Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Article in English | MEDLINE | ID: mdl-38381311

ABSTRACT

Cellulose/ZnO (CZ) nanocomposites are promising antimicrobial materials known for their antibiotic-free nature, biocompatibility, and environmental friendliness. In this study, cellulose fibers extracted from lotus petioles were utilized as a substrate and decorated with various shapes of ZnO nanoparticles (NPs), including small bean, hexagonal ingot-like, long cylindrical, and hexagonal cylinder-shaped NPs. Increasing zinc salt molar concentration resulted in highly crystalline ZnO NPs forming and enhanced interactions between ZnO NPs and -OH groups of cellulose. The thermal stability and UV-visible absorption properties of the CZ samples were influenced by ZnO concentration. Notably, at a ZnO molar ratio of 0.1, the CZ 0.1 sample demonstrated the lowest weight loss, while the optical band gap gradually decreased from 3.0 to 2.45 eV from the CZ 0.01 to CZ 1.0 samples. The CZ nanocomposites exhibited remarkable antibacterial activity against both Staphylococcus aureus (S. aureus, Gram-positive) and Escherichia coli (E. coli, Gram-negative) bacteria under visible light conditions, with a minimum inhibitory concentration (MIC) of 0.005 mg/mL for both bacterial strains. The bactericidal effects increased with higher concentrations of ZnO NPs, even achieving 100% inhibition. Incorporating ZnO NPs onto cellulose fibers derived from lotus plants presents a promising avenue for developing environmentally friendly materials with broad applications in antibacterial and environmental fields.

2.
J Clin Med ; 11(1)2021 Dec 31.
Article in English | MEDLINE | ID: mdl-35011952

ABSTRACT

BACKGROUND: Bleeding events are among the striking complications following transcatheter aortic valve replacement (TAVR), and bleeding prediction models are crucially warranted. Several studies have highlighted that primary hemostasis disorders secondary to persistent loss of high-molecular-weight (HMW) multimers of the von Willebrand factor (vWF) and assessed by adenosine diphosphate closure time (CT-ADP) may be a strong predictor of late major/life-threatening bleeding complications (MLBCs). Pre-existing atrial fibrillation (AF) is a frequent comorbidity in TAVR patients and potentially associated with increased bleeding events after the procedure. OBJECTIVES: This study evaluated the impact of ongoing primary hemostasis disorders, as assessed by post-procedural CT-ADP > 180 s, on clinical events after TAVR among anticoagulated AF patients. METHODS: An ongoing primary hemostasis disorder was defined by post-procedure CT-ADP > 180 s. Bleeding complications were assessed according to the Valve Academic Research Consortium-2 (VARC-2) criteria. The primary endpoint was the occurrence of late MLBCs at one-year follow-up. The secondary endpoint was a composite of mortality, stroke, myocardial infarction, and rehospitalization for heart failure. RESULTS: In total, 384 TAVR patients were included in the analysis. Of these patients, 57 patients (14.8%) had a prolongated CT-ADP > 180 s. Increased MLBCs were observed in patients with CT-ADP > 180 s (35.1% versus 1.2%; p < 0.0001). Conversely, the occurrence of the composite endpoint did not differ between the groups. Multivariate analysis identified CT-ADP > 180 s (HR 28.93; 95% CI 9.74-85.95; p < 0.0001), bleeding history, paradoxical aortic stenosis (AS), and major vascular complications following TAVR as independent predictors of late MLBCs. CONCLUSION: Among patients with anticoagulated AF, a post-procedural CT-ADP > 180 s was identified as a strong independent predictor of late MLBCs. These findings suggest that persistent primary hemostasis disorders contribute to a higher risk of late bleeding events and should be considered for a tailored, risk-adjusted antithrombotic therapy after TAVR.

3.
Mol Immunol ; 59(2): 172-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24632292

ABSTRACT

Serum amyloid A (SAA) has recently been identified by our group as a mitogen for regulatory T cells (Treg). However, the molecular mechanism by which SAA induces Treg proliferation is unknown. Here we provide evidence that IL-1ß and IL-6 are directly involved in the SAA-mediated proliferation of Treg. By engaging its several cognate receptors, SAA induces IL-1ß and IL-6 secretion by monocytes and drives them toward an HLA-DR(hi) HVEM(lo) phenotype resembling immature dendritic cells, which have been implicated in tolerance generation. This monocyte-derived cytokine milieu is required for Treg expansion, as inhibition of IL-1ß and IL-6 abrogate the ability of SAA to induce Treg proliferation. Furthermore, both IL-1ß and IL-6 are required for ERK1/2 and AKT signaling in proliferating Treg. Collectively, these results point to a novel mechanism, by which SAA initiates a monocyte-dependent process that drives mitogenic signals in Treg.


Subject(s)
Interleukin-1beta/immunology , Interleukin-6/immunology , MAP Kinase Signaling System/immunology , Serum Amyloid A Protein/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Proliferation , Cells, Cultured , Dendritic Cells/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , HLA-DR Antigens/immunology , Humans , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Mitogens/immunology , Monocytes/immunology , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/immunology
4.
Arthritis Rheum ; 65(3): 681-92, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23233229

ABSTRACT

OBJECTIVE: In the K/BxN mouse model of inflammatory arthritis, T cells carrying a transgenic T cell receptor initiate disease by helping B cells to produce arthritogenic anti-glucose-6-phosphate isomerase (anti-GPI) autoantibodies. We found that lethally- irradiated lymphocyte-deficient C57BL/6 (B6).g7 (I-A(g7) +) recombinase-activating gene-deficient (Rag(-/-)) mice reconstituted with K/BxN hematopoietic stem and progenitor cells exhibit arthritis by week 4. In contrast, healthy B6.g7 recipients of K/BxN hematopoietic stem and progenitor cells show only mild arthritis, with limited extent and duration. The objective of this study was to investigate the factors responsible for the attenuation of arthritis in B6.g7 recipients. METHODS: Antibody responses were measured by enzyme-linked immunosorbent assay. Fluorescence-activated cell sorting analyses were performed for testing chimerism, expression of markers of activation and suppression, tetramer binding, and intracellular cytokines in CD4+ T cells. Suppressive activity of CD4+ T cells was studied by adoptive transfer. RESULTS: Titers of anti-GPI antibodies in reconstituted B6.g7 mice were ∼60-fold lower than in reconstituted B6.g7 Rag(-/-) mice. Examination of chimerism in the reconstituted B6.g7 mice showed that B cells and myeloid cells in these mice were donor derived, but CD4+ T cells were primarily host derived and enriched for cells expressing the conventional regulatory markers CD25 and FoxP3. Notably, CD4+CD25-FoxP3- T cells expressed markers of suppressive function (CD73 and folate receptor 4), and delayed disease after adoptive transfer. Activation of donor-derived CD4+ T cells was reduced, and thymic deletion of these cells appeared increased. CONCLUSION: Despite myeloablation, host CD4+ T cells having a regulatory phenotype emerge in these mice and attenuate autoimmunity.


Subject(s)
Arthritis/immunology , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cells/immunology , 5'-Nucleotidase/metabolism , Adoptive Transfer , Animals , Arthritis/etiology , Arthritis/pathology , Autoimmune Diseases/etiology , Autoimmune Diseases/pathology , CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Glucose-6-Phosphate Isomerase/genetics , Glucose-6-Phosphate Isomerase/immunology , Hematopoietic Stem Cells/pathology , Homeodomain Proteins/genetics , Immunologic Memory/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Receptors, Cell Surface/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Thymus Gland/cytology , Thymus Gland/immunology , Whole-Body Irradiation
5.
Blood ; 117(14): 3793-8, 2011 Apr 07.
Article in English | MEDLINE | ID: mdl-21325601

ABSTRACT

The acute phase protein serum amyloid A (SAA) has been well characterized as an indicator of inflammation. Nevertheless, its functions in pro versus anti-inflammatory processes remain obscure. Here we provide unexpected evidences that SAA induces the proliferation of the tolerogenic subset of regulatory T cells (T(reg)). Intriguingly, SAA reverses T(reg) anergy via its interaction with monocytes to activate distinct mitogenic pathways in T(reg) but not effector T cells. This selective responsiveness of T(reg) correlates with their diminished expression of SOCS3 and is antagonized by T(reg)-specific induction of this regulator of cytokine signaling. Collectively, these evidences suggest a novel anti-inflammatory role of SAA in the induction of a micro-environment that supports T(reg) expansion at sites of infection or tissue injury, likely to curb (auto)-inflammatory responses.


Subject(s)
Clonal Anergy/drug effects , Monocytes/physiology , Serum Amyloid A Protein/pharmacology , Suppressor of Cytokine Signaling Proteins/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , Animals , Cell Communication/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Child , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Serum Amyloid A Protein/physiology , Signal Transduction/drug effects , Signal Transduction/immunology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Suppressor of Cytokine Signaling Proteins/physiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/physiology
6.
Int Immunol ; 22(8): 705-16, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20547545

ABSTRACT

Several MHC class II alleles linked with autoimmune diseases form unusually low-stability complexes with class II-associated invariant chain peptides (CLIP), leading us to hypothesize that this is an important feature contributing to autoimmune pathogenesis. We recently demonstrated a novel post-endoplasmic reticulum (ER) chaperoning role of the CLIP peptides for the murine class II allele I-E(d). In the current study, we tested the generality of this CLIP chaperone function using a series of invariant chain (Ii) mutants designed to have varying CLIP affinity for I-A(g7). In cells expressing these Ii CLIP mutants, I-A(g7) abundance, turnover and antigen presentation are all subject to regulation by CLIP affinity, similar to I-E(d). However, I-A(g7) undergoes much greater quantitative changes than observed for I-E(d). In addition, we find that Ii with a CLIP region optimized for I-A(g7) binding may be preferentially assembled with I-A(g7) even in the presence of higher levels of wild-type Ii. This finding indicates that, although other regions of Ii interact with class II, CLIP binding to the groove is likely to be a dominant event in assembly of nascent class II molecules with Ii in the ER.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/immunology , Histocompatibility Antigens Class II/immunology , Molecular Chaperones/immunology , Protein Processing, Post-Translational/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Surface/genetics , Antigens, Surface/immunology , B-Lymphocytes/immunology , Cell Line, Tumor , Flow Cytometry , Histocompatibility Antigens Class II/genetics , Immunoblotting , Mice , Mice, Inbred BALB C , Mutation
7.
Immunology ; 131(1): 18-32, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20408893

ABSTRACT

DM catalyses class II-associated invariant chain peptide (CLIP) release, edits the repertoire of peptides bound to major histocompatibility complex (MHC) class II molecules, affects class II structure, and thereby modulates binding of conformation-sensitive anti-class II antibodies. Here, we investigate the ability of DM to enhance the cell surface binding of monomorphic antibodies. We show that this enhancement reflects increases in cell surface class II expression and total cellular abundance, but notably these effects are selective for particular alleles. Evidence from analysis of cellular class II levels after cycloheximide treatment and from pulse-chase experiments indicates that DM increases the half-life of affected alleles. Unexpectedly, the pulse-chase experiments also revealed an early effect of DM on assembly of these alleles. The allelically variant feature that correlates with susceptibility to these DM effects is low affinity for CLIP; DM-dependent changes in abundance are reduced by invariant chain (CLIP) mutants that enhance CLIP binding to class II. We found evidence that DM mediates rescue of peptide-receptive DR0404 molecules from inactive forms in vitro and evidence suggesting that a similar process occurs in cells. Thus, multiple mechanisms, operating along the biosynthetic pathway of class II molecules, contribute to DM-mediated increases in the abundance of low-CLIP-affinity alleles.


Subject(s)
Alleles , Antigens, Differentiation, B-Lymphocyte/metabolism , HLA-D Antigens/metabolism , Histocompatibility Antigens Class II/metabolism , Peptides/metabolism , Animals , Antibodies/metabolism , Antigen Presentation , Antigens, Differentiation, B-Lymphocyte/genetics , B-Lymphocytes/metabolism , Cell Line , Half-Life , Histocompatibility Antigens Class II/genetics , Humans , Mice , Peptides/genetics , Protein Binding , Protein Conformation , Transfection
8.
Immunology ; 130(3): 436-46, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20331476

ABSTRACT

SUMMARY: The expression of major histocompatibility complex class II (MHC II) molecules is post-translationally regulated by endocytic protein turnover. Here, we identified the serine protease cathepsin G (CatG) as an MHC II-degrading protease by in vitro screening and examined its role in MHC II turnover in vivo. CatG, uniquely among endocytic proteases tested, initiated cleavage of detergent-solubilized native and recombinant soluble MHC II molecules. CatG cleaved human leukocyte antigen (HLA)-DR isolated from both HLA-DM-expressing and DM-null cells. Even following CatG cleavage, peptide binding was retained by pre-loaded, soluble recombinant HLA-DR. MHC II cleavage occurred on the loop between fx1 and fx2 of the membrane-proximal beta2 domain. All allelic variants of HLA-DR tested and murine I-A(g7) class II molecules were susceptible, whereas murine I-E(k) and HLA-DM were not, consistent with their altered sequence at the P1' position of the CatG cleavage site. CatG effects were reduced on HLA-DR molecules with DRB mutations in the region implicated in interaction with HLA-DM. In contrast, addition of CatG to intact B-lymphoblastoid cell lines (B-LCLs) did not cause degradation of membrane-bound MHC II. Moreover, inhibition or genetic ablation of CatG in primary antigen-presenting cells did not cause accumulation of MHC II molecules. Thus, in vivo, the CatG cleavage site is sterically inaccessible or masked by associated molecules. A combination of intrinsic and context-dependent proteolytic resistance may allow peptide capture by MHC II molecules in harshly proteolytic endocytic compartments, as well as persistent antigen presentation in acute inflammatory settings with extracellular proteolysis.


Subject(s)
Cathepsin G/chemistry , Cathepsin G/metabolism , Histocompatibility Antigens Class II/metabolism , Protein Processing, Post-Translational , Amino Acid Sequence , Amino Acid Substitution , Animals , B-Lymphocytes/metabolism , Cathepsin G/antagonists & inhibitors , Cathepsin G/genetics , Cathepsins/metabolism , Cell Line , Dendritic Cells/metabolism , HLA-D Antigens/genetics , HLA-D Antigens/metabolism , HLA-DR Antigens/genetics , HLA-DR Antigens/metabolism , HLA-DR1 Antigen/genetics , HLA-DR1 Antigen/metabolism , HLA-DR3 Antigen/genetics , HLA-DR3 Antigen/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/pharmacology , Humans , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Molecular Sequence Data , Peptide Fragments/analysis , Peptide Fragments/metabolism , Peptides/metabolism , Polymorphism, Genetic/genetics , Protein Binding/physiology , Recombinant Proteins/metabolism , Sequence Alignment
9.
J Immunol ; 183(6): 3895-905, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19717517

ABSTRACT

Persistence of even the stealthiest viruses can perturb immune function either to the benefit or detriment of the host. Lymphocytic choriomeningitis virus (LCMV) establishes lifelong, systemic persistence when introduced in utero or at birth. Despite a highly evolved host-pathogen relationship, LCMV cannot escape detection by the innate immune system, which results in chronic stimulation of the type 1 IFN pathway in adult carrier mice. In this study we demonstrate that IFN-beta is chronically up-regulated in peripheral lymphoid and nonlymphoid tissues (but not the CNS) of mice persistently infected from birth with LCMV and that dendritic cells (DCs) represent at least one source of IFN-beta. Interestingly, chronic stimulation of this innate pathway significantly elevated MHC class I expression in the CNS as well as the periphery. Elevated MHC I expression was dependent on IFN-alphabeta receptor but not MyD88-dependent signaling, as only genetic deletion of the former reduced MHC I to normal levels. An increase in circulating virus was also observed in the IFN-alphabeta receptor deficient carrier mice, signifying that type I IFN continually exerts anti-viral pressure during a LCMV carrier state. Finally, to determine whether heightened CNS MHC I could be therapeutically corrected, we purged LCMV carrier mice of their persistent infection using adoptive immunotherapy. This treatment significantly reduced CNS MHC I expression. Collectively, these data demonstrate that even a well adapted pathogen can chronically stimulate the innate immune system and consequently alter the expression of Ag presenting machinery in an immunologically specialized compartment like the CNS.


Subject(s)
Central Nervous System/immunology , Histocompatibility Antigens Class I/genetics , Interferon-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Virus Diseases/immunology , Animals , Chronic Disease , Immunotherapy , Lymphocytic choriomeningitis virus/immunology , Mice , Receptor, Interferon alpha-beta/immunology , Up-Regulation , Virus Diseases/therapy
10.
J Virol ; 83(17): 8905-15, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19553326

ABSTRACT

Persistent viral infections are a major health concern worldwide. During persistent infection, overwhelming viral replication and the rapid loss of antiviral T-cell function can prevent immune-mediated clearance of the infection, and therapies to reanimate the immune response and purge persistent viruses have been largely unsuccessful. Adoptive immunotherapy using memory T cells is a highly successful therapeutic approach to eradicate a persistent viral infection. Understanding precisely how therapeutically administered memory T cells achieve clearance should improve our ability to terminate states of viral persistence in humans. Mice persistently infected from birth with lymphocytic choriomeningitis virus are tolerant to the pathogen at the T-cell level and thus provide an excellent model to evaluate immunotherapeutic regimens. Previously, we demonstrated that adoptively transferred memory T cells require recipient dendritic cells to effectively purge an established persistent viral infection. However, the mechanisms that reactivate and sustain memory T-cell responses during clearance of such an infection remain unclear. Here we establish that therapeutic memory T cells require CD80 and CD86 costimulatory signals to efficiently clear an established persistent viral infection in vivo. Early blockade of costimulatory pathways with CTLA-4-Fc decreased the secondary expansion of virus-specific CD8(+) and CD4(+) memory T cells as well as their ability to produce antiviral cytokines and purge the persistent infection. Late costimulation blockade also reduced virus-specific T-cell numbers, illustrating that sustained interactions with costimulatory molecules is required for efficient T-cell expansion. These findings indicate that antiviral memory T cells require costimulation to efficiently clear a persistent viral infection and that costimulatory pathways can be targeted to modulate the magnitude of an adoptive immunotherapeutic regimen.


Subject(s)
Adoptive Transfer , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Lymphocytic Choriomeningitis/therapy , Lymphocytic choriomeningitis virus/immunology , Animals , B7-1 Antigen/immunology , B7-2 Antigen/immunology , Lymphocyte Activation/immunology , Mice
11.
J Immunol ; 181(2): 1161-9, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18606669

ABSTRACT

Restrictions in the diversity of an adaptive immune repertoire can facilitate viral persistence. Because a host afflicted with an immune deficiency is not likely to purge a persistent infection using endogenous mechanisms, it is important to explore adoptive therapies to supplement the host with a functional immune defense. In this study, we describe a virus carrier state that results from introducing lymphocytic choriomeningitis virus (LCMV) into adult mice possessing a restricted T cell repertoire. On infection of these mice, LCMV establishes systemic persistence, and within the CNS the virus infects astrocytes (and later oligodendrocytes) rather than its traditional parenchymal target neurons. To determine whether LCMV could be purged from a novel target selection in the absence of an endogenous immune repertoire, we adoptively transferred virus-specific memory cells into adult carrier mice. The memory cells purged virus from the periphery as well as the CNS, but they induced fatalities not typically associated with adoptive immunotherapy. When the repertoire of the recipient mice was examined, a deficiency in natural regulatory T cells was noted. We therefore supplemented carrier mice with regulatory T cells and simultaneously performed adoptive immunotherapy. Cotransfer of regulatory T cells significantly reduced mortality while still permitting the antiviral memory cells to purge the persistent infection. These data indicate that regulatory T cells can be used therapeutically to lessen the pathogenicity of virus-specific immune cells in an immunodeficient host. We also propose that the novel carrier state described herein will facilitate the study of immunotherapeutic regimens.


Subject(s)
Central Nervous System/immunology , Immunologic Memory , Immunotherapy, Adoptive , Lymphocytic Choriomeningitis/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes/immunology , Animals , Astrocytes/cytology , Astrocytes/immunology , Astrocytes/virology , Carrier State , Cell Line , Central Nervous System/cytology , Central Nervous System/virology , Lymphocytic Choriomeningitis/therapy , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/physiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Oligodendroglia/cytology , Oligodendroglia/immunology , Oligodendroglia/virology , T-Lymphocytes, Regulatory/metabolism , Virus Replication
12.
Virol J ; 4: 53, 2007 Jun 06.
Article in English | MEDLINE | ID: mdl-17553158

ABSTRACT

Once a virus infection establishes persistence in the central nervous system (CNS), it is especially difficult to eliminate from this specialized compartment. Therefore, it is of the utmost importance to fully understand scenarios during which a persisting virus is ultimately purged from the CNS by the adaptive immune system. Such a scenario can be found following infection of adult mice with an immunosuppressive variant of lymphocytic choriomeningitis virus (LCMV) referred to as clone 13. In this study we demonstrate that following intravenous inoculation, clone 13 rapidly infected peripheral tissues within one week, but more slowly innundated the entire brain parenchyma over the course of a month. During the establishment of persistence, we observed that genetically tagged LCMV-specific cytotoxic T lymphocytes (CTL) progressively lost function; however, the severity of this loss in the CNS was never as substantial as that observed in the periphery. One of the most impressive features of this model system is that the peripheral T cell response eventually regains functionality at ~60-80 days post-infection, and this was associated with a rapid decline in virus from the periphery. Coincident with this "reanimation phase" was a massive influx of CD4 T and B cells into the CNS and a dramatic reduction in viral distribution. In fact, olfactory bulb neurons served as the last refuge for the persisting virus, which was ultimately purged from the CNS within 200 days post-infection. These data indicate that a functionally revived immune response can prevail over a virus that establishes widespread presence both in the periphery and brain parenchyma, and that therapeutic enhancement of an existing response could serve as an effective means to thwart long term CNS persistence.


Subject(s)
Arenaviridae Infections/immunology , Arenaviridae Infections/virology , Central Nervous System/immunology , Central Nervous System/virology , Lymphocytic choriomeningitis virus/immunology , Animals , B-Lymphocytes/immunology , Blood/immunology , Blood/virology , Brain/immunology , Brain/pathology , Brain/virology , CD4-Positive T-Lymphocytes/immunology , Central Nervous System/pathology , Disease Models, Animal , Immune Tolerance , Mice , Mice, Inbred C57BL , Neurons/virology , Olfactory Bulb/virology , Olfactory Nerve/virology , Spleen/virology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , Time Factors , Viremia
13.
J Exp Med ; 203(8): 1963-75, 2006 Aug 07.
Article in English | MEDLINE | ID: mdl-16847068

ABSTRACT

Given the global impact of persistent infections on the human population, it is of the utmost importance to devise strategies to noncytopathically purge tissues of infectious agents. The central nervous system (CNS) poses a unique challenge when considering such strategies, as it is an immunologically specialized compartment that contains a nonreplicative cell population. Administration of exogenously derived pathogen-specific memory T cells (referred to as adoptive immunotherapy) to mice burdened with a persistent lymphocytic choriomeningitis virus (LCMV) infection from birth results in eradication of the pathogen from all tissues, including the CNS. In this study, we sought mechanistic insights into this highly successful therapeutic approach. By monitoring the migration of traceable LCMV-specific memory CD8+ T cells after immunotherapy, it was revealed that cytotoxic T lymphocytes (CTLs) distributed widely throughout the CNS compartment early after immunotherapy, which resulted in a dramatic elevation in the activity of CNS antigen-presenting cells (APCs). Immunotherapy induced microglia activation as well as the recruitment of macrophages and dendritic cells (DCs) into the brain parenchyma. However, DCs emerged as the only CNS APC population capable of inducing memory CTLs to preferentially produce the antiviral cytokine tumor necrosis factor-alpha, a cytokine demonstrated to be required for successful immunotherapeutic clearance. DCs were also found to be an essential element of the immunotherapeutic process because in their absence, memory T cells failed to undergo secondary expansion, and viral clearance was not attained in the CNS. These experiments underscore the importance of DCs in the immunotherapeutic clearance of a persistent viral infection and suggest that strategies to elevate the activation/migration of DCs (especially within the CNS) may facilitate pathogen clearance.


Subject(s)
Antigen Presentation/immunology , Brain/cytology , Dendritic Cells/cytology , Dendritic Cells/immunology , Immunotherapy, Adoptive , Lymphocytic Choriomeningitis/immunology , Animals , Animals, Newborn , Brain/pathology , Brain/virology , Carrier State , Cell Movement , Dendritic Cells/virology , Histocompatibility Antigens Class II/metabolism , Humans , Immunologic Memory/immunology , Kinetics , Lymphocytic choriomeningitis virus/physiology , Mice , Mice, Inbred C57BL , Spleen/cytology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/metabolism
14.
J Immunol ; 173(8): 4779-90, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15470017

ABSTRACT

Although both self- and pathogen-specific T cells can participate in tissue destruction, recent studies have proposed that after viral infection, bystander T cells of an irrelevant specificity can bypass peptide-MHC restriction and contribute to undesired immunopathological consequences. To evaluate the importance of this mechanism of immunopathogenesis, we determined the relative contributions of Ag-specific and bystander CD8+ T cells to the development of CNS disease. Using lymphocytic choriomeningitis virus (LCMV) as a stimulus for T cell recruitment into the CNS, we demonstrate that bystander CD8+ T cells with an activated surface phenotype can indeed be recruited into the CNS over a chronic time window. These cells become anatomically positioned in the CNS parenchyma, and a fraction aberrantly acquires the capacity to produce the effector cytokine, IFN-gamma. However, when directly compared with their virus-specific counterparts, the contribution of bystander T cells to CNS damage was insignificant in nature (even when specifically activated). Although bystander T cells alone failed to cause tissue injury, transferring as few as 1000 naive LCMV-specific CD8+ T cells into a restricted repertoire containing only bystander T cells was sufficient to induce immune-mediated pathology and reconstitute a fatal CNS disease. These studies underscore the importance of specific T cells in the development of immunopathology and subsequent disease. Because of highly restrictive constraints imposed by the host, it is more likely that specific, rather than nonspecific, bystander T cells are the active participants in T cell-mediated diseases that afflict humans.


Subject(s)
Brain/immunology , H-2 Antigens/immunology , Lymphocytic Choriomeningitis/immunology , Peptide Fragments/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Apoptosis , Brain/pathology , Cell Movement , Cytokines/biosynthesis , Histocompatibility Antigen H-2D , Hyaluronan Receptors/analysis , Lymphocytic Choriomeningitis/etiology , Lymphocytic Choriomeningitis/pathology , Mice , Mice, Inbred C57BL , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...