Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Med Sci Educ ; 31(3): 1025-1028, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34457946

ABSTRACT

Medical schools are moving away from traditional lectures in favor of small-group learning. Here we present a game-based activity designed to improve student engagement while serving as a course review. The activity incorporated 32 questions submitted by eleven discipline directors. The 133 student participants reviewed the session positively, rating it highest of the 11 course sessions with an overall quality of a 4.68 (± 0.84) out of a 6-point Likert scale. The students remarked that the activity was fun and engaging, yet long. It reviewed a breadth of content over several specialties in a format that encouraged active, team-based learning.

2.
PLoS One ; 15(9): e0239157, 2020.
Article in English | MEDLINE | ID: mdl-32960910

ABSTRACT

CD4dimCD8bright T cells, a genuine population of CD8+ T cells, are highly activated and cytolytic. Recently, the low affinity IgG Fc fragment receptor CD32a was described as marker of HIV latency while others reported that CD32a is associated with T cell activation. Given that we have previously established that CD4dimCD8bright T cells are highly activated, mediate anti-HIV responses, and are infected by HIV, we assessed here CD32 expression on CD4dimCD8bright T cells in context of HIV. CD32 frequency on peripheral CD4dimCD8bright and CD4+ T cells was determined by flow cytometry among HIV negative and HIV positive patients. We report that among HIV- individuals, mean CD32 percent expression was 60% on CD4dimCD8bright T cells and 17% on CD4+ T cells (p<0.01). Among HIV+ patients, mean CD32 percent expression was 54% on CD4dimCD8bright T cells and 12% on CD4+ T cells (p<0.001). CD32 expression on CD4dimCD8bright T cells did not correlate with CD4 count and viral load and was not different by HIV serostatus. CD32 was also higher on other double positive T cell populations in both HIV negative and HIV positive donors in comparison to their single positive T cell counterpart. Together, these studies indicate that CD32 is enriched on double positive T cells regardless of HIV serostatus. The functional role of CD32 on these double positive T cells remains to be elucidated.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , HIV Infections/blood , Receptors, IgG/metabolism , T-Lymphocyte Subsets/immunology , Anti-HIV Agents/therapeutic use , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Separation , Flow Cytometry , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/virology , HIV-1/isolation & purification , Humans , Receptors, IgG/immunology , T-Lymphocyte Subsets/metabolism , Viral Load
3.
PLoS Pathog ; 16(6): e1008381, 2020 06.
Article in English | MEDLINE | ID: mdl-32525948

ABSTRACT

HIV invades the brain during acute infection. Yet, it is unknown whether long-lived infected brain cells release productive virus that can egress from the brain to re-seed peripheral organs. This understanding has significant implication for the brain as a reservoir for HIV and most importantly HIV interplay between the brain and peripheral organs. Given the sheer number of astrocytes in the human brain and their controversial role in HIV infection, we evaluated their infection in vivo and whether HIV infected astrocytes can support HIV egress to peripheral organs. We developed two novel models of chimeric human astrocyte/human peripheral blood mononuclear cells: NOD/scid-IL-2Rgc null (NSG) mice (huAstro/HuPBMCs) whereby we transplanted HIV (non-pseudotyped or VSVg-pseudotyped) infected or uninfected primary human fetal astrocytes (NHAs) or an astrocytoma cell line (U138MG) into the brain of neonate or adult NSG mice and reconstituted the animals with human peripheral blood mononuclear cells (PBMCs). We also transplanted uninfected astrocytes into the brain of NSG mice and reconstituted with infected PBMCs to mimic a biological infection course. As expected, the xenotransplanted astrocytes did not escape/migrate out of the brain and the blood brain barrier (BBB) was intact in this model. We demonstrate that astrocytes support HIV infection in vivo and egress to peripheral organs, at least in part, through trafficking of infected CD4+ T cells out of the brain. Astrocyte-derived HIV egress persists, albeit at low levels, under combination antiretroviral therapy (cART). Egressed HIV evolved with a pattern and rate typical of acute peripheral infection. Lastly, analysis of human cortical or hippocampal brain regions of donors under cART revealed that astrocytes harbor between 0.4-5.2% integrated HIV gag DNA and 2-7% are HIV gag mRNA positive. These studies establish a paradigm shift in the dynamic interaction between the brain and peripheral organs which can inform eradication of HIV reservoirs.


Subject(s)
Astrocytes , Blood-Brain Barrier , HIV Infections , HIV-1/metabolism , Hippocampus , Virus Release , Animals , Anti-Retroviral Agents/pharmacology , Astrocytes/metabolism , Astrocytes/pathology , Astrocytes/virology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Blood-Brain Barrier/virology , Cell Line, Tumor , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/pathology , HIV-1/genetics , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/virology , Humans , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID
4.
Aging Cell ; 16(5): 956-965, 2017 10.
Article in English | MEDLINE | ID: mdl-28612507

ABSTRACT

Emerging evidence suggests that cell senescence plays an important role in aging-associated diseases including neurodegenerative diseases. HIV leads to a spectrum of neurologic diseases collectively termed HIV-associated neurocognitive disorders (HAND). Drug abuse, particularly methamphetamine (meth), is a frequently abused psychostimulant among HIV+ individuals and its abuse exacerbates HAND. The mechanism by which HIV and meth lead to brain cell dysregulation is not entirely clear. In this study, we evaluated the impact of HIV and meth on astrocyte senescence using in vitro and several animal models. Astrocytes constitute up to 50% of brain cells and play a pivotal role in marinating brain homeostasis. We show here that HIV and meth induce significant senescence of primary human fetal astrocytes, as evaluated by induction of senescence markers (ß-galactosidase and p16INK4A ), senescence-associated morphologic changes, and cell cycle arrest. HIV- and meth-mediated astrocyte senescence was also demonstrated in three small animal models (humanized mouse model of HIV/NSG-huPBMCs, HIV-transgenic rats, and in a meth administration rat model). Senescent astrocytes in turn mediated neuronal toxicity. Further, we show that ß-catenin, a pro-survival/proliferation transcriptional co-activator, is downregulated by HIV and meth in human astrocytes and this downregulation promotes astrocyte senescence while induction of ß-catenin blocks HIV- and meth-mediated astrocyte senescence. These studies, for the first time, demonstrate that HIV and meth induce astrocyte senescence and implicate the ß-catenin pathway as potential therapeutic target to overcome astrocyte senescence.


Subject(s)
AIDS Dementia Complex/genetics , Astrocytes/metabolism , Central Nervous System Stimulants/adverse effects , Methamphetamine/adverse effects , Neurons/metabolism , Substance-Related Disorders/genetics , beta Catenin/genetics , AIDS Dementia Complex/complications , AIDS Dementia Complex/metabolism , AIDS Dementia Complex/physiopathology , Animals , Animals, Genetically Modified , Astrocytes/pathology , Astrocytes/virology , Calcium-Binding Proteins , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression Regulation , HIV-1/pathogenicity , HIV-1/physiology , Humans , Male , Mice , Microfilament Proteins , Neurons/pathology , Neurons/virology , Primary Cell Culture , Rats , Signal Transduction , Substance-Related Disorders/complications , Substance-Related Disorders/metabolism , Substance-Related Disorders/physiopathology , beta Catenin/metabolism
5.
Glia ; 64(1): 105-21, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26337870

ABSTRACT

Multiple sclerosis (MS) frequently starts near the lateral ventricles, which are lined by subventricular zone (SVZ) progenitor cells that can migrate to lesions and contribute to repair. Because MS-induced inflammation may decrease SVZ proliferation and thus limit repair, we studied the role of galectin-3 (Gal-3), a proinflammatory protein. Gal-3 expression was increased in periventricular regions of human MS in post-mortem brain samples and was also upregulated in periventricular regions in a murine MS model, Theiler's murine encephalomyelitis virus (TMEV) infection. Whereas TMEV increased SVZ chemokine (CCL2, CCL5, CCL, and CXCL10) expression in wild type (WT) mice, this was inhibited in Gal-3(-/-) mice. Though numerous CD45+ immune cells entered the SVZ of WT mice after TMEV infection, their numbers were significantly diminished in Gal-3(-/-) mice. TMEV also reduced neuroblast and proliferative SVZ cell numbers in WT mice but this was restored in Gal-3(-/-) mice and was correlated with increased numbers of doublecortin+ neuroblasts in the corpus callosum. In summary, our data showed that loss of Gal-3 blocked chemokine increases after TMEV, reduced immune cell migration into the SVZ, reestablished SVZ proliferation and increased the number of progenitors in the corpus callosum. These results suggest Gal-3 plays a central role in modulating the SVZ neurogenic niche's response to this model of MS.


Subject(s)
Brain/metabolism , Galectin 3/metabolism , Multiple Sclerosis/metabolism , Nervous System Autoimmune Disease, Experimental/metabolism , Neurogenesis , Stem Cell Niche/physiology , Adolescent , Adult , Aged , Animals , Brain/immunology , Brain/pathology , Cell Movement , Child , Female , Galectin 3/genetics , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Nervous System Autoimmune Disease, Experimental/immunology , Nervous System Autoimmune Disease, Experimental/pathology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Poliomyelitis/metabolism , Poliomyelitis/pathology , Theilovirus , Young Adult
6.
J Immunol ; 196(1): 317-27, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26582945

ABSTRACT

The role of CD8(+) T cells in HIV control in the brain and the consequences of such control are unclear. Approximately 3% of peripheral CD8(+) T cells dimly express CD4 on their surface. This population is known as CD4(dim)CD8(bright) T cells. We evaluated the role of CD4(dim)CD8(bright) and CD8 single positive T cells in HIV-infected brain using NOD/SCID/IL-2rcγ(-/-) mice reconstituted with human PBMCs (NSG-huPBMC). All three T cell populations (CD4 single positive, CD8 single positive, and CD4(dim)CD8(bright)) were found in NSG-huPBMC mouse brain within 2 wk of infection. Wnts secreted from astrocytes induced CD4(dim)CD8(bright) T cells by 2-fold in vitro. Injection of highly purified CD8 single positive T cells into mouse brain induced CD4(dim)CD8(bright) T cells by 10-fold, which were proliferative and exhibited a terminally differentiated effector memory phenotype. Brain CD4(dim)CD8(bright) T cells from HIV-infected mice exhibited anti-HIV-specific responses, as demonstrated by induction of CD107ab post exposure to HIV peptide-loaded targets. Further, higher frequency of CD4(dim)CD8(bright) T cells (R = -0.62; p ≤ 0.001), but not CD8 single positive T cells (R = -0.24; p ≤ 0.27), negatively correlated with HIV gag mRNA transcripts in HIV-infected NSG-huPBMC brain. Together, these studies indicate that single positive CD8(+) T cells entering the CNS during HIV infection can give rise to CD4(dim)CD8(bright) T cells, likely through a Wnt signaling-dependent manner, and that these cells are associated with potent anti-HIV control in the CNS. Thus, CD4(dim)CD8(bright) T cells are capable of HIV control in the CNS and may offer protection against HIV-associated neurocognitive disorders.


Subject(s)
Brain/immunology , CD4 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , Wnt Signaling Pathway/immunology , Animals , Astrocytes/metabolism , Brain/cytology , Brain/virology , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cell Movement/immunology , Cells, Cultured , HIV Infections/virology , Heterografts , Interleukin Receptor Common gamma Subunit/genetics , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/transplantation , Lymphocyte Activation/immunology , Lysosomal-Associated Membrane Protein 1/metabolism , Lysosomal-Associated Membrane Protein 2/metabolism , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Spleen/cytology
8.
Glia ; 63(3): 441-51, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25331637

ABSTRACT

HIV-mediated neuropathogenesis is a multifaceted process involving several players, including resident brain cells (neurons, astrocytes, and microglia) and infiltrating cells [peripheral blood mononuclear cells (PBMCs)]. We evaluated the dynamic interaction between astrocytes and infiltrating PBMCs as it impacts HIV in the CNS. We demonstrate that human primary-derived astrocytes (PDAs) predominantly secrete Wnt 1, 2b, 3, 5b, and 10b. Wnts are small secreted glycoproteins that initiate either ß-catenin-dependent or independent signal transduction. The Wnt pathway plays a vital role in the regulation of CNS activities including neurogenesis, neurotransmitter release, synaptic plasticity, and memory consolidation. We show that HIV infection of PDAs altered astrocyte Wnt profile by elevating Wnts 2b and 10b. Astrocyte conditioned media (ACM) inhibited HIV replication in PBMCs by 50%. Removal of Wnts from ACM abrogated its ability to suppress HIV replication in PBMCs. Inversely, PBMCs supernatant activated PDAs, as demonstrated by a 10-fold increase in HLA-DR and a 5-fold increase in IFNγ expression, and enhanced astrocyte susceptibility to HIV by 2-fold, which was mediated by IFNγ in a Stat-3-dependent manner. Collectively, these data demonstrate a dynamic interaction between astrocytes and PBMCs, whereby astrocyte-secreted Wnts exert an anti-HIV effect on infected PBMCs and PBMCs, in turn, secrete IFNγ that enhance astrocyte susceptibility to productive HIV infection and mediate their activation.


Subject(s)
AIDS Dementia Complex/physiopathology , Astrocytes/physiology , Leukocytes, Mononuclear/physiology , AIDS Dementia Complex/virology , Astrocytes/virology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Culture Media, Conditioned , HIV/physiology , HLA-DR Antigens/metabolism , Humans , Interferon-gamma/metabolism , Leukocytes, Mononuclear/virology , STAT3 Transcription Factor/metabolism , Wnt Proteins/metabolism
9.
Curr HIV Res ; 12(3): 164-73, 2014.
Article in English | MEDLINE | ID: mdl-24862328

ABSTRACT

Cells of the monocyte/macrophage lineage are an important target for HIV-1 infection. They are often at anatomical sites linked to HIV-1 transmission and are an important vehicle for disseminating HIV-1 throughout the body, including the central nervous system. Monocytes do not support extensive productive HIV-1 replication, but they become more susceptible to HIV-1infection as they differentiate into macrophages. The mechanisms guiding susceptibility of HIV-1 replication in monocytes versus macrophages are not entirely clear. We determined whether endogenous activity of ß-catenin signaling impacts differential susceptibility of monocytes and monocyte-derived macrophages (MDMs) to productive HIV-1 replication. We show that monocytes have an approximately 4-fold higher activity of ß-catenin signaling than MDMs. Inducing ß-catenin in MDMs suppressed HIV-1 replication by 5-fold while inhibiting endogenous ß-catenin signaling in monocytes by transfecting with a dominant negative mutant for the downstream effector of ß- catenin (TCF-4) promoted productive HIV-1 replication by 6-fold. These findings indicate that ß-catenin/TCF-4 is an important pathway for restricted HIV-1 replication in monocytes and plays a significant role in potentiating HIV-1 replication as monocytes differentiate into macrophages. Targeting this pathway may provide a novel strategy to purge the latent reservoir from monocytes/macrophages, especially in sanctuary sites for HIV-1 such as the central nervous system.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , HIV-1/physiology , Macrophages/immunology , Monocytes/immunology , Signal Transduction , Transcription Factors/metabolism , Virus Replication , beta Catenin/metabolism , HIV-1/immunology , Humans , Macrophages/virology , Monocytes/virology , Transcription Factor 4
10.
AIDS Res Hum Retroviruses ; 30(7): 677-84, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24593860

ABSTRACT

HIV infection is a risk factor for the tumorigenesis including non-AIDS-defining cancers such as those of the gastrointestinal tract. However, the mechanisms underlying such cancer outgrowth are still unknown. Furthermore, combined HIV/cancer studies are difficult to evaluate using primate models or in the clinical patient setting. To understand the mechanisms of tumor outgrowth in the context of HIV infection, we adopted a humanized mouse model permissive to infection and cancer as well as an in vivo humanized mouse challenge with colon cancer in the context of HIV infection. Immunodeficient NOD SCID IL-2R(-/-) mice were immunologically reconstituted by adoptive transfer of 10(7) HIV-negative donor peripheral blood leukocytes and challenged with 10(6) HCT116 human colon cancer cells. A group of mice was treated with antiretroviral therapy. Tumor microenvironment and epithelial tissues in the context of HIV infection were analyzed using immunohistochemistry. We demonstrate that HIV-infected humanized mice develop significantly larger tumors than uninfected mice (p<0.05). Epithelial cell proliferation in HIV-infected mice is significantly enhanced in comparison to proliferation in uninfected mice (p<0.01). Moreover, the activation of ß-catenin, an important step in intestinal epithelial cell proliferation and tumorigenesis, is elevated in the tumors of HIV-infected mice (p<0.0001). Importantly, antiretroviral therapy reverses these pathological processes independently of CD4(+) T cell return. These findings model the ability of HIV infection to result in tumor outgrowth that is evident in HIV-positive patients and lend insight into previously unrecognized mechanisms that may underlie this pathology.


Subject(s)
Gastrointestinal Neoplasms/immunology , Gastrointestinal Neoplasms/pathology , HIV Infections/immunology , HIV-1/immunology , Leukocytes/immunology , Adoptive Transfer , Animals , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/immunology , Carcinogenesis , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Gastrointestinal Neoplasms/virology , HCT116 Cells , HIV Infections/drug therapy , HIV Infections/virology , Humans , Leukocyte Transfusion , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Receptors, Interleukin-2/genetics , beta Catenin/biosynthesis
11.
PLoS One ; 9(3): e92159, 2014.
Article in English | MEDLINE | ID: mdl-24647048

ABSTRACT

Wnts are small secreted glycoproteins that are highly conserved among species. To date, 19 Wnts have been described, which initiate a signal transduction cascade that is either ß-catenin dependent or independent, culminating in the regulation of hundreds of target genes. Extracellular release of Wnts is dependent on lipidation of Wnts by porcupine, a membrane-bound-O-acyltransferase protein in the endoplasmic reticulum. Studies demonstrating the requirement of porcupine for Wnts production are based on cell line and non-human primary cells. We evaluated the requirement for porcupine for Wnts production in human primary astrocytes and CD8+ T cells. Using IWP-2, an inhibitor of porcupine, or siRNA targeting porcupine, we demonstrate that porcupine is not required for the release of Wnt 1, 3, 5b, 6,7a, 10b, and 16a. While IWP had no effect on Wnt 2b release, knockdown of porcupine by siRNA reduced Wnt 2b release by 60%. These data indicate that porcupine-mediated production of Wnts is context dependent and is not required for all Wnts production, suggesting that alternative mechanisms exist for Wnts production.


Subject(s)
Astrocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Membrane Proteins/metabolism , Wnt Proteins/biosynthesis , Acyltransferases , Animals , Cells, Cultured , Fetus/cytology , Humans , Mice , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , beta Catenin/metabolism
12.
Methods Mol Biol ; 900: 381-401, 2012.
Article in English | MEDLINE | ID: mdl-22933080

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) and Theiler's Murine Encephalitis Virus-Induced Demyelinating Disease (TMEV-IDD) are two clinically relevant murine models of multiple sclerosis (MS). Like MS, both are characterized by mononuclear cell infiltration into the CNS and demyelination. EAE is induced by either the administration of myelin protein or peptide in adjuvant or by the adoptive transfer of encephalitogenic T cell blasts into naïve recipients. The relative merits of each of these protocols are compared. Depending on the type of question being asked, different mouse strains and peptides are used. Different disease courses are observed with different strains and different peptides in active EAE. These variations are also addressed. Additionally, issues relevant to clinical grading of EAE in mice are discussed. In addition to EAE induction, useful references for other disease indicators such as DTH, in vitro proliferation, and immunohistochemistry are provided. TMEV-IDD is a useful model for understanding the possible viral etiology of MS. This section provides detailed information on the preparation of viral stocks and subsequent intracerebral infection of mice. Additionally, virus plaque assay and clinical disease assessment are discussed. Recently, recombinant TMEV strains have been created for the study of molecular mimicry which incorporate various 30 amino acid myelin epitopes within the leader region of TMEV.


Subject(s)
Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/virology , Immunologic Techniques/methods , Multiple Sclerosis/pathology , Theilovirus/physiology , Adoptive Transfer , Amino Acid Sequence , Animals , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/immunology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Multiple Sclerosis/immunology , Peptides/chemistry , Vaccination
13.
PLoS One ; 6(10): e26143, 2011.
Article in English | MEDLINE | ID: mdl-22022540

ABSTRACT

FasL expression on human immune cells and cancer cells plays important roles in immune homeostasis and in cancer development. Our previous study suggests that polymorphisms in the FasL promoter can significantly affect the gene expression in human cells. In addition to the functional FasL SNP -844C>T (rs763110), three other SNPs (SNP -756A>G or rs2021837, SNP -478A>T or rs41309790, and SNP -205 C>G or rs74124371) exist in the proximal FasL promoter. In the current study, we established three major FasL hyplotypes in humans. Interestingly, a transcription motif search revealed that the FasL promoter possessed two consensus T-cell factor (TCF/LEF1) binding elements (TBEs), which is either polymorphic (SNP -205C>G) or close to the functional SNP -844C>T. Subsequently, we demonstrate that both FasL TBEs formed complexes with the TCF-4 and ß-catenin transcription factors in vitro and in vivo. Co-transfection of LEF-1 and ß-catenin transcription factors significantly increased FasL promoter activities, suggesting that FasL is a target gene of the ß-catenin/T-cell factor pathway. More importantly, we found that the rare allele (-205G) of the polymorphic FasL TBE (SNP -205C>G) failed to bind the TCF-4 transcription factor and that SNP -205 C>G significantly affected the promoter activity. Furthermore, promoter reporter assays revealed that FasL SNP haplotypes influenced promoter activities in human colon cancer cells and in human T cells. Finally, ß-catenin knockdown significantly decreased the FasL expression in human SW480 colon cancer cells. Collectively, our data suggest that ß-catenin may be involved in FasL gene regulation and that FasL expression is influenced by FasL SNP haplotypes, which may have significant implications in immune response and tumorigenesis.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Fas Ligand Protein/genetics , Haplotypes/genetics , Promoter Regions, Genetic/genetics , Signal Transduction , Transcription Factors/metabolism , beta Catenin/metabolism , Base Sequence , Binding Sites , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Fas Ligand Protein/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lymphoid Enhancer-Binding Factor 1/metabolism , Molecular Sequence Data , Polymorphism, Single Nucleotide/genetics , Protein Binding , T-Lymphocytes/metabolism , Transcription Factor 4 , Up-Regulation/genetics
14.
J Autoimmun ; 36(2): 142-54, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21273044

ABSTRACT

Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease (TMEV-IDD) serves as virus-induced model of chronic progressive multiple sclerosis. Infection of susceptible SJL/J mice leads to life-long CNS virus persistence and a progressive autoimmune demyelinating disease mediated by myelin-specific T cells activated via epitope spreading. In contrast, virus is rapidly cleared by a robust CTL response in TMEV-IDD-resistant C57BL/6 mice. We investigated whether differential induction of regulatory T cells (Tregs) controls susceptibility to TMEV-IDD. Infection of disease-susceptible SJL/J, but not B6 mice, leads to rapid activation and expansion of Tregs resulting in an unfavorable CNS ratio of Treg:Teffector cells. In addition, anti-CD25-induced inactivation of Tregs in susceptible SJL/J, but not resistant B6, mice results in significantly decreased clinical disease concomitant with enhanced anti-viral CD4(+), CD8(+) and antibody responses resulting in decreased CNS viral titers. This is the first demonstration that virus-induced Treg activation regulates susceptibility to autoimmune disease differentially in susceptible and resistant strains of mice and provides a new mechanistic explanation for the etiology of infection-induced autoimmunity.


Subject(s)
Demyelinating Diseases/immunology , Disease Models, Animal , Multiple Sclerosis/immunology , T-Lymphocytes, Regulatory/immunology , Theilovirus/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cardiovirus Infections/immunology , Cardiovirus Infections/metabolism , Cardiovirus Infections/pathology , Cardiovirus Infections/prevention & control , Cell Proliferation , Central Nervous System/immunology , Central Nervous System/pathology , Central Nervous System/virology , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Demyelinating Diseases/prevention & control , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Glucocorticoid-Induced TNFR-Related Protein , Humans , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/immunology , Severity of Illness Index , T-Lymphocytes, Regulatory/metabolism , Theilovirus/drug effects
15.
Virology ; 402(1): 102-11, 2010 Jun 20.
Article in English | MEDLINE | ID: mdl-20381109

ABSTRACT

Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease (TMEV-IDD) is a relevant mouse model of multiple sclerosis. Infection of susceptible SJL/J mice leads to life-long CNS virus persistence and development of a chronic T cell-mediated autoimmune demyelinating disease triggered via epitope spreading to endogenous myelin epitopes. Potent CNS-infiltrating CD8(+) T cell responses to TMEV epitopes have previously been shown to be induced in both disease-susceptible SJL/J and resistant C57BL/6 mice, in which the virus is rapidly cleared. Specific tolerization of SJL CD8(+) T cells specific for the immunodominant TMEV VP3(159)(-)(166) epitope has no effect on viral load or development of clinical TMEV-IDD, but adoptive transfer of activated CD8(+) VP3(159)(-)(166)-specific T cell blasts shortly after TMEV infection to boost the early anti-viral response leads to clearance of CNS virus and protection from subsequent TMEV-IDD. These studies have important implications for vaccine strategies and treatment of chronic infections in humans.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cardiovirus Infections/veterinary , Demyelinating Diseases/prevention & control , T-Lymphocytes, Cytotoxic/immunology , Theilovirus/immunology , Adoptive Transfer , Animals , Cardiovirus Infections/immunology , Cardiovirus Infections/virology , Epitopes, T-Lymphocyte/immunology , Mice , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL
...