101
|
Abstract
Infection with the protozoan parasite Toxoplasma gondii is characterized by asymptomatic latent infection in the central nervous system and skeletal muscle tissue in the majority of immunocompentent individuals. Life-threatening reactivation of the infection in immunocompromized patients originates from rupture of Toxoplasma cysts in the brain. While major progress has been made in our understanding of the immunopathogenesis of infection the mechanism(s) of neuroinvasion of the parasite remains poorly understood. The present review presents the current understanding of blood-brain barrier (patho)physiology and the interaction of Toxoplasma gondii with cells of the blood-brain barrier.
Collapse
Affiliation(s)
- Sabrina M Feustel
- Institute for Microbiology and Hygiene, Charité Medical School, Berlin, Germany
| | | | | |
Collapse
|
102
|
Bhadra R, Gigley JP, Khan IA. The CD8 T-cell road to immunotherapy of toxoplasmosis. Immunotherapy 2012; 3:789-801. [PMID: 21668315 DOI: 10.2217/imt.11.68] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Toxoplasma gondii infection induces a robust CD8 T-cell immunity that is critical for keeping chronic infection under control. In studies using animal models, it has been demonstrated that the absence of this response can compromise the host ability to keep chronic infection under check. Therapeutic agents that facilitate the induction and maintenance of CD8 T-cell response against the pathogen need to be developed. In the last decade, major strides in understanding the development of effector and memory response, particularly in viral and tumor models, have been made. However, factors involved in the generation of effector or memory response against T. gondii infection have not been extensively investigated. This information will be invaluable in designing immunotherapeutic regimens needed for combating this intracellular pathogen that poses a severe risk for pregnant women and immunocompromised individuals.
Collapse
Affiliation(s)
- Rajarshi Bhadra
- Department of Microbiology, Immunology & Tropical Medicine, George Washington University, Washington, DC 20037, USA
| | | | | |
Collapse
|
103
|
Subauste C. Animal models for Toxoplasma gondii infection. CURRENT PROTOCOLS IN IMMUNOLOGY 2012; Chapter 19:19.3.1-19.3.23. [PMID: 22314833 DOI: 10.1002/0471142735.im1903s96] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Toxoplasma gondii is a protozoan of worldwide distribution. This unit describes murine models of acute T. gondii infection, toxoplasmic encephalitis, and Toxoplasma retinochoroiditis. T. gondii infection in SCID mice allows the study of T cell-independent mechanisms of defense. The uracil auxotroph strain cps1-1 and temperature-sensitive mutant strains of T. gondii allow studies of immunization and adoptive transfer. In vivo study of parasite host-interaction is possible with the use of parasites that express fluorescent proteins and model antigens, plus the use of transgenic mice that express the appropriate T cell receptor and fluorescently labeled leukocytes. Parasites that express bioluminescent markers make 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, the maintenance of T. gondii tissue cysts and tachyzoites, as well as preparation of T. gondii lysate antigens.
Collapse
Affiliation(s)
- Carlos Subauste
- Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
104
|
Salmon H, Franciszkiewicz K, Damotte D, Dieu-Nosjean MC, Validire P, Trautmann A, Mami-Chouaib F, Donnadieu E. Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors. J Clin Invest 2012; 122:899-910. [PMID: 22293174 DOI: 10.1172/jci45817] [Citation(s) in RCA: 755] [Impact Index Per Article: 58.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 12/14/2011] [Indexed: 01/01/2023] Open
Abstract
Appropriate localization and migration of T cells is a prerequisite for antitumor immune surveillance. Studies using fixed tumor samples from human patients have shown that T cells accumulate more efficiently in the stroma than in tumor islets, but the mechanisms by which this occurs are unknown. By combining immunostaining and real-time imaging in viable slices of human lung tumors, we revealed that the density and the orientation of the stromal extracellular matrix likely play key roles in controlling the migration of T cells. Active T cell motility, dependent on chemokines but not on β1 or β2 integrins, was observed in loose fibronectin and collagen regions, whereas T cells migrated poorly in dense matrix areas. Aligned fibers in perivascular regions and around tumor epithelial cell regions dictated the migratory trajectory of T cells and restricted them from entering tumor islets. Consistently, matrix reduction with collagenase increased the ability of T cells to contact cancer cells. Thus, the stromal extracellular matrix influences antitumor immunity by controlling the positioning and migration of T cells. Understanding the mechanisms by which this collagen network is generated has the potential to aid in the development of new therapeutics.
Collapse
Affiliation(s)
- Hélène Salmon
- Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
105
|
Evanko SP, Potter-Perigo S, Bollyky PL, Nepom GT, Wight TN. Hyaluronan and versican in the control of human T-lymphocyte adhesion and migration. Matrix Biol 2011; 31:90-100. [PMID: 22155153 DOI: 10.1016/j.matbio.2011.10.004] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 10/10/2011] [Accepted: 10/31/2011] [Indexed: 12/13/2022]
Abstract
The ability of lymphocytes to migrate freely through connective tissues is vital to efficient immune function. How the extracellular matrix (ECM) may affect T-cell adhesion and migration is not well understood. We have examined the adhesion and migration of activated human T-lymphocytes on ECM made by fibroblast-like synoviocytes and lung fibroblasts. These cells were minimally interactive until treated with a viral mimetic, Poly I:C. This treatment promoted myofibroblast formation and engendered a higher-order structured ECM, rich in versican and hyaluronan, to which T-cells avidly adhered in a hyaluronidase-sensitive manner. This Poly I:C-induced matrix impeded T-cell spreading and migration on and through synoviocyte monolayers, while hyaluronidase treatment or adding versican antibody during matrix formation reversed the effect on T-cell migration. Hyaluronidase also reversed the spread myofibroblast morphology. These data suggest that the viscous hyaluronan- and versican-rich matrix binds and constrains T-lymphocytes. Using purified matrix components and solid state matrices of defined composition, we uncovered a role for versican in modulating hyaluronan-T-cell interactions. Versican prevented T-cell binding to soluble hyaluronan, as well as the amoeboid shape change on hyaluronan-coated dishes and T-cell penetration of collagen gels. Together, these data suggest that hyaluronan and versican play a role in T-cell trafficking and function in inflamed tissues.
Collapse
|
106
|
Herz J, Paterka M, Niesner RA, Brandt AU, Siffrin V, Leuenberger T, Birkenstock J, Mossakowski A, Glumm R, Zipp F, Radbruch H. In vivo imaging of lymphocytes in the CNS reveals different behaviour of naïve T cells in health and autoimmunity. J Neuroinflammation 2011; 8:131. [PMID: 21978405 PMCID: PMC3206448 DOI: 10.1186/1742-2094-8-131] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 10/06/2011] [Indexed: 02/08/2023] Open
Abstract
Background Two-photon laser scanning microscopy (TPLSM) has become a powerful tool in the visualization of immune cell dynamics and cellular communication within the complex biological networks of the inflamed central nervous system (CNS). Whereas many previous studies mainly focused on the role of effector or effector memory T cells, the role of naïve T cells as possible key players in immune regulation directly in the CNS is still highly debated. Methods We applied ex vivo and intravital TPLSM to investigate migratory pathways of naïve T cells in the inflamed and non-inflamed CNS. MACS-sorted naïve CD4+ T cells were either applied on healthy CNS slices or intravenously injected into RAG1 -/- mice, which were affected by experimental autoimmune encephalomyelitis (EAE). We further checked for the generation of second harmonic generation (SHG) signals produced by extracellular matrix (ECM) structures. Results By applying TPLSM on living brain slices we could show that the migratory capacity of activated CD4+ T cells is not strongly influenced by antigen specificity and is independent of regulatory or effector T cell phenotype. Naïve T cells, however, cannot find sufficient migratory signals in healthy, non-inflamed CNS parenchyma since they only showed stationary behaviour in this context. This is in contrast to the high motility of naïve CD4+ T cells in lymphoid organs. We observed a highly motile migration pattern for naïve T cells as compared to effector CD4+ T cells in inflamed brain tissue of living EAE-affected mice. Interestingly, in the inflamed CNS we could detect reticular structures by their SHG signal which partially co-localises with naïve CD4+ T cell tracks. Conclusions The activation status rather than antigen specificity or regulatory phenotype is the central requirement for CD4+ T cell migration within healthy CNS tissue. However, under inflammatory conditions naïve CD4+ T cells can get access to CNS parenchyma and partially migrate along inflammation-induced extracellular SHG structures, which are similar to those seen in lymphoid organs. These SHG structures apparently provide essential migratory signals for naïve CD4+ T cells within the diseased CNS.
Collapse
Affiliation(s)
- Josephine Herz
- Klinik für Neurologie, Universitätsklinik Essen, Hufelandstr. 55, 45122 Essen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
107
|
Interferon-gamma- and perforin-mediated immune responses for resistance against Toxoplasma gondii in the brain. Expert Rev Mol Med 2011; 13:e31. [PMID: 22005272 DOI: 10.1017/s1462399411002018] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Toxoplasma gondii is an obligate intracellular protozoan parasite that causes various diseases, including lymphadenitis, congenital infection of fetuses and life-threatening toxoplasmic encephalitis in immunocompromised individuals. Interferon-gamma (IFN-γ)-mediated immune responses are essential for controlling tachyzoite proliferation during both acute acquired infection and reactivation of infection in the brain. Both CD4+ and CD8+ T cells produce this cytokine in response to infection, although the latter has more potent protective activity. IFN-γ can activate microglia, astrocytes and macrophages, and these activated cells control the proliferation of tachyzoites using different molecules, depending on cell type and host species. IFN-γ also has a crucial role in the recruitment of T cells into the brain after infection by inducing expression of the adhesion molecule VCAM-1 on cerebrovascular endothelial cells, and chemokines such as CXCL9, CXCL10 and CCL5. A recent study showed that CD8+ T cells are able to remove T. gondii cysts, which represent the stage of the parasite in chronic infection, from the brain through their perforin-mediated activity. Thus, the resistance to cerebral infection with T. gondii requires a coordinated network using both IFN-γ- and perforin-mediated immune responses. Elucidating how these two protective mechanisms function and collaborate in the brain against T. gondii will be crucial in developing a new method to prevent and eradicate this parasitic infection.
Collapse
|
108
|
John B, Ricart B, Tait Wojno ED, Harris TH, Randall LM, Christian DA, Gregg B, De Almeida DM, Weninger W, Hammer DA, Hunter CA. Analysis of behavior and trafficking of dendritic cells within the brain during toxoplasmic encephalitis. PLoS Pathog 2011; 7:e1002246. [PMID: 21949652 PMCID: PMC3174247 DOI: 10.1371/journal.ppat.1002246] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 07/20/2011] [Indexed: 12/25/2022] Open
Abstract
Under normal conditions the immune system has limited access to the brain; however, during toxoplasmic encephalitis (TE), large numbers of T cells and APCs accumulate within this site. A combination of real time imaging, transgenic reporter mice, and recombinant parasites allowed a comprehensive analysis of CD11c+ cells during TE. These studies reveal that the CNS CD11c+ cells consist of a mixture of microglia and dendritic cells (DCs) with distinct behavior associated with their ability to interact with parasites or effector T cells. The CNS DCs upregulated several chemokine receptors during TE, but none of these individual receptors tested was required for migration of DCs into the brain. However, this process was pertussis toxin sensitive and dependent on the integrin LFA-1, suggesting that the synergistic effect of signaling through multiple chemokine receptors, possibly leading to changes in the affinity of LFA-1, is involved in the recruitment/retention of DCs to the CNS and thus provides new insights into how the immune system accesses this unique site. Toxoplasmic encephalitis (TE), caused by the protozoan parasite Toxoplasma gondii, can be potentially life threatening especially in immuno-compromised individuals. Immune cells including dendritic cells have been shown to accumulate in the brain during chronic toxoplasmosis; however, little is known about their function, their behavior in vivo, and the mechanisms by which they migrate into the brain. In the present studies, we utilize a combination of real time imaging, transgenic reporter mice, and recombinant parasites to reveal the distinct behavior and morphologies of dendritic cells within the brain and their ability to interact with parasites and effector T cells during TE. The CNS DCs were also found to exhibit a unique chemokine receptor expression pattern during infection, and the migration of DCs into the brain was mediated through a pertussis toxin (which blocks signaling downstream of several chemokine receptors) sensitive process and dependent on the integrin LFA-1. There is currently a poor understanding of the events that lead to DC recruitment to the CNS during inflammation in general, and our studies provide new insights into the mechanisms by which antigen-presenting cells gain access to the brain during infection.
Collapse
Affiliation(s)
- Beena John
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Brendon Ricart
- Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Elia D. Tait Wojno
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Tajie H. Harris
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Louise M. Randall
- 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
| | - Beth Gregg
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Daniel Manzoni De Almeida
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Wolfgang Weninger
- The Centenary Institute for Cancer Medicine and Cell Biology, Newtown, Australia
| | - Daniel A. Hammer
- Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
109
|
Ploix CC, Noor S, Crane J, Masek K, Carter W, Lo DD, Wilson EH, Carson MJ. CNS-derived CCL21 is both sufficient to drive homeostatic CD4+ T cell proliferation and necessary for efficient CD4+ T cell migration into the CNS parenchyma following Toxoplasma gondii infection. Brain Behav Immun 2011; 25:883-96. [PMID: 20868739 PMCID: PMC3032828 DOI: 10.1016/j.bbi.2010.09.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Revised: 09/16/2010] [Accepted: 09/16/2010] [Indexed: 12/29/2022] Open
Abstract
Injury, infection and autoimmune triggers increase CNS expression of the chemokine CCL21. Outside the CNS, CCL21 contributes to chronic inflammatory disease and autoimmunity by three mechanisms: recruitment of lymphocytes into injured or infected tissues, organization of inflammatory infiltrates into lymphoid-like structures and promotion of homeostatic CD4+ T-cell proliferation. To test if CCL21 plays the same role in CNS inflammation, we generated transgenic mice with astrocyte-driven expression of CCL21 (GFAP-CCL21 mice). Astrocyte-produced CCL21 was bioavailable and sufficient to support homeostatic CD4+ T-cell proliferation in cervical lymph nodes even in the absence of endogenous CCL19/CCL21. However, lymphocytes and glial-activation were not detected in the brains of uninfected GFAP-CCL21 mice, although CCL21 levels in GFAP-CCL21 brains were higher than levels expressed in inflamed Toxoplasma-infected non-transgenic brains. Following Toxoplasma infection, T-cell extravasation into submeningeal, perivascular and ventricular sites of infected CNS was not CCL21-dependent, occurring even in CCL19/CCL21-deficient mice. However, migration of extravasated CD4+, but not CD8+ T cells from extra-parenchymal CNS sites into the CNS parenchyma was CCL21-dependent. CD4+ T cells preferentially accumulated at perivascular, submeningeal and ventricular spaces in infected CCL21/CCL19-deficient mice. By contrast, greater numbers of CD4+ T cells infiltrated the parenchyma of infected GFAP-CCL21 mice than in wild-type or CCL19/CCL21-deficient mice. Together these data indicate that CCL21 expression within the CNS has the potential to contribute to T cell-mediated CNS pathology via: (a) homeostatic priming of CD4+ T-lymphocytes outside the CNS and (b) by facilitating CD4+ T-cell migration into parenchymal sites following pathogenic insults to the CNS.
Collapse
Affiliation(s)
| | - Shahani Noor
- Division of Biomedical Sciences, Center for Glial-Neuronal Interactions, University of California Riverside
- Graduate Program in Biomedical Sciences, University of California Riverside
| | - Janelle Crane
- Division of Biomedical Sciences, Center for Glial-Neuronal Interactions, University of California Riverside
| | - Kokoechat Masek
- Division of Biomedical Sciences, Center for Glial-Neuronal Interactions, University of California Riverside
| | - Whitney Carter
- Division of Biomedical Sciences, Center for Glial-Neuronal Interactions, University of California Riverside
| | - David D. Lo
- Division of Biomedical Sciences, Center for Glial-Neuronal Interactions, University of California Riverside
| | - Emma H. Wilson
- Division of Biomedical Sciences, Center for Glial-Neuronal Interactions, University of California Riverside
| | - Monica J. Carson
- Division of Biomedical Sciences, Center for Glial-Neuronal Interactions, University of California Riverside
| |
Collapse
|
110
|
Egen JG, Rothfuchs AG, Feng CG, Horwitz MA, Sher A, Germain RN. Intravital imaging reveals limited antigen presentation and T cell effector function in mycobacterial granulomas. Immunity 2011; 34:807-19. [PMID: 21596592 PMCID: PMC3164316 DOI: 10.1016/j.immuni.2011.03.022] [Citation(s) in RCA: 163] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 01/18/2011] [Accepted: 03/02/2011] [Indexed: 01/18/2023]
Abstract
Cell-mediated adaptive immunity is critical for host defense, but little is known about T cell behavior during delivery of effector function. Here we investigate relationships among antigen presentation, T cell motility, and local production of effector cytokines by CD4+ T cells within hepatic granulomas triggered by Bacille Calmette-Guérin or Mycobacterium tuberculosis. At steady-state, only small fractions of mycobacteria-specific T cells showed antigen-induced migration arrest within granulomas, resulting in low-level, polarized secretion of cytokines. However, exogenous antigen elicited rapid arrest and robust cytokine production by the vast majority of effector T cells. These findings suggest that limited antigen presentation and/or recognition within granulomas evoke a muted T cell response drawing on only a fraction of the host's potential effector capacity. Our results provide new insights into the regulation of host-protective functions, especially how antigen availability influences T cell dynamics and, in turn, effector T cell function during chronic infection.
Collapse
Affiliation(s)
- Jackson G. Egen
- Lymphocyte Biology Section, Laboratory of Immunology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Antonio Gigliotti Rothfuchs
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Carl G. Feng
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Marcus A. Horwitz
- Division of Infectious Diseases, Department of Medicine, University of California-Los Angeles School of Medicine, Los Angeles, CA USA
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | - Ronald N. Germain
- Lymphocyte Biology Section, Laboratory of Immunology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
- Program in Systems Immunology and Infectious Disease Modeling, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| |
Collapse
|
111
|
Control of Toxoplasma reactivation by rescue of dysfunctional CD8+ T-cell response via PD-1-PDL-1 blockade. Proc Natl Acad Sci U S A 2011; 108:9196-201. [PMID: 21576466 DOI: 10.1073/pnas.1015298108] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
In this study, we document that Toxoplasma gondii differentiation and reactivation are mediated by systemic CD8 T-cell dysfunction during chronic infection. We demonstrate that CD8(+) T-cell exhaustion occurs despite control of parasitemia during early-chronic toxoplasmosis. During later phases, these cells become exhausted, leading to parasite reactivation and mortality. Concomitant with increased CD8(+) T-cell apoptosis and decreased effector response, this dysfunction is characterized by a graded elevation in expression of inhibitory receptor PD-1 on these cells in both lymphoid and nonlymphoid tissue. Blockade of the PD-1-PDL-1 pathway reinvigorates this suboptimal CD8(+) T-cell response, resulting in control of parasite reactivation and prevention of mortality in chronically infected animals. To the best of our knowledge, this report is unique in showing that exposure to a persistent pathogen despite initial control of parasitemia can lead to CD8(+) T-cell dysfunction and parasite reactivation.
Collapse
|
112
|
Abstract
Viral infections are a major cause of human disease. Although most viruses replicate in peripheral tissues, some have developed unique strategies to move into the nervous system, where they establish acute or persistent infections. Viral infections in the central nervous system (CNS) can alter homeostasis, induce neurological dysfunction and result in serious, potentially life-threatening inflammatory diseases. This Review focuses on the strategies used by neurotropic viruses to cross the barrier systems of the CNS and on how the immune system detects and responds to viral infections in the CNS. A special emphasis is placed on immune surveillance of persistent and latent viral infections and on recent insights gained from imaging both protective and pathogenic antiviral immune responses.
Collapse
Affiliation(s)
- Dorian B McGavern
- National Institute of Neurological Disorders and Stroke, The National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
113
|
Haque A, Best SE, Unosson K, Amante FH, de Labastida F, Anstey NM, Karupiah G, Smyth MJ, Heath WR, Engwerda CR. Granzyme B Expression by CD8+T Cells Is Required for the Development of Experimental Cerebral Malaria. THE JOURNAL OF IMMUNOLOGY 2011; 186:6148-56. [DOI: 10.4049/jimmunol.1003955] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
114
|
Kang SS, Herz J, Kim JV, Nayak D, Stewart-Hutchinson P, Dustin ML, McGavern DB. Migration of cytotoxic lymphocytes in cell cycle permits local MHC I-dependent control of division at sites of viral infection. ACTA ACUST UNITED AC 2011; 208:747-59. [PMID: 21464219 PMCID: PMC3135345 DOI: 10.1084/jem.20101295] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Virus-specific cytotoxic CD8+ T cells are in cell cycle as they transit from lymphoid tissues to sites of infection. After virus infection, cytotoxic T lymphocytes (CTLs) divide rapidly to eradicate the pathogen and prevent the establishment of persistence. The magnitude of an antiviral CTL response is thought to be controlled by the initiation of a cell cycle program within lymphoid tissues. However, it is presently not known whether this division program proceeds during migration or is influenced locally at sites of viral infection. We demonstrate that antiviral CTLs remain in cell cycle while transiting to infected tissues. Up to one third of virus-specific CTLs within blood were found to be in cell cycle after infection with lymphocytic choriomeningitis virus or vesicular stomatitis virus. Using two-photon microscopy, we found that effector CTL divided rapidly upon arrest in the virus-infected central nervous system as well as in meningeal blood vessels. We also observed that MHC I–dependent interactions, but not costimulation, influenced the division program by advancing effector CTL through stages of the cell cycle. These results demonstrate that CTLs are poised to divide in transit and that their numbers can be influenced locally at the site of infection through interactions with cells displaying cognate antigen.
Collapse
Affiliation(s)
- Silvia S Kang
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
115
|
Waite JC, Leiner I, Lauer P, Rae CS, Barbet G, Zheng H, Portnoy DA, Pamer EG, Dustin ML. Dynamic imaging of the effector immune response to listeria infection in vivo. PLoS Pathog 2011; 7:e1001326. [PMID: 21455492 PMCID: PMC3063765 DOI: 10.1371/journal.ppat.1001326] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 02/25/2011] [Indexed: 11/18/2022] Open
Abstract
Host defense against the intracellular pathogen Listeria monocytogenes (Lm) requires innate and adaptive immunity. Here, we directly imaged immune cell dynamics at Lm foci established by dendritic cells in the subcapsular red pulp (scDC) using intravital microscopy. Blood borne Lm rapidly associated with scDC. Myelomonocytic cells (MMC) swarmed around non-motile scDC forming foci from which blood flow was excluded. The depletion of scDC after foci were established resulted in a 10-fold reduction in viable Lm, while graded depletion of MMC resulted in 30-1000 fold increase in viable Lm in foci with enhanced blood flow. Effector CD8+ T cells at sites of infection displayed a two-tiered reduction in motility with antigen independent and antigen dependent components, including stable interactions with infected and non-infected scDC. Thus, swarming MMC contribute to control of Lm prior to development of T cell immunity by direct killing and sequestration from blood flow, while scDC appear to promote Lm survival while preferentially interacting with CD8+ T cells in effector sites.
Collapse
Affiliation(s)
- Janelle C. Waite
- Program in Molecular Pathogenesis, Helen L. and Martin S. Kimmel Center for Biology and Medicine of the Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York City, New York, United States of America
| | - Ingrid Leiner
- Infectious Disease Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, Immunology Program, Sloan-Kettering Institute, New York City, New York, United States of America
| | - Peter Lauer
- Aduro BioTech, Berkeley, California, United States of America
| | - Chris S. Rae
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, United States of America
| | - Gaetan Barbet
- Program in Molecular Pathogenesis, Helen L. and Martin S. Kimmel Center for Biology and Medicine of the Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York City, New York, United States of America
| | - Huan Zheng
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Daniel A. Portnoy
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, United States of America
- School of Public Health, University of California, Berkeley, Berkeley, California, United State of America
| | - Eric G. Pamer
- Infectious Disease Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, Immunology Program, Sloan-Kettering Institute, New York City, New York, United States of America
| | - Michael L. Dustin
- Program in Molecular Pathogenesis, Helen L. and Martin S. Kimmel Center for Biology and Medicine of the Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York City, New York, United States of America
- * E-mail:
| |
Collapse
|
116
|
Fumagalli S, Coles JA, Ejlerskov P, Ortolano F, Bushell TJ, Brewer JM, De Simoni MG, Dever G, Garside P, Maffia P, Carswell HV. In vivo real-time multiphoton imaging of T lymphocytes in the mouse brain after experimental stroke. Stroke 2011; 42:1429-36. [PMID: 21441145 DOI: 10.1161/strokeaha.110.603704] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND PURPOSE To gain a better understanding of T cell behavior after stroke, we have developed real-time in vivo brain imaging of T cells by multiphoton microscopy after middle cerebral artery occlusion. METHODS Adult male hCD2-GFP transgenic mice that exhibit green fluorescent protein-labeled T cells underwent permanent left distal middle cerebral artery occlusion by electrocoagulation (n=6) or sham surgery (n=6) and then multiphoton laser imaging 72 hours later. RESULTS Extravasated T cell number significantly increased after middle cerebral artery occlusion versus sham. Two T cell populations existed after middle cerebral artery occlusion, possibly driven by 2 T cell subpopulations; 1 had significantly lower and the other significantly higher track velocity and displacement rate than sham. CONCLUSIONS The different motilities and behaviors of T cells observed using our imaging approach after stroke could reveal important mechanisms of immune surveillance for future therapeutic exploitations.
Collapse
Affiliation(s)
- Stefano Fumagalli
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
T-cell production of matrix metalloproteinases and inhibition of parasite clearance by TIMP-1 during chronic Toxoplasma infection in the brain. ASN Neuro 2011; 3:e00049. [PMID: 21434872 PMCID: PMC3024837 DOI: 10.1042/an20100027] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Chronic infection with the intracellular protozoan parasite Toxoplasma gondii leads to tissue remodelling in the brain and a continuous requirement for peripheral leucocyte migration within the CNS (central nervous system). In the present study, we investigate the role of MMPs (matrix metalloproteinases) and their inhibitors in T-cell migration into the infected brain. Increased expression of two key molecules, MMP-8 and MMP-10, along with their inhibitor, TIMP-1 (tissue inhibitor of metalloproteinases-1), was observed in the CNS following infection. Analysis of infiltrating lymphocytes demonstrated MMP-8 and -10 production by CD4+ and CD8+ T-cells. In addition, infiltrating T-cells and CNS resident astrocytes increased their expression of TIMP-1 following infection. TIMP-1-deficient mice had a decrease in perivascular accumulation of lymphocyte populations, yet an increase in the proportion of CD4+ T-cells that had trafficked into the CNS. This was accompanied by a reduction in parasite burden in the brain. Taken together, these findings demonstrate a role for MMPs and TIMP-1 in the trafficking of lymphocytes into the CNS during chronic infection in the brain.
Collapse
|
118
|
Norose K, Kikumura A, Luster AD, Hunter CA, Harris TH. CXCL10 is required to maintain T-cell populations and to control parasite replication during chronic ocular toxoplasmosis. Invest Ophthalmol Vis Sci 2011; 52:389-98. [PMID: 20811054 DOI: 10.1167/iovs.10-5819] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
PURPOSE Toxoplasma gondii is a major cause of ocular disease, which can lead to permanent vision loss in humans. T cells are critically involved in parasite control, but little is known about the molecules that promote T-cell trafficking and migration in the retina. Thus, the aim of this study was to image and dissect the T-cell response during chronic toxoplasmic retinochoroiditis. METHODS C57BL/6 mice were infected with the Me49 strain of T. gondii, and T cells that infiltrated the eye were analyzed by flow cytometry and imaged using multiphoton microscopy. IFN-γ, CXCL9, CXCL10, and CXCR3 mRNA levels were measured by real-time PCR. To investigate the role of CXCL10, mice were treated with anti-CXCL10 antibodies, and histopathology and immunohistochemistry were performed to monitor changes in pathology, cellular infiltration, and parasite burden in the eye. RESULTS Infection with T. gondii leads to the infiltration of highly activated motile T cells into the eye. These cells express CXCR3 and are capable of producing IFN-γ and TNF-α, and CD8+ T cells express granzyme B. The expression of CXCL9 and CXCL10 in the retina was significantly upregulated during chronic infection. Treatment of chronically infected mice with anti-CXCL10 antibodies led to decreases in the numbers of CD3+, CD4+, and CD8+ T cells and the amount of IFN-γ mRNA expression in the retina and an increase in replicating parasites and ocular pathology. CONCLUSIONS The maintenance of the T-cell response and the control of T. gondii in the eye during chronic infection is dependent on CXCL10.
Collapse
Affiliation(s)
- Kazumi Norose
- Department of Infection and Host Defense, Graduate School of Medicine, Chiba University, Chiba, Japan
| | | | | | | | | |
Collapse
|
119
|
Balagopalan L, Sherman E, Barr VA, Samelson LE. Imaging techniques for assaying lymphocyte activation in action. Nat Rev Immunol 2011; 11:21-33. [PMID: 21179118 PMCID: PMC3403683 DOI: 10.1038/nri2903] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Imaging techniques have greatly improved our understanding of lymphocyte activation. Technical advances in spatial and temporal resolution and new labelling tools have enabled researchers to directly observe the activation process. Consequently, research using imaging approaches to study lymphocyte activation has expanded, providing an unprecedented level of cellular and molecular detail in the field. As a result, certain models of lymphocyte activation have been verified, others have been revised and yet others have been replaced with new concepts. In this article, we review the current imaging techniques that are used to assess lymphocyte activation in different contexts, from whole animals to single molecules, and discuss the advantages and potential limitations of these methods.
Collapse
Affiliation(s)
- Lakshmi Balagopalan
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | |
Collapse
|
120
|
Millington OR, Myburgh E, Mottram JC, Alexander J. Imaging of the host/parasite interplay in cutaneous leishmaniasis. Exp Parasitol 2010; 126:310-7. [PMID: 20501336 PMCID: PMC3427850 DOI: 10.1016/j.exppara.2010.05.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Revised: 05/17/2010] [Accepted: 05/19/2010] [Indexed: 11/19/2022]
Abstract
An understanding of host-parasite interplay is essential for the development of therapeutics and vaccines. Immunoparasitologists have learned a great deal from 'conventional'in vitro and in vivo approaches, but recent developments in imaging technologies have provided us (immunologists and parasitologists) with the ability to ask new and exciting questions about the dynamic nature of the parasite-immune system interface. These studies are providing us with new insights into the mechanisms involved in the initiation of a Leishmania infection and the consequent induction and regulation of the immune response. Here, we review some of the recent developments and discuss how these observations can be further developed to understand the immunology of cutaneous Leishmania infection in vivo.
Collapse
|
121
|
Wakim LM, Woodward-Davis A, Bevan MJ. Memory T cells persisting within the brain after local infection show functional adaptations to their tissue of residence. Proc Natl Acad Sci U S A 2010; 107:17872-9. [PMID: 20923878 PMCID: PMC2964240 DOI: 10.1073/pnas.1010201107] [Citation(s) in RCA: 437] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The brain is not routinely surveyed by lymphocytes and is defined as an immuno-privileged site. However, viral infection of the brain results in the infiltration and long-term persistence of pathogen-specific CD8(+) T cells. These cells survive without replenishment from the circulation and are referred to as resident memory T cells (Trm). Brain Trm selectively express the integrin CD103, the expression of which is dependent on antigen recognition within the tissue. After clearance of virus, CD8(+) T cells persist in tight clusters, presumably at prior infection hot spots. Antigen persistence is not a prerequisite for T-cell retention, as suggested by the failure to detect viral genomes in the T-cell clusters. Furthermore, we show that an intracranial dendritic cell immunization regimen, which allows the transient introduction of antigen, also results in the generation of memory T cells that persist long term in the brain. Brain Trm die rapidly on isolation from the tissue and fail to undergo recall expansion after adoptive transfer into the bloodstream of antigen-challenged recipients. These ex vivo defects imply a dependency on the local milieu for function and survival. Cumulatively, this work shows that Trm are a specialized population of memory T cells that can be deposited in tissues previously thought to be beyond routine immune surveillance.
Collapse
Affiliation(s)
- Linda M. Wakim
- Department of Immunology, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| | - Amanda Woodward-Davis
- Department of Immunology, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| | - Michael J. Bevan
- Department of Immunology, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| |
Collapse
|
122
|
Renkawitz J, Sixt M. Mechanisms of force generation and force transmission during interstitial leukocyte migration. EMBO Rep 2010; 11:744-50. [PMID: 20865016 DOI: 10.1038/embor.2010.147] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2010] [Accepted: 08/27/2010] [Indexed: 01/12/2023] Open
Abstract
For innate and adaptive immune responses it is essential that inflammatory cells use quick and flexible locomotion strategies. Accordingly, most leukocytes can efficiently infiltrate and traverse almost every physiological or artificial environment. Here, we review how leukocytes might achieve this task mechanistically, and summarize recent findings on the principles of cytoskeletal force generation and transduction at the leading edge of leukocytes. We propose a model in which the cells switch between adhesion-receptor-mediated force transmission and locomotion modes that are based on cellular deformations, but independent of adhesion receptors. This plasticity in migration strategies allows leukocytes to adapt to the geometry and molecular composition of their environment.
Collapse
Affiliation(s)
- Jörg Renkawitz
- Department of Molecular Cell Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | | |
Collapse
|
123
|
John B, Weninger W, Hunter CA. Advances in imaging the innate and adaptive immune response to Toxoplasma gondii. Future Microbiol 2010; 5:1321-8. [PMID: 20860479 PMCID: PMC3032660 DOI: 10.2217/fmb.10.97] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Toxoplasma gondii is an intracellular protozoan parasite that infects a wide variety of warm-blooded hosts and can have devastating effects in the developing fetus as well as the immunocompromised host. An appreciation of how this organism interacts with the host immune system is crucial to understanding the pathogenesis of this disease. The last decade has been marked by the application of various imaging techniques, such as bioluminescent imaging as well as confocal and multiphoton microscopy to study toxoplasmosis. The ability to manipulate parasites to express fluorescent/bioluminescent markers or model antigens/enzymes combined with the development of reporter mice that allow the detection of distinct immune populations have been crucial to the success of many of these studies. These approaches have permitted the visualization of parasites and immune cells in real-time and provided new insights into the nature of host-pathogen interactions. This article highlights some of the advances in imaging techniques, their strengths and weaknesses, and how these techniques have impacted our understanding of the interaction between parasites and various immune populations during toxoplasmosis.
Collapse
Affiliation(s)
- Beena John
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wolfgang Weninger
- The Centenary Institute for Cancer Medicine & Cell Biology, Locked Bag No. 6, Newtown NSW 2042, Australia
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
124
|
Charles E, Joshi S, Ash JD, Fox BA, Farris AD, Bzik DJ, Lang ML, Blader IJ. CD4 T-cell suppression by cells from Toxoplasma gondii-infected retinas is mediated by surface protein PD-L1. Infect Immun 2010; 78:3484-92. [PMID: 20498261 PMCID: PMC2916285 DOI: 10.1128/iai.00117-10] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 02/23/2010] [Accepted: 05/17/2010] [Indexed: 01/22/2023] Open
Abstract
In the inflamed retina, CD4(+) T cells can cause retinal damage when they are not properly regulated. Since tissue expression of major histocompatibility complex (MHC) class II and costimulatory molecules is a key mechanism for regulating effector T cells, we tested the hypothesis that upregulation of these proteins in the retina contributes to the regulation of CD4 T cells. Here we report that in retinas infected with the protozoan parasite Toxoplasma gondii, MHC class II is upregulated on infiltrating leukocytes as well as on resident retinal cells, including photoreceptors. Flow cytometric analysis indicated that B7 costimulatory family members (CD80, CD86, ICOS-L, and programmed death ligand 2 [PD-L2]) were not expressed on class II(+) cells. In contrast, PD-L1 (also named B7-H1 or CD274) was expressed on the majority of both hematopoietic and resident retinal MHC class II-expressing cells. Retinal cells from Toxoplasma-infected animals were able to suppress T-cell activation in a PD-L1-dependent manner. Finally, we demonstrate that the expression of MHC class II and PD-L1 was critically dependent on gamma interferon (IFN-gamma) expression. These data suggest that retinal MHC class II and PD-L1 expression is a novel mechanism by which the retina protects itself from CD4 T-cell-mediated immune damage in ocular toxoplasmosis and other types of retinal immune responses.
Collapse
Affiliation(s)
- Elizabeth Charles
- Department of Microbiology and Immunology, Department of Ophthalmology, University of Oklahoma Health Sciences Center, Arthritis and Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire 03756
| | - Sunil Joshi
- Department of Microbiology and Immunology, Department of Ophthalmology, University of Oklahoma Health Sciences Center, Arthritis and Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire 03756
| | - John D. Ash
- Department of Microbiology and Immunology, Department of Ophthalmology, University of Oklahoma Health Sciences Center, Arthritis and Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire 03756
| | - Barbara A. Fox
- Department of Microbiology and Immunology, Department of Ophthalmology, University of Oklahoma Health Sciences Center, Arthritis and Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire 03756
| | - A. Darise Farris
- Department of Microbiology and Immunology, Department of Ophthalmology, University of Oklahoma Health Sciences Center, Arthritis and Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire 03756
| | - David J. Bzik
- Department of Microbiology and Immunology, Department of Ophthalmology, University of Oklahoma Health Sciences Center, Arthritis and Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire 03756
| | - Mark L. Lang
- Department of Microbiology and Immunology, Department of Ophthalmology, University of Oklahoma Health Sciences Center, Arthritis and Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire 03756
| | - Ira J. Blader
- Department of Microbiology and Immunology, Department of Ophthalmology, University of Oklahoma Health Sciences Center, Arthritis and Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire 03756
| |
Collapse
|
125
|
Mrass P, Petravic J, Davenport MP, Weninger W. Cell-autonomous and environmental contributions to the interstitial migration of T cells. Semin Immunopathol 2010; 32:257-74. [PMID: 20623124 PMCID: PMC2937148 DOI: 10.1007/s00281-010-0212-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Accepted: 05/31/2010] [Indexed: 01/23/2023]
Abstract
A key to understanding the functioning of the immune system is to define the mechanisms that facilitate directed lymphocyte migration to and within tissues. The recent development of improved imaging technologies, most prominently multi-photon microscopy, has enabled the dynamic visualization of immune cells in real-time directly within intact tissues. Intravital imaging approaches have revealed high spontaneous migratory activity of T cells in secondary lymphoid organs and inflamed tissues. Experimental evidence points towards both environmental and cell-intrinsic cues involved in the regulation of lymphocyte motility in the interstitial space. Based on these data, several conceptually distinct models have been proposed in order to explain the coordination of lymphocyte migration both at the single cell and population level. These range from “stochastic” models, where chance is the major driving force, to “deterministic” models, where the architecture of the microenvironment dictates the migratory trajectory of cells. In this review, we focus on recent advances in understanding naïve and effector T cell migration in vivo. In addition, we discuss some of the contradictory experimental findings in the context of theoretical models of migrating leukocytes.
Collapse
Affiliation(s)
- Paulus Mrass
- The Centenary Institute, Locked Bag No. 6, Newtown, New South Wales 2042, Australia.
| | | | | | | |
Collapse
|
126
|
Garside P, Brewer J. In vivo imaging of infection immunology--4I's! Semin Immunopathol 2010; 32:289-96. [PMID: 20607240 DOI: 10.1007/s00281-010-0215-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Accepted: 06/21/2010] [Indexed: 12/30/2022]
Abstract
As predicted by the red queen hypothesis, microbial pathogens are probably the major reason for the evolution of the immune system (Paterson et al., Nature 464:275-278, 2010). In general, at the population level, i.e., for most of us, most of the time, the immune response to infection is highly effective. However, there remain significant challenges with particularly intransigent organisms or those that are crossing species barriers. Thus, in some cases, efforts to develop new and effective vaccines and drugs have met with limited success. To paraphrase Rudyard Kipling, "I keep six honest serving men--they taught me all I know; their names are what, and why, and when and how and where and who". Addressing these key tenets will be key to understanding the interaction between infection and the immune system. This is particularly important, as the early events during induction of an immune response influence the acquisition of effector function and development of memory responses. Our understanding of the interactions of pathogens with the host immune system has largely been derived through in vitro or static in vivo study. This is a significant issue, as the component parts of the immune system do not work in isolation, and their interactions occur in distinct and specialized micro- and macro anatomical locations that can only be assessed in the physiological context, dynamically in vivo. To this end, the increasing availability of genetically manipulable pathogens and high resolution, real-time in vivo imaging over the preceding 5 years has greatly enhanced our ability to understand and evaluate factors involved in host-pathogen interactions in vivo. This article will review the current status of this area, highlight why progress has been faster with some pathogens and tissues (e.g., protozoa and accessible site such as skin), and speculate on what recent developments in biology and imaging will tell us about pathogen-specific immune responses in the future. This will be done by following the chronological development of the infection process from invasion, to recognition, and dissemination.
Collapse
Affiliation(s)
- Paul Garside
- Division of Immunology, Infection & Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK.
| | | |
Collapse
|
127
|
Two-photon microscopy analysis of leukocyte trafficking and motility. Semin Immunopathol 2010; 32:215-25. [PMID: 20603709 PMCID: PMC2937149 DOI: 10.1007/s00281-010-0210-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 05/28/2010] [Indexed: 01/09/2023]
Abstract
During the last several years, live tissue imaging, in particular using two-photon laser microscopy, has advanced our understanding of leukocyte trafficking mechanisms. Studies using this technique are revealing distinct molecular requirements for leukocyte migration in different tissue environments. Also emerging from the studies are the ingenious infrastructures for leukocyte trafficking, which are produced by stromal cells. This review summarizes the recent imaging studies that provided novel mechanistic insights into in vivo leukocyte migration essential for immunosurveillance.
Collapse
|
128
|
|
129
|
Li SC, Tachiki LML, Luo J, Dethlefs BA, Chen Z, Loudon WG. A biological global positioning system: considerations for tracking stem cell behaviors in the whole body. Stem Cell Rev Rep 2010; 6:317-33. [PMID: 20237964 PMCID: PMC2887536 DOI: 10.1007/s12015-010-9130-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Many recent research studies have proposed stem cell therapy as a treatment for cancer, spinal cord injuries, brain damage, cardiovascular disease, and other conditions. Some of these experimental therapies have been tested in small animals and, in rare cases, in humans. Medical researchers anticipate extensive clinical applications of stem cell therapy in the future. The lack of basic knowledge concerning basic stem cell biology-survival, migration, differentiation, integration in a real time manner when transplanted into damaged CNS remains an absolute bottleneck for attempt to design stem cell therapies for CNS diseases. A major challenge to the development of clinical applied stem cell therapy in medical practice remains the lack of efficient stem cell tracking methods. As a result, the fate of the vast majority of stem cells transplanted in the human central nervous system (CNS), particularly in the detrimental effects, remains unknown. The paucity of knowledge concerning basic stem cell biology--survival, migration, differentiation, integration in real-time when transplanted into damaged CNS remains a bottleneck in the attempt to design stem cell therapies for CNS diseases. Even though excellent histological techniques remain as the gold standard, no good in vivo techniques are currently available to assess the transplanted graft for migration, differentiation, or survival. To address these issues, herein we propose strategies to investigate the lineage fate determination of derived human embryonic stem cells (hESC) transplanted in vivo into the CNS. Here, we describe a comprehensive biological Global Positioning System (bGPS) to track transplanted stem cells. But, first, we review, four currently used standard methods for tracking stem cells in vivo: magnetic resonance imaging (MRI), bioluminescence imaging (BLI), positron emission tomography (PET) imaging and fluorescence imaging (FLI) with quantum dots. We summarize these modalities and propose criteria that can be employed to rank the practical usefulness for specific applications. Based on the results of this review, we argue that additional qualities are still needed to advance these modalities toward clinical applications. We then discuss an ideal procedure for labeling and tracking stem cells in vivo, finally, we present a novel imaging system based on our experiments.
Collapse
Affiliation(s)
- Shengwen Calvin Li
- Center for Neuroscience and Stem Cell Research, Children's Hospital of Orange County Research Institute, University of California Irvine, 455 South Main Street, Orange, CA 92868, USA.
| | | | | | | | | | | |
Collapse
|
130
|
Nourshargh S, Hordijk PL, Sixt M. Breaching multiple barriers: leukocyte motility through venular walls and the interstitium. Nat Rev Mol Cell Biol 2010; 11:366-78. [PMID: 20414258 DOI: 10.1038/nrm2889] [Citation(s) in RCA: 424] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The shuttling of leukocytes between the bloodstream and interstitial tissues involves different locomotion strategies that are governed by locally presented soluble and cell-bound signals. Recent studies have furthered our understanding of the rapidly advancing field of leukocyte migration, particularly regarding cellular and subcellular events at the level of the venular wall. Furthermore, emerging cellular models are now addressing the transition from an adherent mode to a non-adherent state, incorporating mechanisms that support an efficient migratory profile of leukocytes in the interstitial tissue beyond the venular wall.
Collapse
Affiliation(s)
- Sussan Nourshargh
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, William Harvey Research Institute, Charterhouse Square, London, UK.
| | | | | |
Collapse
|
131
|
Wilson EH, Weninger W, Hunter CA. Trafficking of immune cells in the central nervous system. J Clin Invest 2010; 120:1368-79. [PMID: 20440079 DOI: 10.1172/jci41911] [Citation(s) in RCA: 394] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The CNS is an immune-privileged environment, yet the local control of multiple pathogens is dependent on the ability of immune cells to access and operate within this site. However, inflammation of the distinct anatomical sites (i.e., meninges, cerebrospinal fluid, and parenchyma) associated with the CNS can also be deleterious. Therefore, control of lymphocyte entry and migration within the brain is vital to regulate protective and pathological responses. In this review, several recent advances are highlighted that provide new insights into the processes that regulate leukocyte access to, and movement within, the brain.
Collapse
Affiliation(s)
- Emma H Wilson
- Division of Biomedical Sciences, University of California, Riverside, California 92521, USA.
| | | | | |
Collapse
|
132
|
Coombes JL, Robey EA. Dynamic imaging of host–pathogen interactions in vivo. Nat Rev Immunol 2010; 10:353-64. [DOI: 10.1038/nri2746] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
133
|
Beattie L, Peltan A, Maroof A, Kirby A, Brown N, Coles M, Smith DF, Kaye PM. Dynamic imaging of experimental Leishmania donovani-induced hepatic granulomas detects Kupffer cell-restricted antigen presentation to antigen-specific CD8 T cells. PLoS Pathog 2010; 6:e1000805. [PMID: 20300603 PMCID: PMC2837408 DOI: 10.1371/journal.ppat.1000805] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Accepted: 02/03/2010] [Indexed: 01/16/2023] Open
Abstract
Kupffer cells (KCs) represent the major phagocytic population within the liver and provide an intracellular niche for the survival of a number of important human pathogens. Although KCs have been extensively studied in vitro, little is known of their in vivo response to infection and their capacity to directly interact with antigen-specific CD8(+) T cells. Here, using a combination of approaches including whole mount and thin section confocal microscopy, adoptive cell transfer and intra-vital 2-photon microscopy, we demonstrate that KCs represent the only detectable population of mononuclear phagocytes within granulomas induced by Leishmania donovani infection that are capable of presenting parasite-derived peptide to effector CD8(+) T cells. This restriction of antigen presentation to KCs within the Leishmania granuloma has important implications for the identification of new candidate vaccine antigens and for the design of novel immuno-therapeutic interventions.
Collapse
Affiliation(s)
- Lynette Beattie
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Adam Peltan
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Asher Maroof
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Alun Kirby
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Najmeeyah Brown
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Mark Coles
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Deborah F. Smith
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| | - Paul M. Kaye
- Centre for Immunology and Infection, Hull York Medical School and Department of Biology, University of York, York, United Kingdom
| |
Collapse
|
134
|
T cell receptor triggering by force. Trends Immunol 2010; 31:1-6. [PMID: 19836999 DOI: 10.1016/j.it.2009.09.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Revised: 09/21/2009] [Accepted: 09/24/2009] [Indexed: 11/18/2022]
Abstract
Antigen recognition through the interaction between the T cell receptor (TCR) and peptide presented by major histocompatibility complex (pMHC) is the first step in T cell-mediated immune responses. How this interaction triggers TCR signalling that leads to T cell activation is still unclear. Taking into account the mechanical stress exerted on the pMHC-TCR interaction at the dynamic interface between T cells and antigen presenting cells (APCs), we propose the so-called receptor deformation model of TCR triggering. In this model, TCR conformational change induced by mechanical forces initiates TCR signalling. The receptor deformation model, for the first time, explains all three aspects of the TCR triggering puzzle: mechanism, specificity, and sensitivity.
Collapse
|
135
|
Abstract
The chemokine receptor CCR7 is a well-established homing receptor for dendritic cells and T cells. Interactions with its ligands, CCL19 and CCL21, facilitate priming of immune responses in lymphoid tissue, yet CCR7-independent immune responses can be generated in the presence of sufficient antigen. In these studies, we investigated the role of CCR7 signaling in the generation of protective immune responses to the intracellular protozoan parasite Toxoplasma gondii. The results demonstrated a significant increase in the expression of CCL19, CCL21, and CCR7 in peripheral and central nervous system (CNS) tissues over the course of infection. Unexpectedly, despite the presence of abundant antigen, CCR7 was an absolute requirement for protective immunity to T. gondii, as CCR7(-/-) mice succumbed to the parasite early in the acute phase of infection. Although serum levels of interleukin 12 (IL-12), IL-6, tumor necrosis factor alpha (TNF-alpha), and IL-10 remained unchanged, there was a significant decrease in CCL2/monocyte chemoattractant protein 1 (MCP-1) and inflammatory monocyte recruitment to the site of infection. In addition, CCR7(-/-) mice failed to produce sufficient gamma interferon (IFN-gamma), a critical Th1-associated effector cytokine required to control parasite replication. As a result, there was increased parasite dissemination and a significant increase in parasite burden in the lungs, livers, and brains of infected mice. Adoptive-transfer experiments revealed that expression of CCR7 on the T-cell compartment alone is sufficient to enable T-cell priming, increase IFN-gamma production, and allow the survival of CCR7(-/-) mice. These data demonstrate an absolute requirement for T-cell expression of CCR7 for the generation of protective immune responses to Toxoplasma infection.
Collapse
|
136
|
Mueller SN, Hickman HD. In vivo imaging of the T cell response to infection. Curr Opin Immunol 2010; 22:293-8. [PMID: 20080040 DOI: 10.1016/j.coi.2009.12.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Accepted: 12/28/2009] [Indexed: 10/20/2022]
Abstract
The induction and execution of a successful anti-pathogen immune response requires a consecutive series of cellular interactions that begin in lymphoid environments and later extend into the periphery. Much of our current knowledge of these events has been gained using ex vivo approaches that yield important static information but do not convey the dynamic nature of cellular behavior in vivo. The application of multiphoton-laser based microscopic analysis to the ongoing immune response has provided new insight into cellular interactions leading to T cell activation and the behavior of primed immune effectors. Here we discuss recent insights on anti-pathogen immune responses revealed using live imaging of both lymphoid and non-lymphoid tissues.
Collapse
Affiliation(s)
- Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria, Australia.
| | | |
Collapse
|
137
|
Cerebral malaria: why experimental murine models are required to understand the pathogenesis of disease. Parasitology 2009; 137:755-72. [PMID: 20028608 DOI: 10.1017/s0031182009991715] [Citation(s) in RCA: 171] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Cerebral malaria is a life-threatening complication of malaria infection. The pathogenesis of cerebral malaria is poorly defined and progress in understanding the condition is severely hampered by the inability to study in detail, ante-mortem, the parasitological and immunological events within the brain that lead to the onset of clinical symptoms. Experimental murine models have been used to investigate the sequence of events that lead to cerebral malaria, but there is significant debate on the merits of these models and whether their study is relevant to human disease. Here we review the current understanding of the parasitological and immunological events leading to human and experimental cerebral malaria, and explain why we believe that studies with experimental models of CM are crucial to define the pathogenesis of the condition.
Collapse
|
138
|
Abstract
Leishmaniasis is a chronic infection in which intracellular parasites avoid destruction by the immune system. Using intravital imaging, Filipe-Santos et al. (2009) demonstrate that some parasitized dendritic cells receive much less attention than others during their choreographed dance with T cells, suggesting that these "wallflowers" could allow for parasite survival.
Collapse
Affiliation(s)
- Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
139
|
Two-photon laser scanning microscopy imaging of intact spinal cord and cerebral cortex reveals requirement for CXCR6 and neuroinflammation in immune cell infiltration of cortical injury sites. J Immunol Methods 2009; 352:89-100. [PMID: 19800886 DOI: 10.1016/j.jim.2009.09.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 09/22/2009] [Accepted: 09/24/2009] [Indexed: 01/10/2023]
Abstract
The mouse spinal cord is an important site for autoimmune and injury models. Skull thinning surgery provides a minimally invasive window for microscopy of the mouse cerebral cortex, but there are no parallel methods for the spinal cord. We introduce a novel, facile and inexpensive method for two-photon laser scanning microscopy of the intact spinal cord in the mouse by taking advantage of the naturally accessible intervertebral space. These are powerful methods when combined with gene-targeted mice in which endogenous immune cells are labeled with green fluorescent protein (GFP). We first demonstrate that generation of the intervertebral window does not elicit a reaction of GFP(+) microglial cells in CX3CR1(gfp/+) mice. We next demonstrate a distinct rostrocaudal migration of GFP(+) immune cells in the spinal cord of CXCR6(gfp/+) mice during active experimental autoimmune encephalomyelitis (EAE). Interestingly, infiltration of the cerebral cortex by GFP(+) cells in these mice required three conditions: EAE induction, cortical injury and expression of CXCR6 on immune cells.
Collapse
|
140
|
Lang T, Lecoeur H, Prina E. Imaging Leishmania development in their host cells. Trends Parasitol 2009; 25:464-73. [DOI: 10.1016/j.pt.2009.07.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Revised: 06/10/2009] [Accepted: 07/07/2009] [Indexed: 12/13/2022]
|
141
|
John B, Harris TH, Tait ED, Wilson EH, Gregg B, Ng LG, Mrass P, Roos DS, Dzierszinski F, Weninger W, Hunter CA. Dynamic Imaging of CD8(+) T cells and dendritic cells during infection with Toxoplasma gondii. PLoS Pathog 2009; 5:e1000505. [PMID: 19578440 PMCID: PMC2700268 DOI: 10.1371/journal.ppat.1000505] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Accepted: 06/05/2009] [Indexed: 12/21/2022] Open
Abstract
To better understand the initiation of CD8+ T cell responses during infection, the primary response to the intracellular parasite Toxoplasma gondii was characterized using 2-photon microscopy combined with an experimental system that allowed visualization of dendritic cells (DCs) and parasite specific CD8+ T cells. Infection with T. gondii induced localization of both these populations to the sub-capsular/interfollicular region of the draining lymph node and DCs were required for the expansion of the T cells. Consistent with current models, in the presence of cognate antigen, the average velocity of CD8+ T cells decreased. Unexpectedly, infection also resulted in modulation of the behavior of non-parasite specific T cells. This TCR-independent process correlated with the re-modeling of the lymph node micro-architecture and changes in expression of CCL21 and CCL3. Infection also resulted in sustained interactions between the DCs and CD8+ T cells that were visualized only in the presence of cognate antigen and were limited to an early phase in the response. Infected DCs were rare within the lymph node during this time frame; however, DCs presenting the cognate antigen were detected. Together, these data provide novel insights into the earliest interaction between DCs and CD8+ T cells and suggest that cross presentation by bystander DCs rather than infected DCs is an important route of antigen presentation during toxoplasmosis. Toxoplasma gondii is a protozoan parasite that can infect a wide range of hosts, including humans. Infection with T. gondii is potentially life threatening in immuno-compromised individuals and it can be detrimental during pregnancy, often leading to abortion of the fetus. Dendritic cells are thought to play a vital role in the development of protective immunity to Toxoplasma gondii through their ability to produce immunological signals such as cytokines and also process and present parasite derived peptides to T cells. However, little is known about the actual interactions between these cell types in an intact organ, such as the lymph node, during infection. Using the technology of live imaging by 2-photon microscopy we have identified a very early window of time during infection when dendritic cells and T cells make sustained contacts with one another, which appears crucial for the generation of protective responses. We also show that substantial changes are induced in the lymph node micro-architecture as a result of infection, which in turn could have effects on immune responses to secondary pathogens. Understanding the interaction between these immune cells in vivo that leads to resistance to active infection would help in the design of better strategies to develop protective immune responses against this pathogen in immuno-compromised individuals.
Collapse
Affiliation(s)
- Beena John
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Tajie H. Harris
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Elia D. Tait
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Emma H. Wilson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Division of Biomedical Sciences, University of California, Riverside, California, United States of America
| | - Beth Gregg
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Lai Guan Ng
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
- The Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales, Australia
| | - Paulus Mrass
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
- The Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales, Australia
| | - David S. Roos
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | | | - Wolfgang Weninger
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
- The Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales, Australia
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
142
|
'Guided' tour of the brain. Nat Rev Immunol 2009. [DOI: 10.1038/nri2509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|