Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
FASEB J ; 30(8): 2945-58, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27130484

RESUMEN

The bioactive sphingolipid sphingosine-1-phosphate (S1P) and the kinase that produces it have been implicated in inflammatory bowel diseases in mice and humans; however, little is known about the role of the 2 S1P-specific phosphohydrolase isoforms, SGPP1 and SGPP2, which catalyze dephosphorylation of S1P to sphingosine. To elucidate their functions, we generated specific knockout mice. Deletion of Sgpp2, which is mainly expressed in the gastrointestinal tract, significantly reduced dextran sodium sulfate (DSS)-induced colitis severity, whereas deletion of ubiquitously expressed Sgpp1 slightly worsened colitis. Moreover, Sgpp1 deletion enhanced expression of multifunctional proinflammatory cytokines, IL-6, TNF-α, and IL-1ß, activation of the transcription factor signal transducer and activator of transcription 3, and immune cell infiltration into the colon. Conversely, Sgpp2-null mice failed to mount a DSS-induced systemic inflammatory response. Of interest, Sgpp2 deficiency suppressed DSS-induced intestinal epithelial cell apoptosis and improved mucosal barrier integrity. Furthermore, down-regulation of Sgpp2 attenuated LPS-induced paracellular permeability in cultured cells and enhanced expression of the adherens junction protein E-cadherin. Finally, in patients with ulcerative colitis, SGPP2 expression was elevated in colitis tissues relative to that in uninvolved tissues. These results indicate that induction of SGPP2 expression contributes to the pathogenesis of colitis by promoting disruption of the mucosal barrier function. SGPP2 may represent a novel therapeutic target in inflammatory bowel disease.-Huang, W.-C., Liang, J., Nagahashi, M., Avni, D., Yamada, A., Maceyka, M., Wolen, A. R., Kordula, T., Milstien, S., Takabe, K., Oravecz, T., Spiegel, S. Sphingosine-1-phosphate phosphatase 2 promotes disruption of mucosal integrity, and contributes to ulcerative colitis in mice and humans.


Asunto(s)
Colitis Ulcerosa/metabolismo , Mucosa Intestinal/patología , Proteínas de la Membrana/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Animales , Cadherinas , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/genética , Sulfato de Dextran/toxicidad , Regulación hacia Abajo , Humanos , Inflamación/metabolismo , Mucosa Intestinal/enzimología , Lipopolisacáridos/toxicidad , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Permeabilidad , Monoéster Fosfórico Hidrolasas/genética
2.
Am J Physiol Lung Cell Mol Physiol ; 311(5): L1000-L1014, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27663991

RESUMEN

Progressive lung disease with early onset is the main cause of mortality and morbidity in cystic fibrosis patients. Here we report a reduction of sphingosine-1-phosphate (S1P) in the lung of unchallenged Cftrtm1EUR F508del CFTR mutant mice. This correlates with enhanced infiltration by inducible nitric oxide synthase (iNOS)-expressing granulocytes, B cells, and T cells. Furthermore, the ratio of macrophage-derived dendritic cells (MoDC) to conventional dendritic cells (cDC) is higher in mutant mouse lung, consistent with unprovoked inflammation. Oral application of a S1P lyase inhibitor (LX2931) increases S1P levels in mutant mouse tissues. This normalizes the lung MoDC/cDC ratio and reduces B and T cell counts. Lung granulocytes are enhanced, but iNOS expression is reduced in this population. Although lung LyC6+ monocytes are enhanced by LX2931, they apparently do not differentiate to MoDC and macrophages. After challenge with bacterial toxins (LPS-fMLP) we observe enhanced levels of proinflammatory cytokines TNF-α, KC, IFNγ, and IL-12 and the inducible mucin MUC5AC in mutant mouse lung, evidence of deficient resolution of inflammation. LX2931 does not prevent transient inflammation or goblet cell hyperplasia after challenge, but it reduces MUC5AC and proinflammatory cytokine levels toward normal values. We conclude that lung pathology in homozygous mice expressing murine F508del CFTR, which represents the most frequent mutation in CF patients, is characterized by abnormal behavior of infiltrating myeloid cells and delayed resolution of induced inflammation. This phenotype can be partially corrected by a S1P lyase inhibitor, providing a rationale for therapeutic targeting of the S1P signaling pathway in CF patients.


Asunto(s)
Aldehído-Liasas/antagonistas & inhibidores , Fibrosis Quística/tratamiento farmacológico , Inhibidores Enzimáticos/uso terapéutico , Imidazoles/uso terapéutico , Oximas/uso terapéutico , Neumonía/tratamiento farmacológico , Aldehído-Liasas/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Fibrosis Quística/diagnóstico por imagen , Fibrosis Quística/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Imidazoles/farmacología , Lipopolisacáridos/farmacología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Lisofosfolípidos/metabolismo , Ratones Endogámicos C57BL , Mucina 5AC/metabolismo , Mutación/genética , Células Mieloides/efectos de los fármacos , Células Mieloides/metabolismo , N-Formilmetionina Leucil-Fenilalanina/farmacología , Oximas/farmacología , Neumonía/diagnóstico por imagen , Neumonía/patología , Glándulas Salivales/efectos de los fármacos , Glándulas Salivales/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Microtomografía por Rayos X
3.
FASEB J ; 29(12): 5018-28, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26324848

RESUMEN

Sphingosine 1-phosphate (S1P) is a pleiotropic bioactive sphingolipid metabolite that regulates numerous processes important for immune responses. S1P is made within cells and must be transported out of cells to exert its effects through activation of 5 specific cell surface GPCRs in an autocrine or paracrine fashion. Spinster 2 (Spns2) transports S1P out of cells, and its deletion in mice reduces circulating levels of S1P, alters immune cell trafficking, and induces lymphopenia. Here we examined the effects of Spns2 deletion on adaptive immune responses and in autoimmune disease models. Airway inflammation and hypersensitivity as well as delayed-type contact hypersensitivity were attenuated in Spns2(-/-) mice. Similarly, Spns2 deletion reduced dextran sodium sulfate- and oxazolone-induced colitis. Intriguingly, Spns2(-/-) mice were protected from the development of experimental autoimmune encephalopathy, a model of the autoimmune disease multiple sclerosis. Deletion of Spns2 also strongly alleviated disease development in collagen-induced arthritis. These results point to a broad role for Spns2-mediated S1P transport in the initiation and development of adaptive immune related disorders.


Asunto(s)
Proteínas de Transporte de Anión/fisiología , Enfermedades Autoinmunes/fisiopatología , Inflamación/fisiopatología , Animales , Proteínas de Transporte de Anión/genética , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados
4.
J Immunol ; 193(1): 85-95, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24890721

RESUMEN

Lysophospholipids have emerged as biologically important chemoattractants capable of directing lymphocyte development, trafficking, and localization. Lysophosphatidic acid (LPA) is a major lysophospholipid found systemically, and its levels are elevated in certain pathological settings, such as cancer and infections. In this study, we demonstrate that BCR signal transduction by mature murine B cells is inhibited upon LPA engagement of the LPA5 (GPR92) receptor via a Gα12/13-Arhgef1 pathway. The inhibition of BCR signaling by LPA5 manifests by impaired intracellular calcium store release and most likely by interfering with inositol 1,4,5-triphosphate receptor activity. We further show that LPA5 also limits Ag-specific induction of CD69 and CD86 expression and that LPA5-deficient B cells display enhanced Ab responses. Thus, these data show that LPA5 negatively regulates BCR signaling, B cell activation, and immune response. Our findings extend the influence of lysophospholipids on immune function and suggest that alterations in LPA levels likely influence adaptive humoral immunity.


Asunto(s)
Formación de Anticuerpos/fisiología , Linfocitos B/inmunología , Inmunidad Humoral/fisiología , Receptores de Antígenos de Linfocitos B/inmunología , Receptores del Ácido Lisofosfatídico/inmunología , Transducción de Señal/inmunología , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/genética , Antígenos de Diferenciación de Linfocitos T/inmunología , Antígeno B7-2/genética , Antígeno B7-2/inmunología , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Subunidades alfa de la Proteína de Unión al GTP G12-G13/inmunología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Ratones , Ratones Noqueados , Receptores de Antígenos de Linfocitos B/genética , Receptores del Ácido Lisofosfatídico/genética , Transducción de Señal/genética
5.
Ann Rheum Dis ; 74(6): 1317-24, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24618263

RESUMEN

OBJECTIVES: To investigate the role of liver X receptors (LXRs) in experimental skin fibrosis and evaluate their potential as novel antifibrotic targets. METHODS: We studied the role of LXRs in bleomycin-induced skin fibrosis, in the model of sclerodermatous graft-versus-host disease (sclGvHD) and in tight skin-1 (Tsk-1) mice, reflecting different subtypes of fibrotic disease. We examined both LXR isoforms using LXRα-, LXRß- and LXR-α/ß-double-knockout mice. Finally, we investigated the effects of LXRs on fibroblasts and macrophages to establish the antifibrotic mode of action of LXRs. RESULTS: LXR activation by the agonist T0901317 had antifibrotic effects in bleomycin-induced skin fibrosis, in the sclGvHD model and in Tsk-1 mice. The antifibrotic activity of LXRs was particularly prominent in the inflammation-driven bleomycin and sclGvHD models. LXRα-, LXRß- and LXRα/ß-double-knockout mice showed a similar response to bleomycin as wildtype animals. Low levels of the LXR target gene ABCA-1 in the skin of bleomycin-challenged and control mice suggested a low baseline activation of the antifibrotic LXR signalling, which, however, could be specifically activated by T0901317. Fibroblasts were not the direct target cells of LXRs agonists, but LXR activation inhibited fibrosis by interfering with infiltration of macrophages and their release of the pro-fibrotic interleukin-6. CONCLUSIONS: We identified LXRs as novel targets for antifibrotic therapies, a yet unknown aspect of these nuclear receptors. Our data suggest that LXR activation might be particularly effective in patients with inflammatory disease subtypes. Activation of LXRs interfered with the release of interleukin-6 from macrophages and, thus, inhibited fibroblast activation and collagen release.


Asunto(s)
Fibroblastos/metabolismo , Interleucina-6/metabolismo , Macrófagos/metabolismo , Receptores Nucleares Huérfanos/genética , Esclerodermia Difusa/metabolismo , Enfermedades de la Piel/metabolismo , Piel/patología , Animales , Antibióticos Antineoplásicos/toxicidad , Bleomicina/toxicidad , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Fibrosis , Humanos , Hidrocarburos Fluorados/farmacología , Receptores X del Hígado , Macrófagos/efectos de los fármacos , Ratones , Ratones Noqueados , Receptores Nucleares Huérfanos/agonistas , Receptores Nucleares Huérfanos/metabolismo , Piel/efectos de los fármacos , Enfermedades de la Piel/inducido químicamente , Sulfonamidas/farmacología
6.
J Immunol ; 191(7): 3789-98, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23997217

RESUMEN

The phenylalanyl-glycyl-glycyl-alanyl-prolyl (FG-GAP) domain plays an important role in protein-protein interactions, including interaction of integrins with their ligands. Integrin-α FG-GAP repeat-containing protein 2 (Itfg2) is a highly conserved protein in vertebrates that carries two FG-GAP domains, but its role in mammalian physiology is unknown. In this article, we show that Itfg2 is an intracellular protein and it plays a critical role in B cell differentiation and development of autoimmunity. Itfg2-deficient mice displayed a phenotype consistent with retention of B cells in the spleen and had a lower concentration of IgG in the blood when compared with wild-type littermates. Itfg2-deficient splenocytes also showed a defect in cell migration in vitro. After immunization with a thymus-dependent Ag, the absence of Itfg2 caused a shift in B cell maturation from the germinal centers to the extrafollicular regions of the spleen and blocked deposition of Ag-specific plasma cells in the bone marrow. In support of hematopoietic cell intrinsic activity of Itfg2, bone marrow transplantation of Itfg2-deficient cells was sufficient to impair germinal center development in wild-type mice. Furthermore, Itfg2 deficiency exacerbated development of autoimmune disease in MRL/lpr lupus-prone mice. These results identify Itfg2 as a novel contributor to B cell differentiation and a negative regulator of the autoimmune response during lupus.


Asunto(s)
Enfermedades Autoinmunes/genética , Linfocitos B/citología , Linfocitos B/metabolismo , Diferenciación Celular/genética , Cadenas alfa de Integrinas/genética , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/metabolismo , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Secuencia de Bases , Diferenciación Celular/inmunología , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Modelos Animales de Enfermedad , Orden Génico , Marcación de Gen , Genotipo , Centro Germinal/inmunología , Centro Germinal/metabolismo , Células Madre Hematopoyéticas/metabolismo , Inmunoglobulinas/sangre , Cadenas alfa de Integrinas/química , Cadenas alfa de Integrinas/metabolismo , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Fenotipo , Alineación de Secuencia , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
7.
Gut ; 63(6): 928-37, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23749550

RESUMEN

OBJECTIVE: Enterochromaffin cell-derived serotonin (5-HT) promotes intestinal inflammation. We tested hypotheses that peripheral tryptophan hydroxylase (TPH) inhibitors, administered orally, block 5-HT biosynthesis and deplete 5-HT from enterochromaffin cells sufficiently to ameliorate intestinal inflammation; moreover, peripheral TPH inhibitors fail to enter the murine enteric nervous system (ENS) or central nervous systems and thus do not affect constitutive gastrointestinal motility. DESIGN: Two peripheral TPH inhibitors, LP-920540 and telotristat etiprate (LX1032; LX1606) were given orally to mice. Effects were measured on 5-HT levels in the gut, blood and brain, 5-HT immunoreactivity in the ENS, gastrointestinal motility and severity of trinitrobenzene sulfonic acid (TNBS)-induced colitis. Quantitation of clinical scores, histological damage and intestinal expression of inflammation-associated cytokines and chemokines with focused microarrays and real-time reverse transcriptase PCR were employed to evaluate the severity of intestinal inflammation. RESULTS: LP-920540 and LX1032 reduced 5-HT significantly in the gut and blood but not in the brain. Neither LP-920540 nor LX1032 decreased 5-HT immunoreactive neurons or fibres in the myenteric plexus and neither altered total gastrointestinal transit time, colonic motility or gastric emptying in mice. In contrast, oral LP-920540 and LX1032 reduced the severity of TNBS-induced colitis; the expression of 24% of 84 genes encoding inflammation-related cytokines and chemokines was lowered at least fourfold and the reduced expression of 17% was statistically significant. CONCLUSIONS: Observations suggest that that peripheral TPH inhibitors uncouple the positive linkage of enterochromaffin cell-derived 5-HT to intestinal inflammation. Because peripheral TPH inhibitors evidently do not enter the murine ENS, they lack deleterious effects on constitutive intestinal motility in mice.


Asunto(s)
Colitis/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Expresión Génica/efectos de los fármacos , Mucosa Intestinal/metabolismo , Neuronas/metabolismo , Serotonina/biosíntesis , Animales , Encéfalo/metabolismo , Colitis/inducido químicamente , Colitis/patología , Citocinas/genética , Motilidad Gastrointestinal/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Plexo Mientérico/metabolismo , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , Pirimidinas/farmacología , Serotonina/sangre , Índice de Severidad de la Enfermedad , Ácido Trinitrobencenosulfónico , Triptófano Hidroxilasa/antagonistas & inhibidores
8.
Blood ; 120(18): 3764-73, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22976955

RESUMEN

Ribosomal protein (RP) mutations in diseases such as 5q- syndrome both disrupt hematopoiesis and increase the risk of developing hematologic malignancy. However, the mechanism by which RP mutations increase cancer risk has remained an important unanswered question. We show here that monoallelic, germline inactivation of the ribosomal protein L22 (Rpl22) predisposes T-lineage progenitors to transformation. Indeed, RPL22 was found to be inactivated in ∼ 10% of human T-acute lymphoblastic leukemias. Moreover, monoallelic loss of Rpl22 accelerates development of thymic lymphoma in both a mouse model of T-cell malignancy and in acute transformation assays in vitro. We show that Rpl22 inactivation enhances transformation potential through induction of the stemness factor, Lin28B. Our finding that Rpl22 inactivation promotes transformation by inducing expression of Lin28B provides the first insight into the mechanistic basis by which mutations in Rpl22, and perhaps some other RP genes, increases cancer risk.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteínas de Unión al ADN/metabolismo , Neoplasias Hematológicas/genética , Proteínas de Unión al ARN/genética , Proteínas Ribosómicas/genética , Linfocitos T/metabolismo , Animales , Ensayo de Cambio de Movilidad Electroforética , Citometría de Flujo , Silenciador del Gen , Neoplasias Hematológicas/metabolismo , Humanos , Immunoblotting , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Blood ; 117(18): 4895-904, 2011 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-21346251

RESUMEN

The human proton coupled folate transporter (PCFT) is involved in low pH-dependent intestinal folate transport. In this report, we describe a new murine model of the hereditary folate malabsorption syndrome that we developed through targeted disruption of the first 3 coding exons of the murine homolog of the PCFT gene. By 4 weeks of age, PCFT-deficient (PCFT(-/-)) mice developed severe macrocytic normochromic anemia and pancytopenia. Immature erythroblasts accumulated in the bone marrow and spleen of PCFT(-/-) mice and failed to differentiate further, showing an increased rate of apoptosis in intermediate erythroblasts and reduced release of reticulocytes. In response to the inefficient hematologic development, the serum of the PCFT(-/-) animals contained elevated concentrations of erythropoietin, soluble transferrin receptor (sCD71), and thrombopoietin. In vivo folate uptake experiments demonstrated a systemic folate deficiency caused by disruption of PCFT-mediated intestinal folate uptake, thus confirming in vivo a critical and nonredundant role of the PCFT protein in intestinal folate transport and erythropoiesis. The PCFT-deficient mouse serves as a model for the hereditary folate malabsorption syndrome and is the most accurate animal model of folate deficiency anemia described to date that closely captures the spectrum of pathology typical of this disease.


Asunto(s)
Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/metabolismo , Transportador de Folato Acoplado a Protón/antagonistas & inhibidores , Transportador de Folato Acoplado a Protón/genética , Animales , Secuencia de Bases , Médula Ósea/patología , Cartilla de ADN/genética , Modelos Animales de Enfermedad , Eritropoyesis/genética , Eritropoyesis/fisiología , Femenino , Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/patología , Marcación de Gen , Humanos , Absorción Intestinal , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Pancitopenia/genética , Pancitopenia/metabolismo , Pancitopenia/patología , Transportador de Folato Acoplado a Protón/deficiencia , Bazo/patología
10.
J Immunol ; 187(2): 664-75, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21690328

RESUMEN

αß and γδ lineage T cells are thought to arise from a common CD4(-)CD8(-) progenitor in the thymus. However, the molecular pathways controlling fate selection and maturation of these two lineages remain poorly understood. We demonstrated recently that a ubiquitously expressed ribosomal protein, Rpl22, is selectively required for the development of αß lineage T cells. Germline ablation of Rpl22 impairs development of αß lineage, but not γδ lineage, T cells through activation of a p53-dependent checkpoint. In this study, we investigate the downstream effectors used by p53 to impair T cell development. We found that many p53 targets were induced in Rpl22(-/-) thymocytes, including miR-34a, PUMA, p21(waf), Bax, and Noxa. Notably, the proapoptotic factor Bim, while not a direct p53 target, was also strongly induced in Rpl22(-/-) T cells. Gain-of-function analysis indicated that overexpression of miR-34a caused a developmental arrest reminiscent of that induced by p53 in Rpl22-deficient T cells; however, only a few p53 targets alleviated developmental arrest when individually ablated by gene targeting or knockdown. Co-elimination of PUMA and Bim resulted in a nearly complete restoration of development of Rpl22(-/-) thymocytes, indicating that p53-mediated arrest is enforced principally through effects on cell survival. Surprisingly, co-elimination of the primary p53 regulators of cell cycle arrest (p21(waf)) and apoptosis (PUMA) actually abrogated the partial rescue caused by loss of PUMA alone, suggesting that the G1 checkpoint protein p21(waf) facilitates thymocyte development in some contexts.


Asunto(s)
Diferenciación Celular/inmunología , Marcación de Gen , Inhibidores de Crecimiento/inmunología , Proteínas Ribosómicas/deficiencia , Subgrupos de Linfocitos T/inmunología , Proteína p53 Supresora de Tumor/fisiología , Animales , Apoptosis/genética , Apoptosis/inmunología , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteína 11 Similar a Bcl2 , Diferenciación Celular/genética , Línea Celular , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Marcación de Gen/métodos , Inhibidores de Crecimiento/genética , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/deficiencia , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/deficiencia , Proteínas Ribosómicas/biosíntesis , Proteínas Ribosómicas/genética , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Timo/inmunología , Timo/metabolismo , Timo/patología , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/deficiencia , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/deficiencia , Proteína X Asociada a bcl-2/biosíntesis
11.
Front Immunol ; 14: 1084887, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37033994

RESUMEN

Background: The immune cell topography of solid tumors has been increasingly recognized as an important predictive factor for progression of disease and response to immunotherapy. The distribution pattern of immune cells in solid tumors is commonly classified into three categories - namely, "Immune inflamed", "Immune desert" and "Immune excluded" - which, to some degree, connect immune cell presence and positioning within the tumor microenvironment to anti-tumor activity. Materials and methods: In this review, we look at the ways immune exclusion has been defined in published literature and identify opportunities to develop consistent, quantifiable definitions, which in turn, will allow better determination of the underlying mechanisms that span cancer types and, ultimately, aid in the development of treatments to target these mechanisms. Results: The definitions of tumor immune phenotypes, especially immune exclusion, have largely been conceptual. The existing literature lacks in consistency when it comes to practically defining immune exclusion, and there is no consensus on a definition. Majority of the definitions use somewhat arbitrary cut-offs in an attempt to place each tumor into a distinct phenotypic category. Tumor heterogeneity is often not accounted for, which limits the practical application of a definition. Conclusions: We have identified two key issues in existing definitions of immune exclusion, establishing clinically relevant cut-offs within the spectrum of immune cell infiltration as well as tumor heterogeneity. We propose an approach to overcome these limitations, by reporting the degree of immune cell infiltration, tying cut-offs to clinically meaningful outcome measures, maximizing the number of regions of a tumor that are analyzed and reporting the degree of heterogeneity. This will allow for a consensus practical definition for operationalizing this categorization into clinical trial and signal-seeking endpoints.


Asunto(s)
Neoplasias , Escape del Tumor , Humanos , Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Microambiente Tumoral/inmunología , Consenso , Escape del Tumor/inmunología
12.
J Immunol ; 185(1): 327-34, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20505138

RESUMEN

Sh2d3c is an adaptor protein that has been implicated in T cell activation and shown to associate with different components of the integrin signaling pathway ex vivo. However, the in vivo significance of Sh2d3c expression in the regulation of the immune response and/or hematopoietic cell lineage development is not known. In this study, we show that expression of Sh2d3c is more critical for development and function of marginal zone B (MZB) cells than for T cell maturation. Mice deficient in Sh2d3c expression (Sh2d3c(-/-)) had a reduced number of MZB cells, and the residual MZB cells failed to properly capture polysaccharide Ags. Activation-induced proliferation, cytokine production, and migration of Sh2d3c(-)(/)(-) splenic B cells were also significantly reduced in vitro compared with wild-type (Sh2d3c(+/+)) cells. In contrast, T cell development and function were largely normal in Sh2d3c(-/-) mice. The thymi of Sh2d3c(-/-) mice showed no maturational abnormalities, the number of splenic T cells was only modestly reduced, and the T cells responded normally to in vitro polyclonal activation. The observed B cell deficiency in the Sh2d3c(-/-) mice led to diminished humoral immune response against thymus-independent type 2, but not thymus-dependent Ags, which highlights the primary in vivo role of Sh2d3c in regulating B cell development and function.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Transducción de Señal/inmunología , Bazo/inmunología , Bazo/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antígenos T-Independientes/inmunología , Subgrupos de Linfocitos B/patología , Señalización del Calcio/genética , Señalización del Calcio/inmunología , Quimiotaxis de Leucocito/genética , Quimiotaxis de Leucocito/inmunología , Femenino , Linfopenia/genética , Linfopenia/inmunología , Linfopenia/patología , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Transducción de Señal/genética , Bazo/patología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Timo/citología , Timo/inmunología , Timo/metabolismo
13.
Transl Res ; 249: 49-73, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35691544

RESUMEN

As an anti-inflammatory strategy, MAPK-activated protein kinase-2 (MK2) inhibition can potentially avoid the clinical failures seen for direct p38 inhibitors, especially tachyphylaxis. CC-99677, a selective targeted covalent MK2 inhibitor, employs a rare chloropyrimidine that bonds to the sulfur of cysteine 140 in the ATP binding site via a nucleophilic aromatic substitutions (SNAr) mechanism. This irreversible mechanism translates biochemical potency to cells shown by potent inhibition of heat shock protein 27 (HSP27) phosphorylation in LPS-activated monocytic THP-1 cells. The cytokine inhibitory profile of CC-99677 differentiates it from known p38 inhibitors, potentially suppressing a p38 pathway inflammatory response while avoiding tachyphylaxis. Dosed orally, CC-99677 is efficacious in a rat model of ankylosing spondylitis. Single doses, 3 to 400 mg, in healthy human volunteers show linear pharmacokinetics and apparent sustained tumor necrosis factor-α inhibition, with a favorable safety profile. These results support further development of CC-99677 for autoimmune diseases like ankylosing spondylitis.


Asunto(s)
Enfermedades Autoinmunes , Espondilitis Anquilosante , Adenosina Trifosfato , Animales , Antiinflamatorios , Enfermedades Autoinmunes/tratamiento farmacológico , Cisteína , Proteínas de Choque Térmico HSP27/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lipopolisacáridos , Proteínas Serina-Treonina Quinasas , Ratas , Azufre , Factor de Necrosis Tumoral alfa , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Mol Med ; 17(7-8): 717-25, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21556483

RESUMEN

Cerebral malaria (CM) is associated with excessive inflammatory responses and endothelial activation. Sphingosine 1-phosphate (S1P) is a signaling sphingolipid implicated in regulating vascular integrity, inflammation and T-cell migration. We hypothesized that altered S1P signaling during malaria contributes to endothelial activation and inflammation, and show that plasma S1P levels were decreased in Ugandan children with CM compared with children with uncomplicated malaria. Using the Plasmodium berghei ANKA (PbA) model of experimental CM (ECM), we demonstrate that humanized S1P lyase (hS1PL)(-/-) mice with reduced S1P lyase activity (resulting in increased bio-available S1P) had improved survival compared with wild-type littermates. Prophylactic and therapeutic treatment of infected mice with compounds that modulate the S1P pathway and are in human trials for other conditions (FTY720 or LX2931) significantly improved survival in ECM. FTY720 treatment improved vascular integrity as indicated by reduced levels of soluble intercellular adhesion molecule (sICAM), increased angiopoietin 1 (Ang1) (regulator of endothelial quiescence) levels, and decreased Evans blue dye leakage into brain parenchyma. Furthermore, treatment with FTY720 decreased IFNγ levels in plasma as well as CD4(+) and CD8(+) T-cell infiltration into the brain. Finally, when administered during infection in combination with artesunate, FTY720 treatment resulted in increased survival to ECM. These findings implicate dysregulation of the S1P pathway in the pathogenesis of human and murine CM and suggest a novel therapeutic strategy to improve clinical outcome in severe malaria.


Asunto(s)
Aldehído-Liasas/metabolismo , Lisofosfolípidos/metabolismo , Malaria Cerebral/metabolismo , Plasmodium berghei/crecimiento & desarrollo , Esfingosina/análogos & derivados , Aldehído-Liasas/antagonistas & inhibidores , Aldehído-Liasas/genética , Animales , Antimaláricos/farmacología , Artemisininas/farmacología , Artesunato , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Moléculas de Adhesión Celular/sangre , Niño , Preescolar , Femenino , Clorhidrato de Fingolimod , Humanos , Imidazoles/farmacología , Inmunosupresores/farmacología , Lactante , Interferón gamma/sangre , Lisofosfolípidos/sangre , Malaria Cerebral/tratamiento farmacológico , Malaria Cerebral/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Oximas/farmacología , Plasmodium berghei/efectos de los fármacos , Glicoles de Propileno/farmacología , Esfingosina/sangre , Esfingosina/metabolismo , Esfingosina/farmacología , Análisis de Supervivencia , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Resultado del Tratamiento
15.
Bioorg Med Chem Lett ; 19(23): 6780-3, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19836229

RESUMEN

A series of potent piperidine-linked cytosine derivatives were prepared as inhibitors of deoxycytidine kinase (dCK). Compound 9h was discovered to be a potent inhibitor of dCK and shows a good combination of cellular potency and pharmacokinetic parameters. Compound 9h blocks the incorporation of radiolabeled cytosine into mouse T-cells in vitro, as well as in vivo in mice following a T-cell challenge.


Asunto(s)
Desoxicitidina Quinasa/antagonistas & inhibidores , Flucitosina/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Animales , Diseño de Fármacos , Flucitosina/síntesis química , Flucitosina/química , Humanos , Ratones , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Estereoisomerismo , Relación Estructura-Actividad
16.
Bioorg Med Chem Lett ; 19(23): 6784-7, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19836232

RESUMEN

A series of deoxycytidine kinase inhibitors was simultaneously optimized for potency and PK properties. A co-crystal structure then allowed merging this series with a high throughput screening hit to afford a highly potent, selective and orally bioavailable inhibitor, compound 10. This compound showed dose dependent inhibition of deoxycytidine kinase in vivo.


Asunto(s)
Desoxicitidina Quinasa/antagonistas & inhibidores , Desoxicitidina/análogos & derivados , Diseño de Fármacos , Inhibidores de Proteínas Quinasas/farmacología , Desoxicitidina/síntesis química , Desoxicitidina/química , Desoxicitidina/farmacología , Relación Dosis-Respuesta a Droga , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Estereoisomerismo , Relación Estructura-Actividad
17.
PLoS One ; 14(8): e0221528, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31437241

RESUMEN

Expressed strongly by myeloid cells, damage-associated molecular pattern (DAMP) proteins S100A8 and S100A9 are found in the serum of patients with infectious and autoimmune diseases. Compared to S100A9, the role of S100A8 is controversial. We investigated its biological activity in collagen-induced arthritis using the first known viable and fertile S100a8-deficient (S100a8-/-) mouse. Although comparable to the wild type (WT) in terms of lymphocyte distribution in blood and in the primary and secondary lymphoid organs, S100a8-/- mice had increased numbers of neutrophils, monocytes and dendritic cells in the blood and bone marrow, and these all expressed myeloid markers such as CD11b, Ly6G and CD86 more strongly. Granulocyte-macrophage common precursors were increased in S100a8-/- bone marrow and yielded greater numbers of macrophages and dendritic cells in culture. The animals also developed more severe arthritic disease leading to aggravated osteoclast activity and bone destruction. These findings were correlated with increased inflammatory cell infiltration and cytokine secretion in the paws. This study suggests that S100A8 is an anti-inflammatory DAMP that regulates myeloid cell differentiation, thereby mitigating the development of experimental arthritis.


Asunto(s)
Artritis Experimental/patología , Calgranulina A/deficiencia , Mielopoyesis , Animales , Artritis Experimental/diagnóstico por imagen , Médula Ósea/patología , Huesos/diagnóstico por imagen , Huesos/patología , Calgranulina A/metabolismo , Cartílago/patología , Diferenciación Celular , Células Dendríticas/metabolismo , Femenino , Eliminación de Gen , Ratones , Células Mieloides/patología
18.
J Leukoc Biol ; 81(4): 860-9, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17289800

RESUMEN

Throughout evolution, mammals have developed an elaborate network of positive and negative regulatory mechanisms, which provide balance between defensive measures against bacterial and viral pathogens and protective measures against unwarranted destruction of the host by the activated immune system. Kinases and phosphatases encompassing the MAPK pathway are key players in the orderly action of pro- and anti-inflammatory processes, forming numerous promiscuous interactions. Several lines of evidence demonstrate that the phosphorylation and activation status of kinases in the MAPK system has crucial impact on the outcome of downstream events that regulate cytokine production. At least 13 members of the family of dual-specificity phosphatases (DUSP) display unique substrate specificities for MAPKs. Despite the considerable amount of information obtained about the contribution of the different DUSP to MAPK-mediated signaling and innate immunity, the interpretation of available data remains problematic. The in vitro and ex vivo findings are often complicated by functional redundancy of signaling molecules and do not always accurately predict the situation in vivo. Until recently, DUSP research has been hampered by the lack of relevant mammalian knockout (KO) models, which is a powerful tool for delineating in vivo function and redundancy in gene families. This situation changed dramatically over the last year, and this review integrates recent insights into the precise biological role of the DUSP family in innate immunity gained from a comprehensive analysis of mammalian KO models.


Asunto(s)
Proteínas de Ciclo Celular/inmunología , Regulación Enzimológica de la Expresión Génica , Proteínas Inmediatas-Precoces/inmunología , Inmunidad Innata , Modelos Biológicos , Fosfoproteínas Fosfatasas/inmunología , Proteínas Tirosina Fosfatasas/inmunología , Animales , Apoptosis , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Movimiento Celular , Fosfatasa 1 de Especificidad Dual , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Noqueados , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Proteína Fosfatasa 1 , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/metabolismo
19.
Mol Cancer Res ; 13(1): 174-85, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25158955

RESUMEN

UNLABELLED: Autotaxin (ENPP2/ATX) and lysophosphatidic acid (LPA) receptors represent two key players in regulating cancer progression. The present study sought to understand the mechanistic role of LPA G protein-coupled receptors (GPCR), not only in the tumor cells but also in stromal cells of the tumor microenvironment. B16F10 melanoma cells predominantly express LPA5 and LPA2 receptors but lack LPA1. LPA dose dependently inhibited invasion of cells across a Matrigel layer. RNAi-mediated knockdown of LPA5 relieved the inhibitory effect of LPA on invasion without affecting basal invasion. This suggests that LPA5 exerts an anti-invasive action in melanoma cells in response to LPA. In addition, both siRNA-mediated knockdown and pharmacologic inhibition of LPA2 reduced the basal rate invasion. Unexpectedly, when probing the role of this GPCR in host tissues, it was found that the incidence of melanoma-derived lung metastasis was greatly reduced in LPA5 knockout (KO) mice compared with wild-type (WT) mice. LPA1-KO but not LPA2-KO mice also showed diminished melanoma-derived lung metastasis, suggesting that host LPA1 and LPA5 receptors play critical roles in the seeding of metastasis. The decrease in tumor cell residence in the lungs of LPA1-KO and LPA5-KO animals was apparent 24 hours after injection. However, KO of LPA1, LPA2, or LPA5 did not affect the subcutaneous growth of melanoma tumors. IMPLICATIONS: These findings suggest that tumor and stromal LPA receptors, in particular LPA1 and LPA5, play different roles in invasion and the seeding of metastasis.


Asunto(s)
Neoplasias Pulmonares/genética , Melanoma Experimental/genética , Receptores del Ácido Lisofosfatídico/genética , Animales , Carcinogénesis/genética , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Melanoma Experimental/patología , Ratones , Ratones Noqueados , Invasividad Neoplásica/genética , Metástasis de la Neoplasia , Hidrolasas Diéster Fosfóricas/genética , Transducción de Señal/genética , Microambiente Tumoral
20.
PLoS One ; 9(5): e98151, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24852423

RESUMEN

Mammalian sterile 20-like kinase 1 (Mst1) is a MAPK kinase kinase kinase which is involved in a wide range of cellular responses, including apoptosis, lymphocyte adhesion and trafficking. The contribution of Mst1 to Ag-specific immune responses and autoimmunity has not been well defined. In this study, we provide evidence for the essential role of Mst1 in T cell differentiation and autoimmunity, using both genetic and pharmacologic approaches. Absence of Mst1 in mice reduced T cell proliferation and IL-2 production in vitro, blocked cell cycle progression, and elevated activation-induced cell death in Th1 cells. Mst1 deficiency led to a CD4+ T cell development path that was biased toward Th2 and immunoregulatory cytokine production with suppressed Th1 responses. In addition, Mst1-/- B cells showed decreased stimulation to B cell mitogens in vitro and deficient Ag-specific Ig production in vivo. Consistent with altered lymphocyte function, deletion of Mst1 reduced the severity of experimental autoimmune encephalomyelitis (EAE) and protected against collagen-induced arthritis development. Mst1-/- CD4+ T cells displayed an intrinsic defect in their ability to respond to encephalitogenic antigens and deletion of Mst1 in the CD4+ T cell compartment was sufficient to alleviate CNS inflammation during EAE. These findings have prompted the discovery of novel compounds that are potent inhibitors of Mst1 and exhibit desirable pharmacokinetic properties. In conclusion, this report implicates Mst1 as a critical regulator of adaptive immune responses, Th1/Th2-dependent cytokine production, and as a potential therapeutic target for immune disorders.


Asunto(s)
Autoinmunidad , Factor de Crecimiento de Hepatocito/genética , Proteínas Proto-Oncogénicas/genética , Linfocitos T/inmunología , Animales , Artritis Reumatoide/inmunología , Secuencia de Bases , Cartilla de ADN , Activación de Linfocitos , Prueba de Cultivo Mixto de Linfocitos , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA