Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Circulation ; 142(7): 688-704, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32466671

RESUMEN

BACKGROUND: Pericytes regulate vessel stabilization and function, and their loss is associated with diseases such as diabetic retinopathy or cancer. Despite their physiological importance, pericyte function and molecular regulation during angiogenesis remain poorly understood. METHODS: To decipher the transcriptomic programs of pericytes during angiogenesis, we crossed Pdgfrb(BAC)-CreERT2 mice into RiboTagflox/flox mice. Pericyte morphological changes were assessed in mural cell-specific R26-mTmG reporter mice, in which low doses of tamoxifen allowed labeling of single-cell pericytes at high resolution. To study the role of phosphoinositide 3-kinase (PI3K) signaling in pericyte biology during angiogenesis, we used genetic mouse models that allow selective inactivation of PI3Kα and PI3Kß isoforms and their negative regulator phosphate and tensin homolog deleted on chromosome 10 (PTEN) in mural cells. RESULTS: At the onset of angiogenesis, pericytes exhibit molecular traits of cell proliferation and activated PI3K signaling, whereas during vascular remodeling, pericytes upregulate genes involved in mature pericyte cell function, together with a remarkable decrease in PI3K signaling. Immature pericytes showed stellate shape and high proliferation, and mature pericytes were quiescent and elongated. Unexpectedly, we demonstrate that PI3Kß, but not PI3Kα, regulates pericyte proliferation and maturation during vessel formation. Genetic PI3Kß inactivation in pericytes triggered early pericyte maturation. Conversely, unleashing PI3K signaling by means of PTEN deletion delayed pericyte maturation. Pericyte maturation was necessary to undergo vessel remodeling during angiogenesis. CONCLUSIONS: Our results identify new molecular and morphological traits associated with pericyte maturation and uncover PI3Kß activity as a checkpoint to ensure appropriate vessel formation. In turn, our results may open new therapeutic opportunities to regulate angiogenesis in pathological processes through the manipulation of pericyte PI3Kß activity.


Asunto(s)
Neovascularización Fisiológica , Pericitos/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Remodelación Vascular , Animales , Ratones , Ratones Transgénicos , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/genética
2.
Angiogenesis ; 24(3): 471-482, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33730293

RESUMEN

Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that is overexpressed in many cancer types and in vivo studies have shown that vascular endothelial cell FAK expression and FAK-phosphorylation at tyrosine (Y) 397, and subsequently FAK-Y861, are important in tumour angiogenesis. Pericytes also play a vital role in regulating tumour blood vessel stabilisation, but the specific involvement of pericyte FAK-Y397 and FAK-Y861 phosphorylation in tumour blood vessels is unknown. Using PdgfrßCre + ;FAKWT/WT, PdgfrßCre + ;FAKY397F/Y397F and PdgfrßCre + ;FAKY861F/Y861F mice, our data demonstrate that Lewis lung carcinoma tumour growth, tumour blood vessel density, blood vessel perfusion and pericyte coverage were affected only in late stage tumours in PdgfrßCre + ;FAKY861F/Y861F but not PdgfrßCre + ;FAKY397F/Y397F mice. Further examination indicates a dual role for pericyte FAK-Y861 phosphorylation in the regulation of tumour vessel regression and also in the control of pericyte derived signals that influence apoptosis in cancer cells. Overall this study identifies the role of pericyte FAK-Y861 in the regulation of tumour vessel regression and tumour growth control and that non-phosphorylatable FAK-Y861F in pericytes reduces tumour growth and blood vessel density.


Asunto(s)
Apoptosis , Carcinoma Pulmonar de Lewis , Quinasa 1 de Adhesión Focal , Mutación Missense , Proteínas de Neoplasias , Neovascularización Patológica , Pericitos/enzimología , Sustitución de Aminoácidos , Animales , Carcinoma Pulmonar de Lewis/enzimología , Carcinoma Pulmonar de Lewis/genética , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica/enzimología , Neovascularización Patológica/genética , Fosforilación
3.
Clin Sci (Lond) ; 135(24): 2667-2689, 2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34807265

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes a broad range of clinical responses including prominent microvascular damage. The capacity of SARS-CoV-2 to infect vascular cells is still debated. Additionally, the SARS-CoV-2 Spike (S) protein may act as a ligand to induce non-infective cellular stress. We tested this hypothesis in pericytes (PCs), which are reportedly reduced in the heart of patients with severe coronavirus disease-2019 (COVID-19). Here we newly show that the in vitro exposure of primary human cardiac PCs to the SARS-CoV-2 wildtype strain or the α and δ variants caused rare infection events. Exposure to the recombinant S protein alone elicited signalling and functional alterations, including: (1) increased migration, (2) reduced ability to support endothelial cell (EC) network formation on Matrigel, (3) secretion of pro-inflammatory molecules typically involved in the cytokine storm, and (4) production of pro-apoptotic factors causing EC death. Next, adopting a blocking strategy against the S protein receptors angiotensin-converting enzyme 2 (ACE2) and CD147, we discovered that the S protein stimulates the phosphorylation/activation of the extracellular signal-regulated kinase 1/2 (ERK1/2) through the CD147 receptor, but not ACE2, in PCs. The neutralisation of CD147, either using a blocking antibody or mRNA silencing, reduced ERK1/2 activation, and rescued PC function in the presence of the S protein. Immunoreactive S protein was detected in the peripheral blood of infected patients. In conclusion, our findings suggest that the S protein may prompt PC dysfunction, potentially contributing to microvascular injury. This mechanism may have clinical and therapeutic implications.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Basigina/metabolismo , Miocardio/enzimología , Pericitos/enzimología , SARS-CoV-2/fisiología , Glicoproteína de la Espiga del Coronavirus/sangre , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , COVID-19/sangre , Células CACO-2 , Muerte Celular , Niño , Preescolar , Citocinas/metabolismo , Femenino , Interacciones Huésped-Patógeno , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Miocardio/citología , Pericitos/virología , Cultivo Primario de Células , Adulto Joven
4.
Am J Physiol Heart Circ Physiol ; 319(5): H1059-H1068, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-33036546

RESUMEN

The prevalence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) quickly reached pandemic proportions, and knowledge about this virus and coronavirus disease 2019 (COVID-19) has expanded rapidly. This review focuses primarily on mechanisms that contribute to acute cardiac injury and dysfunction, which are common in patients with severe disease. The etiology of cardiac injury is multifactorial, and the extent is likely enhanced by preexisting cardiovascular disease. Disruption of homeostatic mechanisms secondary to pulmonary pathology ranks high on the list, and there is growing evidence that direct infection of cardiac cells can occur. Angiotensin-converting enzyme 2 (ACE2) plays a central role in COVID-19 and is a necessary receptor for viral entry into human cells. ACE2 normally not only eliminates angiotensin II (Ang II) by converting it to Ang-(1-7) but also elicits a beneficial response profile counteracting that of Ang II. Molecular analyses of single nuclei from human hearts have shown that ACE2 is most highly expressed by pericytes. Given the important roles that pericytes have in the microvasculature, infection of these cells could compromise myocardial supply to meet metabolic demand. Furthermore, ACE2 activity is crucial for opposing adverse effects of locally generated Ang II, so virus-mediated internalization of ACE2 could exacerbate pathology by this mechanism. While the role of cardiac pericytes in acute heart injury by SARS-CoV-2 requires investigation, expression of ACE2 by these cells has broader implications for cardiac pathophysiology.


Asunto(s)
Betacoronavirus/patogenicidad , Infecciones por Coronavirus/enzimología , Cardiopatías/enzimología , Peptidil-Dipeptidasa A/metabolismo , Pericitos/enzimología , Neumonía Viral/enzimología , Internalización del Virus , Enzima Convertidora de Angiotensina 2 , Animales , COVID-19 , Infecciones por Coronavirus/virología , Cardiopatías/fisiopatología , Cardiopatías/virología , Interacciones Huésped-Patógeno , Humanos , Pandemias , Pericitos/virología , Neumonía Viral/virología , SARS-CoV-2
5.
FASEB J ; 32(5): 2539-2548, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29263022

RESUMEN

Inflammation plays an important role in the pathogenesis of diabetic retinopathy. We have previously demonstrated the effect of cathepsin D (CD) on the mechanical disruption of retinal endothelial cell junctions and increased vasopermeability, as well as increased levels of CD in retinas of diabetic mice. Here, we have also examined the effect of CD on endothelial-pericyte interactions, as well as the effect of dipeptidyl peptidase-4 (DPP4) inhibitor on CD in endothelial-pericyte interactions in vitro and in vivo. Cocultured cells that were treated with pro-CD demonstrated a significant decrease in the expression of platelet-derived growth factor receptor-ß, a tyrosine kinase receptor that is required for pericyte cell survival; N-cadherin, the key adherens junction protein between endothelium and pericytes; and increases in the vessel destabilizing agent, angiopoietin-2. The effect was reversed in cells that were treated with DPP4 inhibitor along with pro-CD. With pro-CD treatment, there was a significant increase in the phosphorylation of the downstream signaling protein, PKC-α, and Ca2+/calmodulin-dependent protein kinase II in endothelial cells and pericytes, which disrupts adherens junction structure and function, and this was significantly reduced with DPP4 inhibitor treatment. Increased CD levels, vasopermeability, and alteration in junctional-related proteins were observed in the retinas of diabetic rats, which were significantly changed with DPP4 inhibitor treatment. Thus, DPP4 inhibitors may be used as potential adjuvant therapeutic agents to treat increased vascular leakage observed in patients with diabetic macular edema.-Monickaraj, F., McGuire, P., Das, A. Cathepsin D plays a role in endothelial-pericyte interactions during alteration of the blood-retinal barrier in diabetic retinopathy.


Asunto(s)
Barrera Hematorretinal/enzimología , Catepsina D/metabolismo , Comunicación Celular , Retinopatía Diabética/enzimología , Células Endoteliales/enzimología , Pericitos/enzimología , Angiopoyetina 2/metabolismo , Animales , Barrera Hematorretinal/patología , Cadherinas/metabolismo , Señalización del Calcio/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Catepsina D/antagonistas & inhibidores , Supervivencia Celular/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Retinopatía Diabética/patología , Células Endoteliales/patología , Masculino , Proteínas del Tejido Nervioso/metabolismo , Pericitos/patología , Ratas , Ratas Sprague-Dawley , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo
6.
J Neurosci ; 37(1): 129-140, 2017 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-28053036

RESUMEN

Blood-brain barrier disruption (BBB) and release of toxic blood molecules into the brain contributes to neuronal injury during stroke and other cerebrovascular diseases. While pericytes are builders and custodians of the BBB in the normal brain, their impact on BBB integrity during ischemia remains unclear. We imaged pericyte-labeled transgenic mice with in vivo two-photon microscopy to examine the relationship between pericytes and blood plasma leakage during photothrombotic occlusion of cortical capillaries. Upon cessation of capillary flow, we observed that plasma leakage occurred with three times greater frequency in regions where pericyte somata adjoined the endothelium. Pericyte somata covered only 7% of the total capillary length in cortex, indicating that a disproportionate amount of leakage occurred from a small fraction of the capillary bed. Plasma leakage was preceded by rapid activation of matrix metalloproteinase (MMP) at pericyte somata, which was visualized at high resolution in vivo using a fluorescent probe for matrix metalloproteinase-2/9 activity, fluorescein isothiocyanate (FITC)-gelatin. Coinjection of an MMP-9 inhibitor, but not an MMP-2 inhibitor, reduced pericyte-associated FITC-gelatin fluorescence and plasma leakage. These results suggest that pericytes contribute to rapid and localized proteolytic degradation of the BBB during cerebral ischemia. SIGNIFICANCE STATEMENT: Pericytes are a key component of the neurovascular unit and are essential for normal BBB function. However, during acute ischemia, we find that pericytes are involved in creating rapid and heterogeneous BBB disruption in the capillary bed. The mechanism by which pericytes contribute to BBB damage warrants further investigation, as it may yield new therapeutic targets for acute stroke injury and other neurological diseases involving capillary flow impairment.


Asunto(s)
Isquemia Encefálica/fisiopatología , Capilares/fisiopatología , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Pericitos/metabolismo , Animales , Barrera Hematoencefálica/fisiología , Isquemia Encefálica/enzimología , Isquemia Encefálica/metabolismo , Capilares/enzimología , Corteza Cerebral/fisiopatología , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Pericitos/enzimología , Inhibidores de Proteasas/farmacología , Accidente Cerebrovascular/enzimología , Accidente Cerebrovascular/fisiopatología
7.
Biochim Biophys Acta Mol Basis Dis ; 1863(4): 929-935, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28131914

RESUMEN

Events responsible for cerebrovascular disease in diabetes are not fully understood. Pericyte loss is an early event that leads to endothelial cell death, microaneurysms, and cognitive impairment. A biochemical mechanism underlying pericyte loss is rapid respiration (oxidative metabolism of glucose). This escalation in respiration results from free influx of glucose into insulin-insensitive tissues in the face of high glucose levels in the blood. Rapid respiration generates superoxide, the precursor to all reactive oxygen species (ROS), and results in pericyte death. Respiration is regulated by carbonic anhydrases (CAs) VA and VB, the two isozymes expressed in mitochondria, and their pharmacologic inhibition with topiramate reduces respiration, ROS, and pericyte death. Topiramate inhibits both isozymes; therefore, in the earlier studies, their individual roles were not discerned. In a recent genetic study, we showed that mitochondrial CA VA plays a significant role in regulation of reactive oxygen species and pericyte death. The role of CA VB was not addressed. In this report, genetic knockdown and overexpression studies confirm that mitochondrial CA VA regulates respiration in pericytes, whereas mitochondrial CA VB does not contribute significantly. Identification of mitochondrial CA VA as a sole regulator of respiration provides a specific target to develop new drugs with fewer side effects that may be better tolerated and can protect the brain from diabetic injury. Since similar events occur in the capillary beds of other insulin-insensitive tissues such as the eye and kidney, these drugs may also slow the onset and progression of diabetic disease in these tissues.


Asunto(s)
Apoptosis , Encéfalo/enzimología , Anhidrasa Carbónica V/metabolismo , Trastornos Cerebrovasculares/enzimología , Angiopatías Diabéticas/prevención & control , Mitocondrias/enzimología , Proteínas Mitocondriales/metabolismo , Pericitos/enzimología , Animales , Encéfalo/patología , Anhidrasa Carbónica V/genética , Línea Celular Transformada , Trastornos Cerebrovasculares/genética , Trastornos Cerebrovasculares/patología , Angiopatías Diabéticas/enzimología , Angiopatías Diabéticas/genética , Angiopatías Diabéticas/patología , Ratones , Mitocondrias/patología , Proteínas Mitocondriales/genética , Pericitos/patología
8.
J Vasc Res ; 51(6): 429-38, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25612841

RESUMEN

BACKGROUND: Pericytes are multifunctional cells surrounding capillaries and postcapillary venules. In brain microvasculature, pericytes play a pivotal role under physiological and pathological conditions by producing reactive oxygen species (ROS). The aims of this study were to elucidate the source of ROS and its regulation in human brain pericytes. METHODS: The expression of Nox enzymes in the cells was evaluated using RT-PCR and western blot. Superoxide production was determined by superoxide dismutase-inhibitable chemiluminescence. Silencing of Nox4 was performed using RNAi, and cell proliferation was evaluated using the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay. RESULTS: Nox4 was predominant among the Nox family in human brain pericytes. Membrane fractions of cells produced superoxide in the presence of NAD(P)H. Superoxide production was almost abolished with diphenileneiodonium, a Nox inhibitor; however, inhibitors of other possible superoxide-producing enzymes had no effect on NAD(P)H-dependent superoxide production. Pericytes expressed angiotensin II (Ang II) receptors, and Ang II upregulated Nox4 expression. Hypoxic conditions also increased the Nox4 expression. Silencing of Nox4 significantly reduced ROS production and attenuated cell proliferation. CONCLUSION: Our study showed that Nox4 is a major superoxide-producing enzyme and that its expression is regulated by Ang II and hypoxic stress in human brain pericytes. In addition, Nox4 may promote cell growth.


Asunto(s)
Encéfalo/irrigación sanguínea , NADPH Oxidasas/metabolismo , Pericitos/enzimología , Superóxidos/metabolismo , Angiotensina II/metabolismo , Animales , Hipoxia de la Célula , Membrana Celular/enzimología , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Humanos , Infarto de la Arteria Cerebral Media/enzimología , Masculino , Ratones , Microvasos/enzimología , NADPH Oxidasa 4 , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , Pericitos/efectos de los fármacos , Interferencia de ARN , Receptores de Angiotensina/metabolismo , Factores de Tiempo , Transfección
9.
Arterioscler Thromb Vasc Biol ; 33(2): 305-10, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23241407

RESUMEN

OBJECTIVE: Vessels in brain arteriovenous malformations are prone to rupture. The underlying pathogenesis is not clear. Hereditary hemorrhagic telangiectasia type 2 patients with activin receptor-like kinase 1 (Alk1) mutation have a higher incidence of brain arteriovenous malformation than the general population. We tested the hypothesis that vascular endothelial growth factor impairs vascular integrity in the Alk1-deficient brain through reduction of mural cell coverage. METHODS AND RESULTS: Adult Alk1(1f/2f) mice (loxP sites flanking exons 4-6) and wild-type mice were injected with 2×10(7) PFU adenovious-cre recombinase and 2×10(9) genome copies of adeno-associated virus-vascular endothelial growth factor to induce focal homozygous Alk1 deletion (in Alk1(1f/2f) mice) and angiogenesis. Brain vessels were analyzed 8 weeks later. Compared with wild-type mice, the Alk1-deficient brain had more fibrin (99±30×10(3) pixels/mm(2) versus 40±13×10(3); P=0.001), iron deposition (508±506 pixels/mm(2) versus 6±49; P=0.04), and Iba1(+) microglia/macrophage infiltration (888±420 Iba1(+) cells/mm(2) versus 240±104 Iba1(+); P=0.001) after vascular endothelial growth factor stimulation. In the angiogenic foci, the Alk1-deficient brain had more α-smooth muscle actin negative vessels (52±9% versus 12±7%, P<0.001), fewer vascular-associated pericytes (503±179/mm(2) versus 931±115, P<0.001), and reduced platelet-derived growth factor receptor-ß expression. CONCLUSIONS: Reduction of mural cell coverage in response to vascular endothelial growth factor stimulation is a potential mechanism for the impairment of vessel wall integrity in hereditary hemorrhagic telangiectasia type 2-associated brain arteriovenous malformation.


Asunto(s)
Receptores de Activinas Tipo I/deficiencia , Vasos Sanguíneos/enzimología , Encéfalo/irrigación sanguínea , Neovascularización Patológica , Pericitos/enzimología , Telangiectasia Hemorrágica Hereditaria/enzimología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Actinas/metabolismo , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo II , Animales , Becaplermina , Vasos Sanguíneos/patología , Dependovirus/genética , Modelos Animales de Enfermedad , Fibrina/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos , Hierro/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Microglía/patología , Pericitos/patología , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Telangiectasia Hemorrágica Hereditaria/genética , Telangiectasia Hemorrágica Hereditaria/patología , Factor A de Crecimiento Endotelial Vascular/genética
10.
Lipids Health Dis ; 13: 156, 2014 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-25292258

RESUMEN

BACKGROUND: Diabetic retinopathy, the main microvascular complications of diabetes and one of the leading causes of blindness worldwide. Interesting reports on the role of inflammatory/proangiogenic high mobility group 1 (HMGB-1) cytokine and phospholipases A2 (PLA2) in neovascularization have diverted our concentration to reveal whether HMGB-1 and PLA2 plays role in diabetic retinopathy. METHODS: We performed our study in streptozotocin (STZ)-induced diabetic rat model. The expression levels of the cytokines, chemokines, and cell adhesion molecules in retinal tissues were evaluated by quantitative RT-PCR. HMGB-1 and PLA2 protein levels along with VEGF, TNF-α, IL-1ß and ICAM-1 levels were also measured. RESULTS: We observed the retinal pericytes, endothelial injury/death and breakdown of blood-retinal barrier (BRB). The protein expression of HMGB-1, PLA2 and IL-1ß were significantly increased in micro vessels from retina of diabetic rats. Diabetic rats had also high retinal levels of VEGF, ICAM-1 and TNF-α. Further investigation revealed that pericyte death is mediated by HMGB-1-induced cytotoxic activity of glial cells, while HMGB-1 can directly mediate endothelial cell death. Similarly, increased expression of PLA2 represents the diabetic mediated alteration of BRB, perhaps up regulating the VEGF. CONCLUSIONS: Our data suggest that HMGB-1 and PLA2 involved in retinal pericyte and endothelial injury and cell death in diabetic retinopathy. From this study, we suggest that HMGB-1 and PLA2 may be interesting targets in managing diabetic retinopathy.


Asunto(s)
Diabetes Mellitus Experimental/enzimología , Retinopatía Diabética/enzimología , Proteína HMGB1/metabolismo , Fosfolipasas A2/metabolismo , Animales , Apoptosis , Quimiocinas/genética , Quimiocinas/metabolismo , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Diabetes Mellitus Experimental/complicaciones , Expresión Génica , Proteína HMGB1/genética , Masculino , Proteínas de la Membrana/metabolismo , Pericitos/enzimología , Fosfolipasas A2/genética , Ratas Sprague-Dawley , Células Ganglionares de la Retina/fisiología , Vasos Retinianos/enzimología , Vasos Retinianos/patología
11.
J Pharmacol Exp Ther ; 344(3): 637-45, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23249625

RESUMEN

Diabetes-associated complications in the microvasculature of the brain are caused by oxidative stress, generated by overproduction of reactive oxygen species from hyperglycemia-induced accelerated oxidative metabolism of glucose. Pericytes, essential for the viability of the microvasculature, are especially susceptible to oxidative stress. Mitochondrial carbonic anhydrases, regulators of the oxidative metabolism of glucose, determine the rate of reactive oxygen species production and inhibition of mitochondrial carbonic anhydrases rescues glucose-induced pericyte loss in the diabetic mouse brain. We hypothesized that high glucose induces intracellular oxidative stress and pericyte apoptosis and that inhibition of mitochondrial carbonic anhydrases protects pericytes from oxidative stress-induced apoptosis. To validate our hypothesis, conditionally immortalized cerebral pericyte (IPC) cultures were established from Immortomice to investigate the effect of high glucose on oxidative stress and pericyte apoptosis. The IPCs expressed pericyte markers and induced high transendothelial electrical resistance and low permeability in brain endothelial cell monolayers comparable with pericytes in primary cultures. The IPCs also secreted cytokines constitutively and in response to lipopolysaccharide similar to pericytes. High glucose caused oxidative stress and apoptosis of these cells, with both oxidative stress and apoptosis significantly reduced after mitochondrial carbonic anhydrase inhibition. These results provide the first evidence that pharmacological inhibition of mitochondrial carbonic anhydrases attenuates pericyte apoptosis caused by high glucose-induced oxidative stress. Carbonic anhydrase inhibitors have a long history of safe clinical use and can be immediately evaluated for this new indication in translational research. Thus, mitochondrial carbonic anhydrases may provide a new therapeutic target for oxidative stress-related illnesses of the brain.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores de Anhidrasa Carbónica/farmacología , Glucosa/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Estrés Oxidativo/efectos de los fármacos , Pericitos/efectos de los fármacos , Animales , Anhidrasas Carbónicas/metabolismo , Células Cultivadas , Cerebro/efectos de los fármacos , Cerebro/enzimología , Cerebro/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Neuroglía/efectos de los fármacos , Neuroglía/enzimología , Pericitos/enzimología , Pericitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo
12.
Circ Res ; 106(5): 932-40, 2010 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-20110534

RESUMEN

RATIONALE: Pathological neovascularization is a critical component of diseases such as proliferative retinopathies, cancer and rheumatoid arthritis, yet much remains to be learned about the underlying causes. Previous studies showed that vascular endothelial growth factor (VEGF)-A activates the membrane-anchored metalloproteinase ADAM17 (a disintegrin and metalloproteinase 17) in endothelial cells, thereby stimulating crosstalk between VEGF receptor 2 and extracellular signal-regulated kinase. These findings raised interesting questions about the role of ADAM17 in angiogenesis and neovascularization in vivo. OBJECTIVE: The objective of this study was to inactivate ADAM17 in endothelial cells or in pericytes to determine how this affects developmental angiogenesis, pathological retinal neovascularization and heterotopic tumor growth. METHODS AND RESULTS: We generated animals in which floxed ADAM17 was removed by Tie2-Cre in endothelial cells, or by smooth muscle (sm) Cre in smooth muscle cells and pericytes. There were no evident developmental defects in either conditional knockout strain, but pathological retinal neovascularization and growth of heterotopically injected tumor cells was reduced in Adam17flox/flox/Tie2-Cre mice, although not in Adam17flox/flox/sm-Cre mice. Moreover, lack of ADAM17 in endothelial cells decreased ex vivo chord formation, and this could be largely restored by addition of the ADAM17 substrate HB-EGF (heparin-binding epidermal growth factor-like growth factor). Finally we found that ADAM17 is important for the VEGF receptor 2 stimulated processing of several receptors with known functions in endothelial cell biology. CONCLUSIONS: These results provide the first evidence for a role for ADAM17 in pathological neovascularization in vivo. Because ADAM17 does not appear to be required for normal developmental angiogenesis or vascular homeostasis, it could emerge as a good target for treatment of pathological neovascularization.


Asunto(s)
Proteínas ADAM/deficiencia , Células Endoteliales/enzimología , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/prevención & control , Neovascularización Patológica/prevención & control , Pericitos/enzimología , Neovascularización Retiniana/prevención & control , Proteínas ADAM/genética , Proteína ADAM17 , Actinas/metabolismo , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Factor de Crecimiento Similar a EGF de Unión a Heparina , Integrasas/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/enzimología , Neovascularización Patológica/genética , Regiones Promotoras Genéticas , Proteínas Tirosina Quinasas Receptoras/genética , Receptor TIE-2 , Neovascularización Retiniana/enzimología , Neovascularización Retiniana/genética , Porcinos , Factores de Tiempo , Carga Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
13.
Invest Ophthalmol Vis Sci ; 62(7): 6, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34086044

RESUMEN

Purpose: To investigate the expression of angiotensin-converting enzyme 2 (ACE2), the receptor for SARS-CoV-2 in human retina. Methods: Human post-mortem eyes from 13 non-diabetic control cases and 11 diabetic retinopathy cases were analyzed for the expression of ACE2. To compare the vascular ACE2 expression between different organs that involve in diabetes, the expression of ACE2 was investigated in renal specimens from nondiabetic and diabetic nephropathy patients. Expression of TMPRSS2, a cell-surface protease that facilitates SARS-CoV-2 entry, was also investigated in human nondiabetic retinas. Primary human retinal endothelial cells (HRECs) and primary human retinal pericytes (HRPCs) were further used to confirm the vascular ACE2 expression in human retina. Results: We found that ACE2 was expressed in multiple nonvascular neuroretinal cells, including the retinal ganglion cell layer, inner plexiform layer, inner nuclear layer, and photoreceptor outer segments in both nondiabetic and diabetic retinopathy specimens. Strikingly, we observed significantly more ACE2 positive vessels in the diabetic retinopathy specimens. By contrast, in another end-stage organ affected by diabetes, the kidney, ACE2 in nondiabetic and diabetic nephropathy showed apical expression of ACE2 tubular epithelial cells, but no endothelial expression in glomerular or peritubular capillaries. Western blot analysis of protein lysates from HRECs and HRPCs confirmed expression of ACE2. TMPRSS2 expression was present in multiple retinal neuronal cells, vascular and perivascular cells, and Müller glia. Conclusions: Together, these results indicate that retina expresses ACE2 and TMPRSS2. Moreover, there are increased vascular ACE2 expression in diabetic retinopathy retinas.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Retinopatía Diabética/enzimología , Receptores Virales/metabolismo , Retina/enzimología , SARS-CoV-2/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Sitios de Unión , Western Blotting , Células Cultivadas , Nefropatías Diabéticas/enzimología , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/virología , Retinopatía Diabética/patología , Retinopatía Diabética/virología , Endotelio Vascular/enzimología , Endotelio Vascular/virología , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Pericitos/enzimología , Pericitos/virología , Vasos Retinianos/enzimología , Vasos Retinianos/patología , Vasos Retinianos/virología , Serina Endopeptidasas/metabolismo
14.
Am J Physiol Regul Integr Comp Physiol ; 299(5): R1317-25, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20686172

RESUMEN

We used the whole cell patch-clamp technique to investigate the regulation of descending vasa recta (DVR) pericyte Ca(2+)-dependent Cl(-) currents (CaCC) by cytoplasmic Ca(2+) concentration ([Ca](CYT)), voltage, and kinase activity. Murine CaCC increased with voltage and electrode Ca(2+) concentration. The current saturated at [Ca](CYT) of ∼1,000 nM and exhibited an EC(50) for Ca(2+) of ∼500 nM, independent of depolarization potential. Activation time constants were between 100 and 200 ms, independent of electrode Ca(2+). Repolarization-related tail currents elicited by stepping from +100 mV to varying test potentials exhibited deactivation time constants of 50-200 ms that increased with voltage when electrode [Ca](CYT) was 1,000 nM. The calmodulin inhibitor N-(6-aminohexyl)-5-chloro-1-naphthalenesulfonamide hydrochloride (W-7, 30 µM) blocked CaCC. The myosin light chain kinase blockers 1-(5-iodonaphthalene-1-sulfonyl)-1H-hexahydro-1,4-diazepine hydrochloride (ML-7, 1-50 µM) and 1-(5-chloronaphthalene-1-sulfonyl)-1H-hexahydro-1,4-diazepine hydrochloride (ML-9, 10 µM) were similarly effective. Resting pericytes were hyperpolarized by ML-7. Pericytes exposed to ANG II (10 nM) depolarized from a baseline of -50 ± 6 to -29 ± 3 mV and were repolarized to -63 ± 7 mV by exposure to 50 µM ML-7. The Ca(2+)/calmodulin-dependent kinase inhibitor KN-93 reduced pericyte CaCC only when it was present in the electrode and extracellular buffer from the time of membrane break-in. We conclude that murine DVR pericytes are modulated by [Ca](CYT), membrane potential, and phosphorylation events, suggesting that Ca(2+)-dependent Cl(-) conductance may be a target for regulation of vasoactivity and medullary blood flow in vivo.


Asunto(s)
Calcio/metabolismo , Cloruros/metabolismo , Aparato Yuxtaglomerular/irrigación sanguínea , Microvasos/enzimología , Pericitos/enzimología , Proteínas Quinasas/metabolismo , Angiotensina II/metabolismo , Animales , Azepinas/farmacología , Bencilaminas/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Calmodulina/antagonistas & inhibidores , Calmodulina/metabolismo , Transporte Iónico , Potenciales de la Membrana , Ratones , Ratones Endogámicos C57BL , Microvasos/efectos de los fármacos , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/metabolismo , Naftalenos/farmacología , Técnicas de Placa-Clamp , Pericitos/efectos de los fármacos , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Sulfonamidas/farmacología , Factores de Tiempo
15.
Mol Vis ; 16: 408-15, 2010 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-20300563

RESUMEN

PURPOSE: An early and significant event in diabetic retinopathy is the loss of retinal microvascular pericytes. Studies were performed to investigate pathways through which an advanced glycation endproduct and tumor necrosis factor (TNF)-alpha stimulate apoptosis in retinal pericytes through the activation of the pro-apoptotic transcription factor Forkhead box O1 (FOXO1). METHODS: Human retinal pericytes were stimulated by carboxymethyllysine (CML)-collagen, an advanced glycation endproduct, or TNF-alpha in vitro. Apoptosis was assessed by measuring cytoplasmic histone-associated DNA. The role of FOXO1 was examined by RNA interference (RNAi), and specific inhibitors were used to investigate the role of p38 and Jun N-terminal kinase mitogen-activated protein kinase (JNK MAP) kinases, Akt, and nuclear factor kappa B (NF-kappaB). Caspase-3 activity was measured with a luminescent substrate, and FOXO1 DNA-binding activity was measured by electrophoretic mobility shift assay (EMSA). RESULTS: TNF-alpha and CML-collagen but not control collagen stimulated apoptosis, caspase-3 activity, and FOXO1 DNA-binding activity in pericytes. Silencing FOXO1 by small interfering RNA prevented apoptosis of pericytes in response to both TNF-alpha and CML-collagen. By use of specific inhibitors, we demonstrated that both FOXO1 activation and subsequent apoptosis was mediated, in part, by p38 and JNK MAP kinases. In contrast Akt and NF-kappaB inhibitors had the opposite effect on pericyte apoptosis. CONCLUSIONS: The results demonstrate pathways through which two different mediators, TNF-alpha and an advanced glycation endproduct, can induce pericyte apoptosis through activation of the transcription factor FOXO1.


Asunto(s)
Apoptosis , Factores de Transcripción Forkhead/metabolismo , Pericitos/citología , Pericitos/metabolismo , Retina/citología , Animales , Apoptosis/efectos de los fármacos , Bovinos , Productos Finales de Glicación Avanzada/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Pericitos/efectos de los fármacos , Pericitos/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
BMC Cancer ; 10: 288, 2010 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-20546609

RESUMEN

BACKGROUND: Decreased expression of the angiogenesis inhibitor ADAMTS1 (ADAM metallopeptidase with thrombospondin type 1 motif, 1) has previously been reported during prostate cancer progression. The aim of this study was to investigate the function of ADAMTS1 in prostate tumors. METHODS: ADAMTS1 was downregulated by shRNA technology in the human prostate cancer cell line LNCaP (androgen-dependent), originally expressing ADAMTS1, and was upregulated by transfection in its subline LNCaP-19 (androgen-independent), expressing low levels of ADAMTS1. Cells were implanted subcutaneously in nude mice and tumor growth, microvessel density (MVD), blood vessel morphology, pericyte coverage and thrombospondin 1 (TSP1) were studied in the tumor xenografts. RESULTS: Modified expression of ADAMTS1 resulted in altered blood vessel morphology in the tumors. Low expression levels of ADAMTS1 were associated with small diameter blood vessels both in LNCaP and LNCaP-19 tumors, while high levels of ADAMTS1 were associated with larger vessels. In addition, TSP1 levels in the tumor xenografts were inversely related to ADAMTS1 expression. MVD and pericyte coverage were not affected. Moreover, upregulation of ADAMTS1 inhibited tumor growth of LNCaP-19, as evidenced by delayed tumor establishment. In contrast, downregulation of ADAMTS1 in LNCaP resulted in reduced tumor growth rate. CONCLUSIONS: The present study demonstrates that ADAMTS1 is an important regulatory factor of angiogenesis and tumor growth in prostate tumors, where modified ADAMTS1 expression resulted in markedly changed blood vessel morphology, possibly related to altered TSP1 levels.


Asunto(s)
Proteínas ADAM/metabolismo , Vasos Sanguíneos/enzimología , Neovascularización Patológica/enzimología , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/enzimología , Trombospondina 1/metabolismo , Proteínas ADAM/genética , Proteína ADAMTS1 , Animales , Vasos Sanguíneos/patología , Línea Celular Tumoral , Proliferación Celular , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Neovascularización Patológica/genética , Pericitos/enzimología , Neoplasias de la Próstata/genética , Interferencia de ARN , Factores de Tiempo , Transfección , Carga Tumoral
17.
Cardiovasc Res ; 116(6): 1097-1100, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32227090

RESUMEN

A new type of pneumonia caused by a novel coronavirus SARS-CoV-2 outbreaks recently in China and spreads into many other countries. This disease, named as COVID-19, is similar to patients infected by SARS-CoV and MERS-CoV, and nearly 20% of patients developed severe condition. Cardiac injury is a prevalent complication of severe patients, exacerbating the disease severity in coronavirus disease 2019 (COVID-19) patients. Angiotensin-converting enzyme 2 (ACE2), the key host cellular receptor of SARS-CoV-2, has been identified in multiple organs, but its cellular distribution in human heart is not illuminated clearly. This study performed the first state-of-art single cell atlas of adult human heart, and revealed that pericytes with high expression of ACE2 might act as the target cardiac cell of SARS-CoV-2. The pericytes injury due to virus infection may result in capillary endothelial cells dysfunction, inducing microvascular dysfunction. And patients with basic heart failure disease showed increased ACE2 expression at both mRNA and protein levels, meaning that if infected by the virus these patients may have higher risk of heart attack and critically ill condition. The finding of this study explains the high rate of severe cases among COVID-19 patients with basic cardiovascular disease; and these results also perhaps provide important reference to clinical treatment of cardiac injury among severe patients infected by SARS-CoV-2.


Asunto(s)
Betacoronavirus/fisiología , Infecciones por Coronavirus/complicaciones , Cardiopatías/etiología , Miocardio/enzimología , Peptidil-Dipeptidasa A/metabolismo , Pericitos/enzimología , Neumonía Viral/complicaciones , Enzima Convertidora de Angiotensina 2 , Betacoronavirus/metabolismo , COVID-19 , Infecciones por Coronavirus/virología , Expresión Génica , Perfilación de la Expresión Génica , Corazón/virología , Humanos , Pandemias , Peptidil-Dipeptidasa A/genética , Pericitos/virología , Neumonía Viral/virología , Receptores de Coronavirus , Receptores Virales/genética , Receptores Virales/metabolismo , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/metabolismo
18.
J Am Heart Assoc ; 9(13): e016047, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32567476

RESUMEN

Background Pro-NTs (precursor of neurotrophins) and their receptor p75 are potential targets for preventing microvascular dysfunction induced by myocardial ischemia-reperfusion injury (IRI). p75ECD (ectodomain of neurotrophin receptor p75) may physiologically produce neurocytoprotective effects by scavenging pro-NTs. We therefore hypothesized that p75ECD may have a cardioprotective effect on IRI through microvascular mechanisms. Methods and Results Myocardial IRI was induced in Sprague-Dawley rats by occluding the left main coronary arteries for 45 minutes before a subsequent relaxation. Compared with the ischemia-reperfusion group, an intravenous injection of p75ECD (3 mg/kg) 5 minutes before reperfusion reduced the myocardial infarct area at 24 hours after reperfusion (by triphenyltetrazolium chloride, 44.9±3.9% versus 34.6±5.7%, P<0.05); improved the left ventricular ejection fraction (by echocardiography), with less myocardial fibrosis (by Masson's staining), and prevented microvascular dysfunction (by immunofluorescence) at 28 days after reperfusion; and reduced myocardial pro-NTs expression at 24 hours and 28 days after reperfusion (by Western blotting). A simulative IRI model using rat microvascular pericytes was established in vitro by hypoxia-reoxygenation (2/6 hours) combined with pro-NTs treatment (3 nmol/L) at R. p75ECD (3 µg/mL) given at R improved pericyte survival (by methyl thiazolyl tetrazolium assay) and attenuated apoptosis (by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labeling). In the reperfused hearts and hypoxia-reoxygenation +pro-NTs-injured pericytes, p75ECD inhibited the expression of p-JNK (phospho of c-Jun N-terminal kinase)/caspase-3 (by Western blotting). SP600125, an inhibitor of JNK, did not enhance the p75ECD-induced infarct-sparing effects and pericyte protection. Conclusions p75ECD may attenuate myocardial IRI via pro-NTs reduction-induced inhibition of p-JNK/caspase-3 pathway of microvascular pericytes in rats.


Asunto(s)
Caspasa 3/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/enzimología , Fragmentos de Péptidos/farmacología , Pericitos/efectos de los fármacos , Receptor de Factor de Crecimiento Nervioso , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis , Masculino , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Pericitos/enzimología , Pericitos/patología , Fosforilación , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Recuperación de la Función , Transducción de Señal , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
19.
Int J Biochem Cell Biol ; 125: 105794, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32562769

RESUMEN

Neurological disorders are associated with increased oxidative stress. Reactive oxidants damage tissue and promote cell death, but it is apparent that oxidants can have more subtle effects on cell function through the modulation of redox-sensitive signalling pathways. Cells of the blood-brain barrier regulate the brain microenvironment but become dysfunctional during neurological disease. The blood-brain barrier is maintained by many cell types, and is modulated by redox-sensitive pathways, ranging from the cytoskeletal elements responsible for establishing a barrier, to growth factor and cytokine signalling pathways that influence neurovascular cells. During neurological disease, blood-brain barrier cells are exposed to exogenously generated oxidants from immune cells, as well as increasing endogenously oxidant production. These oxidants impair the function of the blood-brain barrier, leading to increased leakage and reduced blood flow. Reducing the impact of oxidants on the function of blood-brain barrier cells may provide new strategies for delaying the progression of neurological disease.


Asunto(s)
Barrera Hematoencefálica/citología , Inflamación/metabolismo , Enfermedades del Sistema Nervioso/metabolismo , Estrés Oxidativo/fisiología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Barrera Hematoencefálica/enzimología , Barrera Hematoencefálica/metabolismo , Muerte Celular/efectos de los fármacos , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Humanos , Inflamación/enzimología , Inflamación/inmunología , Microglía/enzimología , Microglía/metabolismo , Enfermedades del Sistema Nervioso/enzimología , Enfermedades del Sistema Nervioso/fisiopatología , Neutrófilos/enzimología , Neutrófilos/metabolismo , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Pericitos/enzimología , Pericitos/metabolismo , Pericitos/patología , Transducción de Señal/genética
20.
J Cell Physiol ; 218(3): 612-22, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19016245

RESUMEN

The blood-brain barrier (BBB), consisting of specialized endothelial cells surrounded by astrocytes and pericytes, plays a crucial role in brain homeostasis. Many cerebrovascular diseases are associated with BBB breakdown and oxygen (O(2)) deprivation constitutes a critical factor that onsets its disruption. We investigated the impact of astrocytes and pericytes on brain endothelial cell permeability and survival during different degrees of O(2) deprivation. Prolonged exposure to 1% O(2) caused barrier breakdown and exposure to 0.1% O(2) dramatically accelerated disruption and induced cell death, mediated at least in part via caspase-3 activation. Reoxygenation allowed only cells exposed to 1% O(2) to re-establish barrier function. Notably co-culture with astrocytes and pericytes substantially enhanced barrier function under normoxic conditions, and produced differential responses during O(2) deprivation. At 1% O(2) astrocytes partially maintained barrier integrity whereas pericytes accelerated its disruption in the short-term, having positive effects only after prolonged exposure. Unexpectedly, at 0.1% O(2) pericytes were more effective than astrocytes in preserving barrier function although the protection afforded by both cells involved inhibition of caspase-3 pathways. Furthermore, cell-specific regulation of auto- and paracrine VEGF signaling pathways were also in part responsible for the differential modulation of barrier function. Our data suggests that cellular cross-talk within the neurovascular unit is crucial for preservation of barrier integrity and that pericytes, not astrocytes, play a significant role during severe and prolonged O(2) deprivation.


Asunto(s)
Astrocitos/citología , Barrera Hematoencefálica/citología , Oxígeno/metabolismo , Pericitos/citología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Caspasa 3/metabolismo , Inhibidores de Caspasas , Muerte Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Línea Celular , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Uniones Intercelulares/efectos de los fármacos , Uniones Intercelulares/metabolismo , Proteínas de la Membrana/metabolismo , Oxígeno/farmacología , Pericitos/efectos de los fármacos , Pericitos/enzimología , Permeabilidad/efectos de los fármacos , Ratas , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA