Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
mBio ; 7(5)2016 09 13.
Article in English | MEDLINE | ID: mdl-27624124

ABSTRACT

UNLABELLED: Apical membrane antigen 1 (AMA1) is a receptor protein on the surface of Toxoplasma gondii that plays a critical role in host cell invasion. The ligand to which T gondii AMA1 (TgAMA1) binds, TgRON2, is secreted into the host cell membrane by the parasite during the early stages of invasion. The TgAMA1-TgRON2 complex forms the core of the "moving junction," a ring-shaped zone of tight contact between the parasite and host cell membranes, through which the parasite pushes itself during invasion. Paradoxically, the parasite also expresses rhomboid proteases that constitutively cleave the TgAMA1 transmembrane domain. How can TgAMA1 function effectively in host cell binding if its extracellular domain is constantly shed from the parasite surface? We show here that when TgAMA1 binds the domain 3 (D3) peptide of TgRON2, its susceptibility to cleavage by rhomboid protease(s) is greatly reduced. This likely serves to maintain parasite-host cell binding at the moving junction, a hypothesis supported by data showing that parasites expressing a hypercleavable version of TgAMA1 invade less efficiently than wild-type parasites do. Treatment of parasites with the D3 peptide was also found to reduce phosphorylation of S527 on the cytoplasmic tail of TgAMA1, and parasites expressing a phosphomimetic S527D allele of TgAMA1 showed an invasion defect. Taken together, these data suggest that TgAMA1-TgRON2 interaction at the moving junction protects TgAMA1 molecules that are actively engaged in host cell penetration from rhomboid-mediated cleavage and generates an outside-in signal that leads to dephosphorylation of the TgAMA1 cytosolic tail. Both of these effects are required for maximally efficient host cell invasion. IMPORTANCE: Nearly one-third of the world's population is infected with the protozoan parasite Toxoplasma gondii, which causes life-threatening disease in neonates and immunocompromised individuals. T. gondii is a member of the phylum Apicomplexa, which includes many other parasites of veterinary and medical importance, such as those that cause coccidiosis, babesiosis, and malaria. Apicomplexan parasites grow within their hosts through repeated cycles of host cell invasion, parasite replication, and host cell lysis. Parasites that cannot invade host cells cannot survive or cause disease. AMA1 is a highly conserved protein on the surface of apicomplexan parasites that is known to be important for invasion, and the work presented here reveals new and unexpected insights into AMA1 function. A more complete understanding of the role of AMA1 in invasion may ultimately contribute to the development of new chemotherapeutics designed to disrupt AMA1 function and invasion-related signaling in this important group of human pathogens.


Subject(s)
Antigens, Protozoan/metabolism , Peptide Hydrolases/metabolism , Protein Processing, Post-Translational , Proteolysis , Protozoan Proteins/metabolism , Toxoplasma/physiology , Cells, Cultured , Fibroblasts/parasitology , Humans , Phosphorylation , Protein Binding
2.
Stem Cells ; 34(7): 1765-75, 2016 07.
Article in English | MEDLINE | ID: mdl-26946228

ABSTRACT

Human embryonic stem cells (hESCs) have an abbreviated G1 phase of the cell cycle that allows rapid proliferation and maintenance of pluripotency. Lengthening of G1 corresponds to loss of pluripotency during differentiation. However, precise mechanisms that link alterations in the cell cycle and early differentiation remain to be defined. We investigated initial stages of mesendodermal lineage commitment in hESCs, and observed a cell cycle pause. Transcriptome profiling identified several genes with known roles in regulation of the G2/M transition that were differentially expressed early during lineage commitment. WEE1 kinase, which blocks entry into mitosis by phosphorylating CDK1 at Y15, was the most highly expressed of these genes. Inhibition of CDK1 phosphorylation by a specific inhibitor of WEE1 restored cell cycle progression by preventing the G2 pause. Directed differentiation of hESCs revealed that cells paused during commitment to the endo- and mesodermal, but not ectodermal, lineages. Functionally, WEE1 inhibition during meso- and endodermal differentiation selectively decreased expression of definitive endodermal markers SOX17 and FOXA2. Our findings identify a novel G2 cell cycle pause that is required for endodermal differentiation and provide important new mechanistic insights into early events of lineage commitment. Stem Cells 2016;34:1765-1775.


Subject(s)
Cell Cycle Checkpoints , Cell Differentiation , Cell Lineage , Embryonic Stem Cells/cytology , G2 Phase , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cluster Analysis , Embryonic Stem Cells/metabolism , Endoderm/cytology , Female , Gene Expression Profiling , Humans , Male , Mesoderm/cytology , Models, Biological , Nuclear Proteins/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Up-Regulation/genetics
3.
J Reprod Immunol ; 100(1): 66-75, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24080233

ABSTRACT

CD4+ CD25+ regulatory T cells (Tregs) strongly influence the early and late autoimmune responses to meiotic germ cell antigens (MGCA) and the gonadal immunopathology in vasectomized mice. This is supported by the published and recently acquired information presented here. Within 24h of unilateral vasectomy (uni-vx) the ipsilateral epididymis undergoes epithelial cell apoptosis followed by necrosis, severe inflammation, and granuloma formation. Unexpectedly, vasectomy alone induced MGCA-specific tolerance. In contrast, uni-vx plus simultaneous Treg depletion resulted in MGCA-specific autoimmune response and bilateral autoimmune orchitis. Both tolerance and autoimmunity were strictly linked to the early epididymal injury. We now discovered that testicular autoimmunity in uni-vx mice did not occur when Treg depletion was delayed by one week. Remarkably, this delayed Treg depletion also prevented tolerance induction. Therefore, tolerance depends on a rapid de novo Treg response to MGCA exposed after vasectomy. Moreover, tolerance was blunted in mice genetically deficient in PD-1 ligand, suggesting the involvement of induced Treg. We conclude that pre-existing natural Treg prevents post-vasectomy autoimmunity, whereas vasectomy-induced Treg maintains post-vasectomy tolerance. We further discovered that vasectomized mice were still resistant to autoimmune orchitis induction for at least 12-16 months; thus, tolerance is long-lasting. Although significant sperm autoantibodies of low titers became detectable in uni-vx mice at 7 months, the antibody titers fluctuated over time, suggesting a dynamic "balance" between the autoimmune and tolerance states. Finally, we observed severe epididymal fibrosis and hypo-spermatogenesis at 12 months after uni-vx: findings of highly critical clinical significance.


Subject(s)
Epididymis/pathology , Orchitis/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Vasectomy , Animals , Autoantibodies/blood , Autoimmunity/genetics , CD4 Antigens/metabolism , Fibrosis/etiology , Humans , Immune Tolerance/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Depletion , Male , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Orchitis/etiology , Programmed Cell Death 1 Receptor/genetics , Vasectomy/adverse effects
4.
PLoS One ; 8(7): e62743, 2013.
Article in English | MEDLINE | ID: mdl-23894272

ABSTRACT

Histamine H(3) receptor (Hrh3/H(3)R) is primarily expressed by neurons in the central nervous system (CNS) where it functions as a presynaptic inhibitory autoreceptor and heteroreceptor. Previously, we identified an H(3)R-mediated central component in susceptibility to experimental allergic encephalomyelitis (EAE), the principal autoimmune model of multiple sclerosis (MS), related to neurogenic control of blood brain barrier permeability and peripheral T cell effector responses. Furthermore, we identified Hrh3 as a positional candidate for the EAE susceptibility locus Eae8. Here, we characterize Hrh3 polymorphisms between EAE-susceptible and resistant SJL and B10.S mice, respectively, and show that Hrh3 isoform expression in the CNS is differentially regulated by acute peripheral inflammatory stimuli in an allele-specific fashion. Next, we show that Hrh3 is not expressed in any subpopulations of the immune compartment, and that secondary lymphoid tissue is anatomically poised to be regulated by central H(3)R signaling. Accordingly, using transcriptome analysis, we show that, inflammatory stimuli elicit unique transcriptional profiles in the lymph nodes of H(3)RKO mice compared to WT mice, which is indicative of negative regulation of peripheral immune responses by central H(3)R signaling. These results further support a functional link between the neurogenic control of T cell responses and susceptibility to CNS autoimmune disease coincident with acute and/or chronic peripheral inflammation. Pharmacological targeting of H(3)R may therefore be useful in preventing the development and formation of new lesions in MS, thereby limiting disease progression.


Subject(s)
Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Genetic Predisposition to Disease/genetics , Receptors, Histamine H3/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Amino Acid Sequence , Animals , Central Nervous System/immunology , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression Regulation , Hematopoiesis/genetics , Hematopoiesis/immunology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Intracellular Space/metabolism , Lymph Nodes/immunology , Male , Mice , Molecular Sequence Data , Polymorphism, Single Nucleotide , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Structure, Tertiary , Receptors, Histamine H3/chemistry
5.
J Immunol ; 191(2): 614-22, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23772030

ABSTRACT

Histamine (HA) is a key regulator of experimental allergic encephalomyelitis (EAE), the autoimmune model of multiple sclerosis. HA exerts its effects through four known G-protein-coupled receptors: H1, H2, H3, and H4 (histamine receptors; H(1-4)R). Using HR-deficient mice, our laboratory has demonstrated that H1R, H2R, H3R, and H4R play important roles in EAE pathogenesis, by regulating encephalitogenic T cell responses, cytokine production by APCs, blood-brain barrier permeability, and T regulatory cell activity, respectively. Histidine decarboxylase-deficient mice (HDCKO), which lack systemic HA, exhibit more severe EAE and increased Th1 effector cytokine production by splenocytes in response to myelin oligodendrocyte gp35-55. In an inverse approach, we tested the effect of depleting systemic canonical HA signaling on susceptibility to EAE by generating mice lacking all four known G-protein-coupled-HRs (H(1-4)RKO mice). In this article, we report that in contrast to HDCKO mice, H(1-4)RKO mice develop less severe EAE compared with wild-type animals. Furthermore, splenocytes from immunized H(1-4)RKO mice, compared with wild-type mice, produce a lower amount of Th1/Th17 effector cytokines. The opposing results seen between HDCKO and H1-4RKO mice suggest that HA may signal independently of H1-4R and support the existence of an alternative HAergic pathway in regulating EAE resistance. Understanding and exploiting this pathway has the potential to lead to new disease-modifying therapies in multiple sclerosis and other autoimmune and allergic diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Histamine/metabolism , Histidine Decarboxylase/genetics , Receptors, Histamine/genetics , Receptors, Histamine/metabolism , Animals , Antigen-Presenting Cells , Blood-Brain Barrier/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cells, Cultured , Cytokines/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/metabolism , Histidine Decarboxylase/deficiency , Histidine Decarboxylase/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/immunology , Myelin-Oligodendrocyte Glycoprotein/pharmacology , Peptide Fragments/pharmacology , Receptors, Histamine/deficiency , Signal Transduction
6.
FASEB J ; 27(5): 1874-86, 2013 May.
Article in English | MEDLINE | ID: mdl-23371066

ABSTRACT

The uterotropic response of the uterus to 17ß-estradiol (E2) is genetically controlled, with marked variation observed depending on the mouse strain studied. Previous genetic studies from our laboratory using inbred mice that are high [C57BL/6J (B6)] or low [C3H/HeJ (C3H)] responders to E2 led to the identification of quantitative trait (QT) loci associated with phenotypic variation in uterine growth and leukocyte infiltration. The mechanisms underlying differential responsiveness to E2, and the genes involved, are unknown. Therefore, we used a microarray approach to show association of distinct E2-regulated transcriptional signatures with genetically controlled high and low responses to E2 and their segregation in (C57BL/6J×C3H/HeJ) F1 hybrids. Among the 6664 E2-regulated transcripts, analysis of cellular functions of those that were strain specific indicated C3H-selective enrichment of apoptosis, consistent with a 7-fold increase in the apoptosis indicator CASP3, and a 2.4-fold decrease in the apoptosis inhibitor Naip1 (Birc1a) in C3H vs. B6 following treatment with E2. In addition, several differentially expressed transcripts reside within our previously identified QT loci, including the ERα-tethering factor Runx1, demonstrated to enhance E2-mediated transcript regulation. The level of RUNX1 in uterine epithelial cells was shown to be 3.5-fold greater in B6 compared to C3H. Our novel insights into the mechanisms underlying the genetic control of tissue sensitivity to estrogen have great potential to advance understanding of individualized effects in physiological and disease states.


Subject(s)
Caspase 3/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Neuronal Apoptosis-Inhibitory Protein/genetics , Transcription, Genetic/genetics , Uterus/physiology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Proliferation/drug effects , Core Binding Factor Alpha 2 Subunit/physiology , Epithelial Cells/metabolism , Female , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred Strains , Peroxidase/genetics , Protein Array Analysis , Quantitative Trait Loci/physiology , Transcription, Genetic/drug effects , Transcriptome , Uterus/drug effects , Uterus/growth & development
7.
Eur J Immunol ; 42(6): 1536-46, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22678907

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system in which histamine (HA) and its receptors have been implicated in disease pathogenesis. HA exerts its effects through four different G protein-coupled receptors designated H(1)-H(4). We previously examined the effects of traditional single HA receptor (HR) knockouts (KOs) in experimental allergic encephalomyelitis (EAE), the autoimmune model of MS. Our results revealed that H(1) R and H(2) R are propathogenic, while H(3) R and H(4) R are antipathogenic. This suggests that combinatorial targeting of HRs may be an effective disease-modifying therapy (DMT) in MS. To test this hypothesis, we generated H(1) H(2) RKO and H(3) H(4) RKO mice and studied them for susceptibility to EAE. Compared with wild-type (WT) mice, H(1) H(2) RKO mice developed a less severe clinical disease course, whereas the disease course of H(3) H(4) RKO mice was more severe. H(1) H(2) RKO mice also developed less neuropathology and disrupted blood brain barrier permeability compared with WT and H(3) H(4) RKO mice. Additionally, splenocytes from immunized H(1) H(2) RKO mice produced less interferon(IFN)-γ and interleukin(IL)-17. These findings support the concept that combined pharmacological targeting of HRs may be an appropriate ancillary DMT in MS and other immunopathologic diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/etiology , Multiple Sclerosis/etiology , Receptors, Histamine/physiology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Polarity , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/physiology , Receptors, Histamine H1/physiology , Receptors, Histamine H2/physiology , Receptors, Histamine H3/physiology , Receptors, Histamine H4
8.
PLoS Genet ; 8(12): e1003140, 2012.
Article in English | MEDLINE | ID: mdl-23300462

ABSTRACT

Experimental autoimmune orchitis (EAO), the principal model of non-infectious testicular inflammatory disease, can be induced in susceptible mouse strains by immunization with autologous testicular homogenate and appropriate adjuvants. As previously established, the genome of DBA/2J mice encodes genes that are capable of conferring dominant resistance to EAO, while the genome of BALB/cByJ mice does not and they are therefore susceptible to EAO. In a genome scan, we previously identified Orch3 as the major quantitative trait locus controlling dominant resistance to EAO and mapped it to chromosome 11. Here, by utilizing a forward genetic approach, we identified kinesin family member 1C (Kif1c) as a positional candidate for Orch3 and, using a transgenic approach, demonstrated that Kif1c is Orch3. Mechanistically, we showed that the resistant Kif1c(D2) allele leads to a reduced antigen-specific T cell proliferative response as a consequence of decreased MHC class II expression by antigen presenting cells, and that the L(578) → P(578) and S(1027) → P(1027) polymorphisms distinguishing the BALB/cByJ and DBA/2J alleles, respectively, can play a role in transcriptional regulation. These findings may provide mechanistic insight into how polymorphism in other kinesins such as KIF21B and KIF5A influence susceptibility and resistance to human autoimmune diseases.


Subject(s)
Disease Resistance/genetics , Genes, Dominant , Kinesins/genetics , Orchitis , Alleles , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Gene Expression , Genes, MHC Class II , Genetic Predisposition to Disease , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Orchitis/genetics , Orchitis/immunology , Quantitative Trait Loci/genetics , Testis/immunology
9.
J Immunol ; 188(2): 541-7, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22147765

ABSTRACT

Histamine is a biogenic amine that mediates multiple physiological processes, including immunomodulatory effects in allergic and inflammatory reactions, and also plays a key regulatory role in experimental allergic encephalomyelitis, the autoimmune model of multiple sclerosis. The pleiotropic effects of histamine are mediated by four G protein-coupled receptors, as follows: Hrh1/H(1)R, Hrh2/H(2)R, Hrh3/H(3)R, and Hrh4/H(4)R. H(4)R expression is primarily restricted to hematopoietic cells, and its role in autoimmune inflammatory demyelinating disease of the CNS has not been studied. In this study, we show that, compared with wild-type mice, animals with a disrupted Hrh4 (H(4)RKO) develop more severe myelin oligodendrocyte glycoprotein (MOG)(35\x{2013}55)-induced experimental allergic encephalomyelitis. Mechanistically, we also show that H(4)R plays a role in determining the frequency of T regulatory (T(R)) cells in secondary lymphoid tissues, and regulates T(R) cell chemotaxis and suppressor activity. Moreover, the lack of H(4)R leads to an impairment of an anti-inflammatory response due to fewer T(R) cells in the CNS during the acute phase of the disease and an increase in the proportion of Th17 cells.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Receptors, G-Protein-Coupled/physiology , Receptors, Histamine/physiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Animals , Blood-Brain Barrier/immunology , CD4 Lymphocyte Count , Cell Membrane Permeability/genetics , Cell Membrane Permeability/immunology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/genetics , Glycoproteins/administration & dosage , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein , Neurons/immunology , Neurons/pathology , Peptide Fragments/administration & dosage , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Receptors, Histamine/deficiency , Receptors, Histamine/genetics , Receptors, Histamine H4 , Severity of Illness Index , T-Lymphocytes, Regulatory/metabolism
10.
Ann Neurol ; 70(6): 887-96, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22190363

ABSTRACT

OBJECTIVE: The major histocompatibility complex (MHC) is the primary genetic contributor to multiple sclerosis (MS) and experimental allergic encephalomyelitis (EAE), but multiple additional interacting loci are required for genetic susceptibility. The identity of most of these non-MHC genes is unknown. In this report, we identify genes within evolutionarily conserved genetic pathways leading to MS and EAE. METHODS: To identify non-MHC binary and quantitative trait loci (BTL/QTL) important in the pathogenesis of EAE, we generated phenotype-selected congenic mice using EAE-resistant B10.S and EAE-susceptible SJL mice. We hypothesized that genes linked to EAE BTL/QTL and MS-GWAS can be identified if they belong to common evolutionarily conserved pathways, which can be identified with a bioinformatic approach using Ingenuity software. RESULTS: Many known BTL/QTL were retained and linked to susceptibility during phenotype selection, the most significant being a region on chromosome 17 distal to H2 (Eae5). We show in pathway analysis that T helper (T(H))-cell differentiation genes are critical for both diseases. Bioinformatic analyses predicted that Eae5 is important in CD4 T-effector and/or Foxp3(+) T-regulatory cells (Tregs), and we found that B10.S-Eae5(SJL) congenic mice have significantly greater numbers of lymph node CD4 and Tregs than B10.S mice. INTERPRETATION: These results support the polygenic model of MS/EAE, whereby MHC and multiple minor loci are required for full susceptibility, and confirm a critical genetic dependence on CD4 T(H)-cell differentiation and function in the pathogenesis of both diseases.


Subject(s)
Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Major Histocompatibility Complex/genetics , Multiple Sclerosis/genetics , T-Lymphocytes, Helper-Inducer/pathology , Animals , CD4-Positive T-Lymphocytes/physiology , Computational Biology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/etiology , Flow Cytometry , Freund's Adjuvant/adverse effects , Genetic Linkage , Genome-Wide Association Study , Mice , Mice, Congenic , Phenotype , Quantitative Trait Loci/genetics , Statistics, Nonparametric
11.
IUBMB Life ; 63(10): 940-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21905200

ABSTRACT

T cell (TC) activation requires the coordinated signaling of the T cell receptor (TCR) and coreceptor molecules, allowing TCs to respond to lower degrees of TCR occupancy. Coreceptor molecules set the threshold for TC activation by controlling different regulatory signaling loops. The Cbl family members prevent undesired activation of T cells by regulating TCR signals. In this report, we show that TC prestimulation by the CD43 coreceptor molecule before TCR engagement inhibits TCR-dependent c-Cbl tyrosine phosphorylation, c-Cbl interaction with the adapter molecule Crk-L and promotes Cbl-b degradation in a PKCθ-dependent manner. Consequently, the prolonged tyrosine phosphorylation and delayed degradation of ZAP-70 and of the ζ chain lead to enhanced mitogen-activated protein kinase activation and robust TC response. These data indicates that CD43-mediated signals lower the threshold for TC activation by restricting the c-Cbl and Cbl-b inhibitory effects on TCR signaling. In addition to the strength and duration of intracellular signals, our data underscore temporality with which certain molecules are engaged as yet another mechanism to fine tune TC signal quality, and ultimately immune function.


Subject(s)
Leukosialin/metabolism , Lymphocyte Activation/physiology , Proto-Oncogene Proteins c-cbl/metabolism , Signal Transduction/physiology , T-Lymphocytes/physiology , Animals , Cell Proliferation , Humans , Immunoblotting , Immunoprecipitation , Jurkat Cells , Lymphocyte Activation/immunology , Mice , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology
12.
Blood ; 118(12): 3290-300, 2011 Sep 22.
Article in English | MEDLINE | ID: mdl-21791428

ABSTRACT

Although several transcription factors have been shown to be critical for the induction and maintenance of IL-17 expression by CD4 Th cells, less is known about the role of nontranscriptional mechanisms. Here we show that the p38 MAPK signaling pathway is essential for in vitro and in vivo IL-17 production by regulating IL-17 synthesis in CD4 T cells through the activation of the eukaryotic translation initiation factor 4E/MAPK-interacting kinase (eIF-4E/MNK) pathway. We also show that p38 MAPK activation is required for the development and progression of both chronic and relapsing-remitting forms of experimental allergic encephalomyelitis (EAE), the principal autoimmune model of multiple sclerosis. Furthermore, we show that regulation of p38 MAPK activity specifically in T cells is sufficient to modulate EAE severity. Thus, mechanisms other than the regulation of gene expression also contribute to Th17 cell effector functions and, potentially, to the pathogenesis of other Th17 cell-mediated diseases.


Subject(s)
Autoimmunity , Encephalomyelitis, Autoimmune, Experimental/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Interleukin-17/biosynthesis , Lymphocyte Activation/drug effects , Signal Transduction/drug effects , Th17 Cells/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cell Proliferation , Cell Separation , Cells, Cultured , Chronic Disease , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/immunology , Female , Flow Cytometry , Humans , Interleukin-17/analysis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Phosphorylation/drug effects , Polymerase Chain Reaction , Th17 Cells/immunology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
13.
Proc Natl Acad Sci U S A ; 108(18): 7511-6, 2011 May 03.
Article in English | MEDLINE | ID: mdl-21502500

ABSTRACT

Vasectomy is a well accepted global contraceptive approach frequently associated with epididymal granuloma and sperm autoantibody formation. To understand the long-term sequelae of vasectomy, we investigated the early immune response in vasectomized mice. Vasectomy leads to rapid epithelial cell apoptosis and necrosis, persistent inflammation, and sperm granuloma formation in the epididymis. Vasectomized B6AF1 mice did not mount autoimmune response but instead developed sperm antigen-specific tolerance, documented as resistance to immunization-induced experimental autoimmune orchitis (EAO) but not experimental autoimmune encephalomyelitis. Strikingly, tolerance switches over to pathologic autoimmune state following concomitant CD4(+)CD25(+)Foxp3(+) regulatory T cell (Treg) depletion: unilaterally vasectomized mice produce dominant autoantibodies to an orchitogenic antigen (zonadhesin), and develop CD4 T-cell- and antibody-dependent bilateral autoimmune orchitis. Therefore, (i) Treg normally prevents spontaneous organ-specific autoimmunity induction by persistent endogenous danger signal, and (ii) autoantigenic stimulation with sterile autoinflammation can lead to tolerance. Finally, postvasectomy tolerance occurs in B6AF1, C57BL/6, and A/J strains. However, C57BL/6 mice resisted EAO after 60% Treg depletion, but developed EAO after 97% Treg reduction. Therefore, variance in intrinsic Treg function--a possible genetic trait--can influence the divergent tolerogenic versus autoimmune response to vasectomy.


Subject(s)
Autoimmunity/immunology , Immune Tolerance/immunology , Spermatozoa/immunology , T-Lymphocytes, Regulatory/immunology , Vasectomy , Animals , Autoantibodies/immunology , Blotting, Western , Cell Proliferation , Electrophoresis, Polyacrylamide Gel , Male , Membrane Proteins/immunology , Mice , Mice, Mutant Strains , Statistics, Nonparametric
14.
J Immunol ; 186(1): 382-9, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21135167

ABSTRACT

Day 3 thymectomy (D3Tx) results in a loss of peripheral tolerance mediated by natural regulatory T cells (nTregs) and development of autoimmune ovarian dysgenesis (AOD) and autoimmune dacryoadenitis (ADA) in A/J and (C57BL/6J × A/J) F(1) hybrids (B6A), but not in C57BL/6J (B6) mice. Previously, using quantitative trait locus (QTL) linkage analysis, we showed that D3Tx-AOD is controlled by five unlinked QTL (Aod1-Aod5) and H2. In this study, using D3Tx B6-Chr(A/J)/NaJ chromosome (Chr) substitution strains, we confirm that QTL on Chr16 (Aod1a/Aod1b), Chr3 (Aod2), Chr1 (Aod3), Chr2 (Aod4), Chr7 (Aod5), and Chr17 (H2) control D3Tx-AOD susceptibility. In addition, we also present data mapping QTL controlling D3Tx-ADA to Chr17 (Ada1/H2), Chr1 (Ada2), and Chr3 (Ada3). Importantly, B6-ChrX(A/J) mice were as resistant to D3Tx-AOD and D3Tx-ADA as B6 mice, thereby excluding Foxp3 as a susceptibility gene in these models. Moreover, we report quantitative differences in the frequency of nTregs in the lymph nodes (LNs), but not spleen or thymus, of AOD/ADA-resistant B6 and AOD/ADA-susceptible A/J, B6A, and B6-Chr17(A/J) mice. Similar results correlating with experimental allergic encephalomyelitis and orchitis susceptibility were seen with B10.S and SJL/J mice. Using H2-congenic mice, we show that the observed difference in frequency of LN nTregs is controlled by Ada1/H2. These data support the existence of an LN-specific, H2-controlled mechanism regulating the prevalence of nTregs in autoimmune disease susceptibility.


Subject(s)
Autoimmune Diseases/immunology , H-2 Antigens/physiology , Lymph Nodes/cytology , Lymph Nodes/immunology , Oophoritis/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Thymectomy , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/surgery , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Chromosomes/genetics , Dacryocystitis/genetics , Dacryocystitis/immunology , Disease Susceptibility/immunology , Female , Genetic Linkage/immunology , Lymph Nodes/metabolism , Lymphocyte Count , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, Transgenic , Oophoritis/genetics , Quantitative Trait Loci/immunology
15.
Cell Immunol ; 260(2): 119-27, 2010.
Article in English | MEDLINE | ID: mdl-19914609

ABSTRACT

The postnatal maternal environment is known to increase susceptibility to a number of autoimmune diseases. Here we asked whether the postnatal maternal environment could influence autoimmune disease development to day 3 thymectomy (d3tx)-induced autoimmune ovarian disease (AOD) and experimental allergic encephalomyelitis (EAE) in cross-fostered A/J and B6 mice. A/J pups foster-nursed by B6 mothers exhibit an increase in autoimmune disease development while cross-fostering B6 pups on A/J mothers did not alter their susceptibility. The increase in AOD incidence seen in foster-nursed d3tx A/J mice correlated with a decrease in the total number of CD4(+) T cells in the lymph nodes of these animals. Analysis of the cellular composition in the milk revealed that B6 mice shed significantly more maternally derived lymphocytes into their milk compared to A/J mothers. These data suggest that there are maternally derived postnatal factors that influence the development of autoimmune disease in A/J mice.


Subject(s)
Animals, Newborn/immunology , Animals, Suckling/immunology , Autoimmune Diseases/immunology , Disease Susceptibility/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Chemokine CXCL1/metabolism , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Immunity, Maternally-Acquired/immunology , Interleukin-13/metabolism , Interleukin-9/metabolism , Lactation/immunology , Lactation/metabolism , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Ovarian Diseases/immunology , Thymectomy
16.
Biol Reprod ; 80(5): 874-81, 2009 May.
Article in English | MEDLINE | ID: mdl-19144956

ABSTRACT

Intrauterine or intraperitoneal administration of lipopolysaccharide (LPS) into normal mice at midgestation induces preterm delivery (PTD) within 24 h through a mechanism dependent on Toll-like receptor signaling and expression of inflammatory cytokines. The exact participants in the cellular network involved in PTD are not known. Although the activities of innate immune cells are thought to be important, the extent to which this process depends on T and B cells has yet to be examined. Mice deficient in T and B cells due to genetic deficiency in the recombination activating gene 1 (Rag1(-/-)) were given LPS intraperitoneally on Day 15 of gestation and found to be susceptible to LPS-induced PTD. This was found to involve many of the inflammatory mediators reported as important in normal mice. Moreover, at a low dose (3 microg), pregnant Rag1(-/-) mice were found to be more susceptible to PTD than a cohort of normal mice on the same genetic background. This increased susceptibility was partially reversed by transfer, on Day 10 of gestation, of whole lymphocytes or purified CD4(+) T cells. Transfer of purified CD4(+) T cells to Rag1(-/-) mice resulted in a uterine draining node population of FOXP3(+) cells, suggesting that these cells may contribute to resistance to LPS-induced PTD. Overall, the data suggest that, although T and B lymphocytes are not critical positive regulators of LPS-induced PTD, CD4(+) T cells play a protective and regulatory role, and thus could be a target for preventive or therapeutic manipulation.


Subject(s)
Lipopolysaccharides/toxicity , Premature Birth/immunology , Premature Birth/prevention & control , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , B-Lymphocytes/immunology , Base Sequence , DNA, Complementary/genetics , Female , Forkhead Transcription Factors/immunology , Genes, RAG-1 , Lipopolysaccharides/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pregnancy , Premature Birth/chemically induced , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/transplantation , Toll-Like Receptor 4/immunology
17.
Am J Pathol ; 173(3): 892-900, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18688020

ABSTRACT

Weibel-Palade bodies within endothelial cells are secretory granules known to release von Willebrand Factor (VWF), P-selectin, chemokines, and other stored molecules following histamine exposure. Mice with a disrupted VWF gene (VWFKO) have endothelial cells that are deficient in Weibel-Palade bodies. These mice were used to evaluate the role of VWF and/or Weibel-Palade bodies in Bordetella pertussis toxin-induced hypersensitivity to histamine, a subphenotype of experimental allergic encephalomyelitis, the principal autoimmune model of multiple sclerosis. No significant differences in susceptibility to histamine between wild-type and VWFKO mice were detected after 3 days; however, histamine sensitivity persisted significantly longer in VWFKO mice. Correspondingly, encephalomyelitis onset was earlier, disease was more severe, and blood brain barrier (BBB) permeability was significantly increased in VWFKO mice, as compared with wild-type mice. Moreover, inflammation was selectively increased in the brains, but not spinal cords, of VWFKO mice as compared with wild-type mice. Early increases in BBB permeability in VWFKO mice were not due to increased encephalitogenic T-cell activity since BBB permeability did not differ in adjuvant-treated VWFKO mice as compared with littermates immunized with encephalitogenic peptide plus adjuvant. Taken together, these data indicate that VWF and/or Weibel-Palade bodies negatively regulate BBB permeability changes and autoimmune inflammatory lesion formation within the brain elicited by peripheral inflammatory stimuli.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/pathology , Capillary Permeability/physiology , Encephalomyelitis, Autoimmune, Experimental/pathology , von Willebrand Factor/metabolism , Adjuvants, Immunologic/metabolism , Animals , Blood-Brain Barrier/pathology , Brain/immunology , Brain/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Histamine/immunology , Hypersensitivity/immunology , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Weibel-Palade Bodies/metabolism , von Willebrand Factor/genetics
18.
J Immunol ; 180(11): 7471-9, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18490747

ABSTRACT

Structural polymorphisms (L263P, M313V, and S331P) in the third intracellular loop of the murine histamine receptor H(1) (H(1)R) are candidates for Bphs, a shared autoimmune disease locus in experimental allergic encephalomyelitis and experimental allergic orchitis. The P-V-P haplotype is associated with increased disease susceptibility (H(1)R(S)) whereas the L-M-S haplotype is associated with less severe disease (H(1)R(R)). In this study, we show that selective re-expression of the H(1)R(S) allele in T cells fully complements experimental allergic encephalomyelitis susceptibility and the production of disease-associated cytokines while selective re-expression of the H(1)R(R) allele does not. Mechanistically, we show that the two H(1)R alleles exhibit differential cell surface expression and altered intracellular trafficking, with the H(1)R(R) allele being retained within the endoplasmic reticulum. Moreover, we show that all three residues (L-M-S) comprising the H(1)R(R) haplotype are required for altered expression. These data are the first to demonstrate that structural polymorphisms influencing cell surface expression of a G protein-coupled receptor in T cells regulates immune functions and autoimmune disease susceptibility.


Subject(s)
Autoimmune Diseases/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Orchitis/genetics , Receptors, Histamine H1/genetics , Alleles , Animals , Autoimmune Diseases/immunology , Cell Line , Cell Membrane/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Endoplasmic Reticulum/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Genetic Predisposition to Disease , Haplotypes , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Orchitis/immunology , Polymorphism, Genetic , Protein Transport , Receptors, Histamine H1/deficiency , Receptors, Histamine H1/immunology , Receptors, Histamine H1/metabolism , T-Lymphocytes/metabolism
19.
J Clin Invest ; 117(11): 3507-18, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17965772

ABSTRACT

Histamine receptor H1 (H1R) is a susceptibility gene in both experimental autoimmune encephalomyelitis (EAE) and experimental autoimmune orchitis (EAO), 2 classical T cell-mediated models of organ-specific autoimmune disease. Here we showed that expression of H1R in naive CD4+ T cells was required for maximal IFN-gamma production but was dispensable for proliferation. Moreover, H1R signaling at the time of TCR ligation was required for activation of p38 MAPK, a known regulator of IFN-gamma expression. Importantly, selective reexpression of H1R in CD4+ T cells fully complemented both the IFN-gamma production and the EAE susceptibility of H1R-deficient mice. These data suggest that the presence of H1R in CD4+ T cells and its interaction with histamine regulates early TCR signals that lead to Th1 differentiation and autoimmune disease.


Subject(s)
Interferon-gamma/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Histamine H1/immunology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Enzyme Activation , Histamine/immunology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orchitis/immunology , Receptors, Histamine H1/genetics , Signal Transduction/physiology , Th1 Cells/cytology , Th1 Cells/immunology
20.
Biochem Biophys Res Commun ; 325(1): 133-43, 2004 Dec 03.
Article in English | MEDLINE | ID: mdl-15522211

ABSTRACT

The turnover of phosphoinositides leading to PKC activation constitutes one of the principal axes of intracellular signaling. In T lymphocytes, the enhanced and prolonged PKC activation resulting from the engagement of the TcR and co-receptor molecules ensures a productive T cell response. The CD43 co-receptor promotes activation and proliferation, by inducing IL-2 secretion and CD69 expression. CD43 engagement has been shown to promote phosphoinositide turnover and DAG production. Moreover, PKC activation was found to be required for the activation of the MAP kinase pathway in response to CD43 ligation. Here we show that CD43 engagement led to the membrane translocation and enzymatic activity of specific PKC isoenzymes: cPKC (alpha/beta), nPKC (epsilon and theta;), aPKC (zeta) and PKCmu. We also show that activation of PKCtheta; resulting from CD43 ligation induced CD69 expression through an ERK-dependent pathway leading to AP-1, NF-kappaB activation and an ERK independent pathway promoting NFAT activation. Together, these data suggest that PKCtheta; plays a critical role in the co-stimulatory functions of CD43 in human T cells.


Subject(s)
Antigens, CD/metabolism , Isoenzymes/metabolism , Lymphocyte Activation , MAP Kinase Signaling System/physiology , Protein Kinase C/metabolism , Sialoglycoproteins/metabolism , T-Lymphocytes/physiology , Animals , Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Line , DNA/metabolism , DNA-Binding Proteins/metabolism , Enzyme Activation , Humans , Lectins, C-Type , Leukosialin , NF-kappa B/metabolism , NFATC Transcription Factors , Nuclear Proteins/metabolism , Protein Kinase C-theta , Protein Transport/physiology , T-Lymphocytes/cytology , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...