Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Immunity ; 37(2): 264-75, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22902234

ABSTRACT

Mature dendritic cells (DCs) are established as unrivaled antigen-presenting cells (APCs) in the initiation of immune responses, whereas steady-state DCs induce peripheral T cell tolerance. Using various genetic approaches, we depleted CD11c(+) DCs in mice and induced autoimmune CNS inflammation. Unexpectedly, mice lacking DCs developed aggravated disease compared to control mice. Furthermore, when we engineered DCs to present a CNS-associated autoantigen in an induced manner, we found robust tolerance that prevented disease, which coincided with an upregulation of the PD-1 receptor on antigen-specific T cells. Additionally, we showed that PD-1 was necessary for DC-mediated induction of regulatory T cells. Our results show that a reduction of DCs interferes with tolerance, resulting in a stronger inflammatory response, and that other APC populations could compensate for the loss of immunogenic APC function in DC-depleted mice.


Subject(s)
Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Immune Tolerance/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigen Presentation/immunology , Autoantigens/immunology , Autoimmunity/immunology , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , CD11c Antigen , Dendritic Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
2.
Proc Natl Acad Sci U S A ; 113(41): E6145-E6152, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27671624

ABSTRACT

Laquinimod is an oral drug currently being evaluated for the treatment of relapsing, remitting, and primary progressive multiple sclerosis and Huntington's disease. Laquinimod exerts beneficial activities on both the peripheral immune system and the CNS with distinctive changes in CNS resident cell populations, especially astrocytes and microglia. Analysis of genome-wide expression data revealed activation of the aryl hydrocarbon receptor (AhR) pathway in laquinimod-treated mice. The AhR pathway modulates the differentiation and function of several cell populations, many of which play an important role in neuroinflammation. We therefore tested the consequences of AhR activation in myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) using AhR knockout mice. We demonstrate that the pronounced effect of laquinimod on clinical score, CNS inflammation, and demyelination in EAE was abolished in AhR-/- mice. Furthermore, using bone marrow chimeras we show that deletion of AhR in the immune system fully abrogates, whereas deletion within the CNS partially abrogates the effect of laquinimod in EAE. These data strongly support the idea that AhR is necessary for the efficacy of laquinimod in EAE and that laquinimod may represent a first-in-class drug targeting AhR for the treatment of multiple sclerosis and other neurodegenerative diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Quinolones/pharmacology , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/metabolism , Animals , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Deletion , Gene Expression , Gene Expression Profiling , Gene Expression Regulation/drug effects , Hepatocytes/metabolism , Humans , Immune System/immunology , Immune System/metabolism , Mice , Mice, Knockout , Receptors, Aryl Hydrocarbon/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcriptome
3.
Hum Mol Genet ; 25(18): 3975-3987, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27466197

ABSTRACT

Pridopidine has demonstrated improvement in Huntington Disease (HD) motor symptoms as measured by secondary endpoints in clinical trials. Originally described as a dopamine stabilizer, this mechanism is insufficient to explain the clinical and preclinical effects of pridopidine. This study therefore explored pridopidine's potential mechanisms of action. The effect of pridopidine versus sham treatment on genome-wide expression profiling in the rat striatum was analysed and compared to the pathological expression profile in Q175 knock-in (Q175 KI) vs Q25 WT mouse models. A broad, unbiased pathway analysis was conducted, followed by testing the enrichment of relevant pathways. Pridopidine upregulated the BDNF pathway (P = 1.73E-10), and its effect on BDNF secretion was sigma 1 receptor (S1R) dependent. Many of the same genes were independently found to be downregulated in Q175 KI mice compared to WT (5.2e-7 < P < 0.04). In addition, pridopidine treatment upregulated the glucocorticoid receptor (GR) response, D1R-associated genes and the AKT/PI3K pathway (P = 1E-10, P = 0.001, P = 0.004, respectively). Pridopidine upregulates expression of BDNF, D1R, GR and AKT/PI3K pathways, known to promote neuronal plasticity and survival, as well as reported to demonstrate therapeutic benefit in HD animal models. Activation of S1R, necessary for its effect on the BDNF pathway, represents a core component of the mode of action of pridopidine. Since the newly identified pathways are downregulated in neurodegenerative diseases, including HD, these findings suggest that pridopidine may exert neuroprotective effects beyond its role in alleviating some symptoms of HD.


Subject(s)
Brain-Derived Neurotrophic Factor/biosynthesis , Corpus Striatum/metabolism , Huntington Disease/drug therapy , Neuroprotective Agents/administration & dosage , Piperidines/administration & dosage , Animals , Brain-Derived Neurotrophic Factor/genetics , Corpus Striatum/drug effects , Corpus Striatum/pathology , Disease Models, Animal , Gene Expression Regulation/genetics , Genome , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Mice , Neuroprotective Agents/metabolism , Rats , Receptors, Dopamine D5/biosynthesis , Receptors, Dopamine D5/genetics , Receptors, Glucocorticoid/biosynthesis , Receptors, Glucocorticoid/genetics , Signal Transduction/drug effects
4.
Immunity ; 29(6): 986-97, 2008 Dec 19.
Article in English | MEDLINE | ID: mdl-19062318

ABSTRACT

Dendritic cells are critically involved in the promotion and regulation of T cell responses. Here, we report a mouse strain that lacks conventional CD11c(hi) dendritic cells (cDCs) because of constitutive cell-type specific expression of a suicide gene. As expected, cDC-less mice failed to mount effective T cell responses resulting in impaired viral clearance. In contrast, neither thymic negative selection nor T regulatory cell generation or T cell homeostasis were markedly affected. Unexpectedly, cDC-less mice developed a progressive myeloproliferative disorder characterized by prominent extramedullary hematopoiesis and increased serum amounts of the cytokine Flt3 ligand. Our data identify a critical role of cDCs in the control of steady-state hematopoiesis, revealing a feedback loop that links peripheral cDCs to myelogenesis through soluble growth factors, such as Flt3 ligand.


Subject(s)
Dendritic Cells/immunology , Membrane Proteins/immunology , Myeloproliferative Disorders/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Proliferation , Dendritic Cells/metabolism , Homeostasis/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Myeloproliferative Disorders/metabolism , Syndrome , T-Lymphocytes, Regulatory/metabolism
5.
Proc Natl Acad Sci U S A ; 107(33): 14745-50, 2010 Aug 17.
Article in English | MEDLINE | ID: mdl-20679228

ABSTRACT

Lymphoid organs are characterized by a complex network of phenotypically distinct dendritic cells (DC) with potentially unique roles in pathogen recognition and immunostimulation. Classical DC (cDC) include two major subsets distinguished in the mouse by the expression of CD8alpha. Here we describe a subset of CD8alpha(+) DC in lymphoid organs of naïve mice characterized by expression of the CX(3)CR1 chemokine receptor. CX(3)CR1(+) CD8alpha(+) DC lack hallmarks of classical CD8alpha(+) DC, including IL-12 secretion, the capacity to cross-present antigen, and their developmental dependence on the transcriptional factor BatF3. Gene-expression profiling showed that CX(3)CR1(+) CD8alpha(+) DC resemble CD8alpha(-) cDC. The microarray analysis further revealed a unique plasmacytoid DC (PDC) gene signature of CX(3)CR1(+) CD8alpha(+) DC. A PDC relationship of the cells is supported further by the fact that they harbor characteristic D-J Ig gene rearrangements and that development of CX(3)CR1(+) CD8alpha(+) DC requires E2-2, the critical transcriptional regulator of PDC. Thus, CX(3)CR1(+) CD8alpha(+) DC represent a unique DC subset, related to but distinct from PDC. Collectively, the expression-profiling data of this study refine the resolution of previous DC definitions, sharpen the border of classical CD8alpha(+) and CD8alpha(-) DC, and should assist the identification of human counterparts of murine DC subsets.


Subject(s)
CD8 Antigens/immunology , Cell Lineage/immunology , Dendritic Cells/immunology , Receptors, Cytokine/immunology , Receptors, HIV/immunology , Animals , Antigen Presentation/immunology , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , Basic-Leucine Zipper Transcription Factors/metabolism , CD8 Antigens/genetics , CD8 Antigens/metabolism , CX3C Chemokine Receptor 1 , Cell Lineage/genetics , Dendritic Cells/metabolism , Female , Flow Cytometry , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunophenotyping , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Receptors, HIV/genetics , Receptors, HIV/metabolism , Repressor Proteins/genetics , Repressor Proteins/immunology , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
6.
Eur J Immunol ; 41(2): 291-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21267999

ABSTRACT

Classical DC (cDC) are required for efficient protective T-cell immunity. Moreover, recent data indicate that cDC also play a critical role in mediating homeostatic proliferation and maintenance of peripheral Treg. Here, we corroborate these findings by defining CD80/CD86 costimulation as an essential molecular component required for the cDC-Treg interactions. In contrast to earlier reports, the reduced Treg compartment of mice lacking cDC or selective CD80/86 expression on cDC, as such, did not render the respective animals prone to systemic lymphocyte hyperactivation or autoimmunity. Rather, we provide evidence that elevated immunoglobulin titers, as well as changes in T-cell subset prevalence and activation status are strictly associated with the nonmalignant myeloproliferative disorder triggered by the absence of cDC.


Subject(s)
B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies/blood , Autoimmunity/immunology , B7-1 Antigen/genetics , B7-2 Antigen/genetics , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Count , Chimera/immunology , Dendritic Cells/metabolism , Forkhead Transcription Factors/metabolism , Immune Tolerance/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloproliferative Disorders/etiology , Myeloproliferative Disorders/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , fms-Like Tyrosine Kinase 3/metabolism
7.
J Clin Invest ; 118(12): 3954-65, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19033665

ABSTRACT

Implantation is a key stage during pregnancy, as the fate of the embryo is often decided upon its first contact with the maternal endometrium. Around this time, DCs accumulate in the uterus; however, their role in pregnancy and, more specifically, implantation, remains unknown. We investigated the function of uterine DCs (uDCs) during implantation using a transgenic mouse model that allows conditional ablation of uDCs in a spatially and temporally regulated manner. Depletion of uDCs resulted in a severe impairment of the implantation process, leading to embryo resorption. Depletion of uDCs also caused embryo resorption in syngeneic and T cell-deficient pregnancies, which argues against a failure to establish immunological tolerance during implantation. Moreover, even in the absence of embryos, experimentally induced deciduae failed to adequately form. Implantation failure was associated with impaired decidual proliferation and differentiation. Dynamic contrast-enhanced MRI revealed perturbed angiogenesis characterized by reduced vascular expansion and attenuated maturation. We suggest therefore that uDCs directly fine-tune decidual angiogenesis by providing two critical factors, sFlt1 and TGF-beta1, that promote coordinated blood vessel maturation. Collectively, uDCs appear to govern uterine receptivity, independent of their predicted role in immunological tolerance, by regulating tissue remodeling and angiogenesis. Importantly, our results may aid in understanding the limited implantation success of embryos transferred following in vitro fertilization.


Subject(s)
Dendritic Cells/immunology , Embryo Implantation/immunology , Embryo, Mammalian/immunology , Endometrium/immunology , Immune Tolerance/physiology , Pregnancy/immunology , Animals , Cell Differentiation/immunology , Cell Proliferation , Dendritic Cells/cytology , Embryo Implantation/genetics , Embryo Loss/genetics , Embryo Loss/immunology , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Endometrium/blood supply , Endometrium/cytology , Female , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/immunology , Pregnancy/genetics , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/immunology , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/immunology
8.
J Neuroimmunol ; 290: 84-95, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26711576

ABSTRACT

Glatiramer acetate (Copaxone®; GA) is a non-biological complex drug for multiple sclerosis. GA modulated thousands of genes in genome-wide expression studies conducted in THP-1 cells and mouse splenocytes. Comparing GA with differently-manufactured glatiramoid Polimunol (Synthon) in mice yielded hundreds of differentially expressed probesets, including biologically-relevant genes (e.g. Il18, adj p<9e-6) and pathways. In human monocytes, 700+ probesets differed between Polimunol and GA, enriching for 130+ pathways including response to lipopolysaccharide (adj. p<0.006). Key differences were confirmed by qRT-PCR (splenocytes) or proteomics (THP-1). These studies demonstrate the complexity of GA's mechanisms of action, and may help inform therapeutic equivalence assessment.


Subject(s)
Glatiramer Acetate/chemistry , Glatiramer Acetate/pharmacology , Spleen/drug effects , Spleen/physiology , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/therapeutic use , Animals , Cell Line , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glatiramer Acetate/therapeutic use , Humans , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Mice , Mice, Inbred BALB C , Monocytes/drug effects , Monocytes/physiology , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology
9.
Sci Rep ; 5: 10191, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25998228

ABSTRACT

Glatiramer Acetate (GA) has provided safe and effective treatment for multiple sclerosis (MS) patients for two decades. It acts as an antigen, yet the precise mechanism of action remains to be fully elucidated, and no validated pharmacokinetic or pharmacodynamic biomarkers exist. In order to better characterize GA's biological impact, genome-wide expression studies were conducted with a human monocyte (THP-1) cell line. Consistent with previous literature, branded GA upregulated anti-inflammatory markers (e.g. IL10), and modulated multiple immune-related pathways. Despite some similarities, significant differences were observed between expression profiles induced by branded GA and Probioglat, a differently-manufactured glatiramoid purported to be a generic GA. Key results were verified using qRT-PCR. Genes (e.g. CCL5, adj. p < 4.1 × 10(-5)) critically involved in pro-inflammatory pathways (e.g. response to lipopolysaccharide, adj. p = 8.7 × 10(-4)) were significantly induced by Probioglat compared with branded GA. Key genes were also tested and confirmed at the protein level, and in primary human monocytes. These observations suggest differential biological impact by the two glatiramoids and warrant further investigation.


Subject(s)
Glatiramer Acetate/pharmacology , Transcriptome/drug effects , Cell Line , Chemokines/genetics , Chemokines/metabolism , Humans , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL