Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncogenesis ; 6(1): e287, 2017 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-28092369

RESUMEN

A subset of clear cell renal cell carcinoma (ccRCC) tumors exhibit a HIF1A gene mutation, yielding two ccRCC tumor types, H1H2 type expressing both HIF1α and HIF2α, and H2 type expressing HIF2α, but not functional HIF1α protein. However, it is unclear how the H1H2 type ccRCC tumors escape HIF1's tumor-suppressive activity. The polybromo-1 (PBRM1) gene coding for the BAF180 protein, a component of the SWItch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeling complex, is inactivated in 40% ccRCCs, the function and mechanism of BAF180 mutation is unknown. Our previous study indicates that BAF180-containing SWI/SNF chromatin remodeling complex is a co-activator for transcription factor HIF to induce HIF target genes. Thus, our questions are if BAF180 is involved in HIF-mediated hypoxia response and if PBRM1/BAF180 mutation has any association with the HIF1A retention in H1H2 type ccRCC. We report here that BAF180 is mutated in H1H2 ccRCC cell lines and tumors, and BAF180 re-expression in H1H2 ccRCC cell lines reduced cell proliferation/survival, indicating that BAF180 has tumor-suppressive role in these cells. However, BAF180 is expressed in HIF1-deficient H2 ccRCC cell lines and tumors, and BAF180 knockdown in H2 type ccRCC cell lines reduced cell proliferation/survival, indicating that BAF180 has tumor-promoting activity in these cells. In addition, our data show that BAF180 functions as co-activator for HIF1- and HIF2-mediated transcriptional response, and BAF180's tumor-suppressive and -promoting activity in ccRCC cell lines depends on co-expression of HIF1 and HIF2, respectively. Thus, our studies reveal that BAF180 function in ccRCC is context dependent, and that mutation of PBRM1/BAF180 serves as an alternative strategy for ccRCC tumors to reduce HIF1 tumor-suppressive activity in H1H2 ccRCC tumors. Our studies define distinct functional subgroups of ccRCCs based on expression of BAF180, and suggest that BAF180 inhibition may be a novel therapeutic target for patients with H2, but not H1H2, ccRCC tumors.

2.
Clin Nephrol ; 73(6): 446-8, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20497757

RESUMEN

AIMS: Recombinant human erythropoietin (rHuEPO) is widely used to treat anemia. A dialysis provider enacted a policy utilizing 20,000 U/ml multi-dose vials for rHuEPO dosing. The purpose of this study was to determine the accuracy and precision in administering small rHuEPO doses from this vial. METHODS: Ten registered nurses (RNs) were selected at random, supplied with a rHuEPO vial refilled with water, and instructed to withdraw the following amounts (1,200 U, 2,400 U, 3,600 U) using standard procedures and assuming the standard rHuEPO concentration of 20,000 U/ml. Samples were drawn up in duplicate and placed into 1.5 ml micro-centrifuge tubes. The volumes were measured using P-100 or P-200 microliter pipettes. The equivalent amount of rHuEPO was calculated by multiplying the volume by 20 U/microl. RESULTS: The rHuEPO dosing errors were large and on occasion greater than 100% at the 1,200 U dose. The variability for each RN, while large, was less than the inter-RN variability (within-RN % error 9.6% vs. 29.8% between-RN % error at the 1,200 unit dose). Errors occurred in both directions, both under- and overdosing. CONCLUSION: Utilizing concentrated rHuEPO resulted in significant dosing errors at low rHuEPO doses. The implications include inaccurate medication administration and disparity between administered and billed dosages. Policy decisions that effect medication administration need to be carefully evaluated to determine their impact on patient well-being and safety.


Asunto(s)
Eritropoyetina/administración & dosificación , Errores de Medicación , Humanos , Inyecciones , Enfermeras y Enfermeros , Proteínas Recombinantes
4.
J Biol Chem ; 276(46): 42737-43, 2001 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-11559711

RESUMEN

Activating mutations in ras genes are frequently associated with non-small cell lung cancer cells (NSCLC) and contribute to transformed growth in these cells. Expression of oncogenic forms of Ras in these cells is associated with increased expression and activity of cytosolic phospholipase A(2) (cPLA(2)) and cyclooxygenase-2 (COX-2), leading to constitutively elevated levels of prostaglandin production. Expression of oncogenic Ras is sufficient to induce these enzymes in normal lung epithelial cells. We have previously reported that the JNK and ERK pathways are necessary for induction of cPLA(2) and have defined a minimal region of the cPLA(2) promoter from -58 to -12 that is required for Ha-Ras-mediated induction. To further characterize the cis-regulatory elements within this region involved in this response, site-directed mutagenesis was used to make mutations at various sites. Three cis-regulatory elements were identified: regions -21/-18, -37/-30, and -55/-53. Mutations in any of these elements decreased basal and Ha-Ras-induced cPLA(2) promoter activity in both normal lung epithelial cells, as well as steady state promoter activity in A549 cells, with a mutation in element -21/-18 completely eliminating all promoter activity. Overexpression studies and gel shift assays indicated that Sp1 may serve as a transcription factor functionally regulating promoter activity by directly interacting with two of the cis-regulatory elements, -21/-18 and -37/-30. Expression of Ha-Ras led to induction of c-Jun protein, which showed functional cooperation with Sp1 in driving promoter activity. Additional unidentified transcription factors bound to the regions from -55/-53 and -37/-34.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Células Epiteliales/enzimología , Neoplasias Pulmonares , Pulmón/citología , Fosfolipasas A/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Factor de Transcripción Sp1/metabolismo , Secuencia de Bases , Línea Celular , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Regiones Promotoras Genéticas , Unión Proteica , Homología de Secuencia de Ácido Nucleico , Transcripción Genética , Transfección , Células Tumorales Cultivadas , Proteínas ras/metabolismo
5.
J Biol Chem ; 276(19): 15631-40, 2001 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-11278727

RESUMEN

Hypoxia has been shown to act as a proliferative stimulus for adventitial fibroblasts of the pulmonary artery. The signaling pathways involved in this growth response, however, remain unclear. We tested the hypothesis that hypoxia-induced proliferation of fibroblasts would be dependent on distinct (compared with serum) activation and utilization patterns of mitogen-activated protein (MAP) kinases initiated by Galpha(i/o) proteins. We found that hypoxia stimulated increases in DNA synthesis and growth of quiescent fibroblasts in the absence of exogenous mitogens and also markedly augmented serum-stimulated growth responses. Hypoxia caused a transient activation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK), the time course and pattern of which was somewhat similar to that induced by serum but which was of lesser magnitude. On the other hand, hypoxia-induced activation of p38 MAP kinase was biphasic, whereas serum-stimulated activation of p38 MAP kinase was transient, and the magnitude of activation was greater for hypoxia compared with that of serum stimulation. ERK1/2, JNK1, and p38 MAP kinase but not JNK2 were necessary for hypoxia-induced proliferation because PD98059, SB202190, and JNK1 antisense oligonucleotides nearly ablated the growth response. JNK2 appeared to act as a negative modulator of hypoxia-induced growth because JNK2 antisense oligonucleotides led to an increase in DNA synthesis. In serum-stimulated cells, antisense JNK1 oligonucleotides and PD98059 had inhibitory effects on proliferation, whereas SB202190 led to an increase in DNA synthesis. Pertussis toxin, which blocks Galpha(i/o)-mediated signaling, markedly attenuated hypoxia-induced DNA synthesis and activation of ERK and JNK but not p38 MAP kinase. We conclude that hypoxia itself can act as a growth promoting stimulus for subsets of bovine neonatal adventitial fibroblasts largely through Galpha(i/o)-mediated activation of a complex network of MAP kinases whose specific contributions to hypoxia-induced proliferation differ from traditional serum-induced growth signals.


Asunto(s)
Hipoxia de la Célula/fisiología , Fibroblastos/fisiología , Proteínas de Unión al GTP/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/fisiología , Animales , Animales Recién Nacidos , Aorta Torácica/citología , Aorta Torácica/fisiología , Bovinos , División Celular/efectos de los fármacos , Células Cultivadas , Medio de Cultivo Libre de Suero , Inhibidores Enzimáticos/farmacología , Fibroblastos/citología , Imidazoles/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos , Cinética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteína Quinasa 8 Activada por Mitógenos , Proteína Quinasa 9 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/genética , Músculo Liso Vascular/citología , Oligodesoxirribonucleótidos Antisentido/farmacología , Toxina del Pertussis , Fosforilación , Arteria Pulmonar/citología , Arteria Pulmonar/fisiología , Piridinas/farmacología , Factores de Virulencia de Bordetella/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos
6.
J Biol Chem ; 276(2): 1226-32, 2001 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-11042196

RESUMEN

Mutations in ras genes have been detected with high frequency in nonsmall cell lung cancer cells (NSCLC) and contribute to transformed growth of these cells. It has previously been shown that expression of oncogenic forms of Ras in these cells is associated with elevated expression of cytosolic phospholipase A(2) (cPLA(2)) and cyclooxygenase-2 (COX-2), resulting in high constitutive levels of prostaglandin production. To determine whether expression of constitutively active Ras is sufficient to induce expression of these enzymes in nontransformed cells, normal lung epithelial cells were transfected with H-Ras. Stable expression of H-Ras increased expression of cPLA(2) and COX-2 protein. Transient transfection with H-Ras increased promoter activity for both enzymes. H-Ras expression also activated all three families of MAP kinase: ERKs, JNKs, and p38 MAP kinase. Expression of constitutively active Raf did not increase either cPLA(2) or COX-2 promoter activity, but inhibition of the ERK pathway with pharmacological agents or expression of dominant negative ERK partially blocked the H-Ras-mediated induction of cPLA(2) promoter activity. Expression of dominant negative JNK kinases decreased cPLA(2) promoter activity in NSCLC cell lines and inhibited H-Ras-mediated induction in normal epithelial cells, whereas expression of constructs encoding constitutively active JNKs increased promoter activity. Inhibition of p38 MAP kinase or NF-kappaB had no effect on cPLA(2) expression. Truncational analysis revealed that the region of the cPLA(2) promoter from -58 to +12 contained sufficient elements to mediate H-Ras induction. We conclude that expression of oncogenic forms of Ras directly increases cPLA(2) expression in normal epithelial cells through activation of the JNK and ERK pathways.


Asunto(s)
Genes ras , Proteínas I-kappa B , Isoenzimas/genética , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfolipasas A/genética , Regiones Promotoras Genéticas , Prostaglandina-Endoperóxido Sintasas/genética , Mucosa Respiratoria/fisiología , Animales , Ciclooxigenasa 2 , Citosol/enzimología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Inducción Enzimática , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Genes Reporteros , Imidazoles/farmacología , Isoenzimas/biosíntesis , Proteínas Quinasas JNK Activadas por Mitógenos , Luciferasas/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Inhibidor NF-kappaB alfa , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Fosfolipasas A/biosíntesis , Fosfolipasas A2 , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Piridinas/farmacología , Ratas , Mucosa Respiratoria/citología , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos
7.
J Biol Chem ; 275(29): 22537-43, 2000 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-10807920

RESUMEN

Exposure of vascular smooth muscle cells to arginine vasopressin (AVP) increases smooth muscle alpha-actin (SM-alpha-actin) expression through activation of the SM- alpha-actin promoter. The goal of this study was to determine the role of the mitogen-activated protein kinase (MAP kinase) family in regulation of SM-alpha-actin expression. AVP activated all three MAP kinase family members: ERKs, JNKs, and p38 MAP kinase. Inhibition of JNKs or p38 decreased AVP-stimulated SM-alpha-actin promoter activity, whereas inhibition of ERKs had no effect. A 150-base pair region of the promoter containing two CArG boxes was sufficient to mediate regulation by vasoconstrictors. Mutations in either CArG box decreased AVP-stimulated promoter activity. Electrophoretic mobility shift assays using oligonucleotides corresponding to either CArG box resulted in a complex of similar mobility whose intensity was increased by AVP. Antibodies against serum response factor (SRF) completely super-shifted this complex, indicating that SRF binds to both CArG boxes. Overexpression of SRF increased basal promoter activity, but activity was still stimulated by AVP. AVP stimulation rapidly increased SRF phosphorylation. These data indicate that both JNKs and p38 participate in regulation of SM- alpha-actin expression. SRF, which binds to two critical CArG boxes in the promoter, represents a potential target of these kinases.


Asunto(s)
Actinas/biosíntesis , Arginina Vasopresina/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/metabolismo , Vasoconstrictores/farmacología , Animales , Células Cultivadas , Activación Enzimática , Proteínas Quinasas JNK Activadas por Mitógenos , Ratas , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos
8.
Physiol Res ; 49(5): 503-17, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11191356

RESUMEN

Vascular repair in response to injury or stress (often referred to as remodeling) is a common complication of many cardiovascular abnormalities including pulmonary hypertension, systemic hypertension, atherosclerosis, vein graft remodeling and restenosis following balloon dilatation of the coronary artery. It is not surprising that repair and remodeling occurs frequently in the vasculature in that exposure of blood, vessels to either excessive hemodynamic stress (e.g. hypertension), noxious blood borne agents (e.g. atherogenic lipids), locally released cytokines, or unusual environmental conditions (e.g. hypoxia), requires readily available mechanisms to counteract these adverse stimuli and to preserve structure and function of the vessel wall. The responses, which were presumably evolutionarily developed to repair an injured tissue, often escape self-limiting control and can result, in the case of blood vessels, in lumen narrowing and obstruction to blood flow. Each cell type (i. e. endothelial cells, smooth muscle cells, and fibroblasts) in the vascular wall plays a specific role in the response to injury. However, while the roles of the endothelial cells and smooth muscle cells (SMC) in vascular remodeling have been extensively studied, relatively little attention has been given to the adventitial fibroblasts. Perhaps this is because the fibroblast is a relatively ill-defined cell which, at least compared to the SMC, exhibits few specific cellular markers. Importantly though, it has been well demonstrated that fibroblasts possess the capacity to express several functions such as migration, rapid proliferation, synthesis of connective tissue components, contraction and cytokine production in response to activation or stimulation. The myriad of responses exhibited by the fibroblasts, especially in response to stimulation, suggest that these cells could play a pivotal role in the repair of injury. This fact has been well documented in the setting of wound healing where a hypoxic environment has been demonstrated to be critical in the cellular responses. As such it is not surprising that fibroblasts may play an important role in the vascular response to hypoxia and/or injury. This paper is intended to provide a brief review of the changes that occur in the adventitial fibroblasts in response to vascular stress (especially hypoxia) and the role the activated fibroblasts might play in hypoxia-mediated pulmonary vascular disease.


Asunto(s)
Fibroblastos/fisiología , Hipoxia/fisiopatología , Circulación Pulmonar , Animales , Vasos Sanguíneos/fisiopatología , Muerte Celular , División Celular , Fibroblastos/patología , Humanos
9.
Arterioscler Thromb Vasc Biol ; 19(12): 2884-93, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10591665

RESUMEN

The arterial media is comprised of heterogeneous smooth muscle cell (SMC) subpopulations with markedly different growth responses to pathophysiological stimuli. Little information exists regarding the intracellular signaling pathways that contribute to these differences. Therefore, we investigated the growth-related signaling pathways in a unique subset of subendothelial SMCs (L1 cells) from normal, mature, bovine arteries and compared them with those in "traditional" SMCs derived from the middle media (L2 SMCs). Subendothelial L1 cells exhibited serum-independent autonomous growth, not observed in L2 SMCs. Autonomous growth of L1 cells was driven largely by the constitutively activated extracellular signal-regulated kinase (ERK-1/2) cascade. Inhibition of upstream activators of ERKs (MAP kinase kinase-1, p21(ras), receptor tyrosine kinases, and Gi protein-coupled receptors) led to suppression of autonomous growth in these cells. L1 cells also exhibited constitutive activation of important downstream targets of ERKs (cytosolic phospholipase A(2), cyclooxygenase-2) and secreted large amounts of prostaglandins. Importantly, L1 cells secreted potent mitogenic factor(s), which could potentially contribute in an autocrine fashion to the constitutive activation of these cells. Our data suggest that unique arterial cells with autonomous growth potential and constitutively activated signaling pathways exist in normal arteries and may contribute selectively to the pathogenesis of vascular diseases.


Asunto(s)
Endotelio Vascular/citología , Endotelio Vascular/enzimología , Sistema de Señalización de MAP Quinasas/fisiología , Angiotensina II/farmacología , Animales , Anticoagulantes/farmacología , Aorta Torácica/citología , Becaplermina , Proteínas Sanguíneas/farmacología , Bovinos , División Celular/efectos de los fármacos , División Celular/fisiología , Tamaño de la Célula/fisiología , Medios de Cultivo Condicionados/farmacología , Medio de Cultivo Libre de Suero/farmacología , Ciclooxigenasa 2 , Dinoprostona/biosíntesis , Endotelina-1/farmacología , Epoprostenol/biosíntesis , Proteínas de Unión al GTP/agonistas , Proteínas de Unión al GTP/antagonistas & inhibidores , Proteínas de Unión al GTP/metabolismo , Regulación Enzimológica de la Expresión Génica , Isoenzimas/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/enzimología , Comunicación Paracrina/efectos de los fármacos , Comunicación Paracrina/fisiología , Fosfolipasas A/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Proteínas Proto-Oncogénicas c-sis , Arteria Pulmonar/citología , Túnica Media/citología , Vasoconstrictores/farmacología
10.
Kidney Int ; 55(6): 2281-8, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10354276

RESUMEN

BACKGROUND: Nitric oxide (NO) and its metabolite, peroxynitrite (ONOO-), are involved in renal tubular cell injury. We postulated that if NO/ONOO- has an effect to reduce cell adhesion to the basement membrane, this may contribute to tubular obstruction and may be partially responsible for the harmful effect of NO on the tubular epithelium during acute renal failure (ARF). METHODS: We examined the effect of the NO donors (z)-1-[2-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1- ium-1, 2-diolate (DETA/NO), spermine NONOate (SpNO), and the ONOO- donor 3-morpholinosydnonimine (SIN-1) on cell-matrix adhesion to collagen types I and IV and fibronectin using three renal tubular epithelial cell lines: LLC-PK1, BSC-1, and OK. RESULTS: In LLC-PK1 cells, DETA/NO (500 microM) had no effect, and SpNO (500 microM) had a modest effect on cell adhesion compared with controls. Exposure to SIN-1 caused a dose-dependent impairment in cell-matrix adhesion. Similar results were obtained in the different cell types and matrix proteins. The effect of SIN-1 (500 microM) on LLC-PK1 cell adhesion was not associated with either cell death or alteration of matrix protein and was attenuated by either the NO scavenger 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide, the superoxide scavenger superoxide dismutase, or the ONOO- scavenger uric acid in a dose-dependent manner. CONCLUSIONS: These results therefore support the possibility that ONOO- generated in the tubular epithelium during ischemia/reperfusion has the potential to impair the adhesion properties of tubular cells, which then may contribute to the tubular obstruction in ARF.


Asunto(s)
Túbulos Renales/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular , Chlorocebus aethiops , Colágeno/metabolismo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Fibronectinas/metabolismo , Depuradores de Radicales Libres/farmacología , Túbulos Renales/citología , Túbulos Renales/metabolismo , Células LLC-PK1 , Molsidomina/análogos & derivados , Molsidomina/farmacología , Nitratos/farmacología , Óxido Nítrico/farmacología , Zarigüeyas , Superóxidos/farmacología , Porcinos
11.
Adv Exp Med Biol ; 474: 231-58, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10635005

RESUMEN

Mammals respond to reduced oxygen concentrations (hypoxia) in many different ways at the systemic, local, cellular and molecular levels. Within the pulmonary circulation, exposure to chronic hypoxia has been demonstrated to illicit increases in pulmonary artery pressure as well as dramatic structural changes in both large and small vessels. It has become increasingly clear that the response to hypoxia in vivo is differentially regulated at the level of specific cell types within the vessel wall. For instance, in large pulmonary blood vessels there is now convincing evidence to suggest that the medial layer is made up of many different subpopulations of smooth muscle cells. In response to hypoxia there are remarkable differences in the proliferative and matrix producing responses of these cells to the hypoxic environment. Some cell populations proliferate and increase matrix protein synthesis, while in other cell populations no apparent change in the proliferative or differentiation state of the cell takes place. In more peripheral vessels, the predominant proliferative changes in response to hypoxia in the pulmonary circulation occur in the adventitial layer rather than in the medial layer. Here again, specific increases in proliferation and matrix protein synthesis take place. Accumulating evidence suggests that the unique responses exhibited by specific cell types of hypoxia in vivo can be modeled in vitro. We have isolated, in culture, specific medial cell populations which demonstrate significant increases in proliferation in response to hypoxia, and others which exhibit no change or, in fact, a decrease in proliferation under hypoxic conditions. We have also isolated and cloned several unique populations of adventitial fibroblasts. There is good evidence that only certain fibroblast populations are capable of responding to hypoxia with an increase in proliferation. We have begun to elucidate the signaling pathways which are activated in those cell populations that exhibit proliferative responses to hypoxia. We show that hypoxia, in the absence of serum or mitogens, specifically activates select members of the protein kinase C isozyme family, as well as members of the mitogen-activated protein kinase (MAPK) family of proteins. This selective activation appears to take place in response to hypoxia only in those cells exhibiting a proliferative response, and antagonists of this pathway inhibit the response. Thus, there appear to be cells within each organ that demonstrate unique responses to hypoxia. A better understanding of why these cells exist and how they specifically transduce hypoxia-mediated signals will lead to a better understanding of how the changes in the pulmonary circulation take place under conditions of chronic hypoxia.


Asunto(s)
Hipoxia de la Célula/fisiología , Regulación de la Expresión Génica , Hipertensión Pulmonar/fisiopatología , Hipoxia/fisiopatología , Músculo Liso Vascular/fisiología , Animales , Humanos , Hipertensión Pulmonar/genética , Mamíferos , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiopatología , Arteria Pulmonar/fisiología , Arteria Pulmonar/fisiopatología
12.
Kidney Int ; 53(6): 1642-6, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9607195

RESUMEN

Nitric oxide (NO) has been shown to be a mediator of hypoxic injury in rat renal proximal tubules (PT). However, the role of NO in hypoxic injury to mouse. PT has not been examined. The aim of the present study was to determine the effect of knockout of nitric oxide synthase (NOS) isoforms on hypoxic injury in mouse PT. Mouse PTs were isolated by collagenase digestion and Percoll centrifugation. The nonselective NOS inhibitor, N-nitro-L-arginine methyl ester (L-NAME, 10 mM), but not its inactive stereoisomer D-NAME, protected against hypoxic injury as assessed by LDH release. Carboxy-imidazolineoxyl N-oxide (carboxy-PTIO, 100 microM), a stable NO scavenger, also afforded cytoprotection against hypoxic injury. To determine the role of the different NOS isoforms in the hypoxic injury, we examined the effect of hypoxia on PT isolated from knockout mice in which either the inducible NOS (iNOS) endothelial NOS (eNOS) or neuronal NOS (nNOS) gene was lacking. PT isolated from iNOS knockout mice were resistant to hypoxic injury compared to wild-type controls. In contrast, PT isolated from both nNOS and eNOS knockout mice were not protected against hypoxic injury. In conclusion, the present study demonstrates that NO is a mediator of hypoxic PT injury in the mouse and that knockout of the iNOS gene is cytoprotective against this hypoxic PT injury.


Asunto(s)
Hipoxia/metabolismo , Túbulos Renales Proximales/metabolismo , Ratones Noqueados/genética , Ratones Noqueados/metabolismo , Óxido Nítrico Sintasa/genética , Animales , Benzoatos/farmacología , Inhibidores Enzimáticos/farmacología , Eliminación de Gen , Imidazoles/farmacología , Técnicas In Vitro , L-Lactato Deshidrogenasa/metabolismo , Ratones , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo I , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III
13.
Front Biosci ; 3: d194-207, 1998 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-9456345

RESUMEN

Arginine vasopressin (AVP) exhibits both acute and long-term effects on vascular smooth muscle cells (VSMC). Acutely, AVP regulates vascular tone and stimulates contraction. Longer term exposure of VSMC to AVP in the absence of other mitogenic agents results in cell hypertrophy without increases in cell number, and increased expression of a number of muscle-specific genes including the smooth muscle form of alpha-actin (SM-alpha-actin). These responses can be distinguished from the proliferative responses seen with growth factors such as platelet-derived growth factor (PDGF), which increase DNA synthesis and cell number and suppress SM-alpha-actin expression. In cultured VSMC, all the effects of AVP are mediated through the V1a receptor which signals through G-proteins. This review examines post-receptor signaling events mediated by AVP in VSMC. AVP rapidly increases intracellular Ca2+ via mobilization of intracellular stores and entry of extracellular Ca2+ via specific cation channels. This pathway, via activation of myosin light chain kinase, is critical for the early contractile response. Increased intracellular Ca2+ also leads to increased arachidonic acid release and eicosanoid production through the action of phospholipase A2. The activation of protein kinases by AVP is examined, focusing on members of the mitogen-activated protein kinase family. These enzymes are likely to play an important role in promoting growth of VSMC as well as modulating their state of differentiation through transcriptional control of muscle-specific gene expression. Recent studies suggesting a role for c-Jun amino terminal kinases in the regulation of smooth muscle-alpha-actin expression are described.


Asunto(s)
Músculo Liso Vascular/metabolismo , Transducción de Señal , Vasopresinas/fisiología , Actinas/genética , Animales , Ácido Araquidónico/metabolismo , Calcio/metabolismo , Eicosanoides/biosíntesis , Expresión Génica/efectos de los fármacos , Humanos , Modelos Biológicos , Músculo Liso Vascular/efectos de los fármacos , Fosforilación , Proteínas Quinasas/metabolismo , Vasopresinas/farmacología
14.
Hepatology ; 27(1): 42-7, 1998 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9425915

RESUMEN

Indirect evidence suggests that the renal and vascular production of prostaglandins is increased in cirrhosis with ascites. However, the activity of the enzymes regulating the prostaglandin pathway has not been investigated in cirrhosis. The aim of the current study was to determine the activity of phospholipase A2 (PLA2), the key enzyme in the regulation of prostaglandin synthesis, in kidney and vascular tissue obtained from rats with carbon tetrachloride-induced cirrhosis and ascites (n = 9) and control rats (n = 6). PLA2 activity was assayed in vitro using [14C]arachidonyl-phosphatidylcholine (PC) and [14C]arachidonyl-phosphatidylethanolamine (PE) as substrates in the presence of Ca2+. Kidneys from cirrhotic rats had significantly higher PLA2 activity compared with control rats, with both PC and PE (35 +/- 5 and 40 +/- 6 vs. 21 +/- 2 and 26 +/- 3 pmol/mg/min, respectively; P < .05 for both). PLA2 activity was increased in the renal cortex as well as in the renal medulla. Fractionation of the kidney extracts by Mono-Q anion-exchange chromatography showed that the elution position of PLA2 activity corresponded to the cytosolic PLA2 isoform (cPLA2). Increased amounts of cPLA2 protein were found in kidney extracts immunoblotted with an anti-cPLA2 antibody However, reverse-transcriptase polymerase chain reaction (RT-PCR) analysis did not detect any difference in cPLA2 mRNA. PLA2 activity was also higher in aortic tissue from cirrhotic rats than in controls (PC 38 +/- 5 vs. 26 +/- 1 and PE 66 +/- 8 vs. 41 +/- 3 pmol/mg/min; P < .05 for both). Incubation of renal and aortic extracts from cirrhotic rats with anti-cPLA2 antibody reduced PLA2 activity by 64% and 88%, respectively. In conclusion, PLA2 activity is increased in kidneys and vascular tissue from cirrhotic rats with ascites. This can be accounted for by an induction of cPLA2, which would mediate, at least in part, the increased renal and vascular production of prostaglandins in cirrhosis.


Asunto(s)
Aorta/enzimología , Citosol/metabolismo , Riñón/enzimología , Cirrosis Hepática Experimental/metabolismo , Fosfolipasas A/metabolismo , Animales , Ascitis/etiología , Cromatografía por Intercambio Iónico , Immunoblotting , Cirrosis Hepática Experimental/complicaciones , Masculino , Fosfolipasas A/genética , Fosfolipasas A2 , Reacción en Cadena de la Polimerasa , Ratas , Ratas Sprague-Dawley , Valores de Referencia , Transcripción Genética
15.
J Biol Chem ; 272(41): 25845-50, 1997 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-9325315

RESUMEN

Prolonged exposure of vascular smooth muscle cells (VSMC) to vasoconstrictors such as vasopressin or angiotensin II induces hypertrophy and increases expression of muscle-specific genes including smooth muscle alpha-actin (SM-alpha-actin). These vasoconstrictors signal through G-proteins, including members of the Gq family. To further investigate the role of Gq family members, VSMC were transfected with a constitutively active mutant of a Gq family member, Galpha16 (Galpha16Q212L). Stable expression of Galpha16Q212L persistently stimulated phospholipase C, resulting in increased basal levels of inositol phosphates. These cells were hypertrophied and expressed elevated levels of SM-alpha-actin compared with wild-type VSMC or cells transfected with a control plasmid (Neo). SM-alpha-actin promoter activity was markedly increased in cells stably or transiently expressing Galpha16Q212L. Basal c-Jun-NH2-terminal kinase (JNK) activity was increased 3-9-fold in cells stably expressing Galpha16Q212L, while basal activity of the p42/44 mitogen-activated protein kinases (ERKs) was unaffected. Transient expression of a kinase inactive JNK kinase partially inhibited induction of SM-alpha-actin promoter activity in response to vasoconstrictors or expression of Galpha16Q212L. These results indicate that expression of constitutively active Galpha16 in VSMC mimics the effects of vasoconstrictors on hypertrophy and muscle-specific gene expression, and activation of JNK may play a role in these responses.


Asunto(s)
Actinas/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteínas de Unión al GTP/metabolismo , Proteínas Quinasas Activadas por Mitógenos , Músculo Liso Vascular/metabolismo , Vasoconstricción , Actinas/genética , Animales , Células Cultivadas , Proteínas Quinasas JNK Activadas por Mitógenos , Regiones Promotoras Genéticas , Ratas , Transfección
16.
Kidney Int ; 52(5): 1271-8, 1997 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9350650

RESUMEN

The effect of glycine on hypoxia- and ionomycin-induced increases in calpain activity in rat proximal tubules was determined. Calpain activity was determined both in vitro and in the intact cell using the fluorescent substrate N-succinyl-Leu-Leu-Val-Tyr-7-amido-4-methyl coumarin (N-succinyl-Leu-Leu-Val-Tyr-AMC) and Western blotting for calpain-specific spectrin breakdown products (BDP), respectively. At 7.5 minutes of hypoxia (prelethal injury model) there was a significant (10-fold) increase in in vitro calpain activity that was not inhibited by glycine. At 15 minutes of hypoxia (postlethal injury model) there was a further increase in calpain activity that was inhibited by glycine. Normoxic tubules incubated with the calcium ionophore ionomycin (5 microM) for two minutes and 10 minutes had a significant increase in calpain activity that was not inhibited by glycine. After 15 minutes of hypoxia in the presence of glycine, there was an increase in calpain-specific spectrin breakdown products (BDP) in both Triton X-100 soluble and cytosolic extracts from proximal tubules. Glycine in concentrations up to 10 mM had no direct effect on the in vitro calpain activity of purified calpains. The present study demonstrates that: (1) prelethal increases in calpain activity stimulated by hypoxia and ionomycin treatment are not affected by glycine; (2) calpain-mediated spectrin breakdown during hypoxia occurs in the presence of glycine; (3) glycine does prevent the additional postlethal increase in calpain activity probably by maintaining membrane integrity to calcium influx.


Asunto(s)
Calpaína/metabolismo , Glicina/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Animales , Calcio/metabolismo , Calpaína/antagonistas & inhibidores , Citoprotección , Ionomicina/farmacología , Túbulos Renales Proximales/enzimología , Masculino , Ratas , Ratas Sprague-Dawley , Espectrina/metabolismo
17.
Biochem Biophys Res Commun ; 237(2): 239-44, 1997 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-9268693

RESUMEN

Application of cyclic mechanical strain to vascular smooth muscle (VSM) cells elicits distinct cellular responses depending on extracellular matrix composition. We now examine activation of p42/p44 MAP kinase (ERK) and c-jun amino terminal kinase (JNK/SAPK) by cyclic (1 Hz) mechanical strain in neonatal rat VSM cells cultured on pronectin or laminin. In cells grown on pronectin, mechanical strain activated both ERKs (peak 10-30 min) and JNK/SAPK (peak 15-30 min). On laminin, mechanical strain induced a comparable activation of JNK/SAPK to that seen on pronectin, but no significant activation of ERKs. In contrast, application of strain to adult VSM cells activated both enzymes independently of extracellular matrix composition. In neonatal VSM cells, cyclic strain induced SM-1 smooth muscle myosin in cells cultured on laminin, but not on pronectin.. Thus in neonatal VSM cells, activation of ERKs and induction of SM-1 myosin by mechanical strain depend on extracellular matrix composition.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Matriz Extracelular/metabolismo , Proteínas Quinasas Activadas por Mitógenos , Músculo Liso Vascular/enzimología , Proteínas Tirosina Quinasas/metabolismo , Envejecimiento/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Activación Enzimática , Proteínas Quinasas JNK Activadas por Mitógenos , Proteína Quinasa 1 Activada por Mitógenos , Proteína Quinasa 3 Activada por Mitógenos , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología , Miosinas/metabolismo , Estimulación Física , Ratas , Ratas Sprague-Dawley
18.
J Biol Chem ; 272(23): 14501-4, 1997 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-9169405

RESUMEN

Mutations in Ras family members that confer oncogenic potential are frequently observed in specific human cancers. We report that human non-small cell lung cancer (NSCLC) lines that harbor oncogenic mutations in Ki-Ras (H460, A549, H2122) synthesized high levels of prostaglandin E2 (PGE2) compared with NSCLC lacking Ras mutations or non-transformed lung epithelial cells (BEAS-2B). This increased PGE2 production was mediated by constitutively high expression of 85-kDa cytosolic phospholipase A2 (cPLA2) and cyclooxygenase 2 (COX-2). The increased expression of cPLA2 protein was mediated through elevated mRNA levels and activation of the cPLA2 promoter. Induction of cPLA2 promoter activity was blocked by expression of dominant-negative forms of Ras. Inhibition of Ras by the farnesyltransferase inhibitor BZA-5B inhibited prostaglandin synthesis in H2122 cells by decreasing expression of both cPLA2 and COX-2. Finally, inhibitors of eicosanoid synthesis blocked anchorage-independent growth of NSCLC lines exhibiting Ki-Ras mutations. These results identify cPLA2 as a novel Ras-inducible regulator of eicosanoid synthesis that participates in cellular transformation.


Asunto(s)
Genes ras , Fosfolipasas A/biosíntesis , Mutación Puntual , Carcinoma de Pulmón de Células no Pequeñas , Línea Celular , Ciclooxigenasa 2 , Citosol/enzimología , Dinoprostona/biosíntesis , Inducción Enzimática , Epitelio , Humanos , Isoenzimas/biosíntesis , Cinética , Pulmón , Neoplasias Pulmonares , Proteínas de la Membrana , Fosfolipasas A2 , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Proteínas Proto-Oncogénicas p21(ras)/genética , Células Tumorales Cultivadas
19.
Biochem J ; 327 ( Pt 3): 709-16, 1997 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-9581546

RESUMEN

Platelet-derived growth factor (PDGF), which is a potent mitogen for vascular smooth-muscle cells (VSMC), also inhibits the expression of specific smooth-muscle proteins, including smooth-muscle alpha-actin (SM-alpha-actin), in these cells. The goal of this study was to identify signalling pathways mediating these distinct effects. In rat aortic VSMC, PDGF caused a rapid activation of Ras and Raf, leading to the activation of mitogen-activated protein kinases (ERKs). Cells stably transfected with constitutively active Ras (H-Ras) expressed low levels of SM-alpha-actin protein. Arginine vasopressin, which stimulated SM-alpha-actin promoter activity in wild-type cells or controls (Neo; transfected with a plasmid lacking an insert), failed to do so in cells transiently expressing H-Ras. The effects of Ras on suppression of SM-alpha-actin expression were not mediated by the Raf/ERK pathway, since cells stably expressing constitutively active Raf (BxB-Raf) had normal levels of SM-alpha-actin protein, and stimulation of SM-alpha-actin promoter activity by vasopressin was unaffected in cells transiently expressing BxB-Raf. Furthermore a specific inhibitor of ERK activation had no effect on SM-alpha-actin expression. Exposure of wild-type VSMC to PDGF, or stable expression of Ras but not Raf, also resulted in constitutive increases in prostaglandin E2 production and cytosolic phospholipase A2 (cPLA2) activity, which was mediated by an increased expression of cPLA2 protein. Transient expression of cPLA2 in wild-type VSMC inhibited the stimulation of SM-alpha-actin promoter activity by vasopressin. These results suggest that PDGF-induced inhibition of SM-alpha-actin expression is mediated through a Ras-dependent/Raf independent pathway involving the induction of cPLA2 and eicosanoid production.


Asunto(s)
Actinas/antagonistas & inhibidores , Regulación de la Expresión Génica , Músculo Liso Vascular/metabolismo , Fosfolipasas A/fisiología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Animales , Citosol/enzimología , Dinoprostona/biosíntesis , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/enzimología , Fosfolipasas A/biosíntesis , Fosfolipasas A2 , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Proteínas Proto-Oncogénicas c-raf/fisiología , Proteínas Proto-Oncogénicas p21(ras)/biosíntesis , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética , Transfección
20.
J Neurochem ; 67(6): 2599-608, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8931495

RESUMEN

Nerve growth cones isolated from fetal rat brain exhibit in their cytosol a robust level of phospholipase A2 activity hydrolyzing phosphatidylinositol (PI) and phosphatidylethanolamine (PE) but not phosphatidylcholine (PC). Western blot analysis with an antibody to the well-characterized cytosolic phospholipase A2 (mol wt 85,000) reveals only trace amounts of this PC- and PE-selective enzyme in growth cones. By gel filtration on Superose 12, growth cone phospholipase A2 activity elutes essentially as two peaks of high molecular mass, at approximately 65 kDa and at well over 100 kDa. Anion exchange chromatography completely separates a PI-selective from a PE-selective activity, indicating the presence of two different, apparently monoselective phospholipase A2 species. The PI-selective enzyme, the predominant phospholipase A2 activity in whole growth cones, is enriched greatly in these structures relative to their parent fractions from fetal brain. This phospholipase A2 is resistant to reducing agents and is found in the cytosol as well as membrane-associated in the presence of Ca2+. However, its catalytic activity is Ca(2+)-independent regardless of whether the enzyme is associated with pure substrate or mixed-lipid growth cone vesicles. The PE-selective phospholipase A2 in growth cones was studied in less detail but shares with the PI-selective enzyme several properties, including intracellular localization, the existence of cytosolic and membrane-associated forms, and Ca2+ independence. Our data indicate growth cones contain two high-molecular-weight forms of phospholipase A2 that share many properties with known, Ca(2+)-independent cytosolic phospholipase A2 species but that appear to be monoselective for PI and PE, respectively. In particular, the PI-selective enzyme may represent a new member of the growing family of cytoplasmic phospholipases A2. The enrichment of the PI-selective phospholipase A2 in growth cones suggests it plays a major role in the regulation of growth cone function.


Asunto(s)
Encéfalo/citología , Neuritas/enzimología , Fosfolipasas A/química , Fosfolipasas A/metabolismo , Animales , Ácido Araquidónico/metabolismo , Calcio/farmacología , Cromatografía en Gel , Cromatografía por Intercambio Iónico , Citosol/enzimología , Feto/citología , Feto/enzimología , Peso Molecular , Neuronas/enzimología , Neuronas/ultraestructura , Fosfatidilinositoles/metabolismo , Fosfolipasas A/efectos de los fármacos , Fosfolipasas A2 , Ratas , Fracciones Subcelulares/enzimología , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA