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










Publication year range
1.
Autoimmun Rev ; 13(12): 1174-81, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25151974

ABSTRACT

T helper 17 (Th17), a distinct subset of CD4(+) T cells with IL-17 as their major cytokine, orchestrate the pathogenesis of inflammatory and autoimmune diseases. Dysregulated Th17 cells contribute to inflammatory and autoimmune diseases. Candidate biologics are in development for targeting IL-17, IL-17 receptors or IL-17 pathways. Several drugs that impact the IL-17 pathway are already in clinical trials for the treatment of autoimmune diseases. In this review we provide evidence for the role of Th17 cells in immune-mediated diseases. An understanding of the role of Th17 in these conditions will provide important insights and unravel novel targets for therapeutic intervention.


Subject(s)
Autoimmunity , Th17 Cells/immunology , Animals , Autoimmune Diseases/immunology , Gonadal Steroid Hormones/immunology , Humans , Inflammation/immunology , Interleukin-17/immunology , Mesenchymal Stem Cells/immunology
2.
Autoimmun Rev ; 12(12): 1160-5, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23860189

ABSTRACT

miRNAs are small non-coding RNA molecules that modulate the expression of multiple protein-encoding genes at the post-transcriptional level. They have recently been recognized as powerful regulators of numerous genes and pathways in the pathogenesis of inflammatory and autoimmune diseases. The targets of most miRNAs remain unknown and their roles in biological processes such as cell differentiation, proliferation, and death (apoptosis) are not clearly understood. In this review we will discuss how certain candidate miRNAs affect inflammatory and immune mediated diseases by regulating their cellular and molecular targets. We focused the influence of gender and sex hormones on miRNA. We believe that understanding the role of miRNAs could shed light on the cause and progression of many inflammatory and autoimmune diseases and eventually lay the groundwork for therapeutic options.


Subject(s)
Autoimmune Diseases/genetics , Gene Expression Regulation , Inflammation/genetics , MicroRNAs/metabolism , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Autoimmunity , Humans , Inflammation/immunology , MicroRNAs/genetics
3.
Autoimmun Rev ; 9(8): 583-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20433954

ABSTRACT

Programmed death 1 (PD-1) and its ligands (PD-L1 and PD-L2) are responsible for inhibitory T cell signaling that helps mediate the mechanisms of tolerance and immune homeostasis. The PD-1:PD-L signaling pathway has been shown to play an important role in a variety of diseases, including autoimmune conditions, chronic infection, and cancer. Recently, investigators have explored the role of sex hormones in modulating the pathway in autoimmune conditions. Exploring the effects of sex hormones on the PD-1:PD-L pathway could shed light on the gender biased nature of many autoimmune conditions as well as aide in the development of therapeutics targeting the immune system.


Subject(s)
Antigens, CD/immunology , Apoptosis Regulatory Proteins/immunology , Autoimmunity/immunology , Gonadal Steroid Hormones/immunology , Sex Characteristics , Animals , B7-H1 Antigen , Female , Humans , Immune Tolerance/immunology , Intercellular Signaling Peptides and Proteins/immunology , Male , Programmed Cell Death 1 Ligand 2 Protein , Programmed Cell Death 1 Receptor , Signal Transduction/immunology , T-Lymphocytes/immunology
4.
Autoimmun Rev ; 9(8): 560-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20385256

ABSTRACT

While CD4(+)CD25(high) regulatory T cells (Tregs) have garnered much attention for their role in the maintenance of immune homeostasis, recent findings have shown that subsets of CD8(+) T cells (CD8(+) Tregs) display immunoregulatory functions as well. Both CD4(+) Tregs and CD8(+) Tregs appear impaired in number and/or function in several autoimmune diseases and in experimental animal models of autoimmunity, suggesting the possibility of immunotherapeutic targeting of these cells for improved management of autoimmune conditions. Our group has developed a strategy to induce CD8(+) Tregs in autoimmune mice through the use of a tolerogenic self-peptide, and new information has been gained on the phenotype, function and role of induced CD8(+) Tregs in autoimmunity. Here we present an overview of the role and mechanisms of action of CD8(+) Tregs in autoimmunity, with a special focus on lupus. We also discuss the potential role of CD8(+) Tregs in other diseases, including chronic infection and cancer.


Subject(s)
Autoimmunity/immunology , CD8-Positive T-Lymphocytes/immunology , Lupus Erythematosus, Systemic/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Humans , Mice
5.
Inflamm Allergy Drug Targets ; 7(4): 253-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19075790

ABSTRACT

A central pathologic mechanism in systemic autoimmune diseases with chronic inflammation such as systemic lupus erythematosus (SLE) is the aberrant production of antibodies against self-components produced by abnormal B cells with the help of hyperactive CD4(+)T cells. One goal for better control of the disease is the limitation of the number of abnormal and hyperactive cells, to prevent and/or attenuate the damaging effects of the pathogenic antibodies on target tissues. Recently, a role of regulatory T cells in the suppression of autoimmune reactivity in diseases including SLE has been recognized. CD4(+)CD25(+) regulatory T cells (Tregs) and CD8(+) inhibitory T (Ti) cells have been found numerically decreased and/or functionally impaired in some patients with active systemic lupus erythematosus. Recent experimental work and preclinical studies have also provided proof-of-concept for the possibility of induction of self-tolerance through the modulation of regulatory/suppressor T cells using self antigen-derived peptides that could promote suppression of the production of pathogenic antibodies. This review explores the mechanisms elicited by the administration of self antigen-derived peptides on the induction of suppression of autoimmune responses, and how this information might lead to future development of new strategies for better management of systemic autoimmune conditions.


Subject(s)
Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Peptides/immunology , Animals , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Humans , Immune Tolerance/drug effects , Lupus Erythematosus, Systemic/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/physiology , Transforming Growth Factor beta/physiology
6.
Hum Immunol ; 69(11): 790-6, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18817829

ABSTRACT

Multiple CD8(+) suppressive T cell (Ts) subtypes are now recognized as essential regulators of the immune system that prevent autoimmunity through secretion of multiple cytokines and the subsequent inhibition of effector lymphocyte function. CD8(+) Ts are an exciting area of study because of the possible therapeutic implications of inducing suppressive cells that are able to subdue or anergize autoimmune manifestations. Current research in systemic lupus erythematosus (SLE), a disease in which most effective therapies are widely immunosuppressive, is often focused on novel and highly targeted ways in which to treat this multiorgan disease. CD8(+) Ts have been impaired in human and murine SLE. Our group and others have utilized tolerogenic peptides to induce and study CD8(+) Ts to understand their function, as well as investigate a possible new SLE therapy. This review will discuss the similarities and differences in CD8(+) Ts subsets, the concept of tolerance as a therapy, and the current understanding of CD8(+) Ts in mouse SLE models.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immune Tolerance , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , Animals , Disease Models, Animal , Humans , Immune Tolerance/drug effects , Immunosuppressive Agents/immunology , Immunosuppressive Agents/pharmacology , Lupus Erythematosus, Systemic/therapy , Lymphocyte Activation/drug effects , Mice , Peptides/immunology , Peptides/pharmacology , Species Specificity
7.
J Immunol ; 180(4): 2069-80, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18250412

ABSTRACT

Systemic lupus erythematosus is an autoimmune disease caused primarily by autoantibodies (including IgG anti-DNA) and immune complexes that cause tissue damage. After tolerization with an artificial peptide (pConsensus, pCons) based on murine anti-DNA IgG sequences containing MHC class I and class II T cell determinants, lupus-prone (NZB x NZW)F(1) female (BWF(1)) mice develop regulatory CD4+CD25+ T cells and inhibitory CD8+ T cells, both of which suppress anti-DNA Ig production and immune glomerulonephritis. In the present work, we show that splenocytes from BWF(1) mice treated with pCons had significant expansion of primarily CD8+ T cells. CD4+ T cells and B cells were each directly suppressed by CD8+ T cells from tolerized mice in a contact-independent manner. Both pCons-induced CD8+CD28+ and CD8+CD28- T cells suppressed production of anti-DNA in vitro. Silencing with small interfering RNA of Foxp3 abrogated the suppression mediated by both CD8+ T cell subsets. Additionally, CD8+ T cells from tolerized mice were weakly cytotoxic against syngeneic B cells from old anti-DNA-producing mice, but not from young mice. Importantly, pCons treatment had dual effects on CD8+ suppressor T cells from tolerized mice, increasing the intracellular expression of Foxp3 while decreasing the surface expression of PD1 molecules. Blocking PD1/PDL1 interactions in the CD8+ T cells from tolerized mice reduced their expression of Foxp3 and their ability to suppress CD4+CD25- proliferation. In contrast, blocking PD1/PDL1 in naive T cells increased Foxp3 expression. Our data suggest that tolerization with pCons activates different subsets of inhibitory/cytotoxic CD8+ T cells whose targets are both CD4+CD25- effector T cells and B cells.


Subject(s)
Antigens, Surface/biosynthesis , Apoptosis Regulatory Proteins/biosynthesis , CD8-Positive T-Lymphocytes/immunology , Consensus Sequence , Forkhead Transcription Factors/biosynthesis , Immune Tolerance , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation/immunology , Peptides/administration & dosage , Amino Acid Sequence , Animals , Antibodies, Blocking/pharmacology , Antigens, Surface/genetics , Antigens, Surface/immunology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation , Crosses, Genetic , Cytotoxicity, Immunologic/genetics , Female , Forkhead Transcription Factors/genetics , Genetic Predisposition to Disease , Immune Tolerance/genetics , Immunoglobulin G/biosynthesis , Immunophenotyping , Lupus Erythematosus, Systemic/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred NZB , Molecular Sequence Data , Peptides/chemical synthesis , Programmed Cell Death 1 Receptor , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Up-Regulation/genetics , Up-Regulation/immunology
8.
J Immunol ; 178(12): 7649-57, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17548601

ABSTRACT

Systemic lupus erythematosus is an autoimmune disease caused by autoantibodies, including IgG anti-DNA. New Zealand Black/New Zealand White F(1) female mice, a model of spontaneous polygenic systemic lupus erythematosus, tolerized with an artificial peptide (pConsensus) based on anti-DNA IgG sequences containing MHC class I and class II T cell determinants, develop regulatory CD4+CD25+ T cells and CD8+ inhibitory T cells (CD8+ Ti), both of which suppress autoantibody production. CD8+ Ti inhibit primarily via secretion of TGF-beta. In the present study, we show that the inhibitory function of CD8+ T cells from tolerized mice is sustained for up to 8 wk and at all times depends on expression of Foxp3. Both CD28-positive and CD28-negative CD8+ T cells contain inhibitory cells, but the expression of mRNA for Foxp3 and for TGF-beta is higher and lasts longer in the CD28- subset. In vitro addition of TGF-beta (in the presence of IL-2) induces Foxp3 expression in a dose-response manner. Gene inhibition or blockade with small interfering RNA of Foxp3 abrogates the ability of the CD8+ Ti to inhibit anti-DNA production and the proliferation of CD4+ Th cells. Moreover, a significant correlation between expression of Foxp3 and ability of CD8+ Ti to secrete TGF-beta is observed. Therefore, CD8+ Ti in this system of tolerance are similar to CD4+CD25+ regulatory T cells in their dependence on expression of Foxp3, and there may be a bidirectional Foxp3/TGF-beta autocrine loop that determines the ability of the CD8+ T cells to control autoimmunity.


Subject(s)
Autoimmunity , CD8-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/metabolism , Immune Tolerance , Lupus Erythematosus, Systemic/immunology , Animals , Antibodies, Antinuclear/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Disease Models, Animal , Female , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/genetics , Immunoglobulin G/biosynthesis , Mice , Mice, Inbred Strains , Peptides/immunology , RNA, Messenger/analysis , RNA, Messenger/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
9.
J Immunol ; 175(11): 7728-37, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16301683

ABSTRACT

Lupus-prone (NZB x NZW)F1 mice spontaneously develop elevated titers of anti-DNA Abs that contain T cell determinants in their V(H) regions. We have previously shown that tolerization with an artificial peptide based on these T cell determinants (pConsensus (pCons)) can block production of anti-DNA Abs and prolong survival of the mice. In this study, we show that this protection depends in part on the generation of peripheral TGFbeta- and Foxp3-expressing inhibitory CD8+ (Ti) cells. These CD8+ Ti cells suppress anti-DNA IgG production both in vitro and in vivo and require up-regulated expression of both Foxp3 and TGFbeta to exert their suppressive function, as indicated by microarray analyses, small interfering RNA inhibition studies, and blocking experiments. Additionally, CD8+ Ti cells from pCons-tolerized mice were longer-lived suppressors that up-regulated expression of Bcl-2 and were more resistant to apoptosis than similar cells from naive mice. These data indicate that clinical suppression of autoimmunity after administration of pCons depends in part on the generation of CD8+ Ti cells that suppress secretion of anti-DNA Ig using mechanisms that include Foxp3, TGFbeta, and resistance to apoptosis.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/biosynthesis , Immune Tolerance/immunology , Lupus Erythematosus, Systemic/prevention & control , Oligopeptides/therapeutic use , Transforming Growth Factor beta/biosynthesis , Adoptive Transfer , Animals , Antibodies, Antinuclear/biosynthesis , Antibodies, Antinuclear/immunology , Apoptosis/physiology , CD8-Positive T-Lymphocytes/drug effects , Female , Flow Cytometry , Forkhead Transcription Factors/immunology , Immunoglobulin Variable Region/immunology , Mice , Mice, Inbred NZB , Microarray Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/immunology
10.
Am J Geriatr Psychiatry ; 12(2): 158-65, 2004.
Article in English | MEDLINE | ID: mdl-15010345

ABSTRACT

OBJECTIVE: Complex trait mapping has been widely used to analyze the genetics of behavior. However, the approach has some disadvantages, including poor gene localization and low replicability. Genome-tagged mice (GTMs) are sets of congenic mouse strains that span the entire mouse genome and are a promising reagent for localization of genes contributing to behavior. METHODS: In order to map behavioral loci of interest, a GTM was investigated in which the middle region of Chromosome 1 from DBA/2J was introgressed onto a C57BL/6J background. The GTM was analyzed for behaviors related to sensorimotor gating, anxiety, depression, pain sensitivity, and learning and memory. RESULTS: The GTM was found to harbor a locus contributing to learning and memory, replicating results from complex trait analysis. CONCLUSIONS: The GTMs should be a valuable resource for mapping and confirmation of loci contributing to complex behavioral traits in the mouse, with ultimate implications for human genomic-based research, as well.


Subject(s)
Behavior, Animal/physiology , Chromosome Mapping/methods , Animals , Anxiety/genetics , Chromosomes, Human, Pair 1/genetics , Depression/genetics , Fear , Humans , Learning/physiology , Locomotion/physiology , Male , Memory/physiology , Mice , Mice, Congenic , Pain/genetics
11.
J Neurosci Res ; 74(4): 562-9, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14598300

ABSTRACT

Identification of behavioral loci through complex trait mapping remains a widely employed approach but suffers from poor gene localization and low replicability. Genome-tagged mice (GTMs) are overlapping sets of congenic strains spanning the whole genome and offer the possibilities of superior mapping power and reproducibility. In this study, three GTM strains each consisting of an average approximately 27 cM DBA/2J genomic intervals introgressed onto a C57BL/6J background were employed for localization of behavioral traits. These GTMs were chosen because the corresponding chromosomal regions had been previously identified as containing loci for learning and memory. Analysis of the GTMs allowed confirmation of the learning and memory loci, and one on chromosome 3 was in addition fine mapped to an 8.8-cM region of overlap between two of the GTMs. Moreover, loci for prepulse inhibition of the startle response, acoustic startle response, and spontaneous locomotor activity were also mapped. These results suggest that the GTMs should be a valuable resource for mapping and confirmation of loci contributing to complex behavioral traits in the mouse.


Subject(s)
Behavior, Animal , Genetics, Behavioral , Genome , Mice, Congenic/genetics , Animals , Chromosome Mapping , Chromosomes/genetics , Learning , Male , Memory , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred Strains , Quantitative Trait, Heritable , Reflex, Startle
12.
J Immunol ; 168(11): 5415-23, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12023334

ABSTRACT

The development of chronic rejection is the major limitation to long-term allograft survival. HLA class I Ags have been implicated to play a role in this process because ligation of class I molecules by anti-HLA Abs stimulates smooth muscle cell and endothelial cell proliferation. In this study, we show that ligation of HLA class I molecules on the surface of human aortic endothelial cells stimulates phosphorylation of Src, focal adhesion kinase, and paxillin. Signaling through class I stimulated Src phosphorylation and mediated fibroblast growth factor receptor (FGFR) translocation to the nucleus. In contrast, Src kinase activity was not involved in class I-mediated transfer of FGFR from cytoplasmic stores to the cell surface. Inhibition of Src protein kinase activity blocked HLA class I-stimulated tyrosine phosphorylation of paxillin and focal adhesion kinase. Furthermore, HLA class I-mediated phosphorylation of the focal adhesion proteins and FGFR expression was inhibited by cytochalasin D and latrunculin A, suggesting a role for the actin cytoskeleton in the signaling process. These findings indicate that anti-HLA Abs have the capacity to transduce activation signals in endothelial cells that may promote the development of chronic rejection.


Subject(s)
Actins/physiology , Cytoskeletal Proteins/metabolism , Endothelium, Vascular/metabolism , Histocompatibility Antigens Class I/physiology , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , src-Family Kinases/metabolism , Biological Transport , Endothelium, Vascular/cytology , Fibroblast Growth Factor 2/pharmacology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Mitogen-Activated Protein Kinases/metabolism , Paxillin , Phosphorylation , Receptors, Fibroblast Growth Factor/metabolism , Tyrosine/metabolism
13.
J Immunol ; 168(9): 4674-81, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11971017

ABSTRACT

A possible protective role of IL-18 in host defense against blood-stage murine malarial infection was studied in BALB/c mice using a nonlethal strain, Plasmodium yoelii 265, and a lethal strain, Plasmodium berghei ANKA. Infection induced an increase in mRNA expression of IL-18, IL-12p40, IFN-gamma, and TNF-alpha in the case of P. yoelii 265 and an increase of IL-18, IL-12p40, and IFN-gamma in the case of P. berghei ANKA. The timing of mRNA expression of IL-18 in both cases was consistent with a role in the induction of IFN-gamma protein expression. Histological examination of spleen and liver tissues from infected controls treated with PBS showed poor cellular inflammatory reaction, massive necrosis, a large number of infected parasitized RBCs, and severe deposition of hemozoin pigment. In contrast, IL-18-treated infected mice showed massive infiltration of inflammatory cells consisting of mononuclear cells and Kupffer cells, decreased necrosis, and decreased deposition of the pigment hemozoin. Treatment with rIL-18 increased serum IFN-gamma levels in mice infected with both parasites, delayed onset of parasitemia, conferred a protective effect, and thus increased survival rate of infected mice. Administration of neutralizing anti-IL-18 Ab exacerbated infection, impaired host resistance and shortened the mean survival of mice infected with P. berghei ANKA. Furthermore, IL-18 knockout mice were more susceptible to P. berghei ANKA than were wild-type C57BL/6 mice. These data suggest that IL-18 plays a protective role in host defense by enhancing IFN-gamma production during blood-stage infection by murine malaria.


Subject(s)
Interleukin-18/physiology , Malaria/immunology , Plasmodium berghei , Plasmodium yoelii , Animals , Cytokines/biosynthesis , Cytokines/blood , Cytokines/genetics , Disease Progression , Disease Susceptibility , Erythrocytes/parasitology , Female , Interferon-gamma/blood , Interleukin-18/genetics , Interleukin-18/pharmacology , Kinetics , Liver/immunology , Liver/pathology , Malaria/blood , Malaria/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Plasmodium berghei/growth & development , Plasmodium yoelii/growth & development , RNA, Messenger/biosynthesis , Spleen/immunology , Survival Analysis , Transcriptional Activation
15.
s.l; s.n; 1998. 6 p. ilus, tab.
Non-conventional in English | Sec. Est. Saúde SP, HANSEN, Hanseníase Leprosy, SESSP-ILSLACERVO, Sec. Est. Saúde SP | ID: biblio-1237420
SELECTION OF CITATIONS
SEARCH DETAIL
...