1
|
Fan X, Zhang N, Zhang X, Li S, Fu J, Sun M, Ma X, Khan MZ, Jiang X. Molecular interactions of Toxoplasma gondii dense granule 23 (GRA23) with host proteins PEX3 and TRAP1. Front Vet Sci 2025; 12:1585261. [PMID: 40417354 PMCID: PMC12098569 DOI: 10.3389/fvets.2025.1585261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 04/14/2025] [Indexed: 05/27/2025] Open
Abstract
Background Toxoplasma gondii, an obligate intracellular protozoan, utilizes dense granule proteins to modulate host cell processes. Dense granule protein 23 (GRA23) facilitates molecular trafficking between the parasitophorous vacuole and host cell cyto-plasm, though its specific host protein interactions remain poorly characterized. Methods This study employed pull-down assays coupled with mass spectrometry to identify host proteins interacting with GRA23. Results Among 35 proteins identified, peroxisomal biogenesis factor 3 (PEX3) and TNF receptor-associated protein 1 (TRAP1) were validated through bimolecular fluorescence complementation (BiFC) and co-immunoprecipitation (Co-IP) assays. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses revealed significant enrichment of these interacting proteins in metabolic pathways and cellular processes related to reproduction, growth, and development. Conclusion The interaction between GRA23 and PEX3 suggests potential parasite modulation of peroxisomal functions, while its association with TRAP1 indicates possible exploitation of host chaperone mechanisms. This study provides the first evidence that GRA23 interacts with host proteins implicated in key cellular functions, offering novel insights into T. gondii pathogenesis and potential therapeutic targets for toxoplasmosis treatment.
Collapse
Affiliation(s)
- Xuewei Fan
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
- Heilongjiang Agricultural Economy Vocational College, Mudanjiang, China
| | - Nan Zhang
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Xichen Zhang
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Shudong Li
- Heilongjiang Agricultural Economy Vocational College, Mudanjiang, China
| | - Jiangming Fu
- Heilongjiang Agricultural Economy Vocational College, Mudanjiang, China
| | - Min Sun
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Xiaoxiao Ma
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | | | - Xin Jiang
- Heilongjiang Agricultural Economy Vocational College, Mudanjiang, China
| |
Collapse
|
2
|
Murillo-Léon M, Bastidas-Quintero AM, Steinfeldt T. Decoding Toxoplasma gondii virulence: the mechanisms of IRG protein inactivation. Trends Parasitol 2024; 40:805-819. [PMID: 39168720 DOI: 10.1016/j.pt.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 08/23/2024]
Abstract
Toxoplasmosis is a common parasitic zoonosis that can be life-threatening in immunocompromised patients. About one-third of the human population is infected with Toxoplasma gondii. Primary infection triggers an innate immune response wherein IFN-γ-induced host cell GTPases, namely IRG and GBP proteins, serve as a vital component for host cell resistance. In the past decades, interest in elucidating the function of these GTPase families in controlling various intracellular pathogens has emerged. Numerous T. gondii effectors were identified to inactivate particular IRG proteins. T. gondii is re-optimizing its effectors to combat IRG function and in this way secures transmission. We discuss the IRG-specific effectors employed by the parasite in murine infections, contributing to a better understanding of T. gondii virulence.
Collapse
Affiliation(s)
- Mateo Murillo-Léon
- Institute of Medical Microbiology and Hygiene, Medical Center University of Freiburg, 79104 Freiburg, Germany; CIBSS, Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany; Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Aura María Bastidas-Quintero
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Institute of Virology, Medical Center University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Tobias Steinfeldt
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Institute of Virology, Medical Center University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
3
|
Tang L, Sabi MM, Fu M, Guan J, Wang Y, Xia T, Zheng K, Qu H, Han B. Host cell manipulation by microsporidia secreted effectors: Insights into intracellular pathogenesis. J Eukaryot Microbiol 2024; 71:e13029. [PMID: 39030770 DOI: 10.1111/jeu.13029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/29/2024] [Accepted: 03/29/2024] [Indexed: 07/22/2024]
Abstract
Microsporidia are prolific producers of effector molecules, encompassing both proteins and nonproteinaceous effectors, such as toxins, small RNAs, and small peptides. These secreted effectors play a pivotal role in the pathogenicity of microsporidia, enabling them to subvert the host's innate immunity and co-opt metabolic pathways to fuel their own growth and proliferation. However, the genomes of microsporidia, despite falling within the size range of bacteria, exhibit significant reductions in both structural and physiological features, thereby affecting the repertoire of secretory effectors to varying extents. This review focuses on recent advances in understanding how microsporidia modulate host cells through the secretion of effectors, highlighting current challenges and proposed solutions in deciphering the complexities of microsporidial secretory effectors.
Collapse
Affiliation(s)
- Liyuan Tang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, China
| | - Musa Makongoro Sabi
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, China
| | - Ming Fu
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, China
| | - Jingyu Guan
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, China
| | - Yongliang Wang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, China
| | - Tian Xia
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, China
| | - Kai Zheng
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, China
| | - Hongnan Qu
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, China
- Shenzhen Research Institute, Shandong University, Shenzhen, Guangdong, China
| | - Bing Han
- Department of Pathogenic Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, China
- Shenzhen Research Institute, Shandong University, Shenzhen, Guangdong, China
| |
Collapse
|
4
|
Sun M, Tang T, He K, Long S. TBC9, an essential TBC-domain protein, regulates early vesicular transport and IMC formation in Toxoplasma gondii. Commun Biol 2024; 7:596. [PMID: 38762629 PMCID: PMC11102469 DOI: 10.1038/s42003-024-06310-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 05/08/2024] [Indexed: 05/20/2024] Open
Abstract
Apicomplexan parasites harbor a complex endomembrane system as well as unique secretory organelles. These complex cellular structures require an elaborate vesicle trafficking system, which includes Rab GTPases and their regulators, to assure the biogenesis and secretory of the organelles. Here we exploit the model apicomplexan organism Toxoplasma gondii that encodes a family of Rab GTPase Activating Proteins, TBC (Tre-2/Bub2/Cdc16) domain-containing proteins. Functional profiling of these proteins in tachyzoites reveals that TBC9 is the only essential regulator, which is localized to the endoplasmic reticulum (ER) in T. gondii strains. Detailed analyses demonstrate that TBC9 is required for normal distribution of proteins targeting to the ER, and the Golgi apparatus in the parasite, as well as for the normal formation of daughter inner membrane complexes (IMCs). Pull-down assays show a strong protein interaction between TBC9 and specific Rab GTPases (Rab11A, Rab11B, and Rab2), supporting the role of TBC9 in daughter IMC formation and early vesicular transport. Thus, this study identifies the only essential TBC domain-containing protein TBC9 that regulates early vesicular transport and IMC formation in T. gondii and potentially in closely related protists.
Collapse
Affiliation(s)
- Ming Sun
- National Key Laboratory of Veterinary Public Health Safety and College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Tao Tang
- National Key Laboratory of Veterinary Public Health Safety and College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Kai He
- National Key Laboratory of Veterinary Public Health Safety and College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Shaojun Long
- National Key Laboratory of Veterinary Public Health Safety and College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
5
|
Bando H, Murata Y, Han Y, Sugi T, Fukuda Y, Bzik DJ, Fox BA, Kato K. Toxoplasma gondii chitinase-like protein TgCLP1 regulates the parasite cyst burden. Front Cell Infect Microbiol 2024; 14:1359888. [PMID: 38828265 PMCID: PMC11140023 DOI: 10.3389/fcimb.2024.1359888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/23/2024] [Indexed: 06/05/2024] Open
Abstract
Toxoplasma, an important intracellular parasite of humans and animals, causes life-threatening toxoplasmosis in immunocompromised individuals. Although Toxoplasma secretory proteins during acute infection (tachyzoite, which divides rapidly and causes inflammation) have been extensively characterized, those involved in chronic infection (bradyzoite, which divides slowly and is surrounded by a cyst wall) remain uncertain. Regulation of the cyst wall is essential to the parasite life cycle, and polysaccharides, such as chitin, in the cyst wall are necessary to sustain latent infection. Toxoplasma secretory proteins during the bradyzoite stage may have important roles in regulating the cyst wall via polysaccharides. Here, we focused on characterizing the hypothetical T. gondii chitinase, chitinase-like protein 1 (TgCLP1). We found that the chitinase-like domain containing TgCLP1 is partially present in the bradyzoite microneme and confirmed, albeit partially, its previous identification in the tachyzoite microneme. Furthermore, although parasites lacking TgCLP1 could convert from tachyzoites to bradyzoites and make an intact cyst wall, they failed to convert from bradyzoites to tachyzoites, indicating that TgCLP1 is necessary for bradyzoite reactivation. Taken together, our findings deepen our understanding of the molecular basis of recrudescence and could contribute to the development of novel strategies for the control of toxoplasmosis.
Collapse
Affiliation(s)
- Hironori Bando
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, Osaki, Miyagi, Japan
- Department of Parasitology, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Yuho Murata
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Yongmei Han
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Tatsuki Sugi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
- Division of Collaboration and Education, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Yasuhiro Fukuda
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, Osaki, Miyagi, Japan
| | - David J. Bzik
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Barbara A. Fox
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Kentaro Kato
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, Osaki, Miyagi, Japan
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| |
Collapse
|
6
|
Zhang ZW, Wang M, Sun LX, Elsheikha HM, Lei CL, Wang JL, Fu BQ, Luo JX, Zhu XQ, Li TT. Trx4, a novel thioredoxin protein, is important for Toxoplasma gondii fitness. Parasit Vectors 2024; 17:178. [PMID: 38576040 PMCID: PMC10996207 DOI: 10.1186/s13071-024-06259-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 03/21/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND To successfully replicate within the host cell, Toxoplasma gondii employs several mechanisms to overcome the host cell defenses and mitigate the harmful effects of the free radicals resulting from its own metabolic processes using effectors such as thioredoxin proteins. In this study, we characterize the location and functions of a newly identified thioredoxin in T. gondii, which was named Trx4. METHODS We characterized the functional role of Trx4 in T. gondii Type I RH and Type II Pru strains by gene knockout and studied its subcellular localization by endogenous protein HA tagging using CRISPR-Cas9 gene editing. The enzyme-catalyzed proximity labeling technique, the TurboID system, was employed to identify the proteins in proximity to Trx4. RESULTS Trx4 was identified as a dense granule protein of T. gondii predominantly expressed in the parasitophorous vacuole (PV) and was partially co-localized with GRA1 and GRA5. Functional analysis showed that deletion of trx4 markedly influenced the parasite lytic cycle, resulting in impaired host cell invasion capacity in both RH and Pru strains. Mutation of Trx domains in Trx4 in RH strain revealed that two Trx domains were important for the parasite invasion. By utilizing the TurboID system to biotinylate proteins in proximity to Trx4, we identified a substantial number of proteins, some of which are novel, and others are previously characterized, predominantly distributed in the dense granules. In addition, we uncovered three novel proteins co-localized with Trx4. Intriguingly, deletion of trx4 did not affect the localization of these three proteins. Finally, a virulence assay demonstrated that knockout of trx4 resulted in a significant attenuation of virulence and a significant reduction in brain cyst loads in mice. CONCLUSIONS Trx4 plays an important role in T. gondii invasion and virulence in Type I RH strain and Type II Pru strain. Combining the TurboID system with CRISPR-Cas9 technique revealed many PV-localized proximity proteins associated with Trx4. These findings suggest a versatile role of Trx4 in mediating the processes that occur in this distinctive intracellular membrane-bound vacuolar compartment.
Collapse
Affiliation(s)
- Zhi-Wei Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
| | - Meng Wang
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
| | - Li-Xiu Sun
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
| | - 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
| | - Cheng-Lin Lei
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
| | - Jin-Lei Wang
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, Sichuan Province, 610213, People's Republic of China
| | - Bao-Quan Fu
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, Sichuan Province, 610213, People's Republic of China
| | - Jian-Xun Luo
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
| | - Xing-Quan Zhu
- Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi Province, 030801, People's Republic of China.
| | - Ting-Ting Li
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China.
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, Sichuan Province, 610213, People's Republic of China.
| |
Collapse
|
7
|
Wang Y, Hollingsworth LR, Sangaré LO, Paredes-Santos TC, Krishnamurthy S, Penn BH, Wu H, Saeij JPJ. Host E3 ubiquitin ligase ITCH mediates Toxoplasma gondii effector GRA35-triggered NLRP1 inflammasome activation and cell-autonomous immunity. mBio 2024; 15:e0330223. [PMID: 38376248 PMCID: PMC10936166 DOI: 10.1128/mbio.03302-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/31/2024] [Indexed: 02/21/2024] Open
Abstract
Toxoplasma gondii is an intracellular parasite that can activate the NLRP1 inflammasome leading to macrophage pyroptosis in Lewis rats, but the underlying mechanism is not well understood. In this study, we performed a genome-wide CRISPR screen and identified the dense granule proteins GRA35, GRA42, and GRA43 as the Toxoplasma effectors mediating cell death in Lewis rat macrophages. GRA35 localizes on the parasitophorous vacuole membrane, where it interacts with the host E3 ubiquitin ligase ITCH. Inhibition of proteasome activity or ITCH knockout prevented pyroptosis in Toxoplasma-infected Lewis rat macrophages, consistent with the "NLRP1 functional degradation model." However, there was no evidence that ITCH directly ubiquitinates or interacts with rat NLRP1. We also found that GRA35-ITCH interaction affected Toxoplasma fitness in IFNγ-activated human fibroblasts, likely due to ITCH's role in recruiting ubiquitin and the parasite-restriction factor RNF213 to the parasitophorous vacuole membrane. These findings identify a new role of host E3 ubiquitin ligase ITCH in mediating effector-triggered immunity, a critical concept that involves recognizing intracellular pathogens and initiating host innate immune responses.IMPORTANCEEffector-triggered immunity represents an innate immune defense mechanism that plays a crucial role in sensing and controlling intracellular pathogen infection. The NLRP1 inflammasome in the Lewis rats can detect Toxoplasma infection, which triggers proptosis in infected macrophages and eliminates the parasite's replication niche. The work reported here revealed that host E3 ubiquitin ligase ITCH is able to recognize and interact with Toxoplasma effector protein GRA35 localized on the parasite-host interface, leading to NLRP1 inflammasome activation in Lewis rat macrophages. Furthermore, ITCH-GRA35 interaction contributes to the restriction of Toxoplasma in human fibroblasts stimulated by IFNγ. Thus, this research provides valuable insights into understanding pathogen recognition and restriction mediated by host E3 ubiquitin ligase.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, California, USA
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - L. Robert Hollingsworth
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, USA
| | - Lamba Omar Sangaré
- Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, California, USA
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Tatiana C. Paredes-Santos
- Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, California, USA
| | - Shruthi Krishnamurthy
- Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, California, USA
| | - Bennett H. Penn
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis Health, Sacramento, California, USA
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeroen P. J. Saeij
- Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, California, USA
| |
Collapse
|
8
|
Lüder CGK. IFNs in host defence and parasite immune evasion during Toxoplasma gondii infections. Front Immunol 2024; 15:1356216. [PMID: 38384452 PMCID: PMC10879624 DOI: 10.3389/fimmu.2024.1356216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/23/2024] [Indexed: 02/23/2024] Open
Abstract
Interferons (IFNs) are a family of cytokines with diverse functions in host resistance to pathogens and in immune regulation. Type II IFN, i.e. IFN-γ, is widely recognized as a major mediator of resistance to intracellular pathogens, including the protozoan Toxoplasma gondii. More recently, IFN-α/β, i.e. type I IFNs, and IFN-λ (type III IFN) have been identified to also play important roles during T. gondii infections. This parasite is a widespread pathogen of humans and animals, and it is a model organism to study cell-mediated immune responses to intracellular infection. Its success depends, among other factors, on the ability to counteract the IFN system, both at the level of IFN-mediated gene expression and at the level of IFN-regulated effector molecules. Here, I review recent advances in our understanding of the molecular mechanisms underlying IFN-mediated host resistance and immune regulation during T. gondii infections. I also discuss those mechanisms that T. gondii has evolved to efficiently evade IFN-mediated immunity. Knowledge of these fascinating host-parasite interactions and their underlying signalling machineries is crucial for a deeper understanding of the pathogenesis of toxoplasmosis, and it might also identify potential targets of parasite-directed or host-directed supportive therapies to combat the parasite more effectively.
Collapse
Affiliation(s)
- Carsten G. K. Lüder
- Institute for Medical Microbiology and Virology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
9
|
Zheng XN, Sun LX, Elsheikha HM, Li TT, Gao J, Wu XJ, Zhang ZW, Wang M, Fu BQ, Zhu XQ, Wang JL. A newly characterized dense granule protein (GRA76) is important for the growth and virulence of Toxoplasma gondii. Int J Parasitol 2024; 54:109-121. [PMID: 37832712 DOI: 10.1016/j.ijpara.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/30/2023] [Accepted: 09/03/2023] [Indexed: 10/15/2023]
Abstract
Pathogenicity of the zoonotic pathogen Toxoplasma gondii largely depends on the secretion of effector proteins into the extracellular milieu and host cell cytosol, including the dense granule proteins (GRAs). The protein-encoding gene TGME49_299780 was previously identified as a contributor to parasite fitness. However, its involvement in parasite growth, virulence and infectivity in vitro and in vivo remains unknown. Here, we comprehensively examined the role of this new protein, termed GRA76, in parasite pathogenicity. Subcellular localization revealed high expression of GRA76 in tachyzoites inside the parasitophorous vacuole (PV). However, its expression was significantly decreased in bradyzoites. A CRISPR-Cas9 approach was used to knock out the gra76 gene in the T. gondii type I RH strain and type II Pru strain. The in vitro plaque assays and intracellular replication showed the involvement of GRA76 in replication of RH and Pru strains. Deletion of the gra76 gene significantly decreased parasite virulence, and reduced the brain cyst burden in mice. Using RNA sequencing, we detected a significant increase in the expression of bradyzoite-associated genes such as BAG1 and LDH2 in the PruΔgra76 strain compared with the wild-type Pru strain. Using an in vitro bradyzoite differentiation assay, we showed that loss of GRA76 significantly increased the propensity for parasites to form bradyzoites. Immunization with PruΔgra76 conferred partial protection against acute and chronic infection in mice. These findings show the important role of GRA76 in the pathogenesis of T. gondii and highlight the potential of PruΔgra76 as a candidate for a live-attenuated vaccine.
Collapse
Affiliation(s)
- Xiao-Nan Zheng
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, People's Republic of China; Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi Province 030801, People's Republic of China
| | - Li-Xiu Sun
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, People's Republic of China; Research Center for Parasites & Vectors, College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan Province 410128, 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
| | - Ting-Ting Li
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, People's Republic of China; Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, Sichuan Province 610213, People's Republic of China
| | - Jin Gao
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, People's Republic of China; Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi Province 030801, People's Republic of China
| | - Xiao-Jing Wu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, People's Republic of China; Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi Province 030801, People's Republic of China
| | - Zhi-Wei Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, People's Republic of China
| | - Meng Wang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, People's Republic of China; Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, Sichuan Province 610213, People's Republic of China
| | - Bao-Quan Fu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, People's Republic of China; Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, Sichuan Province 610213, People's Republic of China
| | - Xing-Quan Zhu
- Laboratory of Parasitic Diseases, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi Province 030801, People's Republic of China.
| | - Jin-Lei Wang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, People's Republic of China; Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, Sichuan Province 610213, People's Republic of China.
| |
Collapse
|
10
|
Rinkenberger N, Rosenberg A, Radke JB, Bhushan J, Tomita T, Weiss LM, Sibley LD. Susceptibility of Toxoplasma gondii to autophagy in human cells relies on multiple interacting parasite loci. mBio 2024; 15:e0259523. [PMID: 38095418 PMCID: PMC10790690 DOI: 10.1128/mbio.02595-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/06/2023] [Indexed: 01/04/2024] Open
Abstract
IMPORTANCE Autophagy is a process used by cells to recycle organelles and macromolecules and to eliminate intracellular pathogens. Previous studies have shown that some stains of Toxoplasma gondii are resistant to autophagy-dependent growth restriction, while others are highly susceptible. Although it is known that autophagy-mediated control requires activation by interferon gamma, the basis for why parasite strains differ in their susceptibility is unknown. Our findings indicate that susceptibility involves at least five unlinked parasite genes on different chromosomes, including several secretory proteins targeted to the parasite-containing vacuole and exposed to the host cell cytosol. Our findings reveal that susceptibility to autophagy-mediated growth restriction relies on differential recognition of parasite proteins exposed at the host-pathogen interface, thus identifying a new mechanism for cell-autonomous control of intracellular pathogens.
Collapse
Affiliation(s)
- Nicholas Rinkenberger
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Alex Rosenberg
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Joshua B. Radke
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Jaya Bhushan
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Tadakimi Tomita
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Louis M. Weiss
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - L. David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
11
|
Seizova S, Ferrel A, Boothroyd J, Tonkin CJ. Toxoplasma protein export and effector function. Nat Microbiol 2024; 9:17-28. [PMID: 38172621 DOI: 10.1038/s41564-023-01563-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 11/16/2023] [Indexed: 01/05/2024]
Abstract
Toxoplasma gondii is a single-celled eukaryotic parasite with a considerable host range that must invade the cells of warm-blooded hosts to survive and replicate. The challenges and opportunities that such a strategy represent have been met by the evolution of effectors that are delivered into host cells, counter host defences and co-opt host cell functions for their own purposes. These effectors are delivered in two waves using distinct machinery for each. In this Review, we focus on understanding the architecture of these protein-export systems and how their protein cargo is recognized and selected. We discuss the recent findings on the role that host manipulation has in latent Toxoplasma infections. We also discuss how these recent findings compare to protein export in the related Plasmodium spp. (the causative agent of malaria) and how this can inform our understanding of host manipulation in the larger Apicomplexa phylum and its evolution.
Collapse
Affiliation(s)
- Simona Seizova
- School of Life Sciences, The University of Dundee, Dundee, UK
| | - Abel Ferrel
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - John Boothroyd
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| | - Christopher J Tonkin
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.
| |
Collapse
|
12
|
Okuma H, Saijo-Hamano Y, Yamada H, Sherif AA, Hashizaki E, Sakai N, Kato T, Imasaki T, Kikkawa S, Nitta E, Sasai M, Abe T, Sugihara F, Maniwa Y, Kosako H, Takei K, Standley DM, Yamamoto M, Nitta R. Structural basis of Irgb6 inactivation by Toxoplasma gondii through the phosphorylation of switch I. Genes Cells 2024; 29:17-38. [PMID: 37984375 PMCID: PMC11448365 DOI: 10.1111/gtc.13080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/12/2023] [Accepted: 10/29/2023] [Indexed: 11/22/2023]
Abstract
Irgb6 is a priming immune-related GTPase (IRG) that counteracts Toxoplasma gondii. It is known to be recruited to the low virulent type II T. gondii parasitophorous vacuole (PV), initiating cell-autonomous immunity. However, the molecular mechanism by which immunity-related GTPases become inactivated after the parasite infection remains obscure. Here, we found that Thr95 of Irgb6 is prominently phosphorylated in response to low virulent type II T. gondii infection. We observed that a phosphomimetic T95D mutation in Irgb6 impaired its localization to the PV and exhibited reduced GTPase activity in vitro. Structural analysis unveiled an atypical conformation of nucleotide-free Irgb6-T95D, resulting from a conformational change in the G-domain that allosterically modified the PV membrane-binding interface. In silico docking corroborated the disruption of the physiological membrane binding site. These findings provide novel insights into a T. gondii-induced allosteric inactivation mechanism of Irgb6.
Collapse
Affiliation(s)
- Hiromichi Okuma
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yumiko Saijo-Hamano
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroshi Yamada
- Department of Neuroscience, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Aalaa Alrahman Sherif
- Department of Genome Informatics, Research Institute for Microbial Diseases, Osaka, Japan
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Emi Hashizaki
- Laboratory of Immunoparasitology, Osaka University, Osaka, Japan
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka, Japan
| | | | - Takaaki Kato
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tsuyoshi Imasaki
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Satoshi Kikkawa
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Eriko Nitta
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Miwa Sasai
- Laboratory of Immunoparasitology, Osaka University, Osaka, Japan
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka, Japan
| | - Tadashi Abe
- Department of Neuroscience, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Fuminori Sugihara
- Core Instrumentation Facility, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yoshimasa Maniwa
- Division of Thoracic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Kohji Takei
- Department of Neuroscience, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Daron M Standley
- Department of Genome Informatics, Research Institute for Microbial Diseases, Osaka, Japan
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Masahiro Yamamoto
- Laboratory of Immunoparasitology, Osaka University, Osaka, Japan
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka, Japan
| | - Ryo Nitta
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
13
|
Wang Y, Hollingsworth LR, Sangaré LO, Paredes-Santos TC, Krishnamurthy S, Penn BH, Wu H, Saeij JPJ. Host E3 ubiquitin ligase ITCH mediates Toxoplasma gondii effector GRA35-triggered NLRP1 inflammasome activation and cell-autonomous immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.13.571530. [PMID: 38168400 PMCID: PMC10760081 DOI: 10.1101/2023.12.13.571530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Toxoplasma gondii is an intracellular parasite that can activate the NLRP1 inflammasome leading to macrophage pyroptosis in Lewis rats, but the underlying mechanism is not well understood. In this study, we performed a genome-wide CRISPR screen and identified the dense granule proteins GRA35, GRA42, and GRA43 as the Toxoplasma effectors mediating cell death in Lewis rat macrophages. GRA35 localizes on the parasitophorous vacuole membrane, where it interacts with the host E3 ubiquitin ligase ITCH. Inhibition of proteasome activity or ITCH knockout prevented pyroptosis in Toxoplasma-infected Lewis rat macrophages, consistent with the "NLRP1 functional degradation model". However, there was no evidence that ITCH directly ubiquitinates or interacts with rat NLRP1. We also found that GRA35-ITCH interaction affected Toxoplasma fitness in IFNγ-activated human fibroblasts, likely due to ITCH's role in recruiting ubiquitin and the parasite-restriction factor RNF213 to the parasitophorous vacuole membrane. These findings identify a new role of host E3 ubiquitin ligase ITCH in mediating effector-triggered immunity, a critical concept that involves recognizing intracellular pathogens and initiating host innate immune responses.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, USA
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - L. Robert Hollingsworth
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
| | - Lamba Omar Sangaré
- Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, USA
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Tatiana C. Paredes-Santos
- Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, USA
| | - Shruthi Krishnamurthy
- Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, USA
| | - Bennett H. Penn
- Department of Internal Medicine, Division of Infectious Diseases, UC Davis Health, Sacramento, CA, USA
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, USA
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
| | - Jeroen P. J. Saeij
- Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, USA
| |
Collapse
|
14
|
Wang QQ, Sun M, Tang T, Lai DH, Liu J, Maity S, He K, Wu XT, Yang J, Li YB, Tang XY, Ding HY, Hide G, Distefano M, Lun ZR, Zhu XQ, Long S. Functional screening reveals Toxoplasma prenylated proteins required for endocytic trafficking and rhoptry protein sorting. mBio 2023; 14:e0130923. [PMID: 37548452 PMCID: PMC10470541 DOI: 10.1128/mbio.01309-23] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/12/2023] [Indexed: 08/08/2023] Open
Abstract
In the apicomplexans, endocytosed cargos (e.g., hemoglobin) are trafficked to a specialized organelle for digestion. This follows a unique endocytotic process at the micropore/cytostome in these parasites. However, the mechanism underlying endocytic trafficking remains elusive, due to the repurposing of classical endocytic proteins for the biogenesis of apical organelles. To resolve this issue, we have exploited the genetic tractability of the model apicomplexan Toxoplasma gondii, which ingests host cytosolic materials (e.g., green fluorescent protein[GFP]). We determined an association between protein prenylation and endocytic trafficking, and using an alkyne-labeled click chemistry approach, the prenylated proteome was characterized. Genome editing, using clustered regularly interspaced short palindromic repaet/CRISPR-associated nuclease 9 (CRISPR/Cas9), was efficiently utilized to generate genetically modified lines for the functional screening of 23 prenylated candidates. This identified four of these proteins that regulate the trafficking of endocytosed GFP vesicles. Among these proteins, Rab1B and YKT6.1 are highly conserved but are non-classical endocytic proteins in eukaryotes. Confocal imaging analysis showed that Rab1B and Ras are substantially localized to both the trans-Golgi network and the endosome-like compartments in the parasite. Conditional knockdown of Rab1B caused a rapid defect in secretory trafficking to the rhoptry bulb, suggesting a trafficking intersection role for the key regulator Rab1B. Further experiments confirmed a critical role for protein prenylation in regulating the stability/activity of these proteins (i.e., Rab1B and YKT6.1) in the parasite. Our findings define the molecular basis of endocytic trafficking and reveal a potential intersection function of Rab1B on membrane trafficking in T. gondii. This might extend to other related protists, including the malarial parasites. IMPORTANCE The protozoan Toxoplasma gondii establishes a permissive niche, in host cells, that allows parasites to acquire large molecules such as proteins. Numerous studies have demonstrated that the parasite repurposes the classical endocytic components for secretory sorting to the apical organelles, leaving the question of endocytic transport to the lysosome-like compartment unclear. Recent studies indicated that endocytic trafficking is likely to associate with protein prenylation in malarial parasites. This information promoted us to examine this association in the model apicomplexan T. gondii and to identify the key components of the prenylated proteome that are involved. By exploiting the genetic tractability of T. gondii and a host GFP acquisition assay, we reveal four non-classical endocytic proteins that regulate the transport of endocytosed cargos (e.g., GFP) in T. gondii. Thus, we extend the principle that protein prenylation regulates endocytic trafficking and elucidate the process of non-classical endocytosis in T. gondii and potentially in other related protists.
Collapse
Affiliation(s)
- Qiang-Qiang Wang
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Ming Sun
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Tao Tang
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| | - De-Hua Lai
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jing Liu
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Sanjay Maity
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kai He
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xi-Ting Wu
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiong Yang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yue-Bao Li
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiao-Yan Tang
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Hui-Yong Ding
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Geoff Hide
- Biomedical Research and Innovation Centre and Environmental Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Salford, United Kingdom
| | - Mark Distefano
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota, USA
| | - Zhao-Rong Lun
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xing-Quan Zhu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi Province, China
| | - Shaojun Long
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
15
|
Tachibana Y, Hashizaki E, Sasai M, Yamamoto M. Host genetics highlights IFN-γ-dependent Toxoplasma genes encoding secreted and non-secreted virulence factors in in vivo CRISPR screens. Cell Rep 2023; 42:112592. [PMID: 37269286 DOI: 10.1016/j.celrep.2023.112592] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/25/2023] [Accepted: 05/17/2023] [Indexed: 06/05/2023] Open
Abstract
Secreted virulence factors of Toxoplasma to survive in immune-competent hosts have been extensively explored by classical genetics and in vivo CRISPR screen methods, whereas their requirements in immune-deficient hosts are incompletely understood. Those of non-secreted virulence factors are further enigmatic. Here we develop an in vivo CRISPR screen system to enrich not only secreted but also non-secreted virulence factors in virulent Toxoplasma-infected C57BL/6 mice. Notably, combined usage of immune-deficient Ifngr1-/- mice highlights genes encoding various non-secreted proteins as well as well-known effectors such as ROP5, ROP18, GRA12, and GRA45 as interferon-γ (IFN-γ)-dependent virulence genes. The screen results suggest a role of GRA72 for normal GRA17/GRA23 localization and the IFN-γ-dependent role of UFMylation-related genes. Collectively, our study demonstrates that host genetics can complement in vivo CRISPR screens to highlight genes encoding IFN-γ-dependent secreted and non-secreted virulence factors in Toxoplasma.
Collapse
Affiliation(s)
- Yuta Tachibana
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Emi Hashizaki
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Miwa Sasai
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan; Department of Immunoparasitology, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan; Department of Immunoparasitology, Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
16
|
Giuliano CJ, Wei KJ, Harling FM, Waldman BS, Farringer MA, Boydston EA, Lan TCT, Thomas RW, Herneisen AL, Sanderlin AG, Coppens I, Dvorin JD, Lourido S. Functional profiling of the Toxoplasma genome during acute mouse infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.05.531216. [PMID: 36945434 PMCID: PMC10028831 DOI: 10.1101/2023.03.05.531216] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Within a host, pathogens encounter a diverse and changing landscape of cell types, nutrients, and immune responses. Examining host-pathogen interactions in animal models can therefore reveal aspects of infection absent from cell culture. We use CRISPR-based screens to functionally profile the entire genome of the model apicomplexan parasite Toxoplasma gondii during mouse infection. Barcoded gRNAs were used to track mutant parasite lineages, enabling detection of bottlenecks and mapping of population structures. We uncovered over 300 genes that modulate parasite fitness in mice with previously unknown roles in infection. These candidates span multiple axes of host-parasite interaction, including determinants of tropism, host organelle remodeling, and metabolic rewiring. We mechanistically characterized three novel candidates, including GTP cyclohydrolase I, against which a small-molecule inhibitor could be repurposed as an antiparasitic compound. This compound exhibited antiparasitic activity against T. gondii and Plasmodium falciparum, the most lethal agent of malaria. Taken together, we present the first complete survey of an apicomplexan genome during infection of an animal host, and point to novel interfaces of host-parasite interaction that may offer new avenues for treatment.
Collapse
Affiliation(s)
| | - Kenneth J. Wei
- Whitehead Institute, Cambridge, MA
- Biology Department, MIT, Cambridge, MA
| | - Faye M. Harling
- Whitehead Institute, Cambridge, MA
- Biology Department, MIT, Cambridge, MA
| | | | - Madeline A. Farringer
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, USA
- Biological Sciences in Public Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | | | | | - Raina W. Thomas
- Whitehead Institute, Cambridge, MA
- Biology Department, MIT, Cambridge, MA
| | - Alice L. Herneisen
- Whitehead Institute, Cambridge, MA
- Biology Department, MIT, Cambridge, MA
| | | | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Jeffrey D. Dvorin
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Sebastian Lourido
- Whitehead Institute, Cambridge, MA
- Biology Department, MIT, Cambridge, MA
| |
Collapse
|
17
|
Wan W, Dong H, Lai DH, Yang J, He K, Tang X, Liu Q, Hide G, Zhu XQ, Sibley LD, Lun ZR, Long S. The Toxoplasma micropore mediates endocytosis for selective nutrient salvage from host cell compartments. Nat Commun 2023; 14:977. [PMID: 36813769 PMCID: PMC9947163 DOI: 10.1038/s41467-023-36571-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 02/03/2023] [Indexed: 02/24/2023] Open
Abstract
Apicomplexan parasite growth and replication relies on nutrient acquisition from host cells, in which intracellular multiplication occurs, yet the mechanisms that underlie the nutrient salvage remain elusive. Numerous ultrastructural studies have documented a plasma membrane invagination with a dense neck, termed the micropore, on the surface of intracellular parasites. However, the function of this structure remains unknown. Here we validate the micropore as an essential organelle for endocytosis of nutrients from the host cell cytosol and Golgi in the model apicomplexan Toxoplasma gondii. Detailed analyses demonstrated that Kelch13 is localized at the dense neck of the organelle and functions as a protein hub at the micropore for endocytic uptake. Intriguingly, maximal activity of the micropore requires the ceramide de novo synthesis pathway in the parasite. Thus, this study provides insights into the machinery underlying acquisition of host cell-derived nutrients by apicomplexan parasites that are otherwise sequestered from host cell compartments.
Collapse
Affiliation(s)
- Wenyan Wan
- National Key Laboratory of Veterinary Public Health Security and School of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Hui Dong
- National Key Laboratory of Veterinary Public Health Security and School of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - De-Hua Lai
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Jiong Yang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Kai He
- National Key Laboratory of Veterinary Public Health Security and School of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Xiaoyan Tang
- National Key Laboratory of Veterinary Public Health Security and School of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Qun Liu
- National Key Laboratory of Veterinary Public Health Security and School of Veterinary Medicine, China Agricultural University, 100193, Beijing, China
| | - Geoff Hide
- Biomedical Research and Innovation Centre and Environmental Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Salford, M5 4WT, UK
| | - Xing-Quan Zhu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - L David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, 63110-1093, USA
| | - Zhao-Rong Lun
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Shaojun Long
- National Key Laboratory of Veterinary Public Health Security and School of Veterinary Medicine, China Agricultural University, 100193, Beijing, China.
| |
Collapse
|
18
|
Singh S, Murillo-León M, Endres NS, Arenas Soto AF, Gómez-Marín JE, Melbert F, Kanneganti TD, Yamamoto M, Campos C, Howard JC, Taylor GA, Steinfeldt T. ROP39 is an Irgb10-specific parasite effector that modulates acute Toxoplasma gondii virulence. PLoS Pathog 2023; 19:e1011003. [PMID: 36603017 PMCID: PMC9848475 DOI: 10.1371/journal.ppat.1011003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 01/18/2023] [Accepted: 11/14/2022] [Indexed: 01/06/2023] Open
Abstract
Toxoplasma gondii (T. gondii) is a zoonotic apicomplexan parasite that is an important cause of clinical disability in humans. On a global scale, one third of the human population is infected with T. gondii. Mice and other small rodents are believed to be responsible for transmission of T. gondii to the domestic cat, its definitive host. Interferon-inducible Immunity-Related GTPases (IRG proteins) are important for control of murine T. gondii infections. Virulence differences between T. gondii strains are linked to polymorphic rhoptry proteins (ROPs) that cooperate to inactivate individual IRG family members. In particular, the pseudokinase ROP5 isoform B is critically important in laboratory strains of mice. We identified T. gondii ROP39 in complex with ROP5B and demonstrate its contribution to acute T. gondii virulence. ROP39 directly targets Irgb10 and inhibits homodimer formation of the GTPase leading to an overall reduction of IRG protein loading onto the parasitophorous vacuolar membrane (PVM). Maintenance of PVM integrity rescues the parasite from IRG protein-mediated clearance in vitro and in vivo. This study identifies a novel T. gondii effector that is important for specific inactivation of the IRG resistance system. Our data reveal that yet unknown T. gondii effectors can emerge from identification of direct interaction partners of ROP5B.
Collapse
Affiliation(s)
- Shishir Singh
- Institute of Virology, Medical Center University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Mateo Murillo-León
- Institute of Virology, Medical Center University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Niklas Sebastian Endres
- Institute of Virology, Medical Center University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Ailan Farid Arenas Soto
- Grupo GEPAMOL, Centro de Investigaciones Biomedicas, Universidad del Quindio, Armenia, Quindio, Colombia
| | - Jorge Enrique Gómez-Marín
- Grupo GEPAMOL, Centro de Investigaciones Biomedicas, Universidad del Quindio, Armenia, Quindio, Colombia
| | - Florence Melbert
- Institute of Virology, Medical Center University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Thirumala-Devi Kanneganti
- Department of Immunology, St. Jude Children´s Research Hospital, Memphis, Tenessee, United States of America
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Immunoparasitology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Claudia Campos
- Fundacao Calouste Gulbekian, Instituto Gulbekian de Ciencia, Oeiras, Portugal
| | | | - Gregory Alan 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
| | - Tobias Steinfeldt
- Institute of Virology, Medical Center University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- * E-mail:
| |
Collapse
|
19
|
A. PORTES JULIANA, C. VOMMARO ROSSIANE, AYRES CALDAS LUCIO, S. MARTINS-DUARTE ERICA. Intracellular life of protozoan Toxoplasma gondii: Parasitophorous vacuole establishment and survival strategies. BIOCELL 2023. [DOI: 10.32604/biocell.2023.026629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
20
|
Dogga SK, Lunghi M, Maco B, Li J, Claudi B, Marq JB, Chicherova N, Kockmann T, Bumann D, Hehl AB, Soldati-Favre D, Hammoudi PM. Importance of aspartyl protease 5 in the establishment of the intracellular niche during acute and chronic infection of Toxoplasma gondii. Mol Microbiol 2022; 118:601-622. [PMID: 36210525 DOI: 10.1111/mmi.14987] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 09/21/2022] [Indexed: 11/06/2022]
Abstract
Virulence and persistence of the obligate intracellular parasite Toxoplasma gondii involve the secretion of effector proteins belonging to the family of dense granule proteins (GRAs) that act notably as modulators of the host defense mechanisms and participate in cyst wall formation. The subset of GRAs residing in the parasitophorous vacuole (PV) or exported into the host cell, undergo proteolytic cleavage in the Golgi upon the action of the aspartyl protease 5 (ASP5). In tachyzoites, ASP5 substrates play central roles in the morphology of the PV and the export of effectors across the translocon complex MYR1/2/3. Here, we used N-terminal amine isotopic labeling of substrates to identify novel ASP5 cleavage products by comparing the N-terminome of wild-type and Δasp5 lines in tachyzoites and bradyzoites. Validated substrates reside within the PV or PVM in an ASP5-dependent manner. Remarkably, Δasp5 bradyzoites are impaired in the formation of the cyst wall in vitro and exhibit a considerably reduced cyst burden in chronically infected animals. More specifically two-photon serial tomography of infected mouse brains revealed a comparatively reduced number and size of the cysts throughout the establishment of persistence in the absence of ASP5.
Collapse
Affiliation(s)
- Sunil Kumar Dogga
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Matteo Lunghi
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Bohumil Maco
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Jiagui Li
- Focal Area Infection Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Beatrice Claudi
- Focal Area Infection Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Jean-Baptiste Marq
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Natalia Chicherova
- Focal Area Infection Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Tobias Kockmann
- Functional Genomics Center Zurich, ETH Zurich/University of Zurich, Zurich, Switzerland
| | - Dirk Bumann
- Focal Area Infection Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Adrian B Hehl
- Institute of Parasitology, University of Zurich, Zurich, Switzerland
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Pierre-Mehdi Hammoudi
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
21
|
Griffith MB, Pearce CS, Heaslip AT. Dense granule biogenesis, secretion, and function in Toxoplasma gondii. J Eukaryot Microbiol 2022; 69:e12904. [PMID: 35302693 PMCID: PMC9482668 DOI: 10.1111/jeu.12904] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Toxoplasma gondii is an obligate intracellular parasite and the causative agent of Toxoplasmosis. A key to understanding and treating the disease lies with determining how the parasite can survive and replicate within cells of its host. Proteins released from specialized secretory vesicles, named the dense granules (DGs), have diverse functions that are critical for adapting the intracellular environment, and are thus key to survival and pathogenicity. In this review, we describe the current understanding and outstanding questions regarding dense granule biogenesis, trafficking, and regulation of secretion. In addition, we provide an overview of dense granule protein ("GRA") function upon secretion, with a focus on proteins that have recently been identified.
Collapse
Affiliation(s)
- Michael B Griffith
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
| | - Camille S Pearce
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
| | - Aoife T Heaslip
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
22
|
Guo G, Cui J, Song L, Tang L, Fan S, Shen B, Fang R, Hu M, Zhao J, Zhou Y. Activation of NF-κB signaling by the dense granule protein GRA15 of a newly isolated type 1 Toxoplasma gondii strain. Parasit Vectors 2022; 15:347. [PMID: 36175964 PMCID: PMC9523984 DOI: 10.1186/s13071-022-05429-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/13/2022] [Indexed: 11/17/2022] Open
Abstract
Background It has been reported that the NF-κB pathway, an important component of host defense system against pathogens infections, can be differentially modulated by different Toxoplasma gondii strains, depending on the polymorphism of the GRA15 protein. The recently isolated Toxoplasma strain T.gHB1 is a type 1 (ToxoDB#10) strain but shows different virulence determination mechanisms compared to the classic type 1 strains like RH (ToxoDB#10). Therefore, it is worth investigating whether the T.gHB1 strain (ToxoDB#10) affects the host NF-κB signaling pathway. Methods The effects of T.gHB1 (ToxoDB#10) on host NF-κB pathway were investigated in HEK293T cells. The GRA15 gene product was analyzed by bioinformatics, and its effect on NF-κB activation was examined by Western blotting and nuclear translocation of p65. Different truncations of T.gHB1 GRA15 were constructed to map the critical domains for NF-κB activation. Results We demonstrated that the NF-κB pathway signaling pathway could be activated by the newly identified type 1 T.gHB1 strain (ToxoDB#10) of Toxoplasma, while the classic type 1 strain RH (ToxoDB#10) did not. T.gHB1 GRA15 possesses only one transmembrane region with an extended C terminal region, which is distinct from that of classic type 1 (ToxoDB#10) and type 2 (ToxoDB#1) strains. T.gHB1 GRA15 could clearly induce IκBα phosphorylation and p65 nuclear translocation. Dual luciferase assays in HEK293T cells revealed a requirement for 194–518 aa of T.gHB1 GRA15 to effectively activate NF-κB. Conclusions The overall results indicated that the newly isolated type 1 isolate T.gHB1 (ToxoDB#10) had a unique GRA15, which could activate the host NF-κB signaling through inducing IκBα phosphorylation and p65 nuclear translocation. These results provide new insights for our understanding of the interaction between Toxoplasma parasites and its hosts. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Guanghao Guo
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Jianmin Cui
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Lindong Song
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Lvqing Tang
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Sijie Fan
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Bang Shen
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Rui Fang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Min Hu
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Junlong Zhao
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Yanqin Zhou
- Key Laboratory Preventive Veterinary of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China. .,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China.
| |
Collapse
|
23
|
Rico-San Román L, Amieva R, Regidor-Cerrillo J, García-Sánchez M, Collantes-Fernández E, Pastor-Fernández I, Saeij JPJ, Ortega-Mora LM, Horcajo P. NcGRA7 and NcROP40 Play a Role in the Virulence of Neospora caninum in a Pregnant Mouse Model. Pathogens 2022; 11:pathogens11090998. [PMID: 36145430 PMCID: PMC9506596 DOI: 10.3390/pathogens11090998] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/24/2022] [Accepted: 08/30/2022] [Indexed: 12/01/2022] Open
Abstract
The intraspecific variability among Neospora caninum isolates in their in vitro behaviour and in vivo virulence has been widely studied. In particular, transcriptomic and proteomic analyses have shown a higher expression/abundance of specific genes/proteins in high-virulence isolates. Consequently, the dense granule protein NcGRA7 and the rhoptry protein NcROP40 were proposed as potential virulence factors. The objective of this study was to characterize the role of these proteins using CRISPR/Cas9 knockout (KO) parasites in a well-established pregnant BALB/c mouse model of N. caninum infection at midgestation. The deletion of NcGRA7 and NcROP40 was associated with a reduction of virulence, as infected dams displayed milder clinical signs, lower parasite burdens in the brain, and reduced mortality rates compared to those infected with the wild-type parasite (Nc-Spain7). Specifically, those infected with the NcGRA7 KO parasites displayed significantly milder clinical signs and a lower brain parasite burden. The median survival time of the pups from dams infected with the two KO parasites was significantly increased, but differences in neonatal mortality rates were not detected. Overall, the present study indicates that the disruption of NcGRA7 considerably impairs virulence in mice, while the impact of NcROP40 deletion was more modest. Further research is needed to understand the role of these virulence factors during N. caninum infection.
Collapse
Affiliation(s)
- Laura Rico-San Román
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Rafael Amieva
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Javier Regidor-Cerrillo
- SALUVET-Innova S.L., Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Marta García-Sánchez
- SALUVET-Innova S.L., Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Esther Collantes-Fernández
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Iván Pastor-Fernández
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Jeroen P. J. Saeij
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Luis Miguel Ortega-Mora
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
- Correspondence: (L.M.O.-M.); (P.H.); Tel.: +34-91-3944098 (P.H.)
| | - Pilar Horcajo
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
- Correspondence: (L.M.O.-M.); (P.H.); Tel.: +34-91-3944098 (P.H.)
| |
Collapse
|
24
|
Disruption of Toxoplasma gondii-Induced Host Cell DNA Replication Is Dependent on Contact Inhibition and Host Cell Type. mSphere 2022; 7:e0016022. [PMID: 35587658 PMCID: PMC9241542 DOI: 10.1128/msphere.00160-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The protozoan Toxoplasma gondii is a highly successful obligate intracellular parasite that, upon invasion of its host cell, releases an array of host-modulating protein effectors to counter host defenses and further its own replication and dissemination. Early studies investigating the impact of T. gondii infection on host cell function revealed that this parasite can force normally quiescent cells to activate their cell cycle program. Prior reports by two independent groups identified the dense granule protein effector HCE1/TEEGR as being solely responsible for driving host cell transcriptional changes through its direct interaction with the cyclin E regulatory complex DP1 and associated transcription factors. Our group independently identified HCE1/TEEGR through the presence of distinct repeated regions found in a number of host nuclear targeted parasite effectors and verified its central role in initiating host cell cycle changes. Additionally, we report here the time-resolved kinetics of host cell cycle transition in response to HCE1/TEEGR, using the fluorescence ubiquitination cell cycle indicator reporter line (FUCCI), and reveal the existence of a block in S-phase progression and host DNA synthesis in several cell lines commonly used in the study of T. gondii. Importantly, we have observed that this S-phase block is not due to additional dense granule effectors but rather is dependent on the host cell line background and contact inhibition status of the host monolayer in vitro. This work highlights intriguing differences in the host response to reprogramming by the parasite and raises interesting questions regarding how parasite effectors differentially manipulate the host cell depending on the in vitro or in vivo context. IMPORTANCEToxoplasma gondii chronically infects approximately one-third of the global population and can produce severe pathology in immunologically immature or compromised individuals. During infection, this parasite releases numerous host-targeted effector proteins that can dramatically alter the expression of a variety of host genes. A better understanding of parasite effectors and their host targets has the potential to not only provide ways to control infection but also inform us about our own basic biology. One host pathway that has been known to be altered by T. gondii infection is the cell cycle, and prior reports have identified a parasite effector, known as HCE1/TEEGR, as being responsible. In this report, we further our understanding of the kinetics of cell cycle transition induced by this effector and show that the capacity of HCE1/TEEGR to induce host cell DNA synthesis is dependent on both the cell type and the status of contact inhibition.
Collapse
|
25
|
Gubbels MJ, Ferguson DJP, Saha S, Romano JD, Chavan S, Primo VA, Michaud C, Coppens I, Engelberg K. Toxoplasma gondii's Basal Complex: The Other Apicomplexan Business End Is Multifunctional. Front Cell Infect Microbiol 2022; 12:882166. [PMID: 35573773 PMCID: PMC9103881 DOI: 10.3389/fcimb.2022.882166] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/30/2022] [Indexed: 01/08/2023] Open
Abstract
The Apicomplexa are famously named for their apical complex, a constellation of organelles at their apical end dedicated to invasion of their host cells. In contrast, at the other end of the cell, the basal complex (BC) has been overshadowed since it is much less prominent and specific functions were not immediately obvious. However, in the past decade a staggering array of functions have been associated with the BC and strides have been made in understanding its structure. Here, these collective insights are supplemented with new data to provide an overview of the understanding of the BC in Toxoplasma gondii. The emerging picture is that the BC is a dynamic and multifunctional complex, with a series of (putative) functions. The BC has multiple roles in cell division: it is the site where building blocks are added to the cytoskeleton scaffold; it exerts a two-step stretch and constriction mechanism as contractile ring; and it is key in organelle division. Furthermore, the BC has numerous putative roles in 'import', such as the recycling of mother cell remnants, the acquisition of host-derived vesicles, possibly the uptake of lipids derived from the extracellular medium, and the endocytosis of micronemal proteins. The latter process ties the BC to motility, whereas an additional role in motility is conferred by Myosin C. Furthermore, the BC acts on the assembly and/or function of the intravacuolar network, which may directly or indirectly contribute to the establishment of chronic tissue cysts. Here we provide experimental support for molecules acting in several of these processes and identify several new BC proteins critical to maintaining the cytoplasmic bridge between divided parasites. However, the dispensable nature of many BC components leaves many questions unanswered regarding its function. In conclusion, the BC in T. gondii is a dynamic and multifunctional structure at the posterior end of the parasite.
Collapse
Affiliation(s)
- Marc-Jan Gubbels
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - David J. P. Ferguson
- Nuffield Department of Clinical Laboratory Science, University of Oxford John Radcliffe Hospital, Oxford, United Kingdom
- Department of Biological and Medical Sciences, Faculty of Health and Life Science, Oxford Brookes University, Oxford, United Kingdom
| | - Sudeshna Saha
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Julia D. Romano
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Suyog Chavan
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Vincent A. Primo
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Cynthia Michaud
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Klemens Engelberg
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| |
Collapse
|
26
|
Suwan E, Chalermwong P, Rucksaken R, Sussadee M, Kaewmongkol S, Udonsom R, Jittapalapong S, Mangkit B. Development and evaluation of indirect enzyme-linked immunosorbent assay using recombinant dense granule antigen 7 protein for the detection of Toxoplasma gondii infection in cats in Thailand. Vet World 2022; 15:602-610. [PMID: 35497967 PMCID: PMC9047132 DOI: 10.14202/vetworld.2022.602-610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 02/08/2022] [Indexed: 01/30/2023] Open
Abstract
Background and Aim: Toxoplasma gondii is recognized as a zoonosis causing toxoplasmosis in animals globally. Cat is a definitive host of T. gondii and sheds oocyst through feces, which can infect human beings and animals through contaminated food ingestion. A precise diagnostic test is essential to prevent T. gondii infection in both humans and animals. This study aimed to develop and evaluate the pETite-dense granule antigen 7(GRA7)-based indirect enzyme-linked immunosorbent assay (ELISA) to detect T. gondii infection in cats. Materials and Methods: T. gondii-GRA7 was cloned and expressed in the Expresso®small ubiquitin-related modifier (SUMO) T7 Cloning and Expression System. The recombinant pETite-GRA7 was purified using HisTrap affinity chromatography and confirmed using Western blot analysis. The recombinant protein was used to develop and evaluate the indirect ELISA for T. gondii infection detection. In total, 200 cat sera were tested using pETite-GRA7-based indirect ELISA and indirect fluorescent antibody test (IFAT). The statistical analysis based on Kappa value, sensitivity, specificity, positive predictive value, negative predictive value, χ2 test, and receiver operating characteristic (ROC) curve was used to evaluate the performance of the test. Results: A 606 bp GRA7 polymerase chain reaction (PCR) product was obtained from T. gondii RH strain genomic DNA. The gene was cloned into the pETite™ vector and transformed to HI-Control Escherichia coli BL21 (DE3) for protein expression. Approximately 35 kDa of recombinant pETite-GRA7 was observed and Western blot analysis showed positive bands against anti-6-His antibody and positive-T. gondii cat serum. A sample of 0.5 μg/mL of pETite-GRA7 was subjected to indirect ELISA to detect T. gondii infection in the cat sera. The results showed sensitivity and specificity of pETite-GRA7-based indirect ELISA at 72% and 96%, respectively. An acceptable diagnostic performance was characterized by high concordant results (94%) and substantial agreement (Kappa value=0.65) with IFAT. The seroprevalence levels of ELISA and IFAT were 10% and 9%, respectively, and were not significantly (p>0.05) different. The expected performance of ELISA at different cutoff points using the ROC curve analysis revealed 89% sensitivity and 92% specificity at the cutoff value of 0.146, with a high overall assay accuracy (area under the curve=0.94). Conclusion: In this study, the pETite™ vector, N-terminal 6xHis SUMO fusion tag, was used to improve the solubility and expression level of GRA7. The recombinant pETite-GRA7 showed enhanced protein solubility and purification without special condition requirements. This pETite-GRA7-based indirect ELISA showed high concordant results and substantial agreement with IFAT. ELISA revealed an acceptable sensitivity and specificity. These initial data obtained from cats’ sera demonstrated that pETite-GRA7-based indirect ELISA could be a useful method for local serological diagnosis of T. gondii infection in cats in Thailand.
Collapse
Affiliation(s)
- Eukote Suwan
- Department of Veterinary Technology, Faculty of Veterinary Technology, Kasetsart University, Bangkok, Thailand
| | - Piangjai Chalermwong
- Department of Veterinary Technology, Faculty of Veterinary Technology, Kasetsart University, Bangkok, Thailand
| | - Rucksak Rucksaken
- Department of Veterinary Technology, Faculty of Veterinary Technology, Kasetsart University, Bangkok, Thailand
| | - Metita Sussadee
- Department of Veterinary Technology, Faculty of Veterinary Technology, Kasetsart University, Bangkok, Thailand
| | - Sarawan Kaewmongkol
- Department of Veterinary Technology, Faculty of Veterinary Technology, Kasetsart University, Bangkok, Thailand
| | - Ruenruetai Udonsom
- Department of Protozoology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | - Bandid Mangkit
- Department of Veterinary Technology, Faculty of Veterinary Technology, Kasetsart University, Bangkok, Thailand
| |
Collapse
|
27
|
Abstract
Toxoplasma gondii is a parasitic protist infecting a wide group of warm-blooded animals, ranging from birds to humans. While this infection is usually asymptomatic in healthy individuals, it can also lead to severe ocular or neurological outcomes in immunocompromised individuals or in developing fetuses. This obligate intracellular parasite has the ability to infect a considerable range of nucleated cells and can propagate in the intermediate host. Yet, under the pressure of the immune system it transforms into an encysted persistent form residing primarily in the brain and muscle tissues. Encysted parasites, which are resistant to current medication, may reactivate and give rise to an acute infection. The clinical outcome of toxoplasmosis depends on a complex balance between the host immune response and parasite virulence factors. Susceptibility to the disease is thus determined by both parasite strains and host species. Recent advances on our understanding of host cell-parasite interactions and parasite virulence have brought new insights into the pathophysiology of T. gondii infection and are summarized here.
Collapse
|
28
|
Subekti DT, Ekawasti F, Desem MI, Azmi Z. Toxoplasma gondii virulence prediction using hierarchical cluster analysis based on coding sequences (CDS) of sag1, gra7 and rop18. J Vet Sci 2021; 22:e88. [PMID: 34854270 PMCID: PMC8636655 DOI: 10.4142/jvs.2021.22.e88] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/09/2021] [Accepted: 09/22/2021] [Indexed: 11/20/2022] Open
Abstract
Toxoplasma gondii consists of three genotypes, namely genotype I, II and III. Based on its virulence, T. gondii can be divided into virulent and avirulent strains. This study intends to evaluate an alternative method for predicting T. gondii virulence using hierarchical cluster analysis based on complete coding sequences (CDS) of sag1, gra7 and rop18 genes. Dendrogram was constructed using UPGMA with a Kimura 80 nucleotide distance measurement. The results showed that the prediction errors of T. gondii virulence using sag1, gra7 and rop18 were 7.41%, 6.89% and 9.1%, respectively. Analysis based on CDS of gra7 and rop18 was able to differentiate avirulent strains into genotypes II and III, whereas sag1 failed to differentiate.
Collapse
Affiliation(s)
- Didik T Subekti
- Indonesia Research Center for Veterinary Science, Bogor, West Java 16124, Indonesia.
| | - Fitrine Ekawasti
- Indonesia Research Center for Veterinary Science, Bogor, West Java 16124, Indonesia
| | | | - Zul Azmi
- Goats Research Station, Sei Putih, North Sumatra 20585, Indonesia
| |
Collapse
|
29
|
Fu Y, Brown KM, Jones NG, Moreno SNJ, Sibley LD. Toxoplasma bradyzoites exhibit physiological plasticity of calcium and energy stores controlling motility and egress. eLife 2021; 10:e73011. [PMID: 34860156 PMCID: PMC8683080 DOI: 10.7554/elife.73011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 12/03/2021] [Indexed: 01/01/2023] Open
Abstract
Toxoplasma gondii has evolved different developmental stages for disseminating during acute infection (i.e., tachyzoites) and establishing chronic infection (i.e., bradyzoites). Calcium ion (Ca2+) signaling tightly regulates the lytic cycle of tachyzoites by controlling microneme secretion and motility to drive egress and cell invasion. However, the roles of Ca2+ signaling pathways in bradyzoites remain largely unexplored. Here, we show that Ca2+ responses are highly restricted in bradyzoites and that they fail to egress in response to agonists. Development of dual-reporter parasites revealed dampened Ca2+ responses and minimal microneme secretion by bradyzoites induced in vitro or harvested from infected mice and tested ex vivo. Ratiometric Ca2+ imaging demonstrated lower Ca2+ basal levels, reduced magnitude, and slower Ca2+ kinetics in bradyzoites compared with tachyzoites stimulated with agonists. Diminished responses in bradyzoites were associated with downregulation of Ca2+-ATPases involved in intracellular Ca2+ storage in the endoplasmic reticulum (ER) and acidocalcisomes. Once liberated from cysts by trypsin digestion, bradyzoites incubated in glucose plus Ca2+ rapidly restored their intracellular Ca2+ and ATP stores, leading to enhanced gliding. Collectively, our findings indicate that intracellular bradyzoites exhibit dampened Ca2+ signaling and lower energy levels that restrict egress, and yet upon release they rapidly respond to changes in the environment to regain motility.
Collapse
Affiliation(s)
- Yong Fu
- Department of Molecular Microbiology, Washington University in St. Louis, School of MedicineSt LouisUnited States
| | - Kevin M Brown
- Department of Molecular Microbiology, Washington University in St. Louis, School of MedicineSt LouisUnited States
| | - Nathaniel G Jones
- Department of Molecular Microbiology, Washington University in St. Louis, School of MedicineSt LouisUnited States
| | - Silvia NJ Moreno
- Center for Tropical and Emerging Global Diseases and Department of Cellular Biology, University of GeorgiaAthensUnited States
| | - L David Sibley
- Department of Molecular Microbiology, Washington University in St. Louis, School of MedicineSt LouisUnited States
| |
Collapse
|
30
|
Lodoen MB, Smith NC, Soldati-Favre D, Ferguson DJP, van Dooren GG. Nanos gigantium humeris insidentes: old papers informing new research into Toxoplasma gondii. Int J Parasitol 2021; 51:1193-1212. [PMID: 34736901 PMCID: PMC10538201 DOI: 10.1016/j.ijpara.2021.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/19/2021] [Accepted: 10/19/2021] [Indexed: 11/25/2022]
Abstract
Since Nicolle, Manceaux and Splendore first described Toxoplasma gondii as a parasite of rodents and rabbits in the early 20th century, a diverse and vigorous research community has been built around studying this fascinating intracellular parasite. In addition to its importance as a pathogen of humans, livestock and wildlife, modern researchers are attracted to T. gondii as a facile experimental system to study many aspects of evolutionary biology, cellular biology, host-microbe interactions, and host immunity. For new researchers entering the field, the extensive literature describing the biology of the parasite, and the interactions with its host, can be daunting. In this review, we examine four foundational studies that describe various aspects of T. gondii biology, presenting a 'journal club'-style analysis of each. We have chosen a paper that established the beguiling life cycle of the parasite (Hutchison et al., 1971), a paper that described key features of its cellular biology that the parasite shares with related organisms (Gustafson et al., 1954), a paper that characterised the origin of the unique compartment in which the parasite resides within host cells (Jones and Hirsch, 1972), and a paper that established a key mechanism in the host immune response to parasite infection (Pfefferkorn, 1984). These interesting and far-reaching studies set the stage for subsequent research into numerous facets of parasite biology. As well as providing new researchers with an entry point into the literature surrounding the parasite, revisiting these studies can remind us of the roots of our discipline, how far we have come, and the new directions in which we might head.
Collapse
Affiliation(s)
- Melissa B Lodoen
- Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA
| | - Nicholas C Smith
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; Research School of Biology, Australian National University, Canberra, ACT 2600, Australia
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, CMU, University of Geneva, Geneva, Switzerland
| | - David J P Ferguson
- Nuffield Department of Clinical Laboratory Science, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK; Department of Biological and Medical Sciences, Faculty of Health and Life Science, Oxford Brookes University, Gipsy Lane, Oxford OX3 0BP, UK
| | - Giel G van Dooren
- Research School of Biology, Australian National University, Canberra, ACT 2600, Australia.
| |
Collapse
|
31
|
Wang ZJ, Yu SM, Gao JM, Zhang P, Hide G, Yamamoto M, Lai DH, Lun ZR. High resistance to Toxoplasma gondii infection in inducible nitric oxide synthase knockout rats. iScience 2021; 24:103280. [PMID: 34765911 PMCID: PMC8571494 DOI: 10.1016/j.isci.2021.103280] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/08/2021] [Accepted: 10/13/2021] [Indexed: 11/29/2022] Open
Abstract
Nitric oxide (NO) is an important immune molecule that acts against extracellular and intracellular pathogens in most hosts. However, after the knockout of inducible nitric oxide synthase (iNOS−/−) in Sprague Dawley (SD) rats, these iNOS−/− rats were found to be completely resistant to Toxoplasma gondii infection. Once the iNOS−/− rat peritoneal macrophages (PMs) were infected with T. gondii, they produced high levels of reactive oxygen species (ROS) triggered by GRA43 secreted by T. gondii, which damaged the parasitophorous vacuole membrane and PM mitochondrial membranes within a few hours post-infection. Further evidence indicated that the high levels of ROS caused mitochondrial superoxide dismutase 2 depletion and induced PM pyroptosis and cell death. This discovery of complete resistance to T. gondii infection, in the iNOS−/−-SD rat, demonstrates a strong link between NO and ROS in immunity to T. gondii infection and showcases a potentially novel and effective backup innate immunity system. iNOS−/−-SD rats show strong resistance to Toxoplasma gondii infection iNOS−/−-SD rat PMs resist T. gondii infection through ROS upregulation The T. gondii infection results in PM pyroptosis in iNOS−/−-SD rats GRAs play a key role in the activation of resistance in iNOS−/−-SD rat PMs
Collapse
Affiliation(s)
- Zhen-Jie Wang
- Guangdong Provincial Key Laboratory of Aquatic Economic Animals, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, The People's Republic of China
| | - Shao-Meng Yu
- Guangdong Provincial Key Laboratory of Aquatic Economic Animals, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, The People's Republic of China
| | - Jiang-Mei Gao
- Department of Parasitology, Key Laboratory of Tropical Disease Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, The People's Republic of China
| | - Peng Zhang
- Guangdong Provincial Key Laboratory of Aquatic Economic Animals, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, The People's Republic of China
| | - Geoff Hide
- Biomedical Research Centre, School of Science, Engineering and Environment, University of Salford, Salford M5 4WT, UK
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - De-Hua Lai
- Guangdong Provincial Key Laboratory of Aquatic Economic Animals, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, The People's Republic of China
| | - Zhao-Rong Lun
- Guangdong Provincial Key Laboratory of Aquatic Economic Animals, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, The People's Republic of China.,Department of Parasitology, Key Laboratory of Tropical Disease Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, The People's Republic of China.,Biomedical Research Centre, School of Science, Engineering and Environment, University of Salford, Salford M5 4WT, UK
| |
Collapse
|
32
|
Frickel EM, Hunter CA. Lessons from Toxoplasma: Host responses that mediate parasite control and the microbial effectors that subvert them. J Exp Med 2021; 218:212714. [PMID: 34670268 PMCID: PMC8532566 DOI: 10.1084/jem.20201314] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/03/2021] [Accepted: 09/29/2021] [Indexed: 11/15/2022] Open
Abstract
The intracellular parasite Toxoplasma gondii has long provided a tractable experimental system to investigate how the immune system deals with intracellular infections. This review highlights the advances in defining how this organism was first detected and the studies with T. gondii that contribute to our understanding of how the cytokine IFN-γ promotes control of vacuolar pathogens. In addition, the genetic tractability of this eukaryote organism has provided the foundation for studies into the diverse strategies that pathogens use to evade antimicrobial responses and now provides the opportunity to study the basis for latency. Thus, T. gondii remains a clinically relevant organism whose evolving interactions with the host immune system continue to teach lessons broadly relevant to host–pathogen interactions.
Collapse
Affiliation(s)
- Eva-Maria Frickel
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, UK
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
33
|
Tomita T, Guevara RB, Shah LM, Afrifa AY, Weiss LM. Secreted Effectors Modulating Immune Responses to Toxoplasma gondii. Life (Basel) 2021; 11:988. [PMID: 34575137 PMCID: PMC8467511 DOI: 10.3390/life11090988] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 12/18/2022] Open
Abstract
Toxoplasma gondii is an obligate intracellular parasite that chronically infects a third of humans. It can cause life-threatening encephalitis in immune-compromised individuals. Congenital infection also results in blindness and intellectual disabilities. In the intracellular milieu, parasites encounter various immunological effectors that have been shaped to limit parasite infection. Parasites not only have to suppress these anti-parasitic inflammatory responses but also ensure the host organism's survival until their subsequent transmission. Recent advancements in T. gondii research have revealed a plethora of parasite-secreted proteins that suppress as well as activate immune responses. This mini-review will comprehensively examine each secreted immunomodulatory effector based on the location of their actions. The first section is focused on secreted effectors that localize to the parasitophorous vacuole membrane, the interface between the parasites and the host cytoplasm. Murine hosts are equipped with potent IFNγ-induced immune-related GTPases, and various parasite effectors subvert these to prevent parasite elimination. The second section examines several cytoplasmic and ER effectors, including a recently described function for matrix antigen 1 (MAG1) as a secreted effector. The third section covers the repertoire of nuclear effectors that hijack transcription factors and epigenetic repressors that alter gene expression. The last section focuses on the translocation of dense-granule effectors and effectors in the setting of T. gondii tissue cysts (the bradyzoite parasitophorous vacuole).
Collapse
Affiliation(s)
- Tadakimi Tomita
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (T.T.); (R.B.G.)
| | - Rebekah B. Guevara
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (T.T.); (R.B.G.)
| | - Lamisha M. Shah
- Department of Biological Science, Lehman College of the City University of New York, Bronx, NY 10468, USA; (L.M.S.); (A.Y.A.)
| | - Andrews Y. Afrifa
- Department of Biological Science, Lehman College of the City University of New York, Bronx, NY 10468, USA; (L.M.S.); (A.Y.A.)
| | - Louis M. Weiss
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (T.T.); (R.B.G.)
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
34
|
Rosenberg A, Sibley LD. Toxoplasma gondii secreted effectors co-opt host repressor complexes to inhibit necroptosis. Cell Host Microbe 2021; 29:1186-1198.e8. [PMID: 34043960 PMCID: PMC8711274 DOI: 10.1016/j.chom.2021.04.016] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 02/22/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023]
Abstract
Toxoplasma gondii translocates effector proteins into its host cell to subvert various host pathways. T. gondii effector TgIST blocks the transcription of interferon-stimulated genes to reduce immune defense. Interferons upregulate numerous genes, including protein kinase R (PKR), which induce necrosome formation to activate mixed-lineage-kinase-domain-like (MLKL) pseudokinase and induce necroptosis. Whether these interferon functions are targeted by Toxoplasma is unknown. Here, we examine secreted effectors that localize to the host cell nucleus and find that the chronic bradyzoite stage secretes effector TgNSM that targets the NCoR/SMRT complex, a repressor for various transcription factors, to inhibit interferon-regulated genes involved in cell death. TgNSM acts with TgIST to block IFN-driven expression of PKR and MLKL, thus preventing host cell necroptotic death and protecting the parasite's intracellular niche. The mechanism of action of TgNSM uncovers a role of NCoR/SMRT in necroptosis, assuring survival of intracellular cysts and chronic infection.
Collapse
Affiliation(s)
- Alex Rosenberg
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63130, USA
| | - L David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63130, USA.
| |
Collapse
|
35
|
Ihara F, Nishikawa Y. Toxoplasma gondii manipulates host cell signaling pathways via its secreted effector molecules. Parasitol Int 2021; 83:102368. [PMID: 33905814 DOI: 10.1016/j.parint.2021.102368] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 03/13/2021] [Accepted: 04/07/2021] [Indexed: 01/07/2023]
Abstract
The obligate intracellular parasite Toxoplasma gondii secretes a vast variety of effector molecules from organelles known as rhoptries (ROPs) and dense granules (GRAs). ROP proteins are released into the cytosol of the host cell where they are directed to the cell nucleus or to the parasitophorous vacuole (PV) membrane. ROPs secrete proteins that enable host cell penetration and vacuole formation by the parasites, as well as hijacking host-immune responses. After invading host cells, T. gondii multiplies within a PV that is maintained by the parasite proteins secreted from GRAs. Most GRA proteins remain within the PV, but some are known to access the host cytosol across the PV membrane, and a few are able to traffic into the host-cell nucleus. These effectors bind to host cell proteins and affect host cell signaling pathways to favor the parasite. Studies on host-pathogen interactions have identified many infection-altered host signal transductions. Notably, the relationship between individual parasite effector molecules and the specific targeting of host-signaling pathways is being elucidated through the advent of forward and reverse genetic strategies. Understanding the complex nature of the host-pathogen interactions underlying how the host-signaling pathway is manipulated by parasite effectors may lead to new molecular biological knowledge and novel therapeutic methods for toxoplasmosis. In this review, we discuss how T. gondii modulates cell signaling pathways in the host to favor its survival.
Collapse
Affiliation(s)
- Fumiaki Ihara
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.
| | - Yoshifumi Nishikawa
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan.
| |
Collapse
|
36
|
Dehydroepiandrosterone Effect on Toxoplasma gondii: Molecular Mechanisms Associated to Parasite Death. Microorganisms 2021; 9:microorganisms9030513. [PMID: 33801356 PMCID: PMC8000356 DOI: 10.3390/microorganisms9030513] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 11/21/2022] Open
Abstract
Toxoplasmosis is a zoonotic disease caused by the apicomplexa protozoan parasite Toxoplasma gondii. This disease is a health burden, mainly in pregnant women and immunocompromised individuals. Dehydroepiandrosterone (DHEA) has proved to be an important molecule that could drive resistance against a variety of infections, including intracellular parasites such as Plasmodium falciparum and Trypanozoma cruzi, among others. However, to date, the role of DHEA on T. gondii has not been explored. Here, we demonstrated for the first time the toxoplasmicidal effect of DHEA on extracellular tachyzoites. Ultrastructural analysis of treated parasites showed that DHEA alters the cytoskeleton structures, leading to the loss of the organelle structure and organization as well as the loss of the cellular shape. In vitro treatment with DHEA reduces the viability of extracellular tachyzoites and the passive invasion process. Two-dimensional (2D) SDS-PAGE analysis revealed that in the presence of the hormone, a progesterone receptor membrane component (PGRMC) with a cytochrome b5 family heme/steroid binding domain-containing protein was expressed, while the expression of proteins that are essential for motility and virulence was highly reduced. Finally, in vivo DHEA treatment induced a reduction of parasitic load in male, but not in female mice.
Collapse
|
37
|
Panas MW, Boothroyd JC. Seizing control: How dense granule effector proteins enable Toxoplasma to take charge. Mol Microbiol 2021; 115:466-477. [PMID: 33400323 PMCID: PMC8344355 DOI: 10.1111/mmi.14679] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/02/2021] [Accepted: 01/03/2021] [Indexed: 12/24/2022]
Abstract
Control of the host cell is crucial to the Apicomplexan parasite, Toxoplasma gondii, while it grows intracellularly. To achieve this goal, these single-celled eukaryotes export a series of effector proteins from organelles known as "dense granules" that interfere with normal cellular processes and responses to invasion. While some effectors are found attached to the outer surface of the parasitophorous vacuole (PV) in which Toxoplasma tachyzoites reside, others are found in the host cell's cytoplasm and yet others make their way into the host nucleus, where they alter host transcription. Among the processes that are severely altered are innate immune responses, host cell cycle, and association with host organelles. The ways in which these crucial processes are altered through the coordinated action of a large collection of effectors is as elegant as it is complex, and is the central focus of the following review; we also discuss the recent advances in our understanding of how dense granule effector proteins are trafficked out of the PV.
Collapse
Affiliation(s)
- Michael W. Panas
- Dept. Microbiology and Immunology, Stanford University School of Medicine, Stanford CA 94305
| | - John C. Boothroyd
- Dept. Microbiology and Immunology, Stanford University School of Medicine, Stanford CA 94305
| |
Collapse
|
38
|
Toxoplasma gondii GRA9 Regulates the Activation of NLRP3 Inflammasome to Exert Anti-Septic Effects in Mice. Int J Mol Sci 2020; 21:ijms21228437. [PMID: 33182702 PMCID: PMC7696177 DOI: 10.3390/ijms21228437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 02/06/2023] Open
Abstract
Dense granule proteins (GRAs) are essential components in Toxoplasma gondii, which are suggested to be promising serodiagnostic markers in toxoplasmosis. In this study, we investigated the function of GRA9 in host response and the associated regulatory mechanism, which were unknown. We found that GRA9 interacts with NLR family pyrin domain containing 3 (NLRP3) involved in inflammation by forming the NLRP3 inflammasome. The C-terminal of GRA9 (GRA9C) is essential for GRA9–NLRP3 interaction by disrupting the NLRP3 inflammasome through blocking the binding of apoptotic speck-containing (ASC)-NLRP3. Notably, Q200 of GRA9C is essential for the interaction of NLRP3 and blocking the conjugation of ASC. Recombinant GRA9C (rGRA9C) showed an anti-inflammatory effect and the elimination of bacteria by converting M1 to M2 macrophages. In vivo, rGRA9C increased the anti-inflammatory and bactericidal effects and subsequent anti-septic activity in CLP- and E. coli- or P. aeruginosa-induced sepsis model mice by increasing M2 polarization. Taken together, our findings defined a role of T. gondii GRA9 associated with NLRP3 in host macrophages, suggesting its potential as a new candidate therapeutic agent for sepsis.
Collapse
|
39
|
Nyonda MA, Hammoudi PM, Ye S, Maire J, Marq JB, Yamamoto M, Soldati-Favre D. Toxoplasma gondii GRA60 is an effector protein that modulates host cell autonomous immunity and contributes to virulence. Cell Microbiol 2020; 23:e13278. [PMID: 33040458 DOI: 10.1111/cmi.13278] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 10/04/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023]
Abstract
Toxoplasma gondii infects virtually any nucleated cell and resides inside a non-phagocytic vacuole surrounded by a parasitophorous vacuolar membrane (PVM). Pivotal to the restriction of T. gondii dissemination upon infection in murine cells is the recruitment of immunity regulated GTPases (IRGs) and guanylate binding proteins (GBPs) to the PVM that leads to pathogen elimination. The virulent T. gondii type I RH strain secretes a handful of effectors including the dense granule protein GRA7, the serine-threonine kinases ROP17 and ROP18, and a pseudo-kinase ROP5, that synergistically inhibit the recruitment of IRGs to the PVM. Here, we characterise GRA60, a novel dense granule effector, which localises to the vacuolar space and PVM and contributes to virulence of RH in mice, suggesting a role in the subversion of host cell defence mechanisms. Members of the host cell IRG defence system Irgb10 and Irga6 are recruited to the PVM of RH parasites lacking GRA60 as observed previously for the avirulent RHΔrop5 mutant, with RH preventing such recruitment. Deletion of GRA60 in RHΔrop5 leads to a recruitment of IRGs comparable to the single knockouts. GRA60 therefore represents a novel parasite effector conferring resistance to IRGs in type I parasites, and found associated to ROP18, a member of the virulence complex.
Collapse
Affiliation(s)
- Mary Akinyi Nyonda
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Pierre-Mehdi Hammoudi
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Shu Ye
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Jessica Maire
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Jean-Baptiste Marq
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Division of Infectious Diseases, Osaka University, Suita, Japan
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
40
|
Wang Y, Sangaré LO, Paredes-Santos TC, Hassan MA, Krishnamurthy S, Furuta AM, Markus BM, Lourido S, Saeij JPJ. Genome-wide screens identify Toxoplasma gondii determinants of parasite fitness in IFNγ-activated murine macrophages. Nat Commun 2020; 11:5258. [PMID: 33067458 PMCID: PMC7567896 DOI: 10.1038/s41467-020-18991-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 09/22/2020] [Indexed: 12/12/2022] Open
Abstract
Macrophages play an essential role in the early immune response against Toxoplasma and are the cell type preferentially infected by the parasite in vivo. Interferon gamma (IFNγ) elicits a variety of anti-Toxoplasma activities in macrophages. Using a genome-wide CRISPR screen we identify 353 Toxoplasma genes that determine parasite fitness in naїve or IFNγ-activated murine macrophages, seven of which are further confirmed. We show that one of these genes encodes dense granule protein GRA45, which has a chaperone-like domain, is critical for correct localization of GRAs into the PVM and secretion of GRA effectors into the host cytoplasm. Parasites lacking GRA45 are more susceptible to IFNγ-mediated growth inhibition and have reduced virulence in mice. Together, we identify and characterize an important chaperone-like GRA in Toxoplasma and provide a resource for the community to further explore the function of Toxoplasma genes that determine fitness in IFNγ-activated macrophages.
Collapse
Affiliation(s)
- Yifan Wang
- grid.27860.3b0000 0004 1936 9684Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
| | - Lamba Omar Sangaré
- grid.27860.3b0000 0004 1936 9684Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
| | - Tatiana C. Paredes-Santos
- grid.27860.3b0000 0004 1936 9684Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
| | - Musa A. Hassan
- grid.4305.20000 0004 1936 7988College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK ,grid.4305.20000 0004 1936 7988The Roslin Institute, The University of Edinburgh, Edinburgh, UK ,grid.4305.20000 0004 1936 7988Center for Tropical Livestock Health and Genetics, The University of Edinburgh, Edinburgh, UK
| | - Shruthi Krishnamurthy
- grid.27860.3b0000 0004 1936 9684Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
| | - Anna M. Furuta
- grid.27860.3b0000 0004 1936 9684Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
| | - Benedikt M. Markus
- grid.270301.70000 0001 2292 6283Whitehead Institute for Biomedical Research, Cambridge, MA USA ,grid.5963.9Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Sebastian Lourido
- grid.270301.70000 0001 2292 6283Whitehead Institute for Biomedical Research, Cambridge, MA USA ,grid.116068.80000 0001 2341 2786Department of Biology, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Jeroen P. J. Saeij
- grid.27860.3b0000 0004 1936 9684Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
| |
Collapse
|
41
|
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: 0.6] [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
|
42
|
Tartarelli I, Tinari A, Possenti A, Cherchi S, Falchi M, Dubey JP, Spano F. During host cell traversal and cell-to-cell passage, Toxoplasma gondii sporozoites inhabit the parasitophorous vacuole and posteriorly release dense granule protein-associated membranous trails. Int J Parasitol 2020; 50:1099-1115. [PMID: 32882286 DOI: 10.1016/j.ijpara.2020.06.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/21/2020] [Accepted: 06/23/2020] [Indexed: 10/23/2022]
Abstract
Toxoplasma gondii has a worldwide distribution and infects virtually all warm-blooded animals, including humans. Ingestion of the environmentally resistant oocyst stage, excreted only in the feces of cats, is central to transmission of this apicomplexan parasite. There is vast literature on the host and T. gondii tachyzoite (proliferative stage of the parasite) but little is known of the host-parasite interaction and conversion of the free-living stage (sporozoite inside the oocyst) to the parasitic stage. Here, we present events that follow invasion of host cells with T. gondii sporozoites by using immunofluorescence (IF) and transmission electron microscopy (TEM). Several human type cell cultures were infected with T. gondii sporozoites of the two genotypes (Type II, ME49 and Type III, VEG) most prevalent worldwide. For the first known time, using anti-rhoptry neck protein 4 (RON4) antibodies, the moving junction was visualized in sporozoites during the invasion process and shortly after its completion. Surprisingly, IF and TEM evaluation revealed that intracellular sporozoites release, at their posterior end, long membranous tails, herein named sporozoite-specific trails (SSTs). Differential permeabilization and IF experiments showed that the SSTs are associated with several dense granule proteins (GRAs) and that their membranous component is of parasite origin. Furthermore, TEM observations demonstrated that SST-associated sporozoites are delimited by a typical parasitophorous vacuole, which is retained during parasite exit from the host cell and during cell-to-cell passage. Our data strongly suggest that host cell traversal by T. gondii sporozoites relies on a novel force-producing mechanism, based on the massive extrusion at the parasite posterior pole of GRA-associated membranous material derived from the same pool of membranes forming the intravacuolar network.
Collapse
Affiliation(s)
- Irene Tartarelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Antonella Tinari
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Alessia Possenti
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Simona Cherchi
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Mario Falchi
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Jitender P Dubey
- U.S. Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, Maryland 20705, United States
| | - Furio Spano
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| |
Collapse
|
43
|
Ihara F, Fereig RM, Himori Y, Kameyama K, Umeda K, Tanaka S, Ikeda R, Yamamoto M, Nishikawa Y. Toxoplasma gondii Dense Granule Proteins 7, 14, and 15 Are Involved in Modification and Control of the Immune Response Mediated via NF-κB Pathway. Front Immunol 2020; 11:1709. [PMID: 32849602 PMCID: PMC7412995 DOI: 10.3389/fimmu.2020.01709] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/26/2020] [Indexed: 11/13/2022] Open
Abstract
Toxoplasma gondii infects almost all warm-blooded animals, including humans, leading to both cellular and humoral immune responses in the host. The virulence of T. gondii is strain specific and is defined by secreted effector proteins that disturb host immunity. Here, we focus on nuclear factor-kappa B (NFκB) signaling, which regulates the induction of T-helper type 1 immunity. A luciferase assay for screening effector proteins, including ROPs and GRAs that have biological activity against an NFκB-dependent reporter plasmid, found that overexpression of GRA7, 14, and 15 of a type II strain resulted in a strong activity. Thus, our study was aimed at understanding the involvement of NFκB in the pathogenesis of toxoplasmosis through a comparative analysis of these three molecules. We found that GRA7 and GRA14 were partially involved in the activation of NFκB, whereas GRA15 was essential for NFκB activation. The deletion of GRA7, GRA14, and GRA15 in the type II Prugniaud (Pru) strain resulted in a defect in the nuclear translocation of RelA. Cells infected with the PruΔgra15 parasite showed reduced phosphorylation of inhibitor-κBα. GRA7, GRA14, and GRA15 deficiency decreased the levels of interleukin-6 in RAW246.7 cells, and RNA-seq analysis revealed that GRA7, GRA14, and GRA15 deficiency predominantly resulted in downregulation of gene expression mediated by NFκB. The virulence of all mutant strains increased, but PruΔgra14 only showed a slight increase in virulence. However, the intra-footpad injection of the highly-virulent type I RHΔgra14 parasites in mice resulted in increased virulence. This study shows that GRA7, 14, and 15-induced host immunity via NFκB limits parasite expansion.
Collapse
Affiliation(s)
- Fumiaki Ihara
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Ragab M Fereig
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan.,Department of Animal Medicine, Faculty of Veterinary Medicine, South Valley University, Qena City, Egypt
| | - Yuu Himori
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Kyohko Kameyama
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Kosuke Umeda
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Sachi Tanaka
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan.,Division of Animal Science, Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Nagano, Japan
| | - Rina Ikeda
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Yoshifumi Nishikawa
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| |
Collapse
|
44
|
Li TT, Wang JL, Liang QL, Sun LX, Zhang HS, Zhang ZW, Zhu XQ, Elsheikha HM. Effect of deletion of gra17 and gra23 genes on the growth, virulence, and immunogenicity of type II Toxoplasma gondii. Parasitol Res 2020; 119:2907-2916. [PMID: 32686022 DOI: 10.1007/s00436-020-06815-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 07/09/2020] [Indexed: 12/20/2022]
Abstract
The protozoan parasite Toxoplasma gondii secretes a number of dense granule proteins (GRAs) from the dense granule organelle to manipulate the host cell. Two of these effector proteins (GRA17 and GRA23) are involved in the trafficking of molecules between the parasitophorous vacuole (PV) and the host cell cytoplasm. However, their roles in establishing chronic infection remain obscured. In this study, CRISPR-Cas9 was used to delete gra17 or gra23 gene in T. gondii Pru strain (type II). The growth, the virulence, the ability to establish chronic infection, and the immunogenicity of the constructed mutant strains were investigated in Kunming mice. Pru:Δgra17 and Pru:Δgra23 mutants developed PVs with abnormal morphology and exhibited reduced growth rate, compared with the wild-type Pru strain. Deletion of gra17 abrogated acute infection and blocked cyst formation. Although the deletion of gra23 caused slight attenuation of the parasite virulence in mice, it caused a significant reduction in cyst formation. Immunization with Pru:Δgra17 induced high levels of IgG (IgG1 and IgG2a) antibodies and cytokines (interleukin-2 [IL-2], IL-10, IL-12, and interferon gamma [IFN-γ]), which conferred significant protection in mice challenged with virulent type I (RH), ToxoDB#9 (PYS) strains, or less virulent type II (Pru) strain of T. gondii. These findings show that GRA17 and GRA23 play important roles in T. gondii chronic infection and that irreversible deletion of gra17 in T. gondii type II Pru strain can be a viable option for stimulating protective immunity to T. gondii infection.
Collapse
Affiliation(s)
- 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, 730046, Gansu Province, People's Republic of China
| | - 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, 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
| | - Li-Xiu Sun
- 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
| | - Hai-Sheng 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, 730046, Gansu Province, People's Republic of China
| | - Zhi-Wei 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, 730046, Gansu 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
| | - 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.
| |
Collapse
|
45
|
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: 28] [Impact Index Per Article: 5.6] [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
|
46
|
Rico-San Román L, Horcajo P, Regidor-Cerrillo J, Fernández-Escobar M, Collantes-Fernández E, Gutiérrez-Blázquez D, Hernáez-Sánchez ML, Saeij JPJ, Ortega-Mora LM. Comparative tachyzoite proteome analyses among six Neospora caninum isolates with different virulence. Int J Parasitol 2020; 50:377-388. [PMID: 32360428 DOI: 10.1016/j.ijpara.2020.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/30/2020] [Accepted: 02/14/2020] [Indexed: 02/06/2023]
Abstract
The biological variability among Neospora caninum isolates has been widely shown, however, the molecular basis that determines this diversity has not been thoroughly elucidated to date. The latest studies have focused on a limited number of isolates. Therefore, the goal of the present study was to compare the proteome of a larger number of N. caninum isolates with different origins and virulence. Label-free LC-MS/MS was used to investigate the tachyzoite proteomic differences among Nc-Bahia, Nc-Spain4H and Nc-Spain7, representing high virulence isolates and Nc-Ger6, Nc-Spain2H and Nc-Spain1H, representing low virulence isolates. Pairwise comparisons between all isolates and between high virulence and low virulence groups identified a subset of proteins with higher abundance in high virulence isolates. These proteins were involved in energy and redox metabolism, and DNA/RNA processing, which might determine the faster growth rates and parasite survival of the high virulence isolates. Highlighted proteins included a predicted member of the rhoptry kinase family ROP20 specific for N. caninum, Bradyzoite pseudokinase 1 and several dense granule proteins. DNA polymerase, which was more abundant in all high virulence isolates in all comparisons, might also be implicated in virulence. These results reveal insights into possible mechanisms involved in specific phenotypic traits and virulence in N. caninum, and the relevance of these candidate proteins for N. caninum virulence deserves further investigation.
Collapse
Affiliation(s)
- Laura Rico-San Román
- SALUVET, Animal Health Department, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Pilar Horcajo
- SALUVET, Animal Health Department, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Javier Regidor-Cerrillo
- SALUVET, Animal Health Department, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain; SALUVET-Innova S.L., Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Mercedes Fernández-Escobar
- SALUVET, Animal Health Department, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Esther Collantes-Fernández
- SALUVET, Animal Health Department, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Dolores Gutiérrez-Blázquez
- Proteomics Unit of Complutense University of Madrid, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - María Luisa Hernáez-Sánchez
- Proteomics Unit of Complutense University of Madrid, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Jeroen P J Saeij
- Department of Pathology, Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA
| | - Luis Miguel Ortega-Mora
- SALUVET, Animal Health Department, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain.
| |
Collapse
|
47
|
Mukhopadhyay D, Sangaré LO, Braun L, Hakimi MA, Saeij JP. Toxoplasma GRA15 limits parasite growth in IFNγ-activated fibroblasts through TRAF ubiquitin ligases. EMBO J 2020; 39:e103758. [PMID: 32293748 DOI: 10.15252/embj.2019103758] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 01/01/2023] Open
Abstract
The protozoan parasite Toxoplasma gondii lives inside a vacuole in the host cytosol where it is protected from host cytoplasmic innate immune responses. However, IFNγ-dependent cell-autonomous immunity can destroy the vacuole and the parasite inside. Toxoplasma strain differences in susceptibility to human IFNγ exist, but the Toxoplasma effector(s) that determine these differences are unknown. We show that in human primary fibroblasts, the polymorphic Toxoplasma-secreted effector GRA15 mediates the recruitment of ubiquitin ligases, including TRAF2 and TRAF6, to the vacuole membrane, which enhances recruitment of ubiquitin receptors (p62/NDP52) and ubiquitin-like molecules (LC3B, GABARAP). This ultimately leads to lysosomal degradation of the vacuole. In murine fibroblasts, GRA15-mediated TRAF6 recruitment mediates the recruitment of immunity-related GTPases and destruction of the vacuole. Thus, we have identified how the Toxoplasma effector GRA15 affects cell-autonomous immunity in human and murine cells.
Collapse
Affiliation(s)
- Debanjan Mukhopadhyay
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Lamba Omar Sangaré
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Laurence Braun
- Institute for Advanced Biosciences, Team Host-Pathogen Interactions and Immunity to Infection, INSERM U1209, CNRS, UMR5309, Université Grenoble Alpes, Grenoble, France
| | - Mohamed-Ali Hakimi
- Institute for Advanced Biosciences, Team Host-Pathogen Interactions and Immunity to Infection, INSERM U1209, CNRS, UMR5309, Université Grenoble Alpes, Grenoble, France
| | - Jeroen Pj Saeij
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| |
Collapse
|
48
|
Wang JL, Bai MJ, Elsheikha HM, Liang QL, Li TT, Cao XZ, Zhu XQ. Novel roles of dense granule protein 12 (GRA12) in Toxoplasma gondii infection. FASEB J 2020; 34:3165-3178. [PMID: 31908049 DOI: 10.1096/fj.201901416rr] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 01/09/2023]
Abstract
Dense granule protein 12 (GRA12) is implicated in a range of processes related to the establishment of Toxoplasma gondii infection, such as the formation of the intravacuolar network (IVN) within the parasitophorous vacuole (PV). This protein is also thought to be important for T. gondii-host interaction, pathogenesis, and immune evasion, but their exact roles remain unknown. In this study, the contributions of GRA12 to the molecular pathogenesis of T. gondii infection were examined in vitro and in vivo. Deletion of GRA12 in type I RH and type II Pru T. gondii strains did not affect the parasite growth and replication in vitro, however, it caused a significant reduction in the parasite virulence and tissue cyst burden in vivo. T. gondii Δgra12 mutants were more vulnerable to be eliminated by host immunity, without the accumulation of immunity-related GTPase a6 (Irga6) onto the PV membrane. The ultrastructure of IVN in Δgra12 mutants appeared normal, suggesting that GRA12 is not required for biogenesis of the IVN. Combined deletion of GRA12 and ROP18 induced more severe attenuation of virulence compared to single Δgra12 or Δrop18 mutant strains. These data suggest a functional association between GRA12 and ROP18 that is revealed by the severe attenuation of virulence in a double mutant relative to the single individual mutations. Future studies are needed to define the molecular basis of this putative association. Collectively these findings indicate that although GRA12 is not essential for the parasite growth and replication in vitro, it contributes to the virulence and growth of T. gondii in mice.
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, P.R. China
| | - Meng-Jie Bai
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, P.R. China
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - 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, P.R. 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, P.R. China
| | - Xue-Zhen Cao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, P.R. 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, P.R. China
| |
Collapse
|
49
|
Rommereim LM, Fox BA, Butler KL, Cantillana V, Taylor GA, Bzik DJ. Rhoptry and Dense Granule Secreted Effectors Regulate CD8 + T Cell Recognition of Toxoplasma gondii Infected Host Cells. Front Immunol 2019; 10:2104. [PMID: 31555296 PMCID: PMC6742963 DOI: 10.3389/fimmu.2019.02104] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/21/2019] [Indexed: 12/21/2022] Open
Abstract
Toxoplasma gondii secretes rhoptry (ROP) and dense granule (GRA) effector proteins to evade host immune clearance mediated by interferon gamma (IFN-γ), immunity-related GTPase (IRG) effectors, and CD8+ T cells. Here, we investigated the role of parasite-secreted effectors in regulating host access to parasitophorous vacuole (PV) localized parasite antigens and their presentation to CD8+ T cells by the major histocompatibility class I (MHC-I) pathway. Antigen presentation of PV localized parasite antigens by MHC-I was significantly increased in macrophages and/or dendritic cells infected with mutant parasites that lacked expression of secreted GRA (GRA2, GRA3, GRA4, GRA5, GRA7, GRA12) or ROP (ROP5, ROP18) effectors. The ability of various secreted GRA or ROP effectors to suppress antigen presentation by MHC-I was dependent on cell type, expression of IFN-γ, or host IRG effectors. The suppression of antigen presentation by ROP5, ROP18, and GRA7 correlated with a role for these molecules in preventing PV disruption by IFN-γ-activated host IRG effectors. However, GRA2 mediated suppression of antigen presentation was not correlated with PV disruption. In addition, the GRA2 antigen presentation phenotypes were strictly co-dependent on the expression of the GRA6 protein. These results show that MHC-I antigen presentation of PV localized parasite antigens was controlled by mechanisms that were dependent or independent of IRG effector mediated PV disruption. Our findings suggest that the GRA6 protein underpins an important mechanism that enhances CD8+ T cell recognition of parasite-infected cells with damaged or ruptured PV membranes. However, in intact PVs, parasite secreted effector proteins that associate with the PV membrane or the intravacuolar network membranes play important roles to actively suppress antigen presentation by MHC-I to reduce CD8+ T cell recognition and clearance of Toxoplasma gondii infected host cells.
Collapse
Affiliation(s)
- Leah M Rommereim
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Barbara A Fox
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Kiah L Butler
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Viviana Cantillana
- Division of Geriatrics, Departments of Medicine, Molecular Genetics and Microbiology, and Immunology, Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, NC, United States
| | - Gregory A Taylor
- Division of Geriatrics, Departments of Medicine, Molecular Genetics and Microbiology, and Immunology, Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, NC, United States.,Geriatric Research, Education and Clinical Center, VA Medical Center, Durham, NC, United States
| | - David J Bzik
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| |
Collapse
|
50
|
Toxoplasma gondii effector TgIST blocks type I interferon signaling to promote infection. Proc Natl Acad Sci U S A 2019; 116:17480-17491. [PMID: 31413201 DOI: 10.1073/pnas.1904637116] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
In contrast to the importance of type II interferon-γ (IFN-γ) in control of toxoplasmosis, the role of type I IFN is less clear. We demonstrate here that TgIST, a secreted effector previously implicated in blocking type II IFN-γ signaling, also blocked IFN-β responses by inhibiting STAT1/STAT2-mediated transcription in infected cells. Consistent with a role for type I IFN in cell intrinsic control, ∆Tgist mutants were more susceptible to growth inhibition by murine and human macrophages activated with IFN-β. Additionally, type I IFN was important for production of IFN-γ by natural killer (NK) cells and recruitment of inflammatory monocytes at the site of infection. Mice lacking type I IFN receptors (Ifnar1-/-) showed increased mortality following infection with wild-type parasites and decreased virulence of ∆Tgist parasites was restored in Ifnar1-/- mice. The findings highlight the importance of type I IFN in control of toxoplasmosis and illuminate a parasite mechanism to counteract the effects of both type I and II IFN-mediated host defenses.
Collapse
|