Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Gastroenterology ; 159(4): 1390-1405.e20, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32534933

RESUMEN

BACKGROUND & AIM: Myosin VB (MYO5B) is an essential trafficking protein for membrane recycling in gastrointestinal epithelial cells. The inactivating mutations of MYO5B cause the congenital diarrheal disease, microvillus inclusion disease (MVID). MYO5B deficiency in mice causes mislocalization of SGLT1 and NHE3, but retained apical function of CFTR, resulting in malabsorption and secretory diarrhea. Activation of lysophosphatidic acid (LPA) receptors can improve diarrhea, but the effect of LPA on MVID symptoms is unclear. We investigated whether LPA administration can reduce the epithelial deficits in MYO5B-knockout mice. METHODS: Studies were conducted with tamoxifen-induced, intestine-specific knockout of MYO5B (VilCreERT2;Myo5bflox/flox) and littermate controls. Mice were given LPA, an LPAR2 agonist (GRI977143), or vehicle for 4 days after a single injection of tamoxifen. Apical SGLT1 and CFTR activities were measured in Üssing chambers. Intestinal tissues were collected, and localization of membrane transporters was evaluated by immunofluorescence analysis in tissue sections and enteroids. RNA sequencing and enrichment analysis were performed with isolated jejunal epithelial cells. RESULTS: Daily administration of LPA reduced villus blunting, frequency of multivesicular bodies, and levels of cathepsins in intestinal tissues of MYO5B-knockout mice compared with vehicle administration. LPA partially restored the brush border height and the localization of SGLT1 and NHE3 in small intestine of MYO5B-knockout mice and enteroids. The SGLT1-dependent short-circuit current was increased and abnormal CFTR activities were decreased in jejunum from MYO5B-knockout mice given LPA compared with vehicle. CONCLUSIONS: LPA may regulate a MYO5B-independent trafficking mechanism and brush border maturation, and therefore be developed for treatment of MVID.


Asunto(s)
Lisofosfolípidos/uso terapéutico , Síndromes de Malabsorción/tratamiento farmacológico , Síndromes de Malabsorción/patología , Microvellosidades/patología , Mucolipidosis/tratamiento farmacológico , Mucolipidosis/patología , Miosina Tipo V/deficiencia , Transportador 1 de Sodio-Glucosa/metabolismo , Animales , Modelos Animales de Enfermedad , Enterocitos/patología , Síndromes de Malabsorción/etiología , Ratones , Ratones Noqueados , Mucolipidosis/etiología
2.
Mol Pharmacol ; 85(1): 62-73, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24136993

RESUMEN

Interstitial adenosine stimulates neovascularization in part through A2B adenosine receptor-dependent upregulation of vascular endothelial growth factor (VEGF). In the current study, we tested the hypothesis that A2B receptors upregulate JunB, which can contribute to stimulation of VEGF production. Using the human microvascular endothelial cell line, human mast cell line, mouse cardiac Sca1-positive stromal cells, and mouse Lewis lung carcinoma (LLC) cells, we found that adenosine receptor-dependent upregulation of VEGF production was associated with an increase in VEGF transcription, activator protein-1 (AP-1) activity, and JunB accumulation in all cells investigated. Furthermore, the expression of JunB, but not the expression of other genes encoding transcription factors from the Jun family, was specifically upregulated. In LLC cells expressing A2A and A2B receptor transcripts, only the nonselective adenosine agonist NECA (5'-N-ethylcarboxamidoadenosine), but not the selective A2A receptor agonist CGS21680 [2-p-(2-carboxyethyl) phenylethylamino-5'-N-ethylcarboxamidoadenosine], significantly increased JunB reporter activity and JunB nuclear accumulation, which were inhibited by the A2B receptor antagonist PSB603 [(8-[4-[4-((4-chlorophenzyl)piperazide-1-sulfonyl)phenyl]]-1-propylxanthine]. Using activators and inhibitors of intracellular signaling, we demonstrated that A2B receptor-dependent accumulation of JunB protein and VEGF secretion share common intracellular pathways. NECA enhanced JunB binding to the murine VEGF promoter, whereas mutation of the high-affinity AP-1 site (-1093 to -1086) resulted in a loss of NECA-dependent VEGF reporter activity. Finally, NECA-dependent VEGF secretion and reporter activity were inhibited by the expression of a dominant negative JunB or by JunB knockdown. Thus, our data suggest an important role of the A2B receptor-dependent upregulation of JunB in VEGF production and possibly other AP-1-regulated events.


Asunto(s)
Receptor de Adenosina A2B/metabolismo , Factores de Transcripción/metabolismo , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Adenosina/metabolismo , Animales , Línea Celular , Núcleo Celular/metabolismo , Humanos , Ratones , Mutación , Regiones Promotoras Genéticas , Interferencia de ARN , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/genética , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/genética
3.
J Immunol ; 187(11): 6120-9, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22039302

RESUMEN

Extracellular adenosine and purine nucleotides are elevated in many pathological situations associated with the expansion of CD11b(+)Gr1(+) myeloid-derived suppressor cells (MDSCs). Therefore, we tested whether adenosinergic pathways play a role in MDSC expansion and functions. We found that A(2B) adenosine receptors on hematopoietic cells play an important role in accumulation of intratumoral CD11b(+)Gr1(high) cells in a mouse Lewis lung carcinoma model in vivo and demonstrated that these receptors promote preferential expansion of the granulocytic CD11b(+)Gr1(high) subset of MDSCs in vitro. Flow cytometry analysis of MDSCs generated from mouse hematopoietic progenitor cells revealed that the CD11b(+)Gr-1(high) subset had the highest levels of CD73 (ecto-5'-nucleotidase) expression (Δmean fluorescence intensity [MFI] of 118.5 ± 16.8), followed by CD11b(+)Gr-1(int) (ΔMFI of 57.9 ± 6.8) and CD11b(+)Gr-1(-/low) (ΔMFI of 12.4 ± 1.0) subsets. Even lower levels of CD73 expression were found on Lewis lung carcinoma tumor cells (ΔMFI of 3.2 ± 0.2). The high levels of CD73 expression in granulocytic CD11b(+)Gr-1(high) cells correlated with high levels of ecto-5'-nucleotidase enzymatic activity. We further demonstrated that the ability of granulocytic MDSCs to suppress CD3/CD28-induced T cell proliferation was significantly facilitated in the presence of the ecto-5'-nucleotidase substrate 5'-AMP. We propose that generation of adenosine by CD73 expressed at high levels on granulocytic MDSCs may promote their expansion and facilitate their immunosuppressive activity.


Asunto(s)
Adenosina/metabolismo , Granulocitos/metabolismo , Células Mieloides/metabolismo , 5'-Nucleotidasa/inmunología , 5'-Nucleotidasa/metabolismo , Adenosina/inmunología , Animales , Antígeno CD11b/inmunología , Antígeno CD11b/metabolismo , Carcinoma Pulmonar de Lewis , Diferenciación Celular/inmunología , Proliferación Celular , Separación Celular , Femenino , Citometría de Flujo , Granulocitos/citología , Granulocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Células Mieloides/citología , Células Mieloides/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Purinérgicos P1/inmunología , Receptores Purinérgicos P1/metabolismo
4.
J Pharmacol Exp Ther ; 341(3): 764-74, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22431204

RESUMEN

The existence of multipotent cardiac stromal cells expressing stem cell antigen (Sca)-1 has been reported, and their proangiogenic properties have been demonstrated in myocardial infarction models. In this study, we tested the hypothesis that stimulation of adenosine receptors on cardiac Sca-1(+) cells up-regulates their secretion of proangiogenic factors. We found that Sca-1 is expressed in subsets of mouse cardiac stromal CD31(-) and endothelial CD31(+) cells. The population of Sca-1(+)CD31(+) endothelial cells was significantly reduced, whereas the population of Sca-1(+)CD31(-) stromal cells was increased 1 week after myocardial infarction, indicating their relative functional importance in this pathophysiological process. An increase in adenosine levels in adenosine deaminase-deficient mice in vivo significantly augmented vascular endothelial growth factor (VEGF) production in cardiac Sca-1(+)CD31(-) stromal cells but not in Sca-1(+)CD31(+) endothelial cells. We found that mouse cardiac Sca-1(+)CD31(-) stromal cells predominantly express mRNA encoding A(2B) adenosine receptors. Stimulation of adenosine receptors significantly increased interleukin (IL)-6, CXCL1 (a mouse ortholog of human IL-8), and VEGF release from these cells. Using conditionally immortalized Sca-1(+)CD31(-) stromal cells obtained from wild-type and A(2B) receptor knockout mouse hearts, we demonstrated that A(2B) receptors are essential for adenosine-dependent up-regulation of their paracrine functions. We found that the human heart also harbors a population of stromal cells similar to the mouse cardiac Sca-1(+)CD31(-) stromal cells that increase release of IL-6, IL-8, and VEGF in response to A(2B) receptor stimulation. Thus, our study identified A(2B) adenosine receptors on cardiac stromal cells as potential targets for up-regulation of proangiogenic factors in the ischemic heart.


Asunto(s)
Antígenos Ly/metabolismo , Proteínas de la Membrana/metabolismo , Células Madre Mesenquimatosas/metabolismo , Comunicación Paracrina/fisiología , Receptor de Adenosina A2B/fisiología , Adenosina/metabolismo , Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/metabolismo , Agammaglobulinemia/metabolismo , Animales , Células Cultivadas , Quimiocina CXCL1/metabolismo , Femenino , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Infarto del Miocardio/metabolismo , Miocardio/citología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , ARN Mensajero/metabolismo , Inmunodeficiencia Combinada Grave/metabolismo , Células Madre/metabolismo , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
JCI Insight ; 6(16)2021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34197342

RESUMEN

Functional loss of myosin Vb (MYO5B) induces a variety of deficits in intestinal epithelial cell function and causes a congenital diarrheal disorder, microvillus inclusion disease (MVID). The impact of MYO5B loss on differentiated cell lineage choice has not been investigated. We quantified the populations of differentiated epithelial cells in tamoxifen-induced, epithelial cell-specific MYO5B-knockout (VilCreERT2 Myo5bfl/fl) mice utilizing digital image analysis. Consistent with our RNA-sequencing data, MYO5B loss induced a reduction in tuft cells in vivo and in organoid cultures. Paneth cells were significantly increased by MYO5B deficiency along with expansion of the progenitor cell zone. We further investigated the effect of lysophosphatidic acid (LPA) signaling on epithelial cell differentiation. Intraperitoneal LPA significantly increased tuft cell populations in both control and MYO5B-knockout mice. Transcripts for Wnt ligands were significantly downregulated by MYO5B loss in intestinal epithelial cells, whereas Notch signaling molecules were unchanged. Additionally, treatment with the Notch inhibitor dibenzazepine (DBZ) restored the populations of secretory cells, suggesting that the Notch pathway is maintained in MYO5B-deficient intestine. MYO5B loss likely impairs progenitor cell differentiation in the small intestine in vivo and in vitro, partially mediated by Wnt/Notch imbalance. Notch inhibition and/or LPA treatment may represent an effective therapeutic approach for treatment of MVID.


Asunto(s)
Síndromes de Malabsorción/genética , Microvellosidades/patología , Mucolipidosis/genética , Miosina Tipo V/deficiencia , Receptores Notch/metabolismo , Vía de Señalización Wnt/genética , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Dibenzazepinas/farmacología , Modelos Animales de Enfermedad , Enterocitos/efectos de los fármacos , Enterocitos/metabolismo , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Yeyuno/citología , Yeyuno/efectos de los fármacos , Yeyuno/patología , Lisofosfolípidos/farmacología , Lisofosfolípidos/uso terapéutico , Síndromes de Malabsorción/tratamiento farmacológico , Síndromes de Malabsorción/patología , Ratones , Ratones Noqueados , Microvellosidades/genética , Mucolipidosis/tratamiento farmacológico , Mucolipidosis/patología , Miosina Tipo V/genética , Organoides , Cultivo Primario de Células , Receptores Notch/antagonistas & inhibidores , Células Madre/fisiología , Vía de Señalización Wnt/efectos de los fármacos
6.
Am J Respir Cell Mol Biol ; 42(5): 564-71, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19556606

RESUMEN

Pharmacologic evidence suggests that activation of A(2B) adenosine receptors results in proinflammatory effects relevant to the progression of asthma, a chronic lung disease associated with elevated interstitial adenosine concentrations in the lung. This concept has been challenged by the finding that genetic removal of A(2B) receptors leads to exaggerated responses in models of acute inflammation. Therefore, the goal of our study was to determine the effects of A(2B) receptor gene ablation in the context of ovalbumin-induced chronic pulmonary inflammation. We found that repetitive airway allergen challenge induced a significant increase in adenosine levels in fluid recovered by bronchoalveolar lavage. Genetic ablation of A(2B) receptors significantly attenuated allergen-induced chronic pulmonary inflammation, as evidenced by a reduction in the number of bronchoalveolar lavage eosinophils and in peribronchial eosinophilic infiltration. The most striking difference in the pulmonary inflammation induced in A(2B) receptor knockout (A(2B)KO) and wild-type mice was the lack of allergen-induced IL-4 release in the airways of A(2B)KO animals, in line with a significant reduction in IL-4 protein and mRNA levels in lung tissue. In addition, attenuation of allergen-induced transforming growth factor-beta release in airways of A(2B)KO mice correlated with reduced airway smooth muscle and goblet cell hyperplasia/hypertrophy. In conclusion, genetic removal of A(2B) adenosine receptors in mice leads to inhibition of allergen-induced chronic pulmonary inflammation and airway remodeling. These findings are in agreement with previous pharmacologic studies suggesting a deleterious role for A(2B) receptor signaling in chronic lung inflammation.


Asunto(s)
Neumonía/metabolismo , Neumonía/prevención & control , Receptor de Adenosina A2B/deficiencia , Adenosina/metabolismo , Alérgenos/inmunología , Animales , Bronquios/inmunología , Bronquios/metabolismo , Bronquios/patología , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Enfermedad Crónica , Modelos Animales de Enfermedad , Eosinófilos/patología , Eliminación de Gen , Regulación de la Expresión Génica , Hipertrofia , Interleucina-4/biosíntesis , Metaplasia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Moco/metabolismo , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor de Adenosina A2B/genética , Receptor de Adenosina A2B/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
7.
Blood ; 112(5): 1822-31, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18559975

RESUMEN

Differentiation of functional dendritic cells (DCs) critically depends on the microenvironment. DCs differentiate in hypoxic tumor sites and inflamed or damaged tissue. Because local concentrations of adenosine reach high physiologically relevant levels in these conditions, we assessed the expression of adenosine receptors and the effect of their activation on differentiation of human monocytes and mouse peritoneal macrophages and hematopoietic progenitor cells (HPCs) into myeloid DCs. Stimulation of adenosine receptors skews DC differentiation toward a distinct cell population characterized by expression of both DC and monocyte/macrophage cell surface markers. Pharmacologic analysis and experiments with cells from A(2B) adenosine receptor knockout mice identified A(2B) receptor as the mediator of adenosine effects on DCs. Unlike normal myeloid DCs, adenosine-differentiated DCs have impaired allostimulatory activity and express high levels of angiogenic, pro-inflammatory, immune suppressor, and tolerogenic factors, including VEGF, IL-8, IL-6, IL-10, COX-2, TGF-beta, and IDO. They promoted tumor growth if injected into tumors implanted in mice. Using adenosine desaminase knockout animals, we showed that DCs with proangiogenic phenotype are highly abundant under conditions associated with elevated levels of extracellular adenosine in vivo. Adenosine signaling through A(2B) receptor is an important factor of aberrant DC differentiation and generation of tolerogenic, angiogenic, and proinflammatory cells.


Asunto(s)
Células Dendríticas/fisiología , Receptores Purinérgicos P1/fisiología , Adenosina/farmacología , Antagonistas del Receptor de Adenosina A2 , Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/genética , Animales , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Cartilla de ADN/genética , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Humanos , Tolerancia Inmunológica , Mediadores de Inflamación/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Experimentales/irrigación sanguínea , Neovascularización Patológica , Antagonistas de Receptores Purinérgicos P1 , Receptor de Adenosina A2B/deficiencia , Receptor de Adenosina A2B/genética , Receptor de Adenosina A2B/fisiología , Receptores Purinérgicos P1/deficiencia , Receptores Purinérgicos P1/genética , Transducción de Señal
8.
Circ Res ; 102(3): 356-63, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18032734

RESUMEN

Intracoronary delivery of endothelial progenitor cells (EPCs) is an emerging concept for the treatment of cardiovascular disease. Enhancement of EPC adhesion to vascular endothelium could improve cell retention within targeted organs. Because extracellular adenosine is elevated at sites of ischemia and stimulates neovascularization, we examined the potential role of adenosine in augmenting EPC retention to cardiac microvascular endothelium. Stimulation of adenosine receptors in murine embryonic EPCs (eEPCs) and cardiac endothelial cells (cECs) rapidly, within minutes, increased eEPC adhesion to cECs under static and flow conditions. Similarly, adhesion of human adult culture-expanded EPCs to human cECs was increased by stimulation of adenosine receptors. Furthermore, adenosine increased eEPC retention in isolated mouse hearts perfused with eEPCs. We determined that eEPCs and cECs preferentially express functional A1 and A2B adenosine receptor subtypes, respectively, and that both subtypes are involved in the regulation of eEPC adhesion to cECs. We documented that the interaction between P-selectin and its ligand (P-selectin glycoprotein ligand-1) plays a role in adenosine-dependent eEPC adhesion to cECs and that stimulation of adenosine receptors in cECs induces rapid cell surface expression of P-selectin. Our results suggest a role for adenosine in vasculogenesis and its potential use to stimulate engraftment in cell-based therapies.


Asunto(s)
Adenosina/farmacología , Células Endoteliales/metabolismo , Neovascularización Fisiológica/fisiología , Receptor de Adenosina A1/biosíntesis , Receptor de Adenosina A2B/biosíntesis , Células Madre/metabolismo , Vasodilatadores/farmacología , Adenosina/metabolismo , Agonistas del Receptor de Adenosina A1 , Agonistas del Receptor de Adenosina A2 , Animales , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/trasplante , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Glicoproteínas de Membrana/biosíntesis , Ratones , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/terapia , Miocardio/citología , Miocardio/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Selectina-P/biosíntesis , Trasplante de Células Madre , Células Madre/citología , Vasodilatadores/metabolismo
9.
J Pharmacol Exp Ther ; 324(2): 694-700, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17965229

RESUMEN

Pharmacological studies suggest that A(2B) adenosine receptors mediate proinflammatory effects of adenosine. This concept was recently challenged by the finding that A(2B) adenosine receptor knockout (A(2B)KO) mice had moderate inflammation due to elevated basal plasma tumor necrosis factor (TNF)-alpha and an exaggerated response to lipopolysaccharide (LPS) challenge. However, it is unclear whether this phenomenon actually reflects the loss of putative taming of proinflammatory cytokine production via activation of A(2B) receptors by endogenous adenosine. In this report, we examined adenosine receptor-dependent regulation of interleukin (IL)-6 and TNF-alpha blood plasma levels in A(2B)KO and wild-type mice in vivo and their release from peritoneal macrophages ex vivo. Stimulation of adenosine receptors with 5'-N-ethylcarboxamidoadenosine (NECA) up-regulated IL-6 and suppressed LPS-induced TNF-alpha in wild-type mice. The selective A(2B) antagonists 3-isobutyl-8-pyrrolidinoxanthine and 8-[4-[((4-cyanophenyl)carbamoylmethyl)oxy]phenyl]-1,3-di(n-propyl)xanthine (MRS 1754) inhibited NECA-induced IL-6 release but not the suppression of LPS-induced TNF-alpha secretion from macrophages. Genetic ablation of A(2B) receptors abrogated NECA-induced increases in IL-6 release from mouse peritoneal macrophages and dramatically reduced the ability of NECA to raise IL-6 plasma levels in vivo. In contrast, the absence of A(2B) adenosine receptors did not affect NECA-induced suppression of LPS-activated TNF-alpha release in macrophages, nor did it reduce the ability of NECA to suppress LPS-induced increase in TNF-alpha plasma levels in vivo. Thus, our results indicate that stimulation of A(2B) receptors up-regulates the proinflammatory cytokine IL-6 and argue against the recently suggested anti-inflammatory role of A(2B) receptors in suppression of LPS-stimulated TNF-alpha production by adenosine.


Asunto(s)
Adenosina/fisiología , Citocinas/metabolismo , Receptor de Adenosina A2B/biosíntesis , Receptor de Adenosina A2B/genética , Adenosina/farmacología , Antagonistas del Receptor de Adenosina A2 , Animales , Inflamación/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
10.
Circ Res ; 92(5): 485-92, 2003 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-12600879

RESUMEN

Adenosine is released during tissue injury, ischemia and tumor growth, and promotes angiogenesis. Because mast cells accumulate in the proximity of new blood vessel development, we examined if they may contribute to adenosine-induced angiogenesis. We found that HMC-1 human mast cells express A2A, A2B, and A3 adenosine receptors. The adenosine agonist NECA (100 micromol/L) increased interleukin-8 (IL-8), vascular endothelial growth factor (VEGF), and angiopoietin-2 mRNA expression. NECA-induced secretion of IL-8 and VEGF was verified by ELISA. A2B receptors mediate VEGF and IL-8 secretion because neither CGS21680 (selective A2A agonist) nor IB-MECA (selective A3 agonist) produced this effect, and it was inhibited by the selective A2B antagonist IPDX but not by the selective A2A antagonist SCH58261 or the selective A3 antagonist MRS1191. In contrast, the selective A3 agonist IB-MECA (EC50 1 nmol/L) stimulated angiopoietin-2 expression. Conditioned media from NECA-activated HMC-1 stimulated human umbilical vein endothelial cell proliferation and migration, and induced capillary tube formation. Capillary formation induced by mast cell-conditioned media was maximal if both HMC-1 A2B and A3 receptors were activated, whereas activation of A2B receptor alone was less effective. Thus, adenosine A2B and A3 receptors act in a functional cooperative fashion to promote angiogenesis by a paracrine mechanism involving the differential expression and secretion of angiogenic factors from human mast cells.


Asunto(s)
Mastocitos/fisiología , Neovascularización Fisiológica , Comunicación Paracrina , Receptores Purinérgicos P1/metabolismo , Adenosina/farmacología , Inductores de la Angiogénesis/biosíntesis , Inductores de la Angiogénesis/genética , Angiopoyetina 2 , Línea Celular , Medios de Cultivo Condicionados/farmacología , Factores de Crecimiento Endotelial/biosíntesis , Factores de Crecimiento Endotelial/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Interleucina-8/biosíntesis , Linfocinas/biosíntesis , Linfocinas/genética , Mastocitos/efectos de los fármacos , ARN Mensajero/biosíntesis , Receptor de Adenosina A2B , Receptor de Adenosina A3 , Receptores Purinérgicos P1/genética , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
11.
Circ Res ; 90(5): 531-8, 2002 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-11909816

RESUMEN

Adenosine has been reported to stimulate or inhibit the release of angiogenic factors depending on the cell type examined. To test the hypothesis that differential expression of adenosine receptor subtypes contributes to endothelial cell heterogeneity, we studied microvascular (HMEC-1) and umbilical vein (HUVEC) human endothelial cells. Based on mRNA level and stimulation of adenylate cyclase, we found that HUVECs preferentially express A2A adenosine receptors and HMEC-1 preferentially express A2B receptors. Neither cells expressed A1 or A3 receptors. The nonselective adenosine agonist 5'-N-ethylcarboxamidoadenosine (NECA) increased expression of interleukin-8 (IL-8), basic fibroblast growth factor (bFGF), and vascular endothelial growth factor (VEGF) in HMEC-1, but had no effect in HUVECs. In contrast, the selective A2A agonist 2-p-(2-carboxyethyl)phenylethylamino-NECA (CGS 21680) had no effect on expression of these angiogenic factors. Cotransfection of each type of adenosine receptors with a luciferase reporter in HMEC-1 showed that A2B receptors, but not A1, A2A, or A3, activated IL-8 and VEGF promoters. These effects were mimicked by constitutively active alphaG(q), alphaG12, and alphaG13, but not alphaG(s) or alphaG(i1-3). Furthermore, stimulation of phospholipase C indicated coupling of A2B receptors to G(q) proteins in HMEC-1. Thus, differential expression of adenosine receptor subtypes contributes to functional heterogeneity of human endothelial cells. A2B receptors, predominantly expressed in human microvascular cells, modulate expression of angiogenic factors via coupling to G(q), and possibly via G12/13.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Endotelio Vascular/metabolismo , Receptores Purinérgicos P1/biosíntesis , Adenilil Ciclasas/metabolismo , Animales , Células CHO , Cricetinae , Factores de Crecimiento Endotelial/genética , Endotelio Vascular/citología , Genes Reporteros , Proteínas de Unión al GTP Heterotriméricas/genética , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Fosfatos de Inositol/metabolismo , Interleucina-8/genética , Luciferasas/genética , Linfocinas/genética , Microcirculación/metabolismo , Neovascularización Fisiológica/fisiología , Regiones Promotoras Genéticas/fisiología , Agonistas del Receptor Purinérgico P1 , ARN Mensajero/metabolismo , Receptor de Adenosina A2A , Receptor de Adenosina A2B , Receptores Purinérgicos P1/genética , Sistemas de Mensajero Secundario/fisiología , Transfección , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Venas/metabolismo
12.
Cell Mol Gastroenterol Hepatol ; 2(2): 131-157, 2016 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27019864

RESUMEN

BACKGROUND AND AIMS: Inactivating mutations in MYO5B cause severe neonatal diarrhea in Microvillus Inclusion Disease. Loss of active MYO5B causes the formation of pathognomonic inclusions and aberrations in brush border enzymes. METHODS: We developed three mouse models of germline, constitutively intestinal targeted and inducible intestinal targeted deletion of MYO5B. The mice were evaluated for enterocyte cellular morphology. RESULTS: Germline MYO5B KO mice showed early diarrhea and failure to thrive with evident microvillus inclusions and loss of apical transporters in the duodenum. IgG was present within inclusions. Apical transporters were lost and inclusions were present in the duodenum, but were nearly absent in the ileum. VillinCre;MYO5BF/F mice showed similar pathology and morphological changes in duodenal enterocytes. In contrast, when MYO5B KO was induced with tamoxifen treatment at 8 weeks of age, VillinCreERT2;MYO5BF/F mice developed severe diarrhea with loss of duodenal brush border enzymes, but few inclusions were observed in enterocytes. However, if tamoxifen is administered to 2-day-old VillinCreERT2;MYO5BF/F mice, prominent microvillus inclusions were observed. CONCLUSIONS: The microvillus inclusions that develop after MYO5B loss reveal the presence of an unrecognized apical membrane trafficking pathway in neonatal duodenal enterocytes. However, the diarrheal pathology after MYO5B loss is due to deficits in transporter presentation at the apical membrane in duodenal enterocytes.

13.
Purinergic Signal ; 5(3): 289-98, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19125355

RESUMEN

In human mast cells and microvascular endothelial cells, the A(2B) adenosine receptor controls at least three independent signaling pathways, i.e., Gs-mediated stimulation of adenylate cyclase, Gq-mediated stimulation of phospholipase Cbeta, and Gs/Gq-independent upregulation of IL-8. Functional analysis of cells transfected with full-length and truncated receptor constructs revealed that the A(2B) receptor C-terminus is important for coupling to Gs and Gq proteins. Removal of the entire cytoplasmic portion in the A(2B) receptor C-terminus rendered it incapable of stimulating adenylate cyclase and phospholipase Cbeta. Conversely, removal of the distal 16 amino acids facilitated signal transduction from the receptor to the downstream Gs but not Gq proteins. However, the A(2B) receptor C-terminus is not essential for upregulation of IL-8. Analysis of chimeric A(2A)/A(2B) receptors demonstrated that only chimeras containing the third intracellular loop of the A(2B) receptor mediated agonist-dependent IL-8 reporter stimulation, suggesting that this domain is important for upregulation of IL-8.

14.
Neoplasia ; 10(9): 987-95, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18714400

RESUMEN

Recent studies suggest that tumor-infiltrating immune cells can benefit the tumor by producing factors that promote angiogenesis and suppress immunity. Because the tumor microenvironment is characterized by high adenosine levels, we hypothesized that the low-affinity A(2B) adenosine receptor located on host immune cells may participate in these effects. In the current study, we tested this hypothesis in a Lewis lung carcinoma isograft model using A(2B) receptor knockout (A(2B)KO) mice. These mice exhibited significantly attenuated tumor growth and longer survival times after inoculation with Lewis lung carcinoma compared to wild type (WT) controls. Lewis lung carcinoma tumors in A(2B)KO mice contained significantly lower levels of vascular endothelial growth factor (VEGF) compared to tumors growing in WT animals. This difference was due to VEGF production by host cells, which comprised 30 +/- 2% of total tumor cell population. Stimulation of adenosine receptors on WT tumor-infiltrating CD45+ immune cells increased VEGF production fivefold, an effect not seen in tumor-associated CD45+ immune cells lacking A(2B) receptors. In contrast, we found no significant difference in VEGF production between CD45- tumor cells isolated from WT and A(2B)KO mice. Thus, our data suggest that tumor cells promote their growth by exploiting A(2B) adenosine receptor-dependent regulation of VEGF in host immune cells.


Asunto(s)
Neoplasias/metabolismo , Receptor de Adenosina A2B/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Carcinoma Pulmonar de Lewis , Antígenos Comunes de Leucocito/metabolismo , Ratones , Neoplasias/irrigación sanguínea , Neovascularización Patológica
15.
J Immunol ; 180(11): 7212-20, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18490720

RESUMEN

Pharmacological studies suggest that A(2B) adenosine receptors mediate proinflammatory effects of adenosine in human mast cells in part by up-regulating production of Th2 cytokines and angiogenic factors. This concept has been recently challenged by the finding that mast cells cultured from bone marrow-derived mast cells (BMMCs) of A(2B) knockout mice display an enhanced degranulation in response to FcepsilonRI stimulation. This finding was interpreted as evidence of anti-inflammatory functions of A(2B) receptors and it was suggested that antagonists with inverse agonist activity could promote activation of mast cells. In this report, we demonstrate that genetic ablation of the A(2B) receptor protein has two distinct effects on BMMCs, one is the previously reported enhancement of Ag-induced degranulation, which is unrelated to adenosine signaling; the other is the loss of adenosine signaling via this receptor subtype that up-regulates IL-13 and vascular endothelial growth factor secretion. Genetic ablation of A(2B) receptors had no effect on A(3) adenosine receptor-dependent potentiation of Ag-induced degranulation in mouse BMMCs, but abrogated A(2B) adenosine receptor-dependent stimulation of IL-13 and vascular endothelial growth factor secretion. Adenosine receptor antagonists MRS1706 and DPCPX with known inverse agonist activity at the A(2B) subtype inhibited IL-13 secretion induced by the adenosine analog NECA, but did not mimic the enhanced Ag-induced degranulation observed in A(2B) knockout BMMCs. Thus, our study confirmed the proinflammatory role of adenosine signaling via A(2B) receptors and the anti-inflammatory actions of A(2B) antagonists in mouse BMMCs.


Asunto(s)
Adenosina Monofosfato/metabolismo , Adenosina/metabolismo , Interleucina-13/biosíntesis , Mastocitos/metabolismo , Receptor de Adenosina A2B/metabolismo , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Antagonistas del Receptor de Adenosina A2 , Adenosina Monofosfato/inmunología , Animales , Células de la Médula Ósea , Degranulación de la Célula , Línea Celular , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Interleucina-13/inmunología , Mastocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Purinas/farmacología , Receptor de Adenosina A2B/genética , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/inmunología , Xantinas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
J Pharmacol Exp Ther ; 320(2): 565-72, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17132813

RESUMEN

Because hypoxia increases extracellular adenosine levels and stimulates angiogenesis, we evaluated the relative roles of reduced oxygen concentrations and adenosine receptor activation in the production of angiogenic factors. In vitro, we analyzed the effects of hypoxia and adenosine on the secretion of angiogenic factors from human microvascular endothelial cells (HMEC-1). To study the effects of hypoxia alone, we scavenged adenosine from the hypoxic medium with adenosine deaminase, and we used the stable adenosine analog 5'-N-ethylcarboxamidoadenosine (NECA) to study the effects of stimulation of adenosine receptors. In the absence of adenosine, hypoxia stimulated vascular endothelial growth factor (VEGF) but not interleukin-8 (IL-8) secretion from HMEC-1. In contrast, NECA stimulated both VEGF and IL-8 secretion. VEGF secretion was increased 1.9 +/- 0.04-fold with NECA (10 microM) and 1.7 +/- 0.1-fold with hypoxia (5% O(2)) but 3.8 +/- 0.1-fold when these two stimuli were combined. Thus, adenosine receptors act in a cooperative fashion with hypoxia to stimulate VEGF and induce IL-8 secretion not stimulated by hypoxia alone. In vivo, antagonism of adenosine receptors with caffeine abrogated VEGF up-regulation induced by local injection of NECA into the mouse hind limb and produced a 46% reduction of neovascularization in a mouse ischemic hind limb model. Our study suggests that adenosine actions are not redundant but rather are complementary to the direct effects of hypoxia. Stimulation of adenosine receptors not only contributes to the overall effect of hypoxia but also has additional actions in the regulation of angiogenic factors. Thus, adenosine receptors represent a potential therapeutic target for regulation of neovascularization.


Asunto(s)
Hipoxia/fisiopatología , Interleucina-8/genética , Neovascularización Fisiológica , Receptores Purinérgicos P1/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Adenosina/metabolismo , Adenosina-5'-(N-etilcarboxamida)/farmacología , Animales , Células Cultivadas , Regulación de la Expresión Génica , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/análisis
17.
Mol Pharmacol ; 70(2): 727-35, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16707627

RESUMEN

Human mast cells express functional A(2A) and A(2B) adenosine receptors. However, only stimulation of A(2B), not A(2A), leads to secretion of interleukin (IL)-4, an important step in adenosine receptor-mediated induction of IgE synthesis by B-cells. In this study, we investigate intracellular pathways that link stimulation of A(2B) receptors to IL-4 up-regulation in HMC-1 mast cells. Both A(2A) and A(2B) receptors couple to G(s) proteins and stimulate adenylate cyclase, but only A(2B) stimulates phospholipase Cbeta through coupling to G(q) proteins leading to activation of protein kinase C and calcium mobilization. Inhibition of phospholipase Cbeta completely blocked A(2B) receptor-dependent IL-4 secretion. The protein kinase C inhibitor 2-{8-[(dimethylamino)-methyl]-6,7,8,9-tetrahydropyrido[1,2-a]indol-3-yl}-3-(1-methyl-1H-indol-3-yl)maleimide (Ro-32-0432) had no effect on A(2B) receptor-mediated IL-4 secretion but inhibited phorbol 12-myristate 13-acetate-stimulated IL-4 secretion. In contrast, chelation of intracellular Ca(2+) inhibited both A(2B) receptor- and ionomycin-dependent IL-4 secretion. This Ca(2+)-sensitive pathway probably includes calcineurin and nuclear factor of activated T cells, because A(2B) receptor-dependent IL-4 secretion was blocked with cyclosporin A or 11R-VIVIT peptide. G(s)-linked pathways also play a role in the A(2B) receptor-dependent stimulation of IL-4 secretion; inhibition of adenylate cyclase or protein kinase A attenuated A(2B) receptor-dependent IL-4 secretion. Although stimulation of adenylate cyclase with forskolin did not increase IL-4 secretion on its own, it potentiated the effect of Pasteurella multocida toxin by 2-fold and ionomycin by 3-fold. Both forskolin and stimulation of A(2B) receptors up-regulated NFATc1 protein levels. We conclude that A(2B) receptors up-regulate IL-4 through G(q) signaling that is potentiated via cross-talk with G(s)-coupled pathways.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gs/fisiología , Interleucina-4/biosíntesis , Mastocitos/metabolismo , Receptor de Adenosina A2B/fisiología , Transducción de Señal/fisiología , Adenosina-5'-(N-etilcarboxamida)/farmacología , Células Cultivadas , Colforsina/farmacología , Humanos , Factores de Transcripción NFATC/análisis , Regulación hacia Arriba
18.
J Immunol ; 172(12): 7726-33, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15187156

RESUMEN

Adenosine provokes bronchoconstriction in asthmatics through acute activation of mast cells, but its potential role in chronic inflammation has not been adequately characterized. We hypothesized that adenosine up-regulates Th2 cytokines in mast cells, thus promoting IgE synthesis by B lymphocytes. We tested this hypothesis in human mast cells (HMC-1) expressing A(2A), A(2B), and A(3) adenosine receptors. The adenosine analog 5'-N-ethylcarboxamidoadenosine (NECA) (10 microM) increased mRNA expression of IL-1beta, IL-3, IL-4, IL-8, and IL-13, but not IL-2 and IFN-gamma. Up-regulation of IL-4 and IL-13 was verified using RT-PCR and ELISA; 10 microM NECA increased IL-13 concentrations in HMC-1 conditioned medium 28-fold, from 7.6 +/- 0.3 to 215 +/- 4 pg/ml, and increased IL-4 concentrations 6-fold, from 19.2 +/- 0.1 to 117 +/- 2 pg/ml. This effect was mediated by A(2B) receptors because neither the selective A(2A) agonist 2-p-(2-carboxyethyl)phenethylamino-NECA nor the selective A(3) agonist N(6)-(3-iodobenzyl)-N-methyl-5'-carbamoyladenosine reproduced it, and the selective A(2B) antagonist 3-isobutyl-8-pyrrolidinoxanthine prevented it. Constitutive expression of CD40 ligand on HMC-1 surface was not altered by NECA. Human B lymphocytes cocultured for 12 days with NECA-stimulated HMC-1 produced 870 +/- 33 pg IgE per 10(6) B cells, whereas lymphocytes cocultured with nonstimulated HMC-1, or cultured alone in the absence or in the presence of NECA, produced no IgE. Thus, we demonstrated induction of IgE synthesis by the interaction between adenosine-stimulated mast cells and B lymphocytes, and suggest that this mechanism is involved in the amplification of the allergic inflammatory responses associated with asthma.


Asunto(s)
Adenosina/farmacología , Linfocitos B/inmunología , Inmunoglobulina E/biosíntesis , Mastocitos/inmunología , Receptor de Adenosina A2B/fisiología , Adenosina-5'-(N-etilcarboxamida)/farmacología , Asma/etiología , Linfocitos B/fisiología , Comunicación Celular/inmunología , Células Cultivadas , Técnicas de Cocultivo , Citocinas/genética , Humanos , Interleucina-13/inmunología , Interleucina-4/inmunología , Mastocitos/fisiología , ARN Mensajero/biosíntesis , Células Th2/inmunología , Regulación hacia Arriba/inmunología
19.
Hypertension ; 44(5): 649-54, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15452028

RESUMEN

We previously reported that adenosine A2B receptor activation stimulates angiogenesis. Because hypoxia is a potent stimulus for the release of both adenosine and angiogenic factors, we tested the hypothesis that hypoxia alters the expression of adenosine receptors toward an "angiogenic" phenotype. We used human umbilical vein endothelial cells (HUVECs) and bronchial smooth muscle cells (BSMCs) because, under normoxic conditions, adenosine does not release vascular endothelial growth factor (VEGF). HUVECs expressed a characteristic A2A phenotype (the selective A2A agonist CGS21680 was as potent as the nonselective agonist 5'-N-ethylcarboxamidoadenosine [NECA] in generating cAMP). Hypoxia (4.6% O2, 3 hours) decreased A2A mRNA from 1.56+/-0.3% to 0.16+/-0.01% of beta-actin expression but increased A2B mRNA from 0.08+/-0.01% to 0.27+/-0.05%. Consistent with changes in receptor expression, CGS21680 failed to increase cAMP in hypoxic HUVECs, whereas NECA remained active (A2B phenotype), and NECA increased VEGF release from 9.5+/-1.0 to 14.2+/-1.2 pg/mL (P<0.05), indicating that increased A2B receptors were functionally coupled to upregulation of VEGF. Hypoxia had similar effects on BSMCs, increasing A2B mRNA by 2.4+/-0.3-fold, from 0.42+/-0.04% to 1.00+/-0.13% of beta-actin. Whereas NECA had no effect on VEGF release in normoxic BSMCs, it increased VEGF release in hypoxic BSMCs, from 74.6+/-9.6 to 188.3+/-16.7 pg/mL (P<0.01), and a selective A2B antagonist, CVT-6694, inhibited this increase. A2B receptors activated a VEGF reporter made unresponsive to hypoxia by mutating its hypoxia-inducible factor-1 (HIF-1) binding element, indicating a mechanism independent of HIF-1. In conclusion, hypoxia modulates the expression of adenosine receptors in human endothelial and smooth muscle cells toward an A2B"angiogenic" phenotype.


Asunto(s)
Hipoxia de la Célula/fisiología , Células Endoteliales/metabolismo , Miocitos del Músculo Liso/metabolismo , Neovascularización Fisiológica , Receptores Purinérgicos P1/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Células Cultivadas , Proteínas de Unión al ADN/fisiología , Regulación hacia Abajo , Expresión Génica , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Proteínas Nucleares/fisiología , Fenotipo , ARN Mensajero , Receptor de Adenosina A2B/fisiología , Factores de Transcripción/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA