Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 10(1): 2924, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31266950

RESUMEN

Fas induces apoptosis in activated T cell to maintain immune homeostasis, but the effects of non-apoptotic Fas signaling on T cells remain unclear. Here we show that Fas promotes TH9 cell differentiation by activating NF-κB via Ca2+-dependent PKC-ß activation. In addition, PKC-ß also phosphorylates p38 to inactivate NFAT1 and reduce NFAT1-NF-κB synergy to promote the Fas-induced TH9 transcription program. Fas ligation exacerbates inflammatory bowel disease by increasing TH9 cell differentiation, and promotes antitumor activity in p38 inhibitor-treated TH9 cells. Furthermore, low-dose p38 inhibitor suppresses tumor growth without inducing systemic adverse effects. In patients with tumor, relatively high TH9 cell numbers are associated with good prognosis. Our study thus implicates Fas in CD4+ T cells as a target for inflammatory bowel disease therapy. Furthermore, simultaneous Fas ligation and low-dose p38 inhibition may be an effective approach for TH9 cell induction and cancer therapy.


Asunto(s)
Diferenciación Celular , Enfermedades Inflamatorias del Intestino/inmunología , Transducción de Señal , Linfocitos T Reguladores/citología , Receptor fas/inmunología , Animales , Citocinas/genética , Citocinas/inmunología , Femenino , Humanos , Enfermedades Inflamatorias del Intestino/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , FN-kappa B/genética , FN-kappa B/inmunología , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/inmunología , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/terapia , Proteína Quinasa C beta/genética , Proteína Quinasa C beta/inmunología , Linfocitos T Reguladores/inmunología , Receptor fas/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
2.
Immunity ; 48(6): 1144-1159.e5, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29884460

RESUMEN

PKCß-null (Prkcb-/-) mice are severely immunodeficient. Here we show that mice whose B cells lack PKCß failed to form germinal centers and plasma cells, which undermined affinity maturation and antibody production in response to immunization. Moreover, these mice failed to develop plasma cells in response to viral infection. At the cellular level, we have shown that Prkcb-/- B cells exhibited defective antigen polarization and mTORC1 signaling. While altered antigen polarization impaired antigen presentation and likely restricted the potential of GC development, defective mTORC1 signaling impaired metabolic reprogramming, mitochondrial remodeling, and heme biosynthesis in these cells, which altogether overwhelmingly opposed plasma cell differentiation. Taken together, our study reveals mechanistic insights into the function of PKCß as a key regulator of B cell polarity and metabolic reprogramming that instructs B cell fate.


Asunto(s)
Linfocitos B/inmunología , Diferenciación Celular/inmunología , Activación de Linfocitos/inmunología , Células Plasmáticas/inmunología , Proteína Quinasa C beta/inmunología , Animales , Hemo/biosíntesis , Ratones , Ratones Noqueados , Mitocondrias/inmunología , Mitocondrias/metabolismo , Células Plasmáticas/citología
3.
Sci Rep ; 6: 22114, 2016 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-26911897

RESUMEN

Protein kinase C (PKC) plays a regulatory role in key pathways in cancer. However, since phosphorylation is a step for classical PKC (cPKC) maturation and does not correlate with activation, there is a lack of tools to detect active PKC in tissue samples. Here, a structure-based rational approach was used to select a peptide to generate an antibody that distinguishes active from inactive cPKC. A peptide conserved in all cPKCs, C2Cat, was chosen since modeling studies based on a crystal structure of PKCß showed that it is localized at the interface between the C2 and catalytic domains of cPKCs in an inactive kinase. Anti-C2Cat recognizes active cPKCs at least two-fold better than inactive kinase in ELISA and immunoprecipitation assays, and detects the temporal dynamics of cPKC activation upon receptor or phorbol stimulation. Furthermore, the antibody is able to detect active PKC in human tissue. Higher levels of active cPKC were observed in the more aggressive triple negative breast cancer tumors as compared to the less aggressive estrogen receptor positive tumors. Thus, this antibody represents a reliable, hitherto unavailable and a valuable tool to study PKC activation in cells and tissues. Similar structure-based rational design strategies can be broadly applied to obtain active-state specific antibodies for other signal transduction molecules.


Asunto(s)
Anticuerpos/metabolismo , Neoplasias de la Mama/metabolismo , Neuroblastoma/metabolismo , Proteína Quinasa C beta/metabolismo , Sitios de Unión/inmunología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Carcinogénesis , Línea Celular Tumoral , Activación Enzimática , Femenino , Humanos , Isoenzimas/inmunología , Estadificación de Neoplasias , Neuroblastoma/inmunología , Neuroblastoma/patología , Fragmentos de Péptidos/inmunología , Conformación Proteica , Dominios Proteicos/genética , Proteína Quinasa C beta/genética , Proteína Quinasa C beta/inmunología , Receptores de Estrógenos/metabolismo , Transducción de Señal , Relación Estructura-Actividad
4.
Immunol Lett ; 168(1): 31-40, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26343793

RESUMEN

ES-62, a glycoprotein secreted by the filarial nematode Acanthocheilonema viteae, has been shown to modulate the immune system through subversion of signal transduction pathways operating in various immune system cells. With respect to human bone marrow-derived mast cells (BMMCs), ES-62 was previously shown to inhibit FcϵRI-mediated mast cell functional responses such as degranulation and pro-inflammatory cytokine release through a mechanism involving the degradation of PKC-α. At the same time, it was noted that the worm product was able to degrade certain other PKC isoforms but the significance of this was uncertain. In this study, we have employed PKC isoform KO mice to investigate the role of PKC-α, -ß -ϵ, and -θ in mouse BMMCs in order to establish their involvement in mast cell-mediated responses and also, if their absence impacts on ES-62's activity. The data obtained support that in response to antigen cross-linking of IgE bound to FcϵRI, pro-inflammatory cytokine release is controlled in part by a partnership between one conventional and one novel isoform with PKC-α and -θ acting as positive regulators of IL-6 and TNF-α production, while PKC-ß and ϵ act as negative regulators of such cytokines. Furthermore, ES-62 appears to target certain other PKC isoforms in addition to PKC-α to inhibit cytokine release and this may enable it to more efficiently inhibit mast cell responses.


Asunto(s)
Células de la Médula Ósea/inmunología , Proteínas del Helminto/inmunología , Mastocitos/inmunología , Proteína Quinasa C/inmunología , Animales , Western Blotting , Células de la Médula Ósea/enzimología , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Proteínas del Helminto/metabolismo , Interleucina-6/inmunología , Interleucina-6/metabolismo , Isoenzimas/genética , Isoenzimas/inmunología , Isoenzimas/metabolismo , Masculino , Mastocitos/enzimología , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Proteína Quinasa C beta/genética , Proteína Quinasa C beta/inmunología , Proteína Quinasa C beta/metabolismo , Proteína Quinasa C-alfa/genética , Proteína Quinasa C-alfa/inmunología , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C-epsilon/genética , Proteína Quinasa C-epsilon/inmunología , Proteína Quinasa C-epsilon/metabolismo , Receptores de IgE/inmunología , Receptores de IgE/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
5.
J Autoimmun ; 61: 17-28, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26005049

RESUMEN

Recently we identified in a wide spectrum of autoimmune diseases frequently occurring proinflammatory autoantibodies directed against progranulin, a direct inhibitor of TNFR1 & 2 and of DR3. In the present study we investigated the mechanisms for the breakdown of self-tolerance against progranulin. Isoelectric focusing identified a second, differentially electrically charged progranulin isoform exclusively present in progranulin-antibody-positive patients. Alkaline phosphatase treatment revealed this additional progranulin isoform to be hyperphosphorylated. Subsequently Ser81, which is located within the epitope region of progranulin-antibodies, was identified as hyperphosphorylated serine residue by site directed mutagenesis of candidate phosphorylation sites. Hyperphosphorylated progranulin was detected exclusively in progranulin-antibody-positive patients during the courses of their diseases. The occurrence of hyperphosphorylated progranulin preceded seroconversions of progranulin-antibodies, indicating adaptive immune response. Utilizing panels of kinase and phosphatase inhibitors, PKCß1 was identified as the relevant kinase and PP1 as the relevant phosphatase for phosphorylation and dephosphorylation of Ser81. In contrast to normal progranulin, hyperphosphorylated progranulin interacted exclusively with inactivated (pThr320) PP1, suggesting inactivated PP1 to cause the detectable occurrence of phosphorylated Ser81 PGRN. Investigation of possible functional alterations of PGRN due to Ser81 phosphorylation revealed, that hyperphosphorylation prevents the interaction and thus direct inhibition of TNFR1, TNFR2 and DR3, representing an additional direct proinflammatory effect. Finally phosphorylation of Ser81 PGRN alters the conversion pattern of PGRN. In conclusion, inactivated PP1 induces hyperphosphorylation of progranulin in a wide spectrum of autoimmune diseases. This hyperphosphorylation prevents direct inhibition of TNFR1, TNFR2 and DR3 by PGRN, alters the conversion of PGRN, and is strongly associated with the occurrence of neutralizing, proinflammatory PGRN-antibodies, indicating immunogenicity of this alternative secondary modification.


Asunto(s)
Autoanticuerpos/inmunología , Péptidos y Proteínas de Señalización Intercelular/inmunología , Precursores de Proteínas/inmunología , Serina/inmunología , Animales , Autoanticuerpos/genética , Autoanticuerpos/metabolismo , Sitios de Unión/genética , Western Blotting , Línea Celular , Línea Celular Tumoral , Citometría de Flujo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mutagénesis Sitio-Dirigida , Fosforilación , Progranulinas , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Isoformas de Proteínas/metabolismo , Proteína Quinasa C beta/genética , Proteína Quinasa C beta/inmunología , Proteína Quinasa C beta/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Miembro 25 de Receptores de Factores de Necrosis Tumoral/inmunología , Miembro 25 de Receptores de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Serina/genética , Serina/metabolismo
6.
J Immunol ; 192(11): 5305-13, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24778447

RESUMEN

The cross-regulation of G protein-coupled receptors (GPCRs) plays an important role in the immune response. Studies from several laboratories have suggested that a hierarchy of sensitivities to cross-desensitization exists for the chemoattractant GPCRs. We carried out experiments to study the capacity of the formyl peptide receptor-1 (FPR1) to desensitize chemokine receptors CCR1 and CCR2. Our results show that activation of FPR1 resulted in the desensitization and partial internalization of CCR1, but not CCR2, in both primary human monocytes and HEK293 cells coexpressing CCR1, CCR2, and FPR1 (HR1R2F cells). The desensitization of CCR1 by FPR1 stimulation was not due to the simple depletion of the Ca(2+) stores, but was dependent on activation of protein kinase C. Furthermore, we found that the cross-desensitization of CCR1 by FPR1 was associated with CCR1 phosphorylation and moderate reduction of CCR1 cell-surface expression. In contrast, CCR2 was not phosphorylated or internalized after FPR1 activation. Additional studies showed that optimal cross talk between FPR1 and CCR1 was dependent on the functional activity of protein kinase Cß. These results provide a mechanistic basis for the capacity of certain GPCR ligands to exert rapid and selective cross-inactivation of other chemoattractant receptors, and suggest that FPR1 is able to exert "traffic control" in the migration of inflammatory cells by rapidly inhibiting the cell responses to potentially "low-priority" chemoattractants such as CCR1 agonists without inhibiting the response to "higher priority" CCR2 chemoattractants.


Asunto(s)
Monocitos/inmunología , Receptores CCR1/inmunología , Receptores CCR2/inmunología , Receptores de Formil Péptido/inmunología , Calcio/inmunología , Regulación de la Expresión Génica/inmunología , Células HEK293 , Humanos , Monocitos/citología , Proteína Quinasa C beta/inmunología
7.
Sci Signal ; 7(313): ra16, 2014 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-24550541

RESUMEN

A major mechanism by which cancers escape control by the immune system is by blocking the differentiation of myeloid cells into dendritic cells (DCs), immunostimulatory cells that activate antitumor T cells. Tumor-dependent activation of signal transducer and activator of transcription 3 (STAT3) signaling in myeloid progenitor cells is thought to cause this block in their differentiation. In addition, a signaling pathway through protein kinase C ßII (PKCßII) is essential for the differentiation of myeloid cells into DCs. We found in humans and mice that breast cancer cells substantially decreased the abundance of PKCßII in myeloid progenitor cells through a mechanism involving the enhanced activation of STAT3 signaling by soluble, tumor-derived factors (TDFs). STAT3 bound to previously undescribed negative regulatory elements within the promoter of PRKCB, which encodes PKCßII. We also found a previously undescribed counter-regulatory mechanism through which the activity of PKCßII inhibited tumor-dependent STAT3 signaling by decreasing the abundance of cell surface receptors, such as cytokine and growth factor receptors, that are activated by TDFs. Together, these data suggest that a previously unrecognized cross-talk mechanism between the STAT3 and PKCßII signaling pathways provides the molecular basis for the tumor-induced blockade in the differentiation of myeloid cells, and suggest that enhancing PKCßII activity may be a therapeutic strategy to alleviate cancer-mediated suppression of the immune system.


Asunto(s)
Neoplasias de la Mama/inmunología , Células Dendríticas/inmunología , Neoplasias Mamarias Animales/inmunología , Proteínas de Neoplasias/inmunología , Proteína Quinasa C beta/inmunología , Factor de Transcripción STAT3/inmunología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Células Dendríticas/metabolismo , Células Dendríticas/patología , Femenino , Humanos , Células K562 , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Animales/terapia , Ratones , Ratones Endogámicos BALB C , Células Progenitoras Mieloides , Proteínas de Neoplasias/genética , Proteína Quinasa C beta/genética , Proteína Quinasa C beta/metabolismo , Elementos de Respuesta/inmunología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología
8.
Inflamm Res ; 62(11): 991-1001, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23974215

RESUMEN

OBJECTIVE AND DESIGN: We designed a study to detect downstream phosphorylation targets of PKCß in MCP-1-induced human monocytes. METHODS: Two-dimensional gel electrophoresis was performed for monocytes treated with MCP-1 in the presence or absence of PKCß antisense oligodeoxyribonucleotides (AS-ODN) or a PKCß inhibitor peptide, followed by phospho- and total protein staining. Proteins that stained less intensely with the phospho-stain, when normalized to the total protein stain, in the presence of PKCß AS-ODN or the PKCß inhibitor peptide, were sequenced. RESULTS: Of the proteins identified, vimentin was consistently identified using both experimental approaches. Upon (32)P-labeling and vimentin immunoprecipitation, increased phosphorylation of vimentin was observed in MCP-1 treated monocytes as compared to the untreated monocytes. Both PKCß AS-ODN and the PKCß inhibitor reduced MCP-1-induced vimentin phosphorylation. The IP of monocytes with anti-vimentin antibody and immunoblotting with a PKCß antibody revealed that increased PKCß becomes associated with vimentin upon MCP-1 activation. Upon MCP-1 treatment, monocytes were shown to secrete vimentin and secretion depended on PKCß expression and activity. CONCLUSIONS: We conclude that vimentin, a major intermediate filament protein, is a phosphorylation target of PKCß in MCP-1-treated monocytes and that PKCß phosphorylation is essential for vimentin secretion. Our recently published studies have implicated vimentin as a potent stimulator of the innate immune receptor Dectin-1 as reported by Thiagarajan et al. (Cardiovasc Res 99:494-504, 2013). Taken together our findings suggest that inhibition of PKCß regulates vimentin secretion and, thereby, its interaction with Dectin-1 and downstream stimulation of superoxide anion production. Thus, PKCß phosphorylation of vimentin likely plays an important role in propagating inflammatory responses.


Asunto(s)
Quimiocina CCL2/inmunología , Monocitos/inmunología , Proteína Quinasa C beta/inmunología , Vimentina/inmunología , Células Cultivadas , Humanos , Fosforilación
9.
Proc Natl Acad Sci U S A ; 110(36): 14735-40, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-23959874

RESUMEN

Disruption of the blood-brain barrier (BBB) is a hallmark of acute inflammatory lesions in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis. This disruption may precede and facilitate the infiltration of encephalitogenic T cells. The signaling events that lead to this BBB disruption are incompletely understood but appear to involve dysregulation of tight-junction proteins such as claudins. Pharmacological interventions aiming at stabilizing the BBB in MS might have therapeutic potential. Here, we show that the orally available small molecule LY-317615, a synthetic bisindolylmaleimide and inhibitor of protein kinase Cß, which is clinically under investigation for the treatment of cancer, suppresses the transmigration of activated T cells through an inflamed endothelial cell barrier, where it leads to the induction of the tight-junction molecules zona occludens-1, claudin 3, and claudin 5 and other pathways critically involved in transendothelial leukocyte migration. Treatment of mice with ongoing experimental autoimmune encephalomyelitis with LY-317615 ameliorates inflammation, demyelination, axonal damage, and clinical symptoms. Although LY-317615 dose-dependently suppresses T-cell proliferation and cytokine production independent of antigen specificity, its therapeutic effect is abrogated in a mouse model requiring pertussis toxin. This abrogation indicates that the anti-inflammatory and clinical efficacy is mainly mediated by stabilization of the BBB, thus suppressing the transmigration of encephalitogenic T cells. Collectively, our data suggest the involvement of endothelial protein kinase Cß in stabilizing the BBB in autoimmune neuroinflammation and imply a therapeutic potential of BBB-targeting agents such as LY-317615 as therapeutic approaches for MS.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/prevención & control , Indoles/farmacología , Proteína Quinasa C beta/antagonistas & inhibidores , Animales , Barrera Hematoencefálica/inmunología , Proliferación Celular/efectos de los fármacos , Claudina-3/inmunología , Claudina-3/metabolismo , Claudina-5/inmunología , Claudina-5/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/prevención & control , Relación Dosis-Respuesta a Droga , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Femenino , Perfilación de la Expresión Génica , Inmunohistoquímica , Indoles/inmunología , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Microscopía Confocal , Proteína Quinasa C beta/inmunología , Proteína Quinasa C beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/inmunología , Uniones Estrechas/metabolismo , Migración Transendotelial y Transepitelial/efectos de los fármacos , Migración Transendotelial y Transepitelial/inmunología , Proteína de la Zonula Occludens-1/inmunología , Proteína de la Zonula Occludens-1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...