Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Am J Physiol Cell Physiol ; 316(5): C741-C752, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30811222

RESUMEN

Endothelial cell (EC) mechanochemical transduction is the process by which mechanical stimuli are sensed by ECs and transduced into biochemical signals and ultimately into physiological responses. Identifying the mechanosensor/mechanochemical transducer(s) and describing the mechanism(s) by which they receive and transmit the signals has remained a central focus within the field. The heterotrimeric G protein, Gαq/11, is proposed to be part of a macromolecular complex together with PECAM-1 at EC junctions and may constitute the mechanochemical transducer as it is rapidly activated within seconds of flow onset. The mechanically activated cation channel Piezo1 has recently been implicated due to its involvement in mediating early responses, such as calcium and ATP release. Here, we investigate the role of Piezo1 in rapid shear stress-induced Gαq/11 activation. We show that flow-induced dissociation of Gαq/11 from PECAM-1 in ECs at 15 s is abrogated by BIM-46187, a selective inhibitor of Gαq/11 activation, suggesting that Gαq/11 activation is required for PECAM-1/Gαq/11 dissociation. Although siRNA knockdown of Piezo1 caused a dramatic decrease in PECAM-1/Gαq/11 association in the basal condition, it had no effect on flow-induced dissociation. Interestingly, siRNA knockdown of Piezo1 caused a marked decrease in PECAM-1 expression. Additionally, selective blockade of Piezo1 with ion channel inhibitors had no effect on flow-induced PECAM-1/Gαq/11 dissociations. Lastly, flow onset caused increased association of Gß1 with Piezo1 as well as with the p101 subunit of phosphoinositide 3-kinase, which were both blocked by the Gßγ inhibitor gallein. Together, our results indicate that flow-induced activation of Piezo1 is not upstream of G protein activation.


Asunto(s)
Células Endoteliales/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Canales Iónicos/metabolismo , Mecanotransducción Celular/fisiología , Estrés Mecánico , Células Cultivadas , Vasos Coronarios/citología , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/metabolismo , Células Endoteliales/efectos de los fármacos , Femenino , Humanos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Masculino , Mecanotransducción Celular/efectos de los fármacos , Resistencia al Corte/efectos de los fármacos , Resistencia al Corte/fisiología , Xantenos/farmacología
2.
Sci Rep ; 8(1): 15957, 2018 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-30374028

RESUMEN

Vascular dysfunction associated with low nitric oxide (NO) biavailability and low plasma L-arginine levels is observed in both human and experimental cerebral malaria (ECM). In ECM, cerebrovascular constriction results in decreased pial blood flow and hypoxia, and administration of NO donors reverses constriction and increases survival. Supplementation of L-arginine, the substrate for NO synthesis by NO synthases, has been considered as a strategy to improve vascular health and act as adjunctive therapy in human severe malaria. We investigated the effect of L-arginine supplementation on pial vascular tonus of mice with ECM after direct superfusion on the brain surface or systemic delivery. Pial arteriolar diameters of Plasmodium berghei-infected mice with implanted cranial windows were measured using intravital microscopy methods, before and after L-arginine administration. Systemic delivery of L-arginine was performed intravenously, at 10, 50, 100 and 200 mg/kg, as bolus injection or slowly through osmotic pumps, combined or not with artesunate. Direct superfusion of L-arginine (10-7M, 10-5M and 10-3M) on the brain surface of mice with ECM resulted in immediate, consistent and dose-dependent dilation of pial arterioles. ECM mice showed marked cerebrovascular constriction that progressively worsened over a 24 h-period after subcutaneous saline bolus administration. L-arginine administration prevented the worsening in pial constriction at all the doses tested, and at 50 mg/kg and 100 mg/kg it induced temporary reversal of vasoconstriction. Slow, continuous delivery of L-arginine by osmotic pumps, or combined bolus administration of artesunate with L-arginine, also prevented worsening of pial constriction and resulted in improved survival of mice with ECM. L-arginine ameliorates pial vasoconstriction in mice with ECM.


Asunto(s)
Arginina/farmacología , Vasoconstricción/efectos de los fármacos , Animales , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Arginina/uso terapéutico , Artesunato/farmacología , Artesunato/uso terapéutico , Arterias Cerebrales/efectos de los fármacos , Arterias Cerebrales/fisiología , Relación Dosis-Respuesta a Droga , Femenino , Malaria Cerebral/tratamiento farmacológico , Malaria Cerebral/mortalidad , Malaria Cerebral/veterinaria , Ratones , Ratones Endogámicos C57BL , Donantes de Óxido Nítrico/farmacología , Donantes de Óxido Nítrico/uso terapéutico , Óxido Nítrico Sintasa/metabolismo , Plasmodium berghei/patogenicidad , Tasa de Supervivencia
3.
Biochem Biophys Res Commun ; 497(1): 220-225, 2018 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-29428723

RESUMEN

Piezo1 is a mechanosensitive cation channel that is activated by shear stress in endothelial cells (ECs). It has been shown to mediate shear-induced EC responses, including increased calcium influx, and vascular functions, such as vascular tone and blood pressure. Yoda1, a selective Piezo1 activator, has been shown to mimic shear-induced responses in ECs. Since shear-induced calcium influx causes Akt and ERK1/2 activation in ECs, we examined the effects of Yoda1 and the role of Piezo1 on their activation. Here, we show that Yoda1 robustly activates Akt and ERK1/2 in ECs. Additionally, the Piezo1 antagonists, gadolinium and ruthenium red, but not GsMTx4, effectively blocks Yoda1-induced Akt activation. Our results suggest that Yoda1-induced Akt and ERK1/2 activation is not dependent on Piezo1.


Asunto(s)
Endopeptidasas/metabolismo , Células Endoteliales/fisiología , Canales Iónicos/metabolismo , Mecanotransducción Celular/fisiología , Tioléster Hidrolasas/metabolismo , Células Cultivadas , Células Endoteliales/citología , Femenino , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Proteína Oncogénica v-akt , Fosforilación
4.
Am J Physiol Cell Physiol ; 312(4): C428-C437, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28148497

RESUMEN

Mechanochemical signal transduction occurs when mechanical forces, such as fluid shear stress, are converted into biochemical responses within the cell. The molecular mechanisms by which endothelial cells (ECs) sense/transduce shear stress into biological signals, including the nature of the mechanosensor, are still unclear. G proteins and G protein-coupled receptors (GPCRs) have been postulated independently to mediate mechanotransduction. In this study, we used in situ proximity ligation assay (PLA) to investigate the role of a specific GPCR/Gαq/11 pair in EC shear stress-induced mechanotransduction. We demonstrated that sphingosine 1-phosphate (S1P) stimulation causes a rapid dissociation at 0.5 min of Gαq/11 from its receptor S1P3, followed by an increased association within 2 min of GPCR kinase-2 (GRK2) and ß-arrestin-1/2 with S1P3 in human coronary artery ECs, which are consistent with GPCR/Gαq/11 activation and receptor desensitization/internalization. The G protein activator AlF4 resulted in increased dissociation of Gαq/11 from S1P3, but no increase in association between S1P3 and either GRK2 or ß-arrestin-1/2. The G protein inhibitor guanosine 5'-(ß-thio) diphosphate (GDP-ß-S) and the S1P3 antagonist VPC23019 both prevented S1P-induced activation. Shear stress also caused the rapid activation within 7 s of S1P3/Gαq/11 There were no increased associations between S1P3 and GRK2 or S1P3 and ß-arrestin-1/2 until 5 min. GDP-ß-S, but not VPC23019, prevented dissociation of Gαq/11 from S1P3 in response to shear stress. Shear stress did not induce rapid dephosphorylation of ß-arrestin-1 or rapid internalization of S1P3, indicating no GPCR activation. These findings suggest that Gαq/11 participates in the sensing/transducing of shear stress independently of GPCR activation in ECs.


Asunto(s)
Células Endoteliales/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Lisofosfolípidos/metabolismo , Mecanotransducción Celular/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Esfingosina/análogos & derivados , Células Cultivadas , Humanos , Masculino , Receptores de Lisoesfingolípidos , Resistencia al Corte/fisiología , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Estrés Mecánico
5.
Am J Physiol Heart Circ Physiol ; 312(3): H485-H500, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28011582

RESUMEN

Vascular endothelial cells play an important role in the regulation of vascular function in response to mechanical stimuli in both healthy and diseased states. Prostaglandin I2 (PGI2) is an important antiatherogenic prostanoid and vasodilator produced in endothelial cells through the action of the cyclooxygenase (COX) isoenzymes COX-1 and COX-2. However, the mechanisms involved in sustained, shear-induced production of COX-2 and PGI2 have not been elucidated but are determined in the present study. We used cultured endothelial cells exposed to steady fluid shear stress (FSS) of 10 dyn/cm2 for 5 h to examine shear stress-induced induction of COX-2/PGI2 Our results demonstrate the relationship between the mechanosensor platelet endothelial cell adhesion molecule-1 (PECAM-1) and the intracellular mechanoresponsive molecules phosphatidylinositol 3-kinase (PI3K), focal adhesion kinase (FAK), and mitogen-activated protein kinase p38 in the FSS induction of COX-2 expression and PGI2 release. Knockdown of PECAM-1 (small interference RNA) expression inhibited FSS-induced activation of α5ß1-integrin, upregulation of COX-2, and release of PGI2 in both bovine aortic endothelial cells (BAECs) and human umbilical vein endothelial cells (HUVECs). Furthermore, inhibition of the PI3K pathway (LY294002) substantially inhibited FSS activation of α5ß1-integrin, upregulation of COX-2 gene and protein expression, and release of PGI2 in BAECs. Inhibition of integrin-associated FAK (PF573228) and MAPK p38 (SB203580) also inhibited the shear-induced upregulation of COX-2. Finally, a PECAM-1-/- mouse model was characterized by reduced COX-2 immunostaining in the aorta and reduced plasma PGI2 levels compared with wild-type mice, as well as complete inhibition of acute flow-induced PGI2 release compared with wild-type animals.NEW & NOTEWORTHY In this study we determined the major mechanotransduction pathway by which blood flow-driven shear stress activates cyclooxygenase-2 (COX-2) and prostaglandin I2 (PGI2) release in endothelial cells. Our work has demonstrated for the first time that COX-2/PGI2 mechanotransduction is mediated by the mechanosensor platelet endothelial cell adhesion molecule-1 (PECAM-1).


Asunto(s)
Ciclooxigenasa 2/biosíntesis , Células Endoteliales/metabolismo , Epoprostenol/biosíntesis , Estrés Mecánico , Animales , Bovinos , Línea Celular , Cilios/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inmunohistoquímica , Integrinas/metabolismo , Péptidos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Transducción de Señal/fisiología , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
J Med Chem ; 58(19): 7895-9, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26367273

RESUMEN

Hybrid products in which the dihydroartemisinin scaffold is combined with NO-donor furoxan and NONOate moieties have been synthesized and studied as potential tools for the treatment of cerebral malaria (CM). The designed products were able to dilate rat aorta strips precontracted with phenylephrine with a NO-dependent mechanism. All hybrid compounds showed preserved antiplasmodial activity in vitro and in vivo against Plasmodium berghei ANKA, comparable to artesunate and artemether. Hybrid 10, selected for additional studies, was capable of increasing survival of mice with late-stage CM from 27.5% to 51.6% compared with artemether. Artemisinin-NO-donor hybrid compounds show promise as potential new drugs for treating cerebral malaria.


Asunto(s)
Antimaláricos/química , Antimaláricos/farmacología , Artemisininas/química , Malaria Cerebral/tratamiento farmacológico , Donantes de Óxido Nítrico/química , Donantes de Óxido Nítrico/farmacología , Animales , Antimaláricos/síntesis química , Arteméter , Artemisininas/farmacología , Artesunato , Técnicas de Química Sintética , Ratones , Terapia Molecular Dirigida/métodos , Relajación Muscular/efectos de los fármacos , Plasmodium berghei/efectos de los fármacos , Ratas , Vasodilatadores/química , Vasodilatadores/farmacología
7.
Sci Signal ; 7(326): ra48, 2014 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-24847117

RESUMEN

Thyroid hormone (TH) is essential for vertebrate development and the homeostasis of most adult tissues, including bone. TH stimulates target gene expression through the nuclear thyroid receptors TRα and TRß; however, TH also has rapid, transcription-independent (nongenomic) effects. We found a previously uncharacterized plasma membrane-bound receptor that was necessary and sufficient for nongenomic TH signaling in several cell types. We determined that this receptor is generated by translation initiation from an internal methionine of TRα, which produces a transcriptionally incompetent protein that is palmitoylated and associates with caveolin-containing plasma membrane domains. TH signaling through this receptor stimulated a pro-proliferative and pro-survival program by increasing the intracellular concentrations of calcium, nitric oxide (NO), and cyclic guanosine monophosphate (cGMP), which led to the sequential activation of protein kinase G II (PKGII), the tyrosine kinase Src, and extracellular signal-regulated kinase (ERK) and Akt signaling. Hypothyroid mice exhibited a cGMP-deficient state with impaired bone formation and increased apoptosis of osteocytes, which was rescued by a direct stimulator of guanylate cyclase. Our results link nongenomic TH signaling to a previously uncharacterized membrane-bound receptor, and identify NO synthase, guanylate cyclase, and PKGII as TH effectors that activate kinase cascades to regulate cell survival and proliferation.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Microdominios de Membrana/metabolismo , Osteocitos/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Hormonas Tiroideas/metabolismo , Adulto , Animales , Células Cultivadas , Humanos , Hipotiroidismo/genética , Hipotiroidismo/metabolismo , Microdominios de Membrana/genética , Ratones , Ratones Transgénicos , Osteogénesis/genética , Ratas , Receptores de Hormona Tiroidea/genética , Hormonas Tiroideas/genética
8.
J Biol Chem ; 289(11): 7413-24, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24497640

RESUMEN

The endothelial cell-cell junction has emerged as a major cell signaling structure that responds to shear stress by eliciting the activation of signaling pathways. Platelet endothelial cell adhesion molecule-1 (PECAM-1) and heterotrimeric G protein subunits Gαq and 11 (Gαq/11) are junctional proteins that have been independently proposed as mechanosensors. Our previous findings suggest that they form a mechanosensitive junctional complex that discriminates between different flow profiles. The nature of the PECAM-1·Gαq/11 interaction is still unclear although it is likely an indirect association. Here, we investigated the role of heparan sulfates (HS) in mediating this interaction and in regulating downstream signaling in response to flow. Co-immunoprecipitation studies show that PECAM-1·Gαq/11 binding is dramatically decreased by competitive inhibition with heparin, pharmacological inhibition with the HS antagonist surfen, and enzymatic removal of HS chains with heparinase III treatment as well as by site-directed mutagenesis of basic residues within the extracellular domain of PECAM-1. Using an in situ proximity ligation assay, we show that endogenous PECAM-1·Gαq/11 interactions in endothelial cells are disrupted by both competitive inhibition and HS degradation. Furthermore, we identified the heparan sulfate proteoglycan syndecan-1 in complexes with PECAM-1 that are rapidly decreased in response to flow. Finally, we demonstrate that flow-induced Akt activation is attenuated in endothelial cells in which PECAM-1 was knocked down and reconstituted with a binding mutant. Taken together, our results indicate that the PECAM-1·Gαq/11 mechanosensitive complex contains an endogenous heparan sulfate proteoglycan with HS chains that is critical for junctional complex assembly and regulating the flow response.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Regulación de la Expresión Génica , Heparitina Sulfato/química , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Sindecano-1/metabolismo , Comunicación Celular , Células Cultivadas , Células Endoteliales/citología , Células HEK293 , Humanos , Mutación , Unión Proteica , Multimerización de Proteína , Estructura Terciaria de Proteína , ARN Interferente Pequeño/metabolismo , Estrés Mecánico , Transfección , Enfermedades Vasculares/metabolismo
9.
J Cell Biochem ; 115(1): 121-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23913776

RESUMEN

Endothelial cells undergo a rapid cell-cell junction inclination following exposure to atheroprotective unidirectional flow. In contrast, atherosclerotic lesions correlate with a heterogeneous distribution of the junctional wall inclination in cells exposed to time-varying, reversing, and oscillatory flow as well as to low mean shear stress. However, the underlying biochemical events by which endothelial cells distinctively respond to unidirectional versus flow reversal remain unclear. Here, we show that the subcellular distribution of flow-induced Akt-1 phosphorylation in endothelial cells lining the mouse aorta varies depending on local hemodynamics. Activated Akt-1 accumulated in perinuclear areas of cells in regions predisposed to disturbed flow but were localized at the cell-cell junction in regions of high unidirectional laminar shear stress. In flow-adapted human endothelial cells, reversal in flow direction was associated within minutes with a subcellular concentration of phosphorylated Akt-1 at the upstream edge of cells. Interestingly, oscillatory flow (with a zero mean shear stress) failed to activate Akt-1, whereas a unidirectional pulsatile flow of similar amplitude induced an increase in Akt-1 phosphorylation. Finally, silencing of the G protein αq/11 subunit abrogated both flow-induced Akt-1 and GSK-3ß activation. Together, these results characterize the existence of a Gαq/11-mediated Akt-1 signaling pathway that is dynamically responsive to flow direction, thereby offering a novel approach to regulating EC dysfunctions in regions subjected to flow reversal.


Asunto(s)
Células Endoteliales/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Aorta/citología , Células Cultivadas , Células Endoteliales/citología , Endotelio Vascular/citología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Ratones , Ratones Endogámicos C57BL , Fosforilación , Estrés Mecánico
10.
Proc Natl Acad Sci U S A ; 110(47): 18820-5, 2013 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-24191050

RESUMEN

Molecular targeting of the two receptor interaction domains of the epigenetic repressor silencing mediator of retinoid and thyroid hormone receptors (SMRT(mRID)) produced a transplantable skeletal syndrome that reduced radial bone growth, increased numbers of bone-resorbing periosteal osteoclasts, and increased bone fracture risk. Furthermore, SMRT(mRID) mice develop spontaneous primary myelofibrosis, a chronic, usually idiopathic disorder characterized by progressive bone marrow fibrosis. Frequently linked to polycythemia vera and chronic myeloid leukemia, myelofibrosis displays high patient morbidity and mortality, and current treatment is mostly palliative. To decipher the etiology of this disease, we identified the thrombopoietin (Tpo) gene as a target of the SMRT-retinoic acid receptor signaling pathway in bone marrow stromal cells. Chronic induction of Tpo in SMRT(mRID) mice results in up-regulation of TGF-ß and PDGF in megakaryocytes, uncontrolled proliferation of bone marrow reticular cells, and fibrosis of the marrow compartment. Of therapeutic relevance, we show that this syndrome can be rescued by retinoid antagonists, demonstrating that the physical interface between SMRT and retinoic acid receptor can be a potential therapeutic target to block primary myelofibrosis disease progression.


Asunto(s)
Médula Ósea/metabolismo , Citocinas/metabolismo , Represión Epigenética/fisiología , Co-Represor 2 de Receptor Nuclear/antagonistas & inhibidores , Mielofibrosis Primaria/tratamiento farmacológico , Transducción de Señal/fisiología , Trombopoyetina/genética , Fosfatasa Alcalina/sangre , Animales , Benzotiazoles , Calcio/sangre , Proliferación Celular/efectos de los fármacos , Cartilla de ADN/genética , Diaminas , Ensayo de Inmunoadsorción Enzimática , Perfilación de la Expresión Génica , Técnicas de Sustitución del Gen , Luciferasas , Megacariocitos/metabolismo , Ratones , Co-Represor 2 de Receptor Nuclear/genética , Compuestos Orgánicos , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Reacción en Cadena de la Polimerasa , Mielofibrosis Primaria/etiología , Quinolinas , Trombopoyetina/biosíntesis , Factor de Crecimiento Transformador beta/metabolismo
11.
Virulence ; 4(8): 686-97, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24128424

RESUMEN

Ischemia and hypoxia have been implicated in cerebral malaria (CM) pathogenesis, although direct measurements of hypoxia have not been conducted. C57BL/6 mice infected with Plasmodium berghei ANKA (PbA) develop a neurological syndrome known as experimental cerebral malaria (ECM), whereas BALB/c mice are resistant to ECM. In this study, intravital microscopy methods were used to quantify hemodynamic changes, vascular/tissue oxygen (O2) tension (PO2), and perivascular pH in vivo in ECM and non-ECM models, employing a closed cranial window model. ECM mice on day 6 of infection showed marked decreases in pial blood flow, vascular (arteriolar, venular), and perivascular PO2, perivascular pH, and systemic hemoglobin levels. Changes were more dramatic in mice with late-stage ECM compared with mice with early-stage ECM. These changes led to drastic decreases in O2 delivery to the brain tissue. In addition, ECM animals required a greater PO2 gradient to extract the same amount of O2 compared with non-infected animals, as the pial tissues extract O2 from the steepest portion of the blood O2 equilibrium curve. ECM animals also showed increased leukocyte adherence in postcapillary venules, and the intensity of adhesion was inversely correlated with blood flow and O2 extraction. PbA-infected BALB/c mice displayed no neurological signs on day 6 and while they did show changes similar to those observed in C57BL/6 mice (decreased pial blood flow, vascular/tissue PO2, perivascular pH, hemoglobin levels), non-ECM animals preserved superior perfusion and oxygenation compared with ECM animals at similar anemia and parasitemia levels, resulting in better O2 delivery and O2 extraction by the brain tissue. In conclusion, direct quantitative assessment of pial hemodynamics and oxygenation in vivo revealed that ECM is associated with severe progressive brain tissue hypoxia and acidosis.


Asunto(s)
Encéfalo/patología , Hipoxia/patología , Malaria Cerebral/patología , Animales , Análisis Químico de la Sangre , Química Encefálica , Modelos Animales de Enfermedad , Humanos , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía , Presión Parcial , Plasmodium berghei/crecimiento & desarrollo
12.
Antimicrob Agents Chemother ; 57(11): 5462-71, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23979751

RESUMEN

Cerebral malaria (CM) is associated with low nitric oxide (NO) bioavailability, cerebrovascular constriction, occlusion, and hypoperfusion. Administration of exogenous NO partially prevents the neurological syndrome and associated vascular pathology in an experimental CM (ECM) mouse model. In this study, we evaluated the effects of transdermal glyceryl trinitrate in preventing ECM and, in combination with artemether, rescuing late-stage ECM mice from mortality. The glyceryl trinitrate and/or artemether effect on survival and clinical recovery was evaluated in C57BL/6 mice infected with P. berghei ANKA. NO synthase (NOS) expression in mouse brain was determined by Western blots. Mean arterial pressure (MAP) and pial arteriolar diameter were monitored using a tail-cuff blood pressure system and a cranial window preparation, respectively. Preventative administration of glyceryl trinitrate at 0.025 mg/h decreased ECM mortality from 67 to 11% and downregulated inducible NOS expression in the brain. When administered as adjunctive rescue therapy with artemether, glyceryl trinitrate increased survival from 47 to 79%. The adjunctive therapy caused a sustained reversal of pial arteriolar vasoconstriction in ECM mice, an effect not observed with artemether alone. Glyceryl trinitrate induced a 13% decrease in MAP in uninfected mice but did not further affect MAP in hypotensive ECM mice. Glyceryl trinitrate, when combined with artemether, was an effective adjunctive rescue treatment for ECM. This treatment ameliorated pial arteriolar vasospasm and did not significantly affect MAP. These results indicate that transdermal glyceryl trinitrate has potential to be considered as a candidate for adjunctive therapy for CM.


Asunto(s)
Antimaláricos/farmacología , Artemisininas/farmacología , Encéfalo/efectos de los fármacos , Malaria Cerebral/tratamiento farmacológico , Nitroglicerina/farmacología , Vasodilatadores/farmacología , Administración Cutánea , Animales , Arteméter , Presión Arterial , Encéfalo/irrigación sanguínea , Encéfalo/parasitología , Sinergismo Farmacológico , Femenino , Expresión Génica/efectos de los fármacos , Malaria Cerebral/mortalidad , Malaria Cerebral/parasitología , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo I/metabolismo , Plasmodium berghei/efectos de los fármacos , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/patogenicidad , Análisis de Supervivencia , Resultado del Tratamiento , Vasoconstricción/efectos de los fármacos
13.
PLoS Pathog ; 9(6): e1003444, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23818850

RESUMEN

Cerebrovascular dysfunction plays a key role in the pathogenesis of cerebral malaria. In experimental cerebral malaria (ECM) induced by Plasmodium berghei ANKA, cerebrovascular dysfunction characterized by vascular constriction, occlusion and damage results in impaired perfusion and reduced cerebral blood flow and oxygenation, and has been linked to low nitric oxide (NO) bioavailability. Here, we directly assessed cerebrovascular function in ECM using a novel cranial window method for intravital microscopy of the pial microcirculation and probed the role of NOS isoforms and phosphorylation patterns in the impaired vascular responses. We show that pial arteriolar responses to endothelial NOS (eNOS) and neuronal NOS (nNOS) agonists (Acetylcholine (ACh) and N-Methyl-D-Aspartate (NMDA)) were blunted in mice with ECM, and could be partially recovered by exogenous supplementation of tetrahydrobiopterin (BH4). Pial arterioles in non-ECM mice infected by Plasmodium berghei NK65 remained relatively responsive to the agonists and were not significantly affected by BH4 treatment. These findings, together with the observed blunting of NO production upon stimulation by the agonists, decrease in total NOS activity, augmentation of lipid peroxidation levels, upregulation of eNOS protein expression, and increase in eNOS and nNOS monomerization in the brain during ECM development strongly indicate a state of eNOS/nNOS uncoupling likely mediated by oxidative stress. Furthermore, the downregulation of Serine 1176 (S1176) phosphorylation of eNOS, which correlated with a decrease in cerebrovascular wall shear stress, implicates hemorheological disturbances in eNOS dysfunction in ECM. Finally, pial arterioles responded to superfusion with the NO donor, S-Nitroso-L-glutathione (GSNO), but with decreased intensity, indicating that not only NO production but also signaling is perturbed during ECM. Therefore, the pathological impairment of eNOS and nNOS functions contribute importantly to cerebrovascular dysfunction in ECM and the recovery of intrinsic functionality of NOS to increase NO bioavailability and restore vascular health represents a target for ECM treatment.


Asunto(s)
Circulación Cerebrovascular , Malaria Cerebral , Microcirculación , Óxido Nítrico/metabolismo , Plasmodium berghei/metabolismo , Acetilcolina/farmacología , Animales , Biopterinas/análogos & derivados , Biopterinas/farmacología , Agonistas Colinérgicos , Agonistas de Aminoácidos Excitadores/farmacología , Femenino , Malaria Cerebral/metabolismo , Malaria Cerebral/parasitología , Malaria Cerebral/fisiopatología , Ratones , N-Metilaspartato/farmacología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo
14.
Malar J ; 12: 138, 2013 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-23617605

RESUMEN

BACKGROUND: Human cerebral malaria (HCM) is a life-threatening complication caused by Plasmodium falciparum infection that continues to be a major global health problem despite optimal anti-malarial treatment. In the experimental model of cerebral malaria (ECM) by Plasmodium berghei ANKA, bolus administration of nimodipine at high doses together with artemether, increases survival of mice with ECM. However, the dose and administration route used is associated with cardiovascular side effects such as hypotension and bradycardia in humans and mice, which could preclude its potential use as adjunctive treatment in HCM. METHODS: In the present study, alternative delivery systems for nimodipine during late-stage ECM in association with artesunate were searched to define optimal protocols to achieve maximum efficacy in increasing survival in rescue therapy while causing the least cardiac side effects. The baseline electrocardiogram (ECG) and arterial pressure characteristics of uninfected control animals and of mice with ECM and its response upon rescue treatment with artesunate associated or not with nimodipine is also analysed. RESULTS: Nimodipine, given at 0.5 mg/kg/day via a slow and continuous delivery system by osmotic pumps, increases survival of mice with ECM when used as adjunctive treatment to artesunate. Mice with ECM showed hypotension and ECG changes, including bradycardia and increases in PR, QRS, QTc and ST interval duration. ECM mice also show increased QTc dispersion, heart rate variability (HRV), RMSSD, low frequency (LF) and high frequency (HF) bands of the power spectrum. Both sympathetic and parasympathetic inputs to the heart were increased, but there was a predominance of sympathetic tone as demonstrated by an increased LF/HF ratio. Nimodipine potentiated bradycardia when given by bolus injection, but not when via osmotic pumps. In addition, nimodipine shortened PR duration and improved HRV, RMSSD, LF and HF powers in mice with ECM. In addition, nimodipine did not increased hypotension or decreased the speed of arterial pressure recovery when used in rescue therapy with artesunate. CONCLUSIONS: These data show that slow and continuous delivery of lower doses of nimodipine improves survival of mice with ECM in rescue therapy with artesunate while showing a safer profile in terms of cardiovascular effects.


Asunto(s)
Antihipertensivos/administración & dosificación , Malaria Cerebral/tratamiento farmacológico , Nimodipina/administración & dosificación , Plasmodium berghei/efectos de los fármacos , Terapia Recuperativa/métodos , Administración Intravenosa , Animales , Antimaláricos/administración & dosificación , Artemisininas/administración & dosificación , Artesunato , Modelos Animales de Enfermedad , Electrocardiografía , Femenino , Malaria Cerebral/parasitología , Ratones , Ratones Endogámicos C57BL , Análisis de Supervivencia , Resultado del Tratamiento
15.
Biochem Biophys Res Commun ; 434(3): 641-6, 2013 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-23583373

RESUMEN

Although several potential mechanosensors/mechanotransducers have been proposed, the precise mechanisms by which ECs sense and respond to mechanical forces and translate them into biochemical signals remains unclear. Here, we report that two major ligand-dependent tyrosine autophosphorylation sites of VEGFR2, Y1175 and Y1214, are rapidly activated by shear stress in human coronary artery endothelial cells (HCAECs). Neutralizing antibody against VEGFR2 not only abrogates flow-induced phosphorylation of these tyrosine residues, but also has a marked inhibitory effect on downstream eNOS activation. In situ proximity ligation assay revealed that VEGF and VEGFR2 are closely associated in HCAECs, and more importantly, this association is increased with flow. Finally, we show that flow-induced VEGFR2 activation is attenuated in the presence of the broad spectrum matrix metalloproteinase (MMP) inhibitor, GM6001. Taken together, our results suggest that a ligand-dependent mechanism involving the activity of MMPs plays a key role in the early, shear stress-induced activation of VEGFR2.


Asunto(s)
Metaloproteinasas de la Matriz/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Anticuerpos Neutralizantes/inmunología , Western Blotting , Células Cultivadas , Vasos Coronarios/citología , Vasos Coronarios/enzimología , Vasos Coronarios/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/enzimología , Endotelio Vascular/metabolismo , Humanos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología
16.
Microcirculation ; 20(5): 394-404, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23279271

RESUMEN

OBJECTIVE: The acute implantation of a cranial window for studying cerebroarteriolar reactivity in living animals involves a highly surgically invasive craniotomy procedure at the time of experimentation, which limits its application in severely ill animals such as in the experimental murine model of cerebral malaria (ECM). To overcome this problem, a chronic window implantation scheme was designed and implemented. METHODS: A partial craniotomy is first performed by creating a skull bone flap in the healthy mice, which are then left to recover for one to two weeks, followed by infection to induce ECM. Uninfected animals are utilized as control. When cranial superfusion is needed, the bone flap is retracted and window implantation completed by assembling a perfusion chamber for compound delivery to the exposed brain surface. The presurgical step is intended to minimize surgical trauma on the day of experimentation. RESULTS: Chronic preparations in uninfected mice exhibited remarkably improved stability over acute ones by significantly reducing periarteriolar tissue damage and enhancing cerebroarteriolar dilator responses. The chronic scheme was successfully implemented in ECM mice, which unveiled novel preliminary insights into impaired cerebroarteriolar reactivity and eNOS dysfunction. CONCLUSION: The chronic scheme presents an innovative approach for advancing our mechanistic understanding on cerebrovascular dysfunction in ECM.


Asunto(s)
Circulación Cerebrovascular , Malaria Cerebral/fisiopatología , Plasmodium berghei , Cráneo/cirugía , Animales , Malaria Cerebral/patología , Ratones , Óxido Nítrico Sintasa de Tipo III/metabolismo
17.
Malar J ; 11: 366, 2012 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-23126583

RESUMEN

BACKGROUND: Plasmodium berghei rodent malaria is a well-known model for the investigation of anti-malarial drug efficacy in vivo. However, the availability of drug in vitro assays in P. berghei is reduced when compared with the spectrum of techniques existing for Plasmodium falciparum. New alternatives to the current manual or automated methods described for P. berghei are attractive. The present study reports a new ELISA drug in vitro assay for P. berghei using two monoclonal antibodies against the parasite lactate dehydrogenase (pLDH). METHODS: This procedure includes a short-in vitro culture, the purification of schizonts and the further generation of synchronized mice infections. Early stages of the parasite are then incubated against different concentrations of anti-malarial drugs using micro-plates. The novelty of this procedure in P. berghei relies on the quantification of the drug activity derived from the amount of pLDH estimated by an ELISA assay using two monoclonal antibodies: 14C1 and 19G7. The IC50s obtained through the ELISA assay were compared with those from the micro-test. RESULTS: The initial parameters of the synchronized samples used in the in vitro assays were a parasitaemia of 0.5% and haematocrit of 1%, with an incubation period of 22 hours at 36.5°C. pLDH detection using a 14C1 coating at 10 µg/ml and 19G7 at 2.5 × 10⁻³ µg/ml provided good readouts of optical densities with low background in negative controls and specific detection levels for all parasite stages. IC50s values derived from the ELISA assay for artesunate, chloroquine, amodiaquine and quinine were: 15, 7, 2, and 144 nM, respectively. When artesunate and chloroquine IC50s were evaluated using the micro-test similar values were obtained. CONCLUSION: This ELISA-based in vitro drug assay is easy to implement, fast, and avoids the use radioisotopes or expensive equipment. The utility of this simple assay for screening anti-malarial drug activity against P. berghei in vitro is demonstrated.


Asunto(s)
Antimaláricos/farmacología , Ensayo de Inmunoadsorción Enzimática/métodos , L-Lactato Deshidrogenasa/análisis , Plasmodium berghei/efectos de los fármacos , Plasmodium berghei/enzimología , Animales , Anticuerpos Monoclonales , Anticuerpos Antiprotozoarios , Evaluación Preclínica de Medicamentos , Resistencia a Medicamentos , Proteínas Fluorescentes Verdes/genética , L-Lactato Deshidrogenasa/inmunología , Malaria/tratamiento farmacológico , Malaria/parasitología , Ratones , Ratones Endogámicos C57BL , Parasitemia/tratamiento farmacológico , Parasitemia/parasitología , Plasmodium berghei/genética , Proteínas Recombinantes/genética
18.
Am J Physiol Cell Physiol ; 303(4): C467-73, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22700794

RESUMEN

Disturbed flow patterns, including reversal in flow direction, are key factors in the development of dysfunctional endothelial cells (ECs) and atherosclerotic lesions. An almost immediate response of ECs to fluid shear stress is the increase in cytosolic calcium concentration ([Ca(2+)](i)). Whether the source of [Ca(2+)](i) is extracellular, released from Ca(2+) intracellular stores, or both is still undefined, though it is likely dependent on the nature of forces involved. We have previously shown that a change in flow direction (retrograde flow) on a flow-adapted endothelial monolayer induces the remodeling of the cell-cell junction along with a dramatic [Ca(2+)](i) burst compared with cells exposed to unidirectional or orthograde flow. The heterotrimeric G protein-α q and 11 subunit (Gα(q/11)) is a likely candidate in effecting shear-induced increases in [Ca(2+)](i) since its expression is enriched at the junction and has been previously shown to be activated within seconds after onset of flow. In flow-adapted human ECs, we have investigated to what extent the Gα(q/11) pathway mediates calcium dynamics after reversal in flow direction. We observed that the elapsed time to peak [Ca(2+)](i) response to a 10 dyn/cm(2) retrograde shear stress was increased by 11 s in cells silenced with small interfering RNA directed against Gα(q/11). A similar lag in [Ca(2+)](i) transient was observed after cells were treated with the phospholipase C (PLC)-ßγ inhibitor, U-73122, or the phosphatidylinositol-specific PLC inhibitor, edelfosine, compared with controls. Lower levels of inositol 1,4,5-trisphosphate accumulation seconds after the onset of flow correlated with the increased lag in [Ca(2+)](i) responses observed with the different treatments. In addition, inhibition of the inositol 1,4,5-trisphosphate receptor entirely abrogated flow-induced [Ca(2+)](i). Taken together, our results identify the Gα(q/11)-PLC pathway as the initial trigger for retrograde flow-induced endoplasmic reticulum calcium store release, thereby offering a novel approach to regulating EC dysfunctions in regions subjected to the reversal of blood flow.


Asunto(s)
Calcio/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Señalización del Calcio/fisiología , Citosol/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Humanos , Receptores de Inositol 1,4,5-Trifosfato/antagonistas & inhibidores , Fosfatos de Inositol/metabolismo , Fosfoinositido Fosfolipasa C/metabolismo , Interferencia de ARN , Sistemas de Mensajero Secundario , Estrés Mecánico
19.
PLoS One ; 7(3): e33336, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22413015

RESUMEN

Interstitial fluid flow (IFF) is a potent regulatory signal in bone. During mechanical loading, IFF is generated through two distinct mechanisms that result in spatially distinct flow profiles: poroelastic interactions within the lacunar-canalicular system, and intramedullary pressurization. While the former generates IFF primarily within the lacunar-canalicular network, the latter generates significant flow at the endosteal surface as well as within the tissue. This gives rise to the intriguing possibility that loading-induced IFF may differentially activate osteocytes or surface-residing cells depending on the generating mechanism, and that sensation of IFF generated via intramedullary pressurization may be mediated by a non-osteocytic bone cell population. To begin to explore this possibility, we used the Dmp1-HBEGF inducible osteocyte ablation mouse model and a microfluidic system for modulating intramedullary pressure (ImP) to assess whether structural adaptation to ImP-driven IFF is altered by partial osteocyte depletion. Canalicular convective velocities during pressurization were estimated through the use of fluorescence recovery after photobleaching and computational modeling. Following osteocyte ablation, transgenic mice exhibited severe losses in bone structure and altered responses to hindlimb suspension in a compartment-specific manner. In pressure-loaded limbs, transgenic mice displayed similar or significantly enhanced structural adaptation to Imp-driven IFF, particularly in the trabecular compartment, despite up to ∼50% of trabecular lacunae being uninhabited following ablation. Interestingly, regression analysis revealed relative gains in bone structure in pressure-loaded limbs were correlated with reductions in bone structure in unpressurized control limbs, suggesting that adaptation to ImP-driven IFF was potentiated by increases in osteoclastic activity and/or reductions in osteoblastic activity incurred independently of pressure loading. Collectively, these studies indicate that structural adaptation to ImP-driven IFF can proceed unimpeded following a significant depletion in osteocytes, consistent with the potential existence of a non-osteocytic bone cell population that senses ImP-driven IFF independently and potentially parallel to osteocytic sensation of poroelasticity-derived IFF.


Asunto(s)
Técnicas de Ablación/métodos , Adaptación Fisiológica , Huesos/fisiología , Huesos/cirugía , Líquido Extracelular/fisiología , Osteocitos , Técnicas de Ablación/instrumentación , Animales , Densidad Ósea , Resorción Ósea/etiología , Femenino , Suspensión Trasera/efectos adversos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Presión
20.
J Neuroimmune Pharmacol ; 7(2): 477-87, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22391863

RESUMEN

Administration of the exogenous nitric oxide (NO) donor dipropylenetriamine-NONOate (DPTA-NO) to mice during Plasmodium berghei ANKA (PbA) infection largely prevents development of experimental cerebral malaria (ECM). However, a high dose (1 mg/mouse twice a day) is necessary and causes potent side effects such as marked hypotension. In the present study we evaluated whether an alternative, physiologically relevant NO donor, S-nitrosoglutathione (GSNO), was able to prevent ECM at lower doses with minimal side effects. Prophylactic treatment with high (3.5 mg), intermediate (0.35 mg) or low (0.035 mg) doses of GSNO decreased incidence of ECM in PbA-infected mice, decreasing also edema, leukocyte accumulation and hemorrhage incidence in the brain. The high dose inhibited parasite growth and also induced transient hypotension. Low and intermediate doses had no or only mild effects on parasitemia, blood pressure, and heart rate compared to saline-treated mice. PbA infection decreased brain total and reduced (GSH) glutathione levels. Brain levels of oxidized (GSSG) glutathione and the GSH/GSSG ratio were positively correlated with temperature and motor behavior. Low and intermediate doses of GSNO failed to restore the depleted brain total glutathione and GSH levels, suggesting that ECM prevention by GSNO was probably related to other effects such as inhibition of inflammation and vascular protection. These results indicate that ECM is associated with depletion of the brain glutathione pool and that GSNO is able to prevent ECM development in a wide range of doses, decreasing brain inflammation and inducing milder cardiovascular side effects.


Asunto(s)
Encéfalo/efectos de los fármacos , Malaria Cerebral/prevención & control , Donantes de Óxido Nítrico/farmacología , S-Nitrosoglutatión/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Glutatión/análisis , Frecuencia Cardíaca/efectos de los fármacos , Malaria Cerebral/patología , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...