Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Biochim Biophys Acta Rev Cancer ; 1878(3): 188892, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37004960

RESUMEN

Vestigial-like 1 (VGLL1) is a recently discovered driver of proliferation and invasion that is expressed in many aggressive human malignancies and is strongly associated with poor prognosis. The VGLL1 gene encodes for a co-transcriptional activator that shows intriguing structural similarity to key activators in the hippo pathway, providing important clues to its functional role. VGLL1 binds to TEAD transcription factors in an analogous fashion to YAP1 but appears to activate a distinct set of downstream gene targets. In mammals, VGLL1 expression is found almost exclusively in placental trophoblasts, cells that share many hallmarks of cancer. Due to its role as a driver of tumor progression, VGLL1 has become a target of interest for potential anticancer therapies. In this review, we discuss VGLL1 from an evolutionary perspective, contrast its role in placental and tumor development, summarize the current knowledge of how signaling pathways can modulate VGLL1 function, and discuss potential approaches for targeting VGLL1 therapeutically.


Asunto(s)
Proteínas de Unión al ADN , Neoplasias , Animales , Femenino , Humanos , Embarazo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Placenta/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción de Dominio TEA , Neoplasias/genética , Mamíferos/metabolismo
2.
Biomedicines ; 10(10)2022 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-36289691

RESUMEN

Lung cancer continues to be the deadliest cancer worldwide. A new strategy of combining chemotherapeutics with naturally occurring anticancer compounds, such as lactoferrin, might improve the efficacy and toxicity of current chemotherapy. The aim of this study was to evaluate the effect of recombinant human lactoferrin (rhLf) in combination with etoposide on anticancer activity in human lung adenocarcinoma cells. In addition, we examined the impact of rhLf on etoposide-induced cytotoxicity of human endothelial cells. We found that treatment of A549 cells with a combination of etoposide and rhLf resulted in significantly greater inhibition of cancer cell growth as compared to etoposide alone. The combination repressed cancer cell growth by cell cycle arrest in the G2/M phase and induction of apoptosis. In contrast to cancer cells, rhLf did not affect endothelial cell viability. Importantly, rhLf significantly diminished the etoposide-induced cytotoxicity of endothelial cells. Analysis of the type of drug interaction based on combination index value showed that rhLf synergized with etoposide to induce anticancer activity. The calculated dose reduction index indicated that the combination treatment reduced a 10-fold of etoposide dose to achieve the same anticancer effect. Our data demonstrate that rhLf enhanced the anticancer activity of etoposide and diminished etoposide-induced cytotoxic effect in endothelial cells.

3.
Science ; 374(6575): 1632-1640, 2021 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-34941392

RESUMEN

Gut bacteria modulate the response to immune checkpoint blockade (ICB) treatment in cancer, but the effect of diet and supplements on this interaction is not well studied. We assessed fecal microbiota profiles, dietary habits, and commercially available probiotic supplement use in melanoma patients and performed parallel preclinical studies. Higher dietary fiber was associated with significantly improved progression-free survival in 128 patients on ICB, with the most pronounced benefit observed in patients with sufficient dietary fiber intake and no probiotic use. Findings were recapitulated in preclinical models, which demonstrated impaired treatment response to anti­programmed cell death 1 (anti­PD-1)­based therapy in mice receiving a low-fiber diet or probiotics, with a lower frequency of interferon-γ­positive cytotoxic T cells in the tumor microenvironment. Together, these data have clinical implications for patients receiving ICB for cancer.


Asunto(s)
Fibras de la Dieta , Microbioma Gastrointestinal , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Melanoma/terapia , Probióticos , Animales , Estudios de Cohortes , Ácidos Grasos Volátiles/análisis , Trasplante de Microbiota Fecal , Heces/química , Heces/microbiología , Femenino , Humanos , Inmunoterapia , Masculino , Melanoma/inmunología , Melanoma/microbiología , Melanoma Experimental/inmunología , Melanoma Experimental/microbiología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Supervivencia sin Progresión , Linfocitos T
4.
Arch Immunol Ther Exp (Warsz) ; 69(1): 33, 2021 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-34748082

RESUMEN

Lung cancer remains the leading cause of cancer death worldwide. Despite the recent advances in cancer treatment, only a subset of patients responds to targeted and immune therapies, and many patients developing resistance after an initial response. Lactoferrin (Lf) is a natural glycoprotein with immunomodulatory and anticancer activities. We produced a novel recombinant human Lf (rhLf) that exhibits glycosylation profile compatible with the natural hLf for potential parenteral therapeutic applications. The aim of this study was to evaluate the anticancer effects of this novel rhLf in human lung adenocarcinoma cells and its mechanisms of action. The results showed a concentration-dependent inhibition of A549 cancer cell growth in response to rhLf. Treatment with 1 mg/ml of rhLf for 24 h and 72 h resulted in a significant inhibition of cancer cell growth by 32% and 25%, respectively. Moreover, rhLf increased fourfold the percentage of early and late apoptotic cells compared to the control. This effect was accompanied by increased levels of caspase-3 activity and cell cycle arrest at the S phase in rhLf-treated cancer cells. Furthermore, rhLf significantly attenuated A549 cell migration. Importantly, treatment of normal human bronchial epithelial (NHBE) cells with rhLf showed the cell viability and morphology comparable to the control. In contrast, chemotherapeutic etoposide induced cytotoxicity in NHBE cells and reduced the cell viability by 40%. These results demonstrate the selective anticancer effects of rhLf against lung adenocarcinoma cells without cytotoxicity on normal human cells. This study highlights a potential for clinical utility of this novel rhLf in patients with lung cancer.


Asunto(s)
Adenocarcinoma del Pulmón , Lactoferrina , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/genética , Proliferación Celular , Células Epiteliales/metabolismo , Humanos , Lactoferrina/metabolismo , Proteínas Recombinantes
5.
Biochem Cell Biol ; 99(1): 47-53, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32476452

RESUMEN

The immunomodulatory nature of lactoferrin (LF) derives from its ability to bridge innate and adaptive immunity in obtaining physiological equilibrium. LF is an attractive molecule for treatment of diseases that compromise immune homeostasis. Oral delivery is a preferable method for LF administration; however, its bioavailability is affected by protein degradation and absorption. The aim of this study was to evaluate the systemic effects of orally and intravenously (IV) administered recombinant human LF (rhLF) on blood cell transcriptome profiling. Rats were administered a single dose of rhLF by gavage or IV. The transcriptome profiles from the control and the rhLF-treated rats after 3, 6, and 24 h were analyzed using a Clariom D microarray. The results showed differentially expressed genes in response to IV as well as oral administered rhLF including coding and noncoding RNAs. Moreover, a comparison of the differentially expressed genes between oral and IV administration of LF, after 6 h, revealed that the majority (72.8%) of the genes altered in response to oral administration of rhLF were the same as for the IV treatment. The pathway profiles showed similarities in up-regulation of specific genes involved in oxidative stress and inflammatory responses for both routes of treatments. These findings provide evidence of the systemic signal transduction effects of orally administered rhLF.


Asunto(s)
Lactoferrina/genética , Administración Oral , Animales , Perfilación de la Expresión Génica , Humanos , Inyecciones Intravenosas , Lactoferrina/administración & dosificación , Masculino , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética
6.
Nat Commun ; 11(1): 661, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-32005826

RESUMEN

High dose interleukin-2 (IL-2) is active against metastatic melanoma and renal cell carcinoma, but treatment-associated toxicity and expansion of suppressive regulatory T cells (Tregs) limit its use in patients with cancer. Bempegaldesleukin (NKTR-214) is an engineered IL-2 cytokine prodrug that provides sustained activation of the IL-2 pathway with a bias to the IL-2 receptor CD122 (IL-2Rß). Here we assess the therapeutic impact and mechanism of action of NKTR-214 in combination with anti-PD-1 and anti-CTLA-4 checkpoint blockade therapy or peptide-based vaccination in mice. NKTR-214 shows superior anti-tumor activity over native IL-2 and systemically expands anti-tumor CD8+ T cells while inducing Treg depletion in tumor tissue but not in the periphery. Similar trends of intratumoral Treg dynamics are observed in a small cohort of patients treated with NKTR-214. Mechanistically, intratumoral Treg depletion is mediated by CD8+ Teff-associated cytokines IFN-γ and TNF-α. These findings demonstrate that NKTR-214 synergizes with T cell-mediated anti-cancer therapies.


Asunto(s)
Carcinoma de Células Renales/tratamiento farmacológico , Interleucina-2/análogos & derivados , Melanoma/tratamiento farmacológico , Polietilenglicoles/administración & dosificación , Profármacos/administración & dosificación , Linfocitos T Reguladores/inmunología , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Linfocitos T CD8-positivos/inmunología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/inmunología , Estudios de Cohortes , Quimioterapia Combinada , Femenino , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-2/administración & dosificación , Interleucina-2/agonistas , Interleucina-2/inmunología , Ipilimumab/administración & dosificación , Activación de Linfocitos/efectos de los fármacos , Melanoma/genética , Melanoma/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores de Interleucina-2/genética , Receptores de Interleucina-2/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
7.
Radiat Res ; 186(2): 162-74, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27388041

RESUMEN

There is increasing evidence that radiation-induced damage to endothelial cells and loss of endothelial function may contribute to both acute radiation syndromes and long-term effects of whole-body nuclear irradiation. Therefore, several drugs are being developed to mitigate the effects of nuclear radiation, most of these drugs will target and protect or regenerate leukocytes and platelets. Our laboratory has demonstrated that TP508, a 23-amino acid thrombin peptide, activates endothelial cells and stem cells to revascularize and regenerate tissues. We now show that TP508 can mitigate radiation-induced damage to endothelial cells in vitro and in vivo. Our in vitro results demonstrate that human endothelial cells irradiation attenuates nitric oxide (NO) signaling, disrupts tube formation and induces DNA double-strand breaks (DSB). TP508 treatment reverses radiation effects on NO signaling, restores tube formation and accelerates the repair of radiation-induced DSB. The radiation-mitigating effects of TP508 on endothelial cells were also seen in CD-1 mice where systemic injection of TP508 stimulated endothelial cell sprouting from aortic explants after 8 Gy irradiation. Systemic doses of TP508 that mitigated radiation-induced endothelial cell damage, also significantly increased survival of CD-1 mice when injected 24 h after 8.5 Gy exposure. These data suggest that increased survival observed with TP508 treatment may be due to its effects on vascular and microvascular endothelial cells. Our study supports the usage of a regenerative drug such as TP508 to activate endothelial cells as a countermeasure for mitigating the effects of nuclear radiation.


Asunto(s)
Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Células Endoteliales/efectos de los fármacos , Células Endoteliales/efectos de la radiación , Fragmentos de Péptidos/farmacología , Trombina/farmacología , Secuencia de Aminoácidos , Animales , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Masculino , Ratones , Óxido Nítrico/biosíntesis , Análisis de Supervivencia
8.
J Vasc Res ; 50(3): 186-96, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23594718

RESUMEN

Revascularization of chronic wounds and ischemic tissue is attenuated by endothelial dysfunction and the inability of angiogenic factors to stimulate angiogenesis. We recently showed that TP508, a nonproteolytic thrombin peptide, increases perfusion and NO-dependent vasodilation in hearts with chronic ischemia and stimulates NO production by endothelial cells. In this study, we investigated systemic in vivo effects of TP508 on VEGF-stimulated angiogenesis in vitro using aortic explants in normoxic and hypoxic conditions. Mice were injected with saline or TP508 and 24 h later aortas were removed and cultured to quantify endothelial sprouting. TP508 injection increased endothelial sprouting and potentiated the in vitro response to VEGF. Exposure of control explants to hypoxia inhibited basal and VEGF-stimulated endothelial cell sprouting. This effect of hypoxia was significantly prevented by TP508 injection. Thus, TP508 systemic administration increases responsiveness of aortic endothelial cells to VEGF and diminishes the effect of chronic hypoxia on endothelial cell sprouting. Studies using human endothelial cells in culture suggest that protective effects of TP508 during hypoxia may involve stimulation of endothelial cell NO production. These data suggest potential clinical benefit of using a combination of systemic TP508 and local VEGF as a therapy for revascularization of ischemic tissue.


Asunto(s)
Inductores de la Angiogénesis , Hipoxia de la Célula/efectos de los fármacos , Fragmentos de Péptidos/administración & dosificación , Trombina/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/farmacología , Secuencia de Aminoácidos , Animales , Aorta Torácica/citología , Aorta Torácica/fisiología , Sinergismo Farmacológico , Quimioterapia Combinada , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Inyecciones Intravenosas , Masculino , Ratones , Ratones Endogámicos ICR , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Trombina/química , Técnicas de Cultivo de Tejidos
9.
J Vasc Res ; 47(3): 203-13, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19893317

RESUMEN

TP508, a 23-amino-acid peptide representing a portion of human thrombin, promotes tissue revascularization and repair. The molecular mechanisms of TP508 action, however, remain unclear. Nitric oxide (NO) plays a crucial role in regulation of angiogenesis and wound healing. We, therefore, investigated TP508 effects on NO production in human endothelial cells. TP508 stimulated a rapid, dose-dependent, 2- to 4-fold increase in NO production. TP508 induced NO release as early as 5 min. Continued exposure to TP508 for 1-24 h increased NO concentrations over controls by 100.5 +/- 9.6 and 463.3 +/- 24.2 nM, respectively. These levels of NO release were similar to those produced in response to vascular endothelial growth factor (VEGF). TP508- and VEGF-induced NO production was decreased by inhibitors of PI-3K (LY294002) and Src (PP2). TP508 stimulated early transient phosphorylation of Src and Akt. In contrast to VEGF, TP508 stimulation of NO release was inhibited by PKC inhibitor (Go6976) and was independent of intracellular calcium mobilization. These results demonstrate that TP508 and VEGF stimulate NO production to similar levels but through distinct pathways. This study provides new insights into the initial molecular mechanisms by which TP508 may stimulate diverse cellular effects leading to tissue revascularization and wound healing.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Óxido Nítrico/metabolismo , Fragmentos de Péptidos/farmacología , Trombina/farmacología , Animales , Calcio/metabolismo , Bovinos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
10.
Am J Physiol Cell Physiol ; 296(5): C1162-70, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19244479

RESUMEN

Coronary artery disease results in progressive vascular stenosis associated with chronic myocardial ischemia. Vascular endothelial growth factor (VEGF) stimulates endothelial cell angiogenic responses to revascularize ischemic tissues; however, the effect of chronic hypoxia on the responsiveness of endothelial cells to VEGF remains unclear. We, therefore, investigated whether hypoxia alters VEGF-stimulated signaling and angiogenic responses in primary human coronary artery endothelial (HCAE) cells. Exposure of HCAE cells to hypoxia (1% O(2)) for 24 h decreased VEGF-stimulated endothelial cell migration ( approximately 82%), proliferation ( approximately 30%), and tube formation. Hypoxia attenuated VEGF-stimulated activation of endothelial nitric oxide (NO) synthase (eNOS) ( approximately 72%) and reduced NO production in VEGF-stimulated cells from 237 +/- 38.8 to 61.3 +/- 28.4 nmol/l. Moreover, hypoxia also decreased the ratio of phosphorylated eNOS to total eNOS in VEGF-stimulated cells by approximately 50%. This effect was not observed in thrombin-stimulated cells, suggesting that hypoxia specifically inhibited VEGF signaling upstream of eNOS phosphorylation. VEGF-induced activation of Akt, ERK1/2, p38, p70S6 kinases, and S6 ribosomal protein was also attenuated in hypoxic cells. Moreover, VEGF-stimulated phosphorylation of VEGF receptor-2 (KDR) at Y996 and Y1175 was decreased by hypoxia. This decrease correlated with a 70 +/- 12% decrease in KDR protein expression. Analysis of mRNA from these cells showed that hypoxia reduced steady-state levels of KDR mRNA by 52 +/- 16% and decreased mRNA stability relative to normoxic cells. Our findings demonstrate that chronic hypoxia attenuates VEGF-stimulated signaling in HCAE cells by specific downregulation of KDR expression. These data provide a novel explanation for the impaired angiogenic responses to VEGF in endothelial cells exposed to chronic hypoxia.


Asunto(s)
Células Endoteliales/metabolismo , Hipoxia/metabolismo , Isquemia Miocárdica/metabolismo , Neovascularización Fisiológica/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Adulto , Capilares/citología , Capilares/metabolismo , División Celular/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Enfermedad Crónica , Vasos Coronarios/citología , Regulación hacia Abajo/fisiología , Células Endoteliales/citología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Expresión Génica/fisiología , Humanos , Hipoxia/patología , Isquemia Miocárdica/patología , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
J Cardiovasc Pharmacol Ther ; 13(3): 214-25, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18757834

RESUMEN

Endothelial dysfunction (ED) is characterized by impaired nitric oxide (NO) signaling, decreased NO-dependent vasodilatation, increased vascular inflammation, and diminished response to angiogenic factors. TP508 (Chrysalin), an angiogenic tissue repair peptide, was tested for potential effects on myocardial revascularization and ED using a porcine model of chronic myocardial ischemia. TP508 increased perfusion in ischemic regions up to16-fold (P < .02) and doubled myocardial wall thickening (P < .02) relative to placebo controls. Ischemic arterioles exhibited impaired NO-mediated vasodilation and diminished NO production. TP508 reversed ischemic effects, increasing NO-mediated vasodilation (P < .05), endothelial nitric oxide synthase (eNOS) expression, and NO production. In human endothelial cells, TP508 stimulated eNOS activation (1.84 +/- 0.2-fold; P < .02), increased NO production (85 +/- 18%; P < .02), and prevented hypoxia-induced eNOS downregulation (P < .01). Thus, TP508 reverses ED both in porcine ischemic hearts and cultured human endothelial cells. These results suggest potential therapeutic benefit of TP508 in myocardial revascularization and treatment of ED-related diseases.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Circulación Coronaria/efectos de los fármacos , Vasos Coronarios/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Isquemia Miocárdica/tratamiento farmacológico , Revascularización Miocárdica/métodos , Fragmentos de Péptidos/farmacología , Animales , Hipoxia de la Célula , Células Cultivadas , Enfermedad Crónica , Angiografía Coronaria , Vasos Coronarios/metabolismo , Vasos Coronarios/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ecocardiografía de Estrés , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Humanos , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Isquemia Miocárdica/fisiopatología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Porcinos , Porcinos Enanos , Trombina , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
12.
Expert Opin Pharmacother ; 9(15): 2717-26, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18803458

RESUMEN

Rusalatide acetate (Chrysalin) is an investigational drug being evaluated for treatment of chronic wounds and fractures. Rusalatide acetate interacts with cell surface receptors to stimulate a cascade of cellular and molecular wound healing events, including activation of nitric oxide signaling. Rusalatide acetate significantly accelerated healing of diabetic foot ulcers and distal radius fractures in Phase I/II clinical trials. Subsequently, in one of the largest Phase III fracture studies to date, rusalatide acetate showed significant acceleration of distal radius fracture healing radiographically but failed to meet its primary clinical endpoint - time to removal of immobilization - within the intent-to-treat population. Subset analysis showed that rusalatide acetate met this primary clinical endpoint and significantly accelerated radiographic healing in osteopenic women. Rusalatide acetate may therefore show its greatest efficacy in healing-impaired patients.


Asunto(s)
Pie Diabético/tratamiento farmacológico , Curación de Fractura/efectos de los fármacos , Fragmentos de Péptidos/uso terapéutico , Trombina/uso terapéutico , Secuencia de Aminoácidos , Ensayos Clínicos como Asunto , Humanos , Datos de Secuencia Molecular , Fragmentos de Péptidos/efectos adversos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacocinética , Fragmentos de Péptidos/farmacología , Trombina/efectos adversos , Trombina/química , Trombina/farmacocinética , Trombina/farmacología
13.
J Immunol ; 180(6): 4182-90, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18322230

RESUMEN

Reversal of eosinophilic inflammation has been an elusive therapeutic goal in the management of asthma pathogenesis. In this regard, GM-CSF is a primary candidate cytokine regulating eosinophil activation and survival in the lung; however, its molecular mechanism of propagation and maintenance of stimulated eosinophil activation is not well understood. In this study, we elucidate those late interactions occurring between the GM-CSF receptor and activated eosinophil signaling molecules. Using coimmunoprecipitation with GM-CSF-stimulated eosinophils, we have identified that the GM-CSF receptor beta-chain (GMRbeta) interacted with ICAM-1 and Shp2 phosphatase, as well as Slp76 and ADAP adaptor proteins. Separate experiments using affinity binding with a tyrosine-phosphorylated peptide containing an ITIM (ICAM-1 residues 480-488) showed binding to Shp2 phosphatase and GMRbeta. However, the interaction of GMRbeta with the phosphorylated ICAM-1-derived peptide was observed only with stimulated eosinophil lysates, suggesting that the interaction of GMRbeta with ICAM-1 required phosphorylated Shp2 and/or phosphorylated GMRbeta. Importantly, we found that inhibition of ICAM-1 in activated eosinophils blocked GM-CSF-induced expression of c-fos, c-myc, IL-8, and TNF-alpha. Moreover, inhibition of ICAM-1 expression with either antisense oligonucleotide or an ICAM-1-blocking Ab effectively inhibited ERK activation and eosinophil survival. We concluded that the interaction between ICAM-1 and the GM-CSF receptor was essential for GM-CSF-induced eosinophil activation and survival. Taken together, these results provide novel mechanistic insights defining the interaction between ICAM-1 and the GM-CSF receptor and highlight the importance of targeting ICAM-1 and GM-CSF/IL-5/IL-3 receptor systems as a therapeutic strategy to counter eosinophilia in asthma.


Asunto(s)
Subunidad beta Común de los Receptores de Citocinas/metabolismo , Eosinófilos/citología , Eosinófilos/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Receptor Cross-Talk , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Transducción de Señal , Supervivencia Celular/inmunología , Células Cultivadas , Subunidad beta Común de los Receptores de Citocinas/fisiología , Eosinófilos/enzimología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Humanos , Molécula 1 de Adhesión Intercelular/fisiología , Péptidos/metabolismo , Péptidos/fisiología , Unión Proteica/inmunología , Subunidades de Proteína/metabolismo , Subunidades de Proteína/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/fisiología , Receptor Cross-Talk/inmunología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Transducción de Señal/inmunología
14.
J Biol Chem ; 279(39): 40400-4, 2004 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-15292208

RESUMEN

Src activity is elevated in a majority of colonic and pancreatic cancers and is associated with late stage aggressive cancers. However, the mechanisms leading to its increased activity remain largely undefined. Agonist binding to the cholecystokinin-2 (CCK2)/gastrin receptor (CCK2R), a G-protein-coupled receptor, increases Src activity in a variety of normal and neoplastic cell lines. Recently, we and others (Hellmich, M. R., Rui, X. L., Hellmich, H. L., Fleming, R. Y., Evers, B. M., and Townsend, C. M., Jr. (2000) J. Biol. Chem. 275, 32122-32128; Ding, W. Q., Kuntz, S. M., and Miller, L. J. (2002) Cancer Res. 62, 947-952; Smith, J. P., Verderame, M. F., McLaughlin, P., Martenis, M., Ballard, E., and Zagon, I. S. (2002) Int. J. Mol. Med. 10, 689-694) have identified a splice variant of CCK2R, called CCK2i4svR, that is expressed in human colorectal and pancreatic cancers but not by cells of the adjacent nonmalignant tissue. Compared with CCK2R, CCK2i4svR contains an additional 69 amino acids within its third intracellular loop (3il) domain. Because CCK2i4svR is the only splice variant expressed in some human colon and pancreatic cancers, we questioned whether CCK2i4svR could regulate Src activity. Stably transfected HEK293 cells were used because, unlike many cancer-derived cells, they have a low level of basal Src activity. We report that, in contrast to CCK2R, CCK2i4svR activates Src kinase in the absence of agonist stimulation. In vitro kinase assay of immunoprecipitated receptor protein showed a 6-8-fold increase in Src kinase activity associated with CCK2i4svR compared with CCK2R. Expression of the 3il domain of the CCK2i4svR alone was sufficient to partially activate Src kinase. Together, these data support the hypothesis that the increased Src activity observed in some pancreatic and colorectal cancers is due, in part, to the co-expression of CCK2i4svR.


Asunto(s)
Receptor de Colecistoquinina B/genética , Familia-src Quinasas/metabolismo , Empalme Alternativo , Aminoácidos/química , Western Blotting , Línea Celular , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Activación Enzimática , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/metabolismo , Neoplasias Pancreáticas/metabolismo , Fosforilación , Plásmidos/metabolismo , Pruebas de Precipitina , Unión Proteica , Estructura Terciaria de Proteína , Receptor de Colecistoquinina B/metabolismo , Factores de Tiempo , Transfección
15.
J Biol Chem ; 279(3): 1853-60, 2004 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-14602717

RESUMEN

Small differences in amplitude, duration, and temporal patterns of change in the concentration of free intracellular Ca2+ ([Ca2+](i)) can profoundly affect cell physiology, altering programs of gene expression, cell proliferation, secretory activity, and cell survival. We report a novel mechanism for amplitude modulation of [Ca2+](i) that involves mitogen-activated protein kinase (MAPK). We show that epidermal growth factor (EGF) potentiates gastrin-(1-17) (G17)-stimulated Ca2+ release from intracellular Ca2+ stores through a MAPK-dependent pathway. G17 activation of the cholecystokinin/gastrin receptor (CCK(2)R), a G protein-coupled receptor, stimulates release of Ca2+ from inositol 1,4,5-triphosphate-sensitive Ca2+ stores. Pretreating rat intestinal epithelial cells expressing CCK(2)R with EGF increased the level of G17-stimulated Ca2+ release from intracellular stores. The stimulatory effect of EGF on CCK(2)R-mediated Ca2+ release requires activation of the MAPK kinase (MEK)1,2/extracellular signal-regulated kinase (ERK)1,2 pathway. Inhibition of the MEK1,2/ERK1,2 pathway by either serum starvation or treatment with selective MEK1,2 inhibitors PD98059 and U0126 or expression of a dominant-negative mutant form of MEK1 decreased the amplitude of the G17-stimulated Ca2+ release response. Activation of the MEK1,2/ERK1,2 pathway either by pretreating cells with EGF or by expression of constitutively active K-ras (K-rasV12G) or MEK1 (MEK1*) increased the amplitude of G17-stimulated Ca2+ release. Although EGF, MEK1*, and K-rasV12G activated the MEK1,2/ERK1,2 pathway, they did not increase [Ca2+](i) in the absence of G17. These data demonstrate that the activation state of the MEK1,2/ERK1,2 pathway can modulate the amplitude of the CCK(2)R-mediated Ca2+ release response and identify a novel mechanism for cross-talk between EGF receptor- and CCK(2)R-regulated signaling pathways.


Asunto(s)
Calcio/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Receptores de Colecistoquinina/fisiología , Animales , Activación Enzimática , Gastrinas/farmacología , Humanos , Fosfatidilinositol 3-Quinasas/fisiología , Fosforilación , Ratas
16.
Cancer Res ; 63(13): 3495-502, 2003 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12839933

RESUMEN

The majority of deaths from prostate cancer occur in patients with androgen-insensitive metastatic disease. An important early event in the development of the metastatic phenotype is the induction of genes that promote angiogenesis, such as vascular endothelial growth factor (VEGF) and interleukin-8 (IL-8), which are released from tumor cells into their microenvironment. Coincident with progression from prostatic carcinoma in situ to metastatic disease is an increase in the number of tumor cells exhibiting neuroendocrine (NE) differentiation. NE cells express a variety of peptide hormones, including the bombesin (BBS)-like peptide, gastrin-releasing peptide (GRP), and its cognate receptor, GRP-R. Although there is a strong positive correlation between the degree of NE differentiation and the metastatic potential of prostate cancers, a mechanistic link between increased expression of peptide hormone receptors, such as GRP-R, and proangiogenic gene expression has not been established. Here we report that BBS stimulates nuclear factor kappa B (NF kappa B) activation and proangiogenic gene expression in the androgen-insensitive prostate cancer cells lines, PC-3 and DU-145. In PC-3 cells, BBS stimulation of GRP-R resulted in the up-regulation of IL-8 and VEGF expression through a NF kappa B-dependent pathway. We show that BBS treatment induced inhibitor of NF kappa B degradation, NF kappa B translocation to the cell nucleus, increased NF kappa B binding to its DNA consensus sequence, and increased IL-8 and VEGF mRNA expression and protein secretion. Treatment with the proteasome inhibitor, MG-132, blocked BBS-stimulated NF kappa B DNA binding, and IL-8 and VEGF expression and secretion. Finally, media collected from PC-3 cell cultures, after BBS treatment, stimulated an NF kappa B-dependent migration of human umbilical vascular endothelial cells in vitro. Together, our data demonstrate a role for BBS and GRP-R in the NF kappa B-dependent up-regulation of proangiogenic gene expression, and suggest a possible molecular mechanism linking NE differentiation and the increased metastatic potential of androgen-insensitive prostate cancers.


Asunto(s)
Bombesina/farmacología , Factores de Crecimiento Endotelial/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/genética , Interleucina-8/genética , Linfocinas/genética , FN-kappa B/metabolismo , Neovascularización Patológica/patología , Neoplasias de la Próstata/patología , Animales , División Celular/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/genética , Transcripción Genética , Trasplante Heterólogo , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
17.
Am J Physiol Lung Cell Mol Physiol ; 283(2): L364-72, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12114198

RESUMEN

Airway epithelial cells represent the primary cell target of respiratory syncytial virus (RSV) infection. They actively participate in the lung immune/inflammatory response that follows RSV infection by expressing chemokines, small chemotactic cytokines that recruit and activate leukocytes. Regulated on activation, normal T cell expressed, and presumably secreted (RANTES) is a member of the CC chemokine subfamily and is strongly chemotactic for T lymphocytes, monocytes, basophils, and eosinophils, cell types that are present or activated in the inflammatory infiltrate that follows RSV infection of the lung. RSV infection of airway epithelial cells induces RANTES expression by increasing gene transcription and stabilizing RNA transcripts. The signaling pathway regulating RANTES gene expression after RSV infection has not been determined. In this study, we examined the role of extracellular signal-regulated kinase (ERK) and p38, members of the mitogen-activated protein (MAP) kinase (MAPK) family, in RSV-induced RANTES production. RSV infection of alveolar epithelial cells induced increased phosphorylation and catalytic activity of ERK and the upstream kinases Raf-1 and MAP ERK kinase. Induction of the MAP signaling cascade required a replication-competent virus. RSV infection of alveolar epithelial cells also induced activation of p38 MAPK. Inhibition of ERK and p38 activation significantly reduced RSV-induced RANTES mRNA and protein secretion without affecting RANTES gene transcription or transcription factor activation. These results indicate that the MAPK signaling cascade regulates RANTES production in alveolar epithelial cells through a posttranscriptional mechanism.


Asunto(s)
Quimiocina CCL5/genética , Regulación de la Expresión Génica/fisiología , Quinasa 1 de Quinasa de Quinasa MAP , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Procesamiento Proteico-Postraduccional , Infecciones por Virus Sincitial Respiratorio/genética , Bronquios/citología , Bronquios/metabolismo , Línea Celular , Quimiocina CCL5/metabolismo , Activación Enzimática/fisiología , Células Epiteliales/metabolismo , Humanos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Alveolos Pulmonares/citología , Alveolos Pulmonares/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/fisiología , Replicación Viral , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA