1
|
Orchanian SB, Zolog S, Tomasello J, Malik R, Shin JH, Koshy A, Lodoen MB. Toxoplasma gondii parasites induce a localized myeloid cell immune response surrounding parasites in the brain during acute infection. mBio 2025:e0081025. [PMID: 40492741 DOI: 10.1128/mbio.00810-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 04/20/2025] [Indexed: 06/12/2025] Open
Abstract
Toxoplasma gondii is a foodborne intracellular parasite that chronically infects the brain. Although T. gondii infection elicits a protective immune response, the nature of the monocyte response in the immediate vicinity of parasites during initial brain infection is not well understood. By infecting mice with T. gondii and comparing areas of the brain containing or not containing parasites, we found an increase in CCR2+ monocytes, IBA1+ myeloid cells, GFAP+ astrocytes, and CD68+ signal near parasites in the brain, indicating immune cell infiltration and phagolysosomal activation in response to the infection. CCR2+CD68+ monocytes were specifically increased near tachyzoites with minimal localization of these cells near cysts in the brain. This monocyte response was also detected near parasite-interacted cells, identified using T. gondii parasites that inject Cre recombinase into "interacted" cells of Ai6 CCR2RFP/+ mice, enabling us to track these events in vivo. The chemokine CCL2 and its transcription factor NF-κB were also upregulated surrounding parasites in the brain; however, the T. gondii effector protein GRA15, which sustains NF-κB activation in infected cells, was not required for CCL2 production, NF-κB activation, or myeloid cell recruitment to parasites in the brain. In contrast, active T. gondii replication played a more significant role, as CCR2+ monocytes were recruited to replication-competent but not replication-deficient T. gondii delivered via intracranial injection into mice. These findings provide novel insights into the drivers of immune cell mobilization and activation in the brain during initial T. gondii central nervous system infection and highlight the importance of parasite replication in this process. IMPORTANCE Toxoplasma gondii is a brain-infecting parasite, and the mobilization of peripheral immune cells to the brain is critical for controlling T. gondii infection. However, the initial events driving these cells to sites of T. gondii infection in the brain are poorly understood. We show that peripheral myeloid immune cell recruitment and activation are specific to areas of the brain containing actively replicating parasites, which are capable of lysing host cells. This local immune response is characterized by focal chemokine production, myeloid cell recruitment, and the activation of phagolysosomal pathways. These highly localized host responses were independent of the parasite effector protein that induces NF-κB activation within infected cells, GRA15. However, the localized monocyte recruitment was dependent on live parasites and active parasite replication. This research highlights the importance of host cell sensing of parasite replication in the brain for immune control of T. gondii infection.
Collapse
Affiliation(s)
- Stephanie B Orchanian
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
- Institute for Immunology, University of California, Irvine, California, USA
| | - Sebastian Zolog
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
- Institute for Immunology, University of California, Irvine, California, USA
| | - Julia Tomasello
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
- Institute for Immunology, University of California, Irvine, California, USA
| | - Rijul Malik
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
- Institute for Immunology, University of California, Irvine, California, USA
| | - Ji-Hun Shin
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
- Institute for Immunology, University of California, Irvine, California, USA
| | - Anita Koshy
- Neuroscience Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona, USA
- Department of Immunobiology, University of Arizona, Tucson, Arizona, USA
- Bio5 Institute, University of Arizona, Tucson, Arizona, USA
- Department of Neurology, University of Arizona, Tucson, Arizona, USA
| | - Melissa B Lodoen
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
- Institute for Immunology, University of California, Irvine, California, USA
| |
Collapse
|
2
|
Fu Y, Tomita T, Weiss LM, West CM, Sibley LD. Toxoplasma chitinase-like protein orchestrates cyst wall glycosylation to facilitate effector export and cyst turnover. Proc Natl Acad Sci U S A 2025; 122:e2416870122. [PMID: 39879244 PMCID: PMC11804682 DOI: 10.1073/pnas.2416870122] [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: 08/19/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
Toxoplasma bradyzoites reside in tissue cysts that undergo cycles of expansion, rupture, and release to foster chronic infection. The glycosylated cyst wall acts as a protective barrier, although the processes responsible for formation, remodeling, and turnover are not understood. Herein, we identify a noncanonical chitinase-like enzyme TgCLP1 that localizes to micronemes and is targeted to the cyst wall after secretion. Genetic deletion of TgCLP1 resulted in a thickened cyst wall that decreased cyst turnover, blocked the export of virulence effectors into host cells, and resulted in failure to persist during chronic infection. Genetic complementation with a series of mutants revealed that the GH19 glycosidase domain was crucial for regulating glycosylation of several glycoproteins in the cyst wall. Overall, our findings reveal that TgCLP1 is a multifunctional survival factor that modifies glycoproteins within the cyst wall to modulate export of virulence effectors and regulate turnover of tissue cysts.
Collapse
Affiliation(s)
- Yong Fu
- Department of Molecular Microbiology, Washington University in St. Louis, School of Medicine, St. Louis, MO63130
| | - Tadakimi Tomita
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY10461
| | - Louis M. Weiss
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY10461
- Department of Medicine (Division of Infectious Diseases), Albert Einstein College of Medicine, Bronx, NY10461
| | - Christopher M. West
- Department of Biochemistry and Molecular Biology, Center for Tropical and Global Diseases, University of Georgia, Athens, GA30602
| | - L. David Sibley
- Department of Molecular Microbiology, Washington University in St. Louis, School of Medicine, St. Louis, MO63130
| |
Collapse
|
3
|
Ahmed T. Lipid nanoparticle mediated small interfering RNA delivery as a potential therapy for Alzheimer's disease. Eur J Neurosci 2024; 59:2915-2954. [PMID: 38622050 DOI: 10.1111/ejn.16336] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/21/2024] [Accepted: 03/14/2024] [Indexed: 04/17/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition that exhibits a gradual decline in cognitive function and is prevalent among a significant number of individuals globally. The use of small interfering RNA (siRNA) molecules in RNA interference (RNAi) presents a promising therapeutic strategy for AD. Lipid nanoparticles (LNPs) have been developed as a delivery vehicle for siRNA, which can selectively suppress target genes, by enhancing cellular uptake and safeguarding siRNA from degradation. Numerous research studies have exhibited the effectiveness of LNP-mediated siRNA delivery in reducing amyloid beta (Aβ) levels and enhancing cognitive function in animal models of AD. The feasibility of employing LNP-mediated siRNA delivery as a therapeutic approach for AD is emphasized by the encouraging outcomes reported in clinical studies for other medical conditions. The use of LNP-mediated siRNA delivery has emerged as a promising strategy to slow down or even reverse the progression of AD by targeting the synthesis of tau phosphorylation and other genes linked to the condition. Improvement of the delivery mechanism and determination of the most suitable siRNA targets are crucial for the efficacious management of AD. This review focuses on the delivery of siRNA through LNPs as a promising therapeutic strategy for AD, based on the available literature.
Collapse
Affiliation(s)
- Tanvir Ahmed
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| |
Collapse
|
4
|
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
|
5
|
Bergersen KV, Ramirez AD, Kavvathas B, Mercer F, Wilson EH. Human neutrophil-like cells demonstrate antimicrobial responses to the chronic cyst form of Toxoplasma gondii. Parasite Immunol 2023; 45:e13011. [PMID: 37776091 PMCID: PMC11246559 DOI: 10.1111/pim.13011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 10/01/2023]
Abstract
The protozoan parasite Toxoplasma gondii infects approximately 2.5 billion people worldwide. Infection induces a rapid dissemination of parasites throughout the body followed by the formation of lifelong cysts within neurons of the host brain. Both stages require a dynamic immune response comprised of both innate and adaptive cells. Neutrophils are a primary responding cell to acute infection and have been observed in the brain during murine chronic infection. Previous studies investigating human neutrophils found that invasion by Toxoplasma tachyzoites inhibits apoptosis of neutrophils, prolonging their survival under inflammatory conditions. Here, we demonstrate the differentiation of two distinct subsets following exposure of human neutrophil-like-cells (HNLC) to Toxoplasma cysts. In vitro stimulation and imaging studies show cyst-specific induction of cytokines and cyst clearance by HNLCs. Further testing demonstrates that aged HNLCs perform less phagocytosis of cysts compared to non-aged HNLCs. In conclusion, this study identifies a novel response of HNLCs to Toxoplasma cysts and may indicate a role for neutrophils in the clearance of cysts during human infection with Toxoplasma.
Collapse
Affiliation(s)
- Kristina V. Bergersen
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States of America
| | - Ashley D. Ramirez
- Department of Biological Sciences, California State Polytechnic University, Pomona
| | - Bill Kavvathas
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States of America
| | - Frances Mercer
- Department of Biological Sciences, California State Polytechnic University, Pomona
| | - Emma H. Wilson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States of America
| |
Collapse
|
6
|
Dupont D, Robert M, Brenier-Pinchart M, Lefevre A, Wallon M, Pelloux H. Toxoplasma gondii, a plea for a thorough investigation of its oncogenic potential. Heliyon 2023; 9:e22147. [PMID: 38034818 PMCID: PMC10685377 DOI: 10.1016/j.heliyon.2023.e22147] [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: 06/19/2023] [Revised: 10/20/2023] [Accepted: 11/05/2023] [Indexed: 12/02/2023] Open
Abstract
It is estimated that 30 % of the world's population harbours the parasite Toxoplasma gondii, particularly in the brain. Beyond its implication in potentially severe opportunistic or congenital infections, this persistence has long been considered as without consequence. However, certain data in animals and humans suggest that this carriage may be linked to various neuropsychiatric or neurodegenerative disorders. The hypothesis of a potential cerebral oncogenicity of the parasite is also emerging. In this personal view, we will present the epidemiological arguments in favour of an association between toxoplasmosis and cerebral malignancy, before considering the points that could underlie a potential causal link. More specifically, we will focus on the brain as the preferred location for T. gondii persistence and the propensity of this parasite to interfere with the apoptosis and cell cycle signalling pathways of their host cell.
Collapse
Affiliation(s)
- D. Dupont
- Institut des Agents Infectieux, Parasitologie Mycologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, 69004, France
- Physiologie intégrée du système d’éveil, Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR 5292, Faculté de Médecine, Université Claude Bernard Lyon 1, Bron, 69500, France
| | - M.G. Robert
- Service de Parasitologie-Mycologie, CHU Grenoble Alpes, Grenoble, 38000, France
- Université Grenoble Alpes, Institut pour l'Avancée des Biosciences (IAB), INSERM U1209-CNRS UMR 5309, Grenoble, 38000, France
| | - M.P. Brenier-Pinchart
- Service de Parasitologie-Mycologie, CHU Grenoble Alpes, Grenoble, 38000, France
- Université Grenoble Alpes, Institut pour l'Avancée des Biosciences (IAB), INSERM U1209-CNRS UMR 5309, Grenoble, 38000, France
| | - A. Lefevre
- Institut des Agents Infectieux, Parasitologie Mycologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, 69004, France
| | - M. Wallon
- Institut des Agents Infectieux, Parasitologie Mycologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, 69004, France
- Physiologie intégrée du système d’éveil, Centre de Recherche en Neurosciences de Lyon, INSERM U1028-CNRS UMR 5292, Faculté de Médecine, Université Claude Bernard Lyon 1, Bron, 69500, France
| | - H. Pelloux
- Service de Parasitologie-Mycologie, CHU Grenoble Alpes, Grenoble, 38000, France
- Université Grenoble Alpes, Institut pour l'Avancée des Biosciences (IAB), INSERM U1209-CNRS UMR 5309, Grenoble, 38000, France
| |
Collapse
|
7
|
Sánchez-Arcila JC, Jensen KDC. Forward Genetics in Apicomplexa Biology: The Host Side of the Story. Front Cell Infect Microbiol 2022; 12:878475. [PMID: 35646724 PMCID: PMC9133346 DOI: 10.3389/fcimb.2022.878475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Forward genetic approaches have been widely used in parasitology and have proven their power to reveal the complexities of host-parasite interactions in an unbiased fashion. Many aspects of the parasite's biology, including the identification of virulence factors, replication determinants, antibiotic resistance genes, and other factors required for parasitic life, have been discovered using such strategies. Forward genetic approaches have also been employed to understand host resistance mechanisms to parasitic infection. Here, we will introduce and review all forward genetic approaches that have been used to identify host factors involved with Apicomplexa infections, which include classical genetic screens and QTL mapping, GWAS, ENU mutagenesis, overexpression, RNAi and CRISPR-Cas9 library screens. Collectively, these screens have improved our understanding of host resistance mechanisms, immune regulation, vaccine and drug designs for Apicomplexa parasites. We will also discuss how recent advances in molecular genetics give present opportunities to further explore host-parasite relationships.
Collapse
Affiliation(s)
- Juan C. Sánchez-Arcila
- Department of Molecular and Cell Biology, University of California Merced, Merced, CA, United States
| | - Kirk D. C. Jensen
- Department of Molecular and Cell Biology, University of California Merced, Merced, CA, United States
- Health Science Research Institute, University of California, Merced, Merced, CA, United States
| |
Collapse
|
8
|
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
|
9
|
Khan T, Khan S, Akhtar M, Ali J, Najmi AK. Empagliflozin nanoparticles attenuates type2 diabetes induced cognitive impairment via oxidative stress and inflammatory pathway in high fructose diet induced hyperglycemic mice. Neurochem Int 2021; 150:105158. [PMID: 34391818 DOI: 10.1016/j.neuint.2021.105158] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 01/21/2023]
Abstract
There is snowballing evidence that type 2 diabetes (T2D) predisposes to neuropathophysiological alterations including oxidative stress and triggered inflammatory responses in brain that eventually culminates into cognitive impairment.Accumulating evidences suggest that SGLT2 inhibitor can be a promising intervention for cognitive decline in T2DM. In the present paper, the potential effects of Empagliflozin (EMPA), a SGLT2 inhibitor, against T2D induced cognitive dysfunction have been explored. The effect of EMPA on array of inflammatory mediators including Interleukin-6(IL-6), Interleukin -1β (IL-1β), and Tumour necrosis factor-α(TNF-α)), neuronal proteins including glycogen synthase kinase-3β (GSK- 3β), Phosphorylated tau (p-tau), amyloid beta (Aβ) (1-40, 1-42) and altered oxidative parameters including SOD, catalase, TBARS was determined in the high fructose diet induced hyperglycaemic mice. The obtained results were compared with EMPA nanoparticles (Nps) formulated in our laboratory and found that EMPA Nps significantly showed reduced levels of inflammatory mediators and oxidative stress. Further, decrease in levels of p-tau, Aβ (1-40) and Aβ (1-42) were also observed with EMPA nanoparticles.Thus, the study has demonstrated that EMPA Nps could be a promising therapy to alleviate the progression of cognitive decline in T2D.
Collapse
Affiliation(s)
- Tahira Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia, Hamdard, New Delhi, India
| | - Sana Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia, Hamdard, New Delhi, India
| | - Mohd Akhtar
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia, Hamdard, New Delhi, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia, Hamdard, New Delhi, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia, Hamdard, New Delhi, India.
| |
Collapse
|
10
|
Insights into the biochemical features and immunogenic epitopes of common bradyzoite markers of the ubiquitous Toxoplasma gondii. INFECTION GENETICS AND EVOLUTION 2021; 95:105037. [PMID: 34390868 DOI: 10.1016/j.meegid.2021.105037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/16/2021] [Accepted: 08/09/2021] [Indexed: 12/17/2022]
Abstract
The widespread distribution of Toxoplasma gondii (T. gondii) infection and its harsh outcomes in pregnant women and immunocompromised patients lead researchers towards vaccination strategies. The present in silico investigation was done to reveal biophysical properties and immunogenic epitopes of six bradyzoite markers for rational vaccine design in future. For this purpose, different web servers were used to predict antigenicity, allergenicity, solubility, physicochemical properties, post-translational modification sites (PTMs), the presence of signal peptide and transmembrane domains. Moreover, the secondary and tertiary structures of the proteins were revealed followed by refinement and validation. Finally, NetCTL server was used to predict cytotoxic T-lymphocyte (CTL) epitopes, with subsequent immunogenicity analysis. Also, IEDB server was utilized to predict helper T-lymphocyte (HTL) epitopes, followed by IFN-γ and IL-4 induction, antigenicity and population coverage analysis. As well, several linear antigenic B-cell epitopes were found, with good water solubility and without allergenicity. Totally, these proteins showed appropriate antigenicity, abundant PTMs as well as many CTL, HTL and B-cell epitopes, which could be directed for future vaccination studies in the context of multi-epitope vaccine design.
Collapse
|
11
|
The Toxoplasma Polymorphic Effector GRA15 Mediates Seizure Induction by Modulating Interleukin-1 Signaling in the Brain. mBio 2021; 12:e0133121. [PMID: 34154412 PMCID: PMC8262954 DOI: 10.1128/mbio.01331-21] [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] [Indexed: 11/20/2022] Open
Abstract
Toxoplasmic encephalitis can develop in individuals infected with the protozoan parasite Toxoplasma gondii and is typified by parasite replication and inflammation within the brain. Patients often present with seizures, but the parasite genes and host pathways involved in seizure development and/or propagation are unknown. We previously reported that seizure induction in Toxoplasma-infected mice is parasite strain dependent. Using quantitative trait locus mapping, we identify four loci in the Toxoplasma genome that potentially correlate with seizure development. In one locus, we identify the polymorphic virulence factor, GRA15, as a Toxoplasma gene associated with onset of seizures. GRA15 was previously shown to regulate host NF-κB-dependent gene expression during acute infections, and we demonstrate a similar role for GRA15 in brains of toxoplasmic encephalitic mice. GRA15 is important for increased expression of interleukin 1 beta (IL-1β) and other IL-1 pathway host genes, which is significant since IL-1 signaling is involved in onset of seizures. Inhibiting IL-1 receptor signaling reduced seizure severity in Toxoplasma-infected mice. These data reveal one mechanism by which seizures are induced during toxoplasmic encephalitis.
Collapse
|
12
|
Lori A, Avramopoulos D, Wang AW, Mulle J, Massa N, Duncan EJ, Powers A, Conneely K, Gillespie CF, Jovanovic T, Ressler KJ, Pearce BD. Polygenic risk scores differentiate schizophrenia patients with toxoplasma gondii compared to toxoplasma seronegative patients. Compr Psychiatry 2021; 107:152236. [PMID: 33721583 DOI: 10.1016/j.comppsych.2021.152236] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 01/28/2021] [Accepted: 02/12/2021] [Indexed: 11/28/2022] Open
Abstract
Schizophrenia (SCZ) is an etiologically heterogeneous disease with genetic and environmental risk factors (e.g., Toxoplasma gondii infection) differing among affected individuals. Distinguishing such risk factors may point to differences in pathophysiological pathways and facilitate the discovery of individualized treatments. Toxoplasma gondii (TOXO) has been implicated in increasing the risk of schizophrenia. To determine whether TOXO-positive individuals with SCZ have a different polygenic risk burden than uninfected people, we applied the SCZ polygenic risk score (SCZ-PRS) derived from the Psychiatric GWAS Consortium separately to the TOXO-positive and TOXO-negative subjects with the diagnosis of SCZ as the outcome variable. The SCZ-PRS does not include variants in the major histocompatibility complex. Of 790 subjects assessed for TOXO, the 662 TOXO-negative subjects (50.8% with SCZ) reached a Bonferroni corrected significant association (p = 0.00017, R2 = 0.023). In contrast, the 128 TOXO-positive individuals (53.1% with SCZ) showed no significant association (p = 0.354) for SCZ-PRS and had a much lower R2 (R2 = 0.007). To account for Type-2 error in the TOXO-positive dataset, we performed a random sampling of the TOXO-negative subpopulation (n = 130, repeated 100 times) to simulate equivalent power between groups: the p-value was <0.05 for SCZ-PRS 55% of the time but was rarely (6% of the time) comparable to the high p-value of the seropositive group at p > 0.354. We found intriguing evidence that the SCZ-PRS predicts SCZ in TOXO-negative subjects, as expected, but not in the TOXO-positive individuals. This result highlights the importance of considering environmental risk factors to distinguish a subgroup with independent or different genetic components involved in the development of SCZ.
Collapse
Affiliation(s)
- Adriana Lori
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, 101 Woodruff Circle, GA 30322, USA
| | | | - Alex W Wang
- Dept. of Epidemiology, Rollins School of Public Health; 1518 Clifton Rd., Atlanta, GA 30322, USA; Philadelphia College of Osteopathic Medicine, Suwanee, GA 30024, USA
| | - Jennifer Mulle
- Department of Human Genetics, Emory University, Atlanta, GA 30322, USA
| | - Nicholas Massa
- Dept. of Epidemiology, Rollins School of Public Health; 1518 Clifton Rd., Atlanta, GA 30322, USA; Atlanta Veterans Affairs Health Care System, Decatur, GA 30033, USA
| | - Erica J Duncan
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, 101 Woodruff Circle, GA 30322, USA; Atlanta Veterans Affairs Health Care System, Decatur, GA 30033, USA
| | - Abigail Powers
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, 101 Woodruff Circle, GA 30322, USA
| | - Karen Conneely
- Department of Human Genetics, Emory University, Atlanta, GA 30322, USA
| | - Charles F Gillespie
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, 101 Woodruff Circle, GA 30322, USA
| | - Tanja Jovanovic
- University of Texas at Austin Dell Medical School, Austin, TX, USA
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - Brad D Pearce
- Dept. of Epidemiology, Rollins School of Public Health; 1518 Clifton Rd., Atlanta, GA 30322, USA.
| |
Collapse
|
13
|
Elsheikha HM, Marra CM, Zhu XQ. Epidemiology, Pathophysiology, Diagnosis, and Management of Cerebral Toxoplasmosis. Clin Microbiol Rev 2021; 34:e00115-19. [PMID: 33239310 PMCID: PMC7690944 DOI: 10.1128/cmr.00115-19] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Toxoplasma gondii is known to infect a considerable number of mammalian and avian species and a substantial proportion of the world's human population. The parasite has an impressive ability to disseminate within the host's body and employs various tactics to overcome the highly regulatory blood-brain barrier and reside in the brain. In healthy individuals, T. gondii infection is largely tolerated without any obvious ill effects. However, primary infection in immunosuppressed patients can result in acute cerebral or systemic disease, and reactivation of latent tissue cysts can lead to a deadly outcome. It is imperative that treatment of life-threatening toxoplasmic encephalitis is timely and effective. Several therapeutic and prophylactic regimens have been used in clinical practice. Current approaches can control infection caused by the invasive and highly proliferative tachyzoites but cannot eliminate the dormant tissue cysts. Adverse events and other limitations are associated with the standard pyrimethamine-based therapy, and effective vaccines are unavailable. In this review, the epidemiology, economic impact, pathophysiology, diagnosis, and management of cerebral toxoplasmosis are discussed, and critical areas for future research are highlighted.
Collapse
Affiliation(s)
- Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Christina M Marra
- Departments of Neurology and Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, People's Republic of China
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi Province, People's Republic of China
| |
Collapse
|
14
|
Deshmukh AS, Gurupwar R, Mitra P, Aswale K, Shinde S, Chaudhari S. Toxoplasma gondii induces robust humoral immune response against cyst wall antigens in chronically infected animals and humans. Microb Pathog 2020; 152:104643. [PMID: 33232762 DOI: 10.1016/j.micpath.2020.104643] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 02/01/2023]
Abstract
Toxoplasma gondii differentiation from proliferating tachyzoites into latent bradyzoites is central to pathogenesis and transmission. Strong humoral immune response has been reported against tachyzoite antigens, however, antibody-mediated response towards bradyzoite antigens is poorly characterized. This work aimed to study the humoral immune response towards bradyzoite and associated cyst wall antigens particularly CST1. The immunoreactivity of 404 goats, 88 sheep and 92 human sera to recombinant (CST1 and SRS9) and native proteins of encysted bradyzoite along with well-established tachyzoite antigens (SAG1 and GRA7) was determined using ELISA, Western blot and immunofluorescence analysis (IFA). ELISA results revealed nearly 50% of sera contain T. gondii specific antibodies. Results were further validated using Western blot and IFA. T. gondii positive sera predominantly recognized the cyst wall besides the known tachyzoite surface antigens. The presence of CST1 antibodies in seropositive samples were in line with the staining patterns which were consistent with CST localization. Notably, T. gondii IgM- IgG+ sera recognize the cyst wall whereas IgM + IgG-sera recognize tachyzoite antigens indicating acute infection consistent with presence of parasite DNA. The study demonstrates a strong humoral response against bradyzoite associated cyst wall antigens across naturally infected animals and humans. CST1 emerged as a key immunomodulatory antigen which may have direct implications for clinical immunodiagnostics.
Collapse
Affiliation(s)
- Abhijit S Deshmukh
- Molecular Parasitology Laboratory, DBT- National Institute of Animal Biotechnology, Hyderabad, India.
| | - Rajkumar Gurupwar
- Molecular Parasitology Laboratory, DBT- National Institute of Animal Biotechnology, Hyderabad, India
| | - Pallabi Mitra
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Kalyani Aswale
- Molecular Parasitology Laboratory, DBT- National Institute of Animal Biotechnology, Hyderabad, India
| | - Shilpshri Shinde
- Department of Veterinary Public Health, College of Veterinary and Animal Sciences, Nagpur, India
| | - Sandeep Chaudhari
- Department of Veterinary Public Health, College of Veterinary and Animal Sciences, Nagpur, India
| |
Collapse
|
15
|
Gareh A, Soliman M, Saleh AA, El-Gohary FA, El-Sherbiny HMM, Mohamed RH, Elmahallawy EK. Epidemiological and Histopathological Investigation of Sarcocystis spp. in Slaughtered Dromedary Camels ( Camelus dromedarius) in Egypt. Vet Sci 2020; 7:vetsci7040162. [PMID: 33120870 PMCID: PMC7711966 DOI: 10.3390/vetsci7040162] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 11/16/2022] Open
Abstract
Sarcocystosis is considered one of the major parasitic diseases with a worldwide distribution. It is caused by the obligatory intracellular protozoan parasites of the genus Sarcocystis. Besides its public health issues, sarcocystosis results in significant economic losses due to its impact on productivity and milk yield. A wide range of final and intermediate hosts have been identified, including mammals, birds, and reptiles; however, few studies have investigated the contribution of camels to maintaining the epidemiological foci of the disease in countries such as Egypt. The present study was conducted to grossly and histopathologically identify the prevalence rate of Sarcocystis spp. in camels (N = 100) from the Aswan Governorate, Egypt. Furthermore, the major risk factors related to the development of sarcocystosis in camels were investigated. Samples from the diaphragm, cardiac muscle, esophagus, and testes of the slaughtered camels were collected. Interestingly, Sarcocystis was detected in 75% of the examined camels. Following the studied variable factors, camels aged 5 years or more were found to be at higher risk, with an infection rate of 87.7% (57 of 65) than those younger than 5 years. The infection rate was 81.4% (57 of 70) in males and 60% (18 of 30) in females. The esophagus was the most affected organ (49%), followed by the diaphragm (26%) and cardiac muscle (17%), whereas none of the testes samples were affected. Taken together, the present study demonstrates the high prevalence of Sarcocystis in the examined camels and suggests the importance of these animals in preserving the epidemiological foci of sarcocystosis in Egypt. Future research should map the circulating strains in Egypt and aim to raise public health awareness about the importance of sarcocystosis and other related zoonotic diseases.
Collapse
Affiliation(s)
- Ahmed Gareh
- Department of Parasitology, Faculty of Veterinary Medicine, Aswan University, Aswan 24101, Egypt;
| | - Mahmoud Soliman
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut 71515, Egypt;
| | - Amira A. Saleh
- Department of Medical Parasitology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt;
| | - Fatma A. El-Gohary
- Department of Hygiene and Zoonoses, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Heba M. M. El-Sherbiny
- Educational Veterinary Hospital, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Ragab H. Mohamed
- Department of Theriogenology, Faculty of Veterinary Medicine, Aswan University, Aswan 24101, Egypt;
| | - Ehab Kotb Elmahallawy
- Department of Biomedical Sciences, University of León (ULE), 24071 León, Spain
- Department of Zoonoses, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt
- Correspondence:
| |
Collapse
|
16
|
Mukhopadhyay D, Arranz-Solís D, Saeij JPJ. Influence of the Host and Parasite Strain on the Immune Response During Toxoplasma Infection. Front Cell Infect Microbiol 2020; 10:580425. [PMID: 33178630 PMCID: PMC7593385 DOI: 10.3389/fcimb.2020.580425] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/11/2020] [Indexed: 01/02/2023] Open
Abstract
Toxoplasma gondii is an exceptionally successful parasite that infects a very broad host range, including humans, across the globe. The outcome of infection differs remarkably between hosts, ranging from acute death to sterile infection. These differential disease patterns are strongly influenced by both host- and parasite-specific genetic factors. In this review, we discuss how the clinical outcome of toxoplasmosis varies between hosts and the role of different immune genes and parasite virulence factors, with a special emphasis on Toxoplasma-induced ileitis and encephalitis.
Collapse
Affiliation(s)
| | | | - Jeroen P. J. Saeij
- Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
17
|
Zhao XY, Ewald SE. The molecular biology and immune control of chronic Toxoplasma gondii infection. J Clin Invest 2020; 130:3370-3380. [PMID: 32609097 PMCID: PMC7324197 DOI: 10.1172/jci136226] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Toxoplasma gondii is an incredibly successful parasite owing in part to its ability to persist within cells for the life of the host. Remarkably, at least 350 host species of T. gondii have been described to date, and it is estimated that 30% of the global human population is chronically infected. The importance of T. gondii in human health was made clear with the first reports of congenital toxoplasmosis in the 1940s. However, the AIDS crisis in the 1980s revealed the prevalence of chronic infection, as patients presented with reactivated chronic toxoplasmosis, underscoring the importance of an intact immune system for parasite control. In the last 40 years, there has been tremendous progress toward understanding the biology of T. gondii infection using rodent models, human cell experimental systems, and clinical data. However, there are still major holes in our understanding of T. gondii biology, including the genes controlling parasite development, the mechanisms of cell-intrinsic immunity to T. gondii in the brain and muscle, and the long-term effects of infection on host homeostasis. The need to better understand the biology of chronic infection is underscored by the recent rise in ocular disease associated with emerging haplotypes of T. gondii and our lack of effective treatments to sterilize chronic infection. This Review discusses the cell types and molecular mediators, both host and parasite, that facilitate persistent T. gondii infection. We highlight the consequences of chronic infection for tissue-specific pathology and identify open questions in this area of host-Toxoplasma interactions.
Collapse
|
18
|
Aging with Toxoplasma gondii results in pathogen clearance, resolution of inflammation, and minimal consequences to learning and memory. Sci Rep 2020; 10:7979. [PMID: 32409672 PMCID: PMC7224383 DOI: 10.1038/s41598-020-64823-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 04/15/2020] [Indexed: 12/31/2022] Open
Abstract
Persistent inflammation has been identified as a contributor to aging-related neurodegenerative disorders such as Alzheimer's disease. Normal aging, in the absence of dementia, also results in gradual cognitive decline and is thought to arise, in part, because of a chronic pro-inflammatory state in the brain. Toxoplasma gondii is an obligate intracellular parasite that establishes a persistent, asymptomatic infection of the central nervous system (CNS) accompanied by a pro-inflammatory immune response in many of its hosts, including humans and rodents. Several studies have suggested that the inflammation generated by certain strains of T. gondii infection can be neuroprotective in the context of a secondary insult like beta-amyloid accumulation or stroke. Given these neuroprotective studies, we hypothesized that a prolonged infection with T. gondii may protect against age-associated decline in cognition. To test this hypothesis, we infected young adult mice with either of two genetically distinct, persistent T. gondii strains (Prugniaud/type II/haplogroup 2 and CEP/type III/haplogroup 3) and monitored mouse weight, survival, and learning and memory over the ensuing 20 months. At the end of the study, we evaluated CNS inflammation and parasite burden in the surviving mice. We found that parasite infection had no impact on age-associated decline in learning and memory and that by 20 months post infection, in the surviving mice, we found no evidence of parasite DNA, cysts, or inflammation in the CNS. In addition, we found that mice infected with type III parasites, which are supposed to be less virulent than the type II parasites, had a lower rate of long-term survival. Collectively, these data indicate that T. gondii may not cause a life-long CNS infection. Rather, parasites are likely slowly cleared from the CNS and infection and parasite clearance neither positively nor negatively impacts learning and memory in aging.
Collapse
|
19
|
The Bradyzoite: A Key Developmental Stage for the Persistence and Pathogenesis of Toxoplasmosis. Pathogens 2020; 9:pathogens9030234. [PMID: 32245165 PMCID: PMC7157559 DOI: 10.3390/pathogens9030234] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 12/19/2022] Open
Abstract
Toxoplasma gondii is a ubiquitous parasitic protist found in a wide variety of hosts, including a large proportion of the human population. Beyond an acute phase which is generally self-limited in immunocompetent individuals, the ability of the parasite to persist as a dormant stage, called bradyzoite, is an important aspect of toxoplasmosis. Not only is this stage not eliminated by current treatments, but it can also reactivate in immunocompromised hosts, leading to a potentially fatal outcome. Yet, despite its critical role in the pathology, the bradyzoite stage is relatively understudied. One main explanation is that it is a considerably challenging model, which essentially has to be derived from in vivo sources. However, recent progress on genetic manipulation and in vitro differentiation models now offers interesting perspectives for tackling key biological questions related to this particularly important developmental stage.
Collapse
|
20
|
Guevara RB, Fox BA, Bzik DJ. Succinylated Wheat Germ Agglutinin Colocalizes with the Toxoplasma gondii Cyst Wall Glycoprotein CST1. mSphere 2020; 5:e00031-20. [PMID: 32132158 PMCID: PMC7056803 DOI: 10.1128/msphere.00031-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 02/16/2020] [Indexed: 11/20/2022] Open
Abstract
The glycosylated mucin domain of the Toxoplasma gondii cyst wall glycoprotein CST1 is heavily stained by Dolichos biflorus agglutinin, a lectin that binds to N-acetylgalactosamine. The cyst wall is also heavily stained by the chitin binding lectin succinylated wheat germ agglutinin (s-WGA), which selectively binds to N-acetylglucosamine-decorated structures. Here, we tracked the localization of N-acetylglucosamine-decorated structures that bind to s-WGA in immature and mature in vitro cysts. s-WGA localization was observed at the cyst periphery 6 h after the differentiation of the tachyzoite-stage parasitophorous vacuole. By day 1 and at all later times after differentiation, s-WGA was localized in a continuous staining pattern at the cyst wall. Coinciding with the maturation of the cyst matrix by day 3 of cyst development, s-WGA also localized in a continuous matrix pattern inside the cyst. s-WGA localized in both the outer and inner layer regions of the cyst wall and in a continuous matrix pattern inside mature 7- and 10-day-old cysts. In addition, s-WGA colocalized in the cyst wall with CST1, suggesting that N-acetylglucosamine- and N-acetylgalactosamine-decorated molecules colocalized in the cyst wall. In contrast to CST1, GRA4, and GRA6, the relative accumulation of the molecules that bind s-WGA in the cyst wall was not dependent on the expression of GRA2. Our results suggest that GRA2-dependent and GRA2-independent mechanisms regulate the trafficking and accumulation of glycosylated molecules that colocalize in the cyst wall.IMPORTANCE Chronic Toxoplasma gondii infection is maintained in the central nervous system by thick-walled cysts. If host immunity wanes, cysts recrudesce and cause severe and often lethal toxoplasmic encephalitis. Currently, there are no therapies to eliminate cysts, and little biological information is available regarding cyst structure(s). Here, we investigated cyst wall molecules recognized by succinylated wheat germ agglutinin (s-WGA), a lectin that specifically binds to N-acetylglucosamine-decorated structures. N-Acetylglucosamine regulates cell signaling and plays structural roles at the cell surface in many organisms. The cyst wall and cyst matrix were heavily stained by s-WGA in mature cysts and were differentially stained during cyst development. The relative accumulation of molecules that bind to s-WGA in the cyst wall was not dependent on the expression of GRA2. Our findings suggest that glycosylated cyst wall molecules gain access to the cyst wall via GRA2-dependent and GRA2-independent mechanisms and colocalize in the cyst wall.
Collapse
Affiliation(s)
- Rebekah B Guevara
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Barbara A Fox
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - David J Bzik
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| |
Collapse
|
21
|
Tuladhar S, Kochanowsky JA, Bhaskara A, Ghotmi Y, Chandrasekaran S, Koshy AA. The ROP16III-dependent early immune response determines the subacute CNS immune response and type III Toxoplasma gondii survival. PLoS Pathog 2019; 15:e1007856. [PMID: 31648279 PMCID: PMC6812932 DOI: 10.1371/journal.ppat.1007856] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/25/2019] [Indexed: 12/27/2022] Open
Abstract
Toxoplasma gondii is an intracellular parasite that persistently infects the CNS and that has genetically distinct strains which provoke different acute immune responses. How differences in the acute immune response affect the CNS immune response is unknown. To address this question, we used two persistent Toxoplasma strains (type II and type III) and examined the CNS immune response at 21 days post infection (dpi). Contrary to acute infection studies, type III-infected mice had higher numbers of total CNS T cells and macrophages/microglia but fewer alternatively activated macrophages (M2s) and regulatory T cells (Tregs) than type II-infected mice. By profiling splenocytes at 5, 10, and 21 dpi, we determined that at 5 dpi type III-infected mice had more M2s while type II-infected mice had more pro-inflammatory macrophages and that these responses flipped over time. To test how these early differences influence the CNS immune response, we engineered the type III strain to lack ROP16 (IIIΔrop16), the polymorphic effector protein that drives the early type III-associated M2 response. IIIΔrop16-infected mice showed a type II-like neuroinflammatory response with fewer infiltrating T cells and macrophages/microglia and more M2s and an unexpectedly low CNS parasite burden. At 5 dpi, IIIΔrop16-infected mice showed a mixed inflammatory response with more pro-inflammatory macrophages, M2s, T effector cells, and Tregs, and decreased rates of infection of peritoneal exudative cells (PECs). These data suggested that type III parasites need the early ROP16-associated M2 response to avoid clearance, possibly by the Immunity-Related GTPases (IRGs), which are IFN-γ- dependent proteins essential for murine defenses against Toxoplasma. To test this possibility, we infected IRG-deficient mice and found that IIIΔrop16 parasites now maintained parental levels of PECs infection. Collectively, these studies suggest that, for the type III strain, rop16III plays a key role in parasite persistence and influences the subacute CNS immune response. Toxoplasma is a ubiquitous intracellular parasite that establishes an asymptomatic brain infection in immunocompetent individuals. However, in the immunocompromised and the developing fetus, Toxoplasma can cause problems ranging from fever to chorioretinitis to severe toxoplasmic encephalitis. Emerging evidence suggests that the genotype of the infecting Toxoplasma strain may influence these outcomes, possibly through the secretion of Toxoplasma strain-specific polymorphic effector proteins that trigger different host cell signaling pathways. While such strain-specific modulation of host cell signaling has been shown to affect acute immune responses, it is unclear how these differences influence the subacute or chronic responses in the CNS, the major organ affected in symptomatic disease. This study shows that genetically distinct strains of Toxoplasma provoke strain-specific CNS immune responses and that, for one strain (type III), acute and subacute immune responses and parasite survival are heavily influenced by a polymorphic parasite gene (rop16III).
Collapse
Affiliation(s)
- Shraddha Tuladhar
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
| | - Joshua A. Kochanowsky
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
| | - Apoorva Bhaskara
- Bio5 Institute, University of Arizona, Tucson, Arizona, United States of America
| | - Yarah Ghotmi
- Bio5 Institute, University of Arizona, Tucson, Arizona, United States of America
- Undergraduate Biology Research Program (UBRP), University of Arizona, Tucson, Arizona, United States of America
| | | | - Anita A. Koshy
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
- Bio5 Institute, University of Arizona, Tucson, Arizona, United States of America
- Department of Neurology, University of Arizona, Tucson, Arizona, United States of America
- * E-mail:
| |
Collapse
|
22
|
Hettiarachchi SD, Zhou Y, Seven E, Lakshmana MK, Kaushik AK, Chand HS, Leblanc RM. Nanoparticle-mediated approaches for Alzheimer's disease pathogenesis, diagnosis, and therapeutics. J Control Release 2019; 314:125-140. [PMID: 31647979 DOI: 10.1016/j.jconrel.2019.10.034] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/16/2019] [Accepted: 10/18/2019] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is an irreversible and progressive neurodegenerative disorder manifested by memory loss and cognitive impairment. Deposition of the amyloid β plaques has been identified as the most common AD pathology; however, the excessive accumulation of phosphorylated or total tau proteins, reactive oxygen species, and higher acetylcholinesterase activity are also strongly associated with Alzheimer's dementia. Several therapeutic approaches targeting these pathogenic mechanisms have failed in clinical or preclinical trials, partly due to the limited bioavailability, poor cell, and blood-brain barrier penetration, and low drug half-life of current regimens. The nanoparticles (NPs)-mediated drug delivery systems improve drug solubility and bioavailability, thus renders as superior alternatives. Moreover, NPs-mediated approaches facilitate multiple drug loading and targeted drug delivery, thereby increasing drug efficacy. However, certain NPs can cause acute toxicity damaging cellular and tissue architecture, therefore, NP material should be carefully selected. In this review, we summarize the recent NPs-mediated studies that exploit various pathologic mechanisms of AD by labeling, identifying, and treating the affected brain pathologies. The disadvantages of the select NP-based deliveries and the future aspects will also be discussed.
Collapse
Affiliation(s)
- Sajini D Hettiarachchi
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
| | - Yiqun Zhou
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
| | - Elif Seven
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
| | - Madepalli K Lakshmana
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA
| | - Ajeet K Kaushik
- Department of Natural Sciences, Division of Sciences, Arts & Mathematics, Florida Polytechnic University, Lakeland, FL 33805-8531, USA
| | - Hitendra S Chand
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA
| | - Roger M Leblanc
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
| |
Collapse
|
23
|
Establishment of BV2 microglia polarization model and its effect on Toxoplasma gondii proliferation. Res Vet Sci 2019; 125:382-389. [PMID: 31404885 DOI: 10.1016/j.rvsc.2019.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 06/08/2019] [Accepted: 08/02/2019] [Indexed: 02/07/2023]
Abstract
Toxoplasma gondii is an intracellular opportunistic, parasitic protozoan. Microglia have been classified into two main types: M1 (classically activated macrophages) and M2 (alternatively activated macrophages). BV2 cells were used in this study, together with lipopolysaccharide (LPS) and interferon (IFN)-γ or interleukin (IL)-4, which were used to induce resting microglia. Expression levels of M1/M2 markers were determined at both mRNA and protein levels, using PCR, western blot, and flow cytometry. Furthermore, cells were infected with T. gondii PLK strain, and the dynamic changes in M1/M2 marker expression levels were determined. An in vitro polarization model was successfully established. Expression of Nos2 and M1-associated markers was significantly upregulated at 12 h post-infection in BV2 cells. Further, the JAK/STAT1 and NF-κB signaling pathways were also activated following T. gondii infection. This demonstrated that T. gondii infection induces M1-type microglial polarization in vitro. The present study demonstrated that T. gondii infection affects microglial activation in vitro and elucidated the effects of activated microglia on T. gondii proliferation. This data may serve as a useful reference for more detailed elucidation of interactions between T. gondii and the innate immune system.
Collapse
|
24
|
Tiwari A, Hannah R, Lutshumba J, Ochiai E, Weiss LM, Suzuki Y. Penetration of CD8 + Cytotoxic T Cells into Large Target, Tissue Cysts of Toxoplasma gondii, Leads to Its Elimination. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1594-1607. [PMID: 31301754 DOI: 10.1016/j.ajpath.2019.04.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 04/29/2019] [Indexed: 10/26/2022]
Abstract
CD8+ cytotoxic T cells kill target cells through direct cell-cell contact. However, it remains unclear how these T cells eliminate a target of large mass. We investigated how CD8+ T cells remove tissue cysts of Toxoplasma gondii, which can grow to the size of >50 μm in diameter within infected cells. Notably, immunohistologic analyses in the brains of infected mice visualized the presence of numbers of CD8+ immune T cells that had migrated halfway through the cyst wall as well as T cells located fully within the cysts. Perforin was required for their invasion and cyst elimination. Cysts invaded by the T cells displayed morphologic deterioration and destruction. Within these deteriorated cysts, granular structures intensely positive for granzyme B were detected in association with T. gondii bradyzoites. Furthermore, the bradyzoites within the destroyed cysts were located within accumulated ionized calcium binding adaptor molecule 1 (Iba1)-positive microglia and Ly6C+ macrophages, suggesting that these phagocytes had phagocytosed those organisms for their eradication. The present study uncovered a previously unappreciated capability of CD8+ cytotoxic T cells to penetrate into a large target, T. gondii cysts, for their elimination. This invasive capability of CD8+ cytotoxic T cells in collaboration with phagocytes appears to be a powerful effector mechanism that functions against not only T. gondii cysts but also other large targets, including solid cancers.
Collapse
Affiliation(s)
- Ashish Tiwari
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Rancie Hannah
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Jenny Lutshumba
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Eri Ochiai
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Louis M Weiss
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Yasuhiro Suzuki
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky; Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, Virginia.
| |
Collapse
|
25
|
El-Kowrany SI, El Ghaffar AESA, Shoheib ZS, Mady RF, Gamea GAM. Evaluation of nitazoxanide as a novel drug for the treatment of acute and chronic toxoplasmosis. Acta Trop 2019; 195:145-154. [PMID: 30986380 DOI: 10.1016/j.actatropica.2019.04.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/11/2019] [Accepted: 04/11/2019] [Indexed: 01/03/2023]
Abstract
Toxoplasmosis is a widespread, neglected disease with significant morbidity and mortality. In search of an effective treatment, nitazoxanide (NTZ) was evaluated in the treatment of acute and chronic toxoplasmosis in experimental mice. For this purpose, mice were infected with 20 cysts (acute infection model) or 10 cysts (chronic infection model) of Toxoplasma gondii (ME 49 strain). Treated mice received NTZ (at doses of 100 and 150 mg/kg), starting from the third day (acute model) or the fifth week (chronic model) post-infection, which continued for 14 consecutive days. The effects of NTZ were evaluated in comparison to the pyrimethamine/sulfadiazine combination. Evaluation included mortality rates, brain cyst count, inflammatory scoring and immunological studies. The latter included estimation of interferon-gamma (IFN-γ) and induced nitric oxide synthase (iNOS). In the acute infection model, NTZ at 100 and 150 mg/kg significantly reduced the number of brain cysts by 78 and 87% compared to the infected untreated controls and reduced the mortality rate to 24 and 20%, respectively, compared with 44% in the infected untreated control. In the chronic infection model, cyst reduction reached 32 and 38% for 100 and 150 mg/kg NTZ treatments, respectively. NTZ was significantly able to reduce inflammation caused by acute and chronic T. gondii infection with slight necrosis and few infiltrating mononuclear cells. Additionally, the immunological analysis revealed that NTZ significantly increased the production of serum IFN-γ and enhanced iNOS production in brain tissue, suggesting an immunomodulatory role for the drug. Based on the findings of the present study, it can be concluded that NTZ is a potential drug for the treatment of acute and chronic toxoplasmosis.
Collapse
|
26
|
Wang AW, Avramopoulos D, Lori A, Mulle J, Conneely K, Powers A, Duncan E, Almli L, Massa N, McGrath J, Schwartz AC, Goes FS, Weng L, Wang R, Yolken R, Ruczinski I, Gillespie CF, Jovanovic T, Ressler K, Pulver AE, Pearce BD. Genome-wide association study in two populations to determine genetic variants associated with Toxoplasma gondii infection and relationship to schizophrenia risk. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:133-147. [PMID: 30610941 DOI: 10.1016/j.pnpbp.2018.12.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 12/20/2018] [Accepted: 12/30/2018] [Indexed: 01/10/2023]
Abstract
T. gondii (TOXO) infects over one billion people worldwide, yet the literature lacks a Genome Wide Association Study (GWAS) focused on genetic variants controlling the persistence of TOXO infection. To identify putative T. gondii susceptibility genes, we performed a GWAS using IgG seropositivity as the outcome variable in a discovery sample (n = 790) from an Ashkenazi dataset, and a second sample of predominately African Americans (The Grady Trauma Project, n = 285). We also performed a meta-analyses of the 2 cohorts. None of the SNPs in these analyses was statistically significant after Bonferroni correction for multiple comparisons. In the Ashkenazi population, the gene region of CHIA (chitinase) showed the most nominally significant association with TOXO. Prior studies have shown that the production of chitinase by macrophages in the brain responding to TOXO infection is crucial for controlling the burden of T. gondii cysts. We found a surprising number of genes involved in neurodevelopment and psychiatric disorders among our top hits even though our outcome variable was TOXO infection. In the meta-analysis combining the Ashkenazi and Grady Trauma Project samples, there was enrichment for genes implicated in schizophrenia spectrum disorders (p < .05). Upon limiting our sample to those without mental illness, two schizophrenia related genes (CNTNAP2, GABAR2) still had significant TOXO-associated variants at the p < .05 level, but did not pass the genome wide significance threshold after correction for multiple comparisons. Using Ingenuity Systems molecular network analysis, we identified molecular nodes suggesting that while different genetic variants associated with TOXO in the two population samples, the molecular pathways for TOXO susceptibility nevertheless converged on common pathways. Molecular nodes in these common pathways included NOTCH1, CD44, and RXRA. Prior studies show that CD44 participates in TOXO-induced immunopathology and that RXRA is instrumental in regulating T-helper immune responses. These data provide new insights into the pathophysiology of this common neurotropic parasite.
Collapse
Affiliation(s)
- Alex W Wang
- Department of Epidemiology, Rollins School of Public Health, 1518 Clifton Rd., Atlanta, GA 30322, United States
| | - Dimitrios Avramopoulos
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Adriana Lori
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| | - Jennifer Mulle
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| | - Karen Conneely
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| | - Abigail Powers
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, United States
| | - Erica Duncan
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, United States; Atlanta Veterans Affairs Medical Center, 1670 Clairmont Rd, Decatur, GA 30033, United States
| | - Lynn Almli
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, United States
| | - Nicholas Massa
- Department of Epidemiology, Rollins School of Public Health, 1518 Clifton Rd., Atlanta, GA 30322, United States; Atlanta Veterans Affairs Medical Center, 1670 Clairmont Rd, Decatur, GA 30033, United States
| | - John McGrath
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ann C Schwartz
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, United States
| | - Fernando S Goes
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lei Weng
- Department of Epidemiology, Rollins School of Public Health, 1518 Clifton Rd., Atlanta, GA 30322, United States
| | - Ruihua Wang
- McKusick Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Robert Yolken
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ingo Ruczinski
- Bloomberg School of Public Heath, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Charles F Gillespie
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, United States
| | - Tanja Jovanovic
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, United States
| | - Kerry Ressler
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, United States; Department of Psychiatry, Harvard School of Medicine, 25 Shattuck St, Boston, MA 02115, United States
| | - Ann E Pulver
- Bloomberg School of Public Heath, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Brad D Pearce
- Department of Epidemiology, Rollins School of Public Health, 1518 Clifton Rd., Atlanta, GA 30322, United States.
| |
Collapse
|
27
|
Characterization of strain-specific phenotypes associated with knockout of dense granule protein 9 in Toxoplasma gondii. Mol Biochem Parasitol 2019; 229:53-61. [PMID: 30849416 DOI: 10.1016/j.molbiopara.2019.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 11/23/2022]
Abstract
Toxoplasma gondii is an obligate intracellular parasite that can invade any nucleated cell of mammals and cause toxoplasmosis. Dense granule proteins play major structural functions within the parasitophorous vacuole (PV) and the cyst wall of T. gondii. Moreover, their particular location within the PV allows them to be involved in various interactions between parasites and the host cells. Dense granule protein 9 (GRA9) gene has been identified in T. gondii, although its role in the lytic cycle remains unclear. In the current study, the function of GRA9 in type I and type II Toxoplasma parasites was characterized. T. gondii GRA9 sequence and its expression were analyzed and derivatives of T. gondii RH and PLK strains with a null mutation in GRA9 were generated using CRISPR/Cas9 system. The phenotypes of GRA9 in wild types, knockout and complemented strains were analyzed in vitro and in vivo using Vero cells and BALB/c mice, respectively. Alignment of the amino acid sequence indicated that RH strain GRA9 contained one amino acid substitution when compared with PLK strain. Western blot analysis revealed that PLK strain had a higher expression level of GRA9 than RH strain. The phenotype analysis revealed that knockout of GRA9 in PLK parasites inhibited the plaque formation and egress from PV. Both the plaque formation and egress ability of PLKΔGRA9 strain were restored by complementation with a synonymous allele of PLK strain GRA9. Mouse experiments demonstrated that loss of GRA9 in PLK strain significantly reduced the pathogenicity of T. gondii. However, there was no phenotypic diferences between RH and RHΔGRA9 strains except the defect in host cell invasion. Overall, T. gondii GRA9 knockout only influenced the growth and virulence of PLK strain. These results indicate that GRA9 may be involved in parasite egress and virulence in mice in a strain-specific manner.
Collapse
|
28
|
Huang WY, Wang YP, Mahmmod YS, Wang JJ, Liu TH, Zheng YX, Zhou X, Zhang XX, Yuan ZG. A Double-Edged Sword: Complement Component 3 in Toxoplasma gondii Infection. Proteomics 2019; 19:e1800271. [PMID: 30515942 DOI: 10.1002/pmic.201800271] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 11/01/2018] [Indexed: 12/14/2022]
Abstract
Sprague Dawley rats and Kunming (KM) mice are artificially infected with type II Toxoplasma gondii strain Prugniaud (Pru) to generate toxoplasmosis, which is a fatal disease mediated by T. gondii invasion of the central nervous system (CNS) by unknown mechanisms. The aim is to explore the mechanism of differential susceptibility of mice and rats to T. gondii infection. Therefore, a strategy of isobaric tags for relative and absolute quantitation (iTRAQ) is established to identify differentially expressed proteins (DEPs) in the rats' and the mice's brains compared to the healthy groups. In KM mice, which is susceptible to T. gondii infection, complement component 3 (C3) is upregulated and the tight junction (TJ) pathway shows a disorder. It is presumed that T. gondii-stimulated C3 disrupts the TJ of the blood-brain barrier in the CNS. This effect allows more T. gondii passing to the brain through the intercellular space.
Collapse
Affiliation(s)
- Wan-Yi Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, Guangdong, P. R. China
| | - Ya-Pei Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, Guangdong, P. R. China
| | - Yasser S Mahmmod
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA), Campus de la Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain.,Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain.,Infectious Diseases, Department of Animal Medicine, Faculty of Veterinary Medicine, Zagazig University, 44511, Zagazig, Sharkia Province, Egypt
| | - Jun-Jie Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, Guangdong, P. R. China
| | - Tang-Hui Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, Guangdong, P. R. China
| | - Yu-Xiang Zheng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, Guangdong, P. R. China
| | - Xue Zhou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, Guangdong, P. R. China
| | - Xiu-Xiang Zhang
- College of Agriculture, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China
| | - Zi-Guo Yuan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou, 510642, Guangdong, P. R. China.,Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, Guangdong, P. R. China
| |
Collapse
|
29
|
Hasby Saad MA, Watany M, Tomoum M, El-Mehy D, Elsheikh M, Sharshar R. Acidic mammalian chitinase tuning after enteric helminths eradication in inflammatory respiratory disease patients. Parasite Immunol 2018; 40:e12583. [DOI: 10.1111/pim.12583] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/07/2018] [Accepted: 08/09/2018] [Indexed: 01/18/2023]
Affiliation(s)
- Marwa A. Hasby Saad
- Medical Parasitology Department; Faculty of Medicine; Tanta University; Tanta Egypt
| | - Mona Watany
- Clinical Pathology Department; Faculty of Medicine; Tanta University; Tanta Egypt
| | - Mohamed Tomoum
- Otorhinolaryngeology Department; Faculty of Medicine; Tanta University; Tanta Egypt
| | - Dalia El-Mehy
- Medical Parasitology Department; Faculty of Medicine; Tanta University; Tanta Egypt
| | - May Elsheikh
- Paediatric Department; Faculty of Medicine; Tanta University; Tanta Egypt
| | - Ragia Sharshar
- Pulmonology Department; Faculty of Medicine; Tanta University; Tanta Egypt
| |
Collapse
|
30
|
Acidic Mammalian Chitinase Negatively Affects Immune Responses during Acute and Chronic Aspergillus fumigatus Exposure. Infect Immun 2018; 86:IAI.00944-17. [PMID: 29712728 DOI: 10.1128/iai.00944-17] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/20/2018] [Indexed: 12/12/2022] Open
Abstract
Chitin is a polysaccharide that provides structure and rigidity to the cell walls of fungi and insects. Mammals possess multiple chitinases, which function to degrade chitin, thereby supporting a role for chitinases in immune defense. However, chitin degradation has been implicated in the pathogenesis of asthma. Here, we determined the impact of acidic mammalian chitinase (AMCase) (Chia) deficiency on host defense during acute exposure to the fungal pathogen Aspergillus fumigatus as well as its contribution to A. fumigatus-associated allergic asthma. We demonstrate that chitin in the fungal cell wall was detected at low levels in A. fumigatus conidia, which emerged at the highest level during hyphal transition. In response to acute A. fumigatus challenge, Chia-/- mice unexpectedly demonstrated lower A. fumigatus lung burdens at 2 days postchallenge. The lower fungal burden correlated with decreased lung interleukin-33 (IL-33) levels yet increased IL-1β and prostaglandin E2 (PGE2) production, a phenotype that we reported previously to promote the induction of IL-17A and IL-22. During chronic A. fumigatus exposure, AMCase deficiency resulted in lower dynamic and airway lung resistance than in wild-type mice. Improved lung physiology correlated with attenuated levels of the proallergic chemokines CCL17 and CCL22. Surprisingly, examination of inflammatory responses during chronic exposure revealed attenuated IL-17A and IL-22 responses, but not type 2 responses, in the absence of AMCase. Collectively, these data suggest that AMCase functions as a negative regulator of immune responses during acute fungal exposure and is a contributor to fungal asthma severity, putatively via the induction of proinflammatory responses.
Collapse
|
31
|
Ngô HM, Zhou Y, Lorenzi H, Wang K, Kim TK, Zhou Y, El Bissati K, Mui E, Fraczek L, Rajagopala SV, Roberts CW, Henriquez FL, Montpetit A, Blackwell JM, Jamieson SE, Wheeler K, Begeman IJ, Naranjo-Galvis C, Alliey-Rodriguez N, Davis RG, Soroceanu L, Cobbs C, Steindler DA, Boyer K, Noble AG, Swisher CN, Heydemann PT, Rabiah P, Withers S, Soteropoulos P, Hood L, McLeod R. Toxoplasma Modulates Signature Pathways of Human Epilepsy, Neurodegeneration & Cancer. Sci Rep 2017; 7:11496. [PMID: 28904337 PMCID: PMC5597608 DOI: 10.1038/s41598-017-10675-6] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 08/14/2017] [Indexed: 12/27/2022] Open
Abstract
One third of humans are infected lifelong with the brain-dwelling, protozoan parasite, Toxoplasma gondii. Approximately fifteen million of these have congenital toxoplasmosis. Although neurobehavioral disease is associated with seropositivity, causality is unproven. To better understand what this parasite does to human brains, we performed a comprehensive systems analysis of the infected brain: We identified susceptibility genes for congenital toxoplasmosis in our cohort of infected humans and found these genes are expressed in human brain. Transcriptomic and quantitative proteomic analyses of infected human, primary, neuronal stem and monocytic cells revealed effects on neurodevelopment and plasticity in neural, immune, and endocrine networks. These findings were supported by identification of protein and miRNA biomarkers in sera of ill children reflecting brain damage and T. gondii infection. These data were deconvoluted using three systems biology approaches: "Orbital-deconvolution" elucidated upstream, regulatory pathways interconnecting human susceptibility genes, biomarkers, proteomes, and transcriptomes. "Cluster-deconvolution" revealed visual protein-protein interaction clusters involved in processes affecting brain functions and circuitry, including lipid metabolism, leukocyte migration and olfaction. Finally, "disease-deconvolution" identified associations between the parasite-brain interactions and epilepsy, movement disorders, Alzheimer's disease, and cancer. This "reconstruction-deconvolution" logic provides templates of progenitor cells' potentiating effects, and components affecting human brain parasitism and diseases.
Collapse
Affiliation(s)
- Huân M Ngô
- The University of Chicago, Chicago, IL, 60637, USA.,Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA.,BrainMicro LLC, New Haven, CT, 06511, USA
| | - Ying Zhou
- The University of Chicago, Chicago, IL, 60637, USA
| | | | - Kai Wang
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Taek-Kyun Kim
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Yong Zhou
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | | | - Ernest Mui
- The University of Chicago, Chicago, IL, 60637, USA
| | | | | | | | - Fiona L Henriquez
- The University of Chicago, Chicago, IL, 60637, USA.,FLH, IBEHR School of Science and Sport, University of the West of Scotland, Paisley, PA1 2BE, UK
| | - Alexandre Montpetit
- Genome Quebec, Montréal, QC H3B 1S6, Canada; McGill University, Montréal, QC H3A 0G4, Canada
| | - Jenefer M Blackwell
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, United Kingdom.,Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Sarra E Jamieson
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | | | | | | | | | | | | | - Charles Cobbs
- California Pacific Medical Center, San Francisco, CA, 94114, USA
| | - Dennis A Steindler
- JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, 02111, USA
| | - Kenneth Boyer
- Rush University Medical Center, Chicago, IL, 60612, USA
| | - A Gwendolyn Noble
- Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Charles N Swisher
- Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | | | - Peter Rabiah
- Northshore University Health System, Evanston, IL, 60201, USA
| | | | | | - Leroy Hood
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Rima McLeod
- The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
32
|
Wohlfert EA, Blader IJ, Wilson EH. Brains and Brawn: Toxoplasma Infections of the Central Nervous System and Skeletal Muscle. Trends Parasitol 2017; 33:519-531. [PMID: 28483381 PMCID: PMC5549945 DOI: 10.1016/j.pt.2017.04.001] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/29/2017] [Accepted: 04/08/2017] [Indexed: 02/06/2023]
Abstract
Toxoplasma gondii is a widespread parasitic pathogen that infects over a third of the world's population. Following an acute infection, the parasite can persist within its mammalian host as intraneuronal or intramuscular cysts. Cysts will occasionally reactivate, and - depending on the host's immune status and site of reactivation - encephalitis or myositis can develop. Because these diseases have high levels of morbidity and can be lethal, it is important to understand how Toxoplasma traffics to these tissues, how the immune response controls parasite burden and contributes to tissue damage, and what mechanisms underlie neurological and muscular pathologies that toxoplasmosis patients present with. This review aims to summarize recent important developments addressing these critical topics.
Collapse
Affiliation(s)
- Elizabeth A Wohlfert
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY, USA.
| | - Ira J Blader
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY, USA.
| | - Emma H Wilson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, USA.
| |
Collapse
|
33
|
Cabral CM, McGovern KE, MacDonald WR, Franco J, Koshy AA. Dissecting Amyloid Beta Deposition Using Distinct Strains of the Neurotropic Parasite Toxoplasma gondii as a Novel Tool. ASN Neuro 2017; 9:1759091417724915. [PMID: 28817954 PMCID: PMC5565021 DOI: 10.1177/1759091417724915] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/29/2017] [Accepted: 06/29/2017] [Indexed: 01/13/2023] Open
Abstract
Genetic and pathologic data suggest that amyloid beta (Aβ), produced by processing of the amyloid precursor protein, is a major initiator of Alzheimer's disease (AD). To gain new insights into Aβ modulation, we sought to harness the power of the coevolution between the neurotropic parasite Toxoplasma gondii and the mammalian brain. Two prior studies attributed Toxoplasma-associated protection against Aβ to increases in anti-inflammatory cytokines (TGF-β and IL-10) and infiltrating phagocytic monocytes. These studies only used one Toxoplasma strain making it difficult to determine if the noted changes were associated with Aβ protection or simply infection. To address this limitation, we infected a third human amyloid precursor protein AD mouse model (J20) with each of the genetically distinct, canonical strains of Toxoplasma (Type I, Type II, or Type III). We then evaluated the central nervous system (CNS) for Aβ deposition, immune cell responses, global cytokine environment, and parasite burden. We found that only Type II infection was protective against Aβ deposition despite both Type II and Type III strains establishing a chronic CNS infection and inflammatory response. Compared with uninfected and Type I-infected mice, both Type II- and Type III-infected mice showed increased numbers of CNS T cells and microglia and elevated pro-inflammatory cytokines, but neither group showed a >2-fold elevation of TGF-β or IL-10. These data suggest that we can now use our identification of protective (Type II) and nonprotective (Type III) Toxoplasma strains to determine what parasite and host factors are linked to decreased Aβ burden rather than simply with infection.
Collapse
Affiliation(s)
| | | | - Wes R. MacDonald
- Undergraduate Biology Research Program, University of Arizona, Tucson, AZ, USA
| | - Jenna Franco
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
| | - Anita A. Koshy
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- Department of Immunobiology, University of Arizona, Tucson, AZ, USA
- Department of Neurology, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
34
|
Klein RS, Hunter CA. Protective and Pathological Immunity during Central Nervous System Infections. Immunity 2017; 46:891-909. [PMID: 28636958 PMCID: PMC5662000 DOI: 10.1016/j.immuni.2017.06.012] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/05/2017] [Accepted: 06/05/2017] [Indexed: 02/08/2023]
Abstract
The concept of immune privilege of the central nervous system (CNS) has dominated the study of inflammatory processes in the brain. However, clinically relevant models have highlighted that innate pathways limit pathogen invasion of the CNS and adaptive immunity mediates control of many neural infections. As protective responses can result in bystander damage, there are regulatory mechanisms that balance protective and pathological inflammation, but these mechanisms might also allow microbial persistence. The focus of this review is to consider the host-pathogen interactions that influence neurotropic infections and to highlight advances in our understanding of innate and adaptive mechanisms of resistance as key determinants of the outcome of CNS infection. Advances in these areas have broadened our comprehension of how the immune system functions in the brain and can readily overcome immune privilege.
Collapse
Affiliation(s)
- Robyn S Klein
- Departments of Medicine, Pathology and Immunology, Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
35
|
Bourgeois Y, Roulin AC, Müller K, Ebert D. Parasitism drives host genome evolution: Insights from thePasteuria ramosa-Daphnia magnasystem. Evolution 2017; 71:1106-1113. [DOI: 10.1111/evo.13209] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 02/16/2017] [Indexed: 01/14/2023]
Affiliation(s)
- Yann Bourgeois
- Zoological Institute; Basel University; Vesalgasse 1 4051 Basel Switzerland
| | - Anne C. Roulin
- Zoological Institute; Basel University; Vesalgasse 1 4051 Basel Switzerland
- Institute of Plant and Microbial Biology; Zollikerstrasse 107 8008 Zürich Switzerland
| | - Kristina Müller
- Zoological Institute; Basel University; Vesalgasse 1 4051 Basel Switzerland
| | - Dieter Ebert
- Zoological Institute; Basel University; Vesalgasse 1 4051 Basel Switzerland
| |
Collapse
|
36
|
Sanfilippo C, Nunnari G, Calcagno A, Malaguarnera L, Blennow K, Zetterberg H, Di Rosa M. The chitinases expression is related to Simian Immunodeficiency Virus Encephalitis (SIVE) and in HIV encephalitis (HIVE). Virus Res 2017; 227:220-230. [PMID: 27794455 DOI: 10.1016/j.virusres.2016.10.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/14/2016] [Accepted: 10/21/2016] [Indexed: 01/15/2023]
Abstract
OBJECTIVES Human Immunodeficiency Virus (HIV) infection can induce neurocognitive complications classified as HIV-associated neurocognitive disorder (HAND). The chitinase family is associated with innate immunity cells and many infectious diseases. METHODS We analyzed microarray datasets obtained from NCBI in order to verify the expression of chitinase family genes in hippocampus of uninfected rhesus macaques versus those with histopathologic evidence of Simian Immunodeficiency Virus Encephalitis (SIVE). Moreover, we have analysed two human microarray datasets to verify the results obtained in macaques hippocampus affected by SIVE. For these studies, we have also used the open source tools Genome-scale Integrated Analysis of gene Networks in Tissues (GIANT) to identify the chitinase genes network. RESULTS CHIT1, CHI3L1 and CHI3L2 levels were significantly increased in SIVE hippocampus as compared to non-infected control specimens. Furthermore, we found a negative correlation between CHIA vs. Brain Viral Load (BVL). These data was confirmed partially in human brain section of HAD/HIVE subjects. Also, we showed that HIV-1 was able to modulate the expression of CHIT1, CHI3L1, CHI3L2 and CHID1 in human macrophages. CONCLUSIONS These results suggest that chitinase gene expression is altered in SIVE and in HAD/HIVE brain sections and call for more studies examining whether this is a protective immunological reaction or a destructive tissue response to encephalitis.
Collapse
Affiliation(s)
- C Sanfilippo
- Section of Neurosciences, Department G.F. Ingrassia, University of Catania, Via Santa Sofia, 78, 95123, Catania, Italy
| | - G Nunnari
- Unit of Infectious Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - A Calcagno
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Italy
| | - L Malaguarnera
- Department of Biomedical and Biotechnological Sciences, University of Catania, Italy
| | - K Blennow
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, University of Gothenburg, Sahlgrenska University Hospital, SE-43180, Mölndal, Sweden
| | - H Zetterberg
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, University of Gothenburg, Sahlgrenska University Hospital, SE-43180, Mölndal, Sweden
| | - M Di Rosa
- Department of Biomedical and Biotechnological Sciences, University of Catania, Italy.
| |
Collapse
|
37
|
Jin RM, Blair SJ, Warunek J, Heffner RR, Blader IJ, Wohlfert EA. Regulatory T Cells Promote Myositis and Muscle Damage in Toxoplasma gondii Infection. THE JOURNAL OF IMMUNOLOGY 2016; 198:352-362. [PMID: 27895180 DOI: 10.4049/jimmunol.1600914] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 11/01/2016] [Indexed: 11/19/2022]
Abstract
The coordination of macrophage polarization is essential for the robust regenerative potential of skeletal muscle. Repair begins with a phase mediated by inflammatory monocytes (IM) and proinflammatory macrophages (M1), followed by polarization to a proregenerative macrophage (M2) phenotype. Recently, regulatory T cells (Tregs) were described as necessary for this M1 to M2 transition. We report that chronic infection with the protozoan parasite Toxoplasma gondii causes a nonresolving Th1 myositis with prolonged tissue damage associated with persistent M1 accumulation. Surprisingly, Treg ablation during chronic infection rescues macrophage homeostasis and skeletal muscle fiber regeneration, showing that Tregs can directly contribute to muscle damage. This study provides evidence that the tissue environment established by the parasite could lead to a paradoxical pathogenic role for Tregs. As such, these findings should be considered when tailoring therapies directed at Tregs in inflammatory settings.
Collapse
Affiliation(s)
- Richard M Jin
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214; and
| | - Sarah J Blair
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214; and
| | - Jordan Warunek
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214; and
| | - Reid R Heffner
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214
| | - Ira J Blader
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214; and
| | - Elizabeth A Wohlfert
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214; and
| |
Collapse
|
38
|
Xiao J, Li Y, Gressitt KL, He H, Kannan G, Schultz TL, Svezhova N, Carruthers VB, Pletnikov MV, Yolken RH, Severance EG. Cerebral complement C1q activation in chronic Toxoplasma infection. Brain Behav Immun 2016; 58:52-56. [PMID: 27109609 PMCID: PMC5067173 DOI: 10.1016/j.bbi.2016.04.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/07/2016] [Accepted: 04/20/2016] [Indexed: 11/19/2022] Open
Abstract
Exposure to the neurotropic parasite, Toxoplasma gondii, causes significant brain and behavioral anomalies in humans and other mammals. Understanding the cellular mechanisms of T. gondii-generated brain pathologies would aid the advancement of novel strategies to reduce disease. Complement factor C1q is part of a classic immune pathway that functions peripherally to tag and remove infectious agents and cellular debris from circulation. In the developing and adult brain, C1q modifies neuronal architecture through synapse marking and pruning. T. gondii exposure and complement activation have both been implicated in the development of complex brain disorders such as schizophrenia. Thus, it seems logical that mechanistically, the physiological pathways associated with these two factors are connected. We employed a rodent model of chronic infection to investigate the extent to which cyst presence in the brain triggers activation of cerebral C1q. Compared to uninfected mice, cortical C1q was highly expressed at both the RNA and protein levels in infected animals bearing a high cyst burden. In these mice, C1q protein localized to cytoplasm, adjacent to GFAP-labeled astrocytes, near degenerating cysts, and in punctate patterns along processes. In summary, our results demonstrated an upregulation of cerebral C1q in response to latent T. gondii infection. Our data preliminarily suggest that this complement activity may aid in the clearance of this parasite from the CNS and in so doing, have consequences for the connectivity of neighboring cells and synapses.
Collapse
Affiliation(s)
- Jianchun Xiao
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Blalock 1105, Baltimore, MD 21287-4933, USA
| | - Ye Li
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Blalock 1105, Baltimore, MD 21287-4933, USA
| | - Kristin L Gressitt
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Blalock 1105, Baltimore, MD 21287-4933, USA
| | - Helen He
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Blalock 1105, Baltimore, MD 21287-4933, USA
| | - Geetha Kannan
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Tracey L Schultz
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Nadezhda Svezhova
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Vern B Carruthers
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Mikhail V Pletnikov
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Blalock 1105, Baltimore, MD 21287-4933, USA
| | - Emily G Severance
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Blalock 1105, Baltimore, MD 21287-4933, USA.
| |
Collapse
|
39
|
Abstract
Ingestion of the obligate intracellular protozoan parasite Toxoplasma gondii causes an acute infection that leads to chronic infection of the host. To facilitate the acute phase of the infection, T. gondii manipulates the host response by secreting rhoptry organelle proteins (ROPs) into host cells during its invasion. A few key ROP proteins with signatures of kinases or pseudokinases (ROPKs) act as virulence factors that enhance parasite survival against host gamma interferon-stimulated innate immunity. However, the roles of these and other ROPK proteins in establishing chronic infection have not been tested. Here, we deleted 26 ROPK gene loci encoding 31 unique ROPK proteins of type II T. gondii and show that numerous ROPK proteins influence the development of chronic infection. Cyst burdens were increased in the Δrop16 knockout strain or moderately reduced in 11 ROPK knockout strains. In contrast, deletion of ROP5, ROP17, ROP18, ROP35, or ROP38/29/19 (ROP38, ROP29, and ROP19) severely reduced cyst burdens. Δrop5 and Δrop18 knockout strains were less resistant to host immunity-related GTPases (IRGs) and exhibited >100-fold-reduced virulence. ROP18 kinase activity and association with the parasitophorous vacuole membrane were necessary for resistance to host IRGs. The Δrop17 strain exhibited a >12-fold defect in virulence; however, virulence was not affected in the Δrop35 or Δrop38/29/19 strain. Resistance to host IRGs was not affected in the Δrop17, Δrop35, or Δrop38/29/19 strain. Collectively, these findings provide the first definitive evidence that the type II T. gondii ROPK proteome functions as virulence factors and facilitates additional mechanisms of host manipulation that are essential for chronic infection and transmission of T. gondii. Reactivation of chronic Toxoplasma gondii infection in individuals with weakened immune systems causes severe toxoplasmosis. Existing treatments for toxoplasmosis are complicated by adverse reactions to chemotherapy. Understanding key parasite molecules required for chronic infection provides new insights into potential mechanisms that can interrupt parasite survival or persistence in the host. This study reveals that key secreted rhoptry molecules are used by the parasite to establish chronic infection of the host. Certain rhoptry proteins were found to be critical virulence factors that resist innate immunity, while other rhoptry proteins were found to influence chronic infection without affecting virulence. This study reveals that rhoptry proteins utilize multiple mechanisms of host manipulation to establish chronic infection of the host. Targeted disruption of parasite rhoptry proteins involved in these biological processes opens new avenues to interfere with chronic infection with the goal to either eliminate chronic infection or to prevent recrudescent infections.
Collapse
|
40
|
Hoeksema MA, Laan LC, Postma JJ, Cummings RD, de Winther MPJ, Dijkstra CD, van Die I, Kooij G. Treatment with Trichuris suis soluble products during monocyte-to-macrophage differentiation reduces inflammatory responses through epigenetic remodeling. FASEB J 2016; 30:2826-36. [PMID: 27095802 DOI: 10.1096/fj.201600343r] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 04/12/2016] [Indexed: 01/09/2023]
Abstract
Helminths have strong immunoregulatory properties that may be exploited in treatment of chronic immune disorders, such as multiple sclerosis and inflammatory bowel disease. Essential players in the pathogenesis of these diseases are proinflammatory macrophages. We present evidence that helminths modulate the function and phenotype of these innate immune cells. We found that soluble products derived from the Trichuris suis (TsSP) significantly affect the differentiation of monocytes into macrophages and their subsequent polarization. TsSPs reduce the expression and production of inflammatory cytokines, including IL-6 and TNF, in human proinflammatory M1 macrophages. TsSPs induce a concomitant anti-inflammatory M2 signature, with increased IL-10 production. Furthermore, they suppress CHIT activity and enhance secretion of matrix metalloproteinase 9. Short-term triggering of monocytes with TsSPs early during monocyte-to-macrophage differentiation imprinted these phenotypic alterations, suggesting long-lasting epigenetic changes. The TsSP-induced effects in M1 macrophages were completely reversed by inhibiting histone deacetylases, which corresponded with decreased histone acetylation at the TNF and IL6 promoters. These results demonstrate that TsSPs have a potent and sustained immunomodulatory effect on human macrophage differentiation and polarization through epigenetic remodeling and provide new insights into the mechanisms by which helminths modulate human immune responses.-Hoeksema, M. A., Laan, L. C., Postma, J. J., Cummings, R. D., de Winther, M. P. J., Dijkstra, C. D., van Die, I., Kooij, G. Treatment with Trichuris suis soluble products during monocyte-to-macrophage differentiation reduces inflammatory responses through epigenetic remodeling.
Collapse
Affiliation(s)
- Marten A Hoeksema
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Lisa C Laan
- Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Juliette J Postma
- Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Richard D Cummings
- National Center for Functional Glycomics, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Menno P J de Winther
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Christine D Dijkstra
- Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Irma van Die
- Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; and
| | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; and
| |
Collapse
|
41
|
Möhle L, Israel N, Paarmann K, Krohn M, Pietkiewicz S, Müller A, Lavrik IN, Buguliskis JS, Schott BH, Schlüter D, Gundelfinger ED, Montag D, Seifert U, Pahnke J, Dunay IR. Chronic Toxoplasma gondii infection enhances β-amyloid phagocytosis and clearance by recruited monocytes. Acta Neuropathol Commun 2016; 4:25. [PMID: 26984535 PMCID: PMC4793516 DOI: 10.1186/s40478-016-0293-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 02/19/2016] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION Alzheimer's disease (AD) is associated with the accumulation of β-amyloid (Aβ) as senile plaques in the brain, thus leading to neurodegeneration and cognitive impairment. Plaque formation depends not merely on the amount of generated Aβ peptides, but more importantly on their effective removal. Chronic infections with neurotropic pathogens, most prominently the parasite Toxoplasma (T.) gondii, are frequent in the elderly, and it has been suggested that the resulting neuroinflammation may influence the course of AD. In the present study, we investigated how chronic T. gondii infection and resulting neuroinflammation affect plaque deposition and removal in a mouse model of AD. RESULTS Chronic infection with T. gondii was associated with reduced Aβ and plaque load in 5xFAD mice. Upon infection, myeloid-derived CCR2(hi) Ly6C(hi) monocytes, CCR2(+) Ly6C(int), and CCR2(+) Ly6C(low) mononuclear cells were recruited to the brain of mice. Compared to microglia, these recruited mononuclear cells showed highly increased phagocytic capacity of Aβ ex vivo. The F4/80(+) Ly6C(low) macrophages expressed high levels of Triggering Receptor Expressed on Myeloid cells 2 (TREM2), CD36, and Scavenger Receptor A1 (SCARA1), indicating phagocytic activity. Importantly, selective ablation of CCR2(+) Ly6C(hi) monocytes resulted in an increased amount of Aβ in infected mice. Elevated insulin-degrading enzyme (IDE), matrix metalloproteinase 9 (MMP9), as well as immunoproteasome subunits β1i/LMP2, β2i/MECL-1, and β5i/LMP7 mRNA levels in the infected brains indicated increased proteolytic Aβ degradation. Particularly, LMP7 was highly expressed by the recruited mononuclear cells in the brain, suggesting a novel mechanism of Aβ clearance. CONCLUSIONS Our results indicate that chronic Toxoplasma infection ameliorates β-amyloidosis in a murine model of AD by activation of the immune system, specifically by recruitment of Ly6C(hi) monocytes and by enhancement of phagocytosis and degradation of soluble Aβ. Our findings provide evidence for a modulatory role of inflammation-induced Aβ phagocytosis and degradation by newly recruited peripheral immune cells in the pathophysiology of AD.
Collapse
Affiliation(s)
- Luisa Möhle
- Institute for Medical Microbiology and Hospital Hygiene, University of Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Nicole Israel
- Institute for Molecular and Clinical Immunology, University of Magdeburg, Magdeburg, Germany
| | - Kristin Paarmann
- Department of Pathology (PAT), Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Neurogenetics, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Markus Krohn
- Department of Pathology (PAT), Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway
| | - Sabine Pietkiewicz
- Department of Translational Inflammation Research, Institute of Experimental Internal Medicine, University of Magdeburg, Magdeburg, Germany
| | - Andreas Müller
- Institute for Molecular and Clinical Immunology, University of Magdeburg, Magdeburg, Germany
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Inna N Lavrik
- Department of Translational Inflammation Research, Institute of Experimental Internal Medicine, University of Magdeburg, Magdeburg, Germany
| | | | - Björn H Schott
- Center for Behavioral Brain Sciences (CBBS), University of Magdeburg, Magdeburg, Germany
- Department of Behavioral Neurology, Leibniz Institute for Neurobiology, Magdeburg, Germany
- Department of Psychiatry and Psychotherapy, Campus Mitte, Charité Universitätsmedizin, Berlin, Germany
| | - Dirk Schlüter
- Institute for Medical Microbiology and Hospital Hygiene, University of Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), University of Magdeburg, Magdeburg, Germany
| | - Eckart D Gundelfinger
- Center for Behavioral Brain Sciences (CBBS), University of Magdeburg, Magdeburg, Germany
- Department of Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology, Magdeburg, Germany
- Medical Faculty, University of Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Dirk Montag
- Neurogenetics, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Ulrike Seifert
- Institute for Molecular and Clinical Immunology, University of Magdeburg, Magdeburg, Germany
| | - Jens Pahnke
- Department of Pathology (PAT), Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway
- University of Lübeck (UzL), LIED, Lübeck, Germany
- Leibniz Institute of Plant Biochemistry (IPB), Halle, Germany
| | - Ildiko Rita Dunay
- Institute for Medical Microbiology and Hospital Hygiene, University of Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.
- Center for Behavioral Brain Sciences (CBBS), University of Magdeburg, Magdeburg, Germany.
| |
Collapse
|
42
|
Obligate bacterial mutualists evolving from environmental bacteria in natural insect populations. Nat Microbiol 2016; 1:15011. [PMID: 27571756 DOI: 10.1038/nmicrobiol.2015.11] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 10/30/2015] [Indexed: 11/09/2022]
Abstract
Diverse organisms are associated with obligate microbial mutualists. How such essential symbionts have originated from free-living ancestors is of evolutionary interest. Here we report that, in natural populations of the stinkbug Plautia stali, obligate bacterial mutualists are evolving from environmental bacteria. Of six distinct bacterial lineages associated with insect populations, two are uncultivable with reduced genomes, four are cultivable with non-reduced genomes, one uncultivable symbiont is fixed in temperate populations, and the other uncultivable symbiont coexists with four cultivable symbionts in subtropical populations. Symbiont elimination resulted in host mortality for all symbionts, while re-infection with any of the symbionts restored normal host growth, indicating that all the symbionts are indispensable and almost equivalent functionally. Some aseptic newborns incubated with environmental soils acquired the cultivable symbionts and normal growth was restored, identifying them as environmental Pantoea spp. Our finding uncovers an evolutionary transition from a free-living lifestyle to obligate mutualism that is currently ongoing in nature.
Collapse
|
43
|
Almeida F, Sardinha-Silva A, da Silva TA, Pessoni AM, Pinzan CF, Alegre-Maller ACP, Cecílio NT, Moretti NS, Damásio ARL, Pedersoli WR, Mineo JR, Silva RN, Roque-Barreira MC. Toxoplasma gondii Chitinase Induces Macrophage Activation. PLoS One 2015; 10:e0144507. [PMID: 26659253 PMCID: PMC4684212 DOI: 10.1371/journal.pone.0144507] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 11/19/2015] [Indexed: 01/06/2023] Open
Abstract
Toxoplasma gondii is an obligate intracellular protozoan parasite found worldwide that is able to chronically infect almost all vertebrate species, especially birds and mammalians. Chitinases are essential to various biological processes, and some pathogens rely on chitinases for successful parasitization. Here, we purified and characterized a chitinase from T. gondii. The enzyme, provisionally named Tg_chitinase, has a molecular mass of 13.7 kDa and exhibits a Km of 0.34 mM and a Vmax of 2.64. The optimal environmental conditions for enzymatic function were at pH 4.0 and 50 °C. Tg_chitinase was immunolocalized in the cytoplasm of highly virulent T. gondii RH strain tachyzoites, mainly at the apical extremity. Tg_chitinase induced macrophage activation as manifested by the production of high levels of pro-inflammatory cytokines, a pathogenic hallmark of T. gondii infection. In conclusion, to our knowledge, we describe for the first time a chitinase of T. gondii tachyzoites and provide evidence that this enzyme might influence the pathogenesis of T. gondii infection.
Collapse
Affiliation(s)
- Fausto Almeida
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP, 14049-900, Brasil
| | - Aline Sardinha-Silva
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP, 14049-900, Brasil
| | - Thiago Aparecido da Silva
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP, 14049-900, Brasil
| | - André Moreira Pessoni
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP, 14049-900, Brasil
| | - Camila Figueiredo Pinzan
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP, 14049-900, Brasil
| | - Ana Claudia Paiva Alegre-Maller
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP, 14049-900, Brasil
| | - Nerry Tatiana Cecílio
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP, 14049-900, Brasil
| | - Nilmar Silvio Moretti
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Sao Paulo, São Paulo, SP, Brasil
| | - André Ricardo Lima Damásio
- Departamento de Bioquímica e Biologia Tecidual, Instituto de Biologia, Universidade de Campinas, Campinas, SP, Brasil
| | - Wellington Ramos Pedersoli
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP, 14040-900, Brasil
| | - José Roberto Mineo
- Laboratorio de Imunoparasitologia, Departamento de Imunologia, Microbiologia e Parasitologia, Universidade Federal de Uberlândia, Av. Pará, 1720, Uberlândia, MG, 38400 902, Brasil
| | - Roberto Nascimento Silva
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP, 14040-900, Brasil
| | - Maria Cristina Roque-Barreira
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Av. Bandeirantes 3900, Ribeirão Preto, SP, 14049-900, Brasil
| |
Collapse
|
44
|
Abstract
Bradyzoite forms of Toxoplasma gondii persist in tissue cysts for the lifetime of an infected host and can reactivate to cause clinical disease. It was thought that in vivo bradyzoites within tissue cysts are biologically inactive dormant forms that rarely replicate. Apparently, consensus was wrong.
Collapse
Affiliation(s)
- Kami Kim
- Departments of Medicine, Microbiology & Immunology, and Pathology, Albert Einstein College of Medicine, Bronx, New York, USA.
| |
Collapse
|
45
|
Novel Approaches Reveal that Toxoplasma gondii Bradyzoites within Tissue Cysts Are Dynamic and Replicating Entities In Vivo. mBio 2015; 6:e01155-15. [PMID: 26350965 PMCID: PMC4600105 DOI: 10.1128/mbio.01155-15] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Despite their critical role in chronic toxoplasmosis, the biology of Toxoplasma gondii bradyzoites is poorly understood. In an attempt to address this gap, we optimized approaches to purify tissue cysts and analyzed the replicative potential of bradyzoites within these cysts. In order to quantify individual bradyzoites within tissue cysts, we have developed imaging software, BradyCount 1.0, that allows the rapid establishment of bradyzoite burdens within imaged optical sections of purified tissue cysts. While in general larger tissue cysts contain more bradyzoites, their relative “occupancy” was typically lower than that of smaller cysts, resulting in a lower packing density. The packing density permits a direct measure of how bradyzoites develop within cysts, allowing for comparisons across progression of the chronic phase. In order to capture bradyzoite endodyogeny, we exploited the differential intensity of TgIMC3, an inner membrane complex protein that intensely labels newly formed/forming daughters within bradyzoites and decays over time in the absence of further division. To our surprise, we were able to capture not only sporadic and asynchronous division but also synchronous replication of all bradyzoites within mature tissue cysts. Furthermore, the time-dependent decay of TgIMC3 intensity was exploited to gain insights into the temporal patterns of bradyzoite replication in vivo. Despite the fact that bradyzoites are considered replicatively dormant, we find evidence for cyclical, episodic bradyzoite growth within tissue cysts in vivo. These findings directly challenge the prevailing notion of bradyzoites as dormant nonreplicative entities in chronic toxoplasmosis and have implications on our understanding of this enigmatic and clinically important life cycle stage. The protozoan Toxoplasma gondii establishes a lifelong chronic infection mediated by the bradyzoite form of the parasite within tissue cysts. Technical challenges have limited even the most basic studies on bradyzoites and the tissue cysts in vivo. Bradyzoites, which are viewed as dormant, poorly replicating or nonreplicating entities, were found to be surprisingly active, exhibiting not only the capacity for growth but also previously unrecognized patterns of replication that point to their being considerably more dynamic than previously imagined. These newly revealed properties force us to reexamine the most basic questions regarding bradyzoite biology and the progression of the chronic phase of toxoplasmosis. By developing new tools and approaches to study the chronic phase at the level of bradyzoites, we expose new avenues to tackle both drug development and a better understanding of events that may lead to reactivated symptomatic disease.
Collapse
|
46
|
Rückerl D, Allen JE. Macrophage proliferation, provenance, and plasticity in macroparasite infection. Immunol Rev 2015; 262:113-33. [PMID: 25319331 PMCID: PMC4324133 DOI: 10.1111/imr.12221] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Macrophages have long been center stage in the host response to microbial infection, but only in the past 10–15 years has there been a growing appreciation for their role in helminth infection and the associated type 2 response. Through the actions of the IL-4 receptor α (IL-4Rα), type 2 cytokines result in the accumulation of macrophages with a distinctive activation phenotype. Although our knowledge of IL-4Rα-induced genes is growing rapidly, the specific functions of these macrophages have yet to be established in most disease settings. Understanding the interplay between IL-4Rα-activated macrophages and the other cellular players is confounded by the enormous transcriptional heterogeneity within the macrophage population and by their highly plastic nature. Another level of complexity is added by the new knowledge that tissue macrophages can be derived either from a resident prenatal population or from blood monocyte recruitment and that IL-4 can increase macrophage numbers through proliferative expansion. Here, we review current knowledge on the contribution of macrophages to helminth killing and wound repair, with specific attention paid to distinct cellular origins and plasticity potential.
Collapse
Affiliation(s)
- Dominik Rückerl
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
47
|
Luu L, Coombes JL. Dynamic two-photon imaging of the immune response to Toxoplasma gondii infection. Parasite Immunol 2015; 37:118-26. [PMID: 25407960 DOI: 10.1111/pim.12161] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 11/07/2014] [Indexed: 12/14/2022]
Abstract
Toxoplasma gondii is a highly successful parasite that can manipulate host immune responses to optimize its persistence and spread. As a result, a highly complex relationship exists between T. gondii and the immune system of the host. Advances in imaging techniques, and in particular, the application of two-photon microscopy to mouse infection models, have made it possible to directly visualize interactions between parasites and the host immune system as they occur in living tissues. Here, we will discuss how dynamic imaging techniques have provided unexpected new insight into (i) how immune responses are dynamically regulated by cells and structures in the local tissue environment, (ii) how protective responses to T. gondii are generated and (iii) how the parasite exploits the immune system for its own benefit.
Collapse
Affiliation(s)
- L Luu
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | | |
Collapse
|
48
|
Blanchard N, Dunay IR, Schlüter D. Persistence of Toxoplasma gondii in the central nervous system: a fine-tuned balance between the parasite, the brain and the immune system. Parasite Immunol 2015; 37:150-8. [PMID: 25573476 DOI: 10.1111/pim.12173] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 12/30/2014] [Indexed: 02/04/2023]
Abstract
Upon infection of humans and animals with Toxoplasma gondii, the parasites persist as intraneuronal cysts that are controlled, but not eliminated by the immune system. In particular, intracerebral T cells are crucial in the control of T. gondii infection and are supported by essential functions from other leukocyte populations. Additionally, brain-resident cells including astrocytes, microglia and neurons contribute to the intracerebral immune response by the production of cytokines, chemokines and expression of immunoregulatory cell surface molecules, such as major histocompatibility (MHC) antigens. However, the in vivo behaviour of these individual cell populations, specifically their interaction during cerebral toxoplasmosis, remains to be elucidated. We discuss here what is known about the function of T cells, recruited myeloid cells and brain-resident cells, with particular emphasis on the potential cross-regulation of these cell populations, in governing cerebral toxoplasmosis.
Collapse
Affiliation(s)
- N Blanchard
- Inserm U1043, Toulouse, France; CNRS U5282, Toulouse, France; Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, UPS, Toulouse, France
| | | | | |
Collapse
|
49
|
Landrith TA, Harris TH, Wilson EH. Characteristics and critical function of CD8+ T cells in the Toxoplasma-infected brain. Semin Immunopathol 2015; 37:261-70. [PMID: 25898888 DOI: 10.1007/s00281-015-0487-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 03/23/2015] [Indexed: 12/13/2022]
Abstract
The rise of the AIDS epidemic made the requirement for T cells in our continuous protection from pathogens critically apparent. The striking frequency with which AIDS patients exhibited profound neurological pathologies brought attention to many chronic infections that are latent within the immune-privileged CNS. One of the most common lethal opportunistic infections of these patients was with the protozoan parasite, Toxoplasma gondii. Reactivation of Toxoplasma cysts within the brain causes massive tissue destruction evidenced as multiple ring-enhancing lesions on MRI and is called toxoplasmic encephalitis (TE). TE is not limited to AIDS patients, but rather is a risk for all severely immunocompromised patients, including recipients of chemotherapy or transplant recipients. The lessons learned from these patient populations are supported by T cell depletion studies in mice. Such experiments have demonstrated that CD4+ and CD8+ T cells are required for protection against TE. Although it is clear that these T cell subsets work synergistically to fight infection, much evidence has been generated that suggests CD8+ T cells play a dominant role in protection during chronic toxoplasmosis. In other models of CNS inflammation, such as intracerebral infection with LCMV and experimental autoimmune encephalomyelitis (EAE), infiltration of T cells into the brain is harmful and even fatal. In the brain of the immunocompetent host, the well-regulated T cell response to T. gondii is therefore an ideal model to understand a controlled inflammatory response to CNS infection. This review will examine our current understanding of CD8+ T cells in the CNS during T. gondii infection in regards to the (1) mechanisms governing entry into the brain, (2) cues that dictate behavior within the brain, and (3) the functional and phenotypic properties exhibited by these cells.
Collapse
Affiliation(s)
- Tyler A Landrith
- Division of Biomedical Sciences, University of California, Riverside, CA, 92521, USA
| | | | | |
Collapse
|
50
|
Koch BEV, Stougaard J, Spaink HP. Keeping track of the growing number of biological functions of chitin and its interaction partners in biomedical research. Glycobiology 2015; 25:469-82. [PMID: 25595947 PMCID: PMC4373397 DOI: 10.1093/glycob/cwv005] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Chitin is a vital polysaccharide component of protective structures in many eukaryotic organisms but seems absent in vertebrates. Chitin or chitin oligomers are therefore prime candidates for non-self-molecules, which are recognized and degraded by the vertebrate immune system. Despite the absence of polymeric chitin in vertebrates, chitinases and chitinase-like proteins (CLPs) are well conserved in vertebrate species. In many studies, these proteins have been found to be involved in immune regulation and in mediating the degradation of chitinous external protective structures of invading pathogens. Several important aspects of chitin immunostimulation have recently been uncovered, advancing our understanding of the complex regulatory mechanisms that chitin mediates. Likewise, the last few years have seen large advances in our understanding of the mechanisms and molecular interactions of chitinases and CLPs in relation to immune response regulation. It is becoming increasingly clear that their function in this context is not exclusive to chitin producing pathogens, but includes bacterial infections and cancer signaling as well. Here we provide an overview of the immune signaling properties of chitin and other closely related biomolecules. We also review the latest literature on chitinases and CLPs of the GH18 family. Finally, we examine the existing literature on zebrafish chitinases, and propose the use of zebrafish as a versatile model to complement the existing murine models. This could especially be of benefit to the exploration of the function of chitinases in infectious diseases using high-throughput approaches and pharmaceutical interventions.
Collapse
Affiliation(s)
- Bjørn E V Koch
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark Leiden University, Institute of Biology, Leiden, The Netherlands
| | - Jens Stougaard
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Herman P Spaink
- Leiden University, Institute of Biology, Leiden, The Netherlands
| |
Collapse
|