Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Nat Immunol ; 18(6): 633-641, 2017 06.
Article in English | MEDLINE | ID: mdl-28459434

ABSTRACT

Microglia and other tissue-resident macrophages within the central nervous system (CNS) have essential roles in neural development, inflammation and homeostasis. However, the molecular pathways underlying their development and function remain poorly understood. Here we report that mice deficient in NRROS, a myeloid-expressed transmembrane protein in the endoplasmic reticulum, develop spontaneous neurological disorders. NRROS-deficient (Nrros-/-) mice show defects in motor functions and die before 6 months of age. Nrros-/- mice display astrogliosis and lack normal CD11bhiCD45lo microglia, but they show no detectable demyelination or neuronal loss. Instead, perivascular macrophage-like myeloid cells populate the Nrros-/- CNS. Cx3cr1-driven deletion of Nrros shows its crucial role in microglial establishment during early embryonic stages. NRROS is required for normal expression of Sall1 and other microglial genes that are important for microglial development and function. Our study reveals a NRROS-mediated pathway that controls CNS-resident macrophage development and affects neurological function.


Subject(s)
Astrocytes/metabolism , Central Nervous System/embryology , Gene Expression Regulation, Developmental , Microglia/metabolism , Myeloid Cells/metabolism , Nervous System Diseases/genetics , Proteins/genetics , Animals , Astrocytes/cytology , Blotting, Western , Central Nervous System/cytology , Flow Cytometry , Immunohistochemistry , Lameness, Animal/genetics , Latent TGF-beta Binding Proteins , Locomotion , Macrophages/cytology , Macrophages/metabolism , Membrane Proteins , Mice , Mice, Knockout , Microglia/cytology , Myeloid Cells/cytology , Posture , Transcription Factors/genetics , Urinary Incontinence/genetics , Urinary Retention/genetics
2.
Nat Immunol ; 12(12): 1238-45, 2011 Oct 16.
Article in English | MEDLINE | ID: mdl-22001828

ABSTRACT

Interleukin 22 (IL-22), which is produced by cells of the T(H)17 subset of helper T cells and other leukocytes, not only enhances proinflammatory innate defense mechanisms in epithelial cells but also provides crucial protection to tissues from damage caused by inflammation and infection. In T(H)17 cells, transforming growth factor-ß (TGF-ß) regulates IL-22 and IL-17 differently. IL-6 alone induces T cells to produce only IL-22, whereas the combination of IL-6 and high concentrations of TGF-ß results in the production of IL-17 but not IL-22 by T cells. Here we identify the transcription factor c-Maf, which is induced by TGF-ß, as a downstream repressor of Il22. We found that c-Maf bound to the Il22 promoter and was both necessary and sufficient for the TGF-ß-dependent suppression of IL-22 production in T(H)17 cells.


Subject(s)
Interleukins/biosynthesis , Proto-Oncogene Proteins c-maf/metabolism , Th17 Cells/immunology , Transforming Growth Factor beta/pharmacology , Animals , Base Sequence , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Binding Sites/genetics , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Interleukins/genetics , Mice , Mice, Inbred BALB C , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Nucleotide Motifs , Promoter Regions, Genetic , Proto-Oncogene Proteins c-maf/genetics , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Th17 Cells/drug effects , Transcription, Genetic , Interleukin-22
3.
Nature ; 518(7539): 417-21, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25470037

ABSTRACT

T-helper type 17 (TH17) cells that produce the cytokines interleukin-17A (IL-17A) and IL-17F are implicated in the pathogenesis of several autoimmune diseases. The differentiation of TH17 cells is regulated by transcription factors such as RORγt, but post-translational mechanisms preventing the rampant production of pro-inflammatory IL-17A have received less attention. Here we show that the deubiquitylating enzyme DUBA is a negative regulator of IL-17A production in T cells. Mice with DUBA-deficient T cells developed exacerbated inflammation in the small intestine after challenge with anti-CD3 antibodies. DUBA interacted with the ubiquitin ligase UBR5, which suppressed DUBA abundance in naive T cells. DUBA accumulated in activated T cells and stabilized UBR5, which then ubiquitylated RORγt in response to TGF-ß signalling. Our data identify DUBA as a cell-intrinsic suppressor of IL-17 production.


Subject(s)
Interleukin-17/biosynthesis , Protein Biosynthesis , Th17 Cells/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Enzyme Stability , Female , Inflammation/genetics , Inflammation/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Signal Transduction , Substrate Specificity , Transforming Growth Factor beta/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Proteases/biosynthesis , Ubiquitin-Specific Proteases/deficiency , Ubiquitin-Specific Proteases/genetics , Ubiquitination
4.
Nature ; 506(7489): 456-62, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24553140

ABSTRACT

Crohn's disease is a debilitating inflammatory bowel disease (IBD) that can involve the entire digestive tract. A single-nucleotide polymorphism (SNP) encoding a missense variant in the autophagy gene ATG16L1 (rs2241880, Thr300Ala) is strongly associated with the incidence of Crohn's disease. Numerous studies have demonstrated the effect of ATG16L1 deletion or deficiency; however, the molecular consequences of the Thr300Ala (T300A) variant remains unknown. Here we show that amino acids 296-299 constitute a caspase cleavage motif in ATG16L1 and that the T300A variant (T316A in mice) significantly increases ATG16L1 sensitization to caspase-3-mediated processing. We observed that death-receptor activation or starvation-induced metabolic stress in human and murine macrophages increased degradation of the T300A or T316A variants of ATG16L1, respectively, resulting in diminished autophagy. Knock-in mice harbouring the T316A variant showed defective clearance of the ileal pathogen Yersinia enterocolitica and an elevated inflammatory cytokine response. In turn, deletion of the caspase-3-encoding gene, Casp3, or elimination of the caspase cleavage site by site-directed mutagenesis rescued starvation-induced autophagy and pathogen clearance, respectively. These findings demonstrate that caspase 3 activation in the presence of a common risk allele leads to accelerated degradation of ATG16L1, placing cellular stress, apoptotic stimuli and impaired autophagy in a unified pathway that predisposes to Crohn's disease.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 3/metabolism , Crohn Disease/genetics , Polymorphism, Single Nucleotide/genetics , Proteolysis , Amino Acid Motifs , Animals , Autophagy/genetics , Autophagy-Related Proteins , Carrier Proteins/chemistry , Caspase 3/deficiency , Caspase 3/genetics , Cell Line , Cells, Cultured , Crohn Disease/pathology , Cytokines/immunology , Enzyme Activation , Female , Food Deprivation , Humans , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Stress, Physiological , Yersinia enterocolitica/immunology
5.
Nature ; 509(7499): 235-9, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24739962

ABSTRACT

Reactive oxygen species (ROS) produced by phagocytes are essential for host defence against bacterial and fungal infections. Individuals with defective ROS production machinery develop chronic granulomatous disease. Conversely, excessive ROS can cause collateral tissue damage during inflammatory processes and therefore needs to be tightly regulated. Here we describe a protein, we termed negative regulator of ROS (NRROS), which limits ROS generation by phagocytes during inflammatory responses. NRROS expression in phagocytes can be repressed by inflammatory signals. NRROS-deficient phagocytes produce increased ROS upon inflammatory challenges, and mice lacking NRROS in their phagocytes show enhanced bactericidal activity against Escherichia coli and Listeria monocytogenes. Conversely, these mice develop severe experimental autoimmune encephalomyelitis owing to oxidative tissue damage in the central nervous system. Mechanistically, NRROS is localized to the endoplasmic reticulum, where it directly interacts with nascent NOX2 (also known as gp91(phox) and encoded by Cybb) monomer, one of the membrane-bound subunits of the NADPH oxidase complex, and facilitates the degradation of NOX2 through the endoplasmic-reticulum-associated degradation pathway. Thus, NRROS provides a hitherto undefined mechanism for regulating ROS production--one that enables phagocytes to produce higher amounts of ROS, if required to control invading pathogens, while minimizing unwanted collateral tissue damage.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Escherichia coli/immunology , Listeria monocytogenes/immunology , Proteins/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Animals , Autoimmunity/genetics , Bone Marrow Cells/cytology , Central Nervous System/metabolism , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/metabolism , Female , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Latent TGF-beta Binding Proteins , Macrophages/cytology , Macrophages/enzymology , Macrophages/immunology , Macrophages/metabolism , Male , Membrane Proteins , Mice , NADPH Oxidases/metabolism , Oxidation-Reduction , Oxidative Stress , Phagocytes/cytology , Phagocytes/immunology , Phagocytes/metabolism , Proteins/genetics , Reactive Oxygen Species/metabolism
6.
Ann Neurol ; 75(1): 50-66, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24027119

ABSTRACT

OBJECTIVE: Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system (CNS), characterized by a global increasing incidence driven by relapsing-remitting disease in females. Investigators have described p38 mitogen-activated protein kinase (MAPK) as a key regulator of inflammatory responses in autoimmunity, but its role in the sexual dimorphism in MS or MS models remains unexplored. METHODS: Toward this end, we used experimental autoimmune encephalomyelitis (EAE), the principal animal model of MS, combined with pharmacologic and genetic inhibition of p38 MAPK activity and transcriptomic analyses. RESULTS: Pharmacologic inhibition of p38 MAPK selectively ameliorated EAE in female mice. Conditional deletion studies demonstrated that p38α signaling in macrophages/myeloid cells, but not T cells or dendritic cells, mediated this sexual dimorphism, which was dependent on the presence of adult sex hormones. Analysis of CNS inflammatory infiltrates showed that female but not male mice lacking p38α in myeloid cells exhibited reduced immune cell activation compared with controls, whereas peripheral T-cell priming was unaffected in both sexes. Transcriptomic analyses of myeloid cells revealed differences in p38α-controlled transcripts comprising female- and male-specific gene modules, with greater p38α dependence of proinflammatory gene expression in females. INTERPRETATION: Our findings demonstrate a key role for p38α in myeloid cells in CNS autoimmunity and uncover important molecular mechanisms underlying sex differences in disease pathogenesis. Taken together, our results suggest that the p38 MAPK signaling pathway represents a novel target for much needed disease-modifying therapies for MS.


Subject(s)
Autoimmunity/physiology , Encephalomyelitis, Autoimmune, Experimental/enzymology , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 14/metabolism , Myeloid Cells/enzymology , Sex Characteristics , Animals , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Enzyme Activation/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinase 14/immunology , Myeloid Cells/immunology , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
7.
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
8.
Infect Immun ; 82(2): 773-82, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24478091

ABSTRACT

Pertussis toxin (PTX) is an AB5-type exotoxin produced by the bacterium Bordetella pertussis, the causative agent of whooping cough. In vivo intoxication with PTX elicits a variety of immunologic and inflammatory responses, including vasoactive amine sensitization (VAAS) to histamine (HA), serotonin (5-HT), and bradykinin (BDK). Previously, by using a forward genetic approach, we identified the HA H1 receptor (Hrh1/H1R) as the gene in mice that controls differential susceptibility to B. pertussis PTX-induced HA sensitization (Bphs). Here we show, by using inbred strains of mice, F1 hybrids, and segregating populations, that, unlike Bphs, PTX-induced 5-HT sensitivity (Bpss) and BDK sensitivity (Bpbs) are recessive traits and are separately controlled by multiple loci unlinked to 5-HT and BDK receptors, respectively. Furthermore, we found that PTX sensitizes mice to HA independently of Toll-like receptor 4, a purported receptor for PTX, and that the VAAS properties of PTX are not dependent upon endothelial caveolae or endothelial nitric oxide synthase. Finally, by using mice deficient in individual Gαi/o G-protein subunits, we demonstrate that Gαi1 and Gαi3 are the critical in vivo targets of ADP-ribosylation underlying VAAS elicited by PTX exposure.


Subject(s)
Amines/metabolism , Bordetella pertussis/pathogenicity , Cardiovascular Agents/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Pertussis Toxin/metabolism , Adenosine Diphosphate Ribose/metabolism , Animals , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Mice , Mice, Knockout
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.
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
11.
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
12.
Proc Natl Acad Sci U S A ; 107(44): 18967-72, 2010 Nov 02.
Article in English | MEDLINE | ID: mdl-20956310

ABSTRACT

Disruption of the blood-brain barrier (BBB) underlies the development of experimental autoimmune encephalomyelitis (EAE) and multiple sclerosis. Environmental factors, such as Bordetella pertussis, are thought to sensitize central endothelium to biogenic amines like histamine, thereby leading to increased BBB permeability. B. pertussis-induced histamine sensitization (Bphs) is a monogenic intermediate phenotype of EAE controlled by histamine H(1) receptor (Hrh1/H(1)R). Here, we transgenically overexpressed H(1)R in endothelial cells of Hrh1-KO (H(1)RKO) mice to test the role of endothelial H(1)R directly in Bphs and EAE. Unexpectedly, transgenic H(1)RKO mice expressing endothelial H(1)R under control of the von Willebrand factor promoter (H(1)RKO-vWF(H1R) Tg) were Bphs-resistant. Moreover, H(1)RKO-vWF(H1R) Tg mice exhibited decreased BBB permeability and enhanced protection from EAE compared with H(1)RKO mice. Thus, contrary to prevailing assumptions, our results show that endothelial H(1)R expression reduces BBB permeability, suggesting that endothelial H(1)R signaling may be important in the maintenance of cerebrovascular integrity.


Subject(s)
Blood-Brain Barrier/metabolism , Capillary Permeability , Encephalomyelitis, Autoimmune, Experimental/metabolism , Endothelium, Vascular/metabolism , Receptors, Histamine H1/metabolism , Signal Transduction , Animals , Bordetella pertussis/genetics , Bordetella pertussis/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Genetic Predisposition to Disease , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Receptors, Histamine H1/genetics , Whooping Cough/genetics , Whooping Cough/metabolism , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
13.
J Clin Invest ; 133(12)2023 06 15.
Article in English | MEDLINE | ID: mdl-37317970

ABSTRACT

While the rapid advancement of immunotherapies has revolutionized cancer treatment, only a small fraction of patients derive clinical benefit. Eradication of large, established tumors appears to depend on engaging and activating both innate and adaptive immune system components to mount a rigorous and comprehensive immune response. Identifying such agents is a high unmet medical need, because they are sparse in the therapeutic landscape of cancer treatment. Here, we report that IL-36 cytokine can engage both innate and adaptive immunity to remodel an immune-suppressive tumor microenvironment (TME) and mediate potent antitumor immune responses via signaling in host hematopoietic cells. Mechanistically, IL-36 signaling modulates neutrophils in a cell-intrinsic manner to greatly enhance not only their ability to directly kill tumor cells but also promote T and NK cell responses. Thus, while poor prognostic outcomes are typically associated with neutrophil enrichment in the TME, our results highlight the pleiotropic effects of IL-36 and its therapeutic potential to modify tumor-infiltrating neutrophils into potent effector cells and engage both the innate and adaptive immune system to achieve durable antitumor responses in solid tumors.


Subject(s)
Adaptive Immunity , Neutrophils , Humans , Cytokines , Immunosuppression Therapy , Immunotherapy
14.
Cell Immunol ; 278(1-2): 27-34, 2012.
Article in English | MEDLINE | ID: mdl-23121973

ABSTRACT

The histamine H(1) receptor (Hrh1/H(1)R) was identified as an autoimmune disease gene in experimental allergic encephalomyelitis (EAE), the principal autoimmune model of multiple sclerosis (MS). Previously, we showed that selective re-expression of H(1)R by endothelial cells or T cells in H(1)RKO mice significantly reduced or complemented EAE severity and cytokine responses, respectively. H(1)R regulates innate immune cells, which in turn influences peripheral and central nervous system CD4(+) T cell effector responses. Therefore, we selectively re-expressed H(1)R in CD11b(+) cells of H(1)RKO mice to test the hypothesis that H(1)R signaling in these cells contributes to EAE susceptibility. We demonstrate that transgenic re-expression of H(1)R by H(1)RKO-CD11b(+) cells neither complements EAE susceptibility nor T cell cytokine responses highlighting the cell-specific effects of Hrh1 in the pathogenesis of EAE and MS, and the need for cell-specific targeting in optimizing therapeutic interventions based on such genes.


Subject(s)
Antigens, CD1/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Receptors, Histamine H1/immunology , Animals , Antigens, CD1/genetics , Cytokines/biosynthesis , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression , Genetic Predisposition to Disease , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Organ Specificity , Receptors, Histamine H1/deficiency , Receptors, Histamine H1/genetics , Severity of Illness Index , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
15.
Cell Immunol ; 272(2): 269-74, 2012.
Article in English | MEDLINE | ID: mdl-22078271

ABSTRACT

Susceptibility to autoimmune myocarditis has been associated with histamine release by mast cells during the innate immune response to coxsackievirus B3 (CVB3) infection. To investigate the contribution of histamine H(1) receptor (H(1)R) signaling to CVB3-induced myocarditis, we assessed susceptibility to the disease in C57BL/6J (B6) H(1)R(-/-) mice. No difference was observed in mortality between CVB3-infected B6 and H(1)R(-/-) mice. However, analysis of their hearts revealed a significant increase in myocarditis in H(1)R(-/-) mice that is not attributed to increased virus replication. Enhanced myocarditis susceptibility correlated with a significant expansion in pathogenic Th1 and Vγ4(+) γδ T cells in the periphery of these animals. Furthermore, an increase in regulatory T cells was observed, yet these cells were incapable of controlling myocarditis in H(1)R(-/-) mice. These data establish a critical role for histamine and H(1)R signaling in regulating T cell responses and susceptibility to CVB3-induced myocarditis in B6 mice.


Subject(s)
Coxsackievirus Infections/immunology , Enterovirus B, Human/immunology , Myocarditis/immunology , Receptors, Histamine H1/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Coxsackievirus Infections/metabolism , Disease Susceptibility , Histamine/metabolism , Male , Mice , Mice, Inbred C57BL , Myocarditis/metabolism , Receptors, Histamine H1/deficiency , Receptors, Histamine H1/metabolism , Signal Transduction , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Virus Replication/immunology
16.
J Immunol ; 182(12): 7776-83, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19494301

ABSTRACT

Genes controlling immunopathologic diseases of differing etiopathology may also influence susceptibility to autoimmune disease. B10.D1-H2(q)/SgJ mice with a 2538 G-->A missense mutation in the tyrosine kinase-2 gene (Tyk2) are susceptible to Toxoplasma gondii yet resistant to autoimmune arthritis, unlike the wild-type B10.Q/Ai substrain. To understand whether Tyk2 is also important in a second autoimmune model, experimental allergic encephalomyelitis (EAE) was induced in B10.D1-H2(q)/SgJ (Tyk2(A)) and B10.Q/Ai (Tyk2(G)) mice with the myelin oligodendrocyte glycoprotein peptide 79-96. B10.D1-H2(q)/SgJ mice were resistant to EAE whereas B10.Q/Ai mice were susceptible, and a single copy of the Tyk2(G) allele conferred EAE susceptibility in F(1) hybrids. Furthermore, EAE resistance in B10.D1-H2(q)/SgJ mice was overridden when pertussis toxin (PTX) was used to mimic the effects of environmental factors derived from infectious agents. Numerous cytokines and chemokines were increased when PTX was included in the immunization protocol. However, only RANTES, IL-6, and IFN-gamma increased significantly with both genetic compensation and PTX treatment. These data indicate that Tyk2 is a shared autoimmune disease susceptibility gene whose genetic contribution to disease susceptibility can be modified by environmental factors. Single nucleotide polymorphisms like the one that distinguishes Tyk2 alleles are of considerable significance given the potential role of gene-by-environment interactions in autoimmune disease susceptibility.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/genetics , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide/genetics , TYK2 Kinase/genetics , TYK2 Kinase/metabolism , Alleles , Animals , Cells, Cultured , Cytokines/biosynthesis , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Myelin Proteins , Myelin-Associated Glycoprotein/pharmacology , Myelin-Oligodendrocyte Glycoprotein , Pertussis Toxin/pharmacology
17.
J Immunol ; 182(4): 1789-93, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19201829

ABSTRACT

Multiple sclerosis is a sexually dimorphic, demyelinating disease of the CNS, and experimental allergic encephalomyelitis (EAE) is its principal autoimmune model. Young male SJL/J mice are relatively resistant to EAE whereas older males and SJL/J females of any age are susceptible. By comparing a wide age range of proteolipid protein peptide 139-151 immunized mice, we found that female disease severity remains constant with age. In contrast, EAE disease severity increases with age in SJL/J males, with young males having significantly less severe disease and older males having significantly more disease than equivalently aged females. To determine whether the Y chromosome contributes to this sexual dimorphism, EAE was induced in consomic SJL/J mice carrying a B10.S Y chromosome (SJL.Y(B10.S)). EAE was significantly more severe in young male SJL.Y(B10.S) mice compared with young male SJL/J mice. These studies show that a Y chromosome-linked polymorphism controls the age-dependent EAE sexual dimorphism observed in SJL/J mice.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Genetic Predisposition to Disease/genetics , Sex Characteristics , Y Chromosome/genetics , Age Factors , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Male , Mice , Seasons
18.
Sci Transl Med ; 13(608)2021 08 25.
Article in English | MEDLINE | ID: mdl-34433637

ABSTRACT

Therapeutic approaches are needed to promote T cell-mediated destruction of poorly immunogenic, "cold" tumors typically associated with minimal response to immune checkpoint blockade (ICB) therapy. Bispecific T cell engager (BiTE) molecules induce redirected lysis of cancer cells by polyclonal T cells and have demonstrated promising clinical activity against solid tumors in some patients. However, little is understood about the key factors that govern clinical responses to these therapies. Using an immunocompetent mouse model expressing a humanized CD3ε chain (huCD3e mice) and BiTE molecules directed against mouse CD19, mouse CLDN18.2, or human EPCAM antigens, we investigated the pharmacokinetic and pharmacodynamic parameters and immune correlates associated with BiTE efficacy across multiple syngeneic solid-tumor models. These studies demonstrated that pretreatment tumor-associated T cell density is a critical determinant of response to BiTE therapy, identified CD8+ T cells as important targets and mediators of BiTE activity, and revealed an antagonistic role for CD4+ T cells in BiTE efficacy. We also identified therapeutic combinations, including ICB and 4-1BB agonism, that synergized with BiTE treatment in poorly T cell-infiltrated, immunotherapy-refractory tumors. In these models, BiTE efficacy was dependent on local expansion of tumor-associated CD8+ T cells, rather than their recruitment from circulation. Our findings highlight the relative contributions of baseline T cell infiltration, local T cell proliferation, and peripheral T cell trafficking for BiTE molecule-mediated efficacy, identify combination strategies capable of overcoming resistance to BiTE therapy, and have clinical relevance for the development of BiTE and other T cell engager therapies.


Subject(s)
Antibodies, Bispecific , Neoplasms , Animals , Antibodies, Bispecific/therapeutic use , Antigens, CD19 , CD3 Complex , CD8-Positive T-Lymphocytes , Claudins , Humans , Immunotherapy , Mice , Neoplasms/drug therapy
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.
Cancer Res ; 80(8): 1656-1668, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31988076

ABSTRACT

The deubiquitinating enzyme BAP1 is mutated in a hereditary cancer syndrome with a high risk for mesothelioma and melanocytic tumors. Here, we show that pancreatic intraepithelial neoplasia driven by oncogenic mutant KrasG12D progressed to pancreatic adenocarcinoma in the absence of BAP1. The Hippo pathway was deregulated in BAP1-deficient pancreatic tumors, with the tumor suppressor LATS exhibiting enhanced ubiquitin-dependent proteasomal degradation. Therefore, BAP1 may limit tumor progression by stabilizing LATS and thereby promoting activity of the Hippo tumor suppressor pathway. SIGNIFICANCE: BAP1 is mutated in a broad spectrum of tumors. Pancreatic Bap1 deficiency causes acinar atrophy but combines with oncogenic Ras to produce pancreatic tumors. BAP1-deficient tumors exhibit deregulation of the Hippo pathway.See related commentary by Brekken, p. 1624.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Hippo Signaling Pathway , Humans , Protein Serine-Threonine Kinases , Signal Transduction , Tumor Suppressor Proteins , Ubiquitin Thiolesterase
SELECTION OF CITATIONS
SEARCH DETAIL