Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
2.
PLoS Pathog ; 20(1): e1011710, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38206985

ABSTRACT

Toxoplasma gondii is an obligate intracellular parasite that infects one-third of the world's human population and establishes infection in the brain. Cerebral immune cell infiltration is critical for controlling the parasite, but little is known about the molecular cues guiding immune cells to the brain during infection. Activated astrocytes produce CCL2, a chemokine that mediates inflammatory monocyte recruitment to tissues by binding to the CCR2 receptor. We detected elevated CCL2 production in the brains of C57BL/6J mice by 15 days after T. gondii infection. Utilizing confocal microscopy and intracellular flow cytometry, we identified microglia and brain-infiltrating myeloid cells as the main producers of CCL2 during acute infection, and CCL2 was specifically produced in regions of parasite infection in the brain. In contrast, astrocytes became the dominant CCL2 producer during chronic T. gondii infection. To determine the role of astrocyte-derived CCL2 in mobilizing immune cells to the brain and controlling T. gondii infection, we generated GFAP-Cre x CCL2fl/fl mice, in which astrocytes are deficient in CCL2 production. We observed significantly decreased immune cell recruitment and increased parasite burden in the brain during chronic, but not acute, infection of mice deficient in astrocyte CCL2 production, without an effect on peripheral immune responses. To investigate potential mechanisms explaining the reduced control of T. gondii infection, we analyzed key antimicrobial and immune players in host defense against T. gondii and detected a reduction in iNOS+ myeloid cells, and T. gondii-specific CD4+ T cells in the knockout mice. These data uncover a critical role for astrocyte-derived CCL2 in immune cell recruitment and parasite control in the brain during chronic, but not acute, T. gondii infection.


Subject(s)
Toxoplasma , Toxoplasmosis , Animals , Humans , Mice , Astrocytes/metabolism , Brain/metabolism , Chemokine CCL2/metabolism , Mice, Inbred C57BL , Mice, Knockout , Toxoplasma/metabolism , Toxoplasmosis/metabolism
3.
Am J Pathol ; 194(2): 225-237, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38065361

ABSTRACT

Cerebral edema frequently develops in the setting of brain infection and can contribute to elevated intracranial pressure, a medical emergency. How excess fluid is cleared from the brain is not well understood. Previous studies have shown that interstitial fluid is transported out of the brain along perivascular channels that collect into the cerebrospinal fluid (CSF)-filled subarachnoid space. CSF is then removed from the central nervous system through venous and lymphatic routes. The current study tested the hypothesis that increasing lymphatic drainage of CSF would promote clearance of cerebral edema fluid during infection with the neurotropic parasite Toxoplasma gondii. Fluorescent microscopy and magnetic resonance imaging was used to show that C57BL/6 mice develop vasogenic edema 4 to 5 weeks after infection with T. gondii. Tracer experiments were used to evaluate how brain infection affects meningeal lymphatic function, which demonstrated a decreased rate in CSF outflow in T. gondii-infected mice. Next, mice were treated with a vascular endothelial growth factor (VEGF)-C-expressing viral vector, which induced meningeal lymphangiogenesis and improved CSF outflow in chronically infected mice. No difference in cerebral edema was observed between mice that received VEGF-C and those that rececived sham treatment. Therefore, although VEGF-C treatment can improve lymphatic outflow in mice infected with T. gondii, this effect does not lead to increased clearance of edema fluid from the brains of these mice.


Subject(s)
Brain Edema , Toxoplasma , Toxoplasmosis , Vascular Endothelial Growth Factor C , Animals , Mice , Brain/pathology , Brain Edema/parasitology , Brain Edema/therapy , Mice, Inbred C57BL , Toxoplasmosis/complications , Toxoplasmosis/therapy , Vascular Endothelial Growth Factor C/therapeutic use
4.
PLoS Pathog ; 18(6): e1010296, 2022 06.
Article in English | MEDLINE | ID: mdl-35727849

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.


Subject(s)
Toxoplasma , Toxoplasmosis , Animals , CD8-Positive T-Lymphocytes , Immunologic Memory , Mice , Receptors, Antigen, T-Cell
5.
PLoS Pathog ; 18(9): e1010637, 2022 09.
Article in English | MEDLINE | ID: mdl-36067217

ABSTRACT

Toxoplasma gondii is a ubiquitous intracellular protozoan parasite that establishes a life-long chronic infection largely restricted to the central nervous system (CNS). Constant immune pressure, notably IFN-γ-STAT1 signaling, is required for preventing fatal pathology during T. gondii infection. Here, we report that abrogation of STAT1 signaling in microglia, the resident immune cells of the CNS, is sufficient to induce a loss of parasite control in the CNS and susceptibility to toxoplasmic encephalitis during the early stages of chronic infection. Using a microglia-specific genetic labeling and targeting system that discriminates microglia from blood-derived myeloid cells that infiltrate the brain during infection, we find that, contrary to previous in vitro reports, microglia do not express inducible nitric-oxide synthase (iNOS) during T. gondii infection in vivo. Instead, transcriptomic analyses of microglia reveal that STAT1 regulates both (i) a transcriptional shift from homeostatic to "disease-associated microglia" (DAM) phenotype conserved across several neuroinflammatory models, including T. gondii infection, and (ii) the expression of anti-parasitic cytosolic molecules that are required for eliminating T. gondii in a cell-intrinsic manner. Further, genetic deletion of Stat1 from microglia during T. gondii challenge leads to fatal pathology despite largely equivalent or enhanced immune effector functions displayed by brain-infiltrating immune populations. Finally, we show that microglial STAT1-deficiency results in the overrepresentation of the highly replicative, lytic tachyzoite form of T. gondii, relative to its quiescent, semi-dormant bradyzoite form typical of chronic CNS infection. Our data suggest an overall protective role of CNS-resident microglia against T. gondii infection, illuminating (i) general mechanisms of CNS-specific immunity to infection (ii) and a clear role for IFN-STAT1 signaling in regulating a microglial activation phenotype observed across diverse neuroinflammatory disease states.


Subject(s)
Encephalitis , STAT1 Transcription Factor , Toxoplasma , Toxoplasmosis, Cerebral , Animals , Brain/pathology , Encephalitis/metabolism , Encephalitis/pathology , Mice , Microglia/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Toxoplasma/metabolism , Toxoplasmosis, Cerebral/metabolism
7.
Immunity ; 42(4): 679-91, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25902482

ABSTRACT

Mutations in MECP2, encoding the epigenetic regulator methyl-CpG-binding protein 2, are the predominant cause of Rett syndrome, a disease characterized by both neurological symptoms and systemic abnormalities. Microglial dysfunction is thought to contribute to disease pathogenesis, and here we found microglia become activated and subsequently lost with disease progression in Mecp2-null mice. Mecp2 was found to be expressed in peripheral macrophage and monocyte populations, several of which also became depleted in Mecp2-null mice. RNA-seq revealed increased expression of glucocorticoid- and hypoxia-induced transcripts in Mecp2-deficient microglia and peritoneal macrophages. Furthermore, Mecp2 was found to regulate inflammatory gene transcription in response to TNF stimulation. Postnatal re-expression of Mecp2 using Cx3cr1(creER) increased the lifespan of otherwise Mecp2-null mice. These data suggest that Mecp2 regulates microglia and macrophage responsiveness to environmental stimuli to promote homeostasis. Dysfunction of tissue-resident macrophages might contribute to the systemic pathologies observed in Rett syndrome.


Subject(s)
CpG Islands/immunology , Epigenesis, Genetic , Macrophages, Peritoneal/immunology , Methyl-CpG-Binding Protein 2/immunology , Microglia/immunology , Rett Syndrome/immunology , Animals , CX3C Chemokine Receptor 1 , DNA Methylation , Disease Models, Animal , Female , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Homeostasis/immunology , Humans , Integrases/genetics , Integrases/immunology , Longevity/immunology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/pathology , Male , Methyl-CpG-Binding Protein 2/deficiency , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Microglia/pathology , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology , Rett Syndrome/genetics , Rett Syndrome/pathology , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology
8.
PLoS Pathog ; 16(10): e1009027, 2020 10.
Article in English | MEDLINE | ID: mdl-33108405

ABSTRACT

It is of great interest to understand how invading pathogens are sensed within the brain, a tissue with unique challenges to mounting an immune response. The eukaryotic parasite Toxoplasma gondii colonizes the brain of its hosts, and initiates robust immune cell recruitment, but little is known about pattern recognition of T. gondii within brain tissue. The host damage signal IL-33 is one protein that has been implicated in control of chronic T. gondii infection, but, like many other pattern recognition pathways, IL-33 can signal peripherally, and the specific impact of IL-33 signaling within the brain is unclear. Here, we show that IL-33 is expressed by oligodendrocytes and astrocytes during T. gondii infection, is released locally into the cerebrospinal fluid of T. gondii-infected animals, and is required for control of infection. IL-33 signaling promotes chemokine expression within brain tissue and is required for the recruitment and/or maintenance of blood-derived anti-parasitic immune cells, including proliferating, IFN-γ-expressing T cells and iNOS-expressing monocytes. Importantly, we find that the beneficial effects of IL-33 during chronic infection are not a result of signaling on infiltrating immune cells, but rather on radio-resistant responders, and specifically, astrocytes. Mice with IL-33 receptor-deficient astrocytes fail to mount an adequate adaptive immune response in the CNS to control parasite burden-demonstrating, genetically, that astrocytes can directly respond to IL-33 in vivo. Together, these results indicate a brain-specific mechanism by which IL-33 is released locally, and sensed locally, to engage the peripheral immune system in controlling a pathogen.


Subject(s)
Astrocytes/immunology , Interleukin-33/immunology , Toxoplasmosis, Cerebral/immunology , Adult , Animals , Astrocytes/metabolism , Astrocytes/physiology , Brain/metabolism , Female , Humans , Immunity , Interferon-gamma/immunology , Interleukin-33/metabolism , Male , Mice , Mice, Inbred C57BL , Monocytes/immunology , Signal Transduction , Toxoplasma/metabolism , Toxoplasma/parasitology , Toxoplasmosis/metabolism , Toxoplasmosis, Cerebral/metabolism
9.
Immunity ; 37(3): 511-23, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22981537

ABSTRACT

Interferon-γ (IFN-γ) promotes a population of T-bet(+) CXCR3(+) regulatory T (Treg) cells that limit T helper 1 (Th1) cell-mediated pathology. Our studies demonstrate that interleukin-27 (IL-27) also promoted expression of T-bet and CXCR3 in Treg cells. During infection with Toxoplasma gondii, a similar population emerged that limited T cell responses and was dependent on IFN-γ in the periphery but on IL-27 at mucosal sites. Transfer of Treg cells ameliorated the infection-induced pathology observed in Il27(-/-) mice, and this was dependent on their ability to produce IL-10. Microarray analysis revealed that Treg cells exposed to either IFN-γ or IL-27 have distinct transcriptional profiles. Thus, IFN-γ and IL-27 have different roles in Treg cell biology and IL-27 is a key cytokine that promotes the development of Treg cells specialized to control Th1 cell-mediated immunity at local sites of inflammation.


Subject(s)
Interferon-gamma/pharmacology , Interleukin-17/pharmacology , Salmonella Infections, Animal/immunology , T-Lymphocytes, Regulatory/drug effects , Toxoplasmosis, Animal/immunology , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Female , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Receptors, CXCR3/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/pathology , Salmonella typhimurium/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Toxoplasma/immunology , Toxoplasmosis, Animal/parasitology , Toxoplasmosis, Animal/pathology
10.
J Immunol ; 202(6): 1755-1766, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30718297

ABSTRACT

Control of chronic CNS infection with the parasite Toxoplasma gondii requires ongoing T cell responses in the brain. Immunosuppressive cytokines are also important for preventing lethal immunopathology during chronic infection. To explore the loss of suppressive cytokines exclusively during the chronic phase of infection, we blocked IL-10R in chronically infected mice. Consistent with previous reports, IL-10R blockade led to severe, fatal tissue destruction associated with widespread changes in the inflammatory response, including increased APC activation, expansion of CD4+ T cells, and neutrophil recruitment to the brain. We then sought to identify regulatory mechanisms contributing to IL-10 production, focusing on ICOS, a molecule implicated in IL-10 production. Unexpectedly, ICOS ligand (ICOSL) blockade led to a local expansion of effector T cells in the brain without affecting IL-10 production or APC activation. Instead, we found that ICOSL blockade led to changes in T cells associated with their proliferation and survival. We observed increased expression of IL-2-associated signaling molecules CD25, STAT5 phosphorylation, Ki67, and Bcl-2 in T cells in the brain, along with decreased apoptosis. Interestingly, increases in CD25 and Bcl-2 were not observed following IL-10R blockade. Also, unlike IL-10R blockade, ICOSL blockade led to an expansion of both CD8+ and CD4+ T cells in the brain, with no expansion of peripheral T cells or neutrophil recruitment to the brain and no severe tissue destruction. Overall, these results suggest that IL-10 and ICOS differentially regulate T cell responses in the brain during chronic T. gondii infection.


Subject(s)
Brain/parasitology , Inducible T-Cell Co-Stimulator Protein/immunology , Interleukin-10/immunology , T-Lymphocytes/immunology , Toxoplasmosis/immunology , Animals , Brain/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL
11.
Nature ; 523(7560): 337-41, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26030524

ABSTRACT

One of the characteristics of the central nervous system is the lack of a classical lymphatic drainage system. Although it is now accepted that the central nervous system undergoes constant immune surveillance that takes place within the meningeal compartment, the mechanisms governing the entrance and exit of immune cells from the central nervous system remain poorly understood. In searching for T-cell gateways into and out of the meninges, we discovered functional lymphatic vessels lining the dural sinuses. These structures express all of the molecular hallmarks of lymphatic endothelial cells, are able to carry both fluid and immune cells from the cerebrospinal fluid, and are connected to the deep cervical lymph nodes. The unique location of these vessels may have impeded their discovery to date, thereby contributing to the long-held concept of the absence of lymphatic vasculature in the central nervous system. The discovery of the central nervous system lymphatic system may call for a reassessment of basic assumptions in neuroimmunology and sheds new light on the aetiology of neuroinflammatory and neurodegenerative diseases associated with immune system dysfunction.


Subject(s)
Central Nervous System/anatomy & histology , Central Nervous System/immunology , Lymphatic Vessels/anatomy & histology , Lymphatic Vessels/immunology , Animals , Central Nervous System/cytology , Cranial Sinuses/anatomy & histology , Female , Humans , Immune Tolerance/immunology , Immunologic Surveillance/immunology , Lymphatic Vessels/cytology , Male , Meninges/anatomy & histology , Meninges/cytology , Meninges/immunology , Mice, Inbred C57BL , T-Lymphocytes/cytology , T-Lymphocytes/immunology
12.
Nat Immunol ; 8(12): 1363-71, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17994025

ABSTRACT

Interleukin 10 (IL-10) has a prominent function in regulating the balance between protective and pathological T cell responses. Consistent with that activity, many sources of this cytokine are found in vivo, including from myeloid cells and a variety of T cell subsets. However, although there are many pathways that regulate innate production of IL-10, the factors that govern its synthesis by the adaptive response are poorly understood. Here we report that IL-27 and IL-6 induced T helper type 1 and type 2 cells, as well as T helper cells that produce IL-17, to secrete IL-10. This effect was dependent on the transcription factors STAT1 and STAT3 for IL-27 and on STAT3 for IL-6. Our studies identify a previously unknown pathway that allows the immune system to temper inflammatory responses.


Subject(s)
Interleukin-10/biosynthesis , Interleukin-17/physiology , Interleukin-6/physiology , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/physiology , T-Lymphocytes/immunology , Animals , Humans , Interleukin-10/metabolism , Interleukin-17/metabolism , Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Helper-Inducer
13.
J Immunol ; 198(10): 4054-4061, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28389591

ABSTRACT

Regulatory T cells (Tregs) play an important role in the CNS during multiple infections, as well as autoimmune inflammation, but the behavior of this cell type in the CNS has not been explored. In mice, infection with Toxoplasma gondii leads to a Th1-polarized parasite-specific effector T cell response in the brain. Similarly, Tregs in the CNS during T. gondii infection are Th1 polarized, as exemplified by their T-bet, CXCR3, and IFN-γ expression. Unlike effector CD4+ T cells, an MHC class II tetramer reagent specific for T. gondii did not recognize Tregs isolated from the CNS. Likewise, TCR sequencing revealed minimal overlap in TCR sequence between effector T cells and Tregs in the CNS. Whereas effector T cells are found in the brain parenchyma where parasites are present, Tregs were restricted to the meninges and perivascular spaces. The use of intravital imaging revealed that activated CD4+ T cells within the meninges were highly migratory, whereas Tregs moved more slowly and were found in close association with CD11c+ cells. To test whether the behavior of Tregs in the meninges is influenced by interactions with CD11c+ cells, mice were treated with anti-LFA-1 Abs to reduce the number of CD11c+ cells in this space. The anti-LFA-1 treatment led to fewer contacts between Tregs and the remaining CD11c+ cells and increased the speed of Treg migration. These data suggest that Tregs are anatomically restricted within the CNS, and their interaction with CD11c+ populations regulates their local behavior during T. gondii infection.


Subject(s)
CD11c Antigen/immunology , Meninges/immunology , T-Lymphocytes, Regulatory/physiology , Toxoplasmosis, Cerebral/immunology , Animals , CD11c Antigen/genetics , CD11c Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Movement , Intravital Microscopy , Lymphocyte Activation , Mice , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Toxoplasma/immunology
14.
Trends Immunol ; 36(10): 569-577, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26431936

ABSTRACT

Whereas the study of the interactions between the immune system and the central nervous system (CNS) has often focused on pathological conditions, the importance of neuroimmune communication in CNS homeostasis and function has become clear over that last two decades. Here we discuss the progression of our understanding of the interaction between the peripheral immune system and the CNS. We examine the notion of immune privilege of the CNS in light of both earlier findings and recent studies revealing a functional meningeal lymphatic system that drains cerebrospinal fluid (CSF) to the deep cervical lymph nodes, and consider the implications of a revised perspective on the immune privilege of the CNS on the etiology and pathology of different neurological disorders.


Subject(s)
Central Nervous System/immunology , Animals , Central Nervous System/physiology , Central Nervous System Diseases/immunology , Humans , Immune System/physiology , Lymphatic Diseases/immunology , Lymphatic System/physiology
15.
Immunity ; 30(2): 300-11, 2009 Feb 20.
Article in English | MEDLINE | ID: mdl-19167248

ABSTRACT

To understand lymphocyte behavior in the brain, we used two-photon microscopy to visualize effector CD8(+) T cells during toxoplasmic encephalitis. These cells displayed multiple behaviors with two distinct populations of cells apparent: one with a constrained pattern of migration and one with a highly migratory subset. The proportion of these populations varied over time associated with changes in antigen availability as well as T cell expression of the inhibitory receptor PD1. Unexpectedly, the movement of infiltrating cells was closely associated with an infection-induced reticular system of fibers. This observation suggests that, whereas in other tissues pre-existing scaffolds exist that guide lymphocyte migration, in the brain specialized structures are induced by inflammation that guide migration of T cells in this immune-privileged environment.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Toxoplasma/immunology , Toxoplasmosis, Cerebral/immunology , Toxoplasmosis, Cerebral/parasitology , Animals , Central Nervous System/immunology , Mice , Rats , Toxoplasmosis, Cerebral/pathology
16.
Nature ; 486(7404): 545-8, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22722867

ABSTRACT

Chemokines have a central role in regulating processes essential to the immune function of T cells, such as their migration within lymphoid tissues and targeting of pathogens in sites of inflammation. Here we track T cells using multi-photon microscopy to demonstrate that the chemokine CXCL10 enhances the ability of CD8+ T cells to control the pathogen Toxoplasma gondii in the brains of chronically infected mice. This chemokine boosts T-cell function in two different ways: it maintains the effector T-cell population in the brain and speeds up the average migration speed without changing the nature of the walk statistics. Notably, these statistics are not Brownian; rather, CD8+ T-cell motility in the brain is well described by a generalized Lévy walk. According to our model, this unexpected feature enables T cells to find rare targets with more than an order of magnitude more efficiency than Brownian random walkers. Thus, CD8+ T-cell behaviour is similar to Lévy strategies reported in organisms ranging from mussels to marine predators and monkeys, and CXCL10 aids T cells in shortening the average time taken to find rare targets.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Movement , Chemokine CXCL10/immunology , Animals , Brain/immunology , Brain/microbiology , Chemokine CXCL10/antagonists & inhibitors , Chemokine CXCL10/genetics , Female , Ligands , Male , Mice , Mice, Inbred C57BL , Models, Immunological , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism , Time Factors , Toxoplasma/growth & development , Toxoplasma/immunology
18.
PLoS Comput Biol ; 11(2): e1004058, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25692801

ABSTRACT

The three-dimensional positions of immune cells can be tracked in live tissues precisely as a function of time using two-photon microscopy. However, standard methods of analysis used in the field and experimental artifacts can bias interpretations and obscure important aspects of cell migration such as directional migration and non-Brownian walk statistics. Therefore, methods were developed for minimizing drift artifacts, identifying directional and anisotropic (asymmetric) migration, and classifying cell migration statistics. These methods were applied to describe the migration statistics of CD8+ T cells in uninflamed lymph nodes. Contrary to current models, CD8+ T cell statistics are not well described by a straightforward persistent random walk model. Instead, a model in which one population of cells moves via Brownian-like motion and another population follows variable persistent random walks with noise reproduces multiple statistical measures of CD8+ T cell migration in the lymph node in the absence of inflammation.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Cell Movement/physiology , Lymph Nodes/cytology , Models, Immunological , Animals , Cells, Cultured , Computational Biology , Computer Simulation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spleen/cytology
19.
Infect Immun ; 82(3): 1343-53, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24396042

ABSTRACT

A chronic infection with the parasite Toxoplasma gondii has previously been shown to protect mice against subsequent viral, bacterial, or protozoal infections. Here we have shown that a chronic T. gondii infection can prevent Plasmodium berghei ANKA-induced experimental cerebral malaria (ECM) in C57BL/6 mice. Treatment with soluble T. gondii antigens (STAg) reduced parasite sequestration and T cell infiltration in the brains of P. berghei-infected mice. Administration of STAg also preserved blood-brain barrier function, reduced ECM symptoms, and significantly decreased mortality. STAg treatment 24 h post-P. berghei infection led to a rapid increase in serum levels of interleukin 12 (IL-12) and gamma interferon (IFN-γ). By 5 days after P. berghei infection, STAg-treated mice had reduced IFN-γ levels compared to those of mock-treated mice, suggesting that reductions in IFN-γ at the time of ECM onset protected against lethality. Using IL-10- and IL-12ßR-deficient mice, we found that STAg-induced protection from ECM is IL-10 independent but IL-12 dependent. Treatment of P. berghei-infected mice with recombinant IL-12 significantly decreased parasitemia and mortality. These data suggest that IL-12, either induced by STAg or injected as a recombinant protein, mediates protection from ECM-associated pathology potentially through early induction of IFN-γ and reduction in parasitemia. These results highlight the importance of early IL-12 induction in protection against ECM.


Subject(s)
Interleukin-12/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Toxoplasma/immunology , Up-Regulation/immunology , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/microbiology , Brain/immunology , Brain/microbiology , CHO Cells , Cricetulus , Interferon-gamma/immunology , Interleukin-10/immunology , Malaria, Cerebral/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Parasitemia/immunology , Parasitemia/microbiology , Receptors, Interleukin-11/immunology , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , Toxoplasmosis/immunology , Toxoplasmosis/microbiology
20.
PLoS Pathog ; 7(9): e1002246, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21949652

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.


Subject(s)
Brain/immunology , Dendritic Cells/immunology , Encephalitis/immunology , Toxoplasma/immunology , Toxoplasmosis, Cerebral/immunology , Adoptive Transfer , Animals , Brain/parasitology , CD11c Antigen/analysis , Cell Movement , Dendritic Cells/cytology , Dendritic Cells/physiology , Encephalitis/parasitology , Lymphocyte Function-Associated Antigen-1/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/parasitology , Pertussis Toxin/pharmacology , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , T-Lymphocytes/immunology , Toxoplasmosis, Cerebral/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL