown function, one conserved lncRNA termed mir17hg encodes the mir17 microRNA gene cluster that has been implicated in down-regulating host cell apoptosis during T. gondii infection. Only a minimal number of transcripts were differentially expressed between MyD88 knockout and wild type cells. However, several immune genes were among the differences. While transcripts for parasite secretory proteins were amongst the most highly expressed T. gondii genes during infection, no differentially expressed parasite genes were identified when comparing infection in MyD88 knockout and wild type host BMDM. CONCLUSIONS The large dataset presented here lays the groundwork for continued studies on both the MyD88-independent immune response and the function of lncRNAs during Toxoplasma gondii infection.
Collapse
Affiliation(s)
- Kayla L Menard
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, NM, USA.
| | - Lijing Bu
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, NM, USA
| | - Eric Y Denkers
- Center for Evolutionary and Theoretical Immunology and Department of Biology, University of New Mexico, Albuquerque, NM, USA.
| |
Collapse
|
55
|
Krishnan A, Soldati-Favre D. Amino Acid Metabolism in Apicomplexan Parasites. Metabolites 2021; 11:61. [PMID: 33498308 PMCID: PMC7909243 DOI: 10.3390/metabo11020061] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/05/2021] [Accepted: 01/14/2021] [Indexed: 12/22/2022] Open
Abstract
Obligate intracellular pathogens have coevolved with their host, leading to clever strategies to access nutrients, to combat the host's immune response, and to establish a safe niche for intracellular replication. The host, on the other hand, has also developed ways to restrict the replication of invaders by limiting access to nutrients required for pathogen survival. In this review, we describe the recent advancements in both computational methods and high-throughput -omics techniques that have been used to study and interrogate metabolic functions in the context of intracellular parasitism. Specifically, we cover the current knowledge on the presence of amino acid biosynthesis and uptake within the Apicomplexa phylum, focusing on human-infecting pathogens: Toxoplasma gondii and Plasmodium falciparum. Given the complex multi-host lifecycle of these pathogens, we hypothesize that amino acids are made, rather than acquired, depending on the host niche. We summarize the stage specificities of enzymes revealed through transcriptomics data, the relevance of amino acids for parasite pathogenesis in vivo, and the role of their transporters. Targeting one or more of these pathways may lead to a deeper understanding of the specific contributions of biosynthesis versus acquisition of amino acids and to design better intervention strategies against the apicomplexan parasites.
Collapse
Affiliation(s)
- Aarti Krishnan
- Department of Microbiology and Molecular Medicine, University of Geneva, CMU, Rue Michel-Servet 1, 1211 Geneva, Switzerland;
| | | |
Collapse
|
56
|
Metabolite salvage and restriction during infection - a tug of war between Toxoplasma gondii and its host. Curr Opin Biotechnol 2020; 68:104-114. [PMID: 33202353 DOI: 10.1016/j.copbio.2020.09.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 09/28/2020] [Indexed: 02/03/2023]
Abstract
The apicomplexans, including the coccidian pathogen Toxoplasma gondii, are obligate intracellular parasites whose growth and development are intricately linked to the metabolism of their host. T. gondii depends on its host for the salvage of energy sources, building blocks, vitamins and cofactors to survive and replicate. Additionally, host metabolites directly impact on the parasite life cycle development by triggering or halting differentiation. Although T. gondii infects a wide range of host cells, it has evolved to modulate and maximally exploit its host's metabolism. In return the host has developed strategies to restrict parasite access to metabolites. Here we discuss recent findings which have shed light on the battle over metabolites between T. gondii and its host.
Collapse
|
57
|
Chen L, Christian DA, Kochanowsky JA, Phan AT, Clark JT, Wang S, Berry C, Oh J, Chen X, Roos DS, Beiting DP, Koshy AA, Hunter CA. The Toxoplasma gondii virulence factor ROP16 acts in cis and trans, and suppresses T cell responses. J Exp Med 2020; 217:133618. [PMID: 31961916 PMCID: PMC7062521 DOI: 10.1084/jem.20181757] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/10/2019] [Accepted: 11/25/2019] [Indexed: 01/19/2023] Open
Abstract
The ability of Toxoplasma gondii to inject the rhoptry kinase ROP16 into host cells results in the activation of the transcription factors STAT3 and STAT6, but it is unclear how these events impact infection. Here, parasites that inject Cre-recombinase with rhoptry proteins were used to distinguish infected macrophages from those only injected with parasite proteins. Transcriptional profiling revealed that injection of rhoptry proteins alone was sufficient to induce an M2 phenotype that is dependent on STAT3 and STAT6, but only infected cells displayed reduced expression of genes associated with antimicrobial activity and protective immunity. In vivo, the absence of STAT3 or STAT6 improved parasite control, while the loss of ROP16 resulted in a marked reduction in parasite numbers and heightened parasite-specific T cell responses. Thus, ROP16 is a virulence factor that can act in cis and trans to promote M2 programs and which limits the magnitude of parasite-specific T cell responses.
Collapse
Affiliation(s)
- Longfei Chen
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China.,Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - David A Christian
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Joshua A Kochanowsky
- Department of Neurology and Department of Immunobiology, BIO5 Institute, University of Arizona, Tucson, AZ
| | - Anthony T Phan
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Joseph T Clark
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Shuai Wang
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Corbett Berry
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jung Oh
- Department of Biology, University of Pennsylvania, Philadelphia, PA
| | - Xiaoguang Chen
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - David S Roos
- Department of Biology, University of Pennsylvania, Philadelphia, PA
| | - Daniel P Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Anita A Koshy
- Department of Neurology and Department of Immunobiology, BIO5 Institute, University of Arizona, Tucson, AZ
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
58
|
Mukhopadhyay D, Arranz-Solís D, Saeij JPJ. Influence of the Host and Parasite Strain on the Immune Response During Toxoplasma Infection. Front Cell Infect Microbiol 2020; 10:580425. [PMID: 33178630 PMCID: PMC7593385 DOI: 10.3389/fcimb.2020.580425] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/11/2020] [Indexed: 01/02/2023] Open
Abstract
Toxoplasma gondii is an exceptionally successful parasite that infects a very broad host range, including humans, across the globe. The outcome of infection differs remarkably between hosts, ranging from acute death to sterile infection. These differential disease patterns are strongly influenced by both host- and parasite-specific genetic factors. In this review, we discuss how the clinical outcome of toxoplasmosis varies between hosts and the role of different immune genes and parasite virulence factors, with a special emphasis on Toxoplasma-induced ileitis and encephalitis.
Collapse
Affiliation(s)
| | | | - Jeroen P. J. Saeij
- Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
59
|
Nie LB, Liang QL, Elsheikha HM, Du R, Zhu XQ, Li FC. Global profiling of lysine 2-hydroxyisobutyrylome in Toxoplasma gondii using affinity purification mass spectrometry. Parasitol Res 2020; 119:4061-4071. [PMID: 33057814 DOI: 10.1007/s00436-020-06923-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/06/2020] [Indexed: 11/28/2022]
Abstract
Lysine 2-hydroxyisobutyrylation (Khib) is a recently discovered and evolutionarily conserved form of protein post-translational modification (PTM) found in mammalian and yeast cells. Previous studies have shown that Khib plays roles in the activity of gene transcription and Khib-containing proteins are closely related to the cellular metabolism. In this study, a global Khib-containing analysis using the latest databases (ToxoDB 46, 8322 sequences, downloaded on April 16, 2020) and sensitive immune-affinity enrichment coupled with liquid chromatography-tandem mass spectrometry was performed. A total of 1078 Khib modification sites across 400 Khib-containing proteins were identified in tachyzoites of Toxoplasma gondii RH strain. Bioinformatics and functional enrichment analysis showed that Khib-modified proteins were associated with various biological processes, such as ribosome, glycolysis/gluconeogenesis, and central carbon metabolism. Interestingly, many proteins of the secretory organelles (e.g., microneme, rhoptry, and dense granule) that play roles in the infection cycle of T. gondii were found to be Khib-modified, suggesting the involvement of Khib in key biological process during T. gondii infection. We also found that histone proteins, key enzymes related to cellular metabolism, and several glideosome components had Khib sites. These results expanded our understanding of the roles of Khib in T. gondii and should promote further investigations of how Khib regulates gene expression and key biological functions in T. gondii.
Collapse
Affiliation(s)
- Lan-Bi Nie
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, Jilin Province, People's Republic of China.,State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu Province, People's Republic of China
| | - Qin-Li Liang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu Province, People's Republic of China
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough, LE12 5RD, UK
| | - Rui Du
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, Jilin Province, People's Republic of China
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu Province, People's Republic of China. .,College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi Province, People's Republic of China.
| | - Fa-Cai Li
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu Province, People's Republic of China.,College of Veterinary Medicine, Southwest University, Chongqing, 400715, People's Republic of China
| |
Collapse
|
60
|
Kongsomboonvech AK, Rodriguez F, Diep AL, Justice BM, Castallanos BE, Camejo A, Mukhopadhyay D, Taylor GA, Yamamoto M, Saeij JPJ, Reese ML, Jensen KDC. Naïve CD8 T cell IFNγ responses to a vacuolar antigen are regulated by an inflammasome-independent NLRP3 pathway and Toxoplasma gondii ROP5. PLoS Pathog 2020; 16:e1008327. [PMID: 32853276 PMCID: PMC7480859 DOI: 10.1371/journal.ppat.1008327] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 09/09/2020] [Accepted: 07/05/2020] [Indexed: 12/31/2022] Open
Abstract
Host resistance to Toxoplasma gondii relies on CD8 T cell IFNγ responses, which if modulated by the host or parasite could influence chronic infection and parasite transmission between hosts. Since host-parasite interactions that govern this response are not fully elucidated, we investigated requirements for eliciting naïve CD8 T cell IFNγ responses to a vacuolar resident antigen of T. gondii, TGD057. Naïve TGD057 antigen-specific CD8 T cells (T57) were isolated from transnuclear mice and responded to parasite-infected bone marrow-derived macrophages (BMDMs) in an antigen-dependent manner, first by producing IL-2 and then IFNγ. T57 IFNγ responses to TGD057 were independent of the parasite’s protein export machinery ASP5 and MYR1. Instead, host immunity pathways downstream of the regulatory Immunity-Related GTPases (IRG), including partial dependence on Guanylate-Binding Proteins, are required. Multiple T. gondii ROP5 isoforms and allele types, including ‘avirulent’ ROP5A from clade A and D parasite strains, were able to suppress CD8 T cell IFNγ responses to parasite-infected BMDMs. Phenotypic variance between clades B, C, D, F, and A strains suggest T57 IFNγ differentiation occurs independently of parasite virulence or any known IRG-ROP5 interaction. Consistent with this, removal of ROP5 is not enough to elicit maximal CD8 T cell IFNγ production to parasite-infected cells. Instead, macrophage expression of the pathogen sensors, NLRP3 and to a large extent NLRP1, were absolute requirements. Other members of the conventional inflammasome cascade are only partially required, as revealed by decreased but not abrogated T57 IFNγ responses to parasite-infected ASC, caspase-1/11, and gasdermin D deficient cells. Moreover, IFNγ production was only partially reduced in the absence of IL-12, IL-18 or IL-1R signaling. In summary, T. gondii effectors and host machinery that modulate parasitophorous vacuolar membranes, as well as NLR-dependent but inflammasome-independent pathways, determine the full commitment of CD8 T cells IFNγ responses to a vacuolar antigen. Parasites are excellent “students” of our immune system as they can deflect, antagonize and confuse the immune response making it difficult to vaccinate against these pathogens. In this report, we analyzed how a widespread parasite of mammals, Toxoplasma gondii, manipulates an immune cell needed for immunity to many intracellular pathogens, the CD8 T cell. Host pathways that govern CD8 T cell production of the immune protective cytokine, IFNγ, were also explored. We hypothesized the secreted T. gondii virulence factor, ROP5, work to inhibit the MHC 1 antigen presentation pathway therefore making it difficult for CD8 T cells to see T. gondii antigens sequestered inside a parasitophorous vacuole. However, manipulation through T. gondii ROP5 does not fully explain how CD8 T cells commit to making IFNγ in response to infection. Importantly, CD8 T cell IFNγ responses to T. gondii require the pathogen sensor NLRP3 to be expressed in the infected cell. Other proteins associated with NLRP3 activation, including members of the conventional inflammasome activation cascade pathway, are only partially involved. Our results identify a novel pathway by which NLRP3 regulates T cell function and underscore the need for NLRP3-activating adjuvants in vaccines aimed at inducing CD8 T cell IFNγ responses to parasites.
Collapse
Affiliation(s)
- Angel K. Kongsomboonvech
- Department of Molecular and Cell Biology, University of California, Merced, Merced, California, United States of America
| | - Felipe Rodriguez
- Department of Molecular and Cell Biology, University of California, Merced, Merced, California, United States of America
| | - Anh L. Diep
- Department of Molecular and Cell Biology, University of California, Merced, Merced, California, United States of America
| | - Brandon M. Justice
- Department of Molecular and Cell Biology, University of California, Merced, Merced, California, United States of America
| | - Brayan E. Castallanos
- Department of Molecular and Cell Biology, University of California, Merced, Merced, California, United States of America
| | - Ana Camejo
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Debanjan Mukhopadhyay
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, California, United States of America
| | - Gregory A. Taylor
- Departments of Medicine; Molecular Genetics and Microbiology; and Immunology; and Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, North Carolina, United States of America
- Geriatric Research, Education, and Clinical Center, Durham VA Health Care System, Durham, North Carolina, United States of America
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Jeroen P. J. Saeij
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, California, United States of America
| | - Michael L. Reese
- Department of Pharmacology, University of Texas, Southwestern Medical Center, Dallas, Texas, United States of America
| | - Kirk D. C. Jensen
- Department of Molecular and Cell Biology, University of California, Merced, Merced, California, United States of America
- Health Sciences Research Institute, University of California, Merced, Merced, California, United States of America
- * E-mail:
| |
Collapse
|
61
|
Lee SH, Jung BK, Song H, Seo HG, Chai JY, Oh BM. Neuroprotective Effect of Chronic Intracranial Toxoplasma gondii Infection in a Mouse Cerebral Ischemia Model. THE KOREAN JOURNAL OF PARASITOLOGY 2020; 58:461-466. [PMID: 32871641 PMCID: PMC7462801 DOI: 10.3347/kjp.2020.58.4.461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 06/30/2020] [Indexed: 11/23/2022]
Abstract
Toxoplasma gondii is an obligate intracellular protozoan parasite that can invade various organs in the host body, including the central nervous system. Chronic intracranial T. gondii is known to be associated with neuroprotection against neurodegenerative diseases through interaction with host brain cells in various ways. The present study investigated the neuroprotective effects of chronic T. gondii infection in mice with cerebral ischemia experimentally produced by middle cerebral artery occlusion (MCAO) surgery. The neurobehavioral effects of cerebral ischemia were assessed by measurement of Garcia score and Rotarod behavior tests. The volume of brain ischemia was measured by triphenyltetrazolium chloride staining. The expression levels of related genes and proteins were determined. After cerebral ischemia, corrected infarction volume was significantly reduced in T. gondii infected mice, and their neurobehavioral function was significantly better than that of the uninfection control group. Chronic T. gondii infection induced the expression of hypoxia-inducible factor 1-alpha (HIF-1α) in the brain before MCAO. T. gondii infection also increased the expression of vascular endothelial growth factor after the cerebral ischemia. It is suggested that chronic intracerebral infection of T. gondii may be a potential preconditioning strategy to reduce neural deficits associated with cerebral ischemia and induce brain ischemic tolerance through the regulation of HIF-1α expression.
Collapse
Affiliation(s)
- Seung Hak Lee
- Department of Rehabilitation Medicine, Asan Medical Center, College of Medicine, University of Ulsan, Seoul 05505, Korea
| | - Bong-Kwang Jung
- Institute of Parasitic Diseases, Korea Association of Health Promotion, Seoul 07653, Korea
| | - Hyemi Song
- Institute of Parasitic Diseases, Korea Association of Health Promotion, Seoul 07653, Korea
| | - Han Gil Seo
- Department of Rehabilitation Medicine, Seoul National University Hospital, 03080 Seoul, Korea
| | - Jong-Yil Chai
- Institute of Parasitic Diseases, Korea Association of Health Promotion, Seoul 07653, Korea.,Department of Tropical Medicine and Parasitology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Byung-Mo Oh
- Department of Rehabilitation Medicine, Seoul National University Hospital, 03080 Seoul, Korea
| |
Collapse
|
62
|
Abo-Al-Ela HG. Toxoplasmosis and Psychiatric and Neurological Disorders: A Step toward Understanding Parasite Pathogenesis. ACS Chem Neurosci 2020; 11:2393-2406. [PMID: 31268676 DOI: 10.1021/acschemneuro.9b00245] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Toxoplasmosis, a disease that disrupts fetal brain development and severely affects the host's brain, has been linked to many behavioral and neurological disorders. There is growing interest in how a single-celled neurotropic parasite, Toxoplasma gondii, can control or change the behavior of the host as well as how it dominates the host's neurons. Secrets beyond these could be answered by decoding the Toxoplasma gondii genome, unravelling the function of genomic sequences, and exploring epigenetics and mRNAs alterations, as well as the postulated mechanisms contributing to various neurological and psychiatric symptoms caused by this parasite. Substantial efforts have been made to elucidate the action of T. gondii on host immunity and the biology of its infection. However, the available studies on the molecular aspects of toxoplasmosis that affect central nervous system (CNS) circuits remain limited, and much research is still needed on this interesting topic. In my opinion, this parasite is a gift for studying the biology of the nervous system and related diseases. We should utilize the unique features of Toxoplasma, such as its abilities to modulate brain physiology, for neurological studies or as a possible tool or approach to cure neurological disease.
Collapse
Affiliation(s)
- Haitham G. Abo-Al-Ela
- Animal Health Research Institute, Agriculture Research Center, Shibin Al-Kom, El-Minufiya 7001, Egypt
| |
Collapse
|
63
|
Zhao XY, Ewald SE. The molecular biology and immune control of chronic Toxoplasma gondii infection. J Clin Invest 2020; 130:3370-3380. [PMID: 32609097 PMCID: PMC7324197 DOI: 10.1172/jci136226] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Toxoplasma gondii is an incredibly successful parasite owing in part to its ability to persist within cells for the life of the host. Remarkably, at least 350 host species of T. gondii have been described to date, and it is estimated that 30% of the global human population is chronically infected. The importance of T. gondii in human health was made clear with the first reports of congenital toxoplasmosis in the 1940s. However, the AIDS crisis in the 1980s revealed the prevalence of chronic infection, as patients presented with reactivated chronic toxoplasmosis, underscoring the importance of an intact immune system for parasite control. In the last 40 years, there has been tremendous progress toward understanding the biology of T. gondii infection using rodent models, human cell experimental systems, and clinical data. However, there are still major holes in our understanding of T. gondii biology, including the genes controlling parasite development, the mechanisms of cell-intrinsic immunity to T. gondii in the brain and muscle, and the long-term effects of infection on host homeostasis. The need to better understand the biology of chronic infection is underscored by the recent rise in ocular disease associated with emerging haplotypes of T. gondii and our lack of effective treatments to sterilize chronic infection. This Review discusses the cell types and molecular mediators, both host and parasite, that facilitate persistent T. gondii infection. We highlight the consequences of chronic infection for tissue-specific pathology and identify open questions in this area of host-Toxoplasma interactions.
Collapse
|
64
|
Mukhopadhyay D, Arranz-Solís D, Saeij JPJ. Toxoplasma GRA15 and GRA24 are important activators of the host innate immune response in the absence of TLR11. PLoS Pathog 2020; 16:e1008586. [PMID: 32453782 PMCID: PMC7274473 DOI: 10.1371/journal.ppat.1008586] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/05/2020] [Accepted: 04/30/2020] [Indexed: 12/13/2022] Open
Abstract
The murine innate immune response against Toxoplasma gondii is predominated by the interaction of TLR11/12 with Toxoplasma profilin. However, mice lacking Tlr11 or humans, who do not have functional TLR11 or TLR12, still elicit a strong innate immune response upon Toxoplasma infection. The parasite factors that determine this immune response are largely unknown. Herein, we investigated two dense granule proteins (GRAs) secreted by Toxoplasma, GRA15 and GRA24, for their role in stimulating the innate immune response in Tlr11-/- mice and in human cells, which naturally lack TLR11/TLR12. Our results show that GRA15 and GRA24 synergistically shape the early immune response and parasite virulence in Tlr11-/- mice, with GRA15 as the predominant effector. Nevertheless, acute virulence in Tlr11-/- mice is still dominated by allelic combinations of ROP18 and ROP5, which are effectors that determine evasion of the immunity-related GTPases. In human macrophages, GRA15 and GRA24 play a major role in the induction of IL12, IL18 and IL1β secretion. We further show that GRA15/GRA24-mediated IL12, IL18 and IL1β secretion activates IFNγ secretion by peripheral blood mononuclear cells (PBMCs), which controls Toxoplasma proliferation. Taken together, our study demonstrates the important role of GRA15 and GRA24 in activating the innate immune response in hosts lacking TLR11.
Collapse
Affiliation(s)
- Debanjan Mukhopadhyay
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - David Arranz-Solís
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Jeroen P. J. Saeij
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
65
|
Toxoplasma gondii dense granule protein GRA24 drives MyD88-independent p38 MAPK activation, IL-12 production and induction of protective immunity. PLoS Pathog 2020; 16:e1008572. [PMID: 32413093 PMCID: PMC7255617 DOI: 10.1371/journal.ppat.1008572] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/28/2020] [Accepted: 04/23/2020] [Indexed: 11/19/2022] Open
Abstract
The apicomplexan Toxoplasma gondii induces strong protective immunity dependent upon recognition by Toll-like receptors (TLR)11 and 12 operating in conjunction with MyD88 in the murine host. However, TLR11 and 12 proteins are not present in humans, inspiring us to investigate MyD88-independent pathways of resistance. Using bicistronic IL-12-YFP reporter mice on MyD88+/+ and MyD88-/- genetic backgrounds, we show that CD11c+MHCII+F4/80- dendritic cells, F4/80+ macrophages, and Ly6G+ neutrophils were the dominant cellular sources of IL-12 in both wild type and MyD88 deficient mice after parasite challenge. Parasite dense granule protein GRA24 induces p38 MAPK activation and subsequent IL-12 production in host macrophages. We show that Toxoplasma triggers an early and late p38 MAPK phosphorylation response in MyD88+/+ and MyD88-/- bone marrow-derived macrophages. Using the uracil auxotrophic Type I T. gondii strain cps1-1, we demonstrate that the late response does not require active parasite proliferation, but strictly depends upon GRA24. By i. p. inoculation with cps1-1 and cps1-1:Δgra24, we identified unique subsets of chemokines and cytokines that were up and downregulated by GRA24. Finally, we demonstrate that cps1-1 triggers a strong host-protective GRA24-dependent Th1 response in the absence of MyD88. Our data identify GRA24 as a major mediator of p38 MAPK activation, IL-12 induction and protective immunity that operates independently of the TLR/MyD88 cascade. Toxoplasma gondii is a protozoan parasite that infects over 1 billion people worldwide. Infection with the parasite is normally asymptomatic and Toxoplasma co-exists with its host in the form of latent cysts in brain and muscle tissue. The balance between immune recognition and immune evasion is likely a key factor in the outcome of this host-parasite interaction. It is therefore important to understand how Toxoplasma triggers immunity, and in particular how the protective cytokine IL-12 is induced during infection. While Toll-like receptor (TLR)/MyD88 signaling is important in mouse resistance to Toxoplasma, this pathway is likely less important in human infection. Here, we report that the parasite dense granule protein GRA24 triggers p38 MAPK activation and IL-12 production independently of TLR/MyD88 signaling. We identify additional cytokines and chemokines that are regulated by GRA24 during in vivo infection. Our data demonstrate that GRA24 initiates a protective MyD88-independent immune response during in vivo infection. The GRA24 molecule provides an example of a parasite molecule whose function is induction of a host protective immune response. From the standpoint of Toxoplasma, this likely reflects an evolutionary adaptation to ensure host survival and simultaneously enable latency to maximize the chance of transmission.
Collapse
|
66
|
Prior Toxoplasma Gondii Infection Ameliorates Liver Fibrosis Induced by Schistosoma Japonicum through Inhibiting Th2 Response and Improving Balance of Intestinal Flora in Mice. Int J Mol Sci 2020; 21:ijms21082711. [PMID: 32295161 PMCID: PMC7216211 DOI: 10.3390/ijms21082711] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/01/2020] [Accepted: 04/09/2020] [Indexed: 02/06/2023] Open
Abstract
Schistosomiasis is an immunopathogenic disease in which a T helper (Th) cell type 2-like response plays vital roles. Hepatic fibrosis is its main pathologic manifestations, which is the leading cause of hepatic cirrhosis. Co-infections of Schistosoma japonicum (Sj) with other pathogens are frequently encountered but are easily ignored in clinical studies, and effective therapeutic interventions are lacking. In this study, we explored the effect of Toxoplasma gondii (Tg) prior infection on Th1/Th2 response, community shifts in gut microbiome (GM), and the pathogenesis of schistosomiasis in murine hosts. Mice were prior infected with Tg before Sj infection. The effects of co-infection on Th1/Th2 response and hepatic fibrosis were analyzed. Furthermore, we investigated this issue by sequencing 16S rRNA from fecal specimens to define the GM profiles during co-infection. Tg prior infection markedly reduced the granuloma size and collagen deposit in livers against Sj infection. Prior infection promoted a shift toward Th1 immune response instead of Th2. Furthermore, Tg infection promoted the expansion of preponderant flora and Clostridiaceae was identified as a feature marker in the GM of the co-infection group. Redundancy analysis (RDA)/canonical correspondence analysis (CCA) results showed that liver fibrosis, Th1/Th2 cytokines were significantly correlated (P < 0.05) with the GM compositions. Tg infection inhibits hepatic fibrosis by downregulating Th2 immune response against Sj infection, and further promotes the GM shifts through "gut-liver axis" in the murine hosts. Our study may provide insights into potential anti-fibrosis strategies in co-infection individuals.
Collapse
|
67
|
Park J, Hunter CA. The role of macrophages in protective and pathological responses to Toxoplasma gondii. Parasite Immunol 2020; 42:e12712. [PMID: 32187690 DOI: 10.1111/pim.12712] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/12/2020] [Accepted: 02/24/2020] [Indexed: 02/06/2023]
Abstract
The ability of Toxoplasma gondii to cause clinical disease in immune-competent and immune-deficient hosts coupled with its ease of use in vitro and availability of murine models has led to its use as a model organism to study how the immune system controls an intracellular infection. This article reviews the studies that established the role of the cytokine IFN-γ in the activation of macrophages to control T gondii and the events that lead to the mobilization and expansion of macrophage populations and their ability to limit parasite replication. Macrophages also have pro-inflammatory functions that promote protective NK and T-cell activities as well as regulatory properties that facilitate the resolution of inflammation. Nevertheless, while macrophages are important in determining the outcome of infection, T gondii has evolved mechanisms to subvert macrophage activation and can utilize their migratory activities to promote dissemination and these two properties underlie the ability of this parasite to persist and cause disease.
Collapse
Affiliation(s)
- Jeongho Park
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA.,Kangwon National University College of Veterinary Medicine and Institute of Veterinary Science, Chuncheon, Korea
| | | |
Collapse
|
68
|
Sasai M, Yamamoto M. Decision by injection without infection. J Exp Med 2020; 217:e20192145. [PMID: 31967639 PMCID: PMC7062535 DOI: 10.1084/jem.20192145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The protozoan pathogen Toxoplasma gondii co-opts host immunity by secreting various effector proteins into host cells. In this issue of JEM, Chen et al. (https://doi.org/10.1084/jem.20181757) report that T. gondii injects an effector called ROP16 without invasion to polarize macrophages toward M2 cells.
Collapse
Affiliation(s)
- Miwa Sasai
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| |
Collapse
|
69
|
Cui W, Wang C, Luo Q, Xing T, Shen J, Wang W. Toxoplasma gondii ROP16 I Deletion: The Exacerbated Impact on Adverse Pregnant Outcomes in Mice. Front Microbiol 2020; 10:3151. [PMID: 32082272 PMCID: PMC7005636 DOI: 10.3389/fmicb.2019.03151] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/29/2019] [Indexed: 01/18/2023] Open
Abstract
Imbalance of Th1 and Th2 response at the maternal-fetal interface is considered as a radical event in the pathogenesis of immunity-related pregnant diseases. It has been demonstrated that the ROP16I, a rhoptry protein of Toxoplasma gondii, and the viable parasite with ROP16I may induce M2 macrophage polarization in host innate immunity and may be involved in the adverse pregnant outcomes. However, the mechanisms by which T. gondii-derived effectors subvert the immune tolerance in the pathology of pregnancy remain unclear. Here, we constructed the RH strain with ROP16I deletion (RHΔrop16) to explore the pathogenesis of abnormal pregnancy. We found that C57BL/6 mice infected with RHΔrop16 exhibited the increased resorption of fetuses and more severe adverse pathology of placentae at the early phase of gestation, as compared to the mice infected with RH wild type (RH WT) parasite. Additionally, RHΔrop16 strain infection significantly promoted M1 macrophage phenotypes of CD80 and CD86, and decreased CD206 expression of M2 macrophages, with upregulation of the iNOS and downregulation of the Arg-1 expression in placental homogenates. Simultaneously, the pro-inflammatory cytokines of IL-12 and TNF-α were elevated whereas the anti-inflammatory cytokine of TGF-β1 was dampened. Moreover, the p38α mitogen-activated protein kinase (p38α MAPK) was notably phosphorylated in placental macrophages infected with both RHΔrop16 and RH WT strains compared with the control. Taken together, our findings indicated that ROP16I deletion of type I RH strain may cause exacerbated adverse pregnant outcomes, which is attributable to subversion of the maternal immune tolerance due to the increased pro-inflammatory cytokines in the pregnant animals. The results also suggest that ROP16I might be a protective factor and other T. gondii-derived molecules might be involved in the M1-Th1 biased pathological process in aberrant pregnancy at the early phase of gestation.
Collapse
Affiliation(s)
- Wen Cui
- Department of Pathogen Biology, Provincial Laboratories of Pathogen Biology and Zoonoses Anhui, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Cong Wang
- Department of Clinical Laboratory, The Second Hospital of Hefei, Hefei, China
| | - Qingli Luo
- Department of Pathogen Biology, Provincial Laboratories of Pathogen Biology and Zoonoses Anhui, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Tian Xing
- The Key Laboratory of Oral Disease Research of Anhui, College and Hospital of Stomatology, Anhui Medical University, Hefei, China
| | - Jilong Shen
- Department of Pathogen Biology, Provincial Laboratories of Pathogen Biology and Zoonoses Anhui, School of Basic Medicine, Anhui Medical University, Hefei, China.,Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei Wang
- Department of Pathogen Biology, Provincial Laboratories of Pathogen Biology and Zoonoses Anhui, School of Basic Medicine, Anhui Medical University, Hefei, China
| |
Collapse
|
70
|
Abstract
The obligate intracellular protozoan parasite Toxoplasma gondii can infect any nucleated cell from a warm-blooded host. However, its interaction with host macrophages plays a critical role in shaping the immune response during infection. Therefore, assessing Toxoplasma-macrophage interactions at a cellular level is important. In this chapter, we describe assays that can be used to characterize Toxoplasma-macrophage interactions. These assays can also be used to evaluate other host-pathogen interactions. We describe multiplex approaches for measuring arginase activity, indoleamine 2,3 dioxygenase activity, cell death, and parasite growth during Toxoplasma-macrophage interactions. These assays can be used to compare how different Toxoplasma strains differ in their interaction with macrophages, and we describe how to properly assess Toxoplasma strain differences in Toxoplasma-macrophage interactions.
Collapse
Affiliation(s)
- Debanjan Mukhopadhyay
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, USA
| | - Jeroen P J Saeij
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
71
|
Hernández-de-Los-Ríos A, Murillo-Leon M, Mantilla-Muriel LE, Arenas AF, Vargas-Montes M, Cardona N, de-la-Torre A, Sepúlveda-Arias JC, Gómez-Marín JE. Influence of Two Major Toxoplasma Gondii Virulence Factors (ROP16 and ROP18) on the Immune Response of Peripheral Blood Mononuclear Cells to Human Toxoplasmosis Infection. Front Cell Infect Microbiol 2019; 9:413. [PMID: 31867288 PMCID: PMC6904310 DOI: 10.3389/fcimb.2019.00413] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/20/2019] [Indexed: 12/16/2022] Open
Abstract
Toxoplasma gondii ROP16 and ROP18 proteins have been identified as important virulence factors for this parasite. Here, we describe the effect of ROP16 and ROP18 proteins on peripheral blood mononuclear cells (PBMCs) from individuals with different clinical status of infection. We evaluated IFN-γ, IL-10, and IL-1β levels in supernatants from PBMCs cultures infected with tachyzoites of the T. gondii wild-type RH strain or with knock-out mutants of the rop16 and rop18 encoding genes (RHΔrop16 and RHΔrop18). Cytokine secretion was compared between PBMCs obtained from seronegative individuals (n = 10), with those with chronic asymptomatic (n = 8), or ocular infection (n = 12). We also evaluated if polymorphisms in the genes encoding for IFN-γ, IL-10, IL-1β, Toll-like receptor 9 (TLR9), and purinoreceptor P2RX7 influenced the production of the encoded proteins after ex vivo stimulation. In individuals with chronic asymptomatic infection, only a moderate effect on IL-10 levels was observed when PBMCs were infected with RHΔrop16, whereas a significant difference in the levels of inflammatory cytokines IFN-γ and IL-1β was observed in seronegative individuals, but this was also dependent on the host's cytokine gene polymorphisms. Infection with ROP16-deficient parasites had a significant effect on IFN-γ production in previously non-infected individuals, suggesting that ROP16 which is considered as a virulence factor plays a role during the primary infection in humans, but not in the secondary immune response.
Collapse
Affiliation(s)
- Alejandro Hernández-de-Los-Ríos
- Grupo de Estudio en Parasitología Molecular (GEPAMOL), Facultad de Ciencias de la Salud, Centro de Investigaciones Biomédicas, Universidad del Quindío, Armenia, Colombia
| | - Mateo Murillo-Leon
- Grupo de Estudio en Parasitología Molecular (GEPAMOL), Facultad de Ciencias de la Salud, Centro de Investigaciones Biomédicas, Universidad del Quindío, Armenia, Colombia
| | - Luz Eliana Mantilla-Muriel
- Grupo Infección e Inmunidad, Facultad de Ciencias de la Salud, Universidad Tecnológica de Pereira, Pereira, Colombia
| | - Ailan Farid Arenas
- Grupo de Estudio en Parasitología Molecular (GEPAMOL), Facultad de Ciencias de la Salud, Centro de Investigaciones Biomédicas, Universidad del Quindío, Armenia, Colombia
| | - Mónica Vargas-Montes
- Grupo de Estudio en Parasitología Molecular (GEPAMOL), Facultad de Ciencias de la Salud, Centro de Investigaciones Biomédicas, Universidad del Quindío, Armenia, Colombia
| | - Néstor Cardona
- Grupo de Estudio en Parasitología Molecular (GEPAMOL), Facultad de Ciencias de la Salud, Centro de Investigaciones Biomédicas, Universidad del Quindío, Armenia, Colombia.,Grupo Infección e Inmunidad, Facultad de Ciencias de la Salud, Universidad Tecnológica de Pereira, Pereira, Colombia.,Universidad Antonio Nariño, Armenia, Colombia
| | - Alejandra de-la-Torre
- Grupo NeURos, Unidad de Inmunología, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogota, Colombia
| | - Juan Carlos Sepúlveda-Arias
- Grupo Infección e Inmunidad, Facultad de Ciencias de la Salud, Universidad Tecnológica de Pereira, Pereira, Colombia
| | - Jorge Enrique Gómez-Marín
- Grupo de Estudio en Parasitología Molecular (GEPAMOL), Facultad de Ciencias de la Salud, Centro de Investigaciones Biomédicas, Universidad del Quindío, Armenia, Colombia
| |
Collapse
|
72
|
Tuladhar S, Kochanowsky JA, Bhaskara A, Ghotmi Y, Chandrasekaran S, Koshy AA. The ROP16III-dependent early immune response determines the subacute CNS immune response and type III Toxoplasma gondii survival. PLoS Pathog 2019; 15:e1007856. [PMID: 31648279 PMCID: PMC6812932 DOI: 10.1371/journal.ppat.1007856] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/25/2019] [Indexed: 12/27/2022] Open
Abstract
Toxoplasma gondii is an intracellular parasite that persistently infects the CNS and that has genetically distinct strains which provoke different acute immune responses. How differences in the acute immune response affect the CNS immune response is unknown. To address this question, we used two persistent Toxoplasma strains (type II and type III) and examined the CNS immune response at 21 days post infection (dpi). Contrary to acute infection studies, type III-infected mice had higher numbers of total CNS T cells and macrophages/microglia but fewer alternatively activated macrophages (M2s) and regulatory T cells (Tregs) than type II-infected mice. By profiling splenocytes at 5, 10, and 21 dpi, we determined that at 5 dpi type III-infected mice had more M2s while type II-infected mice had more pro-inflammatory macrophages and that these responses flipped over time. To test how these early differences influence the CNS immune response, we engineered the type III strain to lack ROP16 (IIIΔrop16), the polymorphic effector protein that drives the early type III-associated M2 response. IIIΔrop16-infected mice showed a type II-like neuroinflammatory response with fewer infiltrating T cells and macrophages/microglia and more M2s and an unexpectedly low CNS parasite burden. At 5 dpi, IIIΔrop16-infected mice showed a mixed inflammatory response with more pro-inflammatory macrophages, M2s, T effector cells, and Tregs, and decreased rates of infection of peritoneal exudative cells (PECs). These data suggested that type III parasites need the early ROP16-associated M2 response to avoid clearance, possibly by the Immunity-Related GTPases (IRGs), which are IFN-γ- dependent proteins essential for murine defenses against Toxoplasma. To test this possibility, we infected IRG-deficient mice and found that IIIΔrop16 parasites now maintained parental levels of PECs infection. Collectively, these studies suggest that, for the type III strain, rop16III plays a key role in parasite persistence and influences the subacute CNS immune response. Toxoplasma is a ubiquitous intracellular parasite that establishes an asymptomatic brain infection in immunocompetent individuals. However, in the immunocompromised and the developing fetus, Toxoplasma can cause problems ranging from fever to chorioretinitis to severe toxoplasmic encephalitis. Emerging evidence suggests that the genotype of the infecting Toxoplasma strain may influence these outcomes, possibly through the secretion of Toxoplasma strain-specific polymorphic effector proteins that trigger different host cell signaling pathways. While such strain-specific modulation of host cell signaling has been shown to affect acute immune responses, it is unclear how these differences influence the subacute or chronic responses in the CNS, the major organ affected in symptomatic disease. This study shows that genetically distinct strains of Toxoplasma provoke strain-specific CNS immune responses and that, for one strain (type III), acute and subacute immune responses and parasite survival are heavily influenced by a polymorphic parasite gene (rop16III).
Collapse
Affiliation(s)
- Shraddha Tuladhar
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
| | - Joshua A. Kochanowsky
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
| | - Apoorva Bhaskara
- Bio5 Institute, University of Arizona, Tucson, Arizona, United States of America
| | - Yarah Ghotmi
- Bio5 Institute, University of Arizona, Tucson, Arizona, United States of America
- Undergraduate Biology Research Program (UBRP), University of Arizona, Tucson, Arizona, United States of America
| | | | - Anita A. Koshy
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
- Bio5 Institute, University of Arizona, Tucson, Arizona, United States of America
- Department of Neurology, University of Arizona, Tucson, Arizona, United States of America
- * E-mail:
| |
Collapse
|
73
|
Hargrave KE, Woods S, Millington O, Chalmers S, Westrop GD, Roberts CW. Multi-Omics Studies Demonstrate Toxoplasma gondii-Induced Metabolic Reprogramming of Murine Dendritic Cells. Front Cell Infect Microbiol 2019; 9:309. [PMID: 31572687 PMCID: PMC6749083 DOI: 10.3389/fcimb.2019.00309] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/12/2019] [Indexed: 01/14/2023] Open
Abstract
Toxoplasma gondii is capable of actively invading almost any mammalian cell type including phagocytes. Early events in phagocytic cells such as dendritic cells are not only key to establishing parasite infection, but conversely play a pivotal role in initiating host immunity. It is now recognized that in addition to changes in canonical immune markers and mediators, alteration in metabolism occurs upon activation of phagocytic cells. These metabolic changes are important for supporting the developing immune response, but can affect the availability of nutrients for intracellular pathogens including T. gondii. However, the interaction of T. gondii with these cells and particularly how infection changes their metabolism has not been extensively investigated. Herein, we use a multi-omics approach comprising transcriptomics and metabolomics validated with functional assays to better understand early events in these cells following infection. Analysis of the transcriptome of T. gondii infected bone marrow derived dendritic cells (BMDCs) revealed significant alterations in transcripts associated with cellular metabolism, activation of T cells, inflammation mediated chemokine and cytokine signaling pathways. Multivariant analysis of metabolomic data sets acquired through non-targeted liquid chromatography mass spectroscopy (LCMS) identified metabolites associated with glycolysis, the TCA cycle, oxidative phosphorylation and arginine metabolism as major discriminants between control uninfected and T. gondii infected cells. Consistent with these observations, glucose uptake and lactate dehydrogenase activity were upregulated in T. gondii infected BMDC cultures compared with control BMDCs. Conversely, BMDC mitochondrial membrane potential was reduced in T. gondii-infected cells relative to mitochondria of control BMDCs. These changes to energy metabolism, similar to what has been described following LPS stimulation of BMDCs and macrophages are often termed the Warburg effect. This metabolic reprogramming of cells has been suggested to be an important adaption that provides energy and precursors to facilitate phagocytosis, antigen processing and cytokine production. Other changes to BMDC metabolism are evident following T. gondii infection and include upregulation of arginine degradation concomitant with increased arginase-1 activity and ornithine and proline production. As T. gondii is an arginine auxotroph the resultant reduced cellular arginine levels are likely to curtail parasite multiplication. These results highlight the complex interplay of BMDCs and parasite metabolism within the developing immune response and the consequences for adaptive immunity and pathogen clearance.
Collapse
Affiliation(s)
- Kerrie E Hargrave
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Stuart Woods
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Owain Millington
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Susan Chalmers
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Gareth D Westrop
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Craig W Roberts
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
74
|
Lopez Corcino Y, Gonzalez Ferrer S, Mantilla LE, Trikeriotis S, Yu JS, Kim S, Hansen S, Portillo JAC, Subauste CS. Toxoplasma gondii induces prolonged host epidermal growth factor receptor signalling to prevent parasite elimination by autophagy: Perspectives for in vivo control of the parasite. Cell Microbiol 2019; 21:e13084. [PMID: 31290228 PMCID: PMC6771541 DOI: 10.1111/cmi.13084] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/03/2019] [Accepted: 07/07/2019] [Indexed: 12/23/2022]
Abstract
Toxoplasma gondii causes retinitis and encephalitis. Avoiding targeting by autophagosomes is key for its survival because T. gondii cannot withstand lysosomal degradation. During invasion of host cells, T. gondii triggers epidermal growth factor receptor (EGFR) signalling enabling the parasite to avoid initial autophagic targeting. However, autophagy is a constitutive process indicating that the parasite may also use a strategy operative beyond invasion to maintain blockade of autophagic targeting. Finding that such a strategy exists would be important because it could lead to inhibition of host cell signalling as a novel approach to kill the parasite in previously infected cells and treat toxoplasmosis. We report that T. gondii induced prolonged EGFR autophosphorylation. This effect was mediated by PKCα/PKCβ ➔ Src because T. gondii caused prolonged activation of these molecules and their knockdown or incubation with inhibitors of PKCα/PKCβ or Src after host cell invasion impaired sustained EGFR autophosphorylation. Addition of EGFR tyrosine kinase inhibitor (TKI) to previously infected cells led to parasite entrapment by LC3 and LAMP-1 and pathogen killing dependent on the autophagy proteins ULK1 and Beclin 1 as well as lysosomal enzymes. Administration of gefitinib (EGFR TKI) to mice with ocular and cerebral toxoplasmosis resulted in disease control that was dependent on Beclin 1. Thus, T. gondii promotes its survival through sustained EGFR signalling driven by PKCα/β ➔ Src, and inhibition of EGFR controls pre-established toxoplasmosis.
Collapse
Affiliation(s)
| | - Shekina Gonzalez Ferrer
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | | | - Sophia Trikeriotis
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jin-Sang Yu
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Steven Kim
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Samuel Hansen
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jose-Andres C Portillo
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Carlos S Subauste
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA.,Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
75
|
Figueiredo CA, Düsedau HP, Steffen J, Gupta N, Dunay MP, Toth GK, Reglodi D, Heimesaat MM, Dunay IR. Immunomodulatory Effects of the Neuropeptide Pituitary Adenylate Cyclase-Activating Polypeptide in Acute Toxoplasmosis. Front Cell Infect Microbiol 2019; 9:154. [PMID: 31192159 PMCID: PMC6546896 DOI: 10.3389/fcimb.2019.00154] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/26/2019] [Indexed: 12/21/2022] Open
Abstract
Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) is an endogenous neuropeptide with distinct functions including the regulation of inflammatory processes. PACAP is able to modify the immune response by directly regulating macrophages and monocytes inhibiting the production of inflammatory cytokines, chemokines and free radicals. Here, we analyzed the effect of exogenous PACAP on peripheral immune cell subsets upon acute infection with the parasite Toxoplasma gondii (T. gondii). PACAP administration was followed by diminished innate immune cell recruitment to the peritoneal cavity of T. gondii-infected mice. PACAP did not directly interfere with parasite replication, instead, indirectly reduced parasite burden in mononuclear cell populations by enhancing their phagocytic capacity. Although proinflammatory cytokine levels were attenuated in the periphery upon PACAP treatment, interleukin (IL)-10 and Transforming growth factor beta (TGF-β) remained stable. While PACAP modulated VPAC1 and VPAC2 receptors in immune cells upon binding, it also increased their expression of brain-derived neurotrophic factor (BDNF). In addition, the expression of p75 neurotrophin receptor (p75NTR) on Ly6Chi inflammatory monocytes was diminished upon PACAP administration. Our findings highlight the immunomodulatory effect of PACAP on peripheral immune cell subsets during acute Toxoplasmosis, providing new insights about host-pathogen interaction and the effects of neuropeptides during inflammation.
Collapse
Affiliation(s)
- Caio Andreeta Figueiredo
- Medical Faculty, Institute of Inflammation and Neurodegeneration, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Henning Peter Düsedau
- Medical Faculty, Institute of Inflammation and Neurodegeneration, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Johannes Steffen
- Medical Faculty, Institute of Inflammation and Neurodegeneration, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Nishith Gupta
- Faculty of Life Sciences, Institute of Biology, Humboldt University, Berlin, Germany
| | - Miklos Pal Dunay
- Department and Clinic of Surgery and Ophthalmology, University of Veterinary Medicine, Budapest, Hungary
| | - Gabor K Toth
- Department of Medical Chemistry, University of Szeged, Szeged, Hungary
| | - Dora Reglodi
- Department of Anatomy, MTA-PTE PACAP Research Team, University of Pecs Medical School, Pecs, Hungary
| | - Markus M Heimesaat
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| | - Ildiko Rita Dunay
- Medical Faculty, Institute of Inflammation and Neurodegeneration, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences - CBBS, Magdeburg, Germany
| |
Collapse
|
76
|
Menard KL, Haskins BE, Denkers EY. Impact of Toxoplasma gondii Infection on Host Non-coding RNA Responses. Front Cell Infect Microbiol 2019; 9:132. [PMID: 31157172 PMCID: PMC6530353 DOI: 10.3389/fcimb.2019.00132] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 04/12/2019] [Indexed: 12/12/2022] Open
Abstract
As an intracellular microbe, Toxoplasma gondii must establish a highly intimate relationship with its host to ensure success as a parasite. Many studies over the last decade-and-a-half have highlighted how the host reshapes its immunoproteome to survive infection, and conversely how the parasite regulates host responses to ensure persistence. The role of host non-protein-coding RNA during infection is a vast and largely unexplored area of emerging interest. The potential importance of this facet of the host-parasite interaction is underscored by current estimates that as much as 80% of the host genome is transcribed into non-translated RNA. Here, we review the current state of knowledge with respect to two major classes of non-coding RNA, microRNA (miRNA) and long non-coding RNA (lncRNA), in the host response to T. gondii infection. These two classes of regulatory RNA are known to have profound and widespread effects on cell function. However, their impact on infection and immunity is not well-understood, particularly for the response to T. gondii. Nevertheless, numerous miRNAs have been identified that are upregulated by Toxoplasma, and emerging evidence suggests a functional role during infection. While the field of lncRNA is in its infancy, it is already clear that Toxoplasma is also a strong trigger for this class of regulatory RNA. Non-coding RNA responses induced by T. gondii are likely to be major determinants of the host's ability to resist infection and the parasite's ability to establish long-term latency.
Collapse
Affiliation(s)
- Kayla L Menard
- Department of Biology, Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM, United States
| | - Breanne E Haskins
- Department of Biology, Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM, United States
| | - Eric Y Denkers
- Department of Biology, Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
77
|
Toxoplasma gondii Modulates the Host Cell Responses: An Overview of Apoptosis Pathways. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6152489. [PMID: 31080827 PMCID: PMC6475534 DOI: 10.1155/2019/6152489] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 03/13/2019] [Accepted: 03/26/2019] [Indexed: 01/29/2023]
Abstract
Infection with Toxoplasma gondii has a major implication in public health. Toxoplasma gondii is an obligate intracellular protozoan parasite that can infect all nucleated cells belonging to a wide range of host species. One of the particularities of this parasite is its invasion and persistence in host cells of immunocompetent people. This infection is usually asymptomatic. In immunocompromised patients, the infection is severe and symptomatic. The mechanisms by which T. gondii persists are poorly studied in humans. In mouse models, many aspects of the interaction between the parasite and the host cells are being studied. Apoptosis is one of these mechanisms that could be modulated by Toxoplasma to persist in host cells. Indeed, Toxoplasma has often been implicated in the regulation of apoptosis and viability mechanisms in both human and murine infection models. Several of these studies centered on the regulation of apoptosis pathways have revealed interference of this parasite with host cell immunity, cell signalling, and invasion mechanisms. This review provides an overview of recent studies concerning the effect of Toxoplasma on different apoptotic pathways in infected host cells.
Collapse
|
78
|
Rochet E, Argy N, Greigert V, Brunet J, Sabou M, Marcellin L, de-la-Torre A, Sauer A, Candolfi E, Pfaff AW. Type I ROP16 regulates retinal inflammatory responses during ocular toxoplasmosis. PLoS One 2019; 14:e0214310. [PMID: 30901349 PMCID: PMC6430381 DOI: 10.1371/journal.pone.0214310] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 03/11/2019] [Indexed: 11/18/2022] Open
Abstract
Ocular toxoplasmosis (OT), mostly retinochorioditis, is a major feature of infection with the protozoan parasite Toxoplasma gondii. The pathophysiology of this infection is still largely elusive; especially mouse models are not yet well developed. In contrast, numerous in vitro studies showed the highly Toxoplasma strain dependent nature of the host-parasite interactions. Some distinct polymorphic virulence factors were characterized, notably the rhoptry protein ROP16. Here, we studied the strain-dependent pathophysiology in our OT mouse model. Besides of two wild type strains of the canonical I (RH, virulent) and II (PRU, avirulent) types, we used genetically engineered parasites, RHΔROP16 and PRU ROP16-I, expressing the type I allele of this virulence factor. We analyzed retinal integrity, parasite proliferation and retinal expression of cytokines. PRU parasites behaved much more virulently in the presence of a type I ROP16. In contrast, knockout of ROP16 in the RH strain led to a decrease of intraocular proliferation, but no difference in retinal pathology. Cytokine quantification in aqueous humor showed strong production of Th1 and inflammatory markers following infection with the two strains containing the ROP16-I allele. In strong contrast, immunofluorescence images showed that actual expression of most cytokines in retinal cells is rapidly suppressed by type I strain infection, with or without the involvement of its homologous ROP16 allele. This demonstrates the particular immune privileged situation of the retina, which is also revealed by the fact that parasite proliferation is nearly exclusively observed outside the retina. In summary, we further developed a promising OT mouse model and demonstrated the specific pathology in retinal tissues.
Collapse
Affiliation(s)
- Elise Rochet
- Institut de Parasitologie et Pathologie Tropicale, EA, Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| | - Nicolas Argy
- Institut de Parasitologie et Pathologie Tropicale, EA, Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| | - Valentin Greigert
- Institut de Parasitologie et Pathologie Tropicale, EA, Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
| | - Julie Brunet
- Institut de Parasitologie et Pathologie Tropicale, EA, Fédération de Médecine Translationnelle, 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
| | - Marcela Sabou
- Institut de Parasitologie et Pathologie Tropicale, EA, Fédération de Médecine Translationnelle, 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
| | - Luc Marcellin
- Service de Pathologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Alejandra de-la-Torre
- Institut de Parasitologie et Pathologie Tropicale, EA, Fédération de Médecine Translationnelle, Université de Strasbourg, Strasbourg, France
- Grupo NeURos, Unidad de Inmunología, Universidad del Rosario, Escuela de Medicina y Ciencias de la Salud, Bogotá, Colombia
| | - Arnaud Sauer
- Service d’Ophtalmologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Ermanno Candolfi
- Institut de Parasitologie et Pathologie Tropicale, EA, Fédération de Médecine Translationnelle, 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 Pathologie Tropicale, EA, Fédération de Médecine Translationnelle, 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
- * E-mail:
| |
Collapse
|
79
|
Wang JL, Zhang NZ, Li TT, He JJ, Elsheikha HM, Zhu XQ. Advances in the Development of Anti-Toxoplasma gondii Vaccines: Challenges, Opportunities, and Perspectives. Trends Parasitol 2019; 35:239-253. [PMID: 30718083 DOI: 10.1016/j.pt.2019.01.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/03/2019] [Accepted: 01/08/2019] [Indexed: 12/14/2022]
Abstract
Important progress has been made in understanding how immunity is elicited against Toxoplasma gondii - a complex pathogen with multiple mechanisms of immune evasion. Many vaccine candidates have been tested using various strategies in animal models. However, none of these strategies has delivered as yet, and important challenges remain in the development of vaccines that can eliminate the tissue cysts and/or fully block vertical transmission. In this review, we provide an overview of the current understanding of the host immune response to T. gondii infection and summarize the key limitations for the development of an effective, safe, and durable toxoplasmosis vaccine. We also discuss how the successes and failures in developing and testing vaccine candidates have provided a roadmap for future vaccine development.
Collapse
Affiliation(s)
- Jin-Lei Wang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, PR China
| | - Nian-Zhang Zhang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, PR China
| | - Ting-Ting Li
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, PR China
| | - Jun-Jun He
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, PR China
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough, LE12 5RD, UK.
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, PR China.
| |
Collapse
|
80
|
Foroutan M, Ghaffarifar F, Sharifi Z, Dalimi A, Jorjani O. Rhoptry antigens as Toxoplasma gondii vaccine target. Clin Exp Vaccine Res 2019; 8:4-26. [PMID: 30775347 PMCID: PMC6369123 DOI: 10.7774/cevr.2019.8.1.4] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 07/10/2018] [Accepted: 01/14/2019] [Indexed: 01/14/2023] Open
Abstract
Toxoplasmosis is a cosmopolitan zoonotic infection, caused by a unicellular protozoan parasite known as Toxoplasma gondii that belongs to the phylum Apicomplexa. It is estimated that over one-third of the world's population has been exposed and are latently infected with the parasite. In humans, toxoplasmosis is predominantly asymptomatic in immunocompetent persons, while among immunocompromised individuals may be cause severe and progressive complications with poor prognosis. Moreover, seronegative pregnant mothers are other risk groups for acquiring the infection. The life cycle of T. gondii is very complex, indicating the presence of a plurality of antigenic epitopes. Despite of great advances, recognize and construct novel vaccines for prevent and control of toxoplasmosis in both humans and animals is still remains a great challenge for researchers to select potential protein sequences as the ideal antigens. Notably, in several past years, constant efforts of researchers have made considerable advances to elucidate the different aspects of the cell and molecular biology of T. gondii mainly on microneme antigens, dense granule antigens, surface antigens, and rhoptry proteins (ROP). These attempts thereby provided great impetus to the present focus on vaccine development, according to the defined subcellular components of the parasite. Although, currently there is no commercial vaccine for use in humans. Among the main identified T. gondii antigens, ROPs appear as a putative vaccine candidate that are vital for invasion procedure as well as survival within host cells. Overall, it is estimated that they occupy about 1%–30% of the total parasite cell volume. In this review, we have summarized the recent progress of ROP-based vaccine development through various strategies from DNA vaccines, epitope or multi epitope-based vaccines, recombinant protein vaccines to vaccines based on live-attenuated vectors and prime-boost strategies in different mouse models.
Collapse
Affiliation(s)
- Masoud Foroutan
- Abadan School of Medical Sciences, Abadan, Iran.,Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Ghaffarifar
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zohreh Sharifi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Abdolhosein Dalimi
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ogholniaz Jorjani
- Laboratory Science Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
81
|
Safronova A, Araujo A, Camanzo ET, Moon TJ, Elliott MR, Beiting DP, Yarovinsky F. Alarmin S100A11 initiates a chemokine response to the human pathogen Toxoplasma gondii. Nat Immunol 2018; 20:64-72. [PMID: 30455460 PMCID: PMC6291348 DOI: 10.1038/s41590-018-0250-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 10/01/2018] [Indexed: 12/31/2022]
Abstract
Toxoplasma gondii is a common protozoan parasite that infects up to one-third of the world’s population. Notably, very little is known about innate immune-sensing mechanisms for this obligate intracellular parasite by human cells. Here, by applying an unbiased biochemical screening approach, we have identified that human monocytes recognized the presence of T. gondii infection via detection of the alarmin S100A11 protein, which is released from parasite-infected cells via caspase-1-dependent mechanisms. S100A11 induced a potent chemokine response to T. gondii via engagement of its receptor RAGE and regulated monocyte recruitment in vivo by inducing expression of the chemokine CCL2. Our experiments have revealed a sensing system for T. gondii by human cells that is based on detection infection-mediated release of alarmin S100A11 and RAGE-dependent induction of CCL2, a crucial chemokine required for host resistance to the parasite.
Collapse
Affiliation(s)
- Alexandra Safronova
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Alessandra Araujo
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Ellie T Camanzo
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Taylor J Moon
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Michael R Elliott
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Daniel P Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Felix Yarovinsky
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
82
|
Cong W, Dottorini T, Khan F, Emes RD, Zhang FK, Zhou CX, He JJ, Zhang XX, Elsheikha HM, Zhu XQ. Acute Toxoplasma Gondii Infection in Cats Induced Tissue-Specific Transcriptional Response Dominated by Immune Signatures. Front Immunol 2018; 9:2403. [PMID: 30405608 PMCID: PMC6202952 DOI: 10.3389/fimmu.2018.02403] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/28/2018] [Indexed: 12/19/2022] Open
Abstract
RNA-sequencing was used to detect transcriptional changes in six tissues of cats, seven days after T. gondii infection. A total of 737 genes were differentially expressed (DEGs), of which 410 were up-regulated and 327 were down-regulated. The liver exhibited 151 DEGs, lung (149 DEGs), small intestine (130 DEGs), heart (123 DEGs), brain (104 DEGs), and spleen (80 DEGs)-suggesting tissue-specific transcriptional patterns. Gene ontology and KEGG analyses identified DEGs enriched in immune pathways, such as cytokine-cytokine receptor interaction, Jak-STAT signaling pathway, NOD-like receptor signaling pathway, NF-kappa B signaling pathway, MAPK signaling pathway, T cell receptor signaling pathway, and the cytosolic DNA sensing pathway. C-X-C motif chemokine 10 (CXCL10) was involved in most of the immune-related pathways. PI3K/Akt expression was down-regulated in all tissues, except the spleen. The genes for phosphatase, indoleamine 2,3-dioxygenase, Hes Family BHLH Transcription Factor 1, and guanylate-binding protein 5, playing various roles in immune defense, were co-expressed across various feline tissues. Multivariate K-means clustering analysis produced seven gene clusters featuring similar gene expression patterns specific to individual tissues, with lung tissue cluster having the largest number of DEGs. These findings suggest the presence of a broad immune defense mechanism across various tissues in cats against acute T. gondii infection.
Collapse
Affiliation(s)
- Wei Cong
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Department of Marine Engineering, Marine College, Shandong University, Weihai, China
| | - Tania Dottorini
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom.,Advanced Data Analysis Centre, University of Nottingham, Loughborough, United Kingdom
| | - Faraz Khan
- Advanced Data Analysis Centre, University of Nottingham, Loughborough, United Kingdom
| | - Richard D Emes
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom.,Advanced Data Analysis Centre, University of Nottingham, Loughborough, United Kingdom
| | - Fu-Kai Zhang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Chun-Xue Zhou
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Jun-Jun He
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiao-Xuan Zhang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
83
|
Menard KL, Haskins BE, Colombo AP, Denkers EY. Toxoplasma gondii Manipulates Expression of Host Long Noncoding RNA during Intracellular Infection. Sci Rep 2018; 8:15017. [PMID: 30301916 PMCID: PMC6177471 DOI: 10.1038/s41598-018-33274-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/26/2018] [Indexed: 12/16/2022] Open
Abstract
Long noncoding RNA (lncRNA) are non-protein-coding transcripts greater than 200 nucleotides that regulate gene expression. The field of transcriptomics is only beginning to understand the role of lncRNA in host defense. Little is known about the role of lncRNA in the response to infection by intracellular pathogens such as Toxoplasma gondii. Using a microarray, we examined the differential expression of 35,923 lncRNAs and 24,881 mRNAs in mouse bone-marrow-derived macrophages during infection with high- and low-virulence T. gondii strains. We found that 1,522 lncRNA molecules were differentially regulated during infection with the high-virulence Type I strain, versus 528 with the less-virulent Type II strain. Of these lncRNAs, 282 were co-regulated with a nearby or overlapping mRNA–including approximately 60 mRNAs with immune-related functions. We validated the microarray for 4 lncRNAs and 4 mRNAs using qRT-PCR. Using deletion strains of T. gondii, we found that the secretory kinase ROP16 controls upregulation of lncRNAs Csf1-lnc and Socs2-lnc, demonstrating that the parasite directly manipulates host lncRNA expression. Given the number of regulated lncRNAs and the magnitude of the expression changes, we hypothesize that these molecules constitute both an additional regulatory layer in the host response to infection and a target for manipulation by T. gondii.
Collapse
Affiliation(s)
- Kayla L Menard
- Department of Biology and Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Breanne E Haskins
- Department of Biology and Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM, 87131, USA
| | | | - Eric Y Denkers
- Department of Biology and Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM, 87131, USA.
| |
Collapse
|
84
|
Cabral GRDA, Wang ZT, Sibley LD, DaMatta RA. Inhibition of Nitric Oxide Production in Activated Macrophages Caused by Toxoplasma gondii Infection Occurs by Distinct Mechanisms in Different Mouse Macrophage Cell Lines. Front Microbiol 2018; 9:1936. [PMID: 30177926 PMCID: PMC6109688 DOI: 10.3389/fmicb.2018.01936] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 07/31/2018] [Indexed: 12/23/2022] Open
Abstract
Toxoplasma gondii, the causative agent of toxoplasmosis, is a widespread intracellular parasite able to infect virtually any nucleated cell. T. gondii infection of activated macrophages inhibits nitric oxide (NO) production; however, parasite effectors responsible for this block have not been defined. Macrophage populations are extremely heterogeneous, responding differently to stimuli and to parasite infection. Here we evaluated the inhibition of NO production caused by T. gondii infection of J774-A1 and RAW 264.7 macrophages and assessed the role of several known parasite virulence factors in this phenotype. Infection of activated macrophages from both macrophage lines reduced NO production, however, the mechanism of this decrease was different. Consistent with previous reports, infected J774-A1 macrophages had reduced iNOS expression and lower number of iNOS positive cells. In contrast, T. gondii infection of RAW 264.7 macrophages did not alter iNOS expression or the number of iNOS positive cells, and yet it led to lower levels of NO production. Deletion of a number of previously defined virulence factors including ROP kinases that disrupt innate immune factors, TgIST which blocks STAT1 activation, as well as the secretory trafficking proteins ASP5 and MYR1, did not alter the phenotype of decreased NO production. Taken together our findings indicate that T. gondii infection inhibits NO production of activated macrophages by different mechanisms that involve reduction of iNOS expression vs. iNOS impairment, and suggest that a novel parasite effector is involved in modulating this important host defense pathway.
Collapse
Affiliation(s)
- Gabriel R de Abreu Cabral
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, United States.,Laboratório de Biologia Celular e Tecidual, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, Brazil
| | - Zi T Wang
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, United States
| | - L D Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Renato A DaMatta
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, United States.,Laboratório de Biologia Celular e Tecidual, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, Brazil
| |
Collapse
|
85
|
Liu L, Jin M, Tao Q, Yu L, Du J, Wang C, Luo Q, Xing T, Xu Y, Shen J, Chu D. Effective Amelioration of Liver Fibrosis Through Lentiviral Vector Carrying Toxoplasma gondii gra15II in Murine Model. Front Immunol 2018; 9:1572. [PMID: 30034399 PMCID: PMC6043635 DOI: 10.3389/fimmu.2018.01572] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 06/25/2018] [Indexed: 11/24/2022] Open
Abstract
Our previous investigations indicated that in vitro polarization of mouse macrophages by Toxoplasma gondii type II strain dense granule protein 15 (GRA15 II ), one of the genotype-associated effectors of T. gondii, induced the phenotypes of classically activated macrophage (M1). Transfusion of the cells to mice may effectively alleviated hepatic fibrosis caused by schistosomiasis. The purpose of the study was to identify whether liver macrophages can be in vivo driven to M1 macrophages by lentiviral vector (LV) carrying GRA15 II gene (LV-gra15 II ) and to explore the potential mechanism by which the LV-gra15 II -activated liver macrophage (LV-gra15 II -M) ameliorates the hepatic fibrosis in schistosomiasis. The mice were treated with LV-gra15 II by hydrodynamic injection via the tail vein followed by challenge of Schistosoma japonicum (S. japonicum). Our experiments showed that LV-gra15 II was successfully delivered to liver macrophages and GRA15II was persistently expressed in the macrophages of mice for at least 2 months. Furthermore, the LV-gra15 II infected macrophages were polarized to M1 macrophages in vivo. Consequently, mice with schistosomiasis receiving LV-gra15 II injection displayed a remarkable amelioration of liver granuloma formation and collagen deposition in association with downregulated expression of transforming growth factor-beta1, arginase 1 (Arg-1), α-smooth muscle actin, and an increased expression of matrix metalloproteinase 13 (MMP13). Simultaneously, no negative effects of liver function and vitality of mice were noted. The in vitro experiments indicated that the C-C motif chemokine ligand 2 and nitric oxide level were elevated in LV-gra15 II -M cultural supernatants; hepatocyte growth factor expression was enhanced in LV-gra15 II -M. In addition, LV-gra15 II -M not only secreted MMP13, which greatly degraded type I collagen, but also induced murine hepatic stellate cell (HSC) line (JS1) apoptosis in the co-culture system. Taken together, we identified for the first time that LV-gra15 II may in vivo drive liver macrophages to M1 macrophage phenotypes, which helps for alteration of the liver fibrotic microenvironment with collagen dissolution, HSC deactivation, apoptosis and hepatocyte protection. Our study gives an insight into the use of gene delivery with parasite-derived immunomodulatory factor as a potential immune cell activating agent to re-equilibrate the other pathogen-induced immune response in some chronic diseases.
Collapse
Affiliation(s)
- Lei Liu
- Department of Pathogen Biology and Provincial Laboratories of Pathogen Biology and Zoonoses, Anhui Medical University, Hefei, China
| | - Mengmeng Jin
- Department of Pathogen Biology and Provincial Laboratories of Pathogen Biology and Zoonoses, Anhui Medical University, Hefei, China
| | - Qing Tao
- Department of Pathogen Biology and Provincial Laboratories of Pathogen Biology and Zoonoses, Anhui Medical University, Hefei, China
| | - Li Yu
- Department of Pathogen Biology and Provincial Laboratories of Pathogen Biology and Zoonoses, Anhui Medical University, Hefei, China
| | - Jian Du
- Department of Pathogen Biology and Provincial Laboratories of Pathogen Biology and Zoonoses, Anhui Medical University, Hefei, China
| | - Cong Wang
- Department of Pathogen Biology and Provincial Laboratories of Pathogen Biology and Zoonoses, Anhui Medical University, Hefei, China
| | - Qingli Luo
- Department of Pathogen Biology and Provincial Laboratories of Pathogen Biology and Zoonoses, Anhui Medical University, Hefei, China
| | - Tian Xing
- Key Laboratory of Oral Disease Research of Anhui Province, Stomatologic Hospital and College, Anhui Medical University, Hefei, China
| | - Yuanhong Xu
- Diagnostic Laboratory of the First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Jilong Shen
- Department of Pathogen Biology and Provincial Laboratories of Pathogen Biology and Zoonoses, Anhui Medical University, Hefei, China
| | - Deyong Chu
- Department of Pathogen Biology and Provincial Laboratories of Pathogen Biology and Zoonoses, Anhui Medical University, Hefei, China
| |
Collapse
|
86
|
Zhang Z, Li Y, Xie Q, Li P, Nan X, Kong L, Zeng D, Ding Z, Wang S. The Molecular Characterization and Immunity Identification of Rhoptry Protein 22 of Toxoplasma gondii
as a DNA Vaccine Candidate Against Toxoplasmosis. J Eukaryot Microbiol 2018; 66:147-157. [PMID: 29858559 DOI: 10.1111/jeu.12639] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/08/2018] [Accepted: 05/18/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Zhenchao Zhang
- School of Basic Medical Sciences; Xinxiang Medical University; Xinxiang Henan 453003 China
| | - Yuhua Li
- The First Affiliated Hospital of Xinxiang Medical University; Xinxiang Henan 453003 China
| | - Qing Xie
- School of Basic Medical Sciences; Xinxiang Medical University; Xinxiang Henan 453003 China
| | - Pengju Li
- School of Basic Medical Sciences; Xinxiang Medical University; Xinxiang Henan 453003 China
| | - Xiaoxu Nan
- School of Stomatology; Xinxiang Medical University; Xinxiang Henan 453003 China
| | - Lingmin Kong
- School of Basic Medical Sciences; Xinxiang Medical University; Xinxiang Henan 453003 China
| | - Dapeng Zeng
- The First Affiliated Hospital of Xinxiang Medical University; Xinxiang Henan 453003 China
| | - Zhifang Ding
- School of Basic Medical Sciences; Xinxiang Medical University; Xinxiang Henan 453003 China
| | - Shuai Wang
- School of Basic Medical Sciences; Xinxiang Medical University; Xinxiang Henan 453003 China
| |
Collapse
|
87
|
Zhang Z, Li Y, Wang M, Xie Q, Li P, Zuo S, Kong L, Wang C, Wang S. Immune Protection of Rhoptry Protein 21 (ROP21) of Toxoplasma gondii as a DNA Vaccine Against Toxoplasmosis. Front Microbiol 2018; 9:909. [PMID: 29867820 PMCID: PMC5951983 DOI: 10.3389/fmicb.2018.00909] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 04/19/2018] [Indexed: 12/31/2022] Open
Abstract
Toxoplasma gondii rhoptry proteins (TgROPs) are the major targets as key molecules for immunodiagnosis as well as immunoprophylaxis because of their initial presentation to the host immune system. In this work, it was aimed at evaluating the protection effect of TgROP21 DNA vaccine on experimental mice subjected to T. gondii challenge. The gene sequence encoding TgROP21 was inserted into the eukaryotic expression vector pVAX I, and western blotting indicates that the lysate of BHK cells transfected with pVAX-TgROP21 was specifically recognized as a band of about 82.6 kDa by serum obtained from a T. gondii infected chicken. The efficacy of intramuscular vaccination of BALB/c mice three times at weeks 0, 2, and 4 with pVAX-ROP21 was analyzed. The levels of IgG, IgG1, and IgG2a among pVAX-ROP21 vaccinated animals were integrally increased. It was uncovered by cytokine profile analyses that IFN-γ was significantly increased, while no significant changes were detected in interleukin-2 (IL-2), interleukin-4 (IL-4), and interleukin-10 (IL-10). Additionally, we found that immunization with pVAX-ROP21 significantly prolonged survival time (13.50 ± 1.65 days) after challenge infection with the virulent T. gondii RH strain, in comparison to those of control animals (died within 10 days). Moreover, the number of brain cysts (1475 ± 163) in the animals subjected to pVAX-TgROP21 vaccination decreased remarkably (P < 0.05) compared to the blank control mice (2333 ± 473), and the size of brain cysts in pVAX-TgROP21 group was significantly smaller than the groups of blank, PBS and pVAXI. It was indicated that intense cell-mediated and humoral immunity was triggered and defense against T. gondii was partially induced after vaccination by TgROP21.
Collapse
Affiliation(s)
- Zhenchao Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yuhua Li
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Mingyong Wang
- Henan Key Laboratory of Immunology and Targeted Therapy, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Qing Xie
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Pengju Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Suqiong Zuo
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Lingmin Kong
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Chenxing Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Shuai Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
88
|
Naor A, Panas MW, Marino N, Coffey MJ, Tonkin CJ, Boothroyd JC. MYR1-Dependent Effectors Are the Major Drivers of a Host Cell's Early Response to Toxoplasma, Including Counteracting MYR1-Independent Effects. mBio 2018; 9:e02401-17. [PMID: 29615509 PMCID: PMC5885026 DOI: 10.1128/mbio.02401-17] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 03/07/2018] [Indexed: 01/08/2023] Open
Abstract
The obligate intracellular parasite Toxoplasma gondii controls its host cell from within the parasitophorous vacuole (PV) by using a number of diverse effector proteins, a subset of which require the aspartyl protease 5 enzyme (ASP5) and/or the recently discovered MYR1 protein to cross the PV membrane. To examine the impact these effectors have in the context of the entirety of the host response to Toxoplasma, we used RNA-Seq to analyze the transcriptome expression profiles of human foreskin fibroblasts infected with wild-type RH (RH-WT), RHΔmyr1, and RHΔasp5 tachyzoites. Interestingly, the majority of the differentially regulated genes responding to Toxoplasma infection are MYR1 dependent. A subset of MYR1 responses were ASP5 independent, and MYR1 function did not require ASP5 cleavage, suggesting the export of some effectors requires only MYR1. Gene set enrichment analysis of MYR1-dependent host responses suggests an upregulation of E2F transcription factors and the cell cycle and a downregulation related to interferon signaling, among numerous others. Most surprisingly, "hidden" responses arising in RHΔmyr1- but not RH-WT-infected host cells indicate counterbalancing actions of MYR1-dependent and -independent activities. The host genes and gene sets revealed here to be MYR1 dependent provide new insight into the parasite's ability to co-opt host cell functions.IMPORTANCEToxoplasma gondii is unique in its ability to successfully invade and replicate in a broad range of host species and cells within those hosts. The complex interplay of effector proteins exported by Toxoplasma is key to its success in co-opting the host cell to create a favorable replicative niche. Here we show that a majority of the transcriptomic effects in tachyzoite-infected cells depend on the activity of a novel translocation system involving MYR1 and that the effectors delivered by this system are part of an intricate interplay of activators and suppressors. Removal of all MYR1-dependent effectors reveals previously unknown activities that are masked or hidden by the action of these proteins.
Collapse
Affiliation(s)
- Adit Naor
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Michael W Panas
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Nicole Marino
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Michael J Coffey
- Division of Infection and Immunity, The Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Australia
| | - Christopher J Tonkin
- Division of Infection and Immunity, The Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Australia
| | - John C Boothroyd
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
89
|
Pan M, Zhou Y, Wang Y, Li L, Song Y, Hou L, Zhao J. Screening and Identification of the Host Proteins Interacting with Toxoplasma gondii Rhoptry Protein ROP16. Front Microbiol 2017; 8:2408. [PMID: 29255456 PMCID: PMC5722834 DOI: 10.3389/fmicb.2017.02408] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 11/20/2017] [Indexed: 01/04/2023] Open
Abstract
Toxoplasma gondii, as a zoonotic protozoan parasite, develops sophisticated strategies to manipulate hosts for efficient intracellular survival. After successful invasion, T. gondii injects many effector proteins into host cells for various purposes. TgROP16 (T. gondii rhoptry protein 16), which is secreted from rhoptries into host cells, can activate the host STAT (signal transducer and activator of transcription) signaling pathway through phosphorylation of STAT3 and STAT6. However, whether there are other host proteins modulated by TgROP16 is currently unknown. In this study, yeast two-hybrid (Y2H) screen was used to look for additional host proteins interacting with TgROP16. Yeast cells expressing a mouse cDNA library cloned into the prey vector were used to mate with yeasts expressing ROP16 without signal peptide. Two mouse proteins, Dnaja1 (DnaJ heat shock protein family member A1) and Gabra4 (gamma-aminobutyric acid A receptor, subunit alpha 4) were identified to interact with ROP16 from this screen. Further analysis suggested that the Predomain of ROP16 played key roles in mediating interactions with these host proteins, whereas the contribution from the Kinase domain was minor. The interactions between Dnaja1 and different parts of ROP16 were also estimated in vivo by co-immunoprecipitation. The results showed that the Predomain of ROP16 was the major region to interact with Dnaja1, which is consistent with the Y2H results. Based on the gene ontology analysis, Dnaja1 is predicted to participate in stress response while Gabra4 is involved in the system development process. The discovery of new host proteins that interact with ROP16 of T. gondii will help us to further investigate the functions of this effector proteins during T. gondii infection.
Collapse
Affiliation(s)
- Ming Pan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yanqin Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory for Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China.,Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Yifan Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Longjiao Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yongle Song
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Lun Hou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Junlong Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory for Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China.,Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| |
Collapse
|
90
|
Yakubu RR, Weiss LM, Silmon de Monerri NC. Post-translational modifications as key regulators of apicomplexan biology: insights from proteome-wide studies. Mol Microbiol 2017; 107:1-23. [PMID: 29052917 DOI: 10.1111/mmi.13867] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/12/2017] [Accepted: 10/16/2017] [Indexed: 12/12/2022]
Abstract
Parasites of the Apicomplexa phylum, such as Plasmodium spp. and Toxoplasma gondii, undergo complex life cycles involving multiple stages with distinct biology and morphologies. Post-translational modifications (PTMs), such as phosphorylation, acetylation and glycosylation, regulate numerous cellular processes, playing a role in every aspect of cell biology. PTMs can occur on proteins at any time in their lifespan and through alterations of target protein activity, localization, protein-protein interactions, among other functions, dramatically increase proteome diversity and complexity. In addition, PTMs can be induced or removed on changes in cellular environment and state. Thus, PTMs are likely to be key regulators of developmental transitions, biology and pathogenesis of apicomplexan parasites. In this review we examine the roles of PTMs in both parasite-specific and conserved eukaryotic processes, and the potential crosstalk between PTMs, that together regulate the intricate lives of these protozoa.
Collapse
Affiliation(s)
- Rama R Yakubu
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10128, USA.,Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10128, USA
| | - Louis M Weiss
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10128, USA.,Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10128, USA
| | - Natalie C Silmon de Monerri
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10128, USA.,Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10128, USA
| |
Collapse
|
91
|
Pierog PL, Zhao Y, Singh S, Dai J, Yap GS, Fitzgerald-Bocarsly P. Toxoplasma gondii Inactivates Human Plasmacytoid Dendritic Cells by Functional Mimicry of IL-10. THE JOURNAL OF IMMUNOLOGY 2017; 200:186-195. [PMID: 29180487 DOI: 10.4049/jimmunol.1701045] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/20/2017] [Indexed: 01/07/2023]
Abstract
Plasmacytoid dendritic cells (pDCs) are the major producers of IFN-α, an antiviral cytokine involved in immunomodulation and control of HIV type 1 replication, whereas Toxoplasma gondii is a life-threatening opportunistic infection in AIDS patients. During infection with HIV type 1, human pDCs decrease in circulation and remaining pDC produce lower amounts of IFN-α in response to viral stimulation. In this study, we investigated the impact of coinfection with T. gondii on the innate virus-directed responses of human pDCs. Using intracellular flow cytometry and fluorescence microscopy, we determined that T. gondii invaded but did not induce IFN-α or TNF-α in human pDC. However, T. gondii inhibited IFN-α and TNF-α produced in response to HSV and HIV, thus functionally inactivating pDC. IFN-α production was inhibited only in cells infected by T. gondii, which inhibited neither uptake of GFP-HSV nor localization of TLR9 in CD71+ endosomes, directing us to investigate downstream events. Using imaging flow cytometry, we found that both T. gondii and IL-10 inhibited virus-induced nuclear translocation, but not phosphorylation, of IFN response factor 7. Blockade of IFN response factor 7 nuclear translocation and inhibition of the IFN-α response was partially reversed by a deficiency in the T. gondii-derived ROP16 kinase, known to directly phosphorylate STAT3, a critical mediator of IL-10's anti-inflammatory effects. Taken together, our results indicate that T. gondii suppresses pDC activation by mimicking IL-10's regulatory effects through an ROP16 kinase-dependent mechanism. Our findings further imply a convergent mechanism of inhibition of TLR signaling by T. gondii and IL-10 and suggest potential negative consequences of HIV/T. gondii coinfection.
Collapse
Affiliation(s)
- Piotr L Pierog
- Department of Pathology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103.,Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103.,Rutgers School of Graduate Studies, Newark, NJ 07103; and
| | - Yanlin Zhao
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103.,Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Sukhwinder Singh
- Department of Pathology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Jihong Dai
- Department of Pathology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103.,Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - George S Yap
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103.,Rutgers School of Graduate Studies, Newark, NJ 07103; and.,Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103
| | - Patricia Fitzgerald-Bocarsly
- Department of Pathology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103; .,Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103.,Rutgers School of Graduate Studies, Newark, NJ 07103; and
| |
Collapse
|
92
|
Portillo JAC, Muniz-Feliciano L, Lopez Corcino Y, Lee SJ, Van Grol J, Parsons SJ, Schiemman WP, Subauste CS. Toxoplasma gondii induces FAK-Src-STAT3 signaling during infection of host cells that prevents parasite targeting by autophagy. PLoS Pathog 2017; 13:e1006671. [PMID: 29036202 PMCID: PMC5658194 DOI: 10.1371/journal.ppat.1006671] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 10/26/2017] [Accepted: 09/26/2017] [Indexed: 02/01/2023] Open
Abstract
Targeting of Toxoplasma gondii by autophagy is an effective mechanism by which host cells kill the protozoan. Thus, the parasite must avoid autophagic targeting to survive. Here we show that the mammalian cytoplasmic molecule Focal Adhesion Kinase (FAK) becomes activated during invasion of host cells. Activated FAK appears to accompany the formation of the moving junction (as assessed by expression the parasite protein RON4). FAK activation was inhibited by approaches that impaired β1 and β3 integrin signaling. FAK caused activation of Src that in turn mediated Epidermal Growth Factor Receptor (EGFR) phosphorylation at the unique Y845 residue. Expression of Src-resistant Y845F EGFR mutant markedly inhibited ROP16-independent activation of STAT3 in host cells. Activation of FAK, Y845 EGFR or STAT3 prevented activation of PKR and eIF2α, key stimulators of autophagy. Genetic or pharmacologic inhibition of FAK, Src, EGFR phosphorylation at Y845, or STAT3 caused accumulation of the autophagy protein LC3 and LAMP-1 around the parasite and parasite killing dependent on autophagy proteins (ULK1 and Beclin 1) and lysosomal enzymes. Parasite killing was inhibited by expression of dominant negative PKR. Thus, T. gondii activates a FAK→Src→Y845-EGFR→STAT3 signaling axis within mammalian cells, thereby enabling the parasite to survive by avoiding autophagic targeting through a mechanism likely dependent on preventing activation of PKR and eIF2α.
Collapse
Affiliation(s)
- Jose-Andres C. Portillo
- Department of Medicine, Division of Infectious Disease and HIV Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
| | - Luis Muniz-Feliciano
- Department of Medicine, Division of Infectious Disease and HIV Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
| | - Yalitza Lopez Corcino
- Department of Medicine, Division of Infectious Disease and HIV Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
| | - So Jung Lee
- Department of Medicine, Division of Infectious Disease and HIV Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
| | - Jennifer Van Grol
- Department of Medicine, Division of Infectious Disease and HIV Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
| | - Sarah J. Parsons
- Department of Microbiology and Cancer Center, University of Virginia, Charlottesville, VA, United States of America
| | - William P. Schiemman
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States of America
| | - Carlos S. Subauste
- Department of Medicine, Division of Infectious Disease and HIV Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, United States of America
- * E-mail:
| |
Collapse
|
93
|
Chakraborty S, Roy S, Mistry HU, Murthy S, George N, Bhandari V, Sharma P. Potential Sabotage of Host Cell Physiology by Apicomplexan Parasites for Their Survival Benefits. Front Immunol 2017; 8:1261. [PMID: 29081773 PMCID: PMC5645534 DOI: 10.3389/fimmu.2017.01261] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/21/2017] [Indexed: 12/26/2022] Open
Abstract
Plasmodium, Toxoplasma, Cryptosporidium, Babesia, and Theileria are the major apicomplexan parasites affecting humans or animals worldwide. These pathogens represent an excellent example of host manipulators who can overturn host signaling pathways for their survival. They infect different types of host cells and take charge of the host machinery to gain nutrients and prevent itself from host attack. The mechanisms by which these pathogens modulate the host signaling pathways are well studied for Plasmodium, Toxoplasma, Cryptosporidium, and Theileria, except for limited studies on Babesia. Theileria is a unique pathogen taking into account the way it modulates host cell transformation, resulting in its clonal expansion. These parasites majorly modulate similar host signaling pathways, however, the disease outcome and effect is different among them. In this review, we discuss the approaches of these apicomplexan to manipulate the host–parasite clearance pathways during infection, invasion, survival, and egress.
Collapse
Affiliation(s)
| | - Sonti Roy
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| | - Hiral Uday Mistry
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| | - Shweta Murthy
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| | - Neena George
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| | | | - Paresh Sharma
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| |
Collapse
|
94
|
Zhou J, Lu G, Wang L, Zhou AH, Han YL, Guo JJ, Song PX, Zhou HY, Cong H, Hou M, He SY. Structuraland antigenic analysis of a new Rhoptry Pseudokinase Gene (ROP54) in Toxoplasma gondii. Acta Parasitol 2017; 62:513-519. [PMID: 28682759 DOI: 10.1515/ap-2017-0061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 03/27/2017] [Indexed: 02/06/2023]
Abstract
Toxoplasma gondii is defined as an obligate intracellular apicomplexan parasite and influences approximatelyone-third of the human all over the world. ROP54 protein is expressed in the rhoptry of Toxoplasma gondii. In the present study, we used SMART software to analyzethe secondary structure of ROP54. The 3D model of ROP54 protein was constructed and analyzed using SWISS-MODEL server and VMD software. The structure results fully showed that ROP54 proteinis an importantmember from the ROP family. Moreover, DNAMAN software and Epitope Database online service were used to analyze liner-B cell epitopes and Th-cell epitopes of the protein. The bioinformatics prediction of ROP54 protein could provide positive information on treatment and vaccine for toxoplasmosis. Furthermore, ROP54 gene was obtained from PCR, and a recombinant eukaryotic expression vector (pEGFP-ROP54) was constructed in the following study. After identification of enzyme digestion, the constructed plasmid was transfected into HEK 293-T cells. The RT-PCR result suggested that the recombinant plasmid could transcribe successfully in HEK 293-T cell.
Collapse
|
95
|
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: 0.6] [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]
|
96
|
Toxoplasma gondii and Neospora caninum induce different host cell responses at proteome-wide phosphorylation events; a step forward for uncovering the biological differences between these closely related parasites. Parasitol Res 2017; 116:2707-2719. [DOI: 10.1007/s00436-017-5579-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 07/31/2017] [Indexed: 10/19/2022]
|
97
|
Ma L, Liu G, Liu J, Li M, Zhang H, Tang D, Liu Q. Neospora caninum ROP16 play an important role in the pathogenicity by phosphorylating host cell STAT3. Vet Parasitol 2017; 243:135-147. [DOI: 10.1016/j.vetpar.2017.04.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 04/10/2017] [Accepted: 04/12/2017] [Indexed: 11/25/2022]
|
98
|
Xue J, Jiang W, Chen Y, Gong F, Wang M, Zeng P, Xia C, Wang Q, Huang K. Thioredoxin reductase from Toxoplasma gondii: an essential virulence effector with antioxidant function. FASEB J 2017; 31:4447-4457. [PMID: 28687608 DOI: 10.1096/fj.201700008r] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 06/12/2017] [Indexed: 12/20/2022]
Abstract
Thioredoxin reductase (TR) can help pathogens resist oxidative-burst injury from host immune cells by maintaining a thioredoxin-reduction state during NADPH consumption. TR is a necessary virulence factor that enables the persistent infection of some parasites. We performed bioinformatics analyses and biochemical assays to characterize the activity, subcellular localization, and genetic ablation of Toxoplasma gondii TR (TgTR), to shed light on its biologic function. We expressed the TgTR protein with an Escherichia coli expression system and analyzed its enzyme activity, reporting a Km for the recombinant TgTR of 11.47-15.57 μM, using NADPH as a substrate, and 130.48-151.09 μM with dithio-bis-nitrobenzoic acid as a substrate. The TgTR sequence shared homology with that of TR, but lacked a selenocysteine residue in the C-terminal region and was thought to contain 2 flavin adenine dinucleotide (FAD) domains and 1 NADPH domain. In addition, immunoelectron microscopy results showed that TgTR was widely dispersed in the cytoplasm, and we observed that parasite antioxidant capacity, invasion efficiency, and proliferation were decreased in TR-knockout (TR-KO) strains in vitro, although this strain still stimulated the release of reactive oxygen species release in mouse macrophages while being more sensitive to H2O2 toxicity in vitro Furthermore, our in vivo results revealed that the survival time of mice infected with the TR-KO strain was significantly prolonged relative to that of mice infected with the wild-type strain. These results suggest that TgTR plays an important role in resistance to oxidative damage and can be considered a virulence factor associated with T. gondii infection.-Xue, J., Jiang, W., Chen, Y., Gong, F., Wang, M., Zeng, P., Xia, C., Wang, Q., Huang, K. Thioredoxin reductase from Toxoplasma gondii: an essential virulence effector with antioxidant function.
Collapse
Affiliation(s)
- Junxin Xue
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China.,College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Shanghai Entry-Exit Inspection and Quarantine Bureau, Shanghai, China
| | - Wei Jiang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Yongjun Chen
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Fengju Gong
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Minyan Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Peng Zeng
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Can Xia
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Quan Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China;
| | - Kehe Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China;
| |
Collapse
|
99
|
Abstract
The development of molecular genetics has greatly enhanced the study of the biology and pathology associated with parasites of the phylum Apicomplexa. We have established a system specifically designed for Neospora caninum, and used this system as a heterologous platform for the expression of foreign genes. Plasmid constructs containing fluorescent proteins or targeted genes of Toxoplasma gondii, driven by N. caninum promoters, have yielded robust expression and correct trafficking of target gene products as assessed by immunofluorescence assays and Western blot analyses. Using this approach, we here demonstrated that N. caninum expressing T. gondii’s GRA15 and ROP16 kinase are biologically active and induced immunological phenotypes consistent with T. gondii strains. N. caninum expressing TgGRA15 differentially disturbed the NF-κB pathway, inducing an increased IL-12 production. On the other hand, N. caninum expressing TgROP16 induced host STAT3 phosphorylation and consequent reduction of IL-12 synthesis. These results indicate that heterologous gene expression in N. caninum is a useful tool for the study of specific gene functions and may allow the identification of antigenic targets responsible for the phenotypic differences observed between these two closely related apicomplexan parasites. Additionally, these observations may prove to be useful for the development of vaccine protocols to control toxoplasmosis and/or neosporosis.
Collapse
|
100
|
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
|