Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 38(17): e70046, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39259502

RESUMEN

Large-conductance, calcium-activated potassium channels (BK channels) and the Na/K-ATPase are expressed universally in vascular smooth muscle. The Na/K-ATPase may act via changes in the intracellular Ca2+ concentration mediated by the Na/Ca exchanger (NCX) and via Src kinase. Both pathways are known to regulate BK channels. Whether BK channels functionally interact in vascular smooth muscle cells with the Na/K-ATPase remains to be elucidated. Thus, this study addressed the hypothesis that BK channels limit ouabain-induced vasocontraction. Rat mesenteric arteries were studied using isometric myography, FURA-2 fluorimetry and proximity ligation assay. The BK channel blocker iberiotoxin potentiated methoxamine-induced contractions. The cardiotonic steroid, ouabain (10-5 M), induced a contractile effect of IBTX at basal tension prior to methoxamine administration and enhanced the pro-contractile effect of IBTX on methoxamine-induced contractions. These facilitating effects of ouabain were prevented by the inhibition of either NCX or Src kinase. Furthermore, inhibition of NCX or Src kinase reduced the BK channel-mediated negative feedback regulation of arterial contraction. The effects of NCX and Src kinase inhibition were independent of each other. Co-localization of the Na/K-ATPase and the BK channel was evident. Our data suggest that BK channels limit ouabain-induced vasocontraction by a dual mechanism involving the NCX and Src kinase signaling. The data propose that the NCX and the Src kinase pathways, mediating the ouabain-induced activation of the BK channel, act in an independent manner.


Asunto(s)
Canales de Potasio de Gran Conductancia Activados por el Calcio , Arterias Mesentéricas , Músculo Liso Vascular , Ouabaína , Intercambiador de Sodio-Calcio , ATPasa Intercambiadora de Sodio-Potasio , Familia-src Quinasas , Animales , Ouabaína/farmacología , Familia-src Quinasas/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Ratas , Masculino , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Vasoconstricción/efectos de los fármacos , Ratas Wistar , Contracción Muscular/efectos de los fármacos
2.
Biochem J ; 481(5): 387-403, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38373073

RESUMEN

The dynamic nature of the microtubule network is dependent in part by post-translational modifications (PTMs) - particularly through acetylation, which stabilizes the microtubule network. Whether PTMs of the microtubule network in vascular smooth muscle cells (VSMCs) contribute to the pathophysiology of hypertension is unknown. The aim of this study was to determine the acetylated state of the microtubule network in the mesenteric arteries of spontaneously hypertensive rats (SHR). Experiments were performed on male normotensive rats and SHR mesenteric arteries. Western blotting and mass spectrometry determined changes in tubulin acetylation. Wire myography was used to investigate the effect of tubacin on isoprenaline-mediated vasorelaxations. Isolated cells from normotensive rats were used for scanning ion conductance microscopy (SICM). Mass spectrometry and Western blotting showed that tubulin acetylation is increased in the mesenteric arteries of the SHR compared with normotensive rats. Tubacin enhanced the ß-adrenoceptor-mediated vasodilatation by isoprenaline when the endothelium was intact, but attenuated relaxations when the endothelium was denuded or nitric oxide production was inhibited. By pre-treating vessels with colchicine to disrupt the microtubule network, we were able to confirm that the effects of tubacin were microtubule-dependent. Using SICM, we examined the cell surface Young's modulus of VSMCs, but found no difference in control, tubacin-treated, or taxol-treated cells. Acetylation of tubulin at Lys40 is elevated in mesenteric arteries from the SHR. Furthermore, this study shows that tubacin has an endothelial-dependent bimodal effect on isoprenaline-mediated vasorelaxation.


Asunto(s)
Anilidas , Ácidos Hidroxámicos , Hipertensión , Tubulina (Proteína) , Ratas , Animales , Masculino , Ratas Endogámicas WKY , Acetilación , Isoproterenol/farmacología , Ratas Endogámicas SHR , Arterias Mesentéricas , Vasodilatación , Microtúbulos , Endotelio Vascular/fisiología
3.
J Biol Chem ; 299(10): 105221, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37660920

RESUMEN

Hypertension is associated with the presence of vascular abnormalities, including remodeling and rarefaction. These processes play an important role in cerebrovascular disease development; however, the mechanistic changes leading to these diseases are not well characterized. Using data-independent acquisition-based mass spectrometry analysis, here we determined the protein changes in cerebral arteries in pre- and early-onset hypertension from the spontaneously hypertensive rat (SHR), a model that resembles essential hypertension in humans. Our analysis identified 125 proteins with expression levels that were significantly upregulated or downregulated in 12-week-old spontaneously hypertensive rats compared to normotensive Wistar Kyoto rats. Using an angiogenesis enrichment analysis, we further identified a critical imbalance in angiogenic proteins that promoted an anti-angiogenic profile in cerebral arteries at early onset of hypertension. In a comparison to previously published data, we demonstrate that this angiogenic imbalance is not present in mesenteric and renal arteries from age-matched SHRs. Finally, we identified two proteins (Fbln5 and Cdh13), whose expression levels were critically altered in cerebral arteries compared to the other arterial beds. The observation of an angiogenic imbalance in cerebral arteries from the SHR reveals critical protein changes in the cerebrovasculature at the early onset of hypertension and provides novel insights into the early pathology of cerebrovascular disease.

4.
FASEB J ; 37(12): e23282, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37994700

RESUMEN

Prorenin and the prorenin receptor ((P)RR) are important, yet controversial, members of the renin-angiotensin-aldosterone system. The ((P)RR) is expressed throughout the body, including the vasculature, however, the direct effect of prorenin on arterial contractility is yet to be determined. Within rat mesenteric arteries, immunostaining and proximity ligation assays were used to determine the interacting partners of (P)RR in freshly isolated vascular smooth muscle cells (VSMCs). Wire myography examined the functional effect of prorenin. Simultaneous changes in [Ca2+ ]i and force were recorded in arteries loaded with Fura-2AM. Spontaneously transient outward currents were recorded via perforated whole-cell patch-clamp configuration in freshly isolated VSMCs. We found that the (P)RR is located within a distance of less than 40 nm from the V-ATPase, caveolin-1, ryanodine receptors, and large conductance Ca2+ -activated K+ channels (BKCa ) in VSMCs. [Ca2+ ]i imaging and isometric tension recordings indicate that 1 nM prorenin enhanced α1-adrenoreceptor-mediated contraction, associated with an increased number of Ca2+ waves, independent of voltage-gated Ca2+ channels activation. Incubation of VSMCs with 1 nM prorenin decreased the amplitude and frequency of spontaneously transient outward currents and attenuated BKCa -mediated relaxation. Inhibition of the V-ATPase with 100 nM bafilomycin prevented prorenin-mediated inhibition of BKCa -derived relaxation. Renin (1 nM) had no effect on BKCa -mediated relaxation. In conclusion, prorenin enhances arterial contractility by inhibition of BKCa and increasing intracellular Ca2+ release. It is likely that this effect is mediated through a local shift in pH upon activation of the (P)RR and stimulation of the V-ATPase.


Asunto(s)
Contracción Muscular , Renina , Ratas , Animales , Miocitos del Músculo Liso , Arterias Mesentéricas , Adenosina Trifosfatasas
5.
FASEB J ; 37(9): e23125, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37535015

RESUMEN

The evergreen plant rosemary (Salvia rosmarinus) has been employed medicinally for centuries as a memory aid, analgesic, spasmolytic, vasorelaxant and antihypertensive, with recent preclinical and clinical evidence rationalizing some applications. Voltage-gated potassium (Kv) channels in the KCNQ (Kv7) subfamily are highly influential in the nervous system, muscle and epithelia. KCNQ4 and KCNQ5 regulate vascular smooth muscle excitability and contractility and are implicated as antihypertensive drug targets. Here, we found that rosemary extract potentiates homomeric and heteromeric KCNQ4 and KCNQ5 activity, resulting in membrane hyperpolarization. Two rosemary diterpenes, carnosol and carnosic acid, underlie the effects and, like rosemary, are efficacious KCNQ-dependent vasorelaxants, quantified by myography in rat mesenteric arteries. Sex- and estrous cycle stage-dependence of the vasorelaxation matches sex- and estrous cycle stage-dependent KCNQ expression. The results uncover a molecular mechanism underlying rosemary vasorelaxant effects and identify new chemical spaces for KCNQ-dependent vasorelaxants.


Asunto(s)
Plantas Medicinales , Rosmarinus , Ratas , Animales , Músculo Liso Vascular/fisiología , Canales de Potasio KCNQ , Vasodilatadores/farmacología
6.
J Biol Chem ; 298(1): 101512, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34929167

RESUMEN

Resistance arteries are small blood vessels that create resistance to blood flow. In hypertension, resistance arteries undergo remodeling, affecting their ability to contract and relax appropriately. To date, no study has mapped the hypertension-related proteomic changes in resistance arteries. Using a novel data-independent acquisition-mass spectrometry (DIA-MS) approach, we determined the proteomic changes in small mesenteric and renal arteries in pre- and early-onset hypertension from the spontaneously hypertensive rat (SHR) model, which represents human primary hypertension. Compared with normotensive controls, mesenteric arteries from 12-week-old SHRs had 286 proteins that were significantly up- or downregulated, whereas 52 proteins were identified as up- or downregulated in mesenteric arteries from 6-week-old SHRs. Of these proteins, 18 were also similarly regulated in SHR renal arteries. Our pathway analyses reveal several novel pathways in the pathogenesis of hypertension. Finally, using a matrisome database, we identified 38 altered extracellular-matrix-associated proteins, many of which have never previously been associated with hypertension. Taken together, this study reveals novel proteins and mechanisms that are associated with early-onset hypertension, thereby providing novel insights into disease progression.


Asunto(s)
Hipertensión , Proteómica , Animales , Hipertensión/metabolismo , Hipertensión/patología , Espectrometría de Masas , Arterias Mesentéricas , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Resistencia Vascular
7.
FASEB J ; 36(9): e22457, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35997997

RESUMEN

Tree and shrub barks have been used as folk medicine by numerous cultures across the globe for millennia, for a variety of indications, including as vasorelaxants and antispasmodics. Here, using electrophysiology and myography, we discovered that the KCNQ5 voltage-gated potassium channel mediates vascular smooth muscle relaxant effects of barks used in Native American folk medicine. Bark extracts (1%) from Birch, Cramp Bark, Slippery Elm, White Oak, Red Willow, White Willow, and Wild Cherry each strongly activated KCNQ5 expressed in Xenopus oocytes. Testing of a subset including both the most and the least efficacious extracts revealed that Red Willow, White Willow, and White Oak KCNQ-dependently relaxed rat mesenteric arteries; in contrast, Black Haw bark neither activated KCNQ5 nor induced vasorelaxation. Two compounds common to the active barks (gallic acid and tannic acid) had similarly potent and efficacious effects on both KCNQ5 activation and vascular relaxation, and this together with KCNQ5 modulation by other tannins provides a molecular basis for smooth muscle relaxation effects of Native American folk medicine bark extracts.


Asunto(s)
Canales de Potasio KCNQ , Vasodilatadores , Animales , Humanos , Arterias Mesentéricas , Ratas , Taninos/farmacología , Vasodilatadores/farmacología , Indio Americano o Nativo de Alaska
8.
Br J Clin Pharmacol ; 89(7): 2179-2189, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36764326

RESUMEN

AIMS: The aim of this study is to examine whether colchicine improves ß adrenoceptor-mediated vasodilation in humans by conducting a double-blinded, placebo-controlled intervention study. Colchicine treatment has known beneficial effects on cardiovascular health and reduces the incidence of cardiovascular disease. Studies in isolated rodent arteries have shown that colchicine can enhance ß adrenoceptor-mediated vasodilation, but this has not been determined in humans. METHODS: Middle-aged men with essential hypertension were randomly assigned firstly to acute treatment with either 0.5 mg colchicine (n = 19) or placebo (n = 12). They were subsequently re-randomized for 3 weeks of treatment with either colchicine 0.5 mg twice daily (n = 16) or placebo (n = 15) followed by a washout period of 48-72 h. The vasodilator responses to isoprenaline, acetylcholine and sodium nitroprusside were determined as well as arterial pressure, arterial compliance and plasma inflammatory markers. RESULTS: Acute colchicine treatment increased isoprenaline (by 38% for the highest dose) as well as sodium nitroprusside (by 29% main effect) -induced vasodilation but had no effect on the response to acetylcholine. The 3-week colchicine treatment followed by a washout period did not induce an accumulated or sustained effect on the ß adrenoceptor response, and there was no effect on arterial pressure, arterial compliance or the level of measured inflammatory markers. CONCLUSION: Colchicine acutely enhances ß adrenoceptor- and nitric oxide-mediated changes in vascular conductance in humans, supporting that the mechanism previously demonstrated in rodents, translates to humans. The results provide novel translational evidence for a transient enhancing effect of colchicine on ß adrenoceptor-mediated vasodilation in humans with essential hypertension. CONDENSED ABSTRACT: Preclinical studies in isolated rodent arteries have shown that colchicine can enhance ß adrenoceptor-mediated vasodilation. Here we show that this effect of colchicine can be translated to humans. Acute colchicine treatment was found to increase both isoprenaline- and sodium nitroprusside-induced vasodilation. The study provides the first translational evidence for a transient ß adrenoceptor-mediated vasodilatory effect of colchicine in humans. The finding of an acute effect suggests that it may be clinically important to maintain an adequate bioavailability of colchicine.


Asunto(s)
Acetilcolina , Vasodilatación , Masculino , Persona de Mediana Edad , Humanos , Nitroprusiato/farmacología , Isoproterenol/farmacología , Acetilcolina/farmacología , Colchicina/farmacología , Hipertensión Esencial , Receptores Adrenérgicos
9.
Proc Natl Acad Sci U S A ; 116(42): 21236-21245, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31570602

RESUMEN

Botanical folk medicines have been used throughout human history to treat common disorders such as hypertension, often with unknown underlying mechanisms. Here, we discovered that hypotensive folk medicines from a genetically diverse range of plant species each selectively activated the vascular-expressed KCNQ5 potassium channel, a feature lacking in the modern synthetic pharmacopeia, whereas nonhypotensive plant extracts did not. Analyzing constituents of the hypotensive Sophora flavescens root, we found that the quinolizidine alkaloid aloperine is a KCNQ-dependent vasorelaxant that potently and isoform-selectively activates KCNQ5 by binding near the foot of the channel voltage sensor. Our findings reveal that KCNQ5-selective activation is a defining molecular mechanistic signature of genetically diverse traditional botanical hypotensives, transcending plant genus and human cultural boundaries. Discovery of botanical KCNQ5-selective potassium channel openers may enable future targeted therapies for diseases including hypertension and KCNQ5 loss-of-function encephalopathy.


Asunto(s)
Canales de Potasio KCNQ/metabolismo , Animales , Masculino , Medicina Tradicional/métodos , Raíces de Plantas/química , Ratas , Ratas Wistar
10.
Cell Physiol Biochem ; 55(S3): 46-64, 2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33667331

RESUMEN

BACKGROUND/AIMS: Tea, produced from the evergreen Camellia sinensis, has reported therapeutic properties against multiple pathologies, including hypertension. Although some studies validate the health benefits of tea, few have investigated the molecular mechanisms of action. The KCNQ5 voltage-gated potassium channel contributes to vascular smooth muscle tone and neuronal M-current regulation. METHODS: We applied electrophysiology, myography, mass spectrometry and in silico docking to determine effects and their underlying molecular mechanisms of tea and its components on KCNQ channels and arterial tone. RESULTS: A 1% green tea extract (GTE) hyperpolarized cells by augmenting KCNQ5 activity >20-fold at resting potential; similar effects of black tea were inhibited by milk. In contrast, GTE had lesser effects on KCNQ2/Q3 and inhibited KCNQ1/E1. Tea polyphenols epicatechin gallate (ECG) and epigallocatechin-3-gallate (EGCG), but not epicatechin or epigallocatechin, isoform-selectively hyperpolarized KCNQ5 activation voltage dependence. In silico docking and mutagenesis revealed that activation by ECG requires KCNQ5-R212, at the voltage sensor foot. Strikingly, ECG and EGCG but not epicatechin KCNQ-dependently relaxed rat mesenteric arteries. CONCLUSION: KCNQ5 activation contributes to vasodilation by tea; ECG and EGCG are candidates for future anti-hypertensive drug development.


Asunto(s)
Catequina/análogos & derivados , Canales de Potasio KCNQ/química , Canal de Potasio KCNQ1/química , Arterias Mesentéricas/efectos de los fármacos , Extractos Vegetales/farmacología , Té/química , Animales , Sitios de Unión , Catequina/química , Catequina/farmacología , Canales de Potasio KCNQ/agonistas , Canales de Potasio KCNQ/genética , Canales de Potasio KCNQ/metabolismo , Canal de Potasio KCNQ1/antagonistas & inhibidores , Canal de Potasio KCNQ1/genética , Canal de Potasio KCNQ1/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Arterias Mesentéricas/fisiología , Leche/química , Simulación del Acoplamiento Molecular , Miografía , Oocitos/citología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Técnicas de Placa-Clamp , Extractos Vegetales/química , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Ratas Wistar , Técnicas de Cultivo de Tejidos , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología , Xenopus laevis
11.
Arterioscler Thromb Vasc Biol ; 40(5): 1207-1219, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32188278

RESUMEN

OBJECTIVE: Intravenous acetaminophen/paracetamol (APAP) is well documented to cause hypotension. Since the patients receiving intravenous APAP are usually critically ill, any severe hemodynamic changes, as with those associated with APAP, can be life-threatening. The mechanism underlying this dangerous iatrogenic effect of APAP was unknown. Approach and Results: Here, we show that intravenous APAP caused transient hypotension in rats, which was attenuated by the Kv7 channel blocker, linopirdine. APAP metabolite N-acetyl-p-benzoquinone imine caused vasodilatation of rat mesenteric arteries ex vivo. This vasodilatation was sensitive to linopirdine and also the calcitonin gene-related peptide antagonist, BIBN 4096. Further investigation revealed N-acetyl-p-benzoquinone imine stimulates calcitonin gene-related peptide release from perivascular nerves, causing a cAMP-dependent activation of Kv7 channels. We also show that N-acetyl-p-benzoquinone imine enhances Kv7.4 and Kv7.5 channels overexpressed in oocytes, suggesting that it can activate Kv7.4 and Kv7.5 channels directly, to elicit vasodilatation. CONCLUSIONS: Direct and indirect activation of Kv7 channels by the APAP metabolite N-acetyl-p-benzoquinone imine decreases arterial tone, which can lead to a drop in blood pressure. Our findings provide a molecular mechanism and potential preventive intervention for the clinical phenomenon of intravenous APAP-dependent transient hypotension.


Asunto(s)
Acetaminofén/toxicidad , Presión Sanguínea/efectos de los fármacos , Hipotensión/inducido químicamente , Canales de Potasio KCNQ/agonistas , Arterias Mesentéricas/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Acetaminofén/metabolismo , Animales , Benzoquinonas , Hipotensión/metabolismo , Hipotensión/fisiopatología , Iminas , Canales de Potasio KCNQ/genética , Canales de Potasio KCNQ/metabolismo , Masculino , Potenciales de la Membrana , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/fisiopatología , Ratas Wistar , Transducción de Señal , Xenopus laevis
12.
FASEB J ; 33(2): 2537-2552, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30289750

RESUMEN

KCNE5 is an X-linked gene encoding KCNE5, an ancillary subunit to voltage-gated potassium (KV) channels. Human KCNE5 mutations are associated with atrial fibrillation (AF)- and Brugada syndrome (BrS)-induced cardiac arrhythmias that can arise from increased potassium current in cardiomyocytes. Seeking to establish underlying molecular mechanisms, we created and studied Kcne5 knockout ( Kcne5-/0) mice. Intracardiac ECG revealed that Kcne5 deletion caused ventricular premature beats, increased susceptibility to induction of polymorphic ventricular tachycardia (60 vs. 24% in Kcne5+/0 mice), and 10% shorter ventricular refractory period. Kcne5 deletion increased mean ventricular myocyte KV current density in the apex and also in the subpopulation of septal myocytes that lack fast transient outward current ( Ito,f). The current increases arose from an apex-specific increase in slow transient outward current-1 ( IKslow,1) (conducted by KV1.5) and Ito,f (conducted by KV4) and an increase in IKslow,2 (conducted by KV2.1) in both apex and septum. Kcne5 protein localized to the intercalated discs in ventricular myocytes, where KV2.1 was also detected in both Kcne5-/0 and Kcne5+/0 mice. In HL-1 cardiac cells and human embryonic kidney cells, KCNE5 and KV2.1 colocalized at the cell surface, but predominantly in intracellular vesicles, suggesting that Kcne5 deletion increases IK,slow2 by reducing KV2.1 intracellular sequestration. The human AF-associated mutation KCNE5-L65F negative shifted the voltage dependence of KV2.1-KCNE5 channels, increasing their maximum current density >2-fold, whereas BrS-associated KCNE5 mutations produced more subtle negative shifts in KV2.1 voltage dependence. The findings represent the first reported native role for Kcne5 and the first demonstrated Kcne regulation of KV2.1 in mouse heart. Increased KV current is a manifestation of KCNE5 disruption that is most likely common to both mouse and human hearts, providing a plausible mechanistic basis for human KCNE5-linked AF and BrS.-David, J.-P., Lisewski, U., Crump, S. M., Jepps, T. A., Bocksteins, E., Wilck, N., Lossie, J., Roepke, T. K., Schmitt, N., Abbott, G. W. Deletion in mice of X-linked, Brugada syndrome- and atrial fibrillation-associated Kcne5 augments ventricular KV currents and predisposes to ventricular arrhythmia.


Asunto(s)
Fibrilación Atrial/complicaciones , Síndrome de Brugada/complicaciones , Genes Ligados a X , Activación del Canal Iónico , Miocitos Cardíacos/patología , Canales de Potasio con Entrada de Voltaje/fisiología , Taquicardia Ventricular/etiología , Animales , Fibrilación Atrial/genética , Síndrome de Brugada/genética , Células Cultivadas , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Potasio/metabolismo , Eliminación de Secuencia , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/patología
13.
Am J Physiol Regul Integr Comp Physiol ; 316(5): R552-R562, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30758982

RESUMEN

Activation of antiapoptotic signaling cascades, such as the reperfusion injury salvage kinase (RISK) and survivor activating factor enhancement (SAFE) pathways, is protective in a variety of tissues in the context of ischemia-reperfusion (IR) injury. Hepatic IR injury causes clinically significant hepatocellular damage in surgical procedures, including liver transplantation and hepatic resection, increasing associated morbidity and mortality. We previously found that the cardiovascular-expressed K+ voltage-gated channel ancillary subunit KCNE4 sex specifically influences the cardiac RISK/SAFE pathway response to IR and that Kcne4 deletion testosterone dependently exacerbates cardiac IR injury. Here, we discovered that germline Kcne4 deletion exacerbates hepatic IR injury damage in 13-mo-old male mice, despite a lack of Kcne4 expression in male mouse liver. Examining RISK/SAFE pathway induction, we found that Kcne4 deletion prevents the hepatic ERK1/2 phosphorylation response to IR injury. Conversely, in 13-mo-old female mice, Kcne4 deletion increased both baseline and post-IR GSK-3ß inhibitory phosphorylation, and pharmacological GSK-3ß inhibition was hepatoprotective. Finally, castration of male mice restored normal hepatic RISK/SAFE pathway responses in Kcne4-/- mice, eliminated Kcne4 deletion-dependent serum alanine aminotransferase elevation, and genotype independently augmented the hepatic post-IR GSK-3ß phosphorylation response. These findings support a role for KCNE4 as a systemic modulator of IR injury response and uncover hormonally influenced, sex-specific, KCNE4-dependent and -independent RISK/SAFE pathway induction.


Asunto(s)
Hepatopatías/prevención & control , Hígado/enzimología , Canales de Potasio con Entrada de Voltaje/deficiencia , Daño por Reperfusión/prevención & control , Animales , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Eliminación de Gen , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Hígado/patología , Hepatopatías/enzimología , Hepatopatías/genética , Hepatopatías/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Orquiectomía , Fosforilación , Canales de Potasio con Entrada de Voltaje/genética , Daño por Reperfusión/enzimología , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Factores Sexuales , Transducción de Señal , Testosterona/metabolismo
14.
Pflugers Arch ; 468(4): 643-54, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26729267

RESUMEN

In isolated human atrial cardiomyocytes, inhibition of K2P3.1 K(+) channels results in action potential (action potential duration (APD)) prolongation. It has therefore been postulated that K2P3.1 (KCNK3), together with K2P9.1 (KCNK9), could represent novel drug targets for the treatment of atrial fibrillation (AF). However, it is unknown whether these findings in isolated cells translate to the whole heart. The purposes of this study were to investigate the expression levels of KCNK3 and KCNK9 in human hearts and two relevant rodent models and determine the antiarrhythmic potential of K2P3.1 inhibition in isolated whole-heart preparations. By quantitative PCR, we found that KCNK3 is predominantly expressed in human atria whereas KCNK9 was not detectable in heart human tissue. No differences were found between patients in AF or sinus rhythm. The expression in guinea pig heart resembled humans whereas rats displayed a more uniform expression of KCNK3 between atria and ventricle. In voltage-clamp experiments, ML365 and A293 were found to be potent and selective inhibitors of K2P3.1, but at pH 7.4, they failed to prolong atrial APD and refractory period (effective refractory period (ERP)) in isolated perfused rat and guinea pig hearts. At pH 7.8, which augments K2P3.1 currents, pharmacological channel inhibition produced a significant prolongation of atrial ERP (11.6 %, p = 0.004) without prolonging ventricular APD but did not display a significant antiarrhythmic effect in our guinea pig AF model (3/8 hearts converted on A293 vs 0/7 hearts in time-matched controls). These results suggest that when K2P3.1 current is augmented, K2P3.1 inhibition leads to atrial-specific prolongation of ERP; however, this ERP prolongation did not translate into significant antiarrhythmic effects in our AF model.


Asunto(s)
Potenciales de Acción , Arritmias Cardíacas/metabolismo , Función Atrial , Proteínas del Tejido Nervioso/metabolismo , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Protones , Periodo Refractario Electrofisiológico , Adolescente , Adulto , Animales , Arritmias Cardíacas/fisiopatología , Células Cultivadas , Femenino , Cobayas , Atrios Cardíacos/citología , Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Masculino , Persona de Mediana Edad , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Canales de Potasio de Dominio Poro en Tándem/antagonistas & inhibidores , Canales de Potasio de Dominio Poro en Tándem/genética , Ratas , Ratas Wistar , Especificidad de la Especie , Función Ventricular
15.
J Vasc Res ; 53(3-4): 138-148, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27710966

RESUMEN

Voltage-gated potassium (Kv) channels formed by Kv7 (KCNQ) α-subunits are recognized as crucial for vascular smooth muscle function, in addition to their established roles in the heart (Kv7.1) and the brain (Kv7.2-5). In vivo, Kv7 α-subunits are often regulated by KCNE subfamily ancillary (ß) subunits. We investigated the effects of targeted germline Kcne4 deletion on mesenteric artery reactivity in adult male and female mice. Kcne4 deletion increased mesenteric artery contractility in response to α-adrenoceptor agonist methoxamine, and decreased responses to Kv7.2-7.5 channel activator ML213, in male but not female mice. In contrast, Kcne4 deletion markedly decreased vasorelaxation in response to isoprenaline in both male and female mice. Kcne4 expression was 2-fold lower in the female versus the male mouse mesenteric artery, and Kcne4 deletion elicited only moderate changes of other Kcne transcripts, with no striking sex-specific differences. However, Kv7.4 protein expression in females was twice that in males, and was reduced in both sexes by Kcne4 deletion. Our findings confirm a crucial role for KCNE4 in regulation of Kv7 channel activity to modulate vascular tone, and provide the first known molecular mechanism for sex-specificity of this modulation that has important implications for vascular reactivity and may underlie sex-specific susceptibility to cardiovascular diseases.


Asunto(s)
Canales de Potasio KCNQ/metabolismo , Músculo Liso Vascular/metabolismo , Canales de Potasio con Entrada de Voltaje/deficiencia , Vasoconstricción , Agonistas de Receptores Adrenérgicos alfa 1/farmacología , Anilidas/farmacología , Animales , Compuestos Bicíclicos con Puentes/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Regulación de la Expresión Génica , Genotipo , Canales de Potasio KCNQ/efectos de los fármacos , Canales de Potasio KCNQ/genética , Masculino , Arterias Mesentéricas/metabolismo , Metoxamina/farmacología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Canales de Potasio con Entrada de Voltaje/genética , Factores Sexuales , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Vasodilatadores/farmacología
16.
J Physiol ; 593(24): 5325-40, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26503181

RESUMEN

KEY POINTS: KCNE4 alters the biophysical properties and cellular localization of voltage-gated potassium channel Kv7.4. KCNE4 is expressed in a variety of arteries and, in mesenteric arteries, co-localizes with Kv7.4, which is important in the control of vascular contractility. Knockdown of KCNE4 leads to reduced Kv7.4 membrane abundance, a depolarized membrane potential and an augmented response to vasoconstrictors. KCNE4 is a key regulator of the function and expression of Kv7.4 in vascular smooth muscle. ABSTRACT: The KCNE ancillary subunits (KCNE1-5) significantly alter the expression and function of voltage-gated potassium channels; however, their role in the vasculature has yet to be determined. The present study aimed to investigate the expression and function of the KCNE4 subunit in rat mesenteric arteries and to determine whether it has a functional impact on the regulation of arterial tone by Kv7 channels. In HEK cells expressing Kv7.4, co-expression of KCNE4 increased the membrane expression of Kv7.4 and significantly altered Kv7.4 current properties. Quantitative PCR analysis of different rat arteries found that the KCNE4 isoform predominated and proximity ligation experiments showed that KCNE4 co-localized with Kv7.4 in mesenteric artery myocytes. Morpholino-induced knockdown of KCNE4 depolarized mesenteric artery smooth muscle cells and resulted in their increased sensitivity to methoxamine being attenuated (mean ± SEM EC50 decreased from 5.7 ± 0.63 µm to 1.6 ± 0.23 µm), which coincided with impaired effects of Kv7 modulators. When KCNE4 expression was reduced, less Kv7.4 expression was found in the membrane of the mesenteric artery myocytes. These data show that KCNE4 is consistently expressed in a variety of arteries, and knockdown of the expression product leads to reduced Kv7.4 membrane abundance, a depolarized membrane potential and an augmented response to vasoconstrictors. The present study is the first to demonstrate an integral role of KCNE4 in regulating the function and expression of Kv7.4 in vascular smooth muscle.


Asunto(s)
Arterias Mesentéricas/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Vasoconstricción , Animales , Células Cultivadas , Células HEK293 , Humanos , Masculino , Potenciales de la Membrana , Arterias Mesentéricas/fisiología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiología , Canales de Potasio con Entrada de Voltaje/genética , Ratas , Ratas Wistar
17.
Arterioscler Thromb Vasc Biol ; 34(4): 887-93, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24558103

RESUMEN

OBJECTIVE: Middle cerebral artery (MCA) diameter is regulated by inherent myogenic activity and the effect of potent vasodilators such as calcitonin gene-related peptide (CGRP). Previous studies showed that MCAs express KCNQ1, 4, and 5 potassium channel genes, and the expression products (Kv7 channels) participate in the myogenic control of MCA diameter. The present study investigated the contribution of Kv7.4 and Kv7.5 isoforms to myogenic and CGRP regulation of MCA diameter and determined whether they were affected in hypertensive animals. APPROACH AND RESULTS: Isometric tension recordings performed on MCA from normotensive rats produced CGRP vasodilations that were inhibited by the pan-Kv7 channel blocker linopirdine (P<0.01) and after transfection of arteries with siRNA against KCNQ4 (P<0.01) but not KCNQ5. However, isobaric myography revealed that myogenic constriction in response to increases in intravascular pressure (20-80 mm Hg) was affected by both KCNQ4 and KCNQ5 siRNA. Proximity ligation assay signals were equally abundant for Kv7.4/Kv7.4 or Kv7.4/Kv7.5 antibody combinations but minimal for Kv7.5/Kv7.5 antibodies or Kv7.4/7.1 combinations. In contrast to systemic arteries, Kv7 function and Kv7.4 abundance in MCA were not altered in hypertensive rats. CONCLUSIONS: This study reveals, for the first time to our knowledge, that in cerebral arteries, Kv7.4 and Kv7.5 proteins exist predominantly as a functional heterotetramer, which regulates intrinsic myogenicity and vasodilation attributed to CGRP. Surprisingly, unlike systemic arteries, Kv7 activity in MCAs is not affected by the development of hypertension, and CGRP-mediated vasodilation is well maintained. As such, cerebrovascular Kv7 channels could be amenable for therapeutic targeting in conditions such as cerebral vasospasm.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/farmacología , Canales de Potasio KCNQ/metabolismo , Arteria Cerebral Media/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Hipertensión/metabolismo , Hipertensión/fisiopatología , Canales KATP/metabolismo , Canales de Potasio KCNQ/antagonistas & inhibidores , Canales de Potasio KCNQ/genética , Masculino , Arteria Cerebral Media/metabolismo , Arteria Cerebral Media/fisiopatología , Bloqueadores de los Canales de Potasio/farmacología , Isoformas de Proteínas , Interferencia de ARN , Ratas , Ratas Endogámicas SHR , Ratas Wistar , Transfección
18.
Bio Protoc ; 14(6): e4961, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38841287

RESUMEN

Understanding protein-protein interactions is crucial for unravelling subcellular protein distribution, contributing to our understanding of cellular organisation. Moreover, interaction studies can reveal insights into the mechanisms that cover protein trafficking within cells. Although various techniques such as Förster resonance energy transfer (FRET), co-immunoprecipitation, and fluorescence microscopy are commonly employed to detect protein interactions, their limitations have led to more advanced techniques such as the in situ proximity ligation assay (PLA) for spatial co-localisation analysis. The PLA technique, specifically employed in fixed cells and tissues, utilises species-specific secondary PLA probes linked to DNA oligonucleotides. When proteins are within 40 nm of each other, the DNA oligonucleotides on the probes interact, facilitating circular DNA formation through ligation. Rolling-circle amplification then produces DNA circles linked to the PLA probe. Fluorescently labelled oligonucleotides hybridise to the circles, generating detectable signals for precise co-localisation analysis. We employed PLA to examine the co-localisation of dynein with the Kv7.4 channel protein in isolated vascular smooth muscle cells from rat mesenteric arteries. This method enabled us to investigate whether Kv7.4 channels interact with dynein, thereby providing evidence of their retrograde transport by the microtubule network. Our findings illustrate that PLA is a valuable tool for studying potential novel protein interactions with dynein, and the quantifiable approach offers insights into whether these interactions are changed in disease.

19.
Sci Rep ; 14(1): 13321, 2024 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-38858497

RESUMEN

Myocardin-related transcription factors (MRTFs: myocardin/MYOCD, MRTF-A/MRTFA, and MRTF-B/MRTFB) suppress production of pro-inflammatory cytokines and chemokines in human smooth muscle cells (SMCs) through sequestration of RelA in the NF-κB complex, but additional mechanisms are likely involved. The cGAS-STING pathway is activated by double-stranded DNA in the cytosolic compartment and acts through TANK-binding kinase 1 (TBK1) to spark inflammation. The present study tested if MRTFs suppress inflammation also by targeting cGAS-STING signaling. Interrogation of a transcriptomic dataset where myocardin was overexpressed using a panel of 56 cGAS-STING cytokines showed the panel to be repressed. Moreover, MYOCD, MRTFA, and SRF associated negatively with the panel in human arteries. RT-qPCR in human bronchial SMCs showed that all MRTFs reduced pro-inflammatory cytokines on the panel. MRTFs diminished phosphorylation of TBK1, while STING phosphorylation was marginally affected. The TBK1 inhibitor amlexanox, but not the STING inhibitor H-151, reduced the anti-inflammatory effect of MRTF-A. Co-immunoprecipitation and proximity ligation assays supported binding between MRTF-A and TBK1 in SMCs. MRTFs thus appear to suppress cellular inflammation in part by acting on the kinase TBK1. This may defend SMCs against pro-inflammatory insults in disease.


Asunto(s)
Inflamación , Miocitos del Músculo Liso , Proteínas Nucleares , Proteínas Serina-Treonina Quinasas , Transactivadores , Humanos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Miocitos del Músculo Liso/metabolismo , Transactivadores/metabolismo , Transactivadores/genética , Inflamación/metabolismo , Inflamación/patología , Transducción de Señal , Citocinas/metabolismo , Fosforilación , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Células Cultivadas
20.
Commun Biol ; 7(1): 1059, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39198706

RESUMEN

Pain and inflammation contribute immeasurably to reduced quality of life, yet modern analgesic and anti-inflammatory therapeutics can cause dependence and side effects. Here, we screened 1444 plant extracts, prepared primarily from native species in California and the United States Virgin Islands, against two voltage-gated K+ channels - T-cell expressed Kv1.3 and nociceptive-neuron expressed Kv7.2/7.3. A subset of extracts both inhibits Kv1.3 and activates Kv7.2/7.3 at hyperpolarized potentials, effects predicted to be anti-inflammatory and analgesic, respectively. Among the top dual hits are witch hazel and fireweed; polymodal modulation of multiple K+ channel types by hydrolysable tannins contributes to their dual anti-inflammatory, analgesic actions. In silico docking and mutagenesis data suggest pore-proximal extracellular linker sequence divergence underlies opposite effects of hydrolysable tannins on different Kv1 isoforms. The findings provide molecular insights into the enduring, widespread medicinal use of witch hazel and fireweed and demonstrate a screening strategy for discovering dual anti-inflammatory, analgesic small molecules.


Asunto(s)
Analgésicos , Antiinflamatorios , Extractos Vegetales , Antiinflamatorios/farmacología , Antiinflamatorios/química , Analgésicos/farmacología , Analgésicos/química , Animales , Extractos Vegetales/farmacología , Extractos Vegetales/química , Humanos , Ratones , Coriandrum/química , Simulación del Acoplamiento Molecular , Plantas Medicinales/química , Bloqueadores de los Canales de Potasio/farmacología , Bloqueadores de los Canales de Potasio/química , Masculino , Taninos/farmacología , Taninos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA