Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Phytother Res ; 35(6): 3167-3180, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33885189

RESUMEN

Sarsasapogenin (Sar), a natural steroidal compound, shows neuroprotection, cognition-enhancement, antiinflammation, antithrombosis effects, and so on. However, whether Sar has ameliorative effects on diabetes-associated cognitive impairment remains unknown. In this study, we found that Sar ameliorated diabetes-associated memory impairment in streptozotocin-induced diabetic rats, evidenced by increased numbers of crossing platform and percentage of time spent in the target quadrant in Morris water maze tests, and suppressed the nucleotide-binding domain and leucine-rich repeat containing protein 1 (NLRP1) inflammasome in hippocampus and cerebral cortex. Furthermore, Sar inhibited advanced glycation end-products and its receptor (AGEs/RAGE) axis and suppressed up-regulation of thrombin receptor protease-activated receptor 1 (PAR-1) in cerebral cortex. On the other hand, Sar mitigated high glucose-induced neuronal damages, NLRP1 inflammasome activation, and PAR-1 up-regulation in high glucose-cultured SH-SY5Y cells, but did not affect thrombin activity. Moreover, the effects of Sar were similar to those of a selective PAR-1 antagonist vorapaxar. Further studies indicated that activation of the NLRP1 inflammasome and NF-κB mediated the effect of PAR-1 up-regulation in high glucose condition by using PAR-1 knockdown assay. In summary, this study demonstrated that Sar prevented memory impairment caused by diabetes, which was achieved through suppressing neuroinflammation from activated NLRP1 inflammasome and NF-κB regulated by cerebral PAR-1. HIGHLIGHTS: Sarsasapogenin ameliorated memory impairment caused by diabetes in rats. Sarsasapogenin mitigated neuronal damages and neuroinflammation by down-regulating cerebral PAR-1. The NLRP1 inflammasome and NF-κB signaling mediated the pro-inflammatory effects of PAR-1. Sarsasapogenin was a pleiotropic neuroprotective agent and memory enhancer in diabetic rodents.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Trastornos de la Memoria/tratamiento farmacológico , Espirostanos/farmacología , Animales , Línea Celular , Regulación hacia Abajo , Hipocampo/efectos de los fármacos , Humanos , Inflamasomas/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , FN-kappa B/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Transducción de Señal/efectos de los fármacos , Estreptozocina
2.
Phytomedicine ; 78: 153314, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32882582

RESUMEN

BACKGROUND: Sarsasapogenin (Sar) shows good effects on diabetic nephropathy (DN) through inhibition of the NLRP3 inflammasome, yet the potential mechanism is not well known. PURPOSE: This study was designed to explore the regulation of thrombin and/or its receptor protease-activated receptor 1 (PAR-1) on the NLRP3 inflammasome and NF-κB signaling in DN condition, and further expounded the molecular mechanism of Sar on DN. METHODS: Streptozotocin-induced diabetic rats were treated by gavage with Sar (0, 20 and 60 mg/kg) for consecutive 10 weeks. Then urine and serum were collected for protein excretion, creatinine, urea nitrogen, and uric acid assay reflecting renal functions, renal tissue sections for periodic acid-Schiff staining and ki67 expression reflecting cell proliferation, and renal cortex for the NLRP3 inflammasome and NF-κB signaling as well as thrombin/PAR-1 signaling. High glucose-cultured human mesangial cells (HMCs) were used to further investigate the effects and mechanisms of Sar. RESULTS: Sar markedly ameliorated the renal functions and mesangial cell proliferation in diabetic rats, and suppressed activation of the NLRP3 inflammasome and NF-κB in renal cortex. Moreover, Sar remarkably down-regulated PAR-1 in protein and mRNA levels but didn't affect thrombin activity in kidney, although thrombin activity was significantly decreased in the renal cortex of diabetic rats. Meanwhile, high glucose induced activation of the NLRP3 inflammasome and NF-κB, and increased PAR-1 expression while didn't change thrombin activity in HMCs; however, Sar co-treatment ameliorated all the above indices. Further studies demonstrated that PAR-1 knockdown attenuated activation of the NLRP3 inflammasome and NF-κB, and Sar addition strengthened these effects in high glucose-cultured HMCs. CONCLUSION: Sar relieved DN in rat through inhibition of the NLRP3 inflammasome and NF-κB by down-regulating PAR-1 in kidney.


Asunto(s)
Nefropatías Diabéticas/tratamiento farmacológico , Células Mesangiales/efectos de los fármacos , Receptor PAR-1/metabolismo , Espirostanos/farmacología , Animales , Glucemia/metabolismo , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Humanos , Inflamasomas/efectos de los fármacos , Riñón/efectos de los fármacos , Riñón/metabolismo , Masculino , Células Mesangiales/metabolismo , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Nefritis/tratamiento farmacológico , Nefritis/metabolismo , Ratas Sprague-Dawley , Receptor PAR-1/genética , Trombina/metabolismo
3.
Methods Mol Biol ; 1824: 371-385, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30039419

RESUMEN

Cardiovascular diseases (CVDs) are becoming major contributors to the burden of disease due to genetic and environmental factors. Despite current standard oral care, cardiovascular risk remains relatively high. A triple antiplatelet therapy with a cyclooxygenase-1 (COX-1) inhibitor, a P2Y12 receptor antagonist, and a protease-activated receptor-1 (PAR-1) antagonist has been established in the secondary prevention of atherothrombosis in patients with acute myocardial infraction and in those with peripheral artery disease. However, due to the combinatorial use of three different drugs, patients receiving this triple therapy are exposed to enhanced risk of bleeding. Conforming to polypharmacology principles, the discovery of a single compound that can simultaneously block the three platelet activation pathways (PAR-1, P2Y12, and COX-1) is of importance. Natural products have served as an inexhaustible source of bioactive compounds presenting a diverse pharmaceutical profile, including anti-inflammatory, antioxidant, anticancer, and antithrombotic activity. Indeed, principal component analysis indicated that natural products have the potential to inhibit the three aforementioned pathways, though existed reports refer to single inhibition mechanism on specific receptor(s) implicated in platelet activation. We thus set out to explore possibilities that take advantage of this potential of natural products and shape the basis to produce novel compounds that could simultaneously target PAR-1, P2Y12, and COX-1 platelet activation pathways. Polyunsaturated fatty acids (PUFAs) have multiple effects leading to improvements in blood pressure and cardiac function and arterial compliance. A promising approach to achieve the desirable goal is the bioconjugation of natural products with PUFAs. Herein, we describe the principles that should be followed to develop molecular hybrids bearing triple antiplatelet activity profile.


Asunto(s)
Plaquetas , Ciclooxigenasa 1 , Inhibidores de la Ciclooxigenasa , Ácidos Grasos Insaturados , Plasma/química , Inhibidores de Agregación Plaquetaria , Receptor PAR-1/antagonistas & inhibidores , Receptores Purinérgicos P2Y12 , Plaquetas/química , Plaquetas/metabolismo , Inhibidores de la Ciclooxigenasa/química , Inhibidores de la Ciclooxigenasa/farmacocinética , Inhibidores de la Ciclooxigenasa/farmacología , Evaluación Preclínica de Medicamentos/métodos , Estabilidad de Medicamentos , Ácidos Grasos Insaturados/química , Ácidos Grasos Insaturados/farmacocinética , Ácidos Grasos Insaturados/farmacología , Humanos , Inhibidores de Agregación Plaquetaria/química , Inhibidores de Agregación Plaquetaria/farmacocinética , Inhibidores de Agregación Plaquetaria/farmacología , Antagonistas del Receptor Purinérgico P2Y/química , Antagonistas del Receptor Purinérgico P2Y/farmacocinética , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptor PAR-1/metabolismo
4.
Br J Cancer ; 117(5): 695-703, 2017 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-28697175

RESUMEN

BACKGROUND: Selective platelet release of pro- or anti-angiogenic factors distinctly regulated angiogenesis. We hypothesised that selective release of platelet angiogenic factors could differently regulate tumour growth. METHODS: Breast cancer cell proliferation, cancer cell-induced endothelial tube formation in vitro, and tumour growth in vivo were studied in the presence of protease-activated receptor 1-stimulated platelet releasate (PAR1-PR; rich in pro-angiogenic factors) or PAR4-PR (rich in anti-angiogenic factors). RESULTS: The PAR1-PR and PAR4-PR supplementation (10%) similarly enhanced cell proliferation of MCF-7 and MDA-MB-231 breast cancer cells. The cancer cells triggered capillary-like tube formation of endothelial cells that was further enhanced by pro-angiogenic factor-rich PAR1-PR. The VEGF, but not SDF-1α, receptor blockade abolished PAR1-PR/PAR4-PR-enhanced cancer cell proliferation. Integrin blockade by RGDS had identical effects as VEGF inhibition. The Src and ERK inhibition diminished, whereas PI3K and PKC blockade abolished platelet releasate-enhanced cancer cell proliferation. Using a model of subcutaneous implantation of MDA-MB-231 cells in nude mice, PAR1-PR enhanced tumour growth more markedly than PAR4-PR, and seemed to achieve the exaggeration by promoting more profound tumour angiogenesis. CONCLUSIONS: Platelet releasate increases breast cancer cell proliferation through VEGF-integrin cooperative signalling. Pro-angiogenic factor-rich platelet releasate enhances cancer cell-induced angiogenesis more markedly, and thus exaggerates tumour growth in vivo.


Asunto(s)
Plaquetas/metabolismo , Neoplasias de la Mama/metabolismo , Integrinas/metabolismo , Neovascularización Patológica/metabolismo , Receptor PAR-1/metabolismo , Receptores de Trombina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adulto , Animales , Plaquetas/efectos de los fármacos , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/patología , Proliferación Celular , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrinas/antagonistas & inhibidores , Células MCF-7 , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Oligopéptidos/farmacología , Compuestos de Fenilurea/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Quinasa C/metabolismo , Quinolinas/farmacología , Receptores CXCR4/antagonistas & inhibidores , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
5.
PLoS One ; 10(2): e0118057, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25693178

RESUMEN

Increased vascular permeability leading to acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) is central to the pathogenesis of heatstroke. Protease-activated receptor 1 (PAR1), the receptor for thrombin, plays a key role in disruption of endothelial barrier function in response to extracellular stimuli. However, the role of PAR1 in heat stress-induced endothelial hyper-permeability is unknown. In this study, we measured PAR1 protein expression in heat-stressed human umbilical venous endothelial cells (HUVECs), investigated the influences of PAR1 on endothelial permeability, F-actin rearrangement, and moesin phosphorylation by inhibiting PAR1 with its siRNA, neutralizing antibody (anti-PAR1), specific inhibitor(RWJ56110), and Xuebijing injection (XBJ), a traditional Chinese medicine used for sepsis treatment, and evaluated the role of PAR1 in heatstroke-related ALI/ARDS in mice by suppressing PAR1 with RWJ56110, anti-PAR1and XBJ. We found that heat stress induced PAR1 protein expression 2h after heat stress in endothelial cells, caused the release of endothelial matrix metalloprotease 1, an activator of PAR1, after 60 or 120 min of heat stimulation, as well as promoted endothelial hyper-permeability and F-actin rearrangement, which were inhibited by suppressing PAR1 with RWJ56110, anti-PAR1 and siRNA. PAR1 mediated moesin phosphorylation, which caused F-actin rearrangement and disruption of endothelial barrier function. To corroborate findings from in vitro experiments, we found that RWJ56110 and the anti-PAR1 significantly decreased lung edema, pulmonary microvascular permeability, protein exudation, and leukocytes infiltrations in heatstroke mice. Additionally, XBJ was found to suppress PAR1-moesin signal pathway and confer protective effects on maintaining endothelial barrier function both in vitro and in vivo heat-stressed model, similar to those observed above with the inhibition of PAR1. These results suggest that PAR1 is a potential therapeutic target in heatstroke.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Calor , Receptor PAR-1/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/metabolismo , Animales , Western Blotting , Lavado Broncoalveolar , Permeabilidad Capilar/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/metabolismo , Transducción de Señal/efectos de los fármacos
6.
Neural Plast ; 2015: 646595, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26783471

RESUMEN

Matrix metalloproteinases (MMPs) are zinc-dependent endopeptidases that play a role in varied forms of developmental and postnatal neuroplasticity. MMP substrates include protease-activated receptor-1 (PAR-1), a G-protein coupled receptor expressed in hippocampus. We examined proliferation and differentiation of adult neural progenitor cells (aNPCs) from hippocampi of mice that overexpress the potent PAR-1 agonist MMP-1. We found that, as compared to aNPCs from littermate controls, MMP-1 tg aNPCs display enhanced proliferation. Under differentiating conditions, these cells give rise to a higher percentage of MAP-2(+) neurons and a reduced number of oligodendrocyte precursors, and no change in the number of astrocytes. The fact that these results are MMP and PAR-1 dependent is supported by studies with distinct antagonists. Moreover, JSH-23, an inhibitor of NF-κB p65 nuclear translocation, counteracted both the proliferation and differentiation changes seen in MMP-1 tg-derived NPCs. In complementary studies, we found that the percentage of Sox2(+) undifferentiated progenitor cells is increased in hippocampi of MMP-1 tg animals, compared to wt mice. Together, these results add to a growing body of data suggesting that MMPs are effectors of hippocampal neuroplasticity in the adult CNS and that the MMP-1/PAR-1 axis may play a role in neurogenesis following physiological and/or pathological stimuli.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Hipocampo/fisiología , Metaloproteinasa 13 de la Matriz/metabolismo , Células-Madre Neurales/fisiología , Receptor PAR-1/metabolismo , Animales , Hipocampo/metabolismo , Masculino , Metaloproteinasa 13 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/metabolismo , Factores de Transcripción SOXB1/metabolismo
7.
J Surg Res ; 194(2): 375-382, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25527361

RESUMEN

BACKGROUND: Recombinant human soluble thrombomodulin (rTM) protects against disseminated intravascular coagulopathy by inhibiting coagulation, inflammation, and apoptosis. This study tests the hypothesis that rTM is hepatoprotective after extensive hepatectomy (Hx) and investigates the mechanisms underlying this effect. MATERIALS AND METHODS: Experiment 1: rats (15 per group) were injected with rTM (1.0 or 2.0 mg/kg) or saline just before 95% Hx and their 7-d survival assessed. Experiment 2: rats were assigned to either a treated (2.0 mg/kg rTM just before Hx) or control group (n = 5 per group). Five rats per group were euthanized immediately after surgery, and at 1, 3, 6, 12, and 24 h postoperatively; serum and liver remnant samples were collected for biochemical and histologic analysis, as well as reverse-transcription polymerase chain reaction and Western blotting. RESULTS: All saline-injected rats died within 52 h of Hx, whereas injection of 2.0 mg/kg rTM prolonged survival (P = 0.003). rTM increased the number of Ki67-positive cells and reduced the number of terminal deoxynucleotidyl transferase dUTP nick-end labeling-positive cells. The number of myeloperoxidase-positive cells and the expression of high-mobility group box 1 protein did not differ. Reverse-transcription polymerase chain reaction revealed that rTM significantly enhanced protease-activated receptor-1 and sphingosine kinase 1 messenger RNA expression and significantly reduced plasminogen activator inhibitor-1 and Bax messenger RNA expression. Immunohistochemistry and Western blotting demonstrated that protease-activated receptor-1 expression 24 h after Hx was significantly higher in rTM-treated than in control rats. CONCLUSIONS: rTM may improve survival after extensive Hx by inhibiting apoptosis and promoting liver regeneration.


Asunto(s)
Hepatectomía/efectos adversos , Fallo Hepático/prevención & control , Regeneración Hepática/efectos de los fármacos , Complicaciones Posoperatorias/prevención & control , Trombomodulina/uso terapéutico , Alanina Transaminasa/sangre , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Evaluación Preclínica de Medicamentos , Hepatectomía/mortalidad , Hepatocitos/efectos de los fármacos , Inmunohistoquímica , Fallo Hepático/etiología , Masculino , Complicaciones Posoperatorias/etiología , Ratas Wistar , Receptor PAR-1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
J Mol Biol ; 426(4): 881-93, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24316004

RESUMEN

Activation of platelets by the serine protease thrombin is a critical event in haemostasis. This process involves the binding of thrombin to glycoprotein Ibα (GpIbα) and cleavage of protease-activated receptors (PARs). The N-terminal extracellular domain of GpIbα contains an acidic peptide stretch that has been identified as the main thrombin binding site, and both anion binding exosites of thrombin have been implicated in GpIbα binding, but it remains unclear how they are involved. This issue is of critical importance for the mechanism of platelet activation by thrombin. If both exosites bind to GpIbα, thrombin could potentially act as a platelet adhesion molecule or receptor dimerisation trigger. Alternatively, if only a single site is involved, GpIbα may serve as a cofactor for PAR-1 activation by thrombin. To determine the involvement of thrombin's two exosites in GpIbα binding, we employed the complementary methods of mutational analysis, binding studies, X-ray crystallography and NMR spectroscopy. Our results indicate that the peptide corresponding to the C-terminal portion of GpIbα and the entire extracellular domain bind exclusively to thrombin's exosite II. The interaction of thrombin with GpIbα thus serves to recruit thrombin activity to the platelet surface while leaving exosite I free for PAR-1 recognition.


Asunto(s)
Complejo GPIb-IX de Glicoproteína Plaquetaria/química , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Trombina/química , Trombina/metabolismo , Clorometilcetonas de Aminoácidos/química , Antitrombinas/química , Sitios de Unión , Cristalografía por Rayos X , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Conformación Proteica , Receptor PAR-1/metabolismo , Trombina/genética
9.
Chin J Integr Med ; 20(2): 116-22, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23504578

RESUMEN

OBJECTIVE: To investigate the neuro-protective effects of baicalin in Wistar rats with focal cerebral ischemic reperfusion injury. METHODS: Ninety adult male Wistar rats weighing 320-350 g were randomly divided into the following groups (n=5): (a) sham control group; (b) vehicle group, subjected to middle cerebral artery occlusion and received vehicle intraperitoneally; (c-e) baicalin groups, which were subjected to the middle cerebral artery occlusion and treated with baicalin 25, 50 and 100 mg/kg, respectively. The neurological scores were determined at postoperative 1, 3 and 7 d after the treatment. The expression of protease-activated receptor-1 (PAR-1), PAR-1 mRNA and Caspase-3 were determined using Western blot, reverse transcription polymerase chain reaction (RTPCR) analysis and immunohistochemistry, respectively. RESULTS: Significant decrease was noted in the neurological score in the baicalin group compared with that of the vehicle group (P<0.01). Additionally, down-regulation of PAR-1 mRNA, PAR-1 and Caspase-3 was observed in the baicalin groups compared with those obtained from the vehicle group (P<0.01). Compared with the low-dose baicalin group (25 mg/kg), remarkable decrease was noted in neurological score, and the expression of PAR-1 mRNA, PAR-1 as well as Caspase-3 in the high-dose group (P<0.05). CONCLUSION: Baicalin showed neuro-protective effects in focal cerebral ischemic reperfusion injury through inhibiting the expression of PAR-1 and apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Flavonoides/farmacología , Flavonoides/uso terapéutico , Receptor PAR-1/antagonistas & inhibidores , Daño por Reperfusión/tratamiento farmacológico , Animales , Isquemia Encefálica/complicaciones , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Caspasa 3/metabolismo , Flavonoides/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Daño por Reperfusión/complicaciones , Daño por Reperfusión/genética , Daño por Reperfusión/patología
10.
J Lipid Res ; 54(11): 3085-97, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23883581

RESUMEN

Oxidized phospholipids (oxPLs) generated nonenzymatically display pleiotropic biological actions in inflammation. Their generation by cellular cyclooxygenases (COXs) is currently unknown. To determine whether platelets generate prostaglandin (PG)-containing oxPLs, then characterize their structures and mechanisms of formation, we applied precursor scanning-tandem mass spectrometry to lipid extracts of agonist-activated human platelets. Thrombin, collagen, or ionophore activation stimulated generation of families of PGs comprising PGE2 and D2 attached to four phosphatidylethanolamine (PE) phospholipids (16:0p/, 18:1p/, 18:0p/, and 18:0a/). They formed within 2 to 5 min of activation in a calcium, phospholipase C, p38 MAP kinases, MEK1, cPLA2, and src tyrosine kinase-dependent manner (28.1 ± 2.3 pg/2 × 108 platelets). Unlike free PGs, they remained cell associated, suggesting an autocrine mode of action. Their generation was inhibited by in vivo aspirin supplementation (75 mg/day) or in vitro COX-1 blockade. Inhibitors of fatty acyl reesterification blocked generation significantly, while purified COX-1 was unable to directly oxidize PE in vitro. This indicates that they form in platelets via rapid esterification of COX-1 derived PGE2/D2 into PE. In summary, COX-1 in human platelets acutely mediates membrane phospholipid oxidation via formation of PG-esterified PLs in response to pathophysiological agonists.


Asunto(s)
Aspirina/farmacología , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Ciclooxigenasa 1/metabolismo , Inhibidores de la Ciclooxigenasa/farmacología , Fosfolípidos/metabolismo , Prostaglandinas/metabolismo , Plaquetas/fisiología , Calcio/metabolismo , Dinoprostona/metabolismo , Relación Dosis-Respuesta a Droga , Esterificación/efectos de los fármacos , Retroalimentación Fisiológica/efectos de los fármacos , Humanos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , MAP Quinasa Quinasa 1/metabolismo , Fosfatidiletanolaminas/metabolismo , Activación Plaquetaria/efectos de los fármacos , Prostaglandina D2/metabolismo , Proteína Quinasa C/metabolismo , Receptor PAR-1/metabolismo , Trombina/metabolismo , Familia-src Quinasas/metabolismo
11.
Neuroreport ; 23(14): 814-8, 2012 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-22889888

RESUMEN

The activation of protease-activated receptor 1 (PAR1) in cultured rat hippocampal neurons triggers synaptic retrograde signaling through the endocannabinoid 2-arachidonoylglycerol, thereby activating the cannabinoid receptor 1 (CB1R). CB1R is a metabotropic receptor activated by marihuana and endocannabinoids that suppresses neurotransmitter release. Also, activation of the CB1R increases rapid eye movement sleep (REMS) and food intake. The lateral hypothalamus is a crucial structure to modulate both feeding and waking. To evaluate the effect of PAR1 stimulation in the lateral hypothalamus on food intake and on the sleep-waking cycle, we implanted rats with electrodes, for recording sleep, and cannulae, to administer S1820, a selective PAR1 agonist peptide, bilaterally into the lateral hypothalamus. To determine whether the effects induced by PAR1 stimulation were mediated by CB1R activation, we administered AM251, a CB1R inverse agonist, to block S1820 effects. Our results show that the stimulation of PAR1 into the lateral hypothalamus increases both food intake and REMS and such effects were prevented by AM251, indicating that PAR1 modulates both food intake and the sleep-waking cycle, in the lateral hypothalamus, through CB1R activation. This study shows novel behavioral changes induced by PAR1 activation and further supports the notion that endocannabinoids are food intake and REMS promoters.


Asunto(s)
Regulación del Apetito/fisiología , Hipotálamo/metabolismo , Receptor Cannabinoide CB1/metabolismo , Receptor PAR-1/metabolismo , Sueño REM/fisiología , Animales , Ingestión de Alimentos/fisiología , Masculino , Ratas , Ratas Wistar
12.
J Endocrinol ; 214(2): 165-75, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22593082

RESUMEN

Progesterone and progestins have been demonstrated to enhance breast cancer cell migration, although the mechanisms are still not fully understood. The protease-activated receptors (PARs) are a family of membrane receptors that are activated by serine proteases in the blood coagulation cascade. PAR1 (F2R) has been reported to be involved in cancer cell migration and overexpressed in breast cancer. We herein demonstrate that PAR1 mRNA and protein are upregulated by progesterone treatment of the breast cancer cell lines ZR-75 and T47D. This regulation is dependent on the progesterone receptor (PR) but does not require PR phosphorylation at serine 294 or the PR proline-rich region mPRO. The increase in PAR1 mRNA was transient, being present at 3  h and returning to basal levels at 18  h. The addition of a PAR1-activating peptide (aPAR1) to cells treated with progesterone resulted in an increase in focal adhesion (FA) formation as measured by the cellular levels of phosphorylated FA kinase. The combined but not individual treatment of progesterone and aPAR1 also markedly increased stress fiber formation and the migratory capacity of breast cancer cells. In agreement with in vitro findings, data mining from the Oncomine platform revealed that PAR1 expression was significantly upregulated in PR-positive breast tumors. Our observation that PAR1 expression and signal transduction are modulated by progesterone provides new insight into how the progestin component in hormone therapies increases the risk of breast cancer in postmenopausal women.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma/patología , Movimiento Celular/efectos de los fármacos , Adhesiones Focales/efectos de los fármacos , Progesterona/farmacología , Receptor PAR-1/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Evaluación Preclínica de Medicamentos , Femenino , Adhesiones Focales/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes erbB-1/fisiología , Humanos , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 1/fisiología , Posmenopausia/genética , Posmenopausia/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/fisiología , Receptor PAR-1/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
13.
Clin Appl Thromb Hemost ; 17(6): E165-70, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21406410

RESUMEN

Ankaferd Blood Stopper (ABS) is a novel topical hemostatic agent with pleiotropic actions indicated in clinical hemorrhages. Protease-activated receptor 1 (PAR-1) is located in the crossroads of hemostasis, inflammation, infection, apoptosis and tumorigenesis. ABS-induced formation of the protein network with vital erythroid aggregation covers the entire physiological hemostatic process. The aim of this study is to assess the effects of ABS on PAR-1 in the Human Umbilical Vein Endothelial Cells (HUVEC) model, in relation to the "ipopolysaccharides (LPS)-challenge" to endothelium. For this purpose, ABS 10 µL and 100 µL, had been applied to HUVEC within the time periods of 5 minutes (min), 25 min, 50 min, 6 hours (h) and 24 h. The cells have lifted from the plastic surface and adhered to each other during theABSapplication to the HUVECs. After 24 hours the cells returned to normal baseline level. We observed dose-dependent reversible PAR-1 down-regulation mediated by ABS inside the human umbilical vein endothelial cells. ABS-induced sustained PAR-1 down-regulation in the presence of LPS. Those findings indicated that ABS hemostatic agent may act as a topical biological response modifier by acting on PAR-1 at the vascular endothelial and cellular level.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Lipopolisacáridos/farmacología , Extractos Vegetales/farmacología , Receptor PAR-1/metabolismo , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Receptor PAR-1/genética
14.
Rejuvenation Res ; 13(1): 27-37, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20230276

RESUMEN

Current immunosuppressive treatments for central nervous system demyelinating diseases fail to prevent long-term motor and cognitive decline in patients. Excitingly, glial cell transplantation arises as a promising complementary strategy to challenge oligodendrocytes loss occurring in myelination disorders. A potential source of new oligodendrocytes is the subventricular zone (SVZ) pool of multipotent neural stem cells. However, this approach has been handicapped by the lack of functional methods for identification and pharmacological analysis of differentiating oligodendrocytes, prior to transplantation. In this study, we questioned whether SVZ-derived oligodendrocytes could be functionally discriminated due to intracellular calcium level ([Ca(2+)](i)) variations following KCl, histamine, and thrombin stimulations. Previously, we have shown that SVZ-derived neurons and immature cells can be discriminated on the basis of their selective [Ca(2+)](i) rise upon KCl and histamine stimulation, respectively. Herein, we demonstrate that O4+ and proteolipid protein-positive (PLP+) oligodendrocytes do not respond to these stimuli, but display a robust [Ca(2+)](i) rise following thrombin stimulation, whereas other cell types are thrombin-insensitive. Thrombin-induced Ca(2+) increase in oligodendrocytes is mediated by protease-activated receptor-1 (PAR-1) activation and downstream signaling through G(q/11) and phospholipase C (PLC), resulting in Ca(2+) recruitment from intracellular compartments. This method allows the analysis of functional properties of oligodendrocytes in living SVZ cultures, which is of major interest for the development of effective grafting strategies in the demyelinated brain. Additionally, it opens new perspectives for the search of new pro-oligodendrogenic factors to be used prior grafting.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Neuronas/citología , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos , Trombina/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Ventrículos Cerebrales/citología , Histamina/farmacología , Ratones , Ratones Endogámicos C57BL , Oligodendroglía/metabolismo , Cloruro de Potasio/farmacología , Receptor PAR-1/metabolismo , Células Madre/metabolismo
15.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 30(12): 1302-5, 2010 Dec.
Artículo en Chino | MEDLINE | ID: mdl-21302496

RESUMEN

OBJECTIVE: To investigate the protective effect and mechanism of baicalin on nerve tissue in rat with intracerebral hemorrhage (ICH). METHODS: Rats were randomly divided into five groups: the sham-operated group, the ICH model group, and the three baicalin treated groups treated respectively with small, medium and large doses of baicalin. ICH rat model was established by injecting collagenase VII into caudate nucleus. Baicalin was given by peritoneal injection to the baicalin treated groups, and saline was given to the other two groups once a day started from 2 h after modeling. Animals were sacrificed in batches on the 1st, 3rd, 5th and 10th day of treatment to take their brains for detecting protease-activated receptor-1 (PAR-1) expression and cell apoptosis in brain tissue surrounding hematoma by Western blot and TUNEL method, respectively. And the water content of brain was estimated by dry-wet weight method. RESULTS: Compared with the model group, the PAR-1 expression and TUNEL-positive cells were significantly reduced in the baicalin treated groups; and brain edema was also significantly reduced (P<0.01). CONCLUSIONS: The up-regulated PAR-1 expression after ICH in rats might play an important role in inducing cell apoptosis and brain edema. Baicalin shows significant protective effect on ICH rats, which may be related to its effects in inhibiting PAR-1 expression and decreasing apoptosis cells, so as to reduce brain edema.


Asunto(s)
Encéfalo/patología , Hemorragia Cerebral/tratamiento farmacológico , Flavonoides/uso terapéutico , Fitoterapia , Receptor PAR-1/metabolismo , Animales , Apoptosis/efectos de los fármacos , Encéfalo/metabolismo , Depresión Química , Flavonoides/farmacología , Masculino , Ratas , Ratas Wistar
16.
Neurogastroenterol Motil ; 22(4): 431-8, e105, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19719510

RESUMEN

BACKGROUND: Protease-activated receptors (PARs), a family member of G-protein coupled receptors, are present and functionally active in a wide variety of cells. The object of this study was to demonstrate the presence and function of PAR-1 and PAR-2 in the dorsal motor nucleus of the vagus (DMV). METHODS: DMNV neurons were isolated from neonatal rat brainstems using micro-dissection and enzymatic digestion. Neurons were cultured in Neurobasal medium A containing 2% B27 supplement. Intracellular calcium concentration ([Ca(2+)](i)) was measured using fura-2 based microspectrometry. Expression of PARs was detected by RT-PCR and immunofluorescent staining. KEY RESULT: Thrombin and PAR-1 agonist peptide activate PAR-1 with a maximum change in [Ca(2+)](i) expressed as DeltaF/F0 of 229 +/- 14% and 137 +/- 7%, respectively. Trypsin and PAR-2 agonist peptide activate PAR-2 with a maximum DeltaF/F0 change of 258 +/- 12% and 242 +/- 10%, respectively. Inhibition of phospholipase C (PLC) by U73312 (1 microm) decreased the maximal change in DeltaF/F0 induced by PAR-1 activation from 140 +/- 17% to 21 +/- 3%, while the PAR-2-mediated maximal change in DeltaF/F0 decreased from 185 +/- 21% to 19 +/- 6%. Blockade of IP3 receptor with 2APB inhibited the maximal change in DeltaF/F0 due to PAR-1 and PAR-2 activation by 72 +/- 13% and 71 +/- 20% respectively. PAR-1 immnuoreactivity was present in DMV neurons. Increase in transcripts for PAR-1 and PAR-2 were detected in DMV tissues derived from IBD rats relative to control animals. CONCLUSIONS & INFERENCES: Our results indicate that PAR-1 and PAR-2 are present in the DMV neurons, and their activation leads to increases in intracellular calcium via signal transduction mechanism that involves activation of PLC and the production of IP3.


Asunto(s)
Tronco Encefálico/metabolismo , Neuronas/metabolismo , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Nervio Vago/metabolismo , Análisis de Varianza , Animales , Calcio/metabolismo , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Neuronas/citología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor PAR-1/genética , Receptor PAR-2/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología
17.
Stroke ; 40(6): 2199-204, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19359644

RESUMEN

BACKGROUND AND PURPOSE: To reduce bleeding and damage to central nervous system tissue in intracerebral hemorrhage, the coagulant effect of thrombin is essential. However, thrombin itself can kill neurons in intracerebral hemorrhage as can the matrix metalloproteinases (MMPs), which are also elevated in this condition, in part due to thrombin-mediated activation of MMPs. It is thus important to understand and block the neurotoxic effects of thrombin without inhibiting its therapeutic outcomes. In this study, we have investigated the relative roles of proteinase activated receptor-1, a thrombin receptor, and MMPs in brain injury induced by thrombin or blood. METHODS: Mice were subjected to stereotactic intracerebral injections of saline, thrombin, and autologous blood, with or without hirudin, a thrombin inhibitor, or GM6001, an MMP inhibitor. Twenty-four hours later, tissue sections were obtained to evaluate the area of brain damage and extent of dying neurons. Data from wild-type mice were compared with results obtained with proteinase activated receptor-1 null mice. RESULTS: In blood-induced damage to the brain parenchyma, both hirudin and GM6001 significantly reduced injury to a comparable extent (>40%) implicating both thrombin and MMPs in neurotoxicity. In proteinase activated receptor-1 null mice, blood-induced brain damage was reduced by 22.6% relative to wild-type animals; by comparison, the blood-induced brain damage was reduced by 48.3% using GM6001. CONCLUSIONS: The neurotoxicity of blood in intracerebral hemorrhage involves both proteinase activated receptor-1 and MMP activation, with the latter appearing more prominent in causing death.


Asunto(s)
Hemorragia Cerebral/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Síndromes de Neurotoxicidad/prevención & control , Receptor PAR-1/metabolismo , Trombina/antagonistas & inhibidores , Trombina/toxicidad , Animales , Antitrombinas/uso terapéutico , Transfusión de Sangre Autóloga , Encéfalo/patología , Muerte Celular/efectos de los fármacos , Hemorragia Cerebral/patología , Dipéptidos/farmacología , Femenino , Gelatina/química , Hirudinas , Masculino , Inhibidores de la Metaloproteinasa de la Matriz , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Síndromes de Neurotoxicidad/patología , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/uso terapéutico , Receptor PAR-1/genética
18.
Oncol Rep ; 21(5): 1261-7, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19360302

RESUMEN

Thrombin has been recently demonstrated to promote hepatocellular carcinoma (HCC) cell migration by activation of the proteinase-activated receptor (PAR) subtypes PAR1 and PAR4 suggesting a role of these proteinase-receptor systems in HCC progression. In this study, we investigated the effect of (-)-epigallocatechin-3-gallate (EGCG), the major polyphenolic compound of green tea on thrombin-PAR1/PAR4-mediated hepatocellular carcinoma cell invasion and p42/p44 MAPKinase activation. In this study we used the permanent liver carcinoma cell line HEP-3B and two primary cultures established from surgically resected HCCs. We found that stimulation of HCC cells with thrombin, the PAR1-selective activating peptide, TFLLRN-NH2, and the PAR4-selective activating peptide, AYPGKF-NH2, increased cell invasion across a Matrigel-coated membrane barrier and stimulated activation of p42/p44 MAPKinase phosphorylation. Both the effects on p42/p44 MAPKinases, and on cell invasiveness induced by thrombin and the PAR1/4 subtype-selective agonist peptides were effectively blocked by EGCG. The results clearly identify EGCG as a potent inhibitor of the thrombin-PAR1/PAR4-p42/p44 MAPKinase invasive signaling axis in hepatocellular carcinoma cells as a previously unrecognized mode of action for EGCG in cancer cells. Moreover, the results suggest that (-)-epigal-locatechin-3-gallate might have therapeutic potential for hepatocellular carcinoma.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Catequina/análogos & derivados , Neoplasias Hepáticas/tratamiento farmacológico , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Trombina/antagonistas & inhibidores , Anticarcinógenos/farmacología , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/patología , Catequina/farmacología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Invasividad Neoplásica , Oligopéptidos/farmacología , Fosforilación , Receptor PAR-1/agonistas , Receptor PAR-1/antagonistas & inhibidores , Receptor PAR-1/metabolismo , Receptores de Trombina/agonistas , Receptores de Trombina/antagonistas & inhibidores , Receptores de Trombina/metabolismo , Té/química , Trombina/farmacología
19.
Integr Cancer Ther ; 7(4): 282-94, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19116224

RESUMEN

The supporting role of proteases in tumor progression and invasion is well known; however, the use of proteases as therapeutic agents has also been demonstrated. In this article, the authors report on the differential effects of exogenous serine proteases on the motility of tumor and normal cells. The treatment of normal and tumor cells with a single dose of pancreatic serine proteases, trypsin (TR) and chymotrypsin (CH), leads to a concentration-dependent response by cells, first accelerating and then slowing mobility. Tumor cells are 10 to 20 times more sensitive to exogenous TR/CH, suggesting that a single dose of proteases may cause discordant movements of normal and tumor cells within the tumor environment. The inhibitory effects of TR on cell motility are contradicted by thrombin (TH), particularly in the regulation of normal cells' migration. The purpose of this investigation was to ascertain the role of protease-activated receptors (PARs) in terms of normal and tumor cell motility. Duplicate treatments with proteases resulted in diminished mobility of both normal and tumor cells. Repeated application of TR and TH in 1-hour treatment intervals initially desensitizes cell surface PARs. However, cell surface PARs reappear regardless of subsequent protease treatments in both normal and tumor cells. The resensitization process is retarded in tumor cells when compared with normal cells. This is evidenced by lower expression of PARs as well as by their relocalization at the tumor cell surfaces. Under these conditions, normal cells remain responsive to exogenous proteases in terms of cell motility. Exogenous proteases do not modulate motility of repeatedly stimulated tumor cells, and consequently, the migration of tumor cells appears disconnected from the PAR signaling pathways. The use of activating peptides in lieu of the cognate proteases for a given PAR system indicated that proteases may act through additional targets not regulated by PAR signaling. We hypothesize that the divergent migration patterns of normal and tumor cells due to exposure to proteases is in part mediated by PARs. Thus, treatment with exogenous proteases may cause rearrangement of the tumor and stromal cells within the tumor microenvironment. Such topographical effects may lead to the inhibition of tumor progression and metastasis development.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Neoplasias/patología , Serina Endopeptidasas/farmacología , Animales , Bovinos , Línea Celular , Línea Celular Tumoral , Quimotripsina/metabolismo , Quimotripsina/farmacología , Perros , Precursores Enzimáticos/metabolismo , Precursores Enzimáticos/farmacología , Expresión Génica , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Oligopéptidos/farmacología , Fragmentos de Péptidos/farmacología , Ratas , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Receptor PAR-2/genética , Receptor PAR-2/metabolismo , Serina Endopeptidasas/metabolismo , Trombina/metabolismo , Trombina/farmacología , Tripsina/metabolismo , Tripsina/farmacología
20.
J Biol Chem ; 280(23): 22172-80, 2005 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-15817447

RESUMEN

Thrombospondin-1 (THBS1) is a large extracellular matrix glycoprotein that affects vasculature systems such as platelet activation, angiogenesis, and wound healing. Increases in THBS1 expression have been liked to disease states including tumor progression, atherosclerosis, and arthritis. The present study focuses on the effects of thrombin activation of the G-protein-coupled, protease-activated receptor-1 (PAR-1) on THBS1 gene expression in the microvascular endothelium. Thrombin-induced changes in gene expression were characterized by microarray analysis of approximately 11,000 different human genes in human microvascular endothelial cells (HMEC-1). Thrombin induced the expression of a set of at least 65 genes including THBS1. Changes in THBS1 mRNA correlated with an increase in the extracellular THBS1 protein concentration. The PAR-1-specific agonist peptide (TFLLRNK-PDK) mimicked thrombin stimulation of THBS1 expression, suggesting that thrombin signaling is through PAR-1. Further studies showed THBS1 expression was sensitive to pertussis toxin and protein kinase C inhibition indicating G(i/o)- and G(q)-mediated pathways. THBS1 up-regulation was also confirmed in human umbilical vein endothelial cells stimulated with thrombin. Analysis of the promoter region of THBS1 and other genes of similar expression profile identified from the microarray predicted an EBOX/EGRF transcription model. Expression of members of each family, MYC and EGR1, respectively, correlated with THBS1 expression. These results suggest thrombin formed at sites of vascular injury increases THBS1 expression into the extracellular matrix via activation of a PAR-1, G(i/o), G(q), EBOX/EGRF-signaling cascade, elucidating regulatory points that may play a role in increased THBS1 expression in disease states.


Asunto(s)
Endotelio Vascular/citología , Regulación de la Expresión Génica , Microcirculación/metabolismo , Trombina/química , Trombospondina 1/biosíntesis , Adenosina Difosfato/química , Algoritmos , Amidas/farmacología , Apoptosis , Células Cultivadas , Análisis por Conglomerados , Medios de Cultivo , Cartilla de ADN/química , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Impedancia Eléctrica , Endotelio Vascular/metabolismo , Ensayo de Inmunoadsorción Enzimática , Matriz Extracelular/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Humanos , Indoles/farmacología , Maleimidas/farmacología , Modelos Biológicos , Hibridación de Ácido Nucleico , Análisis de Secuencia por Matrices de Oligonucleótidos , Péptidos/química , Toxina del Pertussis/farmacología , Regiones Promotoras Genéticas , Unión Proteica , Piridinas/farmacología , ARN/metabolismo , Receptor PAR-1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Trombina/metabolismo , Factores de Tiempo , Venas Umbilicales/citología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA