Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Hypertension ; 81(3): 561-571, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38354270

RESUMEN

BACKGROUND: Small arteries exhibit resting tone, a partially contracted state that maintains arterial blood pressure. In arterial smooth muscle cells, potassium channels control contraction and relaxation. Perivascular adipose tissue (PVAT) has been shown to exert anticontractile effects on the blood vessels. However, the mechanisms by which PVAT signals small arteries, and their relevance remain largely unknown. We aimed to uncover key molecular components in adipose-vascular coupling. METHODS: A wide spectrum of genetic mouse models targeting Kcnq3, Kcnq4, and Kcnq5 genes (Kcnq3-/-, Kcnq4-/-, Kcnq5-/-, Kcnq5dn/dn, Kcnq4-/-/Kcnq5dn/dn, and Kcnq4-/-/Kcnq5-/-), telemetry blood pressure measurements, targeted lipidomics, RNA-Seq profiling, wire-myography, patch-clamp, and sharp-electrode membrane potential measurements was used. RESULTS: We show that PVAT causes smooth muscle cell KV7.5 family of voltage-gated potassium (K+) channels to hyperpolarize the membrane potential. This effect relaxes small arteries and regulates blood pressure. Oxygenation of polyunsaturated fats generates oxylipins, a superclass of lipid mediators. We identified numerous oxylipins released by PVAT, which potentiate vasodilatory action in small arteries by opening smooth muscle cell KV7.5 family of voltage-gated potassium (K+) channels. CONCLUSIONS: Our results reveal a key molecular function of the KV7.5 family of voltage-gated potassium (K+) channels in the adipose-vascular coupling, translating PVAT signals, particularly oxylipins, to the central physiological function of vasoregulation. This novel pathway opens new therapeutic perspectives.


Asunto(s)
Oxilipinas , Vasodilatación , Animales , Ratones , Tejido Adiposo , Canales de Potasio KCNQ/genética , Canales de Potasio KCNQ/metabolismo , Oxilipinas/metabolismo , Potasio/metabolismo
2.
STAR Protoc ; 5(1): 102845, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38294910

RESUMEN

The isolated perfused kidney is a classic ex vivo preparation for studying renal physiology in general and vascular function. Here, we present a protocol for assessing myogenic tone in isolated mouse kidneys as well as vasodilatory and vasoconstrictive responses, expressed as perfusion pressure. We describe steps for pre-operative preparation, kidney and renal artery isolation, and connection of renal artery with glass cannula. We then detail how to measure pressure changes in perfused kidneys and the myogenic tone. For complete details on the use and execution of this protocol, please refer to Cui et al.1.


Asunto(s)
Riñón , Vasoconstricción , Ratones , Animales , Presión Sanguínea/fisiología , Perfusión
3.
Cell Mol Life Sci ; 80(9): 265, 2023 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-37615749

RESUMEN

Transient receptor potential cation channel-6 (TRPC6) gene mutations cause familial focal segmental glomerulosclerosis (FSGS), which is inherited as an autosomal dominant disease. In patients with TRPC6-related FSGS, all mutations map to the N- or C-terminal TRPC6 protein domains. Thus far, the majority of TRPC6 mutations are missense resulting in increased or decreased calcium influx; however, the fundamental molecular mechanisms causing cell injury and kidney pathology are unclear. We report a novel heterozygous TRPC6 mutation (V691Kfs*) in a large kindred with no signs of FSGS despite a largely truncated TRPC6 protein. We studied the molecular effects of V691Kfs* TRPC6 mutant using the tridimensional cryo-EM structure of the tetrameric TRPC6 protein. The results indicated that V691 is localized at the pore-forming transmembrane region affecting the ion conduction pathway, and predicted that V691Kfs* causes closure of the ion-conducting pathway leading to channel inactivation. We assessed the impact of V691Kfs* and two previously reported TRPC6 disease mutants (P112Q and G757D) on calcium influx in cells. Our data show that the V691Kfs* fully inactivated the TRCP6 channel-specific calcium influx consistent with a complete loss-of-function phenotype. Furthermore, the V691Kfs* truncation exerted a dominant negative effect on the full-length TRPC6 proteins. In conclusion, the V691Kfs* non-functional truncated TRPC6 is not sufficient to cause FSGS. Our data corroborate recently characterized TRPC6 loss-of-function and gain-of-function mutants suggesting that one defective TRPC6 gene copy is not sufficient to cause FSGS. We underscore the importance of increased rather than reduced calcium influx through TRPC6 for podocyte cell death.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria , Humanos , Glomeruloesclerosis Focal y Segmentaria/genética , Canal Catiónico TRPC6/genética , Calcio , Mutación con Pérdida de Función , Mutación/genética
4.
Ageing Res Rev ; 84: 101813, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36470339

RESUMEN

The arterial myogenic response is an inherent property of resistance arteries. Myogenic tone is crucial for maintaining a relatively constant blood flow in response to changes in intraluminal pressure and protects delicate organs from excessive blood flow. Although this fundamental physiological phenomenon has been extensively studied, the underlying molecular mechanisms are largely unknown. Recent studies identified a crucial role of mechano-activated angiotensin II type 1 receptors (AT1R) in this process. The development of myogenic response is affected by aging. In this review, we summarize recent progress made to understand the role of AT1R and other mechanosensors in the control of arterial myogenic response. We discuss age-related alterations in myogenic response and possible underlying mechanisms and implications for healthy aging.


Asunto(s)
Envejecimiento , Arterias , Animales , Humanos , Ratones , Arterias/metabolismo , Arterias/fisiología , Ratones Noqueados , Envejecimiento/metabolismo , Envejecimiento/patología
5.
J Am Heart Assoc ; 11(4): e022070, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35132870

RESUMEN

Background Blood pressure and tissue perfusion are controlled in part by the level of intrinsic (myogenic) arterial tone. However, many of the molecular determinants of this response are unknown. We previously found that mice with targeted disruption of the gene encoding the angiotensin II type 1a receptor (AT1AR) (Agtr1a), the major murine angiotensin II type 1 receptor (AT1R) isoform, showed reduced myogenic tone; however, uncontrolled genetic events (in this case, gene ablation) can lead to phenotypes that are difficult or impossible to interpret. Methods and Results We tested the mechanosensitive function of AT1R using tamoxifen-inducible smooth muscle-specific AT1aR knockout (smooth muscle-Agtr1a-/-) mice and studied downstream signaling cascades mediated by Gq/11 and/or ß-arrestins. FR900359, Sar1Ile4Ile8-angiotensin II (SII), TRV120027 and TRV120055 were used as selective Gq/11 inhibitor and biased agonists to activate noncanonical ß-arrestin and canonical Gq/11 signaling of the AT1R, respectively. Myogenic and Ang II-induced constrictions were diminished in the perfused renal vasculature, mesenteric and cerebral arteries of smooth muscle-Agtr1a-/- mice. Similar effects were observed in arteries of global mutant Agtr1a-/- but not Agtr1b-/- mice. FR900359 decreased myogenic tone and angiotensin II-induced constrictions whereas selective biased targeting of AT1R-ß-arrestin signaling pathways had no effects. Conclusions This study demonstrates that myogenic arterial constriction requires Gq/11-dependent signaling pathways of mechanoactivated AT1R but not G protein-independent, noncanonical pathways in smooth muscle cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Receptor de Angiotensina Tipo 1 , Vasoconstricción , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Angiotensina II/metabolismo , Animales , Arterias Cerebrales/metabolismo , Ratones , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , beta-Arrestinas/metabolismo
6.
Front Physiol ; 12: 749709, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34899382

RESUMEN

Aging is an independent risk factor for hypertension, cardiovascular morbidity, and mortality. However, detailed mechanisms linking aging to cardiovascular disease are unclear. We studied the aging effects on the role of perivascular adipose tissue and downstream vasoconstriction targets, voltage-dependent KV7 channels, and their pharmacological modulators (flupirtine, retigabine, QO58, and QO58-lysine) in a murine model. We assessed vascular function of young and old mesenteric arteries in vitro using wire myography and membrane potential measurements with sharp electrodes. We also performed bulk RNA sequencing and quantitative reverse transcription-polymerase chain reaction tests in mesenteric arteries and perivascular adipose tissue to elucidate molecular underpinnings of age-related phenotypes. Results revealed impaired perivascular adipose tissue-mediated control of vascular tone particularly via KV7.3-5 channels with increased age through metabolic and inflammatory processes and release of perivascular adipose tissue-derived relaxation factors. Moreover, QO58 was identified as novel pharmacological vasodilator to activate XE991-sensitive KCNQ channels in old mesenteric arteries. Our data suggest that targeting inflammation and metabolism in perivascular adipose tissue could represent novel approaches to restore vascular function during aging. Furthermore, KV7.3-5 channels represent a promising target in cardiovascular aging.

8.
Aging Cell ; 19(4): e13134, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32187825

RESUMEN

Caveolae position CaV 3.2 (T-type Ca2+ channel encoded by the α-3.2 subunit) sufficiently close to RyR (ryanodine receptors) for extracellular Ca2+ influx to trigger Ca2+ sparks and large-conductance Ca2+ -activated K+ channel feedback in vascular smooth muscle. We hypothesize that this mechanism of Ca2+ spark generation is affected by age. Using smooth muscle cells (VSMCs) from mouse mesenteric arteries, we found that both Cav 3.2 channel inhibition by Ni2+ (50 µM) and caveolae disruption by methyl-ß-cyclodextrin or genetic abolition of Eps15 homology domain-containing protein (EHD2) inhibited Ca2+ sparks in cells from young (4 months) but not old (12 months) mice. In accordance, expression of Cav 3.2 channel was higher in mesenteric arteries from young than old mice. Similar effects were observed for caveolae density. Using SMAKO Cav 1.2-/- mice, caffeine (RyR activator) and thapsigargin (Ca2+ transport ATPase inhibitor), we found that sufficient SR Ca2+ load is a prerequisite for the CaV 3.2-RyR axis to generate Ca2+ sparks. We identified a fraction of Ca2+ sparks in aged VSMCs, which is sensitive to the TRP channel blocker Gd3+ (100 µM), but insensitive to CaV 1.2 and CaV 3.2 channel blockade. Our data demonstrate that the VSMC CaV 3.2-RyR axis is down-regulated by aging. This defective CaV 3.2-RyR coupling is counterbalanced by a Gd3+ sensitive Ca2+ pathway providing compensatory Ca2+ influx for triggering Ca2+ sparks in aged VSMCs.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Senescencia Celular , Músculo Liso Vascular/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Canales de Calcio Tipo T/deficiencia , Masculino , Ratones , Ratones Noqueados , Músculo Liso Vascular/citología
9.
Br J Pharmacol ; 177(5): 1164-1186, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31658366

RESUMEN

BACKGROUND AND PURPOSE: BK channels play important roles in various physiological and pathophysiological processes and thus have been the target of several drug development programmes focused on creating new efficacious BK channel openers, such as the GoSlo-SR compounds. However, the effect of GoSlo-SR compounds on vascular smooth muscle has not been studied. Therefore, we tested the hypothesis that GoSlo-SR compounds dilate arteries exclusively by activating BK channels. EXPERIMENTAL APPROACH: Experiments were performed on rat Gracilis muscle, saphenous, mesenteric and tail arteries using isobaric and isometric myography, sharp microelectrodes, digital droplet PCR and the patch-clamp technique. KEY RESULTS: GoSlo-SR compounds dilated isobaric and relaxed and hyperpolarised isometric vessel preparations and their effects were abolished after (a) functionally eliminating K+ channels by pre-constriction with 50 mM KCl or (b) blocking all K+ channels known to be expressed in vascular smooth muscle. However, these effects were not blocked when BK channels were inhibited. Surprisingly, the Kv 7 channel inhibitor XE991 reduced their effects considerably, but neither Kv 1 nor Kv 2 channel blockers altered the inhibitory effects of GoSlo-SR. However, the combined blockade of BK and Kv 7 channels abolished the GoSlo-SR-induced relaxation. GoSlo-SR compounds also activated Kv 7.4 and Kv 7.5 channels expressed in HEK 293 cells. CONCLUSION AND IMPLICATIONS: This study shows that GoSlo-SR compounds are effective relaxants in vascular smooth muscle and mediate their effects by a combined activation of BK and Kv 7.4/Kv 7.5 channels. Activation of Kv 1, Kv 2 or Kv 7.1 channels or other vasodilator pathways seems not to be involved.


Asunto(s)
Canales de Potasio de Gran Conductancia Activados por el Calcio , Vasodilatación , Animales , Arterias , Células HEK293 , Humanos , Músculo Esquelético , Ratas
10.
Channels (Austin) ; 13(1): 505-519, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31797713

RESUMEN

Vascular smooth muscle cells (VSMCs) of small peripheral arteries contribute to blood pressure control by adapting their contractile state. These adaptations depend on the VSMC cytosolic Ca2+ concentration, regulated by complex local elementary Ca2+ signaling pathways. Ca2+ sparks represent local, transient, rapid calcium release events from a cluster of ryanodine receptors (RyRs) in the sarcoplasmic reticulum. In arterial SMCs, Ca2+ sparks activate nearby calcium-dependent potassium channels, cause membrane hyperpolarization and thus decrease the global intracellular [Ca2+] to oppose vasoconstriction. Arterial SMC Cav1.2 L-type channels regulate intracellular calcium stores content, which in turn modulates calcium efflux through RyRs. Cav3.2 T-type channels contribute to a minor extend to Ca2+ spark generation in certain types of arteries. Their localization within cell membrane caveolae is essential. We summarize present data on local elementary calcium signaling (Ca2+ sparks) in arterial SMCs with focus on RyR isoforms, large-conductance calcium-dependent potassium (BKCa) channels, and cell membrane-bound calcium channels (Cav1.2 and Cav3.2), particularly in caveolar microdomains.


Asunto(s)
Arterias/metabolismo , Calcio/metabolismo , Músculo Liso Vascular/metabolismo , Animales , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo T/genética , Canales de Calcio Tipo T/metabolismo , Señalización del Calcio , Humanos
11.
Front Med (Lausanne) ; 6: 126, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31281816

RESUMEN

Perivascular adipose tissue (PVAT) is implicated in the pathophysiology of cardiovascular disease, especially in obese individuals in which the quantity of renal and visceral PVAT is markedly increased. The control of arterial tone by PVAT has emerged as a relatively new field of experimental hypertension research. The discovery of this prototype of vasoregulation has been mostly inferred from data obtained using wire myography. Currently, there is a major discussion on distinguishing between biological vs. technical replicates in biomedical studies, which resulted in numerous guidelines being published on planning studies and publishing data by societies, journals, and associations. Experimental study designs are determined depending on how the experimentator distinguishes between biological vs. technical replicates. These definitions determine the ultimate standards required for making submissions to certain journals. In this article, we examine possible outcomes of different experimental study designs on PVAT control of arterial tone using isolated arteries. Based on experimental data, we determine the sample size and power of statistical analyses for such experiments. We discuss whether n-values should correspond to the number of arterial rings and analyze the resulting effects if those numbers are averaged to provide a single N-value per animal, or whether the hierarchical statistical method represents an alternative for analyzing such kind of data. Our analyses show that that the data (logEC50) from (+) PVAT to (-) PVAT arteries are clustered. Intraclass correlation (ICC) was 31.4%. Moreover, it appeared that the hierarchical approach was better than regular statistical tests as the analyses revealed by a better goodness of fit (v2-2LL test). Based on our results, we propose to use at least three independent arterial rings from each from three animals or at least seven arterial rings from each from two animals for each group, i.e., (+) PVAT vs. (-) PVAT. Finally, we discuss a clinical situation where distinguishing between biological vs. technical replicates can lead to absurd situations in clinical decision makings. We conclude that discrimination between biological vs. technical replicates is helpful in experimental studies but is difficult to implement in everyday's clinical practice.

12.
Front Med (Lausanne) ; 6: 153, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31355199

RESUMEN

Caveolae, flask-shaped cholesterol-, and glycosphingolipid-rich membrane microdomains, contain caveolin 1, 2, 3 and several structural proteins, in particular Cavin 1-4, EHD2, pacsin2, and dynamin 2. Caveolae participate in several physiological processes like lipid uptake, mechanosensitivity, or signaling events and are involved in pathophysiological changes in the cardiovascular system. They serve as a specific membrane platform for a diverse set of signaling molecules like endothelial nitric oxide synthase (eNOS), and further maintain vascular homeostasis. Lack of caveolins causes the complete loss of caveolae; induces vascular disorders, endothelial dysfunction, and impaired myogenic tone; and alters numerous cellular processes, which all contribute to an increased risk for hypertension. This brief review describes our current knowledge on caveolae in vasculature, with special focus on their pathophysiological role in hypertension.

13.
J Am Heart Assoc ; 8(9): e010090, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-31030596

RESUMEN

Background Hypertension is the major risk factor for cardiovascular disease, the most common cause of death worldwide. Resistance arteries are capable of adapting their diameter independently in response to pressure and flow-associated shear stress. Ryanodine receptors (RyRs) are major Ca2+-release channels in the sarcoplasmic reticulum membrane of myocytes that contribute to the regulation of contractility. Vascular smooth muscle cells exhibit 3 different RyR isoforms (RyR1, RyR2, and RyR3), but the impact of individual RyR isoforms on adaptive vascular responses is largely unknown. Herein, we generated tamoxifen-inducible smooth muscle cell-specific RyR2-deficient mice and tested the hypothesis that vascular smooth muscle cell RyR2s play a specific role in elementary Ca2+ signaling and adaptive vascular responses to vascular pressure and/or flow. Methods and Results Targeted deletion of the Ryr2 gene resulted in a complete loss of sarcoplasmic reticulum-mediated Ca2+-release events and associated Ca2+-activated, large-conductance K+ channel currents in peripheral arteries, leading to increased myogenic tone and systemic blood pressure. In the absence of RyR2, the pulmonary artery pressure response to sustained hypoxia was enhanced, but flow-dependent effects, including blood flow recovery in ischemic hind limbs, were unaffected. Conclusions Our results establish that RyR2-mediated Ca2+-release events in VSCM s specifically regulate myogenic tone (systemic circulation) and arterial adaptation in response to changes in pressure (hypoxic lung model), but not flow. They further suggest that vascular smooth muscle cell-expressed RyR2 deserves scrutiny as a therapeutic target for the treatment of vascular responses in hypertension and chronic vascular diseases.


Asunto(s)
Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Retículo Sarcoplasmático/metabolismo , Animales , Aorta/metabolismo , Aorta/fisiopatología , Arterias/metabolismo , Arterias/fisiopatología , Presión Sanguínea/fisiología , Señalización del Calcio , Miembro Posterior/irrigación sanguínea , Hipoxia/metabolismo , Hipoxia/fisiopatología , Flujometría por Láser-Doppler , Pulmón/irrigación sanguínea , Ratones , Ratones Noqueados , Músculo Liso Vascular/fisiopatología , Miografía , Técnicas de Placa-Clamp , Inhibidores de Fosfodiesterasa/farmacología , Arteria Pulmonar/metabolismo , Arteria Pulmonar/fisiopatología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Vasoconstricción
14.
Arterioscler Thromb Vasc Biol ; 38(10): 2371-2381, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30354206

RESUMEN

Objective- This study examined whether caveolae position CaV3.2 (T-type Ca2+ channel encoded by the α-3.2 subunit) sufficiently close to RyR (ryanodine receptors) for extracellular Ca2+ influx to trigger Ca2+ sparks and large-conductance Ca2+-activated K+ channel feedback. Approach and Results- Using smooth muscle cells from mouse mesenteric arteries, the proximity ligation assay confirmed that CaV3.2 reside within 40 nm of caveolin 1, a key caveolae protein. Methyl-ß-cyclodextrin, a cholesterol depleting agent that disrupts caveolae, suppressed CaV3.2 activity along with large-conductance Ca2+-activated K+-mediated spontaneous transient outward currents in cells from C57BL/6 but not CaV3.2-/- mice. Genetic deletion of caveolin 1, a perturbation that prevents caveolae formation, also impaired spontaneous transient outward current production but did so without impairing Ca2+ channel activity, including CaV3.2. These observations indicate a mistargeting of CaV3.2 in caveolin 1-/- mice, a view supported by a loss of Ni2+-sensitive Ca2+ spark generation and colocalization signal (CaV3.2-RyR) from the proximity ligation assay. Vasomotor and membrane potential measurements confirmed that cellular disruption of the CaV3.2-RyR axis functionally impaired the ability of large-conductance Ca2+-activated K+ to set tone in pressurized caveolin 1-/- arteries. Conclusions- Caveolae play a critical role in protein targeting and preserving the close structural relationship between CaV3.2 and RyR needed to drive negative feedback control in resistance arteries.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Señalización del Calcio , Caveolas/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Canales de Calcio Tipo T/deficiencia , Canales de Calcio Tipo T/genética , Caveolina 1/genética , Caveolina 1/metabolismo , Retroalimentación Fisiológica , Masculino , Potenciales de la Membrana , Arterias Mesentéricas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Vasoconstricción , Vasodilatación
15.
J Physiol ; 596(20): 4863-4877, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30146760

RESUMEN

KEY POINTS: In arterial smooth muscle, Ca2+ sparks are elementary Ca2+ -release events generated by ryanodine receptors (RyRs) to cause vasodilatation by opening maxi Ca2+ -sensitive K+ (BKCa ) channels. This study elucidated the contribution of T-type Cav 3.2 channels in caveolae and their functional interaction with L-type Cav 1.2 channels to trigger Ca2+ sparks in vascular smooth muscle cells (VSMCs). Our data demonstrate that L-type Cav 1.2 channels provide the predominant Ca2+ pathway for the generation of Ca2+ sparks in murine arterial VSMCs. T-type Cav 3.2 channels represent an additional source for generation of VSMC Ca2+ sparks. They are located in pit structures of caveolae to provide locally restricted, tight coupling between T-type Cav 3.2 channels and RyRs to ignite Ca2+ sparks. ABSTRACT: Recent data suggest that T-type Cav 3.2 channels in arterial vascular smooth muscle cells (VSMCs) and pits structure of caveolae could contribute to elementary Ca2+ signalling (Ca2+ sparks) via ryanodine receptors (RyRs) to cause vasodilatation. While plausible, their precise involvement in igniting Ca2+ sparks remains largely unexplored. The goal of this study was to elucidate the contribution of caveolar Cav 3.2 channels and their functional interaction with Cav 1.2 channels to trigger Ca2+ sparks in VSMCs from mesenteric, tibial and cerebral arteries. We used tamoxifen-inducible smooth muscle-specific Cav 1.2-/- (SMAKO) mice and laser scanning confocal microscopy to assess Ca2+ spark generation in VSMCs. Ni2+ , Cd2+ and methyl-ß-cyclodextrin were used to inhibit Cav 3.2 channels, Cav 1.2 channels and caveolae, respectively. Ni2+ (50 µmol L-1 ) and methyl-ß-cyclodextrin (10 mmol L-1 ) decreased Ca2+ spark frequency by ∼20-30% in mesenteric VSMCs in a non-additive manner, but failed to inhibit Ca2+ sparks in tibial and cerebral artery VSMCs. Cd2+ (200 µmol L-1 ) suppressed Ca2+ sparks in mesenteric arteries by ∼70-80%. A similar suppression of Ca2+ sparks was seen in mesenteric artery VSMCs of SMAKO mice. The remaining Ca2+ sparks were fully abolished by Ni2+ or methyl-ß-cyclodextrin. Our data demonstrate that Ca2+ influx through CaV 1.2 channels is the primary means of triggering Ca2+ sparks in murine arterial VSMCs. CaV 3.2 channels, localized to caveolae and tightly coupled to RyR, provide an additional Ca2+ source for Ca2+ spark generation in mesenteric, but not tibial and cerebral, arteries.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo T/metabolismo , Señalización del Calcio , Arterias Mesentéricas/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Caveolas/metabolismo , Células Cultivadas , Masculino , Arterias Mesentéricas/citología , Ratones , Miocitos del Músculo Liso/metabolismo
16.
Sci Rep ; 8(1): 4878, 2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29559678

RESUMEN

Transient receptor potential vanilloid 4 (TRPV4) cation channels are functional in all renal vascular segments and mediate endothelium-dependent vasorelaxation. Moreover, they are expressed in distinct parts of the tubular system and activated by cell swelling. Ischaemia/reperfusion injury (IRI) is characterized by tubular injury and endothelial dysfunction. Therefore, we hypothesised a putative organ protective role of TRPV4 in acute renal IRI. IRI was induced in TRPV4 deficient (Trpv4 KO) and wild-type (WT) control mice by clipping the left renal pedicle after right-sided nephrectomy. Serum creatinine level was higher in Trpv4 KO mice 6 and 24 hours after ischaemia compared to WT mice. Detailed histological analysis revealed that IRI caused aggravated renal tubular damage in Trpv4 KO mice, especially in the renal cortex. Immunohistological and functional assessment confirmed TRPV4 expression in proximal tubular cells. Furthermore, the tubular damage could be attributed to enhanced necrosis rather than apoptosis. Surprisingly, the percentage of infiltrating granulocytes and macrophages were comparable in IRI-damaged kidneys of Trpv4 KO and WT mice. The present results suggest a renoprotective role of TRPV4 during acute renal IRI. Further studies using cell-specific TRPV4 deficient mice are needed to clarify cellular mechanisms of TRPV4 in IRI.


Asunto(s)
Túbulos Renales/metabolismo , Daño por Reperfusión/metabolismo , Canales Catiónicos TRPV/deficiencia , Lesión Renal Aguda/metabolismo , Animales , Apoptosis , Modelos Animales de Enfermedad , Isquemia/patología , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Noqueados , Reperfusión/métodos , Daño por Reperfusión/genética , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo
17.
J Am Heart Assoc ; 7(7)2018 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-29574460

RESUMEN

BACKGROUND: Multiple studies have shown that an NO-induced activation of vascular smooth muscle BK channels contributes to the NO-evoked dilation in many blood vessels. In vivo, NO is released continuously. NO attenuates vessel constrictions and, therefore, exerts an anticontractile effect. It is unknown whether the anticontractile effect of continuously present NO is mediated by BK channels. METHODS AND RESULTS: This study tested the hypothesis that BK channels mediate the vasodilatory effect of continuously present NO. Experiments were performed on rat and mouse tail and rat saphenous arteries using isometric myography and FURA-2 fluorimetry. Continuously present NO donors, as well as endogenous NO, attenuated methoxamine-induced vasoconstrictions. This effect was augmented in the presence of the BK channel blocker iberiotoxin. Moreover, the contractile effect of iberiotoxin was reduced in the presence of NO donors. The effect of the NO donor sodium nitroprusside was abolished by an NO scavenger and by a guanylyl cyclase inhibitor. In addition, the effect of sodium nitroprusside was reduced considerably by a protein kinase G inhibitor, but was not altered by inhibition of H2S generation. Sodium nitroprusside attenuated the intracellular calcium concentration response to methoxamine. Furthermore, sodium nitroprusside strongly reduced methoxamine-induced calcium influx, which depends entirely on L-type calcium channels. It did not affect methoxamine-induced calcium release. CONCLUSIONS: In summary, this study demonstrates the following: (1) continuously present NO evokes a strong anticontractile effect on rat and mouse arteries; (2) the iberiotoxin-induced augmentation of the effect of NO is associated with an NO-induced reduction of the effect of iberiotoxin; and (3) NO evoked a reduction of calcium influx via L-type calcium channels.


Asunto(s)
Arterias/metabolismo , Extremidades/irrigación sanguínea , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Óxido Nítrico/metabolismo , Potasio/metabolismo , Cola (estructura animal)/irrigación sanguínea , Vasodilatación , Animales , Arterias/efectos de los fármacos , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Donantes de Óxido Nítrico/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Ratas Wistar , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
18.
Br J Pharmacol ; 174(2): 150-162, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28000293

RESUMEN

BACKGROUND AND PURPOSE: KV 7.1 voltage-gated potassium channels are expressed in vascular smooth muscle cells (VSMC) of diverse arteries, including mesenteric arteries. Based on pharmacological evidence using R-L3 (KV 7.1 channel opener), HMR1556, chromanol 293B (KV 7.1 channel blockers), stimulation of these channels has been suggested to evoke profound relaxation in various vascular beds of rats. However, the specificity of these drugs in vivo is uncertain. EXPERIMENTAL APPROACH: We used Kcnq1-/- mice and pharmacological tools to determine whether KV 7.1 channels play a role in the regulation of arterial tone. KEY RESULTS: R-L3 produced similar concentration-dependent relaxations (EC50  ~ 1.4 µM) of arteries from wild-type (Kcnq1+/+ ) and Kcnq1-/- mice, pre-contracted with either phenylephrine or 60 mM KCl. This relaxation was not affected by 10 µM chromanol 293B, 10 µM HMR1556 or 30 µM XE991 (pan-KV 7 channel blocker). The anti-contractile effects of the perivascular adipose tissue (PVAT) were normal in Kcnq1-/- arteries. Chromanol 293B and HMR1556 did not affect the anti-contractile effects of (PVAT). Isolated VSMCs from Kcnq1-/- mice exhibited normal peak KV currents. The KV 7.2-5 channel opener retigabine caused similar relaxations in Kcnq1-/- and wild-type vessels. CONCLUSION AND IMPLICATIONS: We conclude that KV 7.1 channels were apparently not involved in the control of arterial tone by α1 -adrenoceptor agonists and PVAT. In addition, R-L3 is an inappropriate pharmacological tool for studying the function of native vascular KV 7.1 channels in mice.


Asunto(s)
Aorta/metabolismo , Canal de Potasio KCNQ1/metabolismo , Animales , Aorta/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Canal de Potasio KCNQ1/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Piperidinas/farmacología , Relación Estructura-Actividad , Tiazoles/farmacología , Compuestos de Tosilo/farmacología
19.
Front Physiol ; 7: 335, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27540364

RESUMEN

The anti-contractile effect of perivascular adipose tissue (PVAT) is an important mechanism in the modulation of vascular tone in peripheral arteries. Recent evidence has implicated the XE991-sensitive voltage-gated KV (KCNQ) channels in the regulation of arterial tone by PVAT. However, until now the in vivo pharmacology of the involved vascular KV channels with regard to XE991 remains undetermined, since XE991 effects may involve Ca(2+) activated BKCa channels and/or voltage-dependent KV1.5 channels sensitive to diphenyl phosphine oxide-1 (DPO-1). In this study, we tested whether KV1.5 channels are involved in the control of mesenteric arterial tone and its regulation by PVAT. Our study was also aimed at extending our current knowledge on the in situ vascular pharmacology of DPO-1 and XE991 regarding KV1.5 and BKCa channels, in helping to identify the nature of K(+) channels that could contribute to PVAT-mediated relaxation. XE991 at 30 µM reduced the anti-contractile response of PVAT, but had no effects on vasocontraction induced by phenylephrine (PE) in the absence of PVAT. Similar effects were observed for XE991 at 0.3 µM, which is known to almost completely inhibit mesenteric artery VSMC KV currents. 30 µM XE991 did not affect BKCa currents in VSMCs. Kcna5 (-/-) arteries and wild-type arteries incubated with 1 µM DPO-1 showed normal vasocontractions in response to PE in the presence and absence of PVAT. KV current density and inhibition by 30 µM XE991 were normal in mesenteric artery VSMCs isolated from Kcna5 (-/-) mice. We conclude that KV channels are involved in the control of arterial vascular tone by PVAT. These channels are present in VSMCs and very potently inhibited by the KCNQ channel blocker XE991. BKCa channels and/or DPO-1 sensitive KV1.5 channels in VSMCs are not the downstream mediators of the XE991 effects on PVAT-dependent arterial vasorelaxation. Further studies will need to be undertaken to examine the role of other KV channels in the phenomenon.

20.
Sci Rep ; 6: 27517, 2016 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-27273292

RESUMEN

Hydrogen sulfide (H2S) is known to act protectively during renal ischemia/reperfusion injury (IRI). However, the role of the endogenous H2S in acute kidney injury (AKI) is largely unclear. Here, we analyzed the role of cystathionine gamma-lyase (CTH) in acute renal IRI using CTH-deficient (Cth(-/-)) mice whose renal H2S levels were approximately 50% of control (wild-type) mice. Although levels of serum creatinine and renal expression of AKI marker proteins were equivalent between Cth(-/-) and control mice, histological analysis revealed that IRI caused less renal tubular damage in Cth(-/-) mice. Flow cytometric analysis revealed that renal population of infiltrated granulocytes/macrophages was equivalent in these mice. However, renal expression levels of certain inflammatory cytokines/adhesion molecules believed to play a role in IRI were found to be lower after IRI only in Cth(-/-) mice. Our results indicate that the systemic CTH loss does not deteriorate but rather ameliorates the immediate AKI outcome probably due to reduced inflammatory responses in the kidney. The renal expression of CTH and other H2S-producing enzymes was markedly suppressed after IRI, which could be an integrated adaptive response for renal cell protection.


Asunto(s)
Lesión Renal Aguda/enzimología , Cistationina gamma-Liasa/metabolismo , Animales , Cistationina gamma-Liasa/genética , Sulfuro de Hidrógeno/metabolismo , Riñón/metabolismo , Ratones , Ratones Transgénicos , ARN Mensajero/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...