Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.388
Filtrar
1.
Sci Adv ; 10(27): eado2365, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38959302

RESUMEN

Pityriasis rubra pilaris (PRP) is a rare inflammatory skin disease with a poorly understood pathogenesis. Through a molecularly driven precision medicine approach and an extensive mechanistic pathway analysis in PRP skin samples, compared to psoriasis, atopic dermatitis, healed PRP, and healthy controls, we identified IL-1ß as a key mediator, orchestrating an NF-κB-mediated IL-1ß-CCL20 axis, including activation of CARD14 and NOD2. Treatment of three patients with the IL-1 antagonists anakinra and canakinumab resulted in rapid clinical improvement and reversal of the PRP-associated molecular signature with a 50% improvement in skin lesions after 2 to 3 weeks. This transcriptional signature was consistent with in vitro stimulation of keratinocytes with IL-1ß. With the central role of IL-1ß underscoring its potential as a therapeutic target, our findings propose a redefinition of PRP as an autoinflammatory keratinization disorder. Further clinical trials are needed to validate the efficacy of IL-1ß antagonists in PRP.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Proteína Antagonista del Receptor de Interleucina 1 , Interleucina-1beta , Queratinocitos , Pitiriasis Rubra Pilaris , Humanos , Pitiriasis Rubra Pilaris/tratamiento farmacológico , Pitiriasis Rubra Pilaris/patología , Pitiriasis Rubra Pilaris/genética , Interleucina-1beta/metabolismo , Interleucina-1beta/antagonistas & inhibidores , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Queratinocitos/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/patología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Masculino , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/antagonistas & inhibidores , Femenino , Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteínas Adaptadoras de Señalización CARD/genética , Piel/patología , Piel/metabolismo , Piel/efectos de los fármacos , Interleucina-1/antagonistas & inhibidores , Interleucina-1/metabolismo , Interleucina-1/genética , Persona de Mediana Edad , Guanilato Ciclasa/metabolismo , Guanilato Ciclasa/antagonistas & inhibidores , Guanilato Ciclasa/genética , Adulto , Transducción de Señal/efectos de los fármacos , Proteínas de la Membrana
2.
J Med Chem ; 64(22): 16650-16674, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34781683

RESUMEN

CARM1 is a protein arginine methyltransferase and acts as a transcriptional coactivator regulating multiple biological processes. Aberrant expression of CARM1 has been related to the progression of multiple types of cancers, and therefore CARM1 was considered as a promising drug target. In the present work, we report the structure-based discovery of a series of N1-(3-(pyrimidin-2-yl)benzyl)ethane-1,2-diamines as potent CARM1 inhibitors, in which compound 43 displays high potency and selectivity. With the advantage of excellent tissue distribution, compound 43 demonstrated good in vivo efficacy for solid tumors. Furthermore, from the detailed immuno-oncology study with MC38 C57BL/6J xenograft model, we confirmed that this chemical probe 43 has profound effects in tumor immunity, which paves the way for future studies on the modulation of arginine post-translational modification that could be utilized in solid tumor treatment and cancer immunotherapy.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Adaptadoras de Señalización CARD/antagonistas & inhibidores , Descubrimiento de Drogas , Guanilato Ciclasa/antagonistas & inhibidores , Inmunoterapia/métodos , Neoplasias/terapia , Animales , Antineoplásicos/química , Proteínas Adaptadoras de Señalización CARD/metabolismo , Guanilato Ciclasa/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Neoplasias/inmunología , Procesamiento Proteico-Postraduccional , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Drug Des Devel Ther ; 14: 1583-1592, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32425504

RESUMEN

BACKGROUND: Non-small cell lung cancer (NSCLC) is a common cause of cancer-related deaths. This study identified the regulatory pattern of gallic acid in NSCLC. METHODS: Human NSCLC cells were treated with different doses of gallic acid, after which, MTT assay and flow cytometry were performed to determine the survival and apoptotic rate of human NSCLC cells. Then, co-immunoprecipitation assay was performed to analyze the relationships between gallic acid, epidermal growth factor receptor (EGFR), and CARM1-PELP1. Next, we analyzed whether PELP1, CARM1 and EGFR were associated with the effects of gallic acid on NSCLC cells by conducting rescue experiments. The expression pattern of phosphorylated EGFR, EGFR, Ki67, as well as Fas, FasL and Caspase 3 proteins in cancer cells or xenografts was measured by Western blot analysis. Lastly, the role of gallic acid in the tumor growth was assessed in nude mice. RESULTS: The ideal dose of gallic acid that presented good suppressive effect on NSCLC cells were 30 µM, 50 µM and 75 µM, respectively. Gallic acid played an inhibiting role in the activation of EGFR, which further reduced the formation of CARM1-PELP1 complex, ultimately repressed the proliferation and elevated apoptosis of NSCLC cells. Meanwhile, CARM1 repression led to decreased growth, proliferation and migration abilities of NSCLC cells. Animal experiments confirmed that gallic acid contributed to the inhibition of tumor growth in vivo. CONCLUSION: To sum up, gallic acid could potentially prevent NSCLC progression via inhibition of EGFR activation and impairment of the binding of CARM1 to PELP1, highlighting a novel therapy to dampen NSCLC progression.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Adaptadoras de Señalización CARD/antagonistas & inhibidores , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Proteínas Co-Represoras/antagonistas & inhibidores , Progresión de la Enfermedad , Ácido Gálico/farmacología , Guanilato Ciclasa/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Factores de Transcripción/antagonistas & inhibidores , Proteínas Adaptadoras de Señalización CARD/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Proteínas Co-Represoras/metabolismo , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Guanilato Ciclasa/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Relación Estructura-Actividad , Factores de Transcripción/metabolismo
4.
J Psychopharmacol ; 34(4): 429-440, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31913077

RESUMEN

BACKGROUND: Endocannabinoid neurotransmission in the bed nucleus of the stria terminalis is involved in the control of cardiovascular responses to stress. However, the local mechanisms involved is this regulation are not known. AIMS: The purpose of this study was to assess an interaction of bed nucleus of the stria terminalis endocannabinoid neurotransmission with local nitrergic signaling, as well as to investigate the involvement of local N-methyl-D-aspartate glutamate receptor and nitric oxide signaling in the control of cardiovascular responses to acute restraint stress by bed nucleus of the stria terminalis endocannabinoid neurotransmission in rats. METHODS: The first protocol evaluated the effect of intra-bed nucleus of the stria terminalis microinjection of the selective cannabinoid receptor type 1 receptor antagonist AM251 in nitrite/nitrate content in the bed nucleus of the stria terminalis following restraint stress. The other protocols evaluated the impact of local pretreatment with the selective N-methyl-D-aspartate glutamate receptor antagonist LY235959, the selective neuronal nitric oxide synthase inhibitor Nω-propyl-L-arginine, the soluble guanylate cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, or the protein kinase G inhibitor KT5823 in restraint-evoked cardiovascular changes following bed nucleus of the stria terminalis treatment with AM251. RESULTS: Bilateral microinjection of AM251 into the bed nucleus of the stria terminalis increased local nitric oxide release during restraint stress. Bed nucleus of the stria terminalis treatment with the cannabinoid receptor type 1 receptor antagonist also enhanced the tachycardia caused by restraint stress, but without affecting arterial pressure increase and sympathetic-mediated cutaneous vasoconstriction. The facilitation of restraint-evoked tachycardia following bed nucleus of the stria terminalis treatment with the cannabinoid receptor type 1 receptor antagonist was completely inhibited by local pretreatment with LY235959, Nω-propyl-L-arginine, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, or KT5823. CONCLUSIONS: Our results provide evidence that bed nucleus of the stria terminalis endocannabinoid neurotransmission inhibits local N-methyl-D-aspartate/neuronal nitric oxide synthase/soluble guanylate cyclase/protein kinase G signaling, and this mechanism is involved in the control of the cardiovascular responses to stress.


Asunto(s)
Hemodinámica/efectos de los fármacos , Receptor Cannabinoide CB1/efectos de los fármacos , Núcleos Septales/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Estrés Psicológico/complicaciones , Estrés Psicológico/tratamiento farmacológico , Animales , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de GMP Cíclico/efectos de los fármacos , Guanilato Ciclasa/antagonistas & inhibidores , Guanilato Ciclasa/efectos de los fármacos , Masculino , Microinyecciones , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo I/efectos de los fármacos , Piperidinas/administración & dosificación , Piperidinas/farmacología , Pirazoles/administración & dosificación , Pirazoles/farmacología , Ratas , Ratas Wistar , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Restricción Física , Transmisión Sináptica/efectos de los fármacos
6.
J Virol ; 93(24)2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31554683

RESUMEN

Host factors play multiple essential roles in the replication and pathogenesis of mammalian neurotropic viruses. However, the cellular proteins of the central nervous system (CNS) involved in avian neurotropic virus infection have not been completely elucidated. Here, we employed a gene microarray to identify caspase recruitment domain-containing protein 11 (CARD11), a lymphoma-associated scaffold protein presenting brain-specific upregulated expression in a virulent neurotropic Newcastle disease virus (NDV)-infected natural host. Chicken primary neuronal cells infected with NDV appeared slightly syncytial and died quickly. CARD11 overexpression inhibited viral replication and delayed cytopathic effects; conversely, depletion of CARD11 enhanced viral replication and cytopathic effects in chicken primary neuronal cells. The inhibition of viral replication by CARD11 could not be blocked with CARD11-Bcl10-MALT1 (CBM) signalosome and NF-κB signaling inhibitors. CARD11 was found to interact directly with the viral phosphoprotein (P) through its CC1 domain and the X domain of P; this X domain also mediated the interaction between P and the viral large polymerase protein (L). The CARD11 CC1 domain and L competitively bound to P via the X domain that hindered the P-L interaction of the viral ribonucleoprotein (RNP) complex, resulting in a reduction of viral polymerase activity in a minigenome assay and inhibition of viral replication. Animal experiments further revealed that CARD11 contributed to viral replication inhibition and neuropathology in infected chicken brains. Taken together, our findings identify CARD11 as a brain-specific antiviral factor of NDV infection in avian species.IMPORTANCE Newcastle disease virus (NDV) substantially impacts the poultry industry worldwide and causes viral encephalitis and neurological disorders leading to brain damage, paralysis, and death. The mechanism of interaction between this neurotropic virus and the avian central nervous system (CNS) is largely unknown. Here, we report that host protein CARD11 presented brain-specific upregulated expression that inhibited NDV replication, which was not due to CARD11-Bcl10-MALT1 (CBM) complex-triggered activation of its downstream signaling pathways. The inhibitory mechanism of viral replication is through the CARD11 CC1 domain, and the viral large polymerase protein (L) competitively interacts with the X domain of the viral phosphoprotein (P), which hampers the P-L interaction, suppressing the viral polymerase activity and viral replication. An in vivo study indicated that CARD11 alleviated neuropathological lesions and reduced viral replication in chicken brains. These results provide insight into the interaction between NDV infection and the host defense in the CNS and a potential antiviral target for viral neural diseases.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/antagonistas & inhibidores , Guanilato Ciclasa/antagonistas & inhibidores , Neuronas/virología , Virus de la Enfermedad de Newcastle/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Proteína 10 de la LLC-Linfoma de Células B/metabolismo , Unión Competitiva , Encéfalo/patología , Encéfalo/virología , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/metabolismo , Pollos , Técnicas de Silenciamiento del Gen , Guanilato Ciclasa/genética , Guanilato Ciclasa/metabolismo , Humanos , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/metabolismo , Enfermedad de Newcastle/virología , Receptor EphB2 , Transducción de Señal
7.
J Biol Chem ; 294(22): 8959-8972, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-30992368

RESUMEN

Protozoan parasites of the phylum Apicomplexa actively move through tissue to initiate and perpetuate infection. The regulation of parasite motility relies on cyclic nucleotide-dependent kinases, but how these kinases are activated remains unknown. Here, using an array of biochemical and cell biology approaches, we show that the apicomplexan parasite Toxoplasma gondii expresses a large guanylate cyclase (TgGC) protein, which contains several upstream ATPase transporter-like domains. We show that TgGC has a dynamic localization, being concentrated at the apical tip in extracellular parasites, which then relocates to a more cytosolic distribution during intracellular replication. Conditional TgGC knockdown revealed that this protein is essential for acute-stage tachyzoite growth, as TgGC-deficient parasites were defective in motility, host cell attachment, invasion, and subsequent host cell egress. We show that TgGC is critical for a rapid rise in cytosolic [Ca2+] and for secretion of microneme organelles upon stimulation with a cGMP agonist, but these deficiencies can be bypassed by direct activation of signaling by a Ca2+ ionophore. Furthermore, we found that TgGC is required for transducing changes in extracellular pH and [K+] to activate cytosolic [Ca2+] flux. Together, the results of our work implicate TgGC as a putative signal transducer that activates Ca2+ signaling and motility in Toxoplasma.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Señalización del Calcio , Guanilato Ciclasa/metabolismo , Proteínas Protozoarias/metabolismo , Toxoplasma/metabolismo , Adenosina Trifosfatasas/genética , Calcio/metabolismo , Ionóforos de Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , GMP Cíclico/metabolismo , Citosol/metabolismo , Guanilato Ciclasa/antagonistas & inhibidores , Guanilato Ciclasa/genética , Concentración de Iones de Hidrógeno , Oligonucleótidos Antisentido/metabolismo , Potasio/metabolismo , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/genética , Pirazoles/farmacología , Pirimidinonas/farmacología , Toxoplasma/crecimiento & desarrollo
8.
Pharmacol Biochem Behav ; 180: 92-100, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30857920

RESUMEN

Simvastatin, one of the lipophilic statins, has been shown to be effective in reducing depression in rodents. The present study aimed to investigate the potential antidepressant-like activity of simvastatin and the possible involvement of NO-cGMP-KATP channels pathway and PPARγ using forced swimming test (FST) in mice. In addition, the interaction between simvastatin and fluoxetine as a reference drug was examined. After assessment of locomotor behavior in the open-field test (OFT), FST was applied for evaluation of depressive behavior in mice. Simvastatin at doses (20, 30, and 40 mg/kg, i.p.) was administrated 30 min before the OFT or FST. To evaluate the involvement of NO-cGMP-KATP channels pathway, mice were pre-treated intraperitoneally with l-arginine (a nitric oxide precursor, 750 mg/kg), L-NAME (a NOS inhibitor, 10 mg/kg), methylene blue (guanylyl cyclase inhibitor, 20 mg/kg), sildenafil (a PDE-5 inhibitor, 5 mg/kg), glibenclamide (ATP-sensitive K+ channel blocker, 1 mg/kg), and diazoxide (K+ channels opener, 10 mg/kg). Moreover, to clarify the probable involvement of PPARγ receptors, pioglitazone, a PPARγ agonist (5 mg/kg, i.p.), and GW9662, a PPARγ antagonist (2 mg/kg, i.p.), were pre-treated with simvastatin. Immobility time was significantly decreased after simvastatin injection. Administration of L-NAME, methylene blue, glibenclamide and pioglitazone in combination with the sub-effective dose of simvastatin (20 mg/kg, i.p.) reduced the immobility time in the FST compared to drugs alone, while co-administration of effective doses of simvastatin (30 mg/kg, i.p.) with l-arginine, sildenafil, diazoxide, and GW9662 prevented the antidepressant-like effect of simvastatin. In addition, simvastatin (20 mg/kg) potentiated the antidepressant-like effect of fluoxetine through the NO pathway. None of the drugs produced any significant alterations in locomotor activity using OFT. These results demonstrated that NO-cGMP-KATP channels pathway and PPARγ receptors may be involved in the antidepressant-like effect of simvastatin.


Asunto(s)
Antidepresivos/farmacología , GMP Cíclico/metabolismo , Canales KATP/metabolismo , Óxido Nítrico/metabolismo , PPAR gamma/metabolismo , Simvastatina/farmacología , Natación/fisiología , Anilidas/farmacología , Animales , Antidepresivos/administración & dosificación , Arginina/administración & dosificación , Arginina/farmacología , Depresión/tratamiento farmacológico , Sinergismo Farmacológico , Fluoxetina/administración & dosificación , Fluoxetina/farmacología , Guanilato Ciclasa/antagonistas & inhibidores , Locomoción/efectos de los fármacos , Masculino , Azul de Metileno/administración & dosificación , Azul de Metileno/farmacología , Ratones , NG-Nitroarginina Metil Éster/administración & dosificación , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , PPAR gamma/agonistas , PPAR gamma/antagonistas & inhibidores , Pioglitazona/farmacología , Simvastatina/administración & dosificación
9.
Circulation ; 139(17): 2032-2048, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30717607

RESUMEN

BACKGROUND: Intraplaque hemorrhage promotes atherosclerosis progression, and erythrocytes may contribute to this process. In this study we examined the effects of red blood cells on smooth muscle cell mineralization and vascular calcification and the possible mechanisms involved. METHODS: Erythrocytes were isolated from human and murine whole blood. Intact and lysed erythrocytes and their membrane fraction or specific erythrocyte components were examined in vitro using diverse calcification assays, ex vivo by using the murine aortic ring calcification model, and in vivo after murine erythrocyte membrane injection into neointimal lesions of hypercholesterolemic apolipoprotein E-deficient mice. Vascular tissues (aortic valves, atherosclerotic carotid artery specimens, abdominal aortic aneurysms) were obtained from patients undergoing surgery. RESULTS: The membrane fraction of lysed, but not intact human erythrocytes promoted mineralization of human arterial smooth muscle cells in culture, as shown by Alizarin red and van Kossa stain and increased alkaline phosphatase activity, and by increased expression of osteoblast-specific transcription factors (eg, runt-related transcription factor 2, osterix) and differentiation markers (eg, osteopontin, osteocalcin, and osterix). Erythrocyte membranes dose-dependently enhanced calcification in murine aortic rings, and extravasated CD235a-positive erythrocytes or Perl iron-positive signals colocalized with calcified areas or osteoblast-like cells in human vascular lesions. Mechanistically, the osteoinductive activity of lysed erythrocytes was localized to their membrane fraction, did not involve membrane lipids, heme, or iron, and was enhanced after removal of the nitric oxide (NO) scavenger hemoglobin. Lysed erythrocyte membranes enhanced calcification to a similar extent as the NO donor diethylenetriamine-NO, and their osteoinductive effects could be further augmented by arginase-1 inhibition (indirectly increasing NO bioavailability). However, the osteoinductive effects of erythrocyte membranes were reduced in human arterial smooth muscle cells treated with the NO scavenger 2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl 3-oxide or following inhibition of NO synthase or the NO receptor soluble guanylate cyclase. Erythrocytes isolated from endothelial NO synthase-deficient mice exhibited a reduced potency to promote calcification in the aortic ring assay and after injection into murine vascular lesions. CONCLUSIONS: Our findings in cells, genetically modified mice, and human vascular specimens suggest that intraplaque hemorrhage with erythrocyte extravasation and lysis promotes osteoblastic differentiation of smooth muscle cells and vascular lesion calcification, and also support a role for erythrocyte-derived NO.


Asunto(s)
Membrana Eritrocítica , Calcificación Vascular/etiología , Animales , Aorta , Diferenciación Celular , Células Cultivadas , Durapatita/metabolismo , Guanilato Ciclasa/antagonistas & inhibidores , Hemorragia/complicaciones , Humanos , Hipercolesterolemia/etiología , Ratones , Ratones Noqueados para ApoE , Miocitos del Músculo Liso/patología , Neointima/patología , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/deficiencia , Técnicas de Cultivo de Órganos , Osteoblastos/patología , Triazenos/toxicidad
10.
J Pharm Biomed Anal ; 164: 326-336, 2019 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-30412806

RESUMEN

Riociguat, a guanyl cyclase inhibitor, is one of its kind drug regimen approved for management of pulmonary arterial hypertension and chronic thromboembolism pulmonary hypertension. Extensive literature review indicates lack of comprehensive reports on its metabolic fate. The present study reports the in vivo and in vitro identification and characterization of metabolites of riociguat, using high-performance liquid chromatography-quadruple time-of-flight tandem mass spectrometry. In vitro studies were conducted by incubating the drug in human and rat liver microsomes in presence of respective cofactors. In vivo studies were undertaken by oral administration of suspension of drug to male Sprague-Dawley rats followed by collection of urine, feces and blood at specific intervals. A total of 18 metabolites were observed in in vivo and in vitro matrices which includes hydroxyl, N-oxide, desmethyl, defluorinated hydroxyl, glucuronides and N-acetyl cysteine conjugates. Presence of N-acetyl cysteine conjugates strongly points towards the formation of a reactive metabolite intermediate trapped through N-acetyl cysteine and can be considered a matter of concern as the reactive metabolites have been known to manifest toxicities. Their presence was mimicked in in vitro samples as well. The toxicological properties of drug and metabolites were evaluated by using ADMET Predictor ™ software.


Asunto(s)
Antihipertensivos/análisis , Guanilato Ciclasa/antagonistas & inhibidores , Pirazoles/análisis , Pirimidinas/análisis , Programas Informáticos , Acetilcisteína/química , Administración Oral , Animales , Antihipertensivos/administración & dosificación , Antihipertensivos/metabolismo , Antihipertensivos/toxicidad , Biotransformación , Cromatografía Líquida de Alta Presión/instrumentación , Cromatografía Líquida de Alta Presión/métodos , Simulación por Computador , Minería de Datos , Humanos , Masculino , Microsomas Hepáticos , Pirazoles/administración & dosificación , Pirazoles/metabolismo , Pirazoles/toxicidad , Pirimidinas/administración & dosificación , Pirimidinas/metabolismo , Pirimidinas/toxicidad , Ratas , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem/instrumentación , Espectrometría de Masas en Tándem/métodos
11.
Cell Physiol Biochem ; 48(4): 1563-1578, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30071522

RESUMEN

BACKGROUND/AIMS: Spinal cord glioma is a highly aggressive malignancy that commonly results in high mortality due to metastasis, high recurrence and limited treatment regimens. This study aims to elucidate the effects of long non-coding RNA LINC01260 (LINC01260) on the proliferation, migration and invasion of spinal cord glioma cells by targeting Caspase recruitment domain family, member 11 (CARD11) via nuclear factor kappa B (NF-κB) signaling. METHODS: The Multi Experiment Matrix (MEM) website was used for target gene prediction, and the DAVID database was used for analysis of the relationship between CARD11 and the NF-κB pathway. In total, 60 cases of glioma tissues and adjacent normal tissues were collected. Human U251 glioma cells were grouped into blank, negative control (NC), LINC01260 vector, CARD11 vector, siRNA-LINC01260, siRNA-CARD11, LINC01260 vector + CARD11 vector and LINC01260 + siRNA-CARD11 groups. A dual-luciferase reporter assay was conducted to verify the target relationship between LINC01260 and CARD11. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blot analysis were employed to assess expression of LINC01260, E-cadherin, p53, CARD11, Ki67, N-cadherin, matrix metalloproteinase (MMP)-9, NF-κBp65 and NF-κBp50. MTT, flow cytometry, wound-healing and Transwell assays were performed to examine cell viability, the cell cycle, apoptosis, invasion and migration. Tumor growth was assessed through xenografts in nude mice. RESULTS: CARD11 was confirmed to be a target gene of LINC01260 and was found to be involved in regulating the NF-κB pathway. Compared with adjacent normal tissues, glioma tissues showed reduced expression of LINC01260 and elevated expression of CARD11 and genes related to apoptosis, invasion and migration; activation of NF-κB signaling was also observed. In contrast to the blank and NC groups, an elevated number of cells arrested in G1 phase, increased apoptosis and reduced cell proliferation, invasion and number of cells arrested in S and G2 phases, as well as tumor growth were found for the LINC01260 vector and siRNA-CARD11 groups. CONCLUSIONS: Our findings demonstrate that overexpression of LINC01260 inhibits spinal cord glioma cell proliferation, migration and invasion by targeting CARD11 via NF-κB signaling suppression.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Proliferación Celular , Guanilato Ciclasa/metabolismo , ARN Largo no Codificante/metabolismo , Transducción de Señal , Animales , Proteínas Adaptadoras de Señalización CARD/antagonistas & inhibidores , Proteínas Adaptadoras de Señalización CARD/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Puntos de Control de la Fase G1 del Ciclo Celular , Glioma/metabolismo , Glioma/patología , Guanilato Ciclasa/antagonistas & inhibidores , Guanilato Ciclasa/genética , Humanos , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Desnudos , FN-kappa B/metabolismo , Interferencia de ARN , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/genética , ARN Interferente Pequeño/metabolismo , Neoplasias de la Médula Espinal/metabolismo , Neoplasias de la Médula Espinal/patología , Proteína p53 Supresora de Tumor/metabolismo
12.
Pflugers Arch ; 470(10): 1521-1542, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29845313

RESUMEN

The ventral medial prefrontal cortex (vMPFC) facilitates the cardiac baroreflex response through N-methyl-D-aspartate (NMDA) receptor activation and nitric oxide (NO) formation by neuronal NO synthase (nNOS) and soluble guanylate cyclase (sGC) triggering. Glutamatergic transmission is modulated by the cannabinoid receptor type 1 (CB1) and transient receptor potential vanilloid type 1 (TRPV1) receptors, which may inhibit or stimulate glutamate release in the brain, respectively. Interestingly, vMPFC CB1 receptors decrease cardiac baroreflex responses, while TRPV1 channels facilitate them. Therefore, the hypothesis of the present study is that the vMPFC NMDA/NO pathway is regulated by both CB1 and TRPV1 receptors in the modulation of cardiac baroreflex activity. In order to test this assumption, we used male Wistar rats that had stainless steel guide cannulae bilaterally implanted in the vMPFC. Subsequently, a catheter was inserted into the femoral artery, for cardiovascular recordings, and into the femoral vein for assessing baroreflex activation. The increase in tachycardic and bradycardic responses observed after the microinjection of a CB1 receptors antagonist into the vMPFC was prevented by an NMDA antagonist as well as by the nNOS and sGC inhibition. NO extracellular scavenging also abolished these responses. These same pharmacological manipulations inhibited cardiac reflex enhancement induced by TRPV1 agonist injection into the area. Based on these results, we conclude that vMPFC CB1 and TRPV1 receptors inhibit or facilitate the cardiac baroreflex activity by stimulating or blocking the NMDA activation and NO synthesis.


Asunto(s)
Barorreflejo , Corazón/fisiología , Corteza Prefrontal/metabolismo , Receptor Cannabinoide CB1/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Antagonistas de Receptores de Cannabinoides/farmacología , Inhibidores Enzimáticos/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Guanilato Ciclasa/antagonistas & inhibidores , Frecuencia Cardíaca , Masculino , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología , Ratas , Ratas Wistar , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Canales Catiónicos TRPV/agonistas
13.
Life Sci ; 201: 130-140, 2018 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-29604271

RESUMEN

We hypothesized that endothelium modulates relaxation induced by a nitric oxide (NO) donor ruthenium complex (TERPY, [Ru(terpy)(bdq)NO]3+) in mesenteric arteries of normotensive and spontaneously hypertensive (SHR) rats in different ways. We analyzed the mechanism involved in TERPY-induced relaxation in the second and third branches of mesenteric arteries and investigated how endothelium contributes to the TERPY vasodilator effect on SHR blood vessels. TERPY induced concentration-dependent relaxation in endothelium-denuded (E-) and endothelium-intact (E+) mesenteric arteries of normotensive rats and SHR. Pretreatment with ODQ (which inhibits soluble guanylyl cyclase) or TEA (tetraethylammonium, which blocks potassium channels) significantly reduced the TERPY vasodilator effect on E- mesenteric arteries of normotensive rats and SHR. The presence of endothelium shifted the concentration-effect curves for TERPY in E+ mesenteric arteries of normotensive rats to the right. Conversely, the presence of endothelium shifted the concentration-effect curves for TERPY in the case of SHR E+ mesenteric arteries to the left, which suggested increased potency. L-NNA, a more selective endothelial NO synthase (eNOS) inhibitor, reduced TERPY potency in SHR. The presence of endothelium and notably of NOS contributed to the TERPY vasodilator action in SHR: TERPY promoted eNOS Ser1177 phosphorylation with consequent NO production and increased soluble guanylyl cyclase activity, which may have directly activated potassium channels.


Asunto(s)
Complejos de Coordinación/farmacología , Endotelio Vascular/efectos de los fármacos , Relajación Muscular/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Rutenio/química , Animales , Relación Dosis-Respuesta a Droga , Guanilato Ciclasa/antagonistas & inhibidores , Masculino , Arterias Mesentéricas/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Oxadiazoles/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Quinoxalinas/farmacología , Ratas , Ratas Endogámicas SHR , Resistencia Vascular/efectos de los fármacos , Vasodilatación/efectos de los fármacos
14.
Clin Exp Hypertens ; 40(5): 414-420, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29027818

RESUMEN

BACKGROUND: This study aimed to examine the effects of nesfatin-1 on thoracic aorta vasoreactivity and to investigate the inotropic and chronotropic effects of nesfatin-1 on the spontaneous contractions of the isolated rat atria. METHODS: Isolated right atria and thoracic aorta were used in organ baths. The reactivity of the thoracic aorta was evaluated by potassium chloride (KCl), phenylephrine (Phe), acetylcholine (ACh), and sodium nitroprusside (SNP). The effects of nesfatin-1 on the spontaneous contractions of the rat atria were also examined. RESULTS: Nesfatin-1 (0.1-100 ng/ml) produced a concentration-dependent relaxation response in rat thoracic aorta. The relaxant responses to nesfatin-1 were inhibited by the removal of endothelium, NO synthase blocker N-nitro-L-arginine methyl ester (L-NAME, 10-4 M), and soluble guanylate cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ, 10-5 M). Nesfatin-1 (10 ng/ml, 30 min) increased the relaxation responses to either ACh or SNP, and the contractile response to both Phe and KCl did not significantly change in the arteries that were incubated with nesfatin-1 compared with the controls. The thoracic aorta contractions induced by the stepwise addition of Ca2+ to a high KCl solution with no Ca2+ were not significantly changed by nesfatin-1. Under calcium-free conditions, the contractions of the thoracic aorta rings incubated with nesfatin-1 in response to Phe were not significantly lower than those of the rings from the control rats. Nesfatin-1 showed positive inotropic and chronotropic effects on rat atria. CONCLUSION: Nesfatin-1 significantly changed the vascular responsiveness in rat thoracic aorta and produced positive inotropic and chronotropic effects on rat atria.


Asunto(s)
Aorta Torácica/fisiología , Función Atrial/efectos de los fármacos , Proteínas de Unión al Calcio/farmacología , Proteínas de Unión al ADN/farmacología , Contracción Muscular/efectos de los fármacos , Relajación Muscular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Proteínas del Tejido Nervioso/farmacología , Acetilcolina/farmacología , Animales , Aorta Torácica/efectos de los fármacos , Calcio/farmacología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Inhibidores Enzimáticos/farmacología , Guanilato Ciclasa/antagonistas & inhibidores , Masculino , Músculo Liso Vascular/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Nitroprusiato/farmacología , Nucleobindinas , Oxadiazoles/farmacología , Fenilefrina/farmacología , Cloruro de Potasio/farmacología , Quinoxalinas/farmacología , Ratas , Vasoconstrictores/farmacología , Vasodilatadores/farmacología
15.
Sci Rep ; 7(1): 17993, 2017 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-29269946

RESUMEN

CARM1 is an arginine methyltransferase with diverse histone and non-histone substrates implicated in the regulation of cellular processes including transcriptional co-activation and RNA processing. CARM1 overexpression has been reported in multiple cancer types and has been shown to modulate oncogenic pathways in in vitro studies. Detailed understanding of the mechanism of action of CARM1 in oncogenesis has been limited by a lack of selective tool compounds, particularly for in vivo studies. We describe the identification and characterization of, to our knowledge, the first potent and selective inhibitor of CARM1 that exhibits anti-proliferative effects both in vitro and in vivo and, to our knowledge, the first demonstration of a role for CARM1 in multiple myeloma (MM). EZM2302 (GSK3359088) is an inhibitor of CARM1 enzymatic activity in biochemical assays (IC50 = 6 nM) with broad selectivity against other histone methyltransferases. Treatment of MM cell lines with EZM2302 leads to inhibition of PABP1 and SMB methylation and cell stasis with IC50 values in the nanomolar range. Oral dosing of EZM2302 demonstrates dose-dependent in vivo CARM1 inhibition and anti-tumor activity in an MM xenograft model. EZM2302 is a validated chemical probe suitable for further understanding the biological role CARM1 plays in cancer and other diseases.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteínas Adaptadoras de Señalización CARD/antagonistas & inhibidores , Inhibidores Enzimáticos/uso terapéutico , Guanilato Ciclasa/antagonistas & inhibidores , Isoxazoles/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Pirimidinas/uso terapéutico , Compuestos de Espiro/uso terapéutico , Animales , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacocinética , Humanos , Técnicas In Vitro , Isoxazoles/farmacocinética , Masculino , Ratones , Trasplante de Neoplasias , Pirimidinas/farmacocinética , Ratas Sprague-Dawley , Compuestos de Espiro/farmacocinética
16.
Biochemistry ; 56(51): 6652-6661, 2017 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-29172459

RESUMEN

Sensory guanylate cyclases (zGCs) in zebrafish photoreceptors are regulated by a family of guanylate cyclase activator proteins (called GCAP1-7). GCAP5 contains two nonconserved cysteine residues (Cys15 and Cys17) that could in principle bind to biologically active transition state metal ions (Zn2+ and Fe2+). Here, we present nuclear magnetic resonance (NMR) and isothermal titration calorimetry (ITC) binding analyses that demonstrate the binding of one Fe2+ ion to two GCAP5 molecules (in a 1:2 complex) with a dissociation constant in the nanomolar range. At least one other Fe2+ binds to GCAP5 with micromolar affinity that likely represents electrostatic Fe2+ binding to the EF-hand loops. The GCAP5 double mutant (C15A/C17A) lacks nanomolar binding to Fe2+, suggesting that Fe2+ at this site is ligated directly by thiolate groups of Cys15 and Cys17. Size exclusion chromatography analysis indicates that GCAP5 forms a dimer in the Fe2+-free and Fe2+-bound states. NMR structural analysis and molecular docking studies suggest that a single Fe2+ ion is chelated by thiol side chains from Cys15 and Cys17 in the GCAP5 dimer, forming an [Fe(SCys)4] complex like that observed previously in two-iron superoxide reductases. Binding of Fe2+ to GCAP5 weakens its ability to activate photoreceptor human GC-E by decreasing GC activity >10-fold. Our results indicate a strong Fe2+-induced inhibition of GC by GCAP5 and suggest that GCAP5 may serve as a redox sensor in visual phototransduction.


Asunto(s)
Compuestos Ferrosos/metabolismo , Proteínas Activadoras de la Guanilato-Ciclasa/metabolismo , Guanilato Ciclasa/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/metabolismo , Proteínas de Pez Cebra/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Guanilato Ciclasa/antagonistas & inhibidores , Proteínas Activadoras de la Guanilato-Ciclasa/química , Proteínas Activadoras de la Guanilato-Ciclasa/genética , Luz , Mutación , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Multimerización de Proteína , Homología de Secuencia de Aminoácido , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/genética
17.
Cell Physiol Biochem ; 43(5): 2088-2101, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29059679

RESUMEN

BACKGROUND/AIMS: Cannabinoids are vasoactive substances that act as key regulators of arterial tone in the blood vessels supplying peripheral tissues and the central nervous system. We therefore investigated the effect of cannabinoids on retinal capillaries and pericytes. METHODS: The effects of cannabinoids on capillary diameters were determined using an ex vivo whole-mount rat retinal model. Western blotting, quantitative PCR, and immunohistochemistry were performed to explore the underlying mechanism. RESULTS: Endogenous cannabinoid 2-arachidonoylglycerol and anandamide and exogenous cannabinoid (R-(+)-WIN55212-2) dilated the noradrenaline-precontracted capillaries in a concentration-dependent manner (1 µM to 0.1 mM). The extent of vasorelaxation was positively correlated with changes in pericyte width. The effects of R-(+)-WIN55212-2 on vasorelaxation and pericyte width were inhibited by a cannabinoid receptor type-1 (CB1) antagonist, AM251 or rimonabant (SR141716A), the nitric oxide synthase inhibitor l-NAME, and the guanylate cyclase inhibitor ODQ. They were also abolished by the removal of the endothelium, but not by the cannabinoid receptor-2 antagonist SR144528, the endothelial cannabinoid receptor antagonist O-1918, or the cyclooxygenase inhibitor indomethacin. CONCLUSION: The exogenous cannabinoid R-(+)-WIN55212-2 promotes the vasorelaxation of pericyte-containing rat retinal capillaries. This effect of R-(+)-WIN55212-2 is dependent on CB1 and the nitric oxide-cyclic guanosine monophosphate pathway, and requires an intact endothelium.


Asunto(s)
Cannabinoides/farmacología , Capilares/efectos de los fármacos , Pericitos/citología , Pericitos/efectos de los fármacos , Retina/citología , Animales , Ácidos Araquidónicos/farmacología , Western Blotting , Canfanos/farmacología , Antagonistas de Receptores de Cannabinoides/farmacología , Endocannabinoides/farmacología , Guanilato Ciclasa/antagonistas & inhibidores , Inmunohistoquímica , Masculino , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Piperidinas/farmacología , Alcamidas Poliinsaturadas/farmacología , Pirazoles/farmacología , Ratas , Ratas Wistar , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/metabolismo , Receptores de Cannabinoides/metabolismo , Retina/efectos de los fármacos , Rimonabant
18.
J Physiol ; 595(24): 7427-7439, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29071734

RESUMEN

KEY POINTS: People with insulin resistance or type 2 diabetes can substantially increase their skeletal muscle glucose uptake during exercise and insulin sensitivity after exercise. Skeletal muscle nitric oxide (NO) is important for glucose uptake during exercise, although how prior exercise increases insulin sensitivity is unclear. In the present study, we examined whether NO is necessary for normal increases in skeletal muscle insulin sensitivity after contraction ex vivo in mouse muscle. The present study uncovers, for the first time, a novel role for NO in the insulin sensitizing effects of ex vivo contraction, which is independent of blood flow. ABSTRACT: The factors regulating the increase in skeletal muscle insulin sensitivity after exercise are unclear. We examined whether nitric oxide (NO) is required for the increase in insulin sensitivity after ex vivo contractions. Isolated C57BL/6J mouse EDL muscles were contracted for 10 min or remained at rest (basal) with or without the NO synthase (NOS) inhibition (NG -monomethyl-l-arginine; l-NMMA; 100 µm). Then, 3.5 h post contraction/basal, muscles were exposed to saline or insulin (120 µU ml-1 ) with or without l-NMMA during the last 30 min. l-NMMA had no effect on basal skeletal muscle glucose uptake. The increase in muscle glucose uptake with insulin (57%) was significantly (P < 0.05) greater after prior contraction (140% increase). NOS inhibition during the contractions had no effect on this insulin-sensitizing effect of contraction, whereas NOS inhibition during insulin prevented the increase in skeletal muscle insulin sensitivity post-contraction. Soluble guanylate cyclase inhibition, protein kinase G (PKG) inhibition or cyclic nucleotide phosphodiesterase inhibition each had no effect on the insulin-sensitizing effect of prior contraction. In conclusion, NO is required for increases in insulin sensitivity several hours after contraction of mouse skeletal muscle via a cGMP/PKG independent pathway.


Asunto(s)
Insulina/metabolismo , Contracción Muscular , Fibras Musculares Esqueléticas/metabolismo , Óxido Nítrico/metabolismo , 2',3'-Nucleótido Cíclico Fosfodiesterasas/antagonistas & inhibidores , Animales , Células Cultivadas , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Glucosa/metabolismo , Guanilato Ciclasa/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/fisiología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Transducción de Señal , omega-N-Metilarginina/farmacología
19.
Circ Res ; 120(7): 1174-1182, 2017 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-28360348

RESUMEN

Endothelial cells lining the vessel wall control important aspects of vascular homeostasis. In particular, the production of endothelium-derived nitric oxide and activation of soluble guanylate cyclase promotes endothelial quiescence and governs vasomotor function and proportional remodeling of blood vessels. Here, we discuss novel approaches to improve endothelial nitric oxide generation and preserve its bioavailability. We also discuss therapeutic opportunities aimed at activation of soluble guanylate cyclase for multiple cardiovascular indications.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , GMP Cíclico/metabolismo , Endotelio Vascular/efectos de los fármacos , Guanilato Ciclasa/antagonistas & inhibidores , Óxido Nítrico/metabolismo , Animales , Endotelio Vascular/metabolismo , Humanos , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Inhibidores de Fosfodiesterasa/farmacología
20.
Auton Autacoid Pharmacol ; 37(1): 3-12, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28332266

RESUMEN

Epoxyeicosatrienoic acids (EETs) are endogenous ligands that undergo hydrolysis by soluble epoxide hydrolase (sEH). The responses of 11, 12-EET in comparison with other vasodilator agonists including carbachol and sodium nitroprusside (SNP) were investigated. The effect of 1-cyclohexyl-3-dodecyl urea (CDU), a sEH, was tested on the vasodilator effect induced by 11, 12-EET in the perfused mesenteric beds isolated from normo-glycaemic and type-1 STZ-diabetic rats. In the perfused mesenteric beds of control and diabetic animals, 11, 12-EET produced vasodilation in a dose-dependent manner. The vasodilator response induced by 11, 12-EET was significantly decreased in tissues obtained from diabetic animals, but this was significantly corrected through inhibition of sEH. The effects of nitric oxide synthase inhibitor, cyclo-oxygenase inhibitor, specific potassium channel inhibitors, soluble guanylyl cyclase inhibitor and transient receptor potential channel V4 inhibitor, on vasodilator response to 11, 12-EET were investigated. In tissues isolated from control animals, vasodilator responses to 11, 12-EET were not inhibited by acute incubation with l-NAME, l-NAME with indomethacin, glibenclamide, iberiotoxin, charybdotoxin, apamin or ODQ. Incubation with the transient receptor potential channel V4 inhibitor ruthenium red caused significantly reduced vasodilator responses induced by 11, 12-EET. In conclusion, results from this study indicate that 11, 12-EET has a vasodilator effect in the perfused mesenteric bed, partly through activation of vanilloid receptor. A strategy to elevate the levels of EETs may have a significant impact in correcting microvascular abnormality associated with diabetes.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Circulación Esplácnica/efectos de los fármacos , Vasodilatadores/farmacología , Ácido 8,11,14-Eicosatrienoico/farmacología , Animales , Inhibidores de la Ciclooxigenasa/farmacología , Diabetes Mellitus Experimental/fisiopatología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Guanilato Ciclasa/antagonistas & inhibidores , Masculino , Óxido Nítrico Sintasa/antagonistas & inhibidores , Perfusión , Bloqueadores de los Canales de Potasio/farmacología , Ratas , Ratas Sprague-Dawley , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA