Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 288
Filtrar
Más filtros

Intervalo de año de publicación
1.
Immunity ; 43(3): 488-501, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26320657

RESUMEN

The intestinal epithelial barrier plays a critical role in the mucosal immunity. However, it remains largely unknown how the epithelial barrier is maintained after damage. Here we show that growth factor FGF2 synergized with interleukin-17 (IL-17) to induce genes for repairing of damaged epithelium. FGF2 or IL-17 deficiency resulted in impaired epithelial proliferation, increased pro-inflammatory microbiota outgrowth, and consequently worse pathology in a DSS-induced colitis model. The dysregulated microbiota in the model induced transforming growth factor beta 1 (TGFß1) expression, which in turn induced FGF2 expression mainly in regulatory T cells. Act1, an essential adaptor in IL-17 signaling, suppressed FGF2-induced ERK activation through binding to adaptor molecule GRB2 to interfere with its association with guanine nucleotide exchange factor SOS1. Act1 preferentially bound to IL-17 receptor complex, releasing its suppressive effect on FGF2 signaling. Thus, microbiota-driven FGF2 and IL-17 cooperate to repair the damaged intestinal epithelium through Act1-mediated direct signaling cross-talk.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/inmunología , Interleucina-17/inmunología , Intestinos/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Colitis/genética , Colitis/inmunología , Colitis/metabolismo , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Perfilación de la Expresión Génica/métodos , Células HEK293 , Células HT29 , Células HeLa , Humanos , Interleucina-17/genética , Interleucina-17/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Intestinos/microbiología , Intestinos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microbiota/genética , Microbiota/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Cicatrización de Heridas/inmunología
2.
Exp Eye Res ; 190: 107823, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31600485

RESUMEN

Administration of RC28-E, a VEGF/bFGF dual decoy receptor (IgG1 Fc-fusion protein), have shown relative therapeutic value in ocular in vivo models, including laser-induced choroidal neovascularization (CNV) in monkeys and streptozotocin (STZ)-induced diabetic retinopathy (DR) in rats. In the present study, we have elucidated the pharmacokinetics profiles of RC28-E at the systemic, vitreous and aqueous humor after administration in a primate model (Macaca fascicularis). Moreover, here we tease out the ocular tissue distribution of RC28-E after intravitreal administration, and we also determine the systemic bioavailability after both intravitreal and intravenous administration. Our results show that RC28-E is rapidly and well-distributed into ocular tissues after intravitreal administration. Drug exposure in choroid and retina was approximately one-quarter and one-twelfth of that in vitreous humor, while its half-life in vitreous and aqueous humor were well-sustained (3.3 and 3.0 days). Remarkably, RC28-E could cross the blood-ocular barrier, and the systemic bioavailability of RC28-E was ~25%. No drug accumulation after multiple administration was noticed, but low titers of antibody produce against RC28-E were detected. Overall, RC28-E exhibited high clinical value due to adequate pharmacokinetic profiling, safety and efficacy.


Asunto(s)
Humor Acuoso/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacocinética , Proteínas Recombinantes de Fusión/farmacocinética , Factor A de Crecimiento Endotelial Vascular/farmacocinética , Cuerpo Vítreo/metabolismo , Animales , Disponibilidad Biológica , Barrera Hematorretinal , Coroides/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Factor 2 de Crecimiento de Fibroblastos/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Inyecciones Intravítreas , Cristalino/metabolismo , Macaca fascicularis , Masculino , Proteínas Recombinantes de Fusión/inmunología , Retina/metabolismo , Distribución Tisular , Factor A de Crecimiento Endotelial Vascular/inmunología
3.
Nanomedicine ; 29: 102254, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32615335

RESUMEN

FGF-2 accumulates in many tumor tissues and is closely related to the development of tumor angiogenesis and the immunosuppressive microenvironment. This study aimed to investigate whether active immunization against FGF-2 could modify antitumor immunity and enhance the efficacy of an HPV16 E7-specific therapeutic vaccine. Combined immunization targeting both FGF-2 and E7 significantly suppressed tumor growth, which was accompanied by significantly increased levels of IFN-γ-expressing splenocytes and effector CD8 T cells and decreased levels of immunosuppressive cells such as regulatory T cells (Tregs) and myeloid-derived suppressor cells(MDSCs) in both the spleen and tumor; in addition, the levels of FGF-2 and neovascularization in tumors were decreased in the mice receiving the combined immunization, and tumor cell apoptosis was promoted. The combination of an HPV16 E7-specific vaccine and active immunization against FGF-2 significantly enhances antitumor immune responses in mice with TC-1 tumors, indicating a promising strategy for tumor immunotherapy.


Asunto(s)
Vacunas contra el Cáncer/farmacología , Factor 2 de Crecimiento de Fibroblastos/inmunología , Neovascularización Patológica/inmunología , Proteínas E7 de Papillomavirus/inmunología , Vacunas contra Papillomavirus/farmacología , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 2 de Crecimiento de Fibroblastos/genética , Humanos , Inmunoterapia , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/prevención & control , Neovascularización Patológica/virología , Proteínas E7 de Papillomavirus/antagonistas & inhibidores , Proteínas E7 de Papillomavirus/genética , Vacunas contra Papillomavirus/inmunología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Vacunación
4.
Int J Mol Sci ; 21(15)2020 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-32731615

RESUMEN

Mesenchymal stem cells (MSCs) have become a promising tool in cellular therapy for restoring immune system haemostasis; however, the success of clinical trials has been impaired by the lack of standardized manufacturing processes. This study aims to determine the suitability of source tissues and culture media for the production of MSC-based advanced therapy medicinal products (ATMPs) and to define parameters to extend the set of release criteria. MSCs were isolated from umbilical cord (UC), bone marrow and lipoaspirate and expanded in three different culture media. MSC phenotype, proliferation capacity and immunosuppressive parameters were evaluated in normal MSCs compared to primed MSCs treated with cytokines mimicking an inflammatory environment. Compared to bone marrow and lipoaspirate, UC-derived MSCs (UC-MSCs) showed the highest proliferative capacity, which was further enhanced by media supplemented with bFGF, while the cells maintained their immunosuppressive characteristics. Moreover, UC-MSCs expanded in the bFGF-enriched medium were the least sensitive to undesirable priming-induced changes in the MSC phenotype. Surface markers and secreted factors were identified to reflect the cell response to inflammatory priming and to be variable among MSCs from different source tissues. This study demonstrates that UC is a favorable cell source for manufacturing MSC-based ATMPs for immunosuppressive applications. UC-MSCs are able to use the bFGF-enriched medium for higher cell yields without the impairment of immunosuppressive parameters and undesirable phenotype changes after inflammatory preconditioning of MSCs before transplantation. Additionally, immunosuppressive parameters were identified to help finding predictors of clinically efficient MSCs in the following clinical trials.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Terapia de Inmunosupresión , Células Madre Mesenquimatosas/inmunología , Cordón Umbilical/inmunología , Diferenciación Celular/inmunología , Factor 2 de Crecimiento de Fibroblastos/inmunología , Humanos , Células Madre Mesenquimatosas/citología , Cordón Umbilical/citología
5.
Bull Exp Biol Med ; 169(4): 467-469, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32910376

RESUMEN

Cultured peritoneal macrophages from intact (control) and BCG-infected (experiment) male BALB/c mice were studied 90 days after infection. Polarization of macrophages by M1 (expression of GM-CSF, IFNγ, and CD16/32) and M2 (expression of bFGF and CD36) differentiation pathways was studied with consideration for their the nuclearity class. Mononuclear cells predominated (90% and higher) in macrophage cultures of both groups and presumably, were presented by mainly epithelioid cells. The results indicated polarization of mononuclear and multinuclear macrophages in the M2 direction under conditions of BCG granulomatosis and a higher initial M2 polarization of binuclear macrophages. In control cultures, the ratio of M2 to M1 macrophages was 0.57, in experimental cultures this ratio was 1.6. It seems that long persistence of Mycobacterium tuberculosis in macrophages served as a factor stimulating the plastic processes and transformation of macrophages into epithelioid cells that form the "core" of granulomas and their enlargement upon incorporation of macrophages.


Asunto(s)
Células Epitelioides/patología , Regulación de la Expresión Génica/inmunología , Macrófagos Peritoneales/patología , Mycobacterium bovis/crecimiento & desarrollo , Tuberculosis/patología , Animales , Antígenos CD36/genética , Antígenos CD36/inmunología , Diferenciación Celular , Transdiferenciación Celular/genética , Transdiferenciación Celular/inmunología , Células Epitelioides/inmunología , Células Epitelioides/microbiología , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Interferón gamma/genética , Interferón gamma/inmunología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/microbiología , Masculino , Ratones , Ratones Endogámicos BALB C , Mycobacterium bovis/patogenicidad , Cultivo Primario de Células , Receptores de IgG/genética , Receptores de IgG/inmunología , Tuberculosis/genética , Tuberculosis/inmunología , Tuberculosis/microbiología
6.
J Biol Chem ; 293(44): 17218-17228, 2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30217817

RESUMEN

Fibroblast growth factor receptor-1 (FGFR1) activity at the plasma membrane is tightly controlled by the availability of co-receptors and competing receptor isoforms. We have previously shown that FGFR1 activity in pancreatic beta-cells modulates a wide range of processes, including lipid metabolism, insulin processing, and cell survival. More recently, we have revealed that co-expression of FGFR5, a receptor isoform that lacks a tyrosine-kinase domain, influences FGFR1 responses. We therefore hypothesized that FGFR5 is a co-receptor to FGFR1 that modulates responses to ligands by forming a receptor heterocomplex with FGFR1. We first show here increased FGFR5 expression in the pancreatic islets of nonobese diabetic (NOD) mice and also in mouse and human islets treated with proinflammatory cytokines. Using siRNA knockdown, we further report that FGFR5 and FGFR1 expression improves beta-cell survival. Co-immunoprecipitation and quantitative live-cell imaging to measure the molecular interaction between FGFR5 and FGFR1 revealed that FGFR5 forms a mixture of ligand-independent homodimers (∼25%) and homotrimers (∼75%) at the plasma membrane. Interestingly, co-expressed FGFR5 and FGFR1 formed heterocomplexes with a 2:1 ratio and subsequently responded to FGF2 by forming FGFR5/FGFR1 signaling complexes with a 4:2 ratio. Taken together, our findings identify FGFR5 as a co-receptor that is up-regulated by inflammation and promotes FGFR1-induced survival, insights that reveal a potential target for intervention during beta-cell pathogenesis.


Asunto(s)
Citocinas/inmunología , Diabetes Mellitus/genética , Células Secretoras de Insulina/inmunología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 5 de Factor de Crecimiento de Fibroblastos/genética , Animales , Diabetes Mellitus/inmunología , Dimerización , Femenino , Factor 2 de Crecimiento de Fibroblastos/inmunología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/química , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/inmunología , Receptor Tipo 5 de Factor de Crecimiento de Fibroblastos/química , Receptor Tipo 5 de Factor de Crecimiento de Fibroblastos/inmunología , Regulación hacia Arriba
7.
J Immunol ; 199(9): 3086-3093, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28924004

RESUMEN

Activated CD4 T cells connect to airway smooth muscle cells (ASMCs) in vitro via lymphocyte-derived membrane conduits (LMCs) structurally similar to membrane nanotubes with unknown intercellular signals triggering their formation. We examined the structure and function of CD4 T cell-derived LMCs, and we established a role for ASMC-derived basic fibroblast growth factor 2 (FGF2b) and FGF receptor (FGFR)1 in LMC formation. Blocking FGF2b's synthesis and FGFR1 function reduced LMC formation. Mitochondrial flux from ASMCs to T cells was partially FGF2b and FGFR1 dependent. LMC formation by CD4 T cells and mitochondrial transfer from ASMCs was increased in the presence of asthmatic ASMCs that expressed more mRNA for FGF2b compared with normal ASMCs. These observations identify ASMC-derived FGF2b as a factor needed for LMC formation by CD4 T cells, affecting intercellular communication.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Comunicación Celular/inmunología , Extensiones de la Superficie Celular/inmunología , Factor 2 de Crecimiento de Fibroblastos/inmunología , Miocitos del Músculo Liso/inmunología , Linfocitos T CD4-Positivos/citología , Humanos , Mitocondrias/inmunología , Miocitos del Músculo Liso/citología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/inmunología , Sistema Respiratorio/citología , Sistema Respiratorio/inmunología
8.
Bull Exp Biol Med ; 164(4): 456-458, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29504104

RESUMEN

The incidence of mono- and multinuclear cells and their expression of pro- and antifibrotic factors were studied in cultured peritoneal macrophages from intact and BCG-infected mice. Generally, the expression of factors increased with an increase in the number of nuclei per cell. However, the expression was higher in macrophages from BCG infected mice, except the cells with 3 and more nuclei, extremely rarely expressing IL-1α in cultures from intact and BCG-infected animals. The number of macrophages with 3 and more nuclei, expressing CatD, was comparable with the number of mono- and binuclear macrophages. Presumably, this was determined by various mechanisms of formation of multinuclear (3-5 and more nuclei) macrophages, for example, by amitosis.


Asunto(s)
Catepsina D/inmunología , Factor 2 de Crecimiento de Fibroblastos/inmunología , Interferón gamma/inmunología , Interleucina-1alfa/inmunología , Macrófagos Peritoneales/inmunología , Metaloproteinasa 13 de la Matriz/inmunología , Infecciones por Mycobacterium/inmunología , Factor de Crecimiento Transformador beta1/inmunología , Animales , Catepsina D/genética , Núcleo Celular , Factor 2 de Crecimiento de Fibroblastos/genética , Regulación de la Expresión Génica , Interferón gamma/genética , Interleucina-1alfa/genética , Macrófagos Peritoneales/microbiología , Macrófagos Peritoneales/patología , Masculino , Metaloproteinasa 13 de la Matriz/genética , Ratones , Ratones Endogámicos BALB C , Infecciones por Mycobacterium/microbiología , Infecciones por Mycobacterium/patología , Mycobacterium bovis/inmunología , Cultivo Primario de Células , Transducción de Señal , Factor de Crecimiento Transformador beta1/genética
9.
Respir Res ; 18(1): 48, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28298222

RESUMEN

Mast cells are accumulated in advanced chronic obstructive pulmonary disease (COPD), and interleukin (IL)-17 signaling plays a role in disease progression. The expression, localization and functional relevance of IL-17 receptor (R)A and IL-17RC was explored in COPD by immunodetection, and functional assays.IL-17RA and IL-17RC was increased in very severe COPD, and expressed by mast cells. Increased secretion of the pro-angiogenic basic fibroblast growth factor and vascular endothelial growth factor was observed in vitro-maintained mast cells stimulated with IL-17A. Expression of these mediators was confirmed in end-stage COPD. Thus, accumulation of mast cells in COPD may contribute to vascular remodeling.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Pulmón/inmunología , Mastocitos/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Receptores de Interleucina-17/inmunología , Receptores de Interleucina/inmunología , Factor A de Crecimiento Endotelial Vascular/inmunología , Anciano , Femenino , Factor 2 de Crecimiento de Fibroblastos/inmunología , Humanos , Masculino , Mastocitos/inmunología , Persona de Mediana Edad , Regulación hacia Arriba/inmunología
10.
J Immunol ; 195(10): 4943-52, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26466960

RESUMEN

Fibroblast growth factor (FGF)2,which is one of the 22 members of the FGF family, functions as an extracellular molecule involved in canonical receptor tyrosine kinase signaling. It has been implicated in angiogenesis and the development of the CNS. In this article, we reveal that cytosolic low m.w. isoform (LMW) FGF2 (18 kDa), not its secreted form, plays an unexpected role in the innate immune response. Cytosolic LMW FGF2 directly associated with inactivated RIG-I under physiological conditions, which enhanced RIG-I protein stability, thereby maintaining basal RIG-I levels. However, during RIG-I activation induced by viral RNA, cytosolic FGF2 bound to the caspase recruitment domains of activated RIG-I, which blocked RIG-I-MAVS complex formation. LMW FGF2 deficiency increased type I IFN production, whereas the overexpression of LMW FGF2 exerted the opposite effect. Cytosolic LMW FGF2 functions as a negative regulator in RIG-I-mediated antiviral signaling. This work provides insight into the role of FGF2 in innate immune response.


Asunto(s)
Citosol/inmunología , ARN Helicasas DEAD-box/inmunología , Factor 2 de Crecimiento de Fibroblastos/inmunología , Inmunidad Innata , Transducción de Señal/inmunología , Virosis/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , Factor 2 de Crecimiento de Fibroblastos/genética , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Ratones , Ratones Noqueados , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Transducción de Señal/genética , Virosis/genética
11.
Cancer Sci ; 107(8): 1141-50, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27251178

RESUMEN

Fibroblast growth factor-2 (FGF-2) is one of the most important angiogenic factors to promote tumor growth, progression and metastasis. Neutralizing antibodies against FGF-2 may suppress the growth of tumor cells by blocking the FGF-2 signaling pathway. In this study, a disulfide-stabilized diabody (ds-Diabody) that specifically targets FGF-2 was designed. Compared to its parent antibody, the introduction of disulphide bonds in the diabody could significantly increase the stability of ds-Diabody and maintain its antigen binding activity. The ds-Diabody against FGF-2 could effectively inhibit the tube formation and migration of vascular endothelial cells and block the proliferation and invasion of human breast cancer cells. In the mouse model of breast cancer xenograft tumors, the ds-Diabody against FGF-2 could significantly inhibit the growth of tumor cells. Moreover, the densities of microvessels stained with CD31 and lymphatic vessels stained with LYVE1 in tumors showed a significant decrease following treatment with the ds-Diabody against FGF-2. Our data indicated that the ds-Diabody against FGF-2 could inhibit tumor angiogenesis, lymphangiogenesis and tumor growth.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Disulfuros/química , Factor 2 de Crecimiento de Fibroblastos/inmunología , Inmunoglobulinas/inmunología , Inmunoglobulinas/farmacología , Multimerización de Proteína , Animales , Células 3T3 BALB , Neoplasias de la Mama/irrigación sanguínea , Capilares/efectos de los fármacos , Capilares/crecimiento & desarrollo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Inmunoglobulinas/química , Inmunoglobulinas/aislamiento & purificación , Linfangiogénesis/efectos de los fármacos , Células MCF-7 , Ratones , Invasividad Neoplásica/prevención & control , Neovascularización Patológica/tratamiento farmacológico , Pichia/genética , Pichia/metabolismo , Transducción de Señal/efectos de los fármacos
12.
Ann Rheum Dis ; 74(1): 260-6, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24249810

RESUMEN

OBJECTIVE: Human bone marrow mesenchymal stromal cells (hBM-MSC) are being applied in tissue regeneration and treatment of autoimmune diseases (AD). Their cellular and immunophenotype depend on isolation and culture conditions which may influence their therapeutic application and reflect their in vivo biological functions. We have further characterised the phenotype induced by fibroblast growth factor 2 (FGF2) on healthy donor hBM-MSC focusing on the osteoimmunological markers osteoprotegerin (OPG), receptor activator of nuclear factor kB (RANK), RANK ligand (RANKL) and HLA-DR and their regulation of expression by the inflammatory cytokines IL1ß and IFNγ. METHODS: RANK, RANKL, OPG and HLA-DR expression in hBM-MSC expanded under specific culture conditions, were measured by RT-PCR and flow cytometry. MAPKs induction by FGF2, IL1ß and IFNγ in hBM-MSC was analysed by immunoblotting and RT-PCR. RESULTS: In hBM-MSC, OPG expression is constitutive and FGF2 independent. RANKL expression depends on FGF2 and ERK1/2 activation. IL1ß and IFNγ activate ERK1/2 but fail to induce RANKL. Only IL1ß induces P38MAPK. The previously described HLA-DR induced by FGF2 through ERK1/2 on hBM-MSC, is suppressed by IL1ß through inhibition of CIITA transcription. HLA-DR induced by IFNγ is not affected by IL1ß in hBM-MSC, but is suppressed in articular chondrocytes and lung fibroblasts. CONCLUSIONS: RANKL expression and IL1ß regulated MHC-class II, both induced via activation of the ERK1/2 signalling pathway, are specific for progenitor hBM-MSC expanded in the presence of FGF2. HLA-DR regulated by IL1ß and ERK1/2 is observed on hBM-MSC during early expansion without FGF2 suggesting previous in vivo acquisition. Stromal progenitor cells with this phenotype could have an osteoimmunological role during bone regeneration.


Asunto(s)
Células de la Médula Ósea/metabolismo , Factor 2 de Crecimiento de Fibroblastos/inmunología , Antígenos HLA-DR/genética , Interferón gamma/inmunología , Interleucina-1beta/inmunología , Células Madre Mesenquimatosas/metabolismo , Osteoprotegerina/genética , Ligando RANK/genética , Receptor Activador del Factor Nuclear kappa-B/genética , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Antígenos HLA-DR/efectos de los fármacos , Humanos , Interferón gamma/metabolismo , Interferón gamma/farmacología , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/inmunología , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Osteoprotegerina/efectos de los fármacos , Osteoprotegerina/metabolismo , Ligando RANK/efectos de los fármacos , Ligando RANK/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Activador del Factor Nuclear kappa-B/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
J Med Assoc Thai ; 98 Suppl 9: S16-22, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26817205

RESUMEN

BACKGROUND: Glial derived neurotrophic factor (GDNF) and basic fibroblast growth factor (FGF-2) protect nigrostriatal dopaminergic (DA) neurons and their projections in animal models of Parkinson's disease (PD). Recent data indicate neuroprotective effects of estrogen in PD animal models through its anti-inflammatory and anti-oxidative effects, yet the hormonal effects on GDNF and FGF-2 expression in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice remain uninvestigated. OBJECTIVE: To determine the effects of 17 beta-estradiol (E2) on DA innervation and the expression ofGDNFandFGF-2 in the striatum of MPTP-treated mice. MATERIAL AND METHOD: Adult male mice were treated with E2 or vehicle for 11 days during which they were injected with MPTP or saline on the sixth day. The striatum was collected on day 11 and processedfor tyrosine hydroxylase (TH), GDNF and FGF-2 immunohistochemistry. Extent ofDA innervation and the expression of GDNF and FGF-2 in the striatum were assessed by measuring optical density of TH, GDNF and FGF-2 immunoreactivity, respectively. RESULTS: MPTP induced loss of DA axons and upregulation of FGF-2 expression, but did not alter GDNF level. E2 alleviated loss of DA axons, increased GDNF level, yet caused no change in FGF-2 level ofthe MPTP-intoxicated animals. CONCLUSION: One possible mechanism by which E2 protects nigrostriatal DA axons against MPTP is through upregulation ofstriatal GDNF.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Estrógenos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/inmunología , Factor Neurotrófico Derivado de la Línea Celular Glial/inmunología , Animales , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Estradiol/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Sustancia Negra/metabolismo , Tirosina 3-Monooxigenasa/farmacología
14.
Respiration ; 87(3): 234-42, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24435103

RESUMEN

BACKGROUND: Dendritic cells (DCs) have a pivotal role in the onset and regulation of innate and adaptive immune responses. Moreover, DCs can interact with angiogenic modulators, resulting in modification of their biology and participation in angiogenesis. OBJECTIVES: This study was designed to evaluate the relationship between the density of DCs, vascularity and expression of angiogenic factors [vascular endothelial growth factor (VEGF), transforming growth factor (TGF)-ß and basic fibroblast growth factor (bFGF)] in the central airways of chronic obstructive pulmonary disease (COPD) patients. METHODS: The study included 20 patients with moderate/severe COPD and 8 healthy control subjects. Bronchial biopsies were evaluated by immunohistochemistry. Specimens were examined for CD83 and CD207 to mark mature and immature DCs, respectively, for collagen IV to evaluate vascularity, and for VEGF, TGF-ß and bFGF. RESULTS: Compared to controls, COPD patients had a significant reduction of CD83+ cells and an increased CD207/CD83 ratio (p < 0.05). Vascularity, VEGF, TGF-ß and bFGF were also significantly increased in COPD patients as compared to controls (p < 0.01). In COPD patients, CD83+ cells were inversely related to VEGF and TGF-ß expression (p < 0.05). Moreover, the CD207/CD83 ratio was positively related to VEGF, TGF-ß and vascularity (p < 0.05). Finally, CD207+ cells were inversely related to FEV1 (p < 0.05). CONCLUSION: Our results show a reduced maturation of DCs in COPD that was related to airway vascularity and angiogenic factors (VEGF and TGF-ß). Additionally, immature DCs were significantly related to disease severity. We propose that the interplay between airway vascular changes, on one hand, and DCs maturation on the other, may play a key role in the pathogenetic mechanisms of COPD.


Asunto(s)
Antígenos CD/metabolismo , Bronquios/metabolismo , Células Dendríticas/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Inmunoglobulinas/metabolismo , Lectinas Tipo C/metabolismo , Lectinas de Unión a Manosa/metabolismo , Glicoproteínas de Membrana/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Anciano , Anciano de 80 o más Años , Bronquios/inmunología , Estudios de Casos y Controles , Células Dendríticas/inmunología , Femenino , Factor 2 de Crecimiento de Fibroblastos/inmunología , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Neovascularización Patológica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Índice de Severidad de la Enfermedad , Factor de Crecimiento Transformador beta/inmunología , Factor A de Crecimiento Endotelial Vascular/inmunología , Antígeno CD83
15.
Int J Cancer ; 130(2): 356-64, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21351090

RESUMEN

The roles of fibroblast growth factor-2 (FGF-2) in the hepatocellular carcinoma (HCC) development are still controversial. In this study, we investigated the expression of FGF-2 in chronic hepatitis (CH) type C patients with or without HCC and the immunoregulation of FGF-2 in NK sensitivity of HCC cells. The FGF-2 expressions were detected in the liver tissues of patients, but not in normal liver. The serum FGF-2 levels of the patients with CH, liver cirrhosis (LC) or HCC were significantly higher than those of healthy volunteers. The serum FGF-2 levels of patients decreased with the progression of chronic liver disease. HCC occurrence of LC patients with high levels of serum FGF-2 was significantly lower than that with low levels of serum FGF-2. Proinflammatory cytokines, such as IL-1ß and IL-6, induced FGF-2 expressions in HCC cells and normal hepatocytes. FGF-2 stimulation resulted in increasing the expression of the membrane-bound major histocompatibility complex class I-related chain A (MICA), an NK activating molecule, and decreasing that of human leukocyte antigen (HLA) class I, an NK inhibitory molecule, on HCC cells. This did not occur with normal hepatocytes. Adding anti-FGF receptor-2 neutralizing antibody resulted in inhibiting the change of MICA and HLA class I expressions on FGF-2 stimulated HCC cells. FGF-2 stimulation on HCC cells resulted in increasing NK sensitivity against HCC cells. These findings indicate that FGF-2 produced by HCC cells or normal hepatocytes of chronic liver disease may play critical roles in eliminating HCC cells by innate immunity.


Asunto(s)
Carcinoma Hepatocelular/inmunología , Factor 2 de Crecimiento de Fibroblastos/inmunología , Células Asesinas Naturales/inmunología , Neoplasias Hepáticas/inmunología , Anciano , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Línea Celular Tumoral , Citocinas/biosíntesis , Citocinas/inmunología , Femenino , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Factor 2 de Crecimiento de Fibroblastos/sangre , Antígenos HLA/biosíntesis , Hepatitis C Crónica/inmunología , Antígenos de Histocompatibilidad Clase I/biosíntesis , Humanos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Masculino , Persona de Mediana Edad , Subfamília C de Receptores Similares a Lectina de Células NK/biosíntesis , Subfamilia K de Receptores Similares a Lectina de Células NK/biosíntesis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/biosíntesis
16.
Cell Immunol ; 273(1): 59-66, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22192476

RESUMEN

Long term survival of the human lung allografts are hindered by chronic rejection, manifested clinically as bronchiolitis obliterans syndrome (BOS). We previously demonstrated significant correlation between the development of antibodies (Abs) to K-α1-tubulin (Kα1T) and BOS. In this study, we investigated the molecular basis for fibrinogenesis mediated by ligation of Kα1T expressed on airway epithelial cells by its specific Abs. Using RT-PCR we demonstrate that normal human bronchial epithelial (NHBE) cells upon ligation of Kα1T with specific Abs caused upregulation of pro-fibrotic growth factors. Western blot analysis of NHBE incubated with Kα1T Abs increased hypoxia inducible factor (HIF-1α). Kα1T Ab-mediated growth factor expression is dependent on HIF-1α as inhibition of HIF-1α returned fibrotic growth factor expression to basal levels. In conclusion, we propose that HIF-1α -mediated upregulation of fibrogenic growth factors induced by ligation of Kα1T Abs is critical for development of fibrosis leading to chronic rejection of lung allograft.


Asunto(s)
Bronquiolitis Obliterante/inmunología , Conexinas/inmunología , Rechazo de Injerto/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Trasplante de Pulmón/inmunología , Transducción de Señal/inmunología , Western Blotting , Línea Celular , Células Epiteliales , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/inmunología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/inmunología , ARN/química , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/inmunología
17.
Clin Exp Allergy ; 42(11): 1604-14, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23106660

RESUMEN

BACKGROUND: Unlike other IL-17 family members, the Th2-derived cytokine IL-25 (IL-17E) induces (promotes) Th2 responses. One or both of the two receptors for IL-25 (IL-17RA, IL-17RB) is expressed on inflammatory cells and tissue structural cells, suggesting that in addition to promoting Th2-type inflammation IL-25 may also act on structural cells at sites of Th2-type inflammation such as in the asthmatic bronchial mucosa to promote remodelling changes. OBJECTIVE: Our previous studies showed elevated expression of IL-25 and IL-17RB immunoreactivity in asthmatic airways with co-localization of the latter to endothelial cells. We therefore hypothesized that IL-25 acts on endothelial cells through this receptor to induce production of the key angiogenic and remodelling cytokine basic fibroblast growth factor (bFGF). METHODS: Polymerase chain reaction (PCR) immunocytochemistry/immunohistochemistry and ELISA were employed to detect expression of IL-17RB, IL-17RA and bFGF by human vascular endothelial cells (HUVEC) and immunoreactivity for IL-25 and bFGF in asthmatic bronchial biopsies. Receptor-blocking antibodies, PCR and an in vitro angiogenesis assay were used to investigate whether IL-25 acts on IL-17RB or IL-17RA to induce bFGF expression and angiogenesis. PCR was also employed to investigate the signalling pathways involved in IL-25-mediated bFGF expression. RESULTS: HUVEC constitutively expressed IL-17RB, IL-17RA and bFGF. Production of the latter was further increased by IL-25, but attenuated after blockade of the IL-17RB, but not the IL-17RA receptor. Neutralization of endogenous VEGF and bFGF completely abrogated IL-25-induced angiogenesis which was also inhibited by blocking IL-17RB, but not IL-17RA. The PI3K-specific inhibitor LY294002 also completely attenuated IL-25-induced bFGF expression. Immunoreactivity for IL-25 and bFGF was elevated in the asthmatic bronchial mucosa and the expression of each correlated with the other. CONCLUSIONS AND CLINICAL RELEVANCE: Our data support the hypothesis that IL-25 contributes to elevated bFGF in asthmatic airways by acting on the endothelial cell IL-17RB receptor through PI3K-signalling pathways. Targeting the pathways might benefit therapy of airways remodelling.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Interleucina-17/farmacología , Receptores de Interleucina-17/metabolismo , Células Cultivadas , Células Endoteliales/inmunología , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Interleucina-17/inmunología , Neovascularización Fisiológica/efectos de los fármacos , Receptores de Interleucina-17/antagonistas & inhibidores , Receptores de Interleucina-17/genética , Transducción de Señal/efectos de los fármacos
18.
Wound Repair Regen ; 20(2): 158-65, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22380688

RESUMEN

Wound healing is a sophisticated biologic process. In the case of hemithyroidectomy, the operation time is relatively short with small tissue damage and without skin excision, and bacterial contamination before, during, and after the operation is uncommon. Here, we explored which cytokine(s) affected the rates of healing of skin wounds after hemithyroidectomy of 29 patients. We assessed the amounts of cytokines (e.g., interleukin-6, platelet-derived growth factor, basic fibroblast growth factor, vascular endothelial growth factor, and tumor necrosis factor-α) in either the preoperative or postoperative lavage fluids, or in the drainage fluids on postoperative days (PODs) 1-8. All of these cytokines showed a similar pattern; after reaching a peak on POD1, the production fell sharply on POD2-8, revealing that wound healing commenced on POD1. The rates of wound healing were inversely related to the levels of histamine in six patients (i.e., those with the three largest and those with the three smallest total volumes of drainage fluid on POD1): high (or low) levels of histamine in the postoperative lavage fluids with low (or high) levels in the drainage fluids on POD1 caused earlier (or the delay of) wound healing, suggesting involvement of histamine in the acceleration and delay of wound healing.


Asunto(s)
Citocinas/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Histamina/metabolismo , Interleucina-6/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Tiroidectomía , Factor de Necrosis Tumoral alfa/metabolismo , Cicatrización de Heridas , Citocinas/inmunología , Drenaje , Ensayo de Inmunoadsorción Enzimática , Líquido Extracelular/metabolismo , Femenino , Factor 2 de Crecimiento de Fibroblastos/inmunología , Histamina/inmunología , Humanos , Interleucina-6/inmunología , Masculino , Factor de Crecimiento Derivado de Plaquetas/inmunología , Irrigación Terapéutica , Tiroidectomía/efectos adversos , Factor de Necrosis Tumoral alfa/inmunología
19.
ScientificWorldJournal ; 2012: 607157, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22997500

RESUMEN

OBJECTIVE: To investigate the expression of NOD2 in human VSMCs, its role in the production of inflammatory cytokines in VSMC and the possible interaction of NOD2-mediated signaling pathway with those mediated by TLR2 and TLR4. METHODS: Human coronary artery smooth muscle cells were stimulated with NOD2 agonist MDP alone or in combination with either TLR2 agonist PAM3 or TLR4 agonist LPSs. The mRNA expression of NOD2 and FGF-2 were measured by RT-PCR. The concentration of IL-8 and TNF-α in the culture supernatants was determined by ELISA. VSMC proliferation ability was analyzed by MTT assay. RESULTS: MDP up regulated the expression of NOD2 mRNA in VSMC in a time-dependent manner, up regulated the expression of FGF-2 mRNA in VSMC, induced the production of IL-8 and TNF-α, and promoted the proliferation of VSMC. Additionally, MDP synergied with LPS and PAM3 to promote the proliferation of VSMC and induce the production of IL-8 and TNF-α. CONCLUSION: The activation of NOD2-mediated innate immune signaling pathway can increase the proliferation ability of VSMC and induce the production of inflammatory cytokines in VSMC. It is also shown a synergistic effect with TLR2- and TLR4-mediated signaling pathways in this process.


Asunto(s)
Músculo Liso Vascular/citología , Miocitos del Músculo Liso/inmunología , Proteína Adaptadora de Señalización NOD2/agonistas , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 4/agonistas , Acetilmuramil-Alanil-Isoglutamina/farmacología , Proliferación Celular , Células Cultivadas , Vasos Coronarios/citología , Vasos Coronarios/inmunología , Medios de Cultivo/metabolismo , Sinergismo Farmacológico , Ensayo de Inmunoadsorción Enzimática , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/inmunología , Humanos , Inmunidad Innata , Interleucina-8/inmunología , Lipopéptidos/farmacología , Lipopolisacáridos/farmacología , Músculo Liso Vascular/inmunología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/inmunología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factores de Tiempo , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/inmunología , Factor de Necrosis Tumoral alfa/inmunología
20.
Growth Factors ; 29(6): 290-7, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21981422

RESUMEN

In this work, a novel vaccine delivery system, biodegradable nanoparticles (NPs) in thermosensitive hydrogel, was investigated. Human basic fibroblast growth factor (bFGF)-loaded NPs (bFGF-NPs) were prepared, and then bFGF-NPs were incorporated into thermosensitive hydrogel to form bFGF-NPs in a hydrogel composite (bFGF-NPs/hydrogel). bFGF-NPs/hydrogel was an injectable sol at ambient temperature, but was converted into a non-flowing gel at body temperature. The in vitro release profile showed that bFGF could be released from bFGF-NPs or bFGF-NPs/hydrogel at an extended period, but the release rate of bFGF-NPs/hydrogel was much lower. In vivo experiments suggested that immunogenicity of bFGF improved significantly after being incorporated into the NPs/hydrogel composite, and strong humoral immunity was maintained for longer than 12 weeks. Furthermore, an in vivo protective anti-tumor immunity assay indicated that immunization with bFGF-NPs/hydrogel could induce significant suppression of the growth and metastases of tumors. Thus, the NPs/hydrogel composite may have great potential application as a novel vaccine delivery system.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Carcinoma Pulmonar de Lewis/terapia , Factor 2 de Crecimiento de Fibroblastos/inmunología , Hidrogeles , Nanocápsulas , Vacunación/métodos , Animales , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/prevención & control , Femenino , Factor 2 de Crecimiento de Fibroblastos/administración & dosificación , Humanos , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA