1
|
Ortiz-Sánchez P, González-Soto S, Villamizar LH, Valencia J, Jiménez E, Sacedón R, Ramírez M, Fariñas I, Varas A, Fernández-Sevilla LM, Vicente Á. Meningeal leukaemic aggregates as foci of cell expansion and chemoresistance in acute lymphoblastic leukaemia metastasis. Cell Oncol (Dordr) 2025; 48:725-741. [PMID: 39937211 DOI: 10.1007/s13402-025-01043-y] [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] [Accepted: 01/30/2025] [Indexed: 02/13/2025] Open
Abstract
PURPOSE Central nervous system (CNS) involvement and/or relapse remains one of the most important causes of morbidity/mortality in paediatric B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) patients. To identify novel therapeutic targets and develop less aggressive therapies, a better understanding of the cellular and molecular microenvironment in leptomeningeal metastases is key. Here, we aimed to investigate the formation of metastatic leptomeningeal aggregates and their relevance to the expansion, survival and chemoresistance acquisition of leukaemia cells. METHODS We used BCP-ALL xenograft mouse models, combined with immunohistofluorescence and flow cytometry, to study the development of CNS metastasis and the contribution of leptomeningeal cells to the organisation of leukaemic aggregates. To in vitro mimic the CNS metastasis, we established co-cultures of three-dimensional (3D) ALL cell spheroids and human leptomeningeal cells (hLMCs) and studied the effects on gene expression, proliferation, cytokine production, and chemoresistance. RESULTS In xenografted mice, ALL cells infiltrated the CNS at an early stage and, after crossing an ER-TR7+ fibroblast-like meningeal cell layer, they proliferated extensively and formed large vascularised leukaemic aggregates supported by a network of podoplanin+ leptomeningeal cells. In leukaemia spheroid-hLMC co-cultures, unlike conventional 2D co-cultures, meningeal cells strongly promoted the proliferation of leukaemic cells and generated a pro-inflammatory microenvironment. Furthermore, in 3D cell aggregates, leukaemic cells also developed chemoresistance, at least in part due to ABC transporter up-regulation. CONCLUSION Our results provide evidence for the formation of metastatic ALL-leptomeningeal cell aggregates, their pro-inflammatory profile and their contribution to leukaemic cell expansion, survival and chemoresistance in the CNS.
Collapse
Affiliation(s)
- Paula Ortiz-Sánchez
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Sara González-Soto
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
- Health Research Institute Hospital 12 de Octubre (Imas12), Madrid, Spain
| | - Luz H Villamizar
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Jaris Valencia
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
- Health Research Institute Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Eva Jiménez
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
- Health Research Institute Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Rosa Sacedón
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
- Health Research Institute Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Manuel Ramírez
- Department of Paediatric Haematology and Oncology, Advanced Therapies Unit, Niño Jesús University Children's Hospital, Madrid, Spain
- Health Research Institute Hospital La Princesa, Madrid, Spain
| | - Isabel Fariñas
- Biomedical Research Network on Neurodegenerative Diseases (CIBERNED), Department of Cell Biology and Biotechnology and Biomedicine Institute (BioTecMed), University of Valencia, Valencia, Spain
| | - Alberto Varas
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain.
- Health Research Institute Hospital Clínico San Carlos (IdISSC), Madrid, Spain.
| | - Lidia M Fernández-Sevilla
- Health Research Institute Hospital Clínico San Carlos (IdISSC), Madrid, Spain.
- Department of Basic Health Sciences, Faculty of Health Sciences, University Rey Juan Carlos, Alcorcón, Spain.
| | - Ángeles Vicente
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain.
- Health Research Institute Hospital 12 de Octubre (Imas12), Madrid, Spain.
| |
Collapse
|
2
|
Eberhard JN, Shallberg LA, Winn A, Chandrasekaran S, Giuliano CJ, Merritt EF, Willis E, Konradt C, Christian DA, Aldridge DL, Bunkofske ME, Jacquet M, Dzierszinski F, Katifori E, Lourido S, Koshy AA, Hunter CA. Immune targeting and host-protective effects of the latent stage of Toxoplasma gondii. Nat Microbiol 2025; 10:992-1005. [PMID: 40148566 PMCID: PMC11964939 DOI: 10.1038/s41564-025-01967-z] [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: 08/18/2022] [Accepted: 02/19/2025] [Indexed: 03/29/2025]
Abstract
Latency is a microbial strategy for persistence. For Toxoplasma gondii the bradyzoite stage forms long-lived cysts critical for transmission, and its presence in neurons is considered important for immune evasion. However, the extent to which cyst formation escapes immune pressure and mediates persistence remained unclear. Here we developed a mathematical model highlighting that bradyzoite-directed immunity contributes to control of cyst numbers. In vivo studies demonstrated that transgenic CD8+ T cells recognized a cyst-derived antigen, and neuronal STAT1 signalling promoted cyst control in mice. Modelling and experiments with parasites unable to form bradyzoites (Δbfd1) revealed that the absence of cyst formation in the central nervous system did not prevent long-term persistence but resulted in increased tachyzoite replication with associated tissue damage and mortality. These findings suggest the latent form of T. gondii is under immune pressure, mitigates infection-induced damage and promotes survival of host and parasite.
Collapse
Affiliation(s)
- Julia N Eberhard
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lindsey A Shallberg
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aaron Winn
- Department of Physics and Astronomy, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Christopher J Giuliano
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emily F Merritt
- Department of Immunology, University of Arizona, Tucson, AZ, USA
| | - Elinor Willis
- Comparative Pathology Core, Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christoph Konradt
- Department of Comparative Pathobiology, Purdue University College of Veterinary Medicine, West Lafayette, IN, USA
| | - David A Christian
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel L Aldridge
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Molly E Bunkofske
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maxime Jacquet
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Florence Dzierszinski
- The Royal Ottawa Mental Health Center, Institute of Mental Health Research, Ottawa, Ontario, Canada
| | - Eleni Katifori
- Department of Physics and Astronomy, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Center for Computational Biology, Flatiron Institute, New York, NY, USA
| | - Sebastian Lourido
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anita A Koshy
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- Department of Immunology, University of Arizona, Tucson, AZ, USA
- Department of Neurology, University of Arizona, Tucson, AZ, USA
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Bonnet V, Maikranz E, Madec M, Vertti-Quintero N, Cuche C, Mastrogiovanni M, Alcover A, Di Bartolo V, Baroud CN. Cancer-on-a-chip model shows that the adenomatous polyposis coli mutation impairs T cell engagement and killing of cancer spheroids. Proc Natl Acad Sci U S A 2024; 121:e2316500121. [PMID: 38442157 PMCID: PMC10945811 DOI: 10.1073/pnas.2316500121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 01/25/2024] [Indexed: 03/07/2024] Open
Abstract
Evaluating the ability of cytotoxic T lymphocytes (CTLs) to eliminate tumor cells is crucial, for instance, to predict the efficiency of cell therapy in personalized medicine. However, the destruction of a tumor by CTLs involves CTL migration in the extra-tumoral environment, accumulation on the tumor, antigen recognition, and cooperation in killing the cancer cells. Therefore, identifying the limiting steps in this complex process requires spatio-temporal measurements of different cellular events over long periods. Here, we use a cancer-on-a-chip platform to evaluate the impact of adenomatous polyposis coli (APC) mutation on CTL migration and cytotoxicity against 3D tumor spheroids. The APC mutated CTLs are found to have a reduced ability to destroy tumor spheroids compared with control cells, even though APC mutants migrate in the extra-tumoral space and accumulate on the spheroids as efficiently as control cells. Once in contact with the tumor however, mutated CTLs display reduced engagement with the cancer cells, as measured by a metric that distinguishes different modes of CTL migration. Realigning the CTL trajectories around localized killing cascades reveals that all CTLs transition to high engagement in the 2 h preceding the cascades, which confirms that the low engagement is the cause of reduced cytotoxicity. Beyond the study of APC mutations, this platform offers a robust way to compare cytotoxic cell efficiency of even closely related cell types, by relying on a multiscale cytometry approach to disentangle complex interactions and to identify the steps that limit the tumor destruction.
Collapse
Affiliation(s)
- Valentin Bonnet
- Institut Pasteur, Department of Genomes and Genetics, Université Paris Cité, Physical Microfluidics and Bioengineering, ParisF-75015, France
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau91120, France
| | - Erik Maikranz
- Institut Pasteur, Department of Genomes and Genetics, Université Paris Cité, Physical Microfluidics and Bioengineering, ParisF-75015, France
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau91120, France
| | - Marianne Madec
- Unité Biologie Cellulaire des Lymphocytes, Institut Pasteur, Department of immunology, Université Paris Cité, INSERM-U1224, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, ParisF-75015, France
- Faculty of Medicine, Department of Pathology and Immunology, University of Geneva, Geneva 4CH-1211, Switzerland
| | - Nadia Vertti-Quintero
- Institut Pasteur, Department of Genomes and Genetics, Université Paris Cité, Physical Microfluidics and Bioengineering, ParisF-75015, France
| | - Céline Cuche
- Unité Biologie Cellulaire des Lymphocytes, Institut Pasteur, Department of immunology, Université Paris Cité, INSERM-U1224, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, ParisF-75015, France
| | - Marta Mastrogiovanni
- Unité Biologie Cellulaire des Lymphocytes, Institut Pasteur, Department of immunology, Université Paris Cité, INSERM-U1224, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, ParisF-75015, France
- Albert Einstein College of Medicine, Department of Developmental and Molecular Biology, New York, NY10461
| | - Andrés Alcover
- Unité Biologie Cellulaire des Lymphocytes, Institut Pasteur, Department of immunology, Université Paris Cité, INSERM-U1224, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, ParisF-75015, France
| | - Vincenzo Di Bartolo
- Unité Biologie Cellulaire des Lymphocytes, Institut Pasteur, Department of immunology, Université Paris Cité, INSERM-U1224, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, ParisF-75015, France
| | - Charles N. Baroud
- Institut Pasteur, Department of Genomes and Genetics, Université Paris Cité, Physical Microfluidics and Bioengineering, ParisF-75015, France
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau91120, France
| |
Collapse
|
4
|
Bolini L, Campos RMP, Spiess DA, Lima-Rosa FL, Dantas DP, Conde L, Mendez-Otero R, Vale AM, Pimentel-Coelho PM. Long-term recruitment of peripheral immune cells to brain scars after a neonatal insult. Glia 2024; 72:546-567. [PMID: 37987116 DOI: 10.1002/glia.24490] [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: 06/15/2023] [Revised: 10/23/2023] [Accepted: 10/31/2023] [Indexed: 11/22/2023]
Abstract
Although brain scars in adults have been extensively studied, there is less data available regarding scar formation during the neonatal period, and the involvement of peripheral immune cells in this process remains unexplored in neonates. Using a murine model of neonatal hypoxic-ischemic encephalopathy (HIE) and confocal microscopy, we characterized the scarring process and examined the recruitment of peripheral immune cells to cortical and hippocampal scars for up to 1 year post-insult. Regional differences in scar formation were observed, including the presence of reticular fibrotic networks in the cortex and perivascular fibrosis in the hippocampus. We identified chemokines with chronically elevated levels in both regions and demonstrated, through a parabiosis-based strategy, the recruitment of lymphocytes, neutrophils, and monocyte-derived macrophages to the scars several weeks after the neonatal insult. After 1 year, however, neutrophils and lymphocytes were absent from the scars. Our data indicate that peripheral immune cells are transient components of HIE-induced brain scars, opening up new possibilities for late therapeutic interventions.
Collapse
Affiliation(s)
- Lukas Bolini
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Daiane Aparecida Spiess
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Frederico Luis Lima-Rosa
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danillo Pereira Dantas
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Conde
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rosalia Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andre M Vale
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | |
Collapse
|
5
|
Vizcarra EA, Ulu A, Landrith TA, Qiu X, Godzik A, Wilson EH. Group 1 metabotropic glutamate receptor expression defines a T cell memory population during chronic Toxoplasma infection that enhances IFN-gamma and perforin production in the CNS. Brain Behav Immun 2023; 114:131-143. [PMID: 37604212 DOI: 10.1016/j.bbi.2023.08.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 08/23/2023] Open
Abstract
Within the brain, a pro-inflammatory response is essential to prevent clinical disease due to Toxoplasma gondii reactivation. Infection in the immunocompromised leads to lethal Toxoplasmic encephalitis while in the immunocompetent, there is persistent low-grade inflammation which is devoid of clinical symptoms. This signifies that there is a well-balanced and regulated inflammatory response to T. gondii in the brain. T cells are the dominant immune cells that prevent clinical disease, and this is mediated through the secretion of effector molecules such as perforins and IFN-γ. The presence of cognate antigen, the expression of survival cytokines, and the alteration of the epigenetic landscape drive the development of memory T cells. However, specific extrinsic signals that promote the formation and maintenance of memory T cells within tissue are poorly understood. During chronic infection, there is an increase in extracellular glutamate that, due to its function as an excitatory neurotransmitter, is normally tightly controlled in the CNS. Here we demonstrate that CD8+ T cells from the T. gondii-infected brain parenchyma are enriched for metabotropic glutamate receptors (mGluR's). Characterization studies determined that mGluR+ expression by CD8+ T cells defines a distinct memory population at the transcriptional and protein level. Finally, using receptor antagonists and agonists we demonstrate mGluR signaling is required for optimal CD8+ T cell production of the effector cytokine IFNγ. This work suggests that glutamate is an important environmental signal of inflammation that promotes T cell function. Understanding glutamate's influence on T cells in the brain can provide insights into the mechanisms that govern protective immunity against CNS-infiltrating pathogens and neuroinflammation.
Collapse
Affiliation(s)
- Edward A Vizcarra
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States
| | - Arzu Ulu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States
| | - Tyler A Landrith
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States
| | - Xinru Qiu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States
| | - Adam Godzik
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States
| | - Emma H Wilson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, United States.
| |
Collapse
|
6
|
S Subauste C, Hubal A. Animal Models for Toxoplasma gondii Infection. Curr Protoc 2023; 3:e871. [PMID: 37695167 PMCID: PMC10621533 DOI: 10.1002/cpz1.871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Toxoplasma gondii is an obligate intracellular protozoan parasite that commonly infects mammals and birds throughout the world. This protocol describes murine models of acute T. gondii infection, toxoplasmic encephalitis and toxoplasma retinochoroiditis. T. gondii infection in severe combined immunodeficient (SCID) mice, deficient in T and B cells, has allowed for the study of T cell-independent mechanisms of defense against intracellular organisms, as described here. The uracil auxotroph strain cps1-1 and temperature-sensitive mutant strains of T. gondii induce protection against challenge with virulent strains of the parasite. They have allowed studies of immunization and adoptive-transfer experiments. A protocol is provided for infection with these mutant strains. The EGS strain of T. gondii has the unique feature of spontaneously forming tissue cysts in cell culture. Dual fluorescent reporter stains of this strain have allowed the study of tachyzoite to bradyzoite transitions in vitro and in vivo. A protocol for in vitro and in vivo growth of this strain and tissue cyst isolation is provided. Genetic manipulation of T. gondii and mice has led to the development of parasites that express fluorescent proteins as well as mice with fluorescently labeled leukocytes. This together with the use of T. gondii that express model antigens and transgenic mice that express the appropriate T cell receptor have facilitated the in vivo study of parasite host-interaction. In addition, parasites that express bioluminescent markers have made it possible to study the dynamics of infection in real time using bioluminescence imaging. Support protocols present methodology for evaluation of progression of infection and immune response to the parasite that includes these newer methodologies. In addition, support protocols address the maintenance of T. gondii tissue cysts and tachyzoites, as well as preparation of T. gondii lysate antigens. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Induction of acute T. gondii infection in mice Basic Protocol 2: Model of toxoplasmic encephalitis and toxoplasma retinochoroiditis in chronically infected mice Basic Protocol 3: Assessment of T. gondii invasion into neural tissue Basic Protocol 4: T. gondii infection in scid/scid (SCID) mice Basic Protocol 5: Infection with the uracil auxotroph strain CPS1-1 or the temperature-sensitive TS-4 strain of T. gondii Basic Protocol 6: In vivo and in vitro maintenance of the EGS strain of T. gondii Support Protocol 1: Assessment of progression of infection and immune response to T. gondii Support Protocol 2: Maintenance of a bank of T. gondii cysts of the ME49 strain Support Protocol 3: Maintenance of T. gondii tachyzoites using human foreskin fibroblasts Support Protocol 4: Maintenance of T. gondii tachyzoites in mice Support Protocol 5: Preparation of T. gondii lysate antigens Support Protocol 6: Isolation of T. gondii tissue cysts from brain.
Collapse
Affiliation(s)
- Carlos S Subauste
- Case Western Reserve University, School of Medicine, Cleveland, Ohio, USA
| | - Alyssa Hubal
- Case Western Reserve University, School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
7
|
Yao PA, Sun HJ, Li XY. Identification of key genes in late-onset major depressive disorder through a co-expression network module. Front Genet 2022; 13:1048761. [PMID: 36561317 PMCID: PMC9763307 DOI: 10.3389/fgene.2022.1048761] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Late-onset major depressive disorder (LOD) increases the risk of disability and suicide in elderly patients. However, the complex pathological mechanism of LOD still remains unclear. We selected 10 LOD patients and 12 healthy control samples from the GSE76826 dataset for statistical analysis. Under the screening criteria, 811 differentially expressed genes (DEGs) were screened. We obtained a total of two most clinically significant modules through the weighted gene co-expression network analysis (WGCNA). Functional analysis of the genes in the most clinically significant modules was performed to explore the potential mechanism of LOD, followed by protein-protein interaction (PPI) analysis and hub gene identification in the core area of the PPI network. Furthermore, we identified immune infiltrating cells using the cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT) algorithm between healthy subjects and LOD patients with the GSE98793 dataset. Next, six hub genes (CD27, IL7R, CXCL1, CCR7, IGLL5, and CD79A) were obtained by intersecting hub genes with DEGs, followed by verifying the diagnostic accuracy with the receiver operating characteristic curve (ROC). In addition, we constructed the least absolute shrinkage and selection operator (LASSO) regression model for hub gene cross-validation. Finally, we found that CD27 and IGLL5 were good diagnostic indicators of LOD, and CD27 may be the key gene of immune function change in LOD. In conclusion, our research shows that the changes in the immune function may be an important mechanism in the development of LOD, which can provide some guidance for the related research of LOD in the future.
Collapse
Affiliation(s)
- Ping-An Yao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China,Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Hai-Ju Sun
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | - Xiao-Yu Li
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China,The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China,*Correspondence: Xiao-Yu Li,
| |
Collapse
|
8
|
T cell surveillance of Toxoplasma gondii: Basic insights into how T cells operate in the central nervous system. Curr Opin Neurobiol 2022; 77:102640. [PMID: 36240583 DOI: 10.1016/j.conb.2022.102640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 01/10/2023]
Abstract
The ability of T cells to operate in the central nervous system (CNS) is required for resistance to multiple pathogens that affect this tissue. The intracellular parasite Toxoplasma gondii has evolved to persist in the CNS and poses unique challenges to the immune system with the need to control parasite replication while balancing the adverse pathology associated with local inflammation. This article reviews the models used to study the response to T. gondii during toxoplasmic encephalitis and highlights some of the broader lessons that are relevant to understanding how T cells function in the CNS.
Collapse
|
9
|
Tu T, Peng Z, Song Z, Ma Y, Zhang H. New insight into DAVF pathology—Clues from meningeal immunity. Front Immunol 2022; 13:858924. [PMID: 36189220 PMCID: PMC9520480 DOI: 10.3389/fimmu.2022.858924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 08/26/2022] [Indexed: 11/13/2022] Open
Abstract
In recent years, with the current access in techniques, studies have significantly advanced the knowledge on meningeal immunity, revealing that the central nervous system (CNS) border acts as an immune landscape. The latest concept of meningeal immune system is a tertiary structure, which is a comprehensive overview of the meningeal immune system from macro to micro. We comprehensively reviewed recent advances in meningeal immunity, particularly the new understanding of the dural sinus and meningeal lymphatics. Moreover, based on the clues from the meningeal immunity, new insights were proposed into the dural arteriovenous fistula (DAVF) pathology, aiming to provide novel ideas for DAVF understanding.
Collapse
Affiliation(s)
- Tianqi Tu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhenghong Peng
- Department of Health Management Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zihao Song
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yongjie Ma
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, China
- *Correspondence: Yongjie Ma, ; Hongqi Zhang,
| | - Hongqi Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, China
- *Correspondence: Yongjie Ma, ; Hongqi Zhang,
| |
Collapse
|
10
|
Shallberg LA, Phan AT, Christian DA, Perry JA, Haskins BE, Beiting DP, Harris TH, Koshy AA, Hunter CA. Impact of secondary TCR engagement on the heterogeneity of pathogen-specific CD8+ T cell response during acute and chronic toxoplasmosis. PLoS Pathog 2022; 18:e1010296. [PMID: 35727849 PMCID: PMC9249239 DOI: 10.1371/journal.ppat.1010296] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 07/01/2022] [Accepted: 05/06/2022] [Indexed: 11/19/2022] Open
Abstract
Initial TCR engagement (priming) of naive CD8+ T cells results in T cell expansion, and these early events influence the generation of diverse effector and memory populations. During infection, activated T cells can re-encounter cognate antigen, but how these events influence local effector responses or formation of memory populations is unclear. To address this issue, OT-I T cells which express the Nur77-GFP reporter of TCR activation were paired with the parasite Toxoplasma gondii that expresses OVA to assess how secondary encounter with antigen influences CD8+ T cell responses. During acute infection, TCR stimulation in affected tissues correlated with parasite burden and was associated with markers of effector cells while Nur77-GFP- OT-I showed signs of effector memory potential. However, both Nur77-GFP- and Nur77-GFP+ OT-I from acutely infected mice formed similar memory populations when transferred into naive mice. During the chronic stage of infection in the CNS, TCR activation was associated with large scale transcriptional changes and the acquisition of an effector T cell phenotype as well as the generation of a population of CD103+ CD69+ Trm like cells. While inhibition of parasite replication resulted in reduced effector responses it did not alter the Trm population. These data sets highlight that recent TCR activation contributes to the phenotypic heterogeneity of the CD8+ T cell response but suggest that this process has a limited impact on memory populations at acute and chronic stages of infection.
Collapse
Affiliation(s)
- Lindsey A. Shallberg
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Anthony T. Phan
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - David A. Christian
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Joseph A. Perry
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Breanne E. Haskins
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Daniel P. Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Tajie H. Harris
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Anita A. Koshy
- Department of Neurology, Department of Immunobiology, and BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
11
|
Kovacs MA, Cowan MN, Babcock IW, Sibley LA, Still K, Batista SJ, Labuzan SA, Sethi I, Harris TH. Meningeal lymphatic drainage promotes T cell responses against Toxoplasma gondii but is dispensable for parasite control in the brain. eLife 2022; 11:80775. [PMID: 36541708 PMCID: PMC9812409 DOI: 10.7554/elife.80775] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
The discovery of meningeal lymphatic vessels that drain the CNS has prompted new insights into how immune responses develop in the brain. In this study, we examined how T cell responses against CNS-derived antigen develop in the context of infection. We found that meningeal lymphatic drainage promotes CD4+ and CD8+ T cell responses against the neurotropic parasite Toxoplasma gondii in mice, and we observed changes in the dendritic cell compartment of the dural meninges that may support this process. Indeed, we found that mice chronically, but not acutely, infected with T. gondii exhibited a significant expansion and activation of type 1 and type 2 conventional dendritic cells (cDC) in the dural meninges. cDC1s and cDC2s were both capable of sampling cerebrospinal fluid (CSF)-derived protein and were found to harbor processed CSF-derived protein in the draining deep cervical lymph nodes. Disrupting meningeal lymphatic drainage via ligation surgery led to a reduction in CD103+ cDC1 and cDC2 number in the deep cervical lymph nodes and caused an impairment in cDC1 and cDC2 maturation. Concomitantly, lymphatic vessel ligation impaired CD4+ and CD8+ T cell activation, proliferation, and IFN-γ production at this site. Surprisingly, however, parasite-specific T cell responses in the brain remained intact following ligation, which may be due to concurrent activation of T cells at non-CNS-draining sites during chronic infection. Collectively, our work reveals that CNS lymphatic drainage supports the development of peripheral T cell responses against T. gondii but remains dispensable for immune protection of the brain.
Collapse
Affiliation(s)
- Michael A Kovacs
- Center for Brain Immunology and Glia, Department of Neuroscience, University of VirginiaCharlottesvilleUnited States
| | - Maureen N Cowan
- Center for Brain Immunology and Glia, Department of Neuroscience, University of VirginiaCharlottesvilleUnited States
| | - Isaac W Babcock
- Center for Brain Immunology and Glia, Department of Neuroscience, University of VirginiaCharlottesvilleUnited States
| | - Lydia A Sibley
- Center for Brain Immunology and Glia, Department of Neuroscience, University of VirginiaCharlottesvilleUnited States
| | - Katherine Still
- Center for Brain Immunology and Glia, Department of Neuroscience, University of VirginiaCharlottesvilleUnited States
| | - Samantha J Batista
- Center for Brain Immunology and Glia, Department of Neuroscience, University of VirginiaCharlottesvilleUnited States
| | - Sydney A Labuzan
- Center for Brain Immunology and Glia, Department of Neuroscience, University of VirginiaCharlottesvilleUnited States
| | - Ish Sethi
- Center for Brain Immunology and Glia, Department of Neuroscience, University of VirginiaCharlottesvilleUnited States
| | - Tajie H Harris
- Center for Brain Immunology and Glia, Department of Neuroscience, University of VirginiaCharlottesvilleUnited States
| |
Collapse
|
12
|
Fowell DJ, Kim M. The spatio-temporal control of effector T cell migration. Nat Rev Immunol 2021; 21:582-596. [PMID: 33627851 PMCID: PMC9380693 DOI: 10.1038/s41577-021-00507-0] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2021] [Indexed: 02/08/2023]
Abstract
Effector T cells leave the lymph nodes armed with specialized functional attributes. Their antigenic targets may be located anywhere in the body, posing the ultimate challenge: how to efficiently identify the target tissue, navigate through a complex tissue matrix and, ultimately, locate the immunological insult. Recent advances in real-time in situ imaging of effector T cell migratory behaviour have revealed a great degree of mechanistic plasticity that enables effector T cells to push and squeeze their way through inflamed tissues. This process is shaped by an array of 'stop' and 'go' guidance signals including target antigens, chemokines, integrin ligands and the mechanical cues of the inflamed microenvironment. Effector T cells must sense and interpret these competing signals to correctly position themselves to mediate their effector functions for complete and durable responses in infectious disease and malignancy. Tuning T cell migration therapeutically will require a new understanding of this complex decision-making process.
Collapse
Affiliation(s)
- Deborah J. Fowell
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute for Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY.,Department of Microbiology and Immunology, Cornell University, Ithaca, NY
| | - Minsoo Kim
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute for Biomedical Sciences, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
13
|
Sbierski-Kind J, Mroz N, Molofsky AB. Perivascular stromal cells: Directors of tissue immune niches. Immunol Rev 2021; 302:10-31. [PMID: 34075598 DOI: 10.1111/imr.12984] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/05/2021] [Accepted: 05/09/2021] [Indexed: 12/12/2022]
Abstract
Perivascular niches are specialized microenvironments where stromal and immune cells interact with vasculature to monitor tissue status. Adventitial perivascular niches surround larger blood vessels and other boundary sites, supporting collections of immune cells, stromal cells, lymphatics, and neurons. Adventitial fibroblasts (AFs), a subtype of mesenchymal stromal cell, are the dominant constituents in adventitial spaces, regulating vascular integrity while organizing the accumulation and activation of a variety of interacting immune cells. In contrast, pericytes are stromal mural cells that support microvascular capillaries and surround organ-specific parenchymal cells. Here, we outline the unique immune and non-immune composition of perivascular tissue immune niches, with an emphasis on the heterogeneity and immunoregulatory functions of AFs and pericytes across diverse organs. We will discuss how perivascular stromal cells contribute to the regulation of innate and adaptive immune responses and integrate immunological signals to impact tissue health and disease.
Collapse
Affiliation(s)
- Julia Sbierski-Kind
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Nicholas Mroz
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA.,Biomedical Sciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Ari B Molofsky
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA.,Diabetes Center, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
14
|
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: 110] [Impact Index Per Article: 27.5] [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
|
15
|
McGovern KE, Nance JP, David CN, Harrison RES, Noor S, Worth D, Landrith TA, Obenaus A, Carson MJ, Morikis D, Wilson EH. SPARC coordinates extracellular matrix remodeling and efficient recruitment to and migration of antigen-specific T cells in the brain following infection. Sci Rep 2021; 11:4549. [PMID: 33633185 PMCID: PMC7907143 DOI: 10.1038/s41598-021-83952-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/03/2021] [Indexed: 01/14/2023] Open
Abstract
Central nervous system (CNS) injury and infection can result in profound tissue remodeling in the brain, the mechanism and purpose of which is poorly understood. Infection with the protozoan parasite Toxoplasma gondii causes chronic infection and inflammation in the brain parenchyma. Control of parasite replication requires the continuous presence of IFNγ-producing T cells to keep T. gondii in its slowly replicating cyst form. During infection, a network of extracellular matrix fibers, revealed using multiphoton microscopy, forms in the brain. The origin and composition of these structures are unknown but the fibers have been observed to act as a substrate for migrating T cells. In this study, we show a critical regulator of extracellular matrix (ECM) remodeling, Secreted Protein, Acidic, Rich in Cysteine (SPARC), is upregulated in the brain during the early phases of infection in the frontal cortex. In the absence of SPARC, a reduced and disordered fibrous network, increased parasite burden, and reduced antigen-specific T cell entry into the brain points to a role for SPARC in T cell recruitment to and migration within the brain. We also report SPARC can directly bind to CCR7 ligands CCL19 and CCL21 but not CXCL10, and enhance migration toward a chemokine gradient. Measurement of T cell behavior points to tissue remodeling being important for access of immune cells to the brain and facilitating cellular locomotion. Together, these data identify SPARC as an important regulatory component of immune cell trafficking and access to the inflamed CNS.
Collapse
Affiliation(s)
- Kathryn E McGovern
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
- BIO5 Institute, Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
| | - J Philip Nance
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Clément N David
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
- Nanostring Technologies, Inc, 530 Fairview Ave N, Seattle, WA, 98109, USA
| | - Reed E S Harrison
- Department of Bioengineering, University of California, Riverside, Riverside, CA, 92521-0129, USA
- UCSD Bioengineering and the Institute for Engineering in Medicine, San Diego, CA, 92093, USA
| | - Shahani Noor
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
- School of Medicine, MSC08, University of New Mexico, Albequerque, NM, 87131, USA
| | - Danielle Worth
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Tyler A Landrith
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
- Ambrey Genetics, Aliso Viejo, CA, 92656, USA
| | - Andre Obenaus
- School of Medicine, University of California, Irvine, Irvine, CA, 92697, USA
| | - Monica J Carson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Dimitrios Morikis
- Department of Bioengineering, University of California, Riverside, Riverside, CA, 92521-0129, USA
| | - Emma H Wilson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA.
| |
Collapse
|
16
|
Ramaglia V, Florescu A, Zuo M, Sheikh-Mohamed S, Gommerman JL. Stromal Cell–Mediated Coordination of Immune Cell Recruitment, Retention, and Function in Brain-Adjacent Regions. THE JOURNAL OF IMMUNOLOGY 2021; 206:282-291. [DOI: 10.4049/jimmunol.2000833] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/27/2020] [Indexed: 12/15/2022]
|
17
|
Afifi MA. The Parasites Caught In-Action: Imaging at the Host-Parasite Interface. J Microsc Ultrastruct 2021; 9:1-6. [PMID: 33850705 PMCID: PMC8030542 DOI: 10.4103/jmau.jmau_1_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 01/02/2020] [Accepted: 01/21/2020] [Indexed: 11/24/2022] Open
Abstract
For many decades, scientists were unable to expose the invisible existence of the parasites in their living hosts, except by scarification and then dissection of the animal model. This process just demonstrates a dead parasite in a dead host. Using this approach, very limited information can be obtained concerning the dynamics of infection and the pathways utilized by the parasite to survive within a hostile host's environment. Introduction of ultra-high-speed imaging techniques, with a time domain of barely few microseconds or even less, has revolutionized the "in vivo dissection" of the parasites. Such methods provide platforms for imaging host-parasite interactions at diverse scales, down to the molecular level. These have complementary advantages and relative assets in investigating host-parasite interactions. Therefore, better elucidation of such interaction may require the usage of more than one approach. Precise in vivo quantification, of the parasite load within the host, and better insight into the kinetics of infection are the two main advantages of the novel imaging procedures. However, imaging parasite-host interplay is still a challenging approach due to many constraints related to the parasite biology, the tissue environment within which the parasites exist, and the logistic technical limitations. This review was planned to assist better understanding of how much the new imaging techniques impacted the recent advances in parasite biology, especially the immunobiology of protozoan parasites.
Collapse
Affiliation(s)
- Mohammed A. Afifi
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Parasitology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
18
|
Fernandes NRJ, Reilly NS, Schrock DC, Hocking DC, Oakes PW, Fowell DJ. CD4 + T Cell Interstitial Migration Controlled by Fibronectin in the Inflamed Skin. Front Immunol 2020; 11:1501. [PMID: 32793204 PMCID: PMC7393769 DOI: 10.3389/fimmu.2020.01501] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/09/2020] [Indexed: 01/21/2023] Open
Abstract
The extracellular matrix (ECM) is extensively remodeled during inflammation providing essential guidance cues for immune cell migration and signals for cell activation and survival. There is increasing interest in the therapeutic targeting of ECM to mitigate chronic inflammatory diseases and enhance access to the tumor microenvironment. T cells utilize the ECM as a scaffold for interstitial migration, dependent on T cell expression of matrix-binding integrins αVβ1/αVβ3 and tissue display of the respective RGD-containing ligands. The specific ECM components that control T cell migration are unclear. Fibronectin (FN), a canonical RGD-containing matrix component, is heavily upregulated in inflamed tissues and in vitro can serve as a substrate for leukocyte migration. However, limited by lack of tools to intravitally visualize and manipulate FN, the specific role of FN in effector T cell migration in vivo is unknown. Here, we utilize fluorescently-tagged FN to probe for FN deposition, and intravital multiphoton microscopy to visualize T cell migration relative to FN in the inflamed ear dermis. Th1 cells were found to migrate along FN fibers, with T cells appearing to actively push or pull against flexible FN fibers. To determine the importance of T cell interactions with FN, we used a specific inhibitor of FN polymerization, pUR4. Intradermal delivery of pUR4 (but not the control peptide) to the inflamed skin resulted in a local reduction in FN deposition. We also saw a striking attenuation of Th1 effector T cell movement at the pUR4 injection site, suggesting FN plays a key role in T cell interstitial migration. In mechanistic studies, pUR4 incubation with FN in vitro resulted in enhanced tethering of T cells to FN matrix, limiting productive migration. In vivo, such tethering led to increased Th1 accumulation in the inflamed dermis. Enhanced Th1 accumulation exacerbated inflammation with increased Th1 activation and IFNγ cytokine production. Thus, our studies highlight the importance of ECM FN fibrils for T cell migration in inflamed tissues and suggest that manipulating local levels of ECM FN may prove beneficial in promoting T cell accumulation in tissues and enhancing local immunity to infection or cancer.
Collapse
Affiliation(s)
- Ninoshka R. J. Fernandes
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Nicholas S. Reilly
- Department of Physics and Astronomy, University of Rochester, Rochester, NY, United States
| | - Dillon C. Schrock
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Denise C. Hocking
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, United States
| | - Patrick W. Oakes
- Department of Physics and Astronomy, University of Rochester, Rochester, NY, United States
- Department of Biology, University of Rochester, Rochester, NY, United States
| | - Deborah J. Fowell
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| |
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
|
Boothroyd JC. What a Difference 30 Years Makes! A Perspective on Changes in Research Methodologies Used to Study Toxoplasma gondii. Methods Mol Biol 2020; 2071:1-25. [PMID: 31758444 DOI: 10.1007/978-1-4939-9857-9_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Toxoplasma gondii is a remarkable species with a rich cell, developmental, and population biology. It is also sometimes responsible for serious disease in animals and humans and the stages responsible for such disease are relatively easy to study in vitro or in laboratory animal models. As a result of all this, Toxoplasma has become the subject of intense investigation over the last several decades, becoming a model organism for the study of the phylum of which it is a member, Apicomplexa. This has led to an ever-growing number of investigators applying an ever-expanding set of techniques to dissecting how Toxoplasma "ticks" and how it interacts with its many hosts. In this perspective piece I first wind back the clock 30 years and then trace the extraordinary pace of methodologies that have propelled the field forward to where we are today. In keeping with the theme of this collection, I focus almost exclusively on the parasite, rather than host side of the equation. I finish with a few thoughts about where the field might be headed-though if we have learned anything, the only sure prediction is that the pace of technological advance will surely continue to accelerate and the future will give us still undreamed of methods for taking apart (and then putting back together) this amazing organism with all its intricate biology. We have so far surely just scratched the surface.
Collapse
Affiliation(s)
- John C Boothroyd
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
21
|
Imaging the dynamic recruitment of monocytes to the blood-brain barrier and specific brain regions during Toxoplasma gondii infection. Proc Natl Acad Sci U S A 2019; 116:24796-24807. [PMID: 31727842 DOI: 10.1073/pnas.1915778116] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Brain infection by the parasite Toxoplasma gondii in mice is thought to generate vulnerability to predation by mechanisms that remain elusive. Monocytes play a key role in host defense and inflammation and are critical for controlling T. gondii However, the dynamic and regional relationship between brain-infiltrating monocytes and parasites is unknown. We report the mobilization of inflammatory (CCR2+Ly6Chi) and patrolling (CX3CR1+Ly6Clo) monocytes into the blood and brain during T. gondii infection of C57BL/6J and CCR2RFP/+CX3CR1GFP/+ mice. Longitudinal analysis of mice using 2-photon intravital imaging of the brain through cranial windows revealed that CCR2-RFP monocytes were recruited to the blood-brain barrier (BBB) within 2 wk of T. gondii infection, exhibited distinct rolling and crawling behavior, and accumulated within the vessel lumen before entering the parenchyma. Optical clearing of intact T. gondii-infected brains using iDISCO+ and light-sheet microscopy enabled global 3D detection of monocytes. Clusters of T. gondii and individual monocytes across the brain were identified using an automated cell segmentation pipeline, and monocytes were found to be significantly correlated with sites of T. gondii clusters. Computational alignment of brains to the Allen annotated reference atlas [E. S. Lein et al., Nature 445:168-176 (2007)] indicated a consistent pattern of monocyte infiltration during T. gondii infection to the olfactory tubercle, in contrast to LPS treatment of mice, which resulted in a diffuse distribution of monocytes across multiple brain regions. These data provide insights into the dynamics of monocyte recruitment to the BBB and the highly regionalized localization of monocytes in the brain during T. gondii CNS infection.
Collapse
|
22
|
Tsitsiklis A, Bangs DJ, Robey EA. CD8+ T Cell Responses to Toxoplasma gondii: Lessons from a Successful Parasite. Trends Parasitol 2019; 35:887-898. [DOI: 10.1016/j.pt.2019.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/22/2019] [Accepted: 08/26/2019] [Indexed: 01/21/2023]
|
23
|
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
|
24
|
Schlüter D, Barragan A. Advances and Challenges in Understanding Cerebral Toxoplasmosis. Front Immunol 2019; 10:242. [PMID: 30873157 PMCID: PMC6401564 DOI: 10.3389/fimmu.2019.00242] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 01/28/2019] [Indexed: 11/22/2022] Open
Abstract
Toxoplasma gondii is a widespread parasitic pathogen that infects over one third of the global human population. The parasite invades and chronically persists in the central nervous system (CNS) of the infected host. Parasite spread and persistence is intimately linked to an ensuing immune response, which does not only limit parasite-induced damage but also may facilitate dissemination and induce parasite-associated immunopathology. Here, we discuss various aspects of toxoplasmosis where knowledge is scarce or controversial and, the recent advances in the understanding of the delicate interplay of T. gondii with the immune system in experimental and clinical settings. This includes mechanisms for parasite passage from the circulation into the brain parenchyma across the blood-brain barrier during primary acute infection. Later, as chronic latent infection sets in with control of the parasite in the brain parenchyma, the roles of the inflammatory response and of immune cell responses in this phase of the disease are discussed. Additionally, the function of brain resident cell populations is delineated, i.e., how neurons, astrocytes and microglia serve both as target cells for the parasite but also actively contribute to the immune response. As the infection can reactivate in the CNS of immune-compromised individuals, we bring up the immunopathogenesis of reactivated toxoplasmosis, including the special case of congenital CNS manifestations. The relevance, advantages and limitations of rodent infection models for the understanding of human cerebral toxoplasmosis are discussed. Finally, this review pinpoints questions that may represent challenges to experimental and clinical science with respect to improved diagnostics, pharmacological treatments and immunotherapies.
Collapse
Affiliation(s)
- Dirk Schlüter
- Hannover Medical School, Institute of Medical Microbiology and Hospital Epidemiology, Hannover, Germany
| | - Antonio Barragan
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
25
|
Hussell T, Lui S, Jagger C, Morgan D, Brand O. The consequence of matrix dysfunction on lung immunity and the microbiome in COPD. Eur Respir Rev 2018; 27:27/148/180032. [PMID: 29950305 DOI: 10.1183/16000617.0032-2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/12/2018] [Indexed: 12/23/2022] Open
Abstract
The pulmonary extracellular matrix (ECM) is a complex network of proteins which primarily defines tissue architecture and regulates various biochemical and biophysical processes. It is a dynamic system comprising two main structures (the interstitial matrix and the basement membrane) which undergo continuous, yet highly regulated, remodelling. This remodelling process is essential for tissue homeostasis and uncontrolled regulation can lead to pathological states including chronic obstructive pulmonary disease (COPD). Altered expression of ECM proteins, as observed in COPD, can contribute to the degradation of alveolar walls and thickening of the small airways which can cause limitations in airflow. Modifications in ECM composition can also impact immune cell migration and retention in the lung with migrating cells becoming entrapped in the diseased airspaces. Furthermore, ECM changes affect the lung microbiome, aggravating and advancing disease progression. A dysbiosis in bacterial diversity can lead to infection, inducing epithelial injury and pro-inflammatory reactions. Here we review the changes noted in the different ECM components in COPD and discuss how an imbalance in microbial commensalism can impact disease development.
Collapse
Affiliation(s)
- Tracy Hussell
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK
| | - Sylvia Lui
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK
| | - Christopher Jagger
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK
| | - David Morgan
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK
| | - Oliver Brand
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, UK
| |
Collapse
|
26
|
Sanecka A, Yoshida N, Kolawole EM, Patel H, Evavold BD, Frickel EM. T Cell Receptor-Major Histocompatibility Complex Interaction Strength Defines Trafficking and CD103 + Memory Status of CD8 T Cells in the Brain. Front Immunol 2018; 9:1290. [PMID: 29922298 PMCID: PMC5996069 DOI: 10.3389/fimmu.2018.01290] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 05/23/2018] [Indexed: 11/30/2022] Open
Abstract
T cell receptor-major histocompatibility complex (TCR-MHC) affinities span a wide range in a polyclonal T cell response, yet it is undefined how affinity shapes long-term properties of CD8 T cells during chronic infection with persistent antigen. Here, we investigate how the affinity of the TCR-MHC interaction shapes the phenotype of memory CD8 T cells in the chronically Toxoplasma gondii-infected brain. We employed CD8 T cells from three lines of transnuclear (TN) mice that harbor in their endogenous loci different T cell receptors specific for the same Toxoplasma antigenic epitope ROP7. The three TN CD8 T cell clones span a wide range of affinities to MHCI-ROP7. These three CD8 T cell clones have a distinct and fixed hierarchy in terms of effector function in response to the antigen measured as proliferation capacity, trafficking, T cell maintenance, and memory formation. In particular, the T cell clone of lowest affinity does not home to the brain. The two higher affinity T cell clones show differences in establishing resident-like memory populations (CD103+) in the brain with the higher affinity clone persisting longer in the host during chronic infection. Transcriptional profiling of naïve and activated ROP7-specific CD8 T cells revealed that Klf2 encoding a transcription factor that is known to be a negative marker for T cell trafficking is upregulated in the activated lowest affinity ROP7 clone. Our data thus suggest that TCR-MHC affinity dictates memory CD8 T cell fate at the site of infection.
Collapse
Affiliation(s)
- Anna Sanecka
- Host-Toxoplasma Interaction Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Nagisa Yoshida
- Host-Toxoplasma Interaction Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Elizabeth Motunrayo Kolawole
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, United States
| | - Harshil Patel
- Bioinformatics and Biostatistics, The Francis Crick Institute, London, United Kingdom
| | - Brian D. Evavold
- Bioinformatics and Biostatistics, The Francis Crick Institute, London, United Kingdom
| | - Eva-Maria Frickel
- Host-Toxoplasma Interaction Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
27
|
Takamura S. Niches for the Long-Term Maintenance of Tissue-Resident Memory T Cells. Front Immunol 2018; 9:1214. [PMID: 29904388 PMCID: PMC5990602 DOI: 10.3389/fimmu.2018.01214] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022] Open
Abstract
Tissue-resident memory T cells (TRM cells) are a population of immune cells that reside in the lymphoid and non-lymphoid organs without recirculation through the blood. These important cells occupy and utilize unique anatomical and physiological niches that are distinct from those for other memory T cell populations, such as central memory T cells in the secondary lymphoid organs and effector memory T cells that circulate through the tissues. CD8+ TRM cells typically localize in the epithelial layers of barrier tissues where they are optimally positioned to act as sentinels to trigger antigen-specific protection against reinfection. CD4+ TRM cells typically localize below the epithelial layers, such as below the basement membrane, and cluster in lymphoid structures designed to optimize interactions with antigen-presenting cells upon reinfection. A key feature of TRM populations is their ability to be maintained in barrier tissues for prolonged periods of time. For example, skin CD8+ TRM cells displace epidermal niches originally occupied by γδ T cells, thereby enabling their stable persistence for years. It is also clear that the long-term maintenance of TRM cells in different microenvironments is dependent on multiple tissue-specific survival cues, although the specific details are poorly understood. However, not all TRM persist over the long term. Recently, we identified a new spatial niche for the maintenance of CD8+ TRM cells in the lung, which is created at the site of tissue regeneration after injury [termed repair-associated memory depots (RAMD)]. The short-lived nature of RAMD potentially explains the short lifespans of CD8+ TRM cells in this particular tissue. Clearly, a better understanding of the niche-dependent maintenance of TRM cells will be important for the development of vaccines designed to promote barrier immunity. In this review, we discuss recent advances in our understanding of the properties and nature of tissue-specific niches that maintain TRM cells in different tissues.
Collapse
Affiliation(s)
- Shiki Takamura
- Department of Immunology, Faculty of Medicine, Kindai University, Osaka, Japan
| |
Collapse
|
28
|
Espinosa-Carrasco G, Le Saout C, Fontanaud P, Michau A, Mollard P, Hernandez J, Schaeffer M. Integrin β1 Optimizes Diabetogenic T Cell Migration and Function in the Pancreas. Front Immunol 2018; 9:1156. [PMID: 29904378 PMCID: PMC5990596 DOI: 10.3389/fimmu.2018.01156] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/08/2018] [Indexed: 01/08/2023] Open
Abstract
T cell search behavior is dictated by their need to encounter their specific antigen to eliminate target cells. However, mechanisms controlling effector T cell motility are highly tissue-dependent. Specifically, how diabetogenic T cells encounter their target beta cells in dispersed islets throughout the pancreas (PA) during autoimmune diabetes remains unclear. Using intra-vital 2-photon microscopy in a mouse model of diabetes, we found that CXCR3 chemokine downregulated CD8+ T cell motility specifically within islets, promoting effector cell confinement to their target sites. By contrast, T cell velocity and directionality in the exocrine tissue were enhanced along blood vessels and extracellular matrix fibers. This guided migration implicated integrin-dependent interactions, since integrin blockade impaired exocrine T cell motility. In addition, integrin β1 blockade decreased CD4+ T cell effector phenotype specifically in the PA. Thus, we unveil an important role for integrins in the PA during autoimmune diabetes that may have important implications for the design of new therapies.
Collapse
Affiliation(s)
- Gabriel Espinosa-Carrasco
- INSERM U1183, Institute for Regenerative Medicine and Biotherapy, University of Montpellier, Montpellier, France.,Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Cécile Le Saout
- INSERM U1183, Institute for Regenerative Medicine and Biotherapy, University of Montpellier, Montpellier, France
| | - Pierre Fontanaud
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Aurélien Michau
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Patrice Mollard
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Javier Hernandez
- INSERM U1183, Institute for Regenerative Medicine and Biotherapy, University of Montpellier, Montpellier, France
| | - Marie Schaeffer
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
29
|
Rua R, McGavern DB. Advances in Meningeal Immunity. Trends Mol Med 2018; 24:542-559. [PMID: 29731353 PMCID: PMC6044730 DOI: 10.1016/j.molmed.2018.04.003] [Citation(s) in RCA: 198] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/08/2018] [Accepted: 04/09/2018] [Indexed: 12/26/2022]
Abstract
The central nervous system (CNS) is an immunologically specialized tissue protected by a blood-brain barrier. The CNS parenchyma is enveloped by a series of overlapping membranes that are collectively referred to as the meninges. The meninges provide an additional CNS barrier, harbor a diverse array of resident immune cells, and serve as a crucial interface with the periphery. Recent studies have significantly advanced our understanding of meningeal immunity, demonstrating how a complex immune landscape influences CNS functions under steady-state and inflammatory conditions. The location and activation state of meningeal immune cells can profoundly influence CNS homeostasis and contribute to neurological disorders, but these cells are also well equipped to protect the CNS from pathogens. In this review, we discuss advances in our understanding of the meningeal immune repertoire and provide insights into how this CNS barrier operates immunologically under conditions ranging from neurocognition to inflammatory diseases.
Collapse
Affiliation(s)
- Rejane Rua
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
30
|
Mrass P, Oruganti SR, Fricke GM, Tafoya J, Byrum JR, Yang L, Hamilton SL, Miller MJ, Moses ME, Cannon JL. ROCK regulates the intermittent mode of interstitial T cell migration in inflamed lungs. Nat Commun 2017; 8:1010. [PMID: 29044117 PMCID: PMC5647329 DOI: 10.1038/s41467-017-01032-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 08/14/2017] [Indexed: 12/27/2022] Open
Abstract
Effector T cell migration through tissues can enable control of infection or mediate inflammatory damage. Nevertheless, the molecular mechanisms that regulate migration of effector T cells within the interstitial space of inflamed lungs are incompletely understood. Here, we show T cell migration in a mouse model of acute lung injury with two-photon imaging of intact lung tissue. Computational analysis indicates that T cells migrate with an intermittent mode, switching between confined and almost straight migration, guided by lung-associated vasculature. Rho-associated protein kinase (ROCK) is required for both high-speed migration and straight motion. By contrast, inhibition of Gαi signaling with pertussis toxin affects speed but not the intermittent migration of lung-infiltrating T cells. Computational modeling shows that an intermittent migration pattern balances both search area and the duration of contacts between T cells and target cells. These data identify that ROCK-dependent intermittent T cell migration regulates tissue-sampling during acute lung injury. ROCK is associated with T cell movement in lymph nodes. Here the authors use an LPS lung damage model and two-photon imaging to show that CD8+ T cells in lung tissue engage in ROCK-dependent fast linear migration alternating with bursts of slower confined migration that together optimize contact with target cells.
Collapse
Affiliation(s)
- Paulus Mrass
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, MSC 08 4660, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Sreenivasa Rao Oruganti
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, MSC 08 4660, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - G Matthew Fricke
- Department of Computer Science, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Justyna Tafoya
- Department of Computer Science, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA.,Department of Mathematics, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Janie R Byrum
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, MSC 08 4660, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Lihua Yang
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Samantha L Hamilton
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Mark J Miller
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Melanie E Moses
- Department of Computer Science, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA.,Department of Biology, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA.,External Faculty, Santa Fe Institute, 1399 Hyde Park Road, Santa Fe, NM, 87501, USA
| | - Judy L Cannon
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, MSC 08 4660, 1 University of New Mexico, Albuquerque, NM, 87131, USA.
| |
Collapse
|
31
|
Attanasio J, Wherry EJ. Costimulatory and Coinhibitory Receptor Pathways in Infectious Disease. Immunity 2017; 44:1052-68. [PMID: 27192569 DOI: 10.1016/j.immuni.2016.04.022] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Indexed: 12/16/2022]
Abstract
Costimulatory and inhibitory receptors play a key role in regulating immune responses to infections. Recent translation of knowledge about inhibitory receptors such as CTLA-4 and PD-1 into the cancer clinic highlights the opportunities to manipulate these pathways to treat human disease. Studies in infectious disease have provided key insights into the specific roles of these pathways and the effects of their manipulation. Here, recent studies are discussed that have addressed how major inhibitory and costimulatory pathways play a role in regulating immune responses during acute and chronic infections. Mechanistic insights from studies of infectious disease provide opportunities to further expand our toolkit to treat cancer and chronic infections in the clinic.
Collapse
Affiliation(s)
- John Attanasio
- Institute for Immunology and Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - E John Wherry
- Institute for Immunology and Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
32
|
Song KH, Lee J, Jung HR, Park H, Doh J. Turning behaviors of T cells climbing up ramp-like structures are regulated by myosin light chain kinase activity and lamellipodia formation. Sci Rep 2017; 7:11533. [PMID: 28912435 PMCID: PMC5599526 DOI: 10.1038/s41598-017-11938-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 09/01/2017] [Indexed: 12/05/2022] Open
Abstract
T cells navigate diverse microenvironments to perform immune responses. Micro-scale topographical structures within the tissues, which may inherently exist in normal tissues or may be formed by inflammation or injury, can influence T cell migration, but how T cell migration is affected by such topographical structures have not been investigated. In this study, we fabricated ramp-like structures with a 5 μm height and various slopes, and observed T cells climbing up the ramp-like structures. T cells encountering the ramp-like structures exhibited MLC accumulation near head-tail junctions contacting the ramp-like structures, and made turns to the direction perpendicular to the ramp-like structures. Pharmacological study revealed that lamellipodia formation mediated by arp2/3 and contractility regulated by myosin light chain kinase (MLCK) were responsible for the intriguing turning behavior of T cells climbing the ramp-like structures. Arp2/3 or MLCK inhibition substantially reduced probability of T cells climbing sharp-edged ramp-like structures, indicating intriguing turning behavior of T cells mediated by lamellipodia formation and MLCK activity may be important for T cells to access inflamed or injured tissues with abrupt topographical changes.
Collapse
Affiliation(s)
- Kwang Hoon Song
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH) San 31, Hyoja-dong, Nam-Gu, Pohang, Gyeongbuk, 790-784, Korea.,Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, PA, 19104, USA
| | - Jaehyun Lee
- School of Interdisciplinary Bioscience and Bioengineering (I-Bio), Pohang University of Science and Technology (POSTECH) San 31, Hyoja-dong, Nam-Gu, Pohang, Gyeongbuk, 790-784, Korea
| | - Hong-Ryul Jung
- School of Interdisciplinary Bioscience and Bioengineering (I-Bio), Pohang University of Science and Technology (POSTECH) San 31, Hyoja-dong, Nam-Gu, Pohang, Gyeongbuk, 790-784, Korea
| | - HyoungJun Park
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH) San 31, Hyoja-dong, Nam-Gu, Pohang, Gyeongbuk, 790-784, Korea
| | - Junsang Doh
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH) San 31, Hyoja-dong, Nam-Gu, Pohang, Gyeongbuk, 790-784, Korea. .,School of Interdisciplinary Bioscience and Bioengineering (I-Bio), Pohang University of Science and Technology (POSTECH) San 31, Hyoja-dong, Nam-Gu, Pohang, Gyeongbuk, 790-784, Korea.
| |
Collapse
|
33
|
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
|
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
|
Lodygin D, Flügel A. Intravital real-time analysis of T-cell activation in health and disease. Cell Calcium 2017; 64:118-129. [DOI: 10.1016/j.ceca.2016.12.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 01/27/2023]
|
36
|
Immune Alterations in CD8+ T Cells Are Associated with Neuronal C-C and C-X-C Chemokine Receptor Regulation Through Adenosine A2A Receptor Signaling in a BTBR T+ Itpr3tf/J Autistic Mouse Model. Mol Neurobiol 2017; 55:2603-2616. [DOI: 10.1007/s12035-017-0548-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 04/07/2017] [Indexed: 11/25/2022]
|
37
|
Landrith TA, Sureshchandra S, Rivera A, Jang JC, Rais M, Nair MG, Messaoudi I, Wilson EH. CD103 + CD8 T Cells in the Toxoplasma-Infected Brain Exhibit a Tissue-Resident Memory Transcriptional Profile. Front Immunol 2017; 8:335. [PMID: 28424687 PMCID: PMC5372813 DOI: 10.3389/fimmu.2017.00335] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/08/2017] [Indexed: 12/20/2022] Open
Abstract
During chronic infection, memory T cells acquire a unique phenotype and become dependent on different survival signals than those needed for memory T cells generated during an acute infection. The distinction between the role of effector and memory T cells in an environment of persistent antigen remains unclear. Here, in the context of chronic Toxoplasma gondii infection, we demonstrate that a population of CD8 T cells exhibiting a tissue-resident memory (TRM) phenotype accumulates within the brain. We show that this population is distributed throughout the brain in both parenchymal and extraparenchymal spaces. Furthermore, this population is transcriptionally distinct and exhibits a transcriptional signature consistent with the TRM observed in acute viral infections. Finally, we establish that the CD103+ TRM population has an intrinsic capacity to produce both IFN-γ and TNF-α, cytokines critical for parasite control within the central nervous system (CNS). The contribution of this population to pro-inflammatory cytokine production suggests an important role for TRM in protective and ongoing immune responses in the infected CNS. Accession number: GSE95105
Collapse
Affiliation(s)
- Tyler A Landrith
- School of Medicine, University of California, Riverside, CA, USA
| | | | - Andrea Rivera
- School of Medicine, University of California, Riverside, CA, USA
| | - Jessica C Jang
- School of Medicine, University of California, Riverside, CA, USA
| | - Maham Rais
- School of Medicine, University of California, Riverside, CA, USA
| | - Meera G Nair
- School of Medicine, University of California, Riverside, CA, USA
| | - Ilhem Messaoudi
- School of Medicine, University of California, Riverside, CA, USA
| | - Emma H Wilson
- School of Medicine, University of California, Riverside, CA, USA
| |
Collapse
|
38
|
Pikor NB, Cupovic J, Onder L, Gommerman JL, Ludewig B. Stromal Cell Niches in the Inflamed Central Nervous System. THE JOURNAL OF IMMUNOLOGY 2017; 198:1775-1781. [DOI: 10.4049/jimmunol.1601566] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 10/19/2016] [Indexed: 11/19/2022]
|
39
|
Bakheet SA, Alzahrani MZ, Nadeem A, Ansari MA, Zoheir KMA, Attia SM, AL-Ayadhi LY, Ahmad SF. Resveratrol treatment attenuates chemokine receptor expression in the BTBR T + tf/J mouse model of autism. Mol Cell Neurosci 2016; 77:1-10. [DOI: 10.1016/j.mcn.2016.09.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/01/2016] [Accepted: 09/28/2016] [Indexed: 02/06/2023] Open
|
40
|
Rossi B, Constantin G. Live Imaging of Immune Responses in Experimental Models of Multiple Sclerosis. Front Immunol 2016; 7:506. [PMID: 27917173 PMCID: PMC5116921 DOI: 10.3389/fimmu.2016.00506] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 11/01/2016] [Indexed: 12/31/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is the most common animal model of multiple sclerosis (MS), a chronic inflammatory autoimmune disease of the central nervous system (CNS) characterized by multifocal perivascular infiltrates that predominantly comprise lymphocytes and macrophages. During EAE, autoreactive T cells first become active in the secondary lymphoid organs upon contact with antigen-presenting cells (APCs), and then gain access to CNS parenchyma, through a compromised blood–brain barrier, subsequently inducing inflammation and demyelination. Two-photon laser scanning microscopy (TPLSM) is an ideal tool for intravital imaging because of its low phototoxicity, deep tissue penetration, and high resolution. In the last decade, TPLSM has been used to visualize the behavior of T cells and their contact with APCs in the lymph nodes (LNs) and target tissues in several models of autoimmune diseases. The leptomeninges and cerebrospinal fluid represent particularly important points for T cell entry into the CNS and reactivation following contact with local APCs during the preclinical phase of EAE. In this review, we highlight recent findings concerning the pathogenesis of EAE and MS, emphasizing the use of TPLSM to characterize T cell activation in the LNs and CNS, as well as the mechanisms of tolerance induction. Furthermore, we discuss how advanced imaging unveils disease mechanisms and helps to identify novel therapeutic strategies to treat CNS autoimmunity and inflammation.
Collapse
Affiliation(s)
- Barbara Rossi
- Section of General Pathology, Department of Medicine, University of Verona , Verona , Italy
| | - Gabriela Constantin
- Section of General Pathology, Department of Medicine, University of Verona , Verona , Italy
| |
Collapse
|
41
|
STAT1 Signaling in Astrocytes Is Essential for Control of Infection in the Central Nervous System. mBio 2016; 7:mBio.01881-16. [PMID: 27834206 PMCID: PMC5101356 DOI: 10.1128/mbio.01881-16] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The local production of gamma interferon (IFN-γ) is important to control Toxoplasma gondii in the brain, but the basis for these protective effects is not fully understood. The studies presented here reveal that the ability of IFN-γ to inhibit parasite replication in astrocytes in vitro is dependent on signal transducer and activator of transcription 1 (STAT1) and that mice that specifically lack STAT1 in astrocytes are unable to limit parasite replication in the central nervous system (CNS). This susceptibility is associated with a loss of antimicrobial pathways and increased cyst formation in astrocytes. These results identify a critical role for astrocytes in limiting the replication of an important opportunistic pathogen. Astrocytes are the most numerous cell type in the brain, and they are activated in response to many types of neuroinflammation, but their function in the control of CNS-specific infection is unclear. The parasite Toxoplasma gondii is one of the few clinically relevant microorganisms that naturally infects astrocytes, and the studies presented here establish that the ability of astrocytes to inhibit parasite replication is essential for the local control of this opportunistic pathogen. Together, these studies establish a key role for astrocytes as effector cells and in the coordination of many aspects of the protective immune response that operates in the brain.
Collapse
|
42
|
Gaylo A, Schrock DC, Fernandes NRJ, Fowell DJ. T Cell Interstitial Migration: Motility Cues from the Inflamed Tissue for Micro- and Macro-Positioning. Front Immunol 2016; 7:428. [PMID: 27790220 PMCID: PMC5063845 DOI: 10.3389/fimmu.2016.00428] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 09/29/2016] [Indexed: 12/22/2022] Open
Abstract
Effector T cells exit the inflamed vasculature into an environment shaped by tissue-specific structural configurations and inflammation-imposed extrinsic modifications. Once within interstitial spaces of non-lymphoid tissues, T cells migrate in an apparent random, non-directional, fashion. Efficient T cell scanning of the tissue environment is essential for successful location of infected target cells or encounter with antigen-presenting cells that activate the T cell's antimicrobial effector functions. The mechanisms of interstitial T cell motility and the environmental cues that may promote or hinder efficient tissue scanning are poorly understood. The extracellular matrix (ECM) appears to play an important scaffolding role in guidance of T cell migration and likely provides a platform for the display of chemotactic factors that may help to direct the positioning of T cells. Here, we discuss how intravital imaging has provided insight into the motility patterns and cellular machinery that facilitates T cell interstitial migration and the critical environmental factors that may optimize the efficiency of effector T cell scanning of the inflamed tissue. Specifically, we highlight the local micro-positioning cues T cells encounter as they migrate within inflamed tissues, from surrounding ECM and signaling molecules, as well as a requirement for appropriate long-range macro-positioning within distinct tissue compartments or at discrete foci of infection or tissue damage. The central nervous system (CNS) responds to injury and infection by extensively remodeling the ECM and with the de novo generation of a fibroblastic reticular network that likely influences T cell motility. We examine how inflammation-induced changes to the CNS landscape may regulate T cell tissue exploration and modulate function.
Collapse
Affiliation(s)
- Alison Gaylo
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, NY, USA
| | - Dillon C. Schrock
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, NY, USA
| | - Ninoshka R. J. Fernandes
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, NY, USA
| | - Deborah J. Fowell
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, NY, USA
| |
Collapse
|
43
|
|
44
|
Elsheikha HM, Büsselberg D, Zhu XQ. The known and missing links between Toxoplasma gondii and schizophrenia. Metab Brain Dis 2016; 31:749-59. [PMID: 27041387 DOI: 10.1007/s11011-016-9822-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/20/2016] [Indexed: 12/14/2022]
Abstract
Toxoplasma gondii, an intracellular protozoan parasite, has a striking predilection for infecting the Central Nervous System and has been linked to an increased incidence of a number of psychiatric diseases. Several in vitro and in vivo studies have shown that T. gondii infection can affect the structure, bioenergetics and function of brain cells, and alters several host cell processes, including dopaminergic, tryptophan-kynurenine, GABAergic, AKT1, Jak/STAT, and vasopressinergic pathways. These mechanisms underlying the neuropathology of latent toxoplasmosis seem to operate also in schizophrenia, supporting the link between the two disorders. Better understanding of the intricate parasite-neuroglial communications holds the key to unlocking the mystery of T. gondii-mediated schizophrenia and offers substantial prospects for the development of disease-modifying therapies.
Collapse
Affiliation(s)
- Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, Leicestershire, LE12 5RD, UK.
| | - Dietrich Büsselberg
- Weill Cornell Medical College in Qatar, Qatar Foundation - Education City, P.O. Box: 24144, Doha, Qatar
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
| |
Collapse
|
45
|
Pearson YE, Lund AW, Lin AWH, Ng CP, Alsuwaidi A, Azzeh S, Gater DL, Teo JCM. Non-invasive single-cell biomechanical analysis using live-imaging datasets. J Cell Sci 2016; 129:3351-64. [PMID: 27422102 DOI: 10.1242/jcs.191205] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/12/2016] [Indexed: 12/31/2022] Open
Abstract
The physiological state of a cell is governed by a multitude of processes and can be described by a combination of mechanical, spatial and temporal properties. Quantifying cell dynamics at multiple scales is essential for comprehensive studies of cellular function, and remains a challenge for traditional end-point assays. We introduce an efficient, non-invasive computational tool that takes time-lapse images as input to automatically detect, segment and analyze unlabeled live cells; the program then outputs kinematic cellular shape and migration parameters, while simultaneously measuring cellular stiffness and viscosity. We demonstrate the capabilities of the program by testing it on human mesenchymal stem cells (huMSCs) induced to differentiate towards the osteoblastic (huOB) lineage, and T-lymphocyte cells (T cells) of naïve and stimulated phenotypes. The program detected relative cellular stiffness differences in huMSCs and huOBs that were comparable to those obtained with studies that utilize atomic force microscopy; it further distinguished naïve from stimulated T cells, based on characteristics necessary to invoke an immune response. In summary, we introduce an integrated tool to decipher spatiotemporal and intracellular dynamics of cells, providing a new and alternative approach for cell characterization.
Collapse
Affiliation(s)
- Yanthe E Pearson
- Department of Applied Mathematics and Sciences, Khalifa University, P.O. Box 127788, Abu Dhabi, UAE
| | - Amanda W Lund
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Alex W H Lin
- Endothelix, Inc., 2500 West Loop, South Houston, TX 77027, USA
| | - Chee P Ng
- Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, Singapore 138602 Mimetas BV, JH Oortweg 19, Leiden 2333 CH, The Netherlands
| | - Aysha Alsuwaidi
- Department of Biomedical Engineering, Khalifa University, P.O. Box 127788, Abu Dhabi, UAE
| | - Sara Azzeh
- Department of Biomedical Engineering, Khalifa University, P.O. Box 127788, Abu Dhabi, UAE
| | - Deborah L Gater
- Department of Applied Mathematics and Sciences, Khalifa University, P.O. Box 127788, Abu Dhabi, UAE
| | - Jeremy C M Teo
- Department of Biomedical Engineering, Khalifa University, P.O. Box 127788, Abu Dhabi, UAE
| |
Collapse
|
46
|
David CN, Frias ES, Szu JI, Vieira PA, Hubbard JA, Lovelace J, Michael M, Worth D, McGovern KE, Ethell IM, Stanley BG, Korzus E, Fiacco TA, Binder DK, Wilson EH. GLT-1-Dependent Disruption of CNS Glutamate Homeostasis and Neuronal Function by the Protozoan Parasite Toxoplasma gondii. PLoS Pathog 2016; 12:e1005643. [PMID: 27281462 PMCID: PMC4900626 DOI: 10.1371/journal.ppat.1005643] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 04/28/2016] [Indexed: 01/09/2023] Open
Abstract
The immune privileged nature of the CNS can make it vulnerable to chronic and latent infections. Little is known about the effects of lifelong brain infections, and thus inflammation, on the neurological health of the host. Toxoplasma gondii is a parasite that can infect any mammalian nucleated cell with average worldwide seroprevalence rates of 30%. Infection by Toxoplasma is characterized by the lifelong presence of parasitic cysts within neurons in the brain, requiring a competent immune system to prevent parasite reactivation and encephalitis. In the immunocompetent individual, Toxoplasma infection is largely asymptomatic, however many recent studies suggest a strong correlation with certain neurodegenerative and psychiatric disorders. Here, we demonstrate a significant reduction in the primary astrocytic glutamate transporter, GLT-1, following infection with Toxoplasma. Using microdialysis of the murine frontal cortex over the course of infection, a significant increase in extracellular concentrations of glutamate is observed. Consistent with glutamate dysregulation, analysis of neurons reveal changes in morphology including a reduction in dendritic spines, VGlut1 and NeuN immunoreactivity. Furthermore, behavioral testing and EEG recordings point to significant changes in neuronal output. Finally, these changes in neuronal connectivity are dependent on infection-induced downregulation of GLT-1 as treatment with the ß-lactam antibiotic ceftriaxone, rescues extracellular glutamate concentrations, neuronal pathology and function. Altogether, these data demonstrate that following an infection with T. gondii, the delicate regulation of glutamate by astrocytes is disrupted and accounts for a range of deficits observed in chronic infection. The protozoan parasite Toxoplasma gondii infects a third of the world’s population and causes a chronic lifelong infection in the brain of the host. The consequences of such an infection are poorly understood. Here, we demonstrate that Toxoplasma infection can induce profound changes in astrocyte physiology leading to significant disruption of neuronal networks. Pathology can be rescued by upregulating the astrocytic glutamate transporter, GLT-1, restoring concentrations of extracellular glutamate and EEG power. We suggest that such global dysregulation of neurotransmitters should be considered when determining the effects of infection on the CNS.
Collapse
Affiliation(s)
- Clément N. David
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, United States of America
| | - Elma S. Frias
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, United States of America
| | - Jenny I. Szu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, United States of America
| | - Philip A. Vieira
- Department of Psychology, University of California, Riverside, Riverside, California, United States of America
| | - Jacqueline A. Hubbard
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, United States of America
| | - Jonathan Lovelace
- Department of Psychology, University of California, Riverside, Riverside, California, United States of America
| | - Marena Michael
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, United States of America
| | - Danielle Worth
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, United States of America
| | - Kathryn E. McGovern
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, United States of America
| | - Iryna M. Ethell
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, United States of America
| | - B. Glenn Stanley
- Department of Psychology, University of California, Riverside, Riverside, California, United States of America
| | - Edward Korzus
- Department of Psychology, University of California, Riverside, Riverside, California, United States of America
| | - Todd A. Fiacco
- Department of Neuroscience, University of California, Riverside, Riverside, California, United States of America
| | - Devin K. Binder
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, United States of America
| | - Emma H. Wilson
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, United States of America
- * E-mail:
| |
Collapse
|
47
|
Jain R, Tikoo S, Weninger W. Recent advances in microscopic techniques for visualizing leukocytes in vivo. F1000Res 2016; 5:F1000 Faculty Rev-915. [PMID: 27239292 PMCID: PMC4874443 DOI: 10.12688/f1000research.8127.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/12/2016] [Indexed: 12/26/2022] Open
Abstract
Leukocytes are inherently motile and interactive cells. Recent advances in intravital microscopy approaches have enabled a new vista of their behavior within intact tissues in real time. This brief review summarizes the developments enabling the tracking of immune responses in vivo.
Collapse
Affiliation(s)
- Rohit Jain
- Immune Imaging Program, The Centenary Institute, University of Sydney, Newtown, NSW 2042, Australia; Discipline of Dermatology, Sydney Medical School, University of Sydney, NSW 2006, Australia
| | - Shweta Tikoo
- Immune Imaging Program, The Centenary Institute, University of Sydney, Newtown, NSW 2042, Australia; Discipline of Dermatology, Sydney Medical School, University of Sydney, NSW 2006, Australia
| | - Wolfgang Weninger
- Immune Imaging Program, The Centenary Institute, University of Sydney, Newtown, NSW 2042, Australia; Discipline of Dermatology, Sydney Medical School, University of Sydney, NSW 2006, Australia; Department of Dermatology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| |
Collapse
|
48
|
Watanabe R, Kakizaki M, Ikehara Y, Togayachi A. Formation of fibroblastic reticular network in the brain after infection with neurovirulent murine coronavirus. Neuropathology 2016; 36:513-526. [PMID: 27121485 PMCID: PMC7167860 DOI: 10.1111/neup.12302] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/03/2016] [Accepted: 03/05/2016] [Indexed: 01/12/2023]
Abstract
cl‐2 virus is an extremely neurovirulent murine coronavirus. However, during the initial phase of infection between 12 and 24 h post‐inoculation (hpi), the viral antigens are detected only in the meninges, followed by viral spread into the ventricular wall before invasion into the brain parenchyma, indicating that the viruses employ a passage between the meninges and ventricular wall as an entry route into the brain parenchyma. At 48 hpi, the passage was found to be constructed by ER‐TR7 antigen (ERag)‐positive fibers (ERfibs) associated with laminin and collagen III between the fourth ventricle and meninges at the cerebellopontine angle. The construct of the fibers mimics the reticular fibers of the fibroblastic reticular network, which comprises a conduit system in the lymphoid organs. In the meninges, ERfibs together with collagen fibers, lining in a striped pattern, made up a pile of thin sheets. In the brain parenchyma, mature ERfibs associated with laminin were found around blood vessels. Besides mature ERfibs, immature Erfibs without associations with other extracellular matrix components like laminin and collagen appeared after infection, suggesting that the CNS creates a unique conduit system for immune communication triggered by viral invasion.
Collapse
Affiliation(s)
- Rihito Watanabe
- Department of Bioinformatics, Graduate School of Engineering, Soka University, Hachioji, Tokyo, Japan
| | - Masatoshi Kakizaki
- Department of Bioinformatics, Graduate School of Engineering, Soka University, Hachioji, Tokyo, Japan
| | - Yuzuru Ikehara
- Research Center For Medical Glycoscience, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Akira Togayachi
- Research Center For Medical Glycoscience, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| |
Collapse
|
49
|
Immune Surveillance of the CNS following Infection and Injury. Trends Immunol 2016; 36:637-650. [PMID: 26431941 DOI: 10.1016/j.it.2015.08.002] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/10/2015] [Accepted: 08/10/2015] [Indexed: 12/24/2022]
Abstract
The central nervous system (CNS) contains a sophisticated neural network that must be constantly surveyed in order to detect and mitigate a diverse array of challenges. The innate and adaptive immune systems actively participate in this surveillance, which is critical for the maintenance of CNS homeostasis and can facilitate the resolution of infections, degeneration, and tissue damage. Infections and sterile injuries represent two common challenges imposed on the CNS that require a prompt immune response. While the inducers of these two challenges differ in origin, the resultant responses orchestrated by the CNS share some overlapping features. Here, we review how the CNS immunologically discriminates between pathogens and sterile injuries, mobilizes an immune reaction, and, ultimately, regulates local and peripherally-derived immune cells to provide a supportive milieu for tissue repair.
Collapse
|
50
|
Gaylo A, Overstreet MG, Fowell DJ. Imaging CD4 T Cell Interstitial Migration in the Inflamed Dermis. J Vis Exp 2016:e53585. [PMID: 27078264 PMCID: PMC4841317 DOI: 10.3791/53585] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The ability of CD4 T cells to carry out effector functions is dependent upon the rapid and efficient migration of these cells in inflamed peripheral tissues through an as-yet undefined mechanism. The application of multiphoton microscopy to the study of the immune system provides a tool to measure the dynamics of immune responses within intact tissues. Here we present a protocol for non-invasive intravital multiphoton imaging of CD4 T cells in the inflamed mouse ear dermis. Use of a custom imaging platform and a venous catheter allows for the visualization of CD4 T cell dynamics in the dermal interstitium, with the ability to interrogate these cells in real-time via the addition of blocking antibodies to key molecular components involved in motility. This system provides advantages over both in vitro models and surgically invasive imaging procedures. Understanding the pathways used by CD4 T cells for motility may ultimately provide insight into the basic function of CD4 T cells as well as the pathogenesis of both autoimmune diseases and pathology from chronic infections.
Collapse
Affiliation(s)
- Alison Gaylo
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Michael G Overstreet
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Deborah J Fowell
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY;
| |
Collapse
|