Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Pharmacol Res ; 63(1): 13-22, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20971192

RESUMEN

Nitric oxide (NO) drives pro-survival responses in vascular cells and limits platelet adhesion, enhancing blood flow and minimizing thrombosis. The matricellular protein thrombospondin-1 (TSP1), through interaction with its receptor CD47, inhibits soluble guanylyl cyclase (sGC) activation by NO in vascular cells. In vascular smooth muscle cells (VSMCs) both intracellular cGMP and cAMP regulate adhesion, contractility, proliferation, and migration. cGMP can regulate cAMP through feedback control of hydrolysis. Inhibition of the cAMP phosphodiesterase-4 selectively interfered with the ability of exogenous TSP1 to block NO-driven VSMC adhesion but not cGMP accumulation, suggesting that cAMP also contributes to VSMC regulation by TSP1. Inhibition of phosphodiesterase-4 was sufficient to elevate cAMP levels, and inhibiting guanylyl cyclase or phosphodiesterase-3, or adding exogenous TSP1 reversed this increase in cAMP. Thus, TSP1 regulates VSMC cAMP levels in part via cGMP-dependent inhibition of phosphodiesterase-3. Additionally basal cAMP levels were consistently elevated in both VSMCs and skeletal muscle from TSP1 null mice, and treating null cells with exogenous TSP1 suppressed cAMP levels to those of wild type cells. TSP1 inhibited both forskolin and isoproterenol stimulated increases in cAMP in VSMCs. TSP1 also abrogated forskolin and isoproterenol stimulated vasodilation. Consistent with its ability to directly limit adenylyl cyclase-activated vasodilation, TSP1 also limited cAMP-induced dephosphorylation of myosin light chain-2. These findings demonstrate that TSP1 limits both cGMP and cAMP signaling pathways and functional responses in VSMCs and arteries, by both phosphodiesterase-dependent cross talk between these second messengers and by inhibition of adenylyl cyclase activation.


Asunto(s)
AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Transducción de Señal , Trombospondina 1/metabolismo , Vasodilatación , Actinas/metabolismo , Animales , Antígeno CD47/metabolismo , Células Cultivadas , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Células Endoteliales/metabolismo , Guanilato Ciclasa , Humanos , Hidrólisis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Óxido Nítrico/metabolismo , Inhibidores de Fosfodiesterasa/farmacología , Ratas , Receptores Citoplasmáticos y Nucleares , Transducción de Señal/efectos de los fármacos , Guanilil Ciclasa Soluble , Trombospondina 1/deficiencia , Trombospondina 1/genética , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
2.
Artículo en Inglés | MEDLINE | ID: mdl-22685691

RESUMEN

CD47 is a widely expressed cell surface receptor that serves as a counter-receptor for signal regulatory protein-α and as a receptor for the secreted matricellular protein thrombospondin-1. Thrombospondin-1 signaling through CD47 regulates cellular signaling pathways that control cell survival, growth, motility, mitochondrial biogenesis, arterial vasoactive responses to physiologic vasodilators and blood flow, and responsiveness to growth factors. Studies employing mice lacking either thrombospondin-1 or CD47 have revealed an important role for this receptor-ligand interaction in tissue responses to injury and stress. These null mice show enhanced recovery from soft tissue fixed ischemic injuries, ischemia reperfusion injuries, and radiation injuries. These studies have led to development of antisense strategies to locally or globally suppress CD47 gene expression. A translation-blocking CD47 morpholino improves tissue survival in skin flap and hindlimb fixed ischemia models, full thickness skin grafts, and a liver ischemia/reperfusion model of organ transplantation in mice. Furthermore, the benefits of morpholino treatment extend to aged mice and mice with dysregulated fat metabolism that characteristically exhibit impaired recovery from ischemic injuries. Activity of the morpholino was also demonstrated for treatment of ischemic injury in miniature pigs. Treatment with the CD47 morpholino protects mice from major effects of ionizing radiation including alopecia, deterioration of muscle function, soft tissue and cutaneous fibrosis, and loss of hematopoietic stem cells in bone marrow. Remarkably, the same treatment does not protect tumors but instead enhances their ablation by irradiation. We discuss prospects for further development of CD47 antisense therapeutics for clinical applications including reconstructive surgery, organ transplantation, angioplasty, and cancer.

3.
Cell Stress Chaperones ; 15(2): 165-81, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19579057

RESUMEN

Thiolutin is a dithiole synthesized by Streptomyces sp. that inhibits endothelial cell adhesion and tumor growth. We show here that thiolutin potently inhibits developmental angiogenesis in zebrafish and vascular outgrowth from tissue explants in 3D cultures. Thiolutin is a potent and selective inhibitor of endothelial cell adhesion accompanied by rapid induction of HSPB1 (Hsp27) phosphorylation. The inhibitory effects of thiolutin on endothelial cell adhesion are transient, potentially due to a compensatory increase in Hsp27 protein levels. Accordingly, heat shock induction of Hsp27 limits the anti-adhesive activity of thiolutin. Thiolutin treatment results in loss of actin stress fibers, increased cortical actin as cells retract, and decreased cellular F-actin. Mass spectrometric analysis of Hsp27 binding partners following immunoaffinity purification identified several regulatory components of the actin cytoskeleton that associate with Hsp27 in a thiolutin-sensitive manner including several components of the Arp2/3 complex. Among these, ArpC1a is a direct binding partner of Hsp27. Thiolutin treatment induces peripheral localization of phosphorylated Hsp27 and Arp2/3. Hsp27 also associates with the intermediate filament components vimentin and nestin. Thiolutin treatment specifically ablates Hsp27 interaction with nestin and collapses nestin filaments. These results provide new mechanistic insights into regulation of cell adhesion and cytoskeletal dynamics by Hsp27.


Asunto(s)
Actinas/metabolismo , Citoesqueleto/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Proteínas de Choque Térmico HSP27/metabolismo , Pez Cebra/metabolismo , Animales , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Colágeno Tipo I/metabolismo , Citoesqueleto/efectos de los fármacos , Embrión no Mamífero/irrigación sanguínea , Embrión no Mamífero/citología , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Células Endoteliales/citología , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Choque Térmico HSP27/genética , Humanos , Ratones , Unión Proteica , Pirrolidinonas/farmacología , Tubulina (Proteína)/metabolismo , Pez Cebra/embriología
5.
Matrix Biol ; 28(2): 110-9, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19284971

RESUMEN

Nitric oxide (NO) locally regulates vascular resistance and blood pressure by modulating blood vessel tone. Thrombospondin-1 signaling via its receptor CD47 locally limits the ability of NO to relax vascular smooth muscle cells and increase regional blood flow in ischemic tissues. To determine whether thrombospondin-1 plays a broader role in central cardiovascular physiology, we examined vasoactive stress responses in mice lacking thrombospondin-1 or CD47. Mice lacking thrombospondin-1 exhibit activity-associated increases in heart rate, central diastolic and mean arterial blood pressure and a constant decrease in pulse pressure. CD47-deficient mice have normal central pulse pressure but elevated resting peripheral blood pressure. Both null mice show exaggerated decreases in peripheral blood pressure and increased cardiac output and ejection fraction in response to NO. Autonomic blockade also induces exaggerated hypotensive responses in awake thrombospondin-1 null and CD47 null mice. Both null mice exhibit a greater hypotensive response to isoflurane, and autonomic blockage under isoflurane anesthesia leads to premature death of thrombospondin-1 null mice. Conversely, the hypertensive response to epinephrine is attenuated in thrombospondin-1 null mice. Thus, the matricellular protein thrombospondin-1 and its receptor CD47 serve as acute physiological regulators of blood pressure and exert a vasopressor activity to maintain global hemodynamics under stress.


Asunto(s)
Presión Sanguínea/fisiología , Antígeno CD47/metabolismo , Frecuencia Cardíaca/fisiología , Corazón/fisiología , Transducción de Señal/fisiología , Estrés Fisiológico/fisiología , Trombospondina 1/metabolismo , Animales , Presión Sanguínea/genética , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Ecocardiografía , Frecuencia Cardíaca/genética , Inmunoensayo , Ratones , Ratones Noqueados , Flujo Sanguíneo Regional , Piel/irrigación sanguínea , Trombospondina 1/deficiencia
6.
Nat Rev Cancer ; 9(3): 182-94, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19194382

RESUMEN

In addition to long-term regulation of angiogenesis, angiogenic growth factor signalling through nitric oxide (NO) acutely controls blood flow and haemostasis. Inhibition of this pathway may account for the hypertensive and pro-thrombotic side effects of the vascular endothelial growth factor antagonists that are currently used for cancer treatment. The first identified endogenous angiogenesis inhibitor, thrombospondin 1, also controls tissue perfusion, haemostasis and radiosensitivity by antagonizing NO signalling. We examine the role of these and other emerging activities of thrombospondin 1 in cancer. Clarifying how endogenous and therapeutic angiogenesis inhibitors regulate vascular NO signalling could facilitate development of more selective inhibitors.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Óxido Nítrico/fisiología , Transducción de Señal , Trombospondina 1/fisiología , Animales , Antígenos CD36/fisiología , GMP Cíclico/fisiología , Hemostasis , Humanos , Neoplasias/etiología , Tolerancia a Radiación , Flujo Sanguíneo Regional
7.
Sci Transl Med ; 1(3): 3ra7, 2009 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-20161613

RESUMEN

Radiation-induced damage of normal tissues restricts the therapeutic doses of ionizing radiation that can be delivered to tumors and thereby limits the effectiveness of radiotherapy. Thrombospondin-1 signaling through its cell surface receptor CD47 limits recovery from several types of stress, and mice lacking either gene are profoundly resistant to radiation injury. We describe strategies to protect normal tissues from radiation damage using CD47 or thrombospondin-1 antibodies, a CD47-binding peptide, or antisense suppression of CD47. A morpholino oligonucleotide targeting CD47 confers radioresistance to human endothelial cells in vitro and protects soft tissue, bone marrow, and tumor-associated leukocytes in irradiated mice. In contrast, CD47 suppression in mice bearing melanoma or squamous lung tumors prior to irradiation result in 89% and 71% smaller tumors, respectively. Thus, inhibiting CD47 signaling maintains the viability of normal tissues following irradiation while increasing the radiosensitivity of tumors.


Asunto(s)
Anticuerpos/inmunología , Antígeno CD47/inmunología , Tolerancia a Radiación , Transducción de Señal , Animales , Apoptosis , Médula Ósea/efectos de la radiación , Antígeno CD47/metabolismo , GMP Cíclico/metabolismo , Humanos , Ratones , Músculo Esquelético/efectos de la radiación , Óxido Nítrico/metabolismo , Trombospondina 1/inmunología
8.
Surgery ; 144(5): 752-61, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19081017

RESUMEN

BACKGROUND: Ischemia-reperfusion (I/R) injury remains a primary complication of transplant surgery, accounting for about 80% of liver transplant failures, and is a major source of morbidity in other pathologic conditions. Activation of endothelium and inflammatory cell recruitment are central to the initiation and promulgation of I/R injury, which can be limited by the bioactive gas nitric oxide (NO). The discovery that thrombsospondin-1 (TSP1), via CD47, limits NO signaling in vascular cells and ischemic injuries in vivo suggested that I/R injury could be another important target of this signaling pathway. METHODS: Wild-type, TSP1-null, and CD47-null mice underwent liver I/R injury. Wild-type animals were pretreated with CD47 or control antibodies before liver I/R injury. Tissue perfusion via laser Doppler imaging, serum enzymes, histology, and immunohistology were assessed. RESULTS: TSP1-null and CD47-null mice subjected to subtotal liver I/R injury showed improved perfusion relative to wild-type mice. Null mice subjected to liver I/R had decreased liver enzyme release and less histologic evidence of injury. Elevated TSP1 expression in liver tissue after I/R injury suggested that preventing its interaction with CD47 could be protective. Thus, pretreatment of wild-type mice using a blocking CD47 antibody improved recovery of tissue perfusion and preserved liver integrity after I/R injury. CONCLUSIONS: Tissue survival and perfusion after liver I/R injury are limited by TSP1 and CD47. Targeting CD47 before I/R injury enhances tissue survival and perfusion in a model of liver I/R injury and suggests therapeutics for enhancing organ survival in transplantation surgery.


Asunto(s)
Antígeno CD47/fisiología , Hígado/cirugía , Daño por Reperfusión/etiología , Daño por Reperfusión/terapia , Transducción de Señal/fisiología , Trombospondina 1/fisiología , Animales , Anticuerpos Monoclonales/uso terapéutico , Femenino , Factores Inmunológicos/uso terapéutico , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Recuperación de la Función , Daño por Reperfusión/metabolismo
9.
Am J Pathol ; 173(4): 1100-12, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18787106

RESUMEN

Radiation, a primary mode of cancer therapy, acutely damages cellular macromolecules and DNA and elicits stress responses that lead to cell death. The known cytoprotective activity of nitric oxide (NO) is blocked by thrombospondin-1, a potent antagonist of NO/cGMP signaling in ischemic soft tissues, suggesting that thrombospondin-1 signaling via its receptor CD47 could correspondingly increase radiosensitivity. We show here that soft tissues in thrombospondin-1-null mice are remarkably resistant to radiation injury. Twelve hours after 25-Gy hindlimb irradiation, thrombospondin-1-null mice showed significantly less cell death in both muscle and bone marrow. Two months after irradiation, skin and muscle units in null mice showed minimal histological evidence of radiation injury and near full retention of mitochondrial function. Additionally, both tissue perfusion and acute vascular responses to NO were preserved in irradiated thrombospondin-1-null hindlimbs. The role of thrombospondin-1 in radiosensitization is specific because thrombospondin-2-null mice were not protected. However, mice lacking CD47 showed radioresistance similar to thrombospondin-1-null mice. Both thrombospondin-1- and CD47-dependent radiosensitization is cell autonomous because vascular cells isolated from the respective null mice showed dramatically increased survival and improved proliferative capacity after irradiation in vitro. Therefore, thrombospondin-1/CD47 antagonists may have selective radioprotective activity for normal tissues.


Asunto(s)
Antígeno CD47/metabolismo , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Trombospondina 1/metabolismo , Supervivencia Tisular/efectos de la radiación , Animales , Apoptosis/efectos de la radiación , Vasos Sanguíneos/patología , Vasos Sanguíneos/efectos de la radiación , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Células Endoteliales/patología , Células Endoteliales/efectos de la radiación , Miembro Posterior/patología , Miembro Posterior/efectos de la radiación , Hipertrofia , Masculino , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Neoplasias/patología , Tolerancia a Radiación/efectos de la radiación , Trombospondinas/metabolismo , Rayos X
10.
Neoplasia ; 10(8): 886-96, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18670646

RESUMEN

Nitric oxide (NO) plays important physiological roles in the vasculature to regulate angiogenesis, blood flow, and hemostasis. In solid tumors, NO is generally acknowledged to mediate angiogenic responses to several growth factors. This contrasts with conflicting evidence that NO can acutely increase tumor perfusion through local vasodilation or diminish perfusion by preferential relaxation of peripheral vascular beds outside the tumor. Because thrombospondin 1 (TSP1) is an important physiological antagonist of NO in vascular cells, we examined whether, in addition to inhibiting tumor angiogenesis, TSP1 can acutely regulate tumor blood flow. We assessed this activity of TSP1 in the context of perfusion responses to NO as a vasodilator and epinephrine as a vasoconstrictor. Nitric oxide treatment of wild type and TSP1 null mice decreased perfusion of a syngeneic melanoma, whereas epinephrine transiently increased tumor perfusion. Acute vasoactive responses were also independent of the level of tumor-expressed TSP1 in a melanoma xenograft, but recovery of basal perfusion was modulated by TSP1 expression. In contrast, overexpression of truncated TSP1 lacking part of its CD47 binding domain lacked this modulating activity. These data indicate that TSP1 primarily regulates long-term vascular responses in tumors, in part, because the tumor vasculature has a limited capacity to acutely respond to vasoactive agents.


Asunto(s)
Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/tratamiento farmacológico , Trombospondina 1/farmacología , Vasoconstrictores/farmacología , Vasodilatadores/farmacología , Animales , Línea Celular Tumoral , Epinefrina/farmacología , Femenino , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/farmacología , Flujo Sanguíneo Regional/efectos de los fármacos , Trombospondina 1/deficiencia , Trombospondina 1/genética , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Proteome Res ; 7(10): 4384-95, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18720982

RESUMEN

Thiolutin is a sulfur-based microbial compound with known activity as an angiogenesis inhibitor. Relative to previously studied angiogenesis inhibitors, thiolutin is a remarkably potent inducer of heat shock protein 27 (Hsp27) phosphorylation. This phosphorylation requires p38 kinase but is independent of increased p38 phosphorylation. To elucidate how thiolutin regulates Hsp27 phosphorylation and ultimately angiogenesis, Hsp27 was immunoprecipitated using nonphosphorylated and phospho-Ser78 specific antibodies from lysates of thiolutin treated and untreated human umbilical vein endothelial cells and analyzed by LC-MS. Separate LC-MS analyses of Lys-C, Lys-C plus trypsin, and Lys-C plus Glu-C digests provided 100% sequence coverage, including the identification of a very large 13 kDa Lys-C fragment using a special sample handling procedure (4 M guanidine HCl) prior to the LC-MS analysis to improve the large peptide recovery. The analysis revealed a novel post-translational modification of Hsp27 involving truncation of the N-terminal Met and acetylation of the penultimate Thr. Analysis of a Glu-C fragment containing two phosphorylation sites, Ser78 and Ser82, and a tryptic fragment containing the other phosphorylation site, Ser15, enabled quantitative stoichiometry of Hsp27 phosphorylation by LC-MS. The strategy revealed details of Hsp27 phosphorylation, including significant di-phosphorylation at both Ser78 and Ser82, that would be difficult to obtain by traditional approaches because oligomerization of the hydrophobic N-terminal region of the molecule prevents efficient enzymatic cleavage. The combination of Western blotting, immunoprecipation, and LC-MS provides a quantitative analysis of thiolutin-stimulated Hsp27 phosphorylation and further defines the role of Hsp27 in the antiangiogenic activities of thiolutin and related dithiolethiones.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Proteínas de Choque Térmico/química , Secuencia de Aminoácidos , Inhibidores de la Angiogénesis/farmacología , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Activación Enzimática , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Inmunoprecipitación , Espectrometría de Masas , Datos de Secuencia Molecular , Fosforilación , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Pirrolidinonas/farmacología , Espectroscopía Infrarroja por Transformada de Fourier , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Ann Surg ; 247(5): 860-8, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18438125

RESUMEN

BACKGROUND: Insufficient tissue perfusion underlies many acute and chronic diseases. Tissue perfusion in turn requires adequate blood flow, determined in large part by the relative state of relaxation or constriction of arterial vessels. Nitric oxide (NO) produced by vascular cells modulates blood flow and tissue perfusion by relaxing and dilating arteries. Recently, we reported that the secreted protein thrombospondin-1 (TSP1), through its cell surface receptor CD47, limits the ability of NO to relax and dilate blood vessels and thus decreases tissue perfusion. In the present study, we tested the hypothesis that blocking TSP1-CD47 signaling increases ischemic tissue survival in random cutaneous porcine flaps. METHODS: Random cutaneous flaps 2 x 10 cm2 were raised in white hairless Yucatan miniature pigs and were treated with a monoclonal antibody to TSP1, an antisense morpholino oligonucleotide to CD47 or control agents and tissue survival assessed. Primary vascular smooth muscle cell cultured from Yucatan pigs were also treated with the same agents +/- and an NO donor (DEA/NO) and cGMP quantified. RESULTS: Antibody blockade of TSP1 or morpholino suppression of CD47 dramatically enhanced survival of random tissue flaps. These responses correlated with increased blood vessel patency and tissue blood flow on vessel injection studies. NO-stimulated cGMP flux in Yucatan vascular smooth muscle cell was abrogated after antibody or morpholino treatment. CONCLUSION: Antibody ligation of TSP1 or antisense morpholino knock down of CD47 greatly increased tissue survival to ischemia. Given the similarity between porcine and human soft tissues these results suggest significant therapeutic potential for people.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígeno CD47/metabolismo , Silenciador del Gen , Colgajos Quirúrgicos/irrigación sanguínea , Trombospondina 1/antagonistas & inhibidores , Supervivencia Tisular/efectos de los fármacos , Animales , Antígeno CD47/efectos de los fármacos , Antígeno CD47/genética , Isquemia , Oligodesoxirribonucleótidos Antisentido/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Porcinos , Porcinos Enanos , Técnicas de Cultivo de Tejidos , Supervivencia Tisular/fisiología
13.
Blood ; 111(2): 613-23, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17890448

RESUMEN

Platelet alpha-granules constitute the major rapidly releasable reservoir of thrombospondin-1 in higher animals. Although some fragments and peptides derived from thrombospondin-1 stimulate or inhibit platelet aggregation, its physiologic function in platelets has remained elusive. We now show that endogenous thrombospondin-1 is necessary for platelet aggregation in vitro in the presence of physiologic levels of nitric oxide (NO). Exogenous NO or elevation of cGMP delays thrombin-induced platelet aggregation under high shear and static conditions, and exogenous thrombospondin-1 reverses this delay. Thrombospondin-1-null murine platelets fail to aggregate in response to thrombin in the presence of exogenous NO or 8Br-cGMP. At physiologic concentrations of the NO synthase substrate arginine, thrombospondin-1-null platelets have elevated basal cGMP. Ligation of CD36 or CD47 is sufficient to block NO-induced cGMP accumulation and mimic the effect of thrombospondin-1 on aggregation. Exogenous thrombospondin-1 also reverses the suppression by NO of alphaIIb/beta3 integrin-mediated platelet adhesion on immobilized fibrinogen, mediated in part by increased GTP loading of Rap1. Thrombospondin-1 also inhibits cGMP-mediated activation of cGMP-dependent protein kinase and thereby prevents phosphorylation of VASP. Thus, release of thrombospondin-1 from alpha-granules during activation provides positive feedback to promote efficient platelet aggregation and adhesion by overcoming the antithrombotic activity of physiologic NO.


Asunto(s)
Plaquetas/metabolismo , GMP Cíclico/metabolismo , Fibrinolíticos/metabolismo , Óxido Nítrico/metabolismo , Agregación Plaquetaria/fisiología , Trombospondina 1/metabolismo , Animales , Arginina/genética , Arginina/metabolismo , Plaquetas/citología , Antígenos CD36/genética , Antígenos CD36/metabolismo , Antígeno CD47/genética , Antígeno CD47/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , GMP Cíclico/antagonistas & inhibidores , GMP Cíclico/genética , GMP Cíclico/farmacología , Fibrinolíticos/farmacología , Recubrimiento Inmunológico/efectos de los fármacos , Recubrimiento Inmunológico/fisiología , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/genética , Péptidos/genética , Péptidos/metabolismo , Péptidos/farmacología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Adhesividad Plaquetaria/efectos de los fármacos , Adhesividad Plaquetaria/genética , Agregación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/genética , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Vesículas Secretoras/genética , Vesículas Secretoras/metabolismo , Resistencia al Corte , Trombina/genética , Trombina/metabolismo , Trombina/farmacología , Trombospondina 1/genética , Trombospondina 1/farmacología , Proteínas de Unión al GTP rap1/genética , Proteínas de Unión al GTP rap1/metabolismo
14.
Ann Surg ; 247(1): 180-90, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18156939

RESUMEN

BACKGROUND: Skin graft survival and healing requires rapid restoration of blood flow to the avascular graft. Failure or delay in the process of graft vascularization is a significant source of morbidity and mortality. One of the primary regulators of blood flow and vessel growth is nitric oxide (NO). The secreted protein thrombospondin-1 (TSP1) limits NO-stimulated blood flow and growth and composite tissue survival to ischemia. We herein demonstrate a role for TSP1 in regulating full thickness skin graft (FTSG) survival. METHODS AND RESULTS: FTSG consistently fail in wild type C57BL/6 mice but survive in mice lacking TSP1 or its receptor CD47. Ablation of the TSP1 receptor CD36, however, did not improve FTSG survival. Remarkably, wild type FTSG survived on TSP1 null or CD47 null mice, indicating that TSP1 expression in the wound bed is the primary determinant of graft survival. FTSG survival in wild type mice could be moderately improved by increasing NO flux, but graft survival was increased significantly through antibody blocking of TSP1 binding to CD47 or antisense morpholino oligonucleotide suppression of CD47. CONCLUSIONS: TSP1 through CD47 limits skin graft survival. Blocking TSP1 binding or suppressing CD47 expression drastically increases graft survival. The therapeutic applications of this approach could include burn patients and the broader group of people requiring grafts or tissue flaps for closure and reconstruction of complex wounds of diverse etiologies.


Asunto(s)
Antígeno CD47/metabolismo , Trasplante de Piel , Trombospondina 1/metabolismo , Análisis de Varianza , Animales , Velocidad del Flujo Sanguíneo , Western Blotting , Supervivencia Celular , Modelos Animales de Enfermedad , Supervivencia de Injerto , Flujometría por Láser-Doppler , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Necrosis/prevención & control , Óxido Nítrico/farmacología
15.
Biochem Pharmacol ; 75(4): 875-82, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18068687

RESUMEN

ABT-510 is a potent mimetic of an anti-angiogenic sequence from the second type 1 repeat of thrombospondin-1. ABT-510 and the original d-Ile mimetic from which it was derived, GDGV(dI)TRIR, are similarly active for inhibiting vascular outgrowth in a B16 melanoma explant assay. Because GDGV(dI)TRIR and thrombospondin-1 modulate nitric oxide signaling by inhibiting the fatty translocase activity of CD36, we examined the ability ABT-510 to modulate fatty acid uptake into vascular cells and downstream nitric oxide/cGMP signaling. Remarkably, ABT-510 is less active than GDGV(dI)TRIR for inhibiting myristic acid uptake into both endothelial and vascular smooth muscle cells. Correspondingly, ABT-510 is less potent than GDGV(dI)TRIR for blocking a myristate-stimulated increase in cell adhesion to collagen and nitric oxide-driven accumulation of cGMP. ABT-510 at concentrations sufficient to inhibit CD36 fatty acid translocase activity synergizes with thrombin in aggregating platelets and blunts the activity of NO to delay aggregation, but again less than GDGV(dI)TRIR. In contrast, ABT-510 is more potent than GDGV(dI)TRIR for inducing caspase activation in vascular cells. Thus, we propose that ABT-510 is a drug with at least two mechanisms of action, and its potent anti-tumor activity may be in part independent of CD36 fatty acid translocase inhibition.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antígenos CD36/metabolismo , Caspasas/metabolismo , Ácidos Grasos/metabolismo , Músculo Liso Vascular , Óxido Nítrico/biosíntesis , Oligopéptidos/farmacología , Trombospondina 1/metabolismo , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular , GMP Cíclico/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Transducción de Señal , Trombospondina 1/genética
16.
J Biol Chem ; 282(21): 15404-15, 2007 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-17416590

RESUMEN

Although CD36 is generally recognized to be an inhibitory signaling receptor for thrombospondin-1 (TSP1), the molecular mechanism for transduction of this signal remains unclear. Based on evidence that myristic acid and TSP1 each modulate endothelial cell nitric oxide signaling in a CD36-dependent manner, we examined the ability of TSP1 to modulate the fatty acid translocase activity of CD36. TSP1 and a CD36 antibody that mimics the activity of TSP1 inhibited myristate uptake. Recombinant TSP1 type 1 repeats were weakly inhibitory, but an anti-angiogenic peptide derived from this domain potently inhibited myristate uptake. This peptide also inhibited membrane translocation of the myristoylated CD36 signaling target Fyn and activation of Src family kinases. Myristate uptake stimulated cGMP synthesis via endothelial nitric-oxide synthase and soluble guanylyl cyclase. CD36 ligands blocked myristate-stimulated cGMP accumulation in proportion to their ability to inhibit myristate uptake. TSP1 also inhibited myristate-stimulated cGMP synthesis by engaging its receptor CD47. Myristate stimulated endothelial and vascular smooth muscle cell adhesion on type I collagen via the NO/cGMP pathway, and CD36 ligands that inhibit myristate uptake blocked this response. Therefore, the fatty acid translocase activity of CD36 elicits proangiogenic signaling in vascular cells, and TSP1 inhibits this response by simultaneously inhibiting fatty acid uptake via CD36 and downstream cGMP signaling via CD47.


Asunto(s)
Antígenos CD36/metabolismo , Ácido Mirístico/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Óxido Nítrico/metabolismo , Péptidos/farmacología , Transducción de Señal/efectos de los fármacos , Trombospondina 1/farmacología , Transporte Biológico Activo/efectos de los fármacos , Transporte Biológico Activo/fisiología , Antígeno CD47/metabolismo , Células Cultivadas , GMP Cíclico/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Proteínas de Transporte de Ácidos Grasos/metabolismo , Humanos , Neovascularización Fisiológica/fisiología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Péptidos/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Transducción de Señal/fisiología , Trombospondina 1/metabolismo , Familia-src Quinasas/metabolismo
17.
J Biol Chem ; 281(36): 26069-80, 2006 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-16835222

RESUMEN

CD36 is necessary for inhibition of some angiogenic responses by the matricellular glycoprotein thrombospondin-1 and is therefore assumed to be the receptor that mediates its anti-angiogenic activities. Although ligation of CD36 by antibodies, recombinant type 1 repeats of thrombospondin-1, or CD36-binding peptides was sufficient to inhibit nitric oxide (NO)-stimulated responses in both endothelial and vascular smooth muscle cells, picomolar concentrations of native thrombospondin-1 similarly inhibited NO signaling in vascular cells from wild-type and CD36-null mice. Ligation of the thrombospondin-1 receptor CD47 by recombinant C-terminal regions of thrombospondin-1, thrombospondin-1 peptides, or CD47 antibodies was also sufficient to inhibit NO-stimulated phenotypic responses and cGMP signaling in vascular cells. Thrombospondin-1 did not inhibit NO signaling in CD47-null vascular cells or NO-stimulated vascular outgrowth from CD47-null muscle explants in three-dimensional cultures. Furthermore, the CD36-binding domain of thrombospondin-1 and anti-angiogenic peptides derived from this domain failed to inhibit NO signaling in CD47-null cells. Therefore, ligation of either CD36 or CD47 is sufficient to inhibit NO-stimulated vascular cell responses and cGMP signaling, but only CD47 is necessary for this activity of thrombospondin-1 at physiological concentrations.


Asunto(s)
Antígeno CD47/metabolismo , Células Endoteliales/metabolismo , Músculo Liso Vascular , Óxido Nítrico , Transducción de Señal/fisiología , Trombospondina 1/metabolismo , Animales , Antígenos CD36/genética , Antígenos CD36/metabolismo , Antígeno CD47/genética , Adhesión Celular/fisiología , Proliferación Celular , Células Cultivadas , GMP Cíclico/metabolismo , Células Endoteliales/citología , Humanos , Técnicas In Vitro , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Neovascularización Fisiológica , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/metabolismo , Péptidos/genética , Péptidos/metabolismo
18.
Free Radic Biol Med ; 40(6): 1028-33, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16540398

RESUMEN

Nitric oxide (NO) is an important regulator of angiogenesis and neovascularization. The nature of endothelial cell motility responses to NO was examined using a Boyden chamber method. NO generated via decomposition of either DEA/NO or DETA/NO produced increases in human umbilical vein endothelial cell (HUVEC) chemotaxis, which were completely abrogated by ODQ, a soluble guanylyl cyclase inhibitor. Measurements of NO either directly by chemiluminescence or its chemistry with diaminofluorescein revealed that chemotaxis was driven by subtle NO gradients between the lower and the upper wells in this system. In addition to diffusion and volatilization from the upper chambers, the data showed that HUVEC consumption of NO contributed to these sustained gradients. Comparison of DEA/NO- and DETA/NO-mediated responses suggested that the persistence of spatial NO gradients is as significant as the absolute magnitude of NO exposure per unit time. The findings suggest that subnanomolar NO gradients are sufficient to mobilize endothelial cell migration into hypoxic tissue during neovascularization events, such as in wound healing and cancer.


Asunto(s)
Quimiotaxis/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Guanilato Ciclasa/fisiología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Difusión , Células Endoteliales/fisiología , Endotelio Vascular/citología , Humanos , Hidrazinas/metabolismo , Neovascularización Fisiológica , Óxido Nítrico/farmacología , Óxidos de Nitrógeno/metabolismo , Oxadiazoles/farmacología , Quinoxalinas/farmacología , Triazenos/metabolismo , Volatilización
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA