Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
4.
Handb Exp Pharmacol ; 243: 329-353, 2017.
Article in English | MEDLINE | ID: mdl-28382469

ABSTRACT

HF patients with signs and symptoms of worsening heart failure (HF), despite optimal medical therapy, have a poor prognosis. The pathways contributing to HF are multiple, probably accounting, in part, for current treatment approaches not being more effective. Stem cells, particularly mesenchymal stem cells (MSCs), have a broad range of activities, making them particularly interesting candidates for a new HF therapeutic. This review presents an overview of the studies examining the efficacy of stem cell studies administered to HF patients, focusing mainly on MSCs. It examines the issues surrounding autologous vs. allogenic stem cells, the results of different routes of administration, and implications deriving from the belief that for stem cells to be effective, they must engraft in the myocardium and exert local effects. Since intravenous administration of stem cells leads to sparse cardiac engraftment, stem cell delivery strategies have uniformly involved catheter-based delivery systems. This becomes problematic in a disease that will almost certainly require delivery of the therapeutic throughout the course of the disease. Importantly, it appears that a critical contributing cause of the progressive cardiac dysfunction experienced by HF patients is the existence of a persistent inflammatory response. Since MSCs exert potent anti-inflammatory effects through paracrine mechanisms, it is possible that intravenous delivery of MSCs may be therapeutically effective. If this concept is valid, it could lead to a transformational change in stem cell delivery strategies.


Subject(s)
Heart Failure/therapy , Mesenchymal Stem Cell Transplantation/methods , Cardiomyopathies/complications , Graft Rejection/prevention & control , Heart Failure/etiology , Humans , Immunosuppressive Agents/therapeutic use , Myocardial Ischemia/complications , Transplantation, Autologous , Transplantation, Homologous
5.
Circ Res ; 120(10): 1598-1613, 2017 May 12.
Article in English | MEDLINE | ID: mdl-28232595

ABSTRACT

RATIONALE: Virtually all mesenchymal stem cell (MSC) studies assume that therapeutic effects accrue from local myocardial effects of engrafted MSCs. Because few intravenously administered MSCs engraft in the myocardium, studies have mainly utilized direct myocardial delivery. We adopted a different paradigm. OBJECTIVE: To test whether intravenously administered MSCs reduce left ventricular (LV) dysfunction both post-acute myocardial infarction and in ischemic cardiomyopathy and that these effects are caused, at least partly, by systemic anti-inflammatory activities. METHODS AND RESULTS: Mice underwent 45 minutes of left anterior descending artery occlusion. Human MSCs, grown chronically at 5% O2, were administered intravenously. LV function was assessed by serial echocardiography, 2,3,5-triphenyltetrazolium chloride staining determined infarct size, and fluorescence-activated cell sorting assessed cell composition. Fluorescent and radiolabeled MSCs (1×106) were injected 24 hours post-myocardial infarction and homed to regions of myocardial injury; however, the myocardium contained only a small proportion of total MSCs. Mice received 2×106 MSCs or saline intravenously 24 hours post-myocardial infarction (n=16 per group). At day 21, we harvested blood and spleens for fluorescence-activated cell sorting and hearts for 2,3,5-triphenyltetrazolium chloride staining. Adverse LV remodeling and deteriorating LV ejection fraction occurred in control mice with large infarcts (≥25% LV). Intravenous MSCs eliminated the progressive deterioration in LV end-diastolic volume and LV end-systolic volume. MSCs significantly decreased natural killer cells in the heart and spleen and neutrophils in the heart. Specific natural killer cell depletion 24 hours pre-acute myocardial infarction significantly improved infarct size, LV ejection fraction, and adverse LV remodeling, changes associated with decreased neutrophils in the heart. In an ischemic cardiomyopathy model, mice 4 weeks post-myocardial infarction were randomized to tail-vein injection of 2×106 MSCs, with injection repeated at week 3 (n=16) versus PBS control (n=16). MSCs significantly increased LV ejection fraction and decreased LV end-systolic volume. CONCLUSIONS: Intravenously administered MSCs for acute myocardial infarction attenuate the progressive deterioration in LV function and adverse remodeling in mice with large infarcts, and in ischemic cardiomyopathy, they improve LV function, effects apparently modulated in part by systemic anti-inflammatory activities.


Subject(s)
Cardiomyopathies/therapy , Mesenchymal Stem Cell Transplantation/methods , Myocardial Infarction/therapy , Myocardial Ischemia/therapy , Ventricular Dysfunction, Left/therapy , Administration, Intravenous , Animals , Cardiomyopathies/immunology , Cardiomyopathies/physiopathology , Cells, Cultured , Humans , Male , Mesenchymal Stem Cells/immunology , Mice , Myocardial Infarction/immunology , Myocardial Infarction/physiopathology , Myocardial Ischemia/immunology , Myocardial Ischemia/physiopathology , Treatment Outcome , Ventricular Dysfunction, Left/immunology , Ventricular Dysfunction, Left/physiopathology
6.
J Am Coll Cardiol ; 67(17): 2050-60, 2016 05 03.
Article in English | MEDLINE | ID: mdl-27126533

ABSTRACT

Treatment of acute myocardial infarction (AMI) has improved significantly in recent years, but many patients have adverse left ventricular (LV) remodeling, a maladaptive change associated with progressive heart failure. Although this change is usually associated with large infarcts, some patients with relatively small infarcts have adverse remodeling, whereas other patients with larger infarcts (who survive the first several days after AMI) do not. This paper reviews the relevant data supporting the hypothesis that individual differences in the intensity of the post-AMI inflammatory response, involving 1 or more inflammatory-modulating pathways, may contribute to adverse LV remodeling. It concludes by outlining how individual variations in the inflammatory response could provide important novel therapeutic targets and strategies.


Subject(s)
Inflammation/physiopathology , Myocardial Infarction/physiopathology , Ventricular Remodeling/physiology , Autoimmune Diseases/complications , Biomarkers/metabolism , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Humans , Infections/complications , Inflammasomes/antagonists & inhibitors , Inflammation/prevention & control , Mesenchymal Stem Cell Transplantation , Myocardium/metabolism , Neovascularization, Physiologic , Neutrophils/metabolism , Polymorphism, Genetic , Stroke Volume/physiology
7.
Cardiovasc Revasc Med ; 17(2): 106-12, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26874740

ABSTRACT

INTRODUCTION: Nanoparticles may serve as a promising means to deliver novel therapeutics to the myocardium following myocardial infarction. We sought to determine whether lipid-based liposomal nanoparticles can be shown through different imaging modalities to specifically target injured myocardium following intravenous injection in an ischemia-reperfusion murine myocardial infarction model. METHODS: Mice underwent ischemia-reperfusion surgery and then either received tail-vein injection with gadolinium- and fluorescent-labeled liposomes or no injection (control). The hearts were harvested 24h later and underwent T1 and T2-weighted ex vivo imaging using a 7 Tesla Bruker magnet. The hearts were then sectioned for immunohistochemistry and optical fluorescent imaging. RESULTS: The mean size of the liposomes was 100nm. T1-weighted signal intensity was significantly increased in the ischemic vs. the non-ischemic myocardium for mice that received liposomes compared with control. Optical imaging demonstrated significant fluorescence within the infarct area for the liposome group compared with control (163±31% vs. 13±14%, p=0.001) and fluorescent microscopy confirmed the presence of liposomes within the ischemic myocardium. CONCLUSIONS: Liposomes traffic to the heart and preferentially home to regions of myocardial injury, enabling improved diagnosis of myocardial injury and could serve as a vehicle for drug delivery.


Subject(s)
Albumins/pharmacokinetics , Contrast Media/pharmacokinetics , Fluorescent Dyes/pharmacokinetics , Gadolinium DTPA/pharmacokinetics , Magnetic Resonance Imaging , Myocardial Infarction/diagnostic imaging , Myocardial Reperfusion Injury/diagnostic imaging , Myocardium/metabolism , Optical Imaging/methods , Phosphatidylethanolamines/pharmacokinetics , Albumins/administration & dosage , Animals , Contrast Media/administration & dosage , Disease Models, Animal , Fluorescent Dyes/administration & dosage , Gadolinium DTPA/administration & dosage , Immunohistochemistry , Injections, Intravenous , Liposomes , Male , Mice , Microscopy, Fluorescence , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Nanoparticles , Particle Size , Phosphatidylethanolamines/administration & dosage , Tissue Distribution
8.
J Am Coll Cardiol ; 66(18): 2038-2047, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26516007

ABSTRACT

Over the past 1.5 decades, numerous stem cell trials have been performed in patients with cardiovascular disease. Although encouraging outcome signals have been reported, these have been small, leading to uncertainty as to whether they will translate into significantly improved outcomes. A reassessment of the rationale for the use of stem cells in cardiovascular disease is therefore timely. Such a rationale should include analyses of why previous trials have not produced significant benefit and address whether mechanisms contributing to disease progression might benefit from known activities of stem cells. The present paper provides such a reassessment, focusing on patients with left ventricular systolic dysfunction, either nonischemic or ischemic. We conclude that many mechanisms contributing to progressive left ventricular dysfunction are matched by stem cell activities that could attenuate the myocardial effect of such mechanisms. This suggests that stem cell strategies may improve patient outcomes and justifies further testing.


Subject(s)
Cardiomyopathy, Dilated , Stem Cell Transplantation , Cardiomyopathy, Dilated/etiology , Cardiomyopathy, Dilated/physiopathology , Cardiomyopathy, Dilated/therapy , Disease Progression , Humans , Myocardial Ischemia/complications , Outcome Assessment, Health Care , Stem Cell Transplantation/methods , Stem Cell Transplantation/statistics & numerical data , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/therapy
9.
BMC Dev Biol ; 14: 24, 2014 May 28.
Article in English | MEDLINE | ID: mdl-24886590

ABSTRACT

BACKGROUND: Chloride Intracellular Channel 4 (CLIC4) is one of seven members in the closely related CLIC protein family. CLIC4 is involved in multiple cellular processes including apoptosis, cellular differentiation, inflammation and endothelial tubulogenesis. Despite over a decade of research, no comprehensive in situ expression analysis of CLIC4 in a living organism has been reported. In order to fulfill this goal, we generated a knock-in mouse to express Green Fluorescent Protein (GFP) from the CLIC4 locus, thus substituting the GFP coding region for CLIC4. We used GFP protein expression to eliminate cross reaction with other CLIC family members. RESULTS: We analyzed CLIC4 expression during embryonic development and adult organs. During mid and late gestation, CLIC4 expression is modulated particularly in fetal brain, heart, thymus, liver and kidney as well as in developing brown adipose tissue and stratifying epidermis. In the adult mouse, CLIC4 is highly expressed globally in vascular endothelial cells as well as in liver, lung alveolar septae, pancreatic acini, spermatogonia, renal proximal tubules, cardiomyocytes and thymic epithelial cells. Neural expression included axonal tracks, olfactory bulb, Purkinje cell layer and dentate gyrus. Renal CLIC4 expression was most pronounced in proximal tubules, although altered renal function was not detected in the absence of CLIC4. Myeloid cells and B cells of the spleen are rich in CLIC4 expression as are CD4 and CD8 positive T cells. CONCLUSIONS: In a comprehensive study detailing CLIC4 expression in situ in a mouse model that excludes cross reaction with other family members, we were able to document previously unreported expression for CLIC4 in developing fetus, particularly the brain. In addition, compartmentalized expression of CLIC4 in specific adult tissues and cells provides a focus to explore potential functions of this protein not addressed previously.


Subject(s)
Chloride Channels/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Mitochondrial Proteins/genetics , Adipose Tissue, Brown/embryology , Adipose Tissue, Brown/growth & development , Adipose Tissue, Brown/metabolism , Animals , Brain/embryology , Brain/growth & development , Brain/metabolism , Chloride Channels/metabolism , Epidermis/embryology , Epidermis/growth & development , Epidermis/metabolism , Fetal Heart/embryology , Fetal Heart/metabolism , Green Fluorescent Proteins/metabolism , Heart/growth & development , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Kidney/embryology , Kidney/growth & development , Kidney/metabolism , Liver/embryology , Liver/growth & development , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Mitochondrial Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thymus Gland/embryology , Thymus Gland/growth & development , Thymus Gland/metabolism
10.
PLoS One ; 8(10): e76115, 2013.
Article in English | MEDLINE | ID: mdl-24146823

ABSTRACT

The role of myeloid derived suppressor cells (MDSCs) in promoting tumorigenesis is well-established, and significant effort is being made to further characterize surface markers on MDSCs both for better diagnosis and as potential targets for therapy. Here we show that the B cell receptor adaptor molecule CD79a is unexpectedly expressed on immature bone marrow myeloid cells, and is upregulated on MDSCs generated in multiple different mouse models of metastatic but not non-metastatic cancer. CD79a on MDSCs is upregulated and activated in response to soluble factors secreted by tumor cells. Activation of CD79a on mouse MDSCs, by crosslinking with a specific antibody, maintained their immature phenotype (CD11b+Gr1+), enhanced their migration, increased their suppressive effect on T cell proliferation, and increased secretion of pro-tumorigenic cytokines such as IL-6 and CCL22. Furthermore, crosslinking CD79a on myeloid cells activated signaling through Syk, BLNK, ERK and STAT3 phosphorylation. In vivo, CD79+ myeloid cells showed enhanced ability to promote primary tumor growth and metastasis. Finally we demonstrate that CD79a is upregulated on circulating myeloid cells from lung cancer patients, and that CD79a+ myeloid cells infiltrate human breast tumors. We propose that CD79a plays a functional role in the tumor promoting effects of myeloid cells, and may represent a novel target for cancer therapy.


Subject(s)
B-Lymphocytes/metabolism , Breast Neoplasms/genetics , CD79 Antigens/genetics , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antibodies/metabolism , Antigen-Antibody Complex/metabolism , B-Lymphocytes/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CD11b Antigen/genetics , CD11b Antigen/metabolism , CD79 Antigens/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Movement , Cell Proliferation , Chemokine CCL22/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Interleukin-6/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Myeloid Cells/metabolism , Myeloid Cells/pathology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Syk Kinase , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
11.
Breast Cancer Res ; 14(4): R109, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22812567

ABSTRACT

INTRODUCTION: Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that is diagnosed in approximately 15% of all human breast cancer (BrCa) patients. Currently, no targeted therapies exist for this subtype of BrCa and prognosis remains poor. Our laboratory has previously identified a proliferation/DNA repair/cell cycle gene signature (Tag signature) that is characteristic of human TNBC. We hypothesize that targeting the dysregulated biological networks in the Tag gene signature will lead to the identification of improved combination therapies for TNBC. METHODS: Cross-species genomic analysis was used to identify human breast cancer cell lines that express the Tag signature. Knock-down of the up-regulated genes in the Tag signature by siRNA identified several genes that are critical for TNBC cell growth. Small molecule inhibitors to two of these genes were analyzed, alone and in combination, for their effects on cell proliferation, cell cycle, and apoptosis in vitro and tumor growth in vivo. Synergy between the two drugs was analyzed by the Chou-Talalay method. RESULTS: A custom siRNA screen was used to identify targets within the Tag signature that are critical for growth of TNBC cells. Ribonucleotide reductase 1 and 2 (RRM1 and 2) and checkpoint kinase 1 (CHK1) were found to be critical targets for TNBC cell survival. Combination therapy, to simultaneously attenuate cell cycle checkpoint control through inhibition of CHK1 while inducing DNA damage with gemcitabine, improved therapeutic efficacy in vitro and in xenograft models of TNBC. CONCLUSIONS: This combination therapy may have translational value for patients with TNBC and improve therapeutic response for this aggressive form of breast cancer.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Protein Kinases/genetics , Ribonucleotide Reductases/antagonists & inhibitors , Triple Negative Breast Neoplasms/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation , Checkpoint Kinase 1 , Cluster Analysis , DNA Damage/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Inhibitory Concentration 50 , Mice , Protein Kinases/metabolism , RNA Interference , RNA, Small Interfering/genetics , Retinoblastoma Protein/metabolism , Ribonucleoside Diphosphate Reductase/antagonists & inhibitors , Ribonucleoside Diphosphate Reductase/genetics , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Burden/drug effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays , Gemcitabine
12.
Int J Mol Med ; 28(5): 727-31, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21887476

ABSTRACT

Uveitis is an inflammation of the middle layer of the eye with a high risk of blindness. The Gi protein associated A3 adenosine receptor (A3AR) is highly expressed in inflammatory cells whereas low expression is found in normal cells. CF101 is a highly specific agonist at the A3AR known to induce a robust anti-inflammatory effect in different experimental animal models. The CF101 mechanism of action entails down-regulation of the NF-κB-TNF-α signaling pathway, resulting in inhibition of pro-inflammatory cytokine production and apoptosis of inflammatory cells. In this study the effect of CF101 on the development of retinal antigen interphotoreceptor retinoid-binding protein (IRBP)-induced experimental autoimmune uveitis (EAU) was assessed. Oral treatment with CF101 (10 µg/kg, twice daily), initiated upon disease onset, improved uveitis clinical score measured by fundoscopy and ameliorated the pathological manifestations of the disease. Shortly after treatment with CF101 A3AR expression levels were down-regulated in the lymph node and spleen cells pointing towards receptor activation. Downstream events included a decrease in PI3K and STAT-1 and proliferation inhibition of IRPB auto-reactive T cells ex vivo. Inhibition of interleukin-2, tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) production and up-regulation of interleukin-10 was found in cultured splenocytes derived from CF101-treated animals. Overall, the present study data point towards a marked anti-inflammatory effect of CF101 in EAU and support further exploration of this small molecule drug for the treatment of uveitis.


Subject(s)
Adenosine A3 Receptor Agonists/therapeutic use , Adenosine/analogs & derivatives , Uveitis/drug therapy , Uveitis/immunology , Adenosine/therapeutic use , Animals , Cell Proliferation/drug effects , Cells, Cultured , Interleukin-10/metabolism , Interleukin-2/metabolism , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Receptor, Adenosine A3/metabolism , STAT1 Transcription Factor/metabolism , Spleen/metabolism , Tumor Necrosis Factor-alpha/metabolism , Uveitis/metabolism
13.
J Immunol ; 182(8): 4624-32, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19342637

ABSTRACT

Experimental autoimmune uveitis (EAU) serves as a model for human autoimmune uveitis and for cell-mediated autoimmunity in general. EAU induced in mice by immunization with the retinal Ag interphotoreceptor retinoid-binding protein in CFA is driven by the Th17 response. Oral calcitriol (1,25-dihydroxyvitamin D(3)) prevented as well as partly reversed disease and suppressed immunological responses. In vitro, calcitriol directly suppressed IL-17 induction in purified naive CD4(+) T cells without inhibiting Th17 lineage commitment, as reflected by unaltered RORgammat, STAT3, and FoxP3 expression. In contrast, in vivo treatment with calcitriol of mice challenged for EAU impaired commitment to the Th17 lineage, as judged by reduction of both RORgammat and IL-17 in CD4(+) T cells. Innate immune response parameters in draining lymph nodes of treated mice were suppressed, as was production of IL-1, IL-6, TNF-alpha, and IL-12/IL-23p40, but not IL-10, by explanted splenic dendritic cells (DC). Finally, supernatants of calcitriol-conditioned bone marrow-derived DC had reduced ability to support Th17 polarization of naive CD4(+) T cells in vitro and in vivo. Thus, calcitriol appears to suppress autoimmunity by inhibiting the Th17 response at several levels, including the ability of DC to support priming of Th17 cells, the ability of CD4(+) T cells to commit to the Th17 lineage, and the ability of committed Th17 T cells to produce IL-17.


Subject(s)
Autoimmunity/drug effects , Autoimmunity/immunology , Calcitriol/pharmacology , Interleukin-17/immunology , Retina/immunology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , Administration, Oral , Animals , Calcitriol/administration & dosage , Cell Lineage/drug effects , Cell Lineage/immunology , Cells, Cultured , Female , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice , Mice, Knockout , Retina/drug effects , T-Lymphocytes, Helper-Inducer/cytology , Uveitis/immunology
14.
J Leukoc Biol ; 84(2): 577-85, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18495789

ABSTRACT

We identified inhibitory peptide analogs (IPAs), capable of immunomodulating experimental autoimmune uveitis (EAU), induced in B10.RIII mice by immunization with the retinal antigen interphotoreceptor-binding protein in CFA. Alanine-substituted peptides of the major pathogenic epitope, residues 161-180, were synthesized. They were tested for immunogenicity, cross-reactivity with the native 161-180 epitope, pathogenicity, and ability to prevent EAU when given in IFA before EAU challenge with native murine (m)161-180. Two peptides, 169A and 171A, were unable to elicit disease but cross-reacted with m161-180 by lymphocyte proliferation. Mice pretreated with either of the substituted peptides failed to develop EAU after challenge with the native epitope, m161-180, and had reduced cellular responses by lymphocyte proliferation and by delayed hypersensitivity. Their cytokine response profile to m161-180 showed reduced antigen-specific IFN-gamma and IL-17, whereas IL-4, IL-5, IL-10, and IL-13 from IPA-protected mice were increased, and serum antibody titers to m161-180 revealed reduced IgG2a and elevated IgG1 isotypes, suggesting a Th2 shift in the response. Protection was transferable with lymphoid cells from protected donors to naïve recipients, who were subsequently immunized for EAU. Thus, IPA pretreatment prevents induction of EAU by skewing the response to a subsequent uveitogenic challenge with the native peptide to a nonpathogenic phenotype, as well as by eliciting transferable regulatory cells.


Subject(s)
Autoimmune Diseases/drug therapy , Peptides/therapeutic use , Retina/immunology , T-Lymphocytes, Regulatory/immunology , Uveitis/drug therapy , Uveitis/immunology , Adoptive Transfer , Amino Acid Sequence , Animals , Autoimmunity , Cytokines/metabolism , Immune Tolerance , Lymphocyte Activation , Mice , Models, Animal , Molecular Sequence Data , Peptides/chemistry , Structure-Activity Relationship
15.
J Exp Med ; 205(4): 799-810, 2008 Apr 14.
Article in English | MEDLINE | ID: mdl-18391061

ABSTRACT

Experimental autoimmune uveitis (EAU) represents autoimmune uveitis in humans. We examined the role of the interleukin (IL)-23-IL-17 and IL-12-T helper cell (Th)1 pathways in the pathogenesis of EAU. IL-23 but not IL-12 was necessary to elicit disease by immunization with the retinal antigen (Ag) interphotoreceptor retinoid-binding protein (IRBP) in complete Freund's adjuvant. IL-17 played a dominant role in this model; its neutralization prevented or reversed disease, and Th17 effector cells induced EAU in the absence of interferon (IFN)-gamma. In a transfer model, however, a polarized Th1 line could induce severe EAU independently of host IL-17. Furthermore, induction of EAU with IRBP-pulsed mature dendritic cells required generation of an IFN-gamma-producing effector response, and an IL-17 response by itself was insufficient to elicit pathology. Finally, genetic deficiency of IL-17 did not abrogate EAU susceptibility. Thus, autoimmune pathology can develop in the context of either a Th17 or a Th1 effector response depending on the model. The data suggest that the dominant effector phenotype may be determined at least in part by conditions present during initial exposure to Ag, including the quality/quantity of Toll-like receptor stimulation and/or type of Ag-presenting cells. These data also raise the possibility that the nonredundant requirement for IL-23 in EAU may extend beyond its role in promoting the Th17 effector response and help provide a balance in the current Th1 versus Th17 paradigm.


Subject(s)
Autoimmune Diseases/enzymology , Autoimmune Diseases/pathology , Autoimmunity/immunology , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/immunology , Animals , Antigens/immunology , Autoimmune Diseases/prevention & control , Cell Line , Dendritic Cells/immunology , Disease Susceptibility , Eye/pathology , Humans , Inflammation Mediators/metabolism , Interferon-gamma/deficiency , Interferon-gamma/immunology , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p35/immunology , Interleukin-17/immunology , Interleukin-23/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , Neutralization Tests , Up-Regulation , Uveitis/immunology , Uveitis/prevention & control
16.
J Immunol ; 180(8): 5167-71, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18390697

ABSTRACT

Th17 cells require IL-6 and TGFbeta for lineage commitment and IL-23 for maintenance. Unexpectedly, naive IL-6(-/-) splenocytes stimulated with anti-CD3 and IL-23 produced normal amounts of IL-17 during the first 24 h of culture. These rapid IL-6-independent IL-17 producers were identified as predominantly DX5(+) TCRbeta(+) NKT cells, and a comparable response could be found using the invariant NKT-specific ligand alpha-galactosylceramide. Human NKT cells also produced IL-17. NKT cells constitutively expressed IL-23R and RORgammat. Ligation of either TCR or IL-23R triggered IL-17 production and both together had a synergistic effect, suggesting independent but convergent pathways. IL-17 production was not restricted to a particular subset of NKT cells but they were NK1.1 negative. Importantly, in vivo administration of alpha-galactosylceramide triggered a rapid IL-17 response in the spleen. These data suggest an important biological role for innate IL-17 production by NKT cells that is rapid and precedes the adaptive IL-17 response.


Subject(s)
Interleukin-17/metabolism , Receptors, Interleukin/metabolism , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/metabolism , T-Lymphocyte Subsets/metabolism , Animals , Cells, Cultured , Humans , Immunity, Innate , Interleukin-17/immunology , Interleukin-23/immunology , Interleukin-23/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3 , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Interleukin/immunology , Receptors, Retinoic Acid/immunology , Receptors, Thyroid Hormone/immunology , T-Lymphocyte Subsets/immunology
17.
Ophthalmic Res ; 40(3-4): 169-74, 2008.
Article in English | MEDLINE | ID: mdl-18421234

ABSTRACT

The mouse model of experimental autoimmune uveitis, induced by immunization of mice with the retinal protein IRBP, was developed in our laboratory 20 years ago and published in 1988. Since that time it has been adopted by many investigators and has given rise to many studies that helped elucidate genetic influences, dissect the basic mechanisms of pathogenesis and test novel immunotherapeutic paradigms. The current overview will summarize the salient features of the experimental autoimmune uveitis model and discuss its mechanisms.


Subject(s)
Autoimmune Diseases/immunology , Disease Models, Animal , Uveitis/immunology , Animals , Autoantigens/immunology , Autoimmune Diseases/genetics , Autoimmunity/genetics , Epitopes/immunology , Genetic Predisposition to Disease , Haplotypes , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/immunology , Mice , Mice, Inbred Strains , Retina/immunology , T-Lymphocytes/immunology , Uveitis/genetics
18.
Semin Immunopathol ; 30(2): 135-43, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18317764

ABSTRACT

Experimental autoimmune uveitis (EAU) in its several variants represents human autoimmune uveitis and has been instrumental in obtaining insights into the basic mechanisms of disease. Studies have uncovered that in addition to CD4+ Th1 cells, uveitis can be induced also by CD8+ T cells. Antibodies may have a secondary role after the blood-retinal barrier has been broken. The role in uveitis of a recently discovered IL-17-producing effector T cell type, Th17, is being intensively studied. Th17 cells elicit EAU, can be found in uveitic eyes along with Th1 cells, and are dominant in some types of EAU. In other types of EAU, Th1 cells have a dominant role. The dominant effector type is at least in part determined by conditions under which initial exposure to self-antigen occurs. These findings shed light on the heterogeneity of human disease and may ultimately help to develop better and more rational treatment strategies for human uveitis.


Subject(s)
Autoimmune Diseases/immunology , Uveitis/immunology , Animals , Autoimmune Diseases/pathology , Humans , Interleukin-12/immunology , Interleukin-23/immunology , T-Lymphocyte Subsets/immunology , Uveitis/pathology
19.
J Immunol ; 179(8): 4979-87, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17911582

ABSTRACT

Experimental systemic lupus erythematosus (SLE) can be induced in mice following immunization with an anti-DNA mAb expressing a major Id, 16/6Id. Treatment with a peptide, designated human CDR1 (hCDR1; Edratide), that is based on the sequence of CDR1 of the 16/6Id ameliorated disease manifestations. In the present study, we investigated the roles of apoptosis and related molecules in BALB/c mice with induced experimental SLE following treatment with hCDR1. A higher state of activation and increased rate of apoptosis were found in lymphocytes of SLE-afflicted mice as compared with healthy controls. The latter effects were associated with up-regulated caspase-8 and caspase-3, and down-regulated Bcl-x(L). The ameliorative effects of hCDR1 were associated with down-regulation of caspase-8 and caspase-3, up-regulation of Bcl-x(L), and a reduced rate of apoptosis. Treatment of diseased mice with an apoptosis-reducing compound that inhibited caspases down-regulated the secretion of the pathogenic cytokine IFN-gamma and lowered the intensity of glomerular immune complex deposits and the levels of proteinuria. Furthermore, coincubation of Bcl-x(L) inhibitors with hCDR1-treated cells abrogated the ability of hCDR1 to reduce the activation state of lymphocytes and to down-regulate the secretion of IL-10 and IFN-gamma. Moreover, the Bcl-x(L)-expressing CD4(+)CD25(+) cells from hCDR1-treated mice induced the expression of Bcl-x(L) in CFSE-labeled CD4(+)CD25(-) cells of the SLE-afflicted mice. Thus, the reduction of apoptosis and the up-regulation of Bcl-x(L), which plays an apparent role in tolerance induction, contribute to at least part of the beneficial effects of hCDR1 on lupus manifestations.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/prevention & control , Nerve Tissue Proteins/physiology , Peptides/physiology , Amino Acid Sequence , Animals , Caspase 3/biosynthesis , Caspase 8/biosynthesis , Caspase Inhibitors , Disease Models, Animal , Down-Regulation/immunology , Female , Humans , Lupus Erythematosus, Systemic/enzymology , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Up-Regulation/immunology , bcl-X Protein/biosynthesis
20.
J Immunol ; 179(8): 5146-58, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17911600

ABSTRACT

The eye is an immunologically privileged organ whose Ags serve as targets for experimental autoimmune uveitis (EAU), a model for human uveitis. We used a hydrodynamic i.v. injection of naked DNA to express the uveitogenic retinal Ag interphotoreceptor retinoid-binding protein (IRBP) in the periphery, thus revoking its immune-privileged status. IRBP was expressed in the liver within hours of administration of as little as 10 microg of IRBP-DNA. Vaccinated mice were highly protected from EAU induced by immunization with IRBP for at least 10 wk after vaccination. Protection was partial in a reversal protocol. Mechanistic studies revealed specific hyporesponsiveness to IRBP without immune deviation, no evidence for apoptosis either by the Fas- or Bcl-2-regulated (mitochondrial) pathway and apparent lack of dependence on CD8(+) cells, IL-10, or TGF-beta. In contrast, depletion of CD25(+) cells after vaccination and before challenge markedly abrogated protection. IRBP-specific CD4(+)CD25(high) T cells could be cultured from vaccinated mice and transferred protection to unvaccinated, EAU-challenged recipients. In vitro characterization of these cells revealed that they are Ag specific, anergic, express FoxP3, CTLA-4, and glucocorticoid-induced TNFR, and suppress by contact. Thus, expression of IRBP in the periphery by DNA vaccination results in tolerance that acts at least in part through induction of IRBP-specific, FoxP3(+)CD4(+)CD25(+) regulatory T cells. DNA vaccination may offer a new approach to Ag-specific therapy of uveitis.


Subject(s)
Autoimmune Diseases/prevention & control , Cell Differentiation/immunology , Eye Proteins/administration & dosage , Eye Proteins/immunology , Retinol-Binding Proteins/administration & dosage , Retinol-Binding Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Uveitis/prevention & control , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Amino Acid Sequence , Animals , Autoimmune Diseases/genetics , Cell Differentiation/genetics , Cells, Cultured , Eye Proteins/genetics , Humans , Injections, Jet , Liver/immunology , Liver/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Retinol-Binding Proteins/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/transplantation , T-Lymphocytes, Regulatory/cytology , Uveitis/genetics , Uveitis/immunology , Vaccines, DNA/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...