Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Clin Invest ; 133(12)2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37317970

RESUMEN

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.


Asunto(s)
Inmunidad Adaptativa , Neutrófilos , Humanos , Citocinas , Terapia de Inmunosupresión , Inmunoterapia
2.
Sci Transl Med ; 13(608)2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34433637

RESUMEN

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.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Animales , Anticuerpos Biespecíficos/uso terapéutico , Antígenos CD19 , Complejo CD3 , Linfocitos T CD8-positivos , Claudinas , Humanos , Inmunoterapia , Ratones , Neoplasias/tratamiento farmacológico
3.
Cancer Res ; 80(8): 1656-1668, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-31988076

RESUMEN

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.


Asunto(s)
Adenocarcinoma , Neoplasias Pancreáticas , Vía de Señalización Hippo , Humanos , Proteínas Serina-Treonina Quinasas , Transducción de Señal , Proteínas Supresoras de Tumor , Ubiquitina Tiolesterasa
4.
Front Immunol ; 10: 1014, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31143179

RESUMEN

Dendritic cells (DCs) efficiently process and present antigens to T cells, and by integrating environmental signals, link innate and adaptive immunity. DCs also control the balance between tolerance and immunity, and are required for T-cell mediated anti-tumor immunity. One subset of classical DCs, cDC1, are particularly important for eliciting CD8 T cells that can kill tumor cells. cDC1s are superior in antigen cross-presentation, a process of presenting exogenous antigens on MHC class I to activate CD8+ T cells. Tumor-associated cDC1s can transport tumor antigen to the draining lymph node and cross-present tumor antigens, resulting in priming and activation of cytotoxic T cells. Although cross-presenting cDC1s are critical for eliciting anti-tumor T cell responses, the role and importance of other DC subsets in anti-tumor immunity is not as well-characterized. Recent literature in other contexts suggests that critical crosstalk between DC subsets can significantly alter biological outcomes, and these DC interactions likely also contribute significantly to tumor-specific immune responses. Therefore, antigen presentation by cDC1s may be necessary but not sufficient for maximal immune responses against cancer. Here, we discuss recent advances in the understanding of DC subset interactions to maximize anti-tumor immunity, and propose that such interactions should be considered for the development of better DC-targeted immunotherapies.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Animales , Presentación de Antígeno , Comunicación Celular , Citocinas/metabolismo , Citotoxicidad Inmunológica , Células Dendríticas/trasplante , Humanos , Neoplasias/inmunología
5.
Sci Immunol ; 3(22)2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29678836

RESUMEN

Loss of function of the nuclear deubiquitinating enzyme BRCA1-associated protein-1 (BAP1) is associated with a wide spectrum of cancers. We report that tamoxifen-induced BAP1 deletion in adult mice resulted in severe thymic atrophy. BAP1 was critical for T cell development at several stages. In the thymus, BAP1 was required for progression through the pre-T cell receptor checkpoint. Peripheral T cells lacking BAP1 demonstrated a defect in homeostatic and antigen-driven expansion. Deletion of BAP1 resulted in suppression of E2F target genes and defects in cell cycle progression, which was dependent on the catalytic activity of BAP1, but did not require its interaction with host cell factor-1 (HCF-1). Loss of BAP1 led to increased monoubiquitination of histone H2A at Lys119 (H2AK119ub) throughout the T cell lineage, in particular in immature thymocytes, but did not alter trimethylation of histone H3 at Lys27 (H3K27me3). Deletion of BAP1 also abrogated B cell development in the bone marrow. Our findings uncover a nonredundant function for BAP1 in maintaining the lymphoid lineage.


Asunto(s)
Linfocitos T/metabolismo , Timocitos/metabolismo , Timo/metabolismo , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética , Animales , Atrofia , Ciclo Celular/genética , Perfilación de la Expresión Génica , Histonas/genética , Histonas/metabolismo , Lisina/genética , Lisina/metabolismo , Ratones Noqueados , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Timo/patología , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación
6.
Nat Immunol ; 18(6): 633-641, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28459434

RESUMEN

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.


Asunto(s)
Astrocitos/metabolismo , Sistema Nervioso Central/embriología , Regulación del Desarrollo de la Expresión Génica , Microglía/metabolismo , Células Mieloides/metabolismo , Enfermedades del Sistema Nervioso/genética , Proteínas/genética , Animales , Astrocitos/citología , Western Blotting , Sistema Nervioso Central/citología , Citometría de Flujo , Inmunohistoquímica , Cojera Animal/genética , Proteínas de Unión a TGF-beta Latente , Locomoción , Macrófagos/citología , Macrófagos/metabolismo , Proteínas de la Membrana , Ratones , Ratones Noqueados , Microglía/citología , Células Mieloides/citología , Postura , Factores de Transcripción/genética , Incontinencia Urinaria/genética , Retención Urinaria/genética
7.
Cell Rep ; 16(2): 583-595, 2016 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-27373151

RESUMEN

We introduce neutron-encoded (NeuCode) amino acid labeling of mice as a strategy for multiplexed proteomic analysis in vivo. Using NeuCode, we characterize an inducible knockout mouse model of Bap1, a tumor suppressor and deubiquitinase whose in vivo roles outside of cancer are not well established. NeuCode proteomics revealed altered metabolic pathways following Bap1 deletion, including profound elevation of cholesterol biosynthetic machinery coincident with reduced expression of gluconeogenic and lipid homeostasis proteins in liver. Bap1 loss increased pancreatitis biomarkers and reduced expression of mitochondrial proteins. These alterations accompany a metabolic remodeling with hypoglycemia, hypercholesterolemia, hepatic lipid loss, and acinar cell degeneration. Liver-specific Bap1 null mice present with fully penetrant perinatal lethality, severe hypoglycemia, and hepatic lipid deficiency. This work reveals Bap1 as a metabolic regulator in liver and pancreas, and it establishes NeuCode as a reliable proteomic method for deciphering in vivo biology.


Asunto(s)
Proteómica/métodos , Proteínas Supresoras de Tumor/fisiología , Ubiquitina Tiolesterasa/fisiología , Animales , Hematopoyesis , Histonas/metabolismo , Marcaje Isotópico , Metabolismo de los Lípidos , Lisina/metabolismo , Masculino , Redes y Vías Metabólicas , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias Hepáticas/metabolismo , Páncreas/metabolismo , Proteoma/metabolismo , Ubiquitinación
8.
Nature ; 518(7539): 417-21, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25470037

RESUMEN

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.


Asunto(s)
Interleucina-17/biosíntesis , Biosíntesis de Proteínas , Células Th17/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Animales , Estabilidad de Enzimas , Femenino , Inflamación/genética , Inflamación/patología , Intestino Delgado/metabolismo , Intestino Delgado/patología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Transducción de Señal , Especificidad por Sustrato , Factor de Crecimiento Transformador beta/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteasas Ubiquitina-Específicas/biosíntesis , Proteasas Ubiquitina-Específicas/deficiencia , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación
9.
Nature ; 509(7499): 235-9, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24739962

RESUMEN

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.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Escherichia coli/inmunología , Listeria monocytogenes/inmunología , Proteínas/metabolismo , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Animales , Autoinmunidad/genética , Células de la Médula Ósea/citología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/patología , Retículo Endoplásmico/enzimología , Retículo Endoplásmico/metabolismo , Femenino , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Proteínas de Unión a TGF-beta Latente , Macrófagos/citología , Macrófagos/enzimología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Proteínas de la Membrana , Ratones , NADPH Oxidasas/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Fagocitos/citología , Fagocitos/inmunología , Fagocitos/metabolismo , Proteínas/genética , Especies Reactivas de Oxígeno/metabolismo
10.
Nature ; 506(7489): 456-62, 2014 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-24553140

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 3/metabolismo , Enfermedad de Crohn/genética , Polimorfismo de Nucleótido Simple/genética , Proteolisis , Secuencias de Aminoácidos , Animales , Autofagia/genética , Proteínas Relacionadas con la Autofagia , Proteínas Portadoras/química , Caspasa 3/deficiencia , Caspasa 3/genética , Línea Celular , Células Cultivadas , Enfermedad de Crohn/patología , Citocinas/inmunología , Activación Enzimática , Femenino , Privación de Alimentos , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mutagénesis Sitio-Dirigida , Estrés Fisiológico , Yersinia enterocolitica/inmunología
11.
Infect Immun ; 82(2): 773-82, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24478091

RESUMEN

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.


Asunto(s)
Aminas/metabolismo , Bordetella pertussis/patogenicidad , Fármacos Cardiovasculares/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Toxina del Pertussis/metabolismo , Adenosina Difosfato Ribosa/metabolismo , Animales , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Ratones , Ratones Noqueados
12.
Ann Neurol ; 75(1): 50-66, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24027119

RESUMEN

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.


Asunto(s)
Autoinmunidad/fisiología , Encefalomielitis Autoinmune Experimental/enzimología , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Células Mieloides/enzimología , Caracteres Sexuales , Animales , Células Cultivadas , Encefalomielitis Autoinmune Experimental/inmunología , Activación Enzimática/fisiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteína Quinasa 14 Activada por Mitógenos/inmunología , Células Mieloides/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
PLoS One ; 8(7): e62743, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23894272

RESUMEN

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.


Asunto(s)
Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Predisposición Genética a la Enfermedad/genética , Receptores Histamínicos H3/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Secuencia de Aminoácidos , Animales , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Regulación de la Expresión Génica , Hematopoyesis/genética , Hematopoyesis/inmunología , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Espacio Intracelular/metabolismo , Ganglios Linfáticos/inmunología , Masculino , Ratones , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Estructura Terciaria de Proteína , Receptores Histamínicos H3/química
14.
J Immunol ; 191(2): 614-22, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23772030

RESUMEN

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.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Histamina/metabolismo , Histidina Descarboxilasa/genética , Receptores Histamínicos/genética , Receptores Histamínicos/metabolismo , Animales , Células Presentadoras de Antígenos , Barrera Hematoencefálica/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular , Células Cultivadas , Citocinas/biosíntesis , Encefalomielitis Autoinmune Experimental/metabolismo , Histidina Descarboxilasa/deficiencia , Histidina Descarboxilasa/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/inmunología , Glicoproteína Mielina-Oligodendrócito/farmacología , Fragmentos de Péptidos/farmacología , Receptores Histamínicos/deficiencia , Transducción de Señal
15.
Cell Immunol ; 278(1-2): 27-34, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23121973

RESUMEN

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.


Asunto(s)
Antígenos CD1/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Receptores Histamínicos H1/inmunología , Animales , Antígenos CD1/genética , Citocinas/biosíntesis , Citocinas/inmunología , Células Dendríticas/inmunología , Células Dendríticas/patología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Expresión Génica , Predisposición Genética a la Enfermedad , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Especificidad de Órganos , Receptores Histamínicos H1/deficiencia , Receptores Histamínicos H1/genética , Índice de Severidad de la Enfermedad , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T/inmunología , Linfocitos T/patología
16.
Science ; 337(6101): 1541-6, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22878500

RESUMEN

De-ubiquitinating enzyme BAP1 is mutated in a hereditary cancer syndrome with increased risk of mesothelioma and uveal melanoma. Somatic BAP1 mutations occur in various malignancies. We show that mouse Bap1 gene deletion is lethal during embryogenesis, but systemic or hematopoietic-restricted deletion in adults recapitulates features of human myelodysplastic syndrome (MDS). Knockin mice expressing BAP1 with a 3xFlag tag revealed that BAP1 interacts with host cell factor-1 (HCF-1), O-linked N-acetylglucosamine transferase (OGT), and the polycomb group proteins ASXL1 and ASXL2 in vivo. OGT and HCF-1 levels were decreased by Bap1 deletion, indicating a critical role for BAP1 in stabilizing these epigenetic regulators. Human ASXL1 is mutated frequently in chronic myelomonocytic leukemia (CMML) so an ASXL/BAP1 complex may suppress CMML. A BAP1 catalytic mutation found in a MDS patient implies that BAP1 loss of function has similar consequences in mice and humans.


Asunto(s)
Transformación Celular Neoplásica , Genes Supresores de Tumor , Leucemia Mielomonocítica Crónica/genética , Síndromes Mielodisplásicos/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/genética , Animales , Trasplante de Médula Ósea , Inmunoprecipitación de Cromatina , Desarrollo Embrionario , Eliminación de Gen , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Hematopoyesis , Factor C1 de la Célula Huésped/metabolismo , Humanos , Leucemia Mielomonocítica Crónica/metabolismo , Leucemia Mielomonocítica Crónica/patología , Ratones , Ratones Noqueados , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/patología , Células Mieloides/citología , Células Mieloides/fisiología , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/fisiología , N-Acetilglucosaminiltransferasas/metabolismo , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/química , Ubiquitina Tiolesterasa/metabolismo
17.
Eur J Immunol ; 42(6): 1536-46, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22678907

RESUMEN

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.


Asunto(s)
Encefalomielitis Autoinmune Experimental/etiología , Esclerosis Múltiple/etiología , Receptores Histamínicos/fisiología , Animales , Linfocitos T CD4-Positivos/inmunología , Polaridad Celular , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Acoplados a Proteínas G/fisiología , Receptores Histamínicos H1/fisiología , Receptores Histamínicos H2/fisiología , Receptores Histamínicos H3/fisiología , Receptores Histamínicos H4
18.
Cell Immunol ; 272(2): 269-74, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22078271

RESUMEN

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.


Asunto(s)
Infecciones por Coxsackievirus/inmunología , Enterovirus Humano B/inmunología , Miocarditis/inmunología , Receptores Histamínicos H1/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Infecciones por Coxsackievirus/metabolismo , Susceptibilidad a Enfermedades , Histamina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Miocarditis/metabolismo , Receptores Histamínicos H1/deficiencia , Receptores Histamínicos H1/metabolismo , Transducción de Señal , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/metabolismo , Células TH1/inmunología , Células TH1/metabolismo , Replicación Viral/inmunología
19.
J Immunol ; 188(2): 541-7, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22147765

RESUMEN

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.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Receptores Acoplados a Proteínas G/fisiología , Receptores Histamínicos/fisiología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Animales , Barrera Hematoencefálica/inmunología , Recuento de Linfocito CD4 , Permeabilidad de la Membrana Celular/genética , Permeabilidad de la Membrana Celular/inmunología , Células Cultivadas , Encefalomielitis Autoinmune Experimental/genética , Glicoproteínas/administración & dosificación , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Glicoproteína Mielina-Oligodendrócito , Neuronas/inmunología , Neuronas/patología , Fragmentos de Péptidos/administración & dosificación , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Receptores Histamínicos/deficiencia , Receptores Histamínicos/genética , Receptores Histamínicos H4 , Índice de Severidad de la Enfermedad , Linfocitos T Reguladores/metabolismo
20.
Nat Immunol ; 12(12): 1238-45, 2011 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-22001828

RESUMEN

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.


Asunto(s)
Interleucinas/biosíntesis , Proteínas Proto-Oncogénicas c-maf/metabolismo , Células Th17/inmunología , Factor de Crecimiento Transformador beta/farmacología , Animales , Secuencia de Bases , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Sitios de Unión/genética , Células Cultivadas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Interleucinas/genética , Ratones , Ratones Endogámicos BALB C , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Motivos de Nucleótidos , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-maf/genética , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Células Th17/efectos de los fármacos , Transcripción Genética , Interleucina-22
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...