Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Immunity ; 49(5): 857-872.e5, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30413363

ABSTRACT

Lineage-committed αß and γδ T cells are thought to originate from common intrathymic multipotent progenitors following instructive T cell receptor (TCR) signals. A subset of lymph node and mucosal Vγ2+ γδ T cells is programmed intrathymically to produce IL-17 (Tγδ17 cells), however the role of the γδTCR in development of these cells remains controversial. Here we generated reporter mice for the Tγδ17 lineage-defining transcription factor SOX13 and identified fetal-origin, intrathymic Sox13+ progenitors. In organ culture developmental assays, Tγδ17 cells derived primarily from Sox13+ progenitors, and not from other known lymphoid progenitors. Single cell transcriptome assays of the progenitors found in TCR-deficient mice demonstrated that Tγδ17 lineage programming was independent of γδTCR. Instead, generation of the lineage committed progenitors and Tγδ17 cells was controlled by TCF1 and SOX13. Thus, T lymphocyte lineage fate can be prewired cell-intrinsically and is not necessarily specified by clonal antigen receptor signals.


Subject(s)
Autoantigens/metabolism , Interleukin-17/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Animals , Autoantigens/genetics , Biomarkers , Gene Expression Profiling , Gene Regulatory Networks , Humans , Immunophenotyping , Mice , Mice, Knockout , Mice, Transgenic , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/immunology , Transcriptome
2.
Proc Natl Acad Sci U S A ; 111(34): 12468-73, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25114223

ABSTRACT

Genetic alterations that reduce the function of the immunoregulatory cytokine IL-10 contribute to colitis in mouse and man. Myeloid cells such as macrophages (MΦs) and dendritic cells (DCs) play an essential role in determining the relative abundance of IL-10 versus inflammatory cytokines in the gut. As such, using small molecules to boost IL-10 production by DCs-MΦs represents a promising approach to increase levels of this cytokine specifically in gut tissues. Toward this end, we screened a library of well-annotated kinase inhibitors for compounds that enhance production of IL-10 by murine bone-marrow-derived DCs stimulated with the yeast cell wall preparation zymosan. This approach identified a number of kinase inhibitors that robustly up-regulate IL-10 production including the Food and Drug Administration (FDA)-approved drugs dasatinib, bosutinib, and saracatinib that target ABL, SRC-family, and numerous other kinases. Correlating the kinase selectivity profiles of the active compounds with their effect on IL-10 production suggests that inhibition of salt-inducible kinases (SIKs) mediates the observed IL-10 increase. This was confirmed using the SIK-targeting inhibitor HG-9-91-01 and a series of structural analogs. The stimulatory effect of SIK inhibition on IL-10 is also associated with decreased production of the proinflammatory cytokines IL-1ß, IL-6, IL-12, and TNF-α, and these coordinated effects are observed in human DCs-MΦs and anti-inflammatory CD11c(+) CX3CR1(hi) cells isolated from murine gut tissue. Collectively, these studies demonstrate that SIK inhibition promotes an anti-inflammatory phenotype in activated myeloid cells marked by robust IL-10 production and establish these effects as a previously unidentified activity associated with several FDA-approved multikinase inhibitors.


Subject(s)
Dendritic Cells/drug effects , Dendritic Cells/immunology , Interleukin-10/biosynthesis , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Aniline Compounds/pharmacology , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Cytokines/biosynthesis , Dasatinib , Dendritic Cells/enzymology , Drug Evaluation, Preclinical , Humans , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/enzymology , Inflammatory Bowel Diseases/immunology , Intestine, Small/drug effects , Intestine, Small/enzymology , Intestine, Small/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/drug effects , Myeloid Cells/enzymology , Myeloid Cells/immunology , Nitriles/pharmacology , Phenylurea Compounds/chemistry , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemistry , Pyrimidines/pharmacology , Quinolines/pharmacology , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , Thiazoles/pharmacology , Transcription Factors/metabolism
3.
J Virol ; 88(18): 10748-57, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25008915

ABSTRACT

UNLABELLED: Retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) are essential intracellular detectors of viral RNA. They contribute to the type I interferon (IFN) response that is crucial for host defense against viral infections. Given the potent antiviral and proinflammatory activities elicited by the type I IFNs, induction of the type I IFN response is tightly regulated. Members of the tripartite motif (TRIM) family of proteins have recently emerged as key regulators of antiviral immunity. We show that TRIM13, an E3 ubiquitin ligase, is expressed in immune cells and is upregulated in bone marrow-derived macrophages upon stimulation with inducers of type I IFN. TRIM13 interacts with MDA5 and negatively regulates MDA5-mediated type I IFN production in vitro, acting upstream of IFN regulatory factor 3. We generated Trim13(-/-) mice and show that upon lethal challenge with encephalomyocarditis virus (EMCV), which is sensed by MDA5, Trim13(-/-) mice produce increased amounts of type I IFNs and survive longer than wild-type mice. Trim13(-/-) murine embryonic fibroblasts (MEFs) challenged with EMCV or poly(I · C) also show a significant increase in beta IFN (IFN-ß) levels, but, in contrast, IFN-ß responses to the RIG-I-detected Sendai virus were diminished, suggesting that TRIM13 may play a role in positively regulating RIG-I function. Together, these results demonstrate that TRIM13 regulates the type I IFN response through inhibition of MDA5 activity and that it functions nonredundantly to modulate MDA5 during EMCV infection. IMPORTANCE: The type I interferon (IFN) response is crucial for host defense against viral infections, and proper regulation of this pathway contributes to maintaining immune homeostasis. Retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) are intracellular detectors of viral RNA that induce the type I IFN response. In this study, we show that expression of the gene tripartite motif 13 (Trim13) is upregulated in response to inducers of type I IFN and that TRIM13 interacts with both MDA5 and RIG-I in vitro. Through the use of multiple in vitro and in vivo model systems, we show that TRIM13 is a negative regulator of MDA5-mediated type I IFN production and may also impact RIG-I-mediated type I IFN production by enhancing RIG-I activity. This places TRIM13 at a key junction within the viral response pathway and identifies it as one of the few known modulators of MDA5 activity.


Subject(s)
Cardiovirus Infections/enzymology , DEAD-box RNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Down-Regulation , Encephalomyocarditis virus/physiology , Interferon-alpha/metabolism , Interferon-beta/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cardiovirus Infections/genetics , Cardiovirus Infections/metabolism , Cardiovirus Infections/virology , DEAD-box RNA Helicases/genetics , DNA-Binding Proteins/genetics , Female , Fibroblasts/enzymology , Fibroblasts/metabolism , Fibroblasts/virology , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon-Induced Helicase, IFIH1 , Interferon-alpha/genetics , Interferon-beta/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics
4.
Stem Cells ; 31(11): 2432-42, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23897760

ABSTRACT

Human embryonic stem cells (hESCs) are considered a potential alternative to cadaveric islets as a source of transplantable cells for treating patients with diabetes. We previously described a differentiation protocol to generate pancreatic progenitor cells from hESCs, composed of mainly pancreatic endoderm (PDX1/NKX6.1-positive), endocrine precursors (NKX2.2/synaptophysin-positive, hormone/NKX6.1-negative), and polyhormonal cells (insulin/glucagon-positive, NKX6.1-negative). However, the relative contributions of NKX6.1-negative versus NKX6.1-positive cell fractions to the maturation of functional ß-cells remained unclear. To address this question, we generated two distinct pancreatic progenitor cell populations using modified differentiation protocols. Prior to transplant, both populations contained a high proportion of PDX1-expressing cells (~85%-90%) but were distinguished by their relatively high (~80%) or low (~25%) expression of NKX6.1. NKX6.1-high and NKX6.1-low progenitor populations were transplanted subcutaneously within macroencapsulation devices into diabetic mice. Mice transplanted with NKX6.1-low cells remained hyperglycemic throughout the 5-month post-transplant period whereas diabetes was reversed in NKX6.1-high recipients within 3 months. Fasting human C-peptide levels were similar between groups throughout the study, but only NKX6.1-high grafts displayed robust meal-, glucose- and arginine-responsive insulin secretion as early as 3 months post-transplant. NKX6.1-low recipients displayed elevated fasting glucagon levels. Theracyte devices from both groups contained almost exclusively pancreatic endocrine tissue, but NKX6.1-high grafts contained a greater proportion of insulin-positive and somatostatin-positive cells, whereas NKX6.1-low grafts contained mainly glucagon-expressing cells. Insulin-positive cells in NKX6.1-high, but not NKX6.1-low grafts expressed nuclear MAFA. Collectively, this study demonstrates that a pancreatic endoderm-enriched population can mature into highly functional ß-cells with only a minor contribution from the endocrine subpopulation.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Homeodomain Proteins/biosynthesis , Insulin-Secreting Cells/cytology , Pancreas/cytology , Animals , Cell Differentiation/physiology , Embryonic Stem Cells/transplantation , Endoderm/cytology , Endoderm/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Humans , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, SCID , Nuclear Proteins , Pancreas/metabolism , Transcription Factors
5.
Diabetologia ; 56(9): 1987-98, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23771205

ABSTRACT

AIMS/HYPOTHESIS: Islet transplantation is a promising cell therapy for patients with diabetes, but it is currently limited by the reliance upon cadaveric donor tissue. We previously demonstrated that human embryonic stem cell (hESC)-derived pancreatic progenitor cells matured under the kidney capsule in a mouse model of diabetes into glucose-responsive insulin-secreting cells capable of reversing diabetes. However, the formation of cells resembling bone and cartilage was a major limitation of that study. Therefore, we developed an improved differentiation protocol that aimed to prevent the formation of off-target mesoderm tissue following transplantation. We also examined how variation within the complex host environment influenced the development of pancreatic progenitors in vivo. METHODS: The hESCs were differentiated for 14 days into pancreatic progenitor cells and transplanted either under the kidney capsule or within Theracyte (TheraCyte, Laguna Hills, CA, USA) devices into diabetic mice. RESULTS: Our revised differentiation protocol successfully eliminated the formation of non-endodermal cell populations in 99% of transplanted mice and generated grafts containing >80% endocrine cells. Progenitor cells developed efficiently into pancreatic endocrine tissue within macroencapsulation devices, despite lacking direct contact with the host environment, and reversed diabetes within 3 months. The preparation of cell aggregates pre-transplant was critical for the formation of insulin-producing cells in vivo and endocrine cell development was accelerated within a diabetic host environment compared with healthy mice. Neither insulin nor exendin-4 therapy post-transplant affected the maturation of macroencapsulated cells. CONCLUSIONS/INTERPRETATION: Efficient differentiation of hESC-derived pancreatic endocrine cells can occur in a macroencapsulation device, yielding glucose-responsive insulin-producing cells capable of reversing diabetes.


Subject(s)
Embryonic Stem Cells/cytology , Insulin-Secreting Cells/cytology , Pancreas/cytology , Stem Cells/cytology , Animals , Cell Line , Embryonic Stem Cells/transplantation , Exenatide , Humans , Immunohistochemistry , Male , Mice , Mice, SCID , Peptides/pharmacology , Venoms/pharmacology
6.
Immunity ; 38(4): 681-93, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23562159

ABSTRACT

How innate lymphoid cells (ILCs) in the thymus and gut become specialized effectors is unclear. The prototypic innate-like γδ T cells (Tγδ17) are a major source of interleukin-17 (IL-17). We demonstrate that Tγδ17 cells are programmed by a gene regulatory network consisting of a quartet of high-mobility group (HMG) box transcription factors, SOX4, SOX13, TCF1, and LEF1, and not by conventional TCR signaling. SOX4 and SOX13 directly regulated the two requisite Tγδ17 cell-specific genes, Rorc and Blk, whereas TCF1 and LEF1 countered the SOX proteins and induced genes of alternate effector subsets. The T cell lineage specification factor TCF1 was also indispensable for the generation of IL-22 producing gut NKp46(+) ILCs and restrained cytokine production by lymphoid tissue inducer-like effectors. These results indicate that similar gene network architecture programs innate sources of IL-17, independent of anatomical origins.


Subject(s)
High Mobility Group Proteins/metabolism , Interleukin-17/biosynthesis , Intestines/immunology , Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Animals , Antigens, Ly/metabolism , Autoantigens/genetics , Autoantigens/metabolism , Cell Differentiation/genetics , Cells, Cultured , Gene Regulatory Networks/immunology , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , High Mobility Group Proteins/genetics , Immunity, Innate/genetics , Interleukin-17/genetics , Interleukins/immunology , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Natural Cytotoxicity Triggering Receptor 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Signal Transduction/immunology , Transcriptional Activation/immunology , Interleukin-22
7.
J Immunol ; 190(6): 2659-69, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23378428

ABSTRACT

The Tec family tyrosine kinase, Itk, regulates signaling downstream of the TCR. The absence of Itk in CD4(+) T cells results in impaired Th2 responses along with defects in maturation, cytokine production, and survival of iNKT cells. Paradoxically, Itk(-/-) mice have spontaneously elevated serum IgE levels, resulting from an expansion of the Vγ1.1(+)Vδ6.3(+) subset of γδ T cells, known as γδ NKT cells. Comparisons between γδ NKT cells and αß iNKT cells showed convergence in the pattern of cell surface marker expression, cytokine profiles, and gene expression, suggesting that these two subsets of NKT cells undergo similar differentiation programs. Hepatic γδ NKT cells have an invariant TCR and are derived predominantly from fetal progenitors that expand in the thymus during the first weeks of life. The adult thymus contains these invariant γδ NKT cells plus a heterogeneous population of Vγ1.1(+)Vδ6.3(+) T cells with diverse CDR3 sequences. This latter population, normally excluded from the liver, escapes the thymus and homes to the liver when Itk is absent. In addition, Itk(-/-) γδ NKT cells persistently express high levels of Zbtb16 (PLZF) and Il4, genes that are normally downregulated in the most mature subsets of NKT cells. These data indicate that Itk signaling is required to prevent the expansion of γδ NKT cells in the adult thymus, to block their emigration, and to promote terminal NKT cell maturation.


Subject(s)
Cell Differentiation/immunology , Cellular Senescence/immunology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Protein-Tyrosine Kinases/physiology , Receptors, Antigen, T-Cell, gamma-delta/biosynthesis , Thymus Gland/enzymology , Thymus Gland/immunology , Animals , Cell Migration Inhibition/immunology , Cell Movement/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/cytology , Thymus Gland/cytology
8.
Diabetes ; 61(8): 2016-29, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22740171

ABSTRACT

Diabetes is a chronic debilitating disease that results from insufficient production of insulin from pancreatic ß-cells. Islet cell replacement can effectively treat diabetes but is currently severely limited by the reliance upon cadaveric donor tissue. We have developed a protocol to efficiently differentiate commercially available human embryonic stem cells (hESCs) in vitro into a highly enriched PDX1+ pancreatic progenitor cell population that further develops in vivo to mature pancreatic endocrine cells. Immature pancreatic precursor cells were transplanted into immunodeficient mice with streptozotocin-induced diabetes, and glycemia was initially controlled with exogenous insulin. As graft-derived insulin levels increased over time, diabetic mice were weaned from exogenous insulin and human C-peptide secretion was eventually regulated by meal and glucose challenges. Similar differentiation of pancreatic precursor cells was observed after transplant in immunodeficient rats. Throughout the in vivo maturation period hESC-derived endocrine cells exhibited gene and protein expression profiles that were remarkably similar to the developing human fetal pancreas. Our findings support the feasibility of using differentiated hESCs as an alternative to cadaveric islets for treating patients with diabetes.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Insulin-Secreting Cells/cytology , Pancreas/cytology , Animals , Cell Line , Diabetes Mellitus, Experimental/therapy , Homeodomain Proteins/biosynthesis , Humans , Insulin/therapeutic use , Insulin-Secreting Cells/metabolism , Male , Mice , Pancreas/embryology , Proprotein Convertases/biosynthesis , Rats , Stem Cells/cytology , Trans-Activators/biosynthesis
9.
Nat Immunol ; 13(5): 511-8, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22473038

ABSTRACT

Innate γδ T cells function in the early phase of immune responses. Although innate γδ T cells have often been studied as one homogenous population, they can be functionally classified into effector subsets on the basis of the production of signature cytokines, analogous to adaptive helper T cell subsets. However, unlike the function of adaptive T cells, γδ effector T cell function correlates with genomically encoded T cell antigen receptor (TCR) chains, which suggests that clonal TCR selection is not the main determinant of the differentiation of γδ effector cells. A high-resolution transcriptome analysis of all emergent γδ thymocyte subsets segregated on the basis of use of the TCR γ-chain or δ-chain indicated the existence of three separate subtypes of γδ effector cells in the thymus. The immature γδ subsets were distinguished by unique transcription-factor modules that program effector function.


Subject(s)
Cell Differentiation/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , Transcriptome/immunology , Age Factors , Animals , CD24 Antigen/immunology , CD24 Antigen/metabolism , Cell Differentiation/genetics , Cell Lineage/immunology , Fetus/cytology , Fetus/immunology , Flow Cytometry , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Models, Immunological , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/metabolism , Principal Component Analysis , Receptors, Antigen, T-Cell, gamma-delta/classification , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism , Transcriptome/genetics
10.
J Immunol ; 185(12): 7156-60, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21068400

ABSTRACT

Various innate-like T cell subsets preferentially reside in specific epithelial tissues as the first line of defense. However, mechanisms regulating their tissue-specific development are poorly understood. Using the prototypical skin intraepithelial γδT cells (sIELs) as a model, we show in this study that a TCR-mediated selection plays an important role in promoting acquisition of a specific skin-homing property by fetal thymic sIEL precursors for their epidermal location, and the skin-homing potential is intrinsically programmed even before the selection. In addition, once localized in the skin, the sIEL precursors develop into sIELs without the requirement of further TCR-ligand interaction. These studies reveal that development of the tissue-specific lymphocytes is a hard-wired process that targets them to specific tissues for proper functions.


Subject(s)
Immunity, Innate/physiology , Models, Immunological , Receptors, Antigen, T-Cell, gamma-delta , Skin/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Animals , Mice , Mice, Inbred BALB C , Mice, Knockout , Organ Specificity/genetics , Organ Specificity/immunology , Skin/cytology , T-Lymphocytes/cytology , Thymus Gland/cytology
11.
Semin Immunol ; 22(4): 222-7, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20451409

ABSTRACT

The mechanism of T cell precursor commitment to the gammadelta or alphabeta T cell lineage remains unclear. While TCR signal strength has emerged as a key factor in lineage commitment based on TCR transgenic models, the entire TCR repertoire may not possess the same discriminatory power. A counterbalance to the TCR as the lineage determinant is the pre-existing heterogeneity in gene expression among precursors, which suggests that single precursors are unlikely to respond homogeneously to a given instructive signal.


Subject(s)
Cell Lineage , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Humans , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Signal Transduction , T-Lymphocytes/metabolism
13.
J Neuroimmunol ; 190(1-2): 112-20, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17919740

ABSTRACT

Little is known about antibody production within the central nervous system, called intrathecal antibody production (ITAbP). Mice infected with Theiler's murine encephalomyocarditis virus (TMEV) develop an immune-mediated demyelinating disease(TMEV-IDD), characterized by progressive weakness associated with robust ITAbP, CNS inflammation and demyelination. TMEV-IDD represents an excellent model of human multiple sclerosis (MS), especially its progressive disability. The mechanism of the weakness in this disease is unknown but there is considerable evidence that it is immune-mediated. Our hypothesis was that the disability in the model is induced by robust ITAbP by antibody-secreting cells(ASCs) in the CNS. We further hypothesized that these ASCs are predominantly CD138+ plasma cells, driven by the persistence of virus at high levels in the central nervous system (CNS) as well as high levels of B-cell activating factor (BAFF). In order to test these hypotheses we infected mice with TMEV, and correlated measures of ITAbP with disability. Measures of ITAbP were ELISpot and IgG gene expression, while disability was tested by Rotarod. We found that disability was highly correlated with ITAbP, assayed by number of CNS ASCs as well as amount of IgG mRNA. CNS ASCs were primarily CD138+, and produced high levels of IgG enriched for reactivity to TMEV. There were high levels of BAFF production in the CNS, but these levels were only minimally higher in infected mice than in uninfected controls. TMEV and IgG RNA were distributed throughout the spinal cord. These data are the first demonstrating that ITAbP is highly correlated with disability in TMEV infection, an excellent model of human MS. Our data raise the possibility that ITAbP contributes significantly to disability, both in this model and in MS.


Subject(s)
Antibodies/immunology , Central Nervous System/cytology , Central Nervous System/immunology , Multiple Sclerosis/immunology , Plasma Cells/immunology , Plasma Cells/metabolism , Animals , B-Cell Activating Factor/immunology , Cardiovirus Infections/immunology , Cardiovirus Infections/physiopathology , Cardiovirus Infections/virology , Central Nervous System/physiopathology , Disease Models, Animal , Disease Progression , Female , Immunoglobulin G/immunology , Mice , Multiple Sclerosis/physiopathology , Multiple Sclerosis/virology , Phenotype , Plasma Cells/virology , RNA, Messenger/metabolism , Spinal Cord/cytology , Spinal Cord/immunology , Spinal Cord/physiopathology , Syndecan-1/immunology , Theilovirus/immunology
14.
J Immunol ; 178(8): 5076-85, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17404290

ABSTRACT

IFN-beta effectively controls clinical exacerbations and magnetic resonance imaging activity in most multiple sclerosis patients. However, its mechanism of action has not been yet fully elucidated. In this study we used DNA microarrays to analyze the longitudinal transcriptional profile of blood cells within a week of IFN-beta administration. Using differential expression and gene ontology analyses we found evidence of a general decrease in the cellular activity of T lymphocytes resembling the endogenous antiviral response of IFNs. In contrast, most of the differentially expressed genes (DEGs) from untreated individuals were involved in cellular physiological processes. We then used mutual information (MI) to build networks of coregulated genes in both treated and untreated individuals. Interestingly, the connectivity distribution (k) of networks generated with high MI values displayed scale-free properties. Conversely, the observed k for networks generated with suboptimal MI values approximated a Poisson distribution, suggesting that MI captures biologically relevant interactions. Gene networks from individuals treated with IFN-beta revealed a tight core of immune- and apoptosis-related genes associated with higher values of MI. In contrast, networks obtained from untreated individuals primarily reflected cellular housekeeping functions. Finally, we trained a neural network to reverse engineer the directionality of the main interactions observed at the biological process level. This is the first study that incorporates network analysis to investigate gene regulation in response to a therapeutic drug in humans. Implications of this method in the creation of personalized models of response to therapy are discussed.


Subject(s)
Gene Regulatory Networks , Interferon-beta/pharmacology , Transcription, Genetic/drug effects , Gene Expression Profiling , Humans
15.
J Neuroimmunol ; 185(1-2): 57-63, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17343922

ABSTRACT

Although the central nervous system (CNS) is thought to be immunoprivileged, under special circumstances it can produce antibody. Antibody production within the CNS, called intrathecal antibody production (ITAbP), is a prominent feature of neurological infections and inflammatory diseases, and is thought to possibly contribute to disease in multiple sclerosis (MS), but it has not been extensively studied. We investigated ITAbP in a viral model of MS. ELISpot, real-time RT-PCR for IgG mRNA in CNS tissue, and CSF analysis were used to assess ITAbP in two types of SJL mice infected with one of two strains of Theiler's murine encephalomyelitis virus (TMEV). The amplitude of ITAbP increased during the first 4 months of infection. TMEV viral load remained high during the course of the infection, which likely was the main stimulus for ITAbP, since within samples of infected CNS tissues, levels of IgG gene expression were highly correlated with viral RNA levels, and a large percentage of intrathecally produced antibody was directed against TMEV. This study provides the first extensive analysis of ITAbP in TMEV infection, and demonstrates that, in this animal model of MS, antibody production within the CNS is likely driven by the presence of the causative pathogen.


Subject(s)
Antibodies, Viral/cerebrospinal fluid , Central Nervous System/immunology , Immunoglobulin G/cerebrospinal fluid , Maus Elberfeld virus/immunology , Multiple Sclerosis/immunology , Animals , Antibodies, Viral/biosynthesis , Antibody Formation , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Immunoglobulin G/biosynthesis , Mice , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
16.
Curr Opin Immunol ; 19(2): 169-75, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17291740

ABSTRACT

The divergence of alphabeta and gammadelta T cells from a common precursor in the thymus is regulated by multiple cell-intrinsic and cell-extrinsic factors, most of which are not well defined. Recent studies have provided crucial data regarding the precise timing of lineage commitment and some clarification on the extent of the involvement of Notch and T-cell receptor signaling in this process. Combined with new insights into the differential regulation of molecular pathways active in alphabeta and gammadelta precursors, these data have led to the generation of a revised model of lineage commitment.


Subject(s)
Lymphopoiesis , Multipotent Stem Cells/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/immunology , Thymus Gland/immunology , Animals , Cell Lineage/genetics , Lymphopoiesis/genetics , Mice , Multipotent Stem Cells/chemistry , Receptors, Antigen, T-Cell, alpha-beta/analysis , Receptors, Antigen, T-Cell, gamma-delta/analysis , Receptors, Notch/physiology , Thymus Gland/cytology
17.
Science ; 315(5809): 230-3, 2007 Jan 12.
Article in English | MEDLINE | ID: mdl-17218525

ABSTRACT

alphabeta and gammadelta T cells originate from a common, multipotential precursor population in the thymus, but the molecular mechanisms regulating this lineage-fate decision are unknown. We have identified Sox13 as a gammadelta-specific gene in the immune system. Using Sox13 transgenic mice, we showed that this transcription factor promotes gammadelta T cell development while opposing alphabeta T cell differentiation. Conversely, mice deficient in Sox13 expression exhibited impaired development of gammadelta T cells but not alphabeta T cells. One mechanism of SOX13 function is the inhibition of signaling by the developmentally important Wnt/T cell factor (TCF) pathway. Our data thus reveal a dominant pathway regulating the developmental fate of these two lineages of T lymphocytes.


Subject(s)
Autoantigens/metabolism , High Mobility Group Proteins/metabolism , Lymphopoiesis , Receptors, Antigen, T-Cell, alpha-beta/analysis , Receptors, Antigen, T-Cell, gamma-delta/analysis , T-Lymphocyte Subsets/cytology , Animals , Autoantigens/genetics , CD4 Antigens/genetics , Cell Line , Cell Lineage , Cell Proliferation , Embryonic Development , Gene Expression Profiling , Gene Expression Regulation , Gene Rearrangement, T-Lymphocyte , High Mobility Group Proteins/genetics , Humans , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell, gamma-delta/genetics , Signal Transduction , T Cell Transcription Factor 1/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Wnt Proteins/metabolism
18.
Neurology ; 66(3): 444-6, 2006 Feb 14.
Article in English | MEDLINE | ID: mdl-16476953

ABSTRACT

Some interferon beta (IFNbeta)-treated patients with multiple sclerosis develop antibody-mediated decreased bioactivity with resultant loss of therapeutic effect. The authors developed real-time multiplex reverse transcriptase PCR to measure expression of three IFNbeta-inducible genes to directly assess IFNbeta bioactivity in patients with neutralizing antibodies (NAbs). The three genes responded in tandem. Correlation of NAb level with bioactivity at low/moderate NAb levels was poor, indicating that for such patients, direct measurement of IFNbeta bioactivity is most reliable.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Gene Expression Regulation/drug effects , Interferon-beta/therapeutic use , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Neutralization Tests , 2',5'-Oligoadenylate Synthetase/genetics , Case-Control Studies , GTP-Binding Proteins/genetics , Gene Expression , Humans , Multiple Sclerosis/drug therapy , Myxovirus Resistance Proteins , Oxidoreductases Acting on CH-CH Group Donors , Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
19.
J Neuroimmunol ; 173(1-2): 56-68, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16387369

ABSTRACT

Intrathecal antibody (ITAb) production is a common feature of neurological diseases, yet very little is known about its mechanisms. Because ITAb is prominent in human Lyme neuroborreliosis (LNB), in the present study we established a mouse model of LNB to study ITAb production. We injected different strains of Borrelia burgdorferi into a variety of mouse strains by the intracerebral (i.c.) route to develop the model. Spirochetal infection and ITAb production were identified by complementary methods. This study demonstrates that the mouse model of LNB can be utilized to test hypotheses related to the mechanisms of ITAb production.


Subject(s)
Antibodies, Bacterial/cerebrospinal fluid , Brain Diseases/immunology , Immunoglobulin G/cerebrospinal fluid , Lyme Neuroborreliosis/immunology , Animals , Antibody Formation , Borrelia burgdorferi/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunoblotting , Injections, Intraventricular , Male , Mice , Reverse Transcriptase Polymerase Chain Reaction
20.
J Neuroimmunol ; 166(1-2): 180-8, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16005084

ABSTRACT

Many multiple sclerosis (MS) patients treated with IFNbeta develop anti-IFNbeta antibodies, which can interfere with the bioactivity of the injected cytokine, i.e., antibody-mediated decreased bioactivity (ADB). The precise levels of anti-IFNbeta antibodies inducing decreased bioactivity is unknown. We repeatedly used a bioactivity measure, gene expression of MxA or GEM, and correlated bioactivity with measures of binding and neutralizing antibodies. The binding antibody assay was a capture ELISA, and the neutralizing antibody (NAb) assay was a cytopathic effect (CPE) assay. 27% (17/64) of patients repeatedly sampled developed critical ADB. Bioactivity as determined by GEM correlated negatively with NAb titer, and bioactivity that had been lost with the development of NAbs returned if NAb levels diminished. These data reveal that the GEM assay is a useful adjunct in the management of MS patients treated with IFNbeta, and that lost bioactivity returns when anti-IFNbeta antibody levels diminish.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antibodies/immunology , Interferon-beta/immunology , Interferon-beta/therapeutic use , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Adult , Antibodies/blood , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , False Negative Reactions , Female , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Gene Expression , Humans , Interferon-beta/metabolism , Male , Myxovirus Resistance Proteins , RNA, Messenger/blood , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...