Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Pulm Circ ; 8(3): 2045894018778156, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29718758

RESUMEN

Pulmonary arterial (PA) wall modifications are key pathological features of pulmonary hypertension (PH). Although such abnormalities correlate with heightened phosphorylation of c-Jun N-terminal kinases 1/2 (JNK1/2) in a rat model of PH, the contribution of specific JNK isoforms to the pathophysiology of PH is unknown. Hence, we hypothesized that activation of either one, or both JNK isoforms regulates PA remodeling in PH. We detected increased JNK1/2 phosphorylation in the thickened vessels of PH patients' lungs compared to that in lungs of healthy individuals. JNK1/2 phosphorylation paralleled a marked reduction in MAP kinase phosphatase 1 (JNK dephosphorylator) expression in patients' lungs. Association of JNK1/2 activation with vascular modification was confirmed in the calf model of severe hypoxia-induced PH. To ascertain the role of each JNK isoform in pathophysiology of PH, wild-type (WT), JNK1 null (JNK1-/-), and JNK2 null (JNK2-/-) mice were exposed to chronic hypoxia (10% O2 for six weeks) to develop PH. In hypoxic WT lungs, an increase in JNK1/2 phosphorylation was associated with PH-like pathology. Hallmarks of PH pathophysiology, i.e. excessive accumulation of extracellular matrix and vessel muscularization with medial wall thickening, was also detected in hypoxic JNK1-/- lungs, but not in hypoxia-exposed JNK2-/- lungs. However, hypoxia-induced increases in right ventricular systolic pressure (RVSP) and in right ventricular hypertrophy (RVH) were similar in all three genotypes. Our findings suggest that JNK2 participates in PA remodeling (but likely not in vasoconstriction) in murine hypoxic PH and that modulating JNK2 actions might quell vascular abnormalities and limit the course of PH.

2.
Indian J Clin Biochem ; 29(2): 167-73, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24757298

RESUMEN

Reference intervals (RIs) of serum thyroid stimulating hormone (TSH) and free thyroxine (fT4) were determined in 402 healthy pregnant women by enzyme-linked immunosorbent assay (ELISA) technique after partitioning them into three trimesters. The reference population was chosen from a study population of 610 pregnant females by applying strict inclusion and exclusion criteria. The assays were done using proper quality control measures. RIs were calculated from the central 95 % of the distribution of TSH and fT4 values located between the lower reference limit of 2.5 percentile and upper reference limit of 97.5 percentile value 0.90 confidence intervals for the upper and lower reference limits were also determined. The reference intervals for TSH were 0.25-3.35 µIU/ml for the first trimester; 0.78-4.96 µIU/ml for the second trimester and 0.89-4.6 µIU/ml for the third trimester. Similarly, the reference intervals for fT4 for first, second and third trimesters were 0.64-2.0, 0.53-2.12 and 0.64-1.98 ng/dl respectively. The values thus obtained varied from those provided by the kit literature. In comparison to our derived reference intervals, the reference data from kit manufacturer under-diagnosed both subclinical hypo- and hyper-thyroidism within our pregnant reference population.

3.
PLoS One ; 9(4): e96043, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24763616

RESUMEN

RATIONALE: Lack of an experimental model of portopulmonary hypertension (POPH) has been a major obstacle in understanding of pathophysiological mechanisms underlying the disease. OBJECTIVE: We investigated the effects of CCl4-mediated cirrhosis on the pulmonary vasculature, as an initial step towards an improved understanding of POPH. METHODS AND RESULTS: Male C57BL/6 mice received intraperitoneal injection of either sterile olive oil or CCl4 3 times/week for 12 weeks. Cirrhosis and portal hypertension were confirmed by evidence of bridging fibrosis and nodule formation in CCl4-treated liver determined by trichrome/picrosirius red staining and an increase in spleen weight/body weight ratio, respectively. Staining for the oxidative stress marker, 4-hydroxynonenal (4-HNE), was strong in the liver but was absent in the lung, suggesting that CCl4 did not directly induce oxidative injury in the lung. Pulmonary acceleration time (PAT) and the ratio of PAT/pulmonary ejection time (PET) measured by echocardiography were significantly decreased in cirrhotic mice. Increase in right ventricle (RV) weight/body weight as well as in the weight ratio of RV/(left ventricle + septum) further demonstrated the presence of pathological changes in the pulmonary circulation in these mice. Histological examination revealed that lungs of cirrhotic mice have excessive accumulation of perivascular collagen and thickening of the media of the pulmonary artery. CONCLUSION: Collectively, our data demonstrate that chronic CCl4 treatment induces pathological changes in pulmonary circulation in cirrhotic mice. We propose that this murine cirrhotic model provides an exceptional tool for future studies of the molecular mechanisms mediating pulmonary vascular diseases associated with cirrhosis and for evaluation of novel therapeutic interventions.


Asunto(s)
Tetracloruro de Carbono , Hipertensión Pulmonar/patología , Cirrosis Hepática/patología , Pulmón/patología , Animales , Colágeno/metabolismo , Modelos Animales de Enfermedad , Ácidos Grasos Insaturados/metabolismo , Hidroxiácidos/metabolismo , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/fisiopatología , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/fisiopatología , Pulmón/metabolismo , Pulmón/fisiopatología , Masculino , Ratones Endogámicos C57BL , Estrés Oxidativo
4.
Am J Physiol Cell Physiol ; 306(7): C648-58, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24500281

RESUMEN

Epithelial cells are key players in the pathobiology of numerous hypoxia-induced lung diseases. The mechanisms mediating such hypoxic responses of epithelial cells are not well characterized. Earlier studies reported that hypoxia stimulates protein kinase C (PKC)δ activation in renal cancer cells and an increase in expression of a heparin-binding growth factor, midkine (MK), in lung alveolar epithelial cells. We reasoned that hypoxia might regulate MK levels via a PKCδ-dependent pathway and hypothesized that PKCδ-driven MK expression is required for hypoxia-induced lung epithelial cell proliferation and differentiation. Replication of human lung epithelial cells (A549) was significantly increased by chronic hypoxia (1% O2) and was dependent on expression of PKCδ. Hypoxia-induced proliferation of epithelial cells was accompanied by translocation of PKCδ from Golgi into the nuclei. Marked attenuation in MK protein levels by rottlerin, a pharmacological antagonist of PKC, and by small interfering RNA-targeting PKCδ, revealed that PKCδ is required for MK expression in both normoxic and hypoxic lung epithelial cells. Sequestering MK secreted into the culture media with a neutralizing antibody reduced hypoxia-induced proliferation demonstrating that an increase in MK release from cells is linked with epithelial cell division under hypoxia. In addition, recombinant MK accelerated transition of hypoxic epithelial cells to cells of mesenchymal phenotype characterized by elongated morphology and increased expression of mesenchymal markers, α-smooth muscle actin, and vimentin. We conclude that PKCδ/MK axis mediates hypoxic proliferation and differentiation of lung epithelial cells. Manipulation of PKCδ and MK activity in epithelial cells might be beneficial for the treatment of hypoxia-mediated lung diseases.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Células Epiteliales/enzimología , Pulmón/enzimología , Factores de Crecimiento Nervioso/metabolismo , Proteína Quinasa C-delta/metabolismo , Actinas/metabolismo , Transporte Activo de Núcleo Celular , Anticuerpos Neutralizantes/farmacología , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Hipoxia de la Célula , Línea Celular Tumoral , Núcleo Celular/enzimología , Proliferación Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Transición Epitelial-Mesenquimal , Aparato de Golgi/enzimología , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Midkina , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Factores de Crecimiento Nervioso/inmunología , Fenotipo , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/genética , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , Proteínas Recombinantes/metabolismo , Transducción de Señal , Factores de Tiempo , Transfección , Vimentina/metabolismo
5.
Pulm Circ ; 3(2): 447-8, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24015350
6.
Dev Neurosci ; 35(4): 293-305, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23751520

RESUMEN

Reactive oxygen species (ROS) have been reported to affect neural stem cell self-renewal and therefore may be important for normal development and may influence neurodegenerative processes when ROS activity is elevated. To determine if increasing production of superoxide, via activation of NADPH oxidase (Nox), increases neural stem cell proliferation, 100 nM angiotensin II (Ang II) - a strong stimulator of Nox - was applied to cultures of a murine neural stem cell line, C17.2. Twelve hours following a single treatment with Ang II, there was a doubling of the number of neural stem cells. This increase in neural stem cell numbers was preceded by a gradual elevation of superoxide levels (detected by dihydroethidium fluorescence) from the steady state at 0, 5, and 30 min and gradually increasing from 1 h to the maximum at 12 h, and returning to baseline at 24 h. Ang II-dependent proliferation was blocked by the antioxidant N-acetyl-L-cysteine. Confocal microscopy revealed the presence of two sources of intracellular ROS in C17.2 cells: (i) mitochondrial and (ii) extramitochondrial; the latter indicative of the involvement of one or more specific isoforms of Nox. Of the Nox family, mRNA expression for one member, Nox4, is abundant in neural stem cell cultures, and Ang II treatment resulted in elevation of the relative levels of Nox4 protein. SiRNA targeting of Nox4 mRNA reduced both the constitutive and Ang II-induced Nox4 protein levels and attenuated Ang II-driven increases in superoxide levels and stem cell proliferation. Our findings are consistent with our hypothesis that Ang II-induced proliferation of neural stem cells occurs via Nox4-generated superoxide, suggesting that an Ang II/Nox4 axis is an important regulator of neural stem cell self-renewal and as such may fine-tune normal, stress- or disease-modifying neurogenesis.


Asunto(s)
Angiotensina II/farmacología , Proliferación Celular/efectos de los fármacos , NADPH Oxidasas/metabolismo , Células-Madre Neurales/efectos de los fármacos , Superóxidos/metabolismo , Animales , Western Blotting , Recuento de Células , Células Cultivadas , Interpretación Estadística de Datos , Ratones , Microscopía Confocal , NADPH Oxidasa 4 , NADPH Oxidasas/genética , Células-Madre Neurales/ultraestructura , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
7.
Cardiovasc Res ; 95(3): 356-65, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22735370

RESUMEN

AIMS: Pulmonary hypertension (PH) is a devastating condition for which no disease-modifying therapies exist. PH is recognized as proliferative disease of the pulmonary artery (PA). In the experimental newborn calf model of hypoxia-induced PH, adventitial fibroblasts in the PA wall exhibit a heightened replication index. Because elevated platelet-derived growth factor ß receptor (PDGFß-R) signalling is associated with PH, we tested the hypothesis that the activation of PDGFß-R contributes to fibroblast proliferation and adventitial remodelling in PH. METHODS AND RESULTS: Newborn calves were exposed to either ambient air (P(B) = 640 mmHg) (Neo-C) or high altitude (P(B) = 445 mm Hg) (Neo-PH) for 2 weeks. PDGFß-R phosphorylation was markedly elevated in PA adventitia of Neo-PH calves as well as in cultured PA fibroblasts isolated from Neo-PH animals. PDGFß-R activation with PDGF-BB stimulated higher replication in Neo-PH cells compared with that of control fibroblasts. PDGF-BB-induced proliferation was dependent on reactive oxygen species generation and extracellular signal-regulated kinase1/2 activation in both cell populations; however, only Neo-PH cell division via PDGFß-R activation displayed a unique dependence on c-Jun N-terminal kinase1 (JNK1) stimulation as the blockade of JNK1 with SP600125, a pharmacological antagonist of the JNK pathway, and JNK1-targeted siRNA selectively blunted Neo-PH cell proliferation. CONCLUSIONS: Our data strongly suggest that hypoxia-induced modified cells engage the PDGFß-R-JNK1 axis to confer distinctively heightened proliferation and adventitial remodelling in PH.


Asunto(s)
Adventicia/enzimología , Proliferación Celular , Fibroblastos/enzimología , Hipertensión Pulmonar/enzimología , Hipoxia/enzimología , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Arteria Pulmonar/enzimología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Adventicia/patología , Altitud , Animales , Animales Recién Nacidos , Becaplermina , Bovinos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Activación Enzimática , Hipertensión Pulmonar Primaria Familiar , Fibroblastos/patología , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Hipoxia/etiología , Hipoxia/genética , Hipoxia/patología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 8 Activada por Mitógenos/genética , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-sis/metabolismo , Arteria Pulmonar/patología , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección
8.
Pulm Circ ; 2(4): 415-33, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23372926

RESUMEN

Genetically modified mouse models have unparalleled power to determine the mechanisms behind different processes involved in the molecular and physiologic etiology of various classes of human pulmonary hypertension (PH). Processes known to be involved in PH for which there are extensive mouse models available include the following: (1) Regulation of vascular tone through secreted vasoactive factors; (2) regulation of vascular tone through potassium and calcium channels; (3) regulation of vascular remodeling through alteration in metabolic processes, either through alteration in substrate usage or through circulating factors; (4) spontaneous vascular remodeling either before or after development of elevated pulmonary pressures; and (5) models in which changes in tone and remodeling are primarily driven by inflammation. PH development in mice is of necessity faster and with different physiologic ramifications than found in human disease, and so mice make poor models of natural history of PH. However, transgenic mouse models are a perfect tool for studying the processes involved in pulmonary vascular function and disease, and can effectively be used to test interventions designed against particular molecular pathways and processes involved in disease.

9.
J Neuroinflammation ; 8: 129, 2011 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-21975039

RESUMEN

BACKGROUND: Reactive oxygen species (ROS), superoxide and hydrogen peroxide (H2O2), are necessary for appropriate responses to immune challenges. In the brain, excess superoxide production predicts neuronal cell loss, suggesting that Parkinson's disease (PD) with its wholesale death of dopaminergic neurons in substantia nigra pars compacta (nigra) may be a case in point. Although microglial NADPH oxidase-produced superoxide contributes to dopaminergic neuron death in an MPTP mouse model of PD, this is secondary to an initial die off of such neurons, suggesting that the initial MPTP-induced death of neurons may be via activation of NADPH oxidase in neurons themselves, thus providing an early therapeutic target. METHODS: NADPH oxidase subunits were visualized in adult mouse nigra neurons and in N27 rat dopaminergic cells by immunofluorescence. NADPH oxidase subunits in N27 cell cultures were detected by immunoblots and RT-PCR. Superoxide was measured by flow cytometric detection of H2O2-induced carboxy-H2-DCFDA fluorescence. Cells were treated with MPP+ (MPTP metabolite) following siRNA silencing of the Nox2-stabilizing subunit p22phox, or simultaneously with NADPH oxidase pharmacological inhibitors or with losartan to antagonize angiotensin II type 1 receptor-induced NADPH oxidase activation. RESULTS: Nigral dopaminergic neurons in situ expressed three subunits necessary for NADPH oxidase activation, and these as well as several other NADPH oxidase subunits and their encoding mRNAs were detected in unstimulated N27 cells. Overnight MPP+ treatment of N27 cells induced Nox2 protein and superoxide generation, which was counteracted by NADPH oxidase inhibitors, by siRNA silencing of p22phox, or losartan. A two-wave ROS cascade was identified: 1) as a first wave, mitochondrial H2O2 production was first noted at three hours of MPP+ treatment; and 2) as a second wave, H2O2 levels were further increased by 24 hours. This second wave was eliminated by pharmacological inhibitors and a blocker of protein synthesis. CONCLUSIONS: A two-wave cascade of ROS production is active in nigral dopaminergic neurons in response to neurotoxicity-induced superoxide. Our findings allow us to conclude that superoxide generated by NADPH oxidase present in nigral neurons contributes to the loss of such neurons in PD. Losartan suppression of nigral-cell superoxide production suggests that angiotensin receptor blockers have potential as PD preventatives.


Asunto(s)
1-Metil-4-fenilpiridinio/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Línea Celular , Neuronas Dopaminérgicas/citología , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/metabolismo , Femenino , Herbicidas/farmacología , Peróxido de Hidrógeno/farmacología , Losartán/farmacología , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasas/química , NADPH Oxidasas/genética , Oxidantes/farmacología , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Sustancia Negra/citología
10.
Am J Pathol ; 178(1): 98-109, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21224048

RESUMEN

Although mitogen-activated protein kinase phosphatase-1 (MKP-1) is a key deactivator of MAP kinases, known effectors of lung vessel formation, whether it plays a role in the expression of proangiogenic vascular endothelial growth factor (VEGF) in hypoxic lung is unknown. We therefore hypothesized that MKP-1 is a crucial modulator of hypoxia-stimulated vessel development by regulating lung VEGF levels. Wild-type MKP-1(+/+), heterozygous MKP-1(+/-), and deficient MKP-1(-/-) mice were exposed to sea level (SL), Denver altitude (DA) (1609 m [5280 feet]), and severe high altitude (HYP) (∼5182 m [∼17,000 feet]) for 6 weeks. Hypoxia enhanced phosphorylation of p38 MAP kinase, a substrate of MKP-1, as well as α smooth muscle actin (αSMA) expression in vessels, respiratory epithelium, and interstitium of phosphatase-deficient lung. αSMA-positive vessel (<50 µm outside diameter) densities were markedly reduced, whereas vessel wall thickness was increased in hypoxic MKP-1(-/-) lung. Mouse embryonic fibroblasts (MEFs) of all three genotypes were isolated to pinpoint the mechanism involved in hypoxia-induced vascular abnormalities of MKP-1(-/-) lung. Sustained phosphorylation of p38 MAP kinase was observed in MKP-1-null MEFs in response to hypoxia exposure. Although hypoxia up-regulated VEGF levels in MKP-1(+/+) MEFs eightfold, only a 70% increase in VEGF expression was observed in MKP-1-deficient cells. Therefore, our data strongly suggest that MKP-1 might be the key regulator of vascular densities through the regulation of VEGF levels in hypoxic lung.


Asunto(s)
Fosfatasa 1 de Especificidad Dual/fisiología , Hipoxia/enzimología , Pulmón/irrigación sanguínea , Neovascularización Fisiológica , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Actinas/metabolismo , Animales , Fosfatasa 1 de Especificidad Dual/genética , Hipoxia/fisiopatología , Antígeno Ki-67/metabolismo , Pulmón/fisiología , Ratones , Ratones Mutantes , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Stem Cell Res Ther ; 1(5): 36, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21122109

RESUMEN

INTRODUCTION: Regulated neurotransmitter actions in the mammalian central nervous system determine brain function and control peripheral organs and behavior. Although drug-seeking behaviors, including alcohol consumption, depend on central neurotransmission, modification of neurotransmitter actions in specific brain nuclei remains challenging. Herein, we report a novel approach for neurotransmission modification in vivo by transplantation of stem cells engineered to take up the neurotransmitter dopamine (DA) efficiently through the action of the human dopamine transporter (hDAT). As a functional test in mice, we used voluntary alcohol consumption, which is known to release DA in nucleus accumbens (NAC), an event hypothesized to help maintain drug-seeking behavior. We reasoned that reducing extracellular DA levels, by engrafting into NAC DA-sequestering stem cells expressing hDAT, would alter alcohol intake. METHODS: We have generated a neural stem cell line stably expressing the hDAT. Uptake kinetics of DA were determined to select a clone for transplantation. These genetically modified stem cells (or cells transfected with a construct lacking the hDAT sequence) were transplanted bilaterally into the NAC of wild-type mice trained to consume 10% alcohol in a two-bottle free-choice test for alcohol consumption. Alcohol intake was then ascertained for 1 week after transplantation, and brain sections through the NAC were examined for surviving grafted cells. RESULTS: Modified stem cells expressed hDAT and uptaken DA selectively via hDAT. Mice accustomed to drinking 10% ethanol by free choice reduced their alcohol consumption after being transplanted with hDAT-expressing stem cells. By contrast, control stem cells lacked that effect. Histologic examination revealed surviving stem cells in the NAC of all engrafted brains. CONCLUSIONS: Our findings represent proof of principle suggesting that genetically engineered stem cells can be useful for exploring the role of neurotransmitters (or other signaling molecules) in alcohol consumption and potentially in other aspects of brain function.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Comportamiento de Búsqueda de Drogas/fisiología , Células-Madre Neurales/metabolismo , Consumo de Bebidas Alcohólicas/metabolismo , Animales , Células Cultivadas , Dopamina/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/biosíntesis , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/trasplante , Núcleo Accumbens/metabolismo , Ratas , Ratas Sprague-Dawley , Transfección , beta-Galactosidasa/metabolismo
12.
Cardiovasc Res ; 78(3): 440-8, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18218684

RESUMEN

AIMS: Cultured fibroblasts of hypoxia-stimulated remodelled pulmonary artery (PA) adventitia proliferate at a greater rate compared with those of normal adventitia. Since protein kinase C (PKC) zeta is a replication repressor of normal adventitial fibroblasts, we hypothesized that loss of the repressor activity of PKCzeta might contribute to increased rate of proliferation in adventitial cells of remodelled PA. METHODS AND RESULTS: Isolated PA adventitial fibroblasts of neonatal control (Fib-C) and chronic hypoxia-exposed (Fib-H) calves were used to test our hypothesis. For evaluation of the role of PKCzeta in hypoxia-induced vascular adventitial remodelling, expression and activation of PKCzeta were also examined in lung sections of Fib-C and Fib-H animals by immunoperoxidase staining. Although constitutively active PKCzeta expression attenuated DNA synthesis in Fib-C, it stimulated proliferation in Fib-H. PKCzeta-specific myristoylated pseudosubstrate peptide inhibitor (PKCzeta-PI) induced replication in Fib-C, whereas the inhibitor blocked DNA synthesis in Fib-H. Hypoxia stimulated PKCzeta as well as MAP kinase kinase (MEK)1/2 and extracellular signal-regulated kinase (ERK)1/2 phosphorylation in Fib-H cells. However, ERK1/2 activation was mediated by both MEK1/2-dependent and MEK1/2-independent PKCzeta-regulated mechanisms in hypoxia-exposed Fib-H. PKCzeta was selectively activated in the adventitial cells of the remodelled vascular wall, as demonstrated by strong immunoreactivity against the anti-phosphoPKCzeta antibody in the Fib-H lung sections. CONCLUSION: PKCzeta acts as a replication repressor in Fib-C cells; however, the same isozyme mediates Fib-H proliferation. Thus, chronic exposure to hypoxia leads to the emergence of cells lacking anti-replication activity of PKCzeta in the PA adventitia.


Asunto(s)
Proliferación Celular , Tejido Conectivo/enzimología , Fibroblastos/metabolismo , Hipoxia/enzimología , Proteína Quinasa C/metabolismo , Arteria Pulmonar/enzimología , Animales , Bovinos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Tejido Conectivo/efectos de los fármacos , Tejido Conectivo/patología , Modelos Animales de Enfermedad , Activación Enzimática , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Hipoxia/patología , Técnicas para Inmunoenzimas , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Péptidos/farmacología , Fenotipo , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/patología , Factores de Tiempo
13.
Neurochem Res ; 33(5): 886-901, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-17992568

RESUMEN

Among various types of neurons affected in Parkinson's disease, dopamine (DA) neurons of the substantia nigra undergo the most pronounced degeneration. Products of DA oxidation and consequent cellular damage have been hypothesized to contribute to neuronal death. To examine whether elevated intracellular DA will selectively predispose the dopaminergic subpopulation of nigral neurons to damage by an oxidative insult, we first cultured rat primary mesencephalic cells in the presence of rotenone to elevate reactive oxygen species. Although MAP2(+) neurons were more sensitive to rotenone-induced toxicity than type 1 astrocytes, rotenone affected equally both DA (TH(+)) neurons and MAP2(+) neurons. In contrast, when intracellular DA concentration was elevated, DA neurons became selectively sensitized to rotenone. Raising intracellular DA levels in primary DA neurons resulted in dopaminergic neuron death in the presence of subtoxic concentrations of rotenone. Furthermore, mitochondrial superoxide dismutase mimetic, manganese (III) meso-tetrakis (4-benzoic acid) porphyrin, blocked activation of caspase-3, and consequent cell death. Our results demonstrate that an inhibitor of mitochondrial complex I and increased cytosolic DA may cooperatively lead to conditions of elevated oxidative stress and thereby promote selective demise of dopaminergic neurons.


Asunto(s)
Apoptosis/efectos de los fármacos , Dopamina/farmacología , Neuronas/efectos de los fármacos , Rotenona/toxicidad , Animales , Dopamina/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Inmunohistoquímica , Neuronas/metabolismo , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley
14.
Physiology (Bethesda) ; 21: 134-45, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16565479

RESUMEN

An increasing volume of experimental data indicates that the adventitial fibroblast, in both the pulmonary and systemic circulations, is a critical regulator of vascular wall function in health and disease. A rapidly emerging concept is that the vascular adventitia acts as biological processing center for the retrieval, integration, storage, and release of key regulators of vessel wall function. In response to stress or injury, resident adventitial cells can be activated and reprogrammed to exhibit different functional and structural behaviors. In fact, under certain conditions, the adventitial compartment may be considered the principal injury-sensing tissue of the vessel wall. In response to vascular stresses such as overdistension and hypoxia, the adventitial fibroblast is activated and undergoes phenotypic changes, which include proliferation, differentiation, upregulation of contractile and extracellular matrix proteins, and release of factors that directly affect medial smooth muscle cell tone and growth and that stimulate recruitment of inflammatory and progenitor cells to the vessel wall. Each of these changes in fibroblast phenotype modulates either directly or indirectly changes in overall vascular function and structure. The purpose of this review is to present the current evidence demonstrating that the adventitial fibroblast acts as a key regulator of pulmonary vascular function and structure from the "outside-in."


Asunto(s)
Tejido Conectivo/fisiología , Endotelio Vascular/fisiología , Fibroblastos/fisiología , Arteria Pulmonar/fisiología , Venas Pulmonares/fisiología , Animales , Comunicación Celular , Diferenciación Celular , Proliferación Celular , Tejido Conectivo/patología , Endotelio Vascular/patología , Matriz Extracelular/fisiología , Fibroblastos/patología , Humanos , Hipoxia/etiología , Hipoxia/patología , Hipoxia/fisiopatología , Enfermedades Pulmonares/etiología , Enfermedades Pulmonares/patología , Enfermedades Pulmonares/fisiopatología , Arteria Pulmonar/patología , Circulación Pulmonar/fisiología , Venas Pulmonares/patología , Células Madre/patología , Células Madre/fisiología
15.
Mol Biol Cell ; 17(4): 1995-2008, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16467381

RESUMEN

We have previously found that hypoxia stimulates proliferation of vascular fibroblasts through Galphai-mediated activation of ERK1/2. Here, we demonstrate that hypoxia also activates the atypical protein kinase Czeta (PKCzeta) isozyme and stimulates the expression of ERK1/2-specific phosphatase, MAP kinase phosphatase-1 (MKP-1), which attenuates ERK1/2-mediated proliferative signals. Replication repressor activity is unique to PKCzeta because the blockade of classical and novel PKC isozymes does not affect fibroblast proliferation. PKCzeta is phosphorylated upon prolonged (24 h) exposure to hypoxia, whereas ERK1/2, the downstream kinases, are maximally activated in fibroblasts exposed to acute (10 min) hypoxia. However, PKCzeta blockade results in persistent ERK1/2 phosphorylation and marked increase in hypoxia-induced replication. Similarly prolonged ERK1/2 phosphorylation and increase in hypoxia-stimulated proliferation are also observed upon blockade of MKP-1 activation. Because of the parallel suppressive actions of PKCzeta and MKP-1 on ERK1/2 phosphorylation and proliferation, the role of PKCzeta in the regulation of MKP-1 expression was evaluated. PKCzeta attenuation reduces MKP-1 expression, whereas PKCzeta overexpression increases MKP-1 levels. In conclusion, our results indicate for the first time that hypoxia activates PKCzeta, which acts as a terminator of ERK1/2 activation through the regulation of downstream target, MKP-1 expression and thus serves to limit hypoxia-induced proliferation of fibroblasts.


Asunto(s)
Vasos Sanguíneos/citología , Proteínas de Ciclo Celular/metabolismo , Hipoxia/enzimología , Proteínas Inmediatas-Precoces/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteína Quinasa C/fisiología , Proteínas Tirosina Fosfatasas/metabolismo , Animales , Vasos Sanguíneos/enzimología , Bovinos , Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/genética , Núcleo Celular/enzimología , Proliferación Celular , Células Cultivadas , Fosfatasa 1 de Especificidad Dual , Activación Enzimática , Fibroblastos/enzimología , Proteínas Inmediatas-Precoces/análisis , Proteínas Inmediatas-Precoces/genética , Fosfoproteínas Fosfatasas/análisis , Fosfoproteínas Fosfatasas/genética , Fosforilación , Proteína Quinasa C/análisis , Proteína Quinasa C/antagonistas & inhibidores , Proteína Fosfatasa 1 , Proteínas Tirosina Fosfatasas/análisis , Proteínas Tirosina Fosfatasas/genética
17.
Am J Physiol Cell Physiol ; 286(2): C416-25, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14561589

RESUMEN

Activation of the alpha-smooth muscle actin (alpha-SMA) gene during the conversion of fibroblasts into myofibroblasts is an essential feature of various fibrotic conditions. Microvascular compromise and thus local environmental hypoxia are important components of the fibrotic response. The present study was thus undertaken to test the hypothesis that hypoxia can induce transdifferentiation of vascular fibroblasts into myofibroblasts and also to evaluate potential signaling mechanisms governing this process. We found that hypoxia significantly upregulates alpha-SMA protein levels in bovine pulmonary artery adventitial fibroblasts. Increased alpha-SMA expression is controlled at the transcriptional level because the alpha-SMA gene promoter activity, assayed via a luciferase reporter, was markedly increased in transfected fibroblasts exposed to hypoxia. Hypoxic induction of the alpha-SMA gene was mimicked by overexpression of constitutively active Galphai2 (alphai2Q205L) but not Galpha16 (alpha-16Q212L). Blockade of hypoxia-induced alpha-SMA expression with pertussis toxin, a Galphai antagonist, confirmed a role for Galphai in the hypoxia-induced transdifferentiation process. c-Jun NH2-terminal kinase (JNK) inhibitor II and SB202190, but not U0126, also attenuated alpha-SMA expression in hypoxic fibroblasts, suggesting the importance of JNK in the differentiation process. Hypoxia-induced increase in bromodeoxyuridine incorporation, which occurred concomitantly with hypoxia-induced differentiation, was blocked by U0126, suggesting that DNA synthesis and alpha-SMA expression take place through simultaneously activated parallel signaling pathways. Neutralizing antibody against transforming growth factor-beta1 blocked only 30% of the hypoxia-induced alpha-SMA promoter activity. Taken together, our results suggest that hypoxia induces differentiation of vascular fibroblasts into myofibroblasts by upregulating the expression of alpha-SMA, and this increase in alpha-SMA level occurs through Galphai- and JNK-dependent signaling pathways.


Asunto(s)
Fibroblastos/patología , Hipoxia/patología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Arteria Pulmonar/patología , Actinas/genética , Actinas/metabolismo , Animales , Animales Recién Nacidos , Anticuerpos/farmacología , Bromodesoxiuridina/metabolismo , Bovinos , Diferenciación Celular , Células Cultivadas , Subunidad alfa de la Proteína de Unión al GTP Gi2 , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Liso/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta1 , Regulación hacia Arriba
18.
Chest ; 122(6 Suppl): 326S-334S, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12475810

RESUMEN

Substantial experimental evidence supports the idea that the fibroblast may play a significant role in the vascular response to injury, especially under hypoxic conditions. Fibroblasts have the ability to rapidly respond to hypoxic stress and to modulate their function to adapt rapidly to local vascular needs. Fibroblasts appear to be uniquely equipped to proliferate, transdifferentiate, and migrate under hypoxic conditions. Proliferative responses to hypoxia depend on the activation of Galpha(i) and Gq kinase family members, and on the subsequent stimulation of protein kinase C and mitogen-activated protein kinase family members. Extracellular nucleotides (eg, adenosine triphosphate [ATP]) are likely to be increased in the hypoxic adventitial compartment and can act as autocrine/paracrine modifiers of the hypoxia-induced proliferative response. The proliferative effects of ATP appear to be mediated largely through G-protein-coupled P2Y receptors in fetal and neonatal fibroblasts. Hypoxia, acting through Galpha(iota)-coupled pathways, also can directly up-regulate alpha-smooth muscle actin expression in fibroblast subpopulations, suggesting that hypoxia may play a direct role in mediating the "transdifferentiation" of fibroblasts into myofibroblasts in the vessel wall. In addition, chronic hypoxia causes stable (at least in vitro) phenotypic changes in fibroblasts that appear to be associated with changes in the signaling pathways used to elicit proliferation. However, it is also becoming clear that, similar to the heterogeneity described for vascular smooth muscle cells, numerous fibroblast subtypes exist in the vessel wall, and that each may respond in unique ways to hypoxia and other stimuli and thus serve special functions in response to injury. In fact, adventitia may be considered to be compartments in which cells with "stem-cell-like" characteristics reside. Future work is needed to determine more precisely the role of the fibroblast in the wide variety of vascular complications observed in many humans diseases, and in the genes and gene products that confer unique properties to this important vascular cell.


Asunto(s)
Hipoxia de la Célula/fisiología , Fibroblastos/metabolismo , Pulmón/fisiopatología , Arteria Pulmonar/fisiopatología , Adulto , Animales , Vasos Sanguíneos/fisiopatología , Bovinos , Diferenciación Celular/fisiología , Niño , Espacio Extracelular , Fibroblastos/clasificación , Fibroblastos/fisiología , Humanos , Lactante , Pulmón/irrigación sanguínea , Sistema de Señalización de MAP Quinasas/fisiología , Nucleótidos/fisiología , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...