Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Intervalo de año de publicación
1.
Front Immunol ; 12: 695947, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34168658

RESUMEN

Multiple Sclerosis (MS) is a debilitating central nervous system disorder associated with inflammatory T cells. Activation and expansion of inflammatory T cells is thought to be behind MS relapses and influence disease severity. Protein arginine N-methyltransferase 5 (PRMT5) is a T cell activation-induced enzyme that symmetrically dimethylates proteins and promotes T cell proliferation. However, the mechanism behind PRMT5-mediated control of T cell proliferation and whether PRMT5 contributes to diseases severity is unclear. Here, we evaluated the role of PRMT5 on cyclin/cdk pairs and cell cycle progression, as well as PRMT5's link to disease severity in an animal model of relapsing-remitting MS. Treatment of T helper 1 (mTh1) cells with the selective PRMT5 inhibitor, HLCL65, arrested activation-induced T cell proliferation at the G1 stage of the cell cycle, suggesting PRMT5 promotes cell cycle progression in CD4+ T cells. The Cyclin E1/Cdk2 pair promoting G1/S progression was also decreased after PRMT5 inhibition, as was the phosphorylation of retinoblastoma. In the SJL mouse relapsing-remitting model of MS, the highest PRMT5 expression in central nervous system-infiltrating cells corresponded to peak and relapse timepoints. PRMT5 expression also positively correlated with increasing CD4 Th cell composition, disease severity and Cyclin E1 expression. These data indicate that PRMT5 promotes G1/S cell cycle progression and suggest that this effect influences disease severity and/or progression in the animal model of MS. Modulating PRMT5 levels may be useful for controlling T cell expansion in T cell-mediated diseases including MS.


Asunto(s)
Ciclo Celular , Proliferación Celular , Ciclina E/metabolismo , Encefalomielitis Autoinmune Experimental/enzimología , Esclerosis Múltiple Recurrente-Remitente/enzimología , Proteínas Oncogénicas/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Células TH1/enzimología , Animales , Quinasa 2 Dependiente de la Ciclina/metabolismo , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Genes Codificadores de los Receptores de Linfocitos T , Ratones Transgénicos , Esclerosis Múltiple Recurrente-Remitente/inmunología , Esclerosis Múltiple Recurrente-Remitente/patología , Fosforilación , Proteína de Retinoblastoma/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal , Células TH1/inmunología , Células TH1/patología
2.
J Clin Invest ; 130(4): 1683-1698, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32091410

RESUMEN

Protein arginine methyltransferase 5 (PRMT5) catalyzes symmetric dimethylation (SDM) of arginine, a posttranslational modification involved in oncogenesis and embryonic development. However, the role and mechanisms by which PRMT5 modulates Th cell polarization and autoimmune disease have not yet been elucidated. Here, we found that PRMT5 promoted SREBP1 SDM and the induction of cholesterol biosynthetic pathway enzymes that produce retinoid-related orphan receptor (ROR) agonists that activate RORγt. Specific loss of PRMT5 in the CD4+ Th cell compartment suppressed Th17 differentiation and protected mice from developing experimental autoimmune encephalomyelitis (EAE). We also found that PRMT5 controlled thymic and peripheral homeostasis in the CD4+ Th cell life cycle and invariant NK (iNK) T cell development and CD8+ T cell maintenance. This work demonstrates that PRMT5 expression in recently activated T cells is necessary for the cholesterol biosynthesis metabolic gene expression program that generates RORγt agonistic activity and promotes Th17 differentiation and EAE. These results point to Th PRMT5 and its downstream cholesterol biosynthesis pathway as promising therapeutic targets in Th17-mediated diseases.


Asunto(s)
Autoinmunidad , Diferenciación Celular/inmunología , Colesterol/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Proteína-Arginina N-Metiltransferasas/inmunología , Células Th17/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Diferenciación Celular/genética , Colesterol/genética , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Ratones , Ratones Transgénicos , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/patología , 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/inmunología , Proteína-Arginina N-Metiltransferasas/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/inmunología , Células Th17/patología
3.
Front Immunol ; 10: 524, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30941147

RESUMEN

Multiple sclerosis is an autoimmune disease of the central nervous system (CNS) mediated by CD4+ T cells and modeled via experimental autoimmune encephalomyelitis (EAE). Inhibition of PRMT5, the major Type II arginine methyltransferase, suppresses pathogenic T cell responses and EAE. PRMT5 is transiently induced in proliferating memory inflammatory Th1 cells and during EAE. However, the mechanisms driving PRMT5 protein induction and repression as T cells expand and return to resting is currently unknown. Here, we used naive mouse and memory mouse and human Th1/Th2 cells as models to identify mechanisms controlling PRMT5 protein expression in initial and recall T cell activation. Initial activation of naive mouse T cells resulted in NF-κB-dependent transient Prmt5 transcription and NF-κB, mTOR and MYC-dependent PRMT5 protein induction. In murine memory Th cells, transcription and miRNA loss supported PRMT5 induction to a lesser extent than in naive T cells. In contrast, NF-κB/MYC/mTOR-dependent non-transcriptional PRMT5 induction played a major role. These results highlight the importance of the NF-κB/mTOR/MYC axis in PRMT5-driven pathogenic T cell expansion and may guide targeted therapeutic strategies for MS.


Asunto(s)
Activación de Linfocitos/genética , FN-kappa B/genética , Proteína-Arginina N-Metiltransferasas/genética , Proteínas Proto-Oncogénicas c-myc/genética , Serina-Treonina Quinasas TOR/genética , Transcripción Genética/genética , Animales , Línea Celular , Encefalomielitis Autoinmune Experimental/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Esclerosis Múltiple/genética , Células TH1/fisiología , Células Th2/fisiología
4.
J Immunol ; 198(4): 1439-1451, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28087667

RESUMEN

In the autoimmune disease multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE), expansion of pathogenic, myelin-specific Th1 cell populations drives active disease; selectively targeting this process may be the basis for a new therapeutic approach. Previous studies have hinted at a role for protein arginine methylation in immune responses, including T cell-mediated autoimmunity and EAE. However, a conclusive role for the protein arginine methyltransferase (PRMT) enzymes that catalyze these reactions has been lacking. PRMT5 is the main PRMT responsible for symmetric dimethylation of arginine residues of histones and other proteins. PRMT5 drives embryonic development and cancer, but its role in T cells, if any, has not been investigated. In this article, we show that PRMT5 is an important modulator of CD4+ T cell expansion. PRMT5 was transiently upregulated during maximal proliferation of mouse and human memory Th cells. PRMT5 expression was regulated upstream by the NF-κB pathway, and it promoted IL-2 production and proliferation. Blocking PRMT5 with novel, highly selective small molecule PRMT5 inhibitors severely blunted memory Th expansion, with preferential suppression of Th1 cells over Th2 cells. In vivo, PRMT5 blockade efficiently suppressed recall T cell responses and reduced inflammation in delayed-type hypersensitivity and clinical disease in EAE mouse models. These data implicate PRMT5 in the regulation of adaptive memory Th cell responses and suggest that PRMT5 inhibitors may be a novel therapeutic approach for T cell-mediated inflammatory disease.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Memoria Inmunológica , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/fisiología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Animales , Citocinas/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/metabolismo , Regulación de la Expresión Génica , Humanos , Inflamación , Interleucina-2/biosíntesis , Interleucina-2/inmunología , Activación de Linfocitos , Metilación , Ratones , FN-kappa B/inmunología , Proteína-Arginina N-Metiltransferasas/genética , Células TH1/inmunología , Células Th2/inmunología , Regulación hacia Arriba
5.
Clin Immunol ; 176: 12-22, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28039018

RESUMEN

Recent studies implicate innate immunity to systemic lupus erythematosus (SLE) pathogenesis. Toll-like receptor (TLR)8 is estrogen-regulated and binds viral ssRNA to stimulate innate immune responses, but recent work indicates that microRNA (miR)-21 within extracellular vesicles (EVs) can also trigger this receptor. Our objective was to examine TLR8 expression/activation to better understand sex-biased responses involving TLR8 in SLE. Our data identify an estrogen response element that promotes STAT1 expression and demonstrate STAT1-dependent transcriptional activation of TLR8 with estrogen stimulation. In lieu of viral ssRNA activation, we explored EV-encapsulated miR-21 as an endogenous ligand and observed induction of both TLR8 and cytokine expression in vitro. Moreover, extracellular miR detection was found predominantly within EVs. Thus, just as a cytokine or chemokine, EV-encapsulated miR-21 can act as an inflammatory signaling molecule, or miRokine, by virtue of being an endogenous ligand of TLR8. Collectively, our data elucidates a novel innate inflammatory pathway in SLE.


Asunto(s)
Estrógenos/metabolismo , Lupus Eritematoso Sistémico/metabolismo , MicroARNs/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/fisiología , Receptor Toll-Like 8/metabolismo , Línea Celular Tumoral , Quimiocinas/metabolismo , Humanos , Inmunidad Innata/inmunología , Inflamación/inmunología , Inflamación/metabolismo , Ligandos , Lupus Eritematoso Sistémico/inmunología , Células MCF-7
6.
PLoS One ; 11(7): e0159724, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27447824

RESUMEN

Inflammatory M1 spectrum macrophages protect from infection but can cause inflammatory disease and tissue damage, whereas alternatively activated/M2 spectrum macrophages reduce inflammation and promote tissue repair. Modulation of macrophage phenotype may be therapeutically beneficial and requires further understanding of the molecular programs that control macrophage differentiation. A potential mechanism by which macrophages differentiate may be through microRNA (miRNA), which bind to messenger RNA and post-transcriptionally modify gene expression, cell phenotype and function. We hypothesized that the inflammation-associated miRNA, miR-155, would be required for typical development of macrophage inflammatory state. miR-155 was rapidly up-regulated over 100-fold in inflammatory M1(LPS + IFN-γ), but not M2(IL-4), macrophages. Inflammatory genes Inos, Il1b and Tnfa and their corresponding protein or enzymatic products were reduced up to 72% in miR-155 knockout mouse M1(LPS + IFN-γ) macrophages, but miR-155 deficiency did not affect expression of the M2-associated gene Arg1 in M2(IL-4) macrophages. Additionally, a miR-155 oligonucleotide inhibitor efficiently suppressed Inos and Tnfa gene expression in wild-type M1(LPS + IFN-γ) macrophages. Comparative transcriptional profiling of unstimulated and M1(LPS + IFN-γ) macrophages derived from wild-type (WT) and miR-155 knockout (KO) mice revealed that half (approximately 650 genes) of the signature we previously identified in WT M1(LPS + IFN-γ) macrophages was dependent on miR-155. Real-Time PCR of independent datasets confirmed that miR-155 contributed to suppression of its validated mRNA targets Inpp5d, Tspan14, Ptprj and Mafb and induction of Inos, Il1b, Tnfa, Il6 and Il12. Overall, these data indicate that miR-155 plays an essential role in driving the inflammatory phenotype of M1(LPS+ IFN-γ) macrophages.


Asunto(s)
Regulación de la Expresión Génica , Inflamación/genética , Macrófagos/metabolismo , MicroARNs/genética , Transcriptoma , Animales , Biomarcadores , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Inflamación/inmunología , Inflamación/metabolismo , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Ratones , Fenotipo , Interferencia de ARN , ARN Mensajero/genética
7.
PLoS One ; 10(12): e0145342, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26699615

RESUMEN

Classically (M1) and alternatively activated (M2) macrophages exhibit distinct phenotypes and functions. It has been difficult to dissect macrophage phenotypes in vivo, where a spectrum of macrophage phenotypes exists, and also in vitro, where low or non-selective M2 marker protein expression is observed. To provide a foundation for the complexity of in vivo macrophage phenotypes, we performed a comprehensive analysis of the transcriptional signature of murine M0, M1 and M2 macrophages and identified genes common or exclusive to either subset. We validated by real-time PCR an M1-exclusive pattern of expression for CD38, G-protein coupled receptor 18 (Gpr18) and Formyl peptide receptor 2 (Fpr2) whereas Early growth response protein 2 (Egr2) and c-Myc were M2-exclusive. We further confirmed these data by flow cytometry and show that M1 and M2 macrophages can be distinguished by their relative expression of CD38 and Egr2. Egr2 labeled more M2 macrophages (~70%) than the canonical M2 macrophage marker Arginase-1, which labels 24% of M2 macrophages. Conversely, CD38 labeled most (71%) in vitro M1 macrophages. In vivo, a similar CD38+ population greatly increased after LPS exposure. Overall, this work defines exclusive and common M1 and M2 signatures and provides novel and improved tools to distinguish M1 and M2 murine macrophages.


Asunto(s)
Biomarcadores/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Inflamación/metabolismo , Activación de Macrófagos/fisiología , Macrófagos/metabolismo , Transcriptoma , Animales , Citometría de Flujo , Técnicas para Inmunoenzimas , Inflamación/inducido químicamente , Inflamación/genética , Lipopolisacáridos/toxicidad , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Fenotipo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
J Biol Chem ; 287(27): 23119-27, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22593584

RESUMEN

Nicotinic acetylcholine receptor (nAChR) cell surface expression levels are modulated during nicotine dependence and multiple disorders of the nervous system, but the mechanisms underlying nAChR trafficking remain unclear. To determine the role of cysteine residues, including their palmitoylation, on neuronal α4 nAChR subunit maturation and cell surface trafficking, the cysteines in the two intracellular regions of the receptor were replaced with serines using site-directed mutagenesis. Palmitoylation is a post-translational modification that regulates membrane receptor trafficking and function. Metabolic labeling with [(3)H]palmitate determined that the cysteine in the cytoplasmic loop between transmembrane domains 1 and 2 (M1-M2) is palmitoylated. When this cysteine is mutated to a serine, producing a depalmitoylated α4 nAChR, total protein expression decreases, but surface expression increases compared with wild-type α4 levels, as determined by Western blotting and enzyme-linked immunoassays, respectively. The cysteines in the M3-M4 cytoplasmic loop do not appear to be palmitoylated, but replacing all of the cysteines in the loop with serines increases total and cell surface expression. When all of the intracellular cysteines in both loops are mutated to serines, there is no change in total expression, but there is an increase in surface expression. Calcium accumulation assays and high affinity binding for [(3)H]epibatidine determined that all mutants retain functional activity. Thus, our results identify a novel palmitoylation site on cysteine 273 in the M1-M2 loop of the α4 nAChR and determine that cysteines in both intracellular loops are regulatory factors in total and cell surface protein expression of the α4ß2 nAChR.


Asunto(s)
Lipoilación/fisiología , Neuronas/fisiología , Transporte de Proteínas/fisiología , Receptores Nicotínicos/metabolismo , Animales , Calcio/metabolismo , Técnicas de Cocultivo , Cisteína/genética , Citoplasma/metabolismo , Células HEK293 , Hipocampo/citología , Humanos , Neuronas/citología , Palmitatos/metabolismo , Palmitatos/farmacología , Cultivo Primario de Células , Estructura Terciaria de Proteína , Ratas , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores Nicotínicos/química , Receptores Nicotínicos/genética , Tritio
9.
J Neurosci ; 26(4): 1179-89, 2006 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-16436605

RESUMEN

Peripheral myelin protein 22 (PMP22) is a tetraspan membrane glycoprotein, the misexpression of which is associated with hereditary demyelinating neuropathies. Myelinating Schwann cells (SCs) produce the highest levels of PMP22, yet the function of the protein in peripheral nerve biology is unresolved. To investigate the potential roles of PMP22, we engineered a novel knock-out (-/-) mouse line by replacing the first two coding exons of pmp22 with the lacZ reporter. PMP22-deficient mice show strong beta-galactosidase reactivity in peripheral nerves, cartilage, intestines, and lungs, whereas phenotypically they display the characteristics of tomaculous neuropathy. In the absence of PMP22, myelination of peripheral nerves is delayed, and numerous axon-SC profiles show loose basal lamina, suggesting altered interactions of the glial cells with the extracellular matrix. The levels of beta4 integrin, a molecule involved in the linkage between SCs and the basal lamina, are severely reduced in nerves of PMP22-deficient mice. During early stages of myelination, PMP22 and beta4 integrin are coexpressed at the cell surface and can be coimmunoprecipitated together with laminin and alpha6 integrin. In agreement, in clone A colonic carcinoma cells, epitope-tagged PMP22 forms a complex with beta4 integrin. Together, these data indicate that PMP22 is a binding partner in the integrin/laminin complex and is involved in mediating the interaction of SCs with the extracellular environment.


Asunto(s)
Integrina alfa6beta4/metabolismo , Proteínas de la Mielina/metabolismo , Células de Schwann/metabolismo , Adenocarcinoma/química , Adenocarcinoma/patología , Animales , Membrana Basal/metabolismo , Membrana Basal/ultraestructura , Sitios de Unión , Línea Celular Tumoral/química , Técnicas de Cocultivo , Neoplasias del Colon/química , Neoplasias del Colon/patología , Exones/genética , Ganglios Espinales/citología , Humanos , Integrina alfa6beta4/química , Operón Lac , Laminina/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Complejos Multiproteicos , Proteínas de la Mielina/química , Proteínas de la Mielina/deficiencia , Proteínas de la Mielina/genética , Proteínas de la Mielina/fisiología , Especificidad de Órganos , Enfermedades del Sistema Nervioso Periférico/genética , Enfermedades del Sistema Nervioso Periférico/metabolismo , Enfermedades del Sistema Nervioso Periférico/patología , Fenotipo , Unión Proteica , Mapeo de Interacción de Proteínas , Ratas , Células de Schwann/ultraestructura , Nervio Ciático/metabolismo , Nervio Ciático/ultraestructura
10.
Mol Biol Cell ; 16(3): 1142-51, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15635102

RESUMEN

Peripheral myelin protein 22 (PMP22) is associated with a subset of hereditary peripheral neuropathies. Although predominantly recognized as a transmembrane constituent of peripheral nerve myelin, PMP22 is localized to epithelial and endothelial cell-cell junctions, where its function remains unknown. In this report, we investigated the role of PMP22 in epithelial biology. Expression of human PMP22 (hPMP22) slows cell growth and induces a flattened morphology in Madin-Darby canine kidney (MDCK) cells. The transepithelial electrical resistance (TER) and paracellular flux of MDCK monolayers are elevated by hPMP22 expression. After calcium switch, peptides corresponding to the second, but not the first, extracellular loop of PMP22 perturb the recovery of TER and paracellular flux. Finally, subsequent to wounding, epithelial monolayers expressing hPMP22 fail to migrate normally. These results indicate that PMP22 is capable of modulating several aspects of epithelial cell biology, including junctional permeability and wound closure.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/patología , Proteínas de la Mielina/metabolismo , Animales , Biotinilación , Western Blotting , Calcio/metabolismo , Comunicación Celular , Línea Celular , Movimiento Celular , Proliferación Celular , Perros , Impedancia Eléctrica , Epitelio/metabolismo , Epitelio/patología , Fase G1 , Crecimiento , Uniones Intercelulares/metabolismo , Microscopía Fluorescente , Vaina de Mielina/metabolismo , Péptidos/química , Permeabilidad , Fase de Descanso del Ciclo Celular , Fase S , Transgenes , Cicatrización de Heridas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA