Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Ther ; 31(3): 686-700, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36641624

RESUMEN

Several studies have shown the influence of commensal microbes on T cell function, specifically in the setting of checkpoint immunotherapy for cancer. In this study, we investigated how vancomycin-induced gut microbiota dysbiosis affects chimeric antigen receptor (CAR) T immunotherapy using multiple preclinical models as well as clinical correlates. In two murine tumor models, hematopoietic CD19+-A20 lymphoma and CD19+-B16 melanoma, mice receiving vancomycin in combination with CD19-directed CAR T cell (CART-19) therapy displayed increased tumor control and tumor-associated antigens (TAAs) cross-presentation compared with CART-19 alone. Fecal microbiota transplant from human healthy donors to pre-conditioned mice recapitulated the results obtained in naive gut microbiota mice. Last, B cell acute lymphoblastic leukemia patients treated with CART-19 and exposed to oral vancomycin showed higher CART-19 peak expansion compared with unexposed patients. These results substantiate the role of the gut microbiota on CAR T cell therapy and suggest that modulation of the gut microbiota using vancomycin may improve outcomes after CAR T cell therapy across tumor types.


Asunto(s)
Microbioma Gastrointestinal , Receptores Quiméricos de Antígenos , Humanos , Ratones , Animales , Receptores de Antígenos de Linfocitos T/genética , Reactividad Cruzada , Vancomicina/farmacología , Inmunoterapia , Linfocitos T , Inmunoterapia Adoptiva/métodos , Receptores Quiméricos de Antígenos/genética , Antígenos CD19
2.
Nat Commun ; 12(1): 4365, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34272369

RESUMEN

Activating RAS missense mutations are among the most prevalent genomic alterations observed in human cancers and drive oncogenesis in the three most lethal tumor types. Emerging evidence suggests mutant KRAS (mKRAS) may be targeted immunologically, but mKRAS epitopes remain poorly defined. Here we employ a multi-omics approach to characterize HLA class I-restricted mKRAS epitopes. We provide proteomic evidence of mKRAS epitope processing and presentation by high prevalence HLA class I alleles. Select epitopes are immunogenic enabling mKRAS-specific TCRαß isolation. TCR transfer to primary CD8+ T cells confers cytotoxicity against mKRAS tumor cell lines independent of histologic origin, and the kinetics of lytic activity correlates with mKRAS peptide-HLA class I complex abundance. Adoptive transfer of mKRAS-TCR engineered CD8+ T cells leads to tumor eradication in a xenograft model of metastatic lung cancer. This study validates mKRAS peptides as bona fide epitopes facilitating the development of immune therapies targeting this oncoprotein.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Carcinogénesis/inmunología , Epítopos de Linfocito T/inmunología , Neoplasias Pulmonares/inmunología , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Traslado Adoptivo , Alelos , Animales , Carcinogénesis/genética , Línea Celular Tumoral , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Mutación , Péptidos/genética , Péptidos/inmunología , Proteómica , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cell ; 183(1): 126-142.e17, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32961131

RESUMEN

CD19-directed immunotherapies are clinically effective for treating B cell malignancies but also cause a high incidence of neurotoxicity. A subset of patients treated with chimeric antigen receptor (CAR) T cells or bispecific T cell engager (BiTE) antibodies display severe neurotoxicity, including fatal cerebral edema associated with T cell infiltration into the brain. Here, we report that mural cells, which surround the endothelium and are critical for blood-brain-barrier integrity, express CD19. We identify CD19 expression in brain mural cells using single-cell RNA sequencing data and confirm perivascular staining at the protein level. CD19 expression in the brain begins early in development alongside the emergence of mural cell lineages and persists throughout adulthood across brain regions. Mouse mural cells demonstrate lower levels of Cd19 expression, suggesting limitations in preclinical animal models of neurotoxicity. These data suggest an on-target mechanism for neurotoxicity in CD19-directed therapies and highlight the utility of human single-cell atlases for designing immunotherapies.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Células Epiteliales/metabolismo , Inmunoterapia Adoptiva/efectos adversos , Animales , Anticuerpos Biespecíficos/inmunología , Antígenos CD19/inmunología , Linfocitos B/inmunología , Barrera Hematoencefálica/inmunología , Encéfalo/inmunología , Encéfalo/metabolismo , Línea Celular Tumoral , Citotoxicidad Inmunológica , Humanos , Inmunoterapia/efectos adversos , Inmunoterapia/métodos , Inmunoterapia Adoptiva/métodos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Músculo Liso Vascular/metabolismo , Neoplasias , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Análisis de la Célula Individual/métodos , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Arterioscler Thromb Vasc Biol ; 38(6): 1297-1308, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29674474

RESUMEN

OBJECTIVE: To test the hypothesis that loss of IL-19 (interleukin-19) exacerbates atherosclerosis. APPROACH AND RESULTS: Il19-/- mice were crossed into Ldlr-/- (low-density lipoprotein receptor knock out) mice. Double knockout (dKO) mice had increased plaque burden in aortic arch and root compared with Ldlr-/- controls after 14 weeks of high-fat diet (HFD). dKO mice injected with 10 ng/g per day rmIL-19 had significantly less plaque compared with controls. qRT-PCR and Western blot analysis revealed dKO mice had increased systemic and intraplaque polarization of T cells and macrophages to proinflammatory Th1 and M1 phenotypes, and also significantly increased TNF (tumor necrosis factor)-α expression in spleen and aortic arch compared with Ldlr-/- controls. Bone marrow transplantation suggests that immune cells participate in IL-19 protection. Bone marrow-derived macrophages and vascular smooth muscle cells isolated from dKO mice had a significantly greater expression of inflammatory cytokine mRNA and protein compared with controls. Spleen and aortic arch from dKO mice had significantly increased expression of the mRNA stability protein HuR (human antigen R). Bone marrow-derived macrophage and vascular smooth muscle cell isolated from dKO mice also had greater HuR abundance. HuR stabilizes proinflammatory transcripts by binding AU-rich elements in the 3' untranslated region. Cytokine and HuR mRNA stability were increased in dKO bone marrow-derived macrophage and vascular smooth muscle cell, which was rescued by addition of IL-19 to these cells. IL-19-induced expression of miR133a, which targets and reduced HuR abundance; miR133a levels were lower in dKO mice compared with controls. CONCLUSIONS: These data indicate that IL-19 is an atheroprotective cytokine which decreases the abundance of HuR, leading to reduced inflammatory mRNA stability.


Asunto(s)
Aorta Torácica/metabolismo , Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Proteína 1 Similar a ELAV/metabolismo , Eliminación de Gen , Interleucina-10/deficiencia , Estabilidad del ARN , ARN Mensajero/metabolismo , Receptores de LDL/deficiencia , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/prevención & control , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/prevención & control , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Proteína 1 Similar a ELAV/genética , Femenino , Predisposición Genética a la Enfermedad , Interleucina-10/administración & dosificación , Interleucina-10/genética , Interleucinas , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fenotipo , Placa Aterosclerótica , Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/genética , Receptores de LDL/genética , Factor de Necrosis Tumoral alfa/metabolismo
5.
J Mol Cell Cardiol ; 105: 38-48, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28257760

RESUMEN

The transformation of vascular smooth muscle cells [VSMC] into foam cells leading to increased plaque size and decreased stability is a key, yet understudied step in atherogenesis. We reported that Interleukin-19 (IL-19), a novel, anti-inflammatory cytokine, attenuates atherosclerosis by anti-inflammatory effects on VSMC. In this work we report that IL-19 induces expression of miR133a, a muscle-specific miRNA, in VSMC. Although previously unreported, we report that miR133a can target and reduce mRNA abundance, mRNA stability, and protein expression of Low Density Lipoprotein Receptor Adaptor Protein 1, (LDLRAP1), an adaptor protein which functions to internalize the LDL receptor. Mutations in this gene lead to LDL receptor malfunction and cause the Autosomal Recessive Hypercholesterolemia (ARH) disorder in humans. Herein we show that IL-19 reduces lipid accumulation in VSMC, and LDLRAP1 expression and oxLDL uptake in a miR133a-dependent mechanism. We show that LDLRAP1 is expressed in plaque and neointimal VSMC of mouse and human injured arteries. Transfection of miR133a and LDLRAP1 siRNA into VSMC reduces their proliferation and uptake of oxLDL. miR133a is significantly increased in plasma from hyperlipidemic compared with normolipidemic patients. Expression of miR133a in IL-19 stimulated VSMC represents a previously unrecognized link between vascular lipid metabolism and inflammation, and may represent a therapeutic opportunity to combat vascular inflammatory diseases.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Células Endoteliales/metabolismo , Interleucinas/metabolismo , Lipoproteínas LDL/metabolismo , MicroARNs/genética , Miocitos del Músculo Liso/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular , Proliferación Celular , Células Cultivadas , Colesterol/metabolismo , Regulación de la Expresión Génica , Humanos , Hiperlipidemias/genética , Hiperlipidemias/metabolismo , Ratones , Interferencia de ARN , ARN Mensajero/genética
6.
Am J Physiol Cell Physiol ; 310(11): C931-41, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27053520

RESUMEN

Neovascularization and inflammation are independent biological processes but are linked in response to injury. The role of inflammation-dampening cytokines in the regulation of angiogenesis remains to be clarified. The purpose of this work was to test the hypothesis that IL-19 can induce angiogenesis in the absence of tissue hypoxia and to identify potential mechanisms. Using the aortic ring model of angiogenesis, we found significantly reduced sprouting capacity in aortic rings from IL-19(-/-) compared with wild-type mice. Using an in vivo assay, we found that IL-19(-/-) mice respond to vascular endothelial growth factor (VEGF) significantly less than wild-type mice and demonstrate decreased capillary formation in Matrigel plugs. IL-19 signals through the IL-20 receptor complex, and IL-19 induces IL-20 receptor subunit expression in aortic rings and cultured human vascular smooth muscle cells, but not endothelial cells, in a peroxisome proliferator-activated receptor-γ-dependent mechanism. IL-19 activates STAT3, and IL-19 angiogenic activity in aortic rings is STAT3-dependent. Using a quantitative RT-PCR screening assay, we determined that IL-19 has direct proangiogenic effects on aortic rings by inducing angiogenic gene expression. M2 macrophages participate in angiogenesis, and IL-19 has indirect angiogenic effects, as IL-19-stimulated bone marrow-derived macrophages secrete proangiogenic factors that induce greater sprouting of aortic rings than unstimulated controls. Using a quantitative RT-PCR screen, we determined that IL-19 induces expression of angiogenic cytokines in bone marrow-derived macrophages. Together, these data suggest that IL-19 can promote angiogenesis in the absence of hypoxia by at least two distinct mechanisms: 1) direct effects on vascular cells and 2) indirect effects by stimulation of macrophages.


Asunto(s)
Aorta Torácica/metabolismo , Interleucina-10/metabolismo , Neovascularización Fisiológica , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/inmunología , Células Cultivadas , Colágeno/farmacología , Medios de Cultivo Condicionados/metabolismo , Combinación de Medicamentos , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Genotipo , Humanos , Interleucina-10/deficiencia , Interleucina-10/genética , Interleucinas , Laminina/farmacología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/inmunología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , PPAR gamma/genética , PPAR gamma/metabolismo , Fenotipo , Proteoglicanos/farmacología , Interferencia de ARN , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Transfección , Factor A de Crecimiento Endotelial Vascular/farmacología
7.
Am J Pathol ; 186(5): 1361-74, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26952642

RESUMEN

Atherosclerosis regression is an important clinical goal, and treatments that can reverse atherosclerotic plaque formation are actively being sought. Our aim was to determine whether administration of exogenous IL-19, a Th2 cytokine, could attenuate progression of preformed atherosclerotic plaque and to identify molecular mechanisms. LDLR(-/-) mice were fed a Western diet for 12 weeks, then administered rIL-19 or phosphate-buffered saline concomitant with Western diet for an additional 8 weeks. Analysis of atherosclerosis burden showed that IL-19-treated mice were similar to baseline, in contrast to control mice which showed a 54% increase in plaque, suggesting that IL-19 halted the progression of atherosclerosis. Plaque characterization showed that IL-19-treated mice had key features of atherosclerosis regression, including a reduction in macrophage content and an enrichment in markers of M2 macrophages. Mechanistic studies revealed that IL-19 promotes the activation of key pathways leading to M2 macrophage polarization, including STAT3, STAT6, Kruppel-like factor 4, and peroxisome proliferator-activated receptor γ, and can reduce cytokine-induced inflammation in vivo. We identified a novel role for IL-19 in regulating macrophage lipid metabolism through peroxisome proliferator-activated receptor γ-dependent regulation of scavenger receptor-mediated cholesterol uptake and ABCA1-mediated cholesterol efflux. These data show that IL-19 can halt progression of preformed atherosclerotic plaques by regulating both macrophage inflammation and cholesterol homeostasis and implicate IL-19 as a link between inflammation and macrophage cholesterol metabolism.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Colesterol/metabolismo , Interleucina-10/farmacología , Macrófagos/metabolismo , Placa Aterosclerótica/tratamiento farmacológico , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Biomarcadores/metabolismo , Dieta Occidental , Progresión de la Enfermedad , Femenino , Inflamación , Interleucinas , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Metabolismo de los Lípidos/fisiología , Macrófagos/efectos de los fármacos , Masculino , Ratones Noqueados , PPAR gamma/metabolismo , Factores de Transcripción STAT/metabolismo , Transfección
8.
Macrophage (Houst) ; 2(1): e562, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26029742

RESUMEN

Hypoxia in ischemic limbs typically initiates angiogenic and inflammatory factors to promote angiogenesis in attempt to restore perfusion, and revascularization involves multiple cell types and systems. Macrophage display phenotype plasticity, and can polarize in response to local and systemic cytokine stimuli. M2 macrophage are known to play an important role in angiogenesis and wound healing. While accepted that many pro-inflammatory cytokines induce angiogenesis, the effects of anti-inflammatory interleukins on initiation of angiogenesis are less clear. Interleukin-19 [IL-19] is a presumed anti-inflammatory cytokine, with unknown effects on macrophage polarization. In our recent study, we used several experimental approaches and determined that IL-19 regulated neovascularization in the murine hind-limb ischemia model. In addition to endothelial cells, we found that IL-19 could target and polarize macrophage to the M2 phenotype. IL-19 could induce expression of angiogenic, and reduce expression of anti-angiogenic cytokines in these cells. This is the first study to demonstrate that IL-19 could polarize macrophage, and potentially identifies IL-19 as a therapy to induce angiogenesis in ischemic tissue.

9.
J Mol Cell Cardiol ; 79: 21-31, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25450612

RESUMEN

Hypoxia in ischemic limbs typically initiates angiogenic and inflammatory factors to promote angiogenesis in attempt to restore perfusion. There is a gap in our knowledge concerning the role of anti-inflammatory interleukins in angiogenesis, macrophage polarization, and endothelial cell activation. Interleukin-19 is a unique anti-inflammatory Th2 cytokine that promotes angiogenic effects in cultured endothelial cells (EC); the purpose of this study was to characterize a role for IL-19 in restoration of blood flow in hind-limb ischemia, and define potential mechanisms. Hind limb ischemia was induced by femoral artery ligation, and perfusion quantitated using Laser Doppler Perfusion Imaging (LDPI). Wild type mice which received i.p. injections of rIL-19 (10ng/g/day) showed significantly increased levels of perfusion compared to PBS controls. LDPI values were significantly decreased in IL-19(-/-) mice when compared to wild type mice. IL-19(-/-) mice injected with rIL-19 had significantly increased LDPI compared with PBS control mice. Significantly increased capillary density was quantitated in rIL-19 treated mice, and significantly less capillary density in IL-19(-/-) mice. Multiple cell types participate in IL-19 induced angiogenesis. IL-19 treatment of human microvascular EC induced expression of angiogenic cytokines. M2 macrophage marker and VEGF-A expression were significantly increased in macrophage and the spleen from rIL-19 injected mice, and M1 marker expression was significantly increased in the spleen from IL-19(-/-) compared with controls. Plasma VEGF-A levels are higher in rIL-19 injected mice. IL-19 decreased the expression of anti-angiogenic IL-12 in the spleen and macrophage. This study is the first to implicate IL-19 as a novel pro-angiogenic interleukin and suggests therapeutic potential for this cytokine.


Asunto(s)
Polaridad Celular , Células Endoteliales/metabolismo , Miembro Posterior/irrigación sanguínea , Interleucina-10/metabolismo , Isquemia/patología , Macrófagos/citología , Neovascularización Fisiológica , Animales , Capilares/metabolismo , Capilares/patología , Regulación de la Expresión Génica , Miembro Posterior/patología , Humanos , Interleucina-10/deficiencia , Subunidad p40 de la Interleucina-12/metabolismo , Interleucinas , Isquemia/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Neovascularización Fisiológica/genética , Fenotipo , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Am J Pathol ; 184(7): 2134-43, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24814101

RESUMEN

We tested the hypothesis that IL-19, a putative member of the type 2 helper T-cell family of anti-inflammatory interleukins, can attenuate intimal hyperplasia and modulate the vascular smooth muscle cell (VSMC) response to injury. Ligated carotid artery of IL-19 knockout (KO) mice demonstrated a significantly higher neointima/intima ratio compared with wild-type (WT) mice (P = 0.04). More important, the increased neointima/intima ratio in the KO could be reversed by injection of 10 ng/g per day recombinant IL-19 into the KO mouse (P = 0.04). VSMCs explanted from IL-19 KO mice proliferated significantly more rapidly than WT. This could be inhibited by addition of IL-19 to KO VSMCs (P = 0.04 and P < 0.01). IL-19 KO VSMCs migrated more rapidly compared with WT (P < 0.01). Interestingly, there was no type 1 helper T-cell polarization in the KO mouse, but there was significantly greater leukocyte infiltrate in the ligated artery in these mice compared with WT. IL-19 KO VSMCs expressed significantly greater levels of inflammatory mRNA, including IL-1ß, tumor necrosis factor α, and monocyte chemoattractant protein-1 in response to tumor necrosis factor α stimulation (P < 0.01 for all). KO VSMCs expressed greater adhesion molecule expression and adherence to monocytes. Together, these data indicate that IL-19 is a previously unrecognized counterregulatory factor for VSMCs, and its expression is an important protective mechanism in regulation of vascular restenosis.


Asunto(s)
Interleucina-10/metabolismo , Miocitos del Músculo Liso/metabolismo , Neointima/patología , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Femenino , Hiperplasia/patología , Interleucinas , Ligadura , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Proteínas Recombinantes/metabolismo , Túnica Íntima/patología
11.
Arterioscler Thromb Vasc Biol ; 33(10): 2316-24, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23950143

RESUMEN

OBJECTIVE: Interleukin-19 (IL-19) is a putative Th2, anti-inflammatory interleukin. Its expression and potential role in atherogenesis are unknown. IL-19 is not detected in normal artery and is expressed to a greater degree in plaque from symptomatic versus asymptomatic patients, suggesting a compensatory counter-regulatory function. We tested whether IL-19 could reduce atherosclerosis in susceptible mice and identified plausible mechanisms. APPROACH AND RESULTS: LDLR(-/-) mice fed an atherogenic diet and injected with either 1.0 or 10.0 ng/g per day recombinant mouse IL-19 had significantly less plaque area in the aortic arch compared with controls (P<0.0001). Weight gain, cholesterol, and triglyceride levels were not significantly different. Gene expression in splenocytes from IL-19-treated mice demonstrated immune cell Th2 polarization, with decreased expression of T-bet, interferon-γ, interleukin-1ß, and interleukin-12ß and increased expression of GATA3 and FoxP3 mRNA. A greater percentage of lymphocytes were Th2 polarized in IL-19-treated mice. Cellular characterization of plaque by immunohistochemistry demonstrated that IL-19-treated mice have significantly less macrophage infiltrate compared with controls (P<0.001). Intravital microscopy revealed significantly less leukocyte adhesion in wild-type mice injected with IL-19 and fed an atherogenic diet compared with controls. Treatment of cultured endothelial cells, vascular smooth muscle cells, and bone marrow-derived macrophages with IL-19 resulted in a significant decrease in chemokine mRNA and mRNA stability protein human antigen R. CONCLUSIONS: These data suggest that IL-19 is a potent inhibitor of experimental atherosclerosis, with diverse mechanisms including immune cell polarization, decrease in macrophage adhesion, and decrease in gene expression. This may identify IL-19 as a novel therapeutic to limit vascular inflammation.


Asunto(s)
Aorta/efectos de los fármacos , Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Interleucina-10/farmacología , Anciano , Animales , Aorta/inmunología , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Biomarcadores/sangre , Enfermedades de las Arterias Carótidas/inmunología , Enfermedades de las Arterias Carótidas/patología , Células Cultivadas , Colesterol/sangre , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-10/metabolismo , Interleucinas/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/inmunología , Placa Aterosclerótica , Receptores de LDL/deficiencia , Receptores de LDL/genética , Proteínas Recombinantes/farmacología , Bazo/efectos de los fármacos , Bazo/inmunología , Células Th2/efectos de los fármacos , Células Th2/inmunología , Factores de Tiempo , Triglicéridos/sangre
12.
Autoimmunity ; 45(4): 279-89, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22283427

RESUMEN

Although it is evident that there is complex interplay among genetic and environmental factors contributing to systemic autoimmunity, the events inciting autoreactivity are incompletely understood. Previously we demonstrated that MRL-MpJ mice posses a genetic background susceptible to autoimmunity development under conditions of altered inhibitory signaling. To gain better understanding of the influence of exogenous factors on autoreactivity in susceptible individuals, young MRL-MpJ mice were challenged with a single injection of heterologous protein and evaluated for evidence of autoimmunity. We found that MRL-MpJ mice developed high titer serum reactivity to DNA within 1 week of protein administration reaching maximal levels within 1 month. Importantly, the level of autoimmunity was sustained for an extended period of time (6 months). This was accompanied by a substantial increase in germinal center B cell and plasma cell numbers. In contrast, control mice showed no change in autoreactivity or lymphocyte homeostasis. Autoimmunity was dependent on marginal zone B cells as their depletion reduced serum auto-reactivity after challenge, thus suggesting immune stimulation with heterologous proteins can precipitate loss of B cell tolerance and autoimmunity in genetically prone individuals. This model may provide an important tool to further investigate the mechanisms whereby environmental stimuli trigger autoimmune reactivity in susceptible hosts.


Asunto(s)
Anticuerpos Antinucleares/sangre , Autoinmunidad , Lupus Eritematoso Sistémico/inmunología , Células Plasmáticas/inmunología , Animales , Anticuerpos Antinucleares/inmunología , ADN/inmunología , Femenino , Centro Germinal/inmunología , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr
13.
J Biol Chem ; 287(4): 2477-84, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22158875

RESUMEN

Heme oxygenase-1 (HO-1) has potent anti-inflammatory activity and recognized vascular protective effects. We have recently described the expression and vascular protective effects of an anti-inflammatory interleukin (IL-19), in vascular smooth muscle cells (VSMC) and injured arteries. The objective of this study was to link the anti-inflammatory effects of IL-19 with HO-1 expression in resident vascular cells. IL-19 induced HO-1 mRNA and protein in cultured human VSMC, as assayed by quantitative RT-PCR, immunoblot, and ELISA. IL-19 does not induce HO-1 mRNA or protein in human endothelial cells. IL-19 activates STAT3 in VSMC, and IL-19-induced HO-1 expression is significantly reduced by transfection of VSMC with STAT3 siRNA or mutation of the consensus STAT binding site in the HO-1 promoter. IL-19 treatment can significantly reduce ROS-induced apoptosis, as assayed by Annexin V flow cytometry. IL-19 significantly reduced ROS concentrations in cultured VSMC. The IL-19-induced reduction in ROS concentration is attenuated when HO-1 is reduced by siRNA, indicating that the IL-19-driven decrease in ROS is mediated by HO-1 expression. IL-19 reduces vascular ROS in vivo in mice treated with TNFα. This points to IL-19 as a potential therapeutic for vascular inflammatory diseases and a link for two previously unassociated protective processes: Th2 cytokine-induced anti-inflammation and ROS reduction.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/fisiología , Hemo-Oxigenasa 1/biosíntesis , Interleucinas/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Células Cultivadas , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/inmunología , Humanos , Interleucinas/genética , Interleucinas/inmunología , Ratones , Músculo Liso Vascular/inmunología , Miocitos del Músculo Liso/inmunología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Mensajero/inmunología , Especies Reactivas de Oxígeno/inmunología , Elementos de Respuesta/fisiología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Factor de Transcripción STAT3/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Vasculitis/genética , Vasculitis/inmunología , Vasculitis/metabolismo
14.
Am J Physiol Cell Physiol ; 300(4): C896-906, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21209363

RESUMEN

Vascular smooth muscle cell (VSMC) migration is an important cellular event in multiple vascular diseases, including atherosclerosis, restenosis, and transplant vasculopathy. Little is known regarding the effects of anti-inflammatory interleukins on VSMC migration. This study tested the hypothesis that an anti-inflammatory Th2 interleukin, interleukin-19 (IL-19), could decrease VSMC motility. IL-19 significantly decreased platelet-derived growth factor (PDGF)-stimulated VSMC chemotaxis in Boyden chambers and migration in scratch wound assays. IL-19 significantly decreased VSMC spreading in response to PDGF. To determine the molecular mechanism(s) for these cellular effects, we examined the effect of IL-19 on activation of proteins that regulate VSMC cytoskeletal dynamics and locomotion. IL-19 decreased PDGF-driven activation of several cytoskeletal regulatory proteins that play an important role in smooth muscle cell motility, including heat shock protein-27 (HSP27), myosin light chain (MLC), and cofilin. IL-19 decreased PDGF activation of the Rac1 and RhoA GTPases, important integrators of migratory signals. IL-19 was unable to inhibit VSMC migration nor was able to inhibit activation of cytoskeletal regulatory proteins in VSMC transduced with a constitutively active Rac1 mutant (RacV14), suggesting that IL-19 inhibits events proximal to Rac1 activation. Together, these data are the first to indicate that IL-19 can have important inhibitory effects on VSMC motility and activation of cytoskeletal regulatory proteins. This has important implications for the use of anti-inflammatory cytokines in the treatment of vascular occlusive disease.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/efectos de los fármacos , Interleucinas/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/fisiología , Adolescente , Adulto , Antiinflamatorios/farmacología , Células Cultivadas , Proteínas del Citoesqueleto/genética , Citoesqueleto/metabolismo , Proteínas de Choque Térmico HSP27/metabolismo , Humanos , Masculino , Miocitos del Músculo Liso/citología , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/farmacología , Adulto Joven , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
15.
Arterioscler Thromb Vasc Biol ; 31(1): 167-75, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20966397

RESUMEN

OBJECTIVE: To characterize the expression and function of interleukin (IL) 19, a recently described T-helper 2 anti-inflammatory IL, on endothelial cell (EC) pathophysiological features. METHODS AND RESULTS: The expression and effects of anti-inflammatory ILs on EC activation and development of angiogenesis are uncharacterized. We demonstrate by immunohistochemistry and immunoblot that IL-19 is expressed in inflamed, but not normal, human coronary endothelium and can be induced in cultured human ECs by serum and basic fibroblast growth factor. IL-19 is mitogenic and chemotactic, and it promotes EC spreading. IL-19 activates the signaling proteins STAT3, p44/42, and Rac1. In functional ex vivo studies, IL-19 promotes cordlike structure formation of cultured ECs and enhances microvessel sprouting in the mouse aortic ring assay. IL-19 induces tube formation in gelatinous protein (Matrigel) plugs in vivo. CONCLUSIONS: To our knowledge, these data are the first to report expression of the anti-inflammatory agent, IL-19, in ECs; and the first to indicate that IL-19 is mitogenic and chemotactic for ECs and can induce the angiogenic potential of ECs.


Asunto(s)
Células Endoteliales/metabolismo , Inflamación/prevención & control , Interleucinas/metabolismo , Neovascularización Fisiológica , Animales , Western Blotting , Proliferación Celular , Forma de la Célula , Células Cultivadas , Quimiotaxis , Células Endoteliales/inmunología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Inmunohistoquímica , Inflamación/inmunología , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Microvasos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Factores de Tiempo , Proteína de Unión al GTP rac1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...