Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Cell Physiol ; 238(5): 976-991, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36852591

RESUMEN

Voltage-dependent potassium channel Kv1.3 plays a key role on T-cell activation; however, lack of reliable antibodies has prevented its accurate detection under endogenous circumstances. To overcome this limitation, we created a Jurkat T-cell line with endogenous Kv1.3 channel tagged, to determine the expression, location, and changes upon activation of the native Kv1.3 channels. CRISPR-Cas9 technique was used to insert a Flag-Myc peptide at the C terminus of the KCNA3 gene. Basal or activated channel expression was studied using western blot analysis and imaging techniques. We identified two isoforms of Kv1.3 other than the canonical channel (54 KDa) differing on their N terminus: a longer isoform (70 KDa) and a truncated isoform (43 KDa). All three isoforms were upregulated after T-cell activation. We focused on the functional characterization of the truncated isoform (short form, SF), because it has not been previously described and could be present in the available Kv1.3-/- mice models. Overexpression of SF in HEK cells elicited small amplitude Kv1.3-like currents, which, contrary to canonical Kv1.3, did not induce HEK proliferation. To explore the role of endogenous SF isoform in a native system, we generated both a knockout Jurkat clone and a clone expressing only the SF isoform. Although the canonical isoform (long form) localizes mainly at the plasma membrane, SF remains intracellular, accumulating perinuclearly. Accordingly, SF Jurkat cells did not show Kv1.3 currents and exhibited depolarized resting membrane potential (VM ), decreased Ca2+ influx, and a reduction in the [Ca2+ ]i increase upon stimulation. Functional characterization of these Kv1.3 channel isoforms showed their differential contribution to signaling pathways involved in formation of the immunological synapse. We conclude that alternative translation initiation generates at least three endogenous Kv1.3 channel isoforms in T cells that exhibit different functional roles. For some of these functions, Kv1.3 proteins do not need to form functional plasma membrane channels.


Asunto(s)
Canal de Potasio Kv1.3 , Animales , Humanos , Ratones , Línea Celular , Membrana Celular/metabolismo , Células Jurkat , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Canal de Potasio Kv1.3/genética , Canal de Potasio Kv1.3/metabolismo
2.
J Cell Physiol ; 236(6): 4330-4347, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33230847

RESUMEN

The voltage-dependent potassium channel Kv1.3 has been implicated in proliferation in many cell types, based on the observation that Kv1.3 blockers inhibited proliferation. By modulating membrane potential, cell volume, and/or Ca2+ influx, K+  channels can influence cell cycle progression. Also, noncanonical channel functions could contribute to modulate cell proliferation independent of K+ efflux. The specificity of the requirement of Kv1.3 channels for proliferation suggests the involvement of molecule-specific interactions, but the underlying mechanisms are poorly identified. Heterologous expression of Kv1.3 channels in HEK cells has been shown to increase proliferation independently of K+ fluxes. Likewise, some of the molecular determinants of Kv1.3-induced proliferation have been located in the C-terminus region, where individual point mutations of putative phosphorylation sites (Y447A and S459A) abolished Kv1.3-induced proliferation. Here, we investigated the mechanisms linking Kv1.3 channels to proliferation exploring the correlation between Kv1.3 voltage-dependent molecular dynamics and cell cycle progression. Using transfected HEK cells, we analyzed both the effect of changes in resting membrane potential on Kv1.3-induced proliferation and the effect of mutated Kv1.3 channels with altered voltage dependence of gating. We conclude that voltage-dependent transitions of Kv1.3 channels enable the activation of proliferative pathways. We also found that Kv1.3 associated with IQGAP3, a scaffold protein involved in proliferation, and that membrane depolarization facilitates their interaction. The functional contribution of Kv1.3-IQGAP3 interplay to cell proliferation was demonstrated both in HEK cells and in vascular smooth muscle cells. Our data indicate that voltage-dependent conformational changes of Kv1.3 are an essential element in Kv1.3-induced proliferation.


Asunto(s)
Proliferación Celular , Activación del Canal Iónico , Canal de Potasio Kv1.3/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Células HEK293 , Humanos , Canales KATP/genética , Canales KATP/metabolismo , Canal de Potasio Kv1.3/química , Canal de Potasio Kv1.3/genética , Potenciales de la Membrana , Mutación , Conformación Proteica , Transducción de Señal , Relación Estructura-Actividad
3.
Arterioscler Thromb Vasc Biol ; 39(12): e273-e286, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31597447

RESUMEN

OBJECTIVE: We have previously described that changes in the expression of Kv channels associate to phenotypic modulation (PM), so that Kv1.3/Kv1.5 ratio is a landmark of vascular smooth muscle cells phenotype. Moreover, we demonstrated that the Kv1.3 functional expression is relevant for PM in several types of vascular lesions. Here, we explore the efficacy of Kv1.3 inhibition for the prevention of remodeling in human vessels, and the mechanisms linking the switch in Kv1.3 /Kv1.5 ratio to PM. Approach and Results: Vascular remodeling was explored using organ culture and primary cultures of vascular smooth muscle cells obtained from human vessels. We studied the effects of Kv1.3 inhibition on serum-induced remodeling, as well as the impact of viral vector-mediated overexpression of Kv channels or myocardin knock-down. Kv1.3 blockade prevented remodeling by inhibiting proliferation, migration, and extracellular matrix secretion. PM activated Kv1.3 via downregulation of Kv1.5. Hence, both Kv1.3 blockers and Kv1.5 overexpression inhibited remodeling in a nonadditive fashion. Finally, myocardin knock-down induced vessel remodeling and Kv1.5 downregulation and myocardin overexpression increased Kv1.5, while Kv1.5 overexpression inhibited PM without changing myocardin expression. CONCLUSIONS: We demonstrate that Kv1.5 channel gene is a myocardin-regulated, vascular smooth muscle cells contractile marker. Kv1.5 downregulation upon PM leaves Kv1.3 as the dominant Kv1 channel expressed in dedifferentiated cells. We demonstrated that the inhibition of Kv1.3 channel function with selective blockers or by preventing Kv1.5 downregulation can represent an effective, novel strategy for the prevention of intimal hyperplasia and restenosis of the human vessels used for coronary angioplasty procedures.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , Vasos Coronarios/patología , Regulación de la Expresión Génica , Canal de Potasio Kv1.3/genética , Canal de Potasio Kv1.5/genética , Músculo Liso Vascular/metabolismo , Proteínas Nucleares/genética , Transactivadores/genética , Células Cultivadas , Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/patología , Vasos Coronarios/metabolismo , Vasos Coronarios/fisiopatología , Humanos , Inmunohistoquímica , Canal de Potasio Kv1.3/antagonistas & inhibidores , Canal de Potasio Kv1.3/biosíntesis , Canal de Potasio Kv1.5/biosíntesis , Músculo Liso Vascular/patología , Proteínas Nucleares/biosíntesis , Técnicas de Cultivo de Órganos , Fenotipo , ARN/genética , Transactivadores/biosíntesis , Remodelación Vascular
4.
Am J Physiol Cell Physiol ; 314(1): C27-C42, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28931540

RESUMEN

Kv1.3 channels are involved in the switch to proliferation of normally quiescent cells, being implicated in the control of cell cycle in many different cell types and in many different ways. They modulate membrane potential controlling K+ fluxes, sense changes in potential, and interact with many signaling molecules through their intracellular domains. From a mechanistic point of view, we can describe the role of Kv1.3 channels in proliferation with at least three different models. In the "membrane potential model," membrane hyperpolarization resulting from Kv1.3 activation provides the driving force for Ca2+ influx required to activate Ca2+-dependent transcription. This model explains most of the data obtained from several cells from the immune system. In the "voltage sensor model," Kv1.3 channels serve mainly as sensors that transduce electrical signals into biochemical cascades, independently of their effect on membrane potential. Kv1.3-dependent proliferation of vascular smooth muscle cells (VSMCs) could fit this model. Finally, in the "channelosome balance model," the master switch determining proliferation may be related to the control of the Kv1.3 to Kv1.5 ratio, as described in glial cells and also in VSMCs. Since the three mechanisms cannot function independently, these models are obviously not exclusive. Nevertheless, they could be exploited differentially in different cells and tissues. This large functional flexibility of Kv1.3 channels surely gives a new perspective on their functions beyond their elementary role as ion channels, although a conclusive picture of the mechanisms involved in Kv1.3 signaling to proliferation is yet to be reached.


Asunto(s)
Proliferación Celular , Canal de Potasio Kv1.3/metabolismo , Potasio/metabolismo , Animales , Señalización del Calcio , Proliferación Celular/efectos de los fármacos , Humanos , Activación del Canal Iónico , Canal de Potasio Kv1.3/antagonistas & inhibidores , Canal de Potasio Kv1.3/química , Canal de Potasio Kv1.3/genética , Potenciales de la Membrana , Modelos Biológicos , Bloqueadores de los Canales de Potasio/farmacología , Conformación Proteica , Relación Estructura-Actividad
5.
Microcirculation ; 25(1)2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29110368

RESUMEN

Kv channels are present in virtually all VSMCs and strongly influence contractile responses. However, they are also instrumental in the proliferative, migratory, and secretory functions of synthetic, dedifferentiated VSMCs upon PM. In fact, Kv channels not only contribute to all these processes but also are active players in the phenotypic switch itself. This review is focused on the role(s) of Kv channels in VSMC proliferation, which is one of the best characterized functions of dedifferentiated VSMCs. VSMC proliferation is a complex process requiring specific Kv channels at specific time and locations. Their identification is further complicated by their large diversity and the differences in expression across vascular beds. Of interest, both conserved changes in some Kv channels and vascular bed-specific regulation of others seem to coexist and participate in VSMC proliferation through complementary mechanisms. Such a system will add flexibility to the process while providing the required robustness to preserve this fundamental cellular response.


Asunto(s)
Proliferación Celular , Músculo Liso Vascular/citología , Canales de Potasio con Entrada de Voltaje/fisiología , Animales , Humanos , Remodelación Vascular
6.
J Physiol ; 595(5): 1497-1513, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27861908

RESUMEN

KEY POINTS: Canonical transient receptor potential (TRPC)3 and TRPC6 channels of vascular smooth muscle cells (VSMCs) mediate stretch- or agonist-induced cationic fluxes, contributing to membrane potential and vascular tone. Native TRPC3/C6 channels can form homo- or heterotetrameric complexes, which can hinder individual TRPC channel properties. The possibility that the differences in their association pattern may change their contribution to vascular tone in hypertension is unexplored. Functional characterization of heterologously expressed channels showed that TRPC6-containing complexes exhibited Pyr3/Pyr10-sensitive currents, whereas TRPC3-mediated currents were blocked by anti-TRPC3 antibodies. VSMCs from hypertensive (blood pressure high; BPH) mice have larger cationic basal currents insensitive to Pyr10 and sensitive to anti-TRPC3 antibodies. Consistently, myography studies showed a larger Pyr3/10-induced vasodilatation in BPN (blood pressure normal) mesenteric arteries. We conclude that the increased TRPC3 channel expression in BPH VSMCs leads to changes in TRPC3/C6 heteromultimeric assembly, with a higher TRPC3 channel contribution favouring depolarization of hypertensive VSMCs. ABSTRACT: Increased vascular tone in essential hypertension involves a sustained rise in total peripheral resistance. A model has been proposed in which the combination of membrane depolarization and higher L-type Ca2+ channel activity generates augmented Ca2+ influx into vascular smooth muscle cells (VSMCs), contraction and vasoconstriction. The search for culprit ion channels responsible for membrane depolarization has provided several candidates, including members of the canonical transient receptor potential (TRPC) family. TRPC3 and TRPC6 are diacylglycerol-activated, non-selective cationic channels contributing to stretch- or agonist-induced depolarization. Conflicting information exists regarding changes in TRPC3/TRPC6 functional expression in hypertension. However, although TRPC3-TRPC6 channels can heteromultimerize, the possibility that differences in their association pattern may change their functional contribution to vascular tone is largely unexplored. We probe this hypothesis using a model of essential hypertension (BPH mice; blood pressure high) and its normotensive control (BPN mice; blood pressure normal). First, non-selective cationic currents through homo- and heterotetramers recorded from transfected Chinese hamster ovary cells indicated that TRPC currents were sensitive to the selective antagonist Pyr10 only when TRPC6 was present, whereas intracellular anti-TRPC3 antibody selectively blocked TRPC3-mediated currents. In mesenteric VSMCs, basal and agonist-induced currents were more sensitive to Pyr3 and Pyr10 in BPN cells. Consistently, myography studies showed a larger Pyr3/10-induced vasodilatation in BPN mesenteric arteries. mRNA and protein expression data supported changes in TRPC3 and TRPC6 proportions and assembly, with a higher TRPC3 channel contribution in BPH VSMCs that could favour cell depolarization. These differences in functional and pharmacological properties of TRPC3 and TRPC6 channels, depending on their assembly, could represent novel therapeutical opportunities.


Asunto(s)
Hipertensión/fisiopatología , Miocitos del Músculo Liso/fisiología , Canales Catiónicos TRPC/fisiología , Animales , Aorta/fisiología , Células CHO , Cricetulus , Hipertensión Esencial , Arteria Femoral/fisiología , Arterias Mesentéricas/fisiología , Ratones , Músculo Liso Vascular/fisiología , Pirazoles/farmacología , Canales Catiónicos TRPC/genética , Canal Catiónico TRPC6
7.
J Biol Chem ; 291(7): 3569-80, 2016 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-26655221

RESUMEN

Changes in voltage-dependent potassium channels (Kv channels) associate to proliferation in many cell types, including transfected HEK293 cells. In this system Kv1.5 overexpression decreases proliferation, whereas Kv1.3 expression increases it independently of K(+) fluxes. To identify Kv1.3 domains involved in a proliferation-associated signaling mechanism(s), we constructed chimeric Kv1.3-Kv1.5 channels and point-mutant Kv1.3 channels, which were expressed as GFP- or cherry-fusion proteins. We studied their trafficking and functional expression, combining immunocytochemical and electrophysiological methods, and their impact on cell proliferation. We found that the C terminus is necessary for Kv1.3-induced proliferation. We distinguished two residues (Tyr-447 and Ser-459) whose mutation to alanine abolished proliferation. The insertion into Kv1.5 of a sequence comprising these two residues increased proliferation rate. Moreover, Kv1.3 voltage-dependent transitions from closed to open conformation induced MEK-ERK1/2-dependent Tyr-447 phosphorylation. We conclude that the mechanisms for Kv1.3-induced proliferation involve the accessibility of key docking sites at the C terminus. For one of these sites (Tyr-447) we demonstrated the contribution of MEK/ERK-dependent phosphorylation, which is regulated by voltage-induced conformational changes.


Asunto(s)
Canal de Potasio Kv1.3/agonistas , Sistema de Señalización de MAP Quinasas , Procesamiento Proteico-Postraduccional , Sustitución de Aminoácidos , Proliferación Celular , Células HEK293 , Humanos , Canal de Potasio Kv1.3/química , Canal de Potasio Kv1.3/genética , Canal de Potasio Kv1.3/metabolismo , Canal de Potasio Kv1.5/agonistas , Canal de Potasio Kv1.5/química , Canal de Potasio Kv1.5/genética , Canal de Potasio Kv1.5/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/antagonistas & inhibidores , MAP Quinasa Quinasa 2/genética , MAP Quinasa Quinasa 2/metabolismo , Mutagénesis Insercional , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fosforilación , Mutación Puntual , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Interferencia de ARN , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Tirosina/metabolismo
8.
Arterioscler Thromb Vasc Biol ; 35(6): 1489-97, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25857312

RESUMEN

OBJECTIVE: Actin dynamics in vascular smooth muscle is known to regulate contractile differentiation and may play a role in the pathogenesis of vascular disease. However, the list of genes regulated by actin polymerization in smooth muscle remains incomprehensive. Thus, the objective of this study was to identify actin-regulated genes in smooth muscle and to demonstrate the role of these genes in the regulation of vascular smooth muscle phenotype. APPROACH AND RESULTS: Mouse aortic smooth muscle cells were treated with an actin-stabilizing agent, jasplakinolide, and analyzed by microarrays. Several transcripts were upregulated including both known and previously unknown actin-regulated genes. Dystrophin and synaptopodin 2 were selected for further analysis in models of phenotypic modulation and vascular disease. These genes were highly expressed in differentiated versus synthetic smooth muscle and their expression was promoted by the transcription factors myocardin and myocardin-related transcription factor A. Furthermore, the expression of both synaptopodin 2 and dystrophin was significantly reduced in balloon-injured human arteries. Finally, using a dystrophin mutant mdx mouse and synaptopodin 2 knockdown, we demonstrate that these genes are involved in the regulation of smooth muscle differentiation and function. CONCLUSIONS: This study demonstrates novel genes that are promoted by actin polymerization, that regulate smooth muscle function, and that are deregulated in models of vascular disease. Thus, targeting actin polymerization or the genes controlled in this manner can lead to novel therapeutic options against vascular pathologies that involve phenotypic modulation of smooth muscle cells.


Asunto(s)
Actinas/metabolismo , Distrofina/genética , Proteínas de Microfilamentos/genética , Músculo Liso Vascular/metabolismo , Enfermedades Vasculares/genética , Enfermedades Vasculares/metabolismo , Animales , Arterias/lesiones , Células Cultivadas , Expresión Génica , Humanos , Ratones Endogámicos mdx , Ratones Noqueados , Contracción Muscular , Relajación Muscular , Polimerizacion , Transcripción Genética
9.
Pflugers Arch ; 467(8): 1711-22, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25208915

RESUMEN

Phenotypic modulation (PM) of vascular smooth muscle cells (VSMCs) is central to the process of intimal hyperplasia which constitutes a common pathological lesion in occlusive vascular diseases. Changes in the functional expression of Kv1.5 and Kv1.3 currents upon PM in mice VSMCs have been found to contribute to cell migration and proliferation. Using human VSMCs from vessels in which unwanted remodeling is a relevant clinical complication, we explored the contribution of the Kv1.5 to Kv1.3 switch to PM. Changes in the expression and the functional contribution of Kv1.3 and Kv1.5 channels were studied in contractile and proliferating VSMCs obtained from human donors. Both a Kv1.5 to Kv1.3 switch upon PM and an anti-proliferative effect of Kv1.3 blockers on PDGF-induced proliferation were observed in all vascular beds studied. When investigating the signaling pathways modulated by the blockade of Kv1.3 channels, we found that anti-proliferative effects of Kv1.3 blockers on human coronary artery VSMCs were occluded by selective inhibition of MEK/ERK and PLCγ signaling pathways, but were unaffected upon blockade of PI3K/mTOR pathway. The temporal course of the anti-proliferative effects of Kv1.3 blockers indicates that they have a role in the late signaling events essential for the mitogenic response to growth factors. These findings establish the involvement of Kv1.3 channels in the PM of human VSMCs. Moreover, as current therapies to prevent restenosis rely on mTOR blockers, our results provide the basis for the development of novel, more specific therapies.


Asunto(s)
Proliferación Celular , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Canal de Potasio Kv1.3/antagonistas & inhibidores , Canal de Potasio Kv1.3/genética , Canal de Potasio Kv1.3/metabolismo , Canal de Potasio Kv1.5/genética , Canal de Potasio Kv1.5/metabolismo , Potenciales de la Membrana , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Fenotipo , Inhibidores de Fosfodiesterasa/farmacología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Factores de Tiempo
10.
J Physiol ; 591(24): 6175-91, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24167226

RESUMEN

Hypertension is a clinical syndrome characterized by increased arterial tone. Although the mechanisms are varied, the generally accepted view is that increased CaV1.2 channel function is a common feature of this pathological condition. Here, we investigated the mechanisms underlying vascular dysfunction in a mouse model of genetic hypertension. Contrary to expectation, we found that whole-cell CaV1.2 currents (ICa) were lower in hypertensive (BPH line) than normotensive (BPN line) myocytes. However, local CaV1.2 sparklet activity was higher in BPH cells, suggesting that the relatively low ICa in these cells was produced by a few hyperactive CaV1.2 channels. Furthermore, our data suggest that while the lower expression of the pore-forming α1c subunit of CaV1.2 currents underlies the lower ICa in BPH myocytes, the increased sparklet activity was due to a different composition in the auxiliary subunits of the CaV1.2 complexes. ICa currents in BPN cells were produced by channels composed of α1c/α2δ/ß3 subunits, while in BPH myocytes currents were probably generated by the opening of channels formed by α1c/α2δ/ß2 subunits. In addition, Ca(2+) sparks evoked large conductance, Ca(2+)-activated K(+) (BK) currents of lower magnitude in BPH than in BPN myocytes, because BK channels were less sensitive to Ca(2+). Our data are consistent with a model in which a decrease in the global number of CaV1.2 currents coexist with the existence of a subpopulation of highly active channels that dominate the resting Ca(2+) influx. The decrease in BK channel activity makes the hyperpolarizing brake ineffective and leads BPH myocytes to a more contracted resting state.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Calcio/metabolismo , Regulación hacia Abajo , Hipertensión/metabolismo , Miocitos del Músculo Liso/metabolismo , Potenciales de Acción , Animales , Canales de Calcio Tipo L/genética , Señalización del Calcio , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Ratones , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/fisiología , Subunidades de Proteína/metabolismo
11.
Arterioscler Thromb Vasc Biol ; 32(5): 1299-307, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22383699

RESUMEN

OBJECTIVE: Phenotypic modulation of vascular smooth muscle cells has been associated with a decreased expression of all voltage-dependent potassium channel (Kv)1 channel encoding genes but Kcna3 (which encodes Kv1.3 channels). In fact, upregulation of Kv1.3 currents seems to be important to modulate proliferation of mice femoral vascular smooth muscle cells in culture. This study was designed to explore if these changes in Kv1 expression pattern constituted a landmark of phenotypic modulation across vascular beds and to investigate the mechanisms involved in the proproliferative function of Kv1.3 channels. METHODS AND RESULTS: Changes in Kv1.3 and Kv1.5 channel expression were reproduced in mesenteric and aortic vascular smooth muscle cells, and their correlate with protein expression was electrophysiologicaly confirmed using selective blockers. Heterologous expression of Kv1.3 and Kv1.5 channels in HEK cells has opposite effects on the proliferation rate. The proproliferative effect of Kv1.3 channels was reproduced by "poreless" mutants but disappeared when voltage-dependence of gating was suppressed. CONCLUSIONS: These findings suggest that the signaling cascade linking Kv1.3 functional expression to cell proliferation is activated by the voltage-dependent conformational change of the channels without needing ion conduction. Additionally, the conserved upregulation of Kv1.3 on phenotypic modulation in several vascular beds makes this channel a good target to control unwanted vascular remodeling.


Asunto(s)
Regulación de la Expresión Génica , Canal de Potasio Kv1.3/genética , Músculo Liso Vascular/fisiología , ARN Mensajero/genética , Vasoconstricción/fisiología , Animales , Western Blotting , Proliferación Celular , Células Cultivadas , Canal de Potasio Kv1.3/biosíntesis , Ratones , Músculo Liso Vascular/citología , Fenotipo , Reacción en Cadena de la Polimerasa
12.
iScience ; 26(8): 107473, 2023 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-37575201

RESUMEN

The functions of macrophages are tightly regulated by their metabolic state. However, the role of the mitochondrial electron transport chain (ETC) in macrophage functions remains understudied. Here, we provide evidence that the succinate dehydrogenase (SDH)/complex II (CII) is required for respiration and plays a role in controlling effector responses in macrophages. We find that the absence of the catalytic subunits Sdha and Sdhb in macrophages impairs their ability to effectively stabilize HIF-1α and produce the pro-inflammatory cytokine IL-1ß in response to LPS stimulation. We also arrive at the novel result that both subunits are essential for the LPS-driven production of IL-10, a potent negative feedback regulator of the macrophage inflammatory response. This phenomenon is explained by the fact that the absence of Sdha and Sdhb leads to the inhibition of Stat3 tyrosine phosphorylation, caused partially by the excessive accumulation of mitochondrial reactive oxygen species (mitoROS) in the knockout cells.

13.
J Physiol ; 590(23): 6075-91, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22966162

RESUMEN

The increased vascular tone that defines essential hypertension is associated with depolarization of vascular smooth muscle cells (VSMCs) and involves a change in the expression profile of ion channels promoting arterial contraction. As a major regulator of VSMC resting membrane potential (V(M)), K(+) channel activity is an important determinant of vascular tone and vessel diameter. However, hypertension-associated changes in the expression and/or modulation of K(+) channels are poorly defined, due to their large molecular diversity and their bed-specific pattern of expression. Moreover, the impact of these changes on the integrated vessel function and their contribution to the development of altered vascular tone under physiological conditions need to be confirmed. Hypertensive (BPH) and normotensive (BPN) mice strains obtained by phenotypic selection were used to explore whether changes in the functional expression of VSMC inward rectifier K(+) channels contribute to the more depolarized resting V(M) and the increased vascular reactivity of hypertensive arteries. We determined the expression levels of inward rectifier K(+) channel mRNA in several vascular beds from BPN and BPH animals, and their functional contribution to VSMC excitability and vascular tone in mesenteric arteries. We found a decrease in the expression of Kir2.1, Kir4.1, Kir6.x and SUR2 mRNA in BPH VSMCs, and a decreased functional contribution of both K(IR) and K(ATP) channels in isolated BPH VSMCs. However, only the effect of K(ATP) channel modulators was impaired when exploring vascular tone, suggesting that decreased functional expression of K(ATP) channels may be an important element in the remodelling of VSMCs in essential hypertension.


Asunto(s)
Hipertensión/fisiopatología , Arterias Mesentéricas/fisiología , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/fisiología , Canales de Potasio de Rectificación Interna/fisiología , Transportadoras de Casetes de Unión a ATP/fisiología , Animales , Ratones , Subunidades de Proteína/fisiología , Receptores de Droga/fisiología , Receptores de Sulfonilureas
14.
Arterioscler Thromb Vasc Biol ; 30(6): 1203-11, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20299686

RESUMEN

OBJECTIVE: Vascular smooth muscle cells (VSMCs) contribute significantly to occlusive vascular diseases by virtue of their ability to switch to a noncontractile, migratory, and proliferating phenotype. Although the participation of ion channels in this phenotypic modulation (PM) has been described previously, changes in their expression are poorly defined because of their large molecular diversity. We obtained a global portrait of ion channel expression in contractile versus proliferating mouse femoral artery VSMCs, and explored the functional contribution to the PM of the most relevant changes that we observed. METHODS AND RESULTS: High-throughput real-time polymerase chain reaction of 87 ion channel genes was performed in 2 experimental paradigms: an in vivo model of endoluminal lesion and an in vitro model of cultured VSMCs obtained from explants. mRNA expression changes showed a good correlation between the 2 proliferative models, with only 2 genes, Kv1.3 and Kvbeta2, increasing their expression on proliferation. The functional characterization demonstrates that Kv1.3 currents increased in proliferating VSMC and that their selective blockade inhibits migration and proliferation. CONCLUSIONS: These findings establish the involvement of Kv1.3 channels in the PM of VSMCs, providing a new therapeutical target for the treatment of intimal hyperplasia.


Asunto(s)
Proliferación Celular , Canal de Potasio Kv1.3/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Análisis por Conglomerados , Modelos Animales de Enfermedad , Arteria Femoral/metabolismo , Arteria Femoral/patología , Perfilación de la Expresión Génica , Genotipo , Hiperplasia , Canal de Potasio Kv1.3/antagonistas & inhibidores , Canal de Potasio Kv1.3/genética , Potenciales de la Membrana , Ratones , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Fenotipo , Bloqueadores de los Canales de Potasio/farmacología , ARN Mensajero/metabolismo , Canales de Potasio de la Superfamilia Shaker/genética , Canales de Potasio de la Superfamilia Shaker/metabolismo , Regulación hacia Arriba , Vasoconstricción
15.
Mol Metab ; 53: 101306, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34298200

RESUMEN

OBJECTIVES: Restenosis after vessel angioplasty due to dedifferentiation of the vascular smooth muscle cells (VSMCs) limits the success of surgical treatment of vascular occlusions. Type 2 diabetes (T2DM) has a major impact on restenosis, with patients exhibiting more aggressive forms of vascular disease and poorer outcomes after surgery. Kv1.3 channels are critical players in VSMC proliferation. Kv1.3 blockers inhibit VSMCs MEK/ERK signalling and prevent vessel restenosis. We hypothesize that dysregulation of microRNAs (miR) play critical roles in adverse remodelling, contributing to Kv1.3 blockers efficacy in T2DM VSMCs. METHODS AND RESULTS: We used clinically relevant in vivo models of vascular risk factors (VRF) and vessels and VSMCs from T2DM patients. RESUKTS: Human T2DM vessels showed increased remodelling, and changes persisted in culture, with augmented VSMCs migration and proliferation. Moreover, there were downregulation of PI3K/AKT/mTOR and upregulation of MEK/ERK pathways, with increased miR-126 expression. The inhibitory effects of Kv1.3 blockers on remodelling were significantly enhanced in T2DM VSMCs and in VRF model. Finally, miR-126 overexpression confered "diabetic" phenotype to non-T2DM VSMCs by downregulating PI3K/AKT axis. CONCLUSIONS: miR-126 plays crucial roles in T2DM VSMC metabolic memory through activation of MEK/ERK pathway, enhancing the efficacy of Kv1.3 blockers in the prevention of restenosis in T2DM patients.


Asunto(s)
Reestenosis Coronaria/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Epigénesis Genética/genética , Canal de Potasio Kv1.3/metabolismo , MicroARNs/metabolismo , Músculo Liso Vascular/metabolismo , Anciano , Animales , Reestenosis Coronaria/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Femenino , Humanos , Canal de Potasio Kv1.3/antagonistas & inhibidores , Masculino , Ratones , MicroARNs/genética , Músculo Liso Vascular/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología
16.
Acta Biomater ; 115: 264-274, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32771595

RESUMEN

Coronary artery disease (CAD) is the most common cardiovascular disorder. Vascular surgery strategies for coronary revascularization (either percutaneous or open) show a high rate of failure because of restenosis of the vessel, due to phenotypic switch of vascular smooth muscle cells (VSMCs) leading to proliferation and migration. We have previously reported that the inhibition of Kv1.3 channel function with selective blockers represents an effective strategy for the prevention of restenosis in human vessels used for coronary angioplasty procedures. However, delivery systems for controlled release of these drugs have not been investigated. Here we tested the efficacy of several formulations of elastin like recombinamers (ELRs) hydrogels to deliver the Kv1.3 blocker PAP-1 in various restenosis models. The dose and time course of PAP-1 release from ELRs click hydrogels was able to inhibit human VSMC proliferation in vitro as well as remodeling of human vessels in organ culture and restenosis in in vivo models. We conclude that this combination of active compound and advanced delivery method could improve the outcomes of vascular surgery in patients. STATEMENT OF SIGNIFICANCE: Vascular surgery strategies for coronary revascularization show a high rate of failure, because of occlusion (restenosis) of the vessel, due to vascular smooth muscle cells proliferation and migration. We have previously reported that blockers of Kv1.3 channels represent an effective anti-restenosis therapy, but delivery systems for their controlled release have not being explored. Here we tested the efficacy of several formulations of elastin like recombinamers (ELRs) hydrogels to deliver the Kv1.3 blocker PAP-1 in various restenosis models, both in vivo and in vitro, and also in human vessels. We demonstrated that combination of active compound and advanced delivery method could improve the outcomes of vascular surgery in patients.


Asunto(s)
Elastina , Músculo Liso Vascular , Proliferación Celular , Células Cultivadas , Humanos , Hiperplasia/tratamiento farmacológico , Hiperplasia/patología , Hiperplasia/prevención & control , Músculo Liso Vascular/patología
17.
J Physiol ; 587(3): 625-40, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19074965

RESUMEN

Essential hypertension involves a gradual and sustained increase in total peripheral resistance, reflecting an increased vascular tone. This change associates with a depolarization of vascular myocytes, and relies on a change in the expression profile of voltage-dependent ion channels (mainly Ca(2+) and K(+) channels) that promotes arterial contraction. However, changes in expression and/or modulation of voltage-dependent K(+) channels (Kv channels) are poorly defined, due to their large molecular diversity and their vascular bed-specific expression. Here we endeavor to characterize the molecular and functional expression of Kv channels in vascular smooth muscle cells (VSMCs) and their regulation in essential hypertension, by using VSMCs from resistance (mesenteric) or conduit (aortic) arteries obtained from a hypertensive inbred mice strain, BPH, and the corresponding normotensive strain, BPN. Real-time PCR reveals a differential distribution of Kv channel subunits in the different vascular beds as well as arterial bed-specific changes under hypertension. In mesenteric arteries, the most conspicuous change was the de novo expression of Kv6.3 (Kcng3) mRNA in hypertensive animals. The functional relevance of this change was studied by using patch-clamp techniques. VSMCs from BPH arteries were more depolarized than BPN ones, and showed significantly larger capacitance values. Moreover, Kv current density in BPH VSMCs is decreased mainly due to the diminished contribution of the Kv2 component. The kinetic and pharmacological profile of Kv2 currents suggests that the expression of Kv6.3 could contribute to the natural development of hypertension.


Asunto(s)
Hipertensión/genética , Hipertensión/fisiopatología , Músculo Liso Vascular/fisiopatología , Canales de Potasio con Entrada de Voltaje/metabolismo , Animales , Aorta/metabolismo , Línea Celular , Perfilación de la Expresión Génica , Hipertensión/metabolismo , Indoles/farmacología , Activación del Canal Iónico/efectos de los fármacos , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/metabolismo , Ratones , Músculo Liso Vascular/efectos de los fármacos , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Canales de Potasio Shab/antagonistas & inhibidores , Canales de Potasio Shab/metabolismo , Canales de Potasio de la Superfamilia Shaker/antagonistas & inhibidores , Canales de Potasio de la Superfamilia Shaker/metabolismo , Triterpenos/farmacología
18.
Front Physiol ; 9: 89, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29483881

RESUMEN

Patients with chronic kidney disease (CKD) have a markedly increased incidence of cardiovascular disease (CVD). The high concentration of circulating uremic toxins and alterations in mineral metabolism and hormone levels produce vascular wall remodeling and significant vascular damage. Medial calcification is an early vascular event in CKD patients and is associated to apoptosis or necrosis and trans-differentiation of vascular smooth muscle cells (VSMC) to an osteogenic phenotype. VSMC obtained from bovine or rat aorta and cultured in the presence of increased inorganic phosphate (Pi) have been extensively used to study these processes. In this study we used human aortic VSMC primary cultures to compare the effects of increased Pi to treatment with serum obtained from uremic patients. Uremic serum induced calcification, trans-differentiation and phenotypic remodeling even with normal Pi levels. In spite of similar calcification kinetics, there were fundamental differences in osteochondrogenic marker expression and alkaline phosphatase induction between Pi and uremic serum-treated cells. Moreover, high Pi induced a dramatic decrease in cell viability, while uremic serum preserved it. In summary, our data suggests that primary cultures of human VSMC treated with serum from uremic patients provides a more informative model for the study of vascular calcification secondary to CKD.

19.
Circ Res ; 97(12): 1280-7, 2005 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-16269658

RESUMEN

Vascular smooth muscle cells (VSMCs) perform diverse functions that can be classified into contractile and synthetic (or proliferating). All of these functions can be fulfilled by the same cell because of its capacity of phenotypic modulation in response to environmental changes. The resting membrane potential is a key determinant for both contractile and proliferating functions. Here, we have explored the expression of voltage-dependent K+ (Kv) channels in contractile (freshly dissociated) and proliferating (cultured) VSMCs obtained from human uterine arteries to establish their contribution to the functional properties of the cells and their possible participation in the phenotypic switch. We have studied the expression pattern (both at the mRNA and at the protein level) of Kvalpha subunits in both preparations as well as their functional contribution to the K+ currents of VSMCs. Our results indicate that phenotypic remodeling associates with a change in the expression and distribution of Kv channels. Whereas Kv currents in contractile VSMCs are mainly performed by Kv1 channels, Kv3.4 is the principal contributor to K+ currents in cultured VSMCs. Furthermore, selective blockade of Kv3.4 channels resulted in a reduced proliferation rate, suggesting a link between Kv channels expression and phenotypic remodeling.


Asunto(s)
Proliferación Celular , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Canales de Potasio con Entrada de Voltaje/fisiología , Útero/irrigación sanguínea , Células Cultivadas , Femenino , Humanos , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Fenotipo , Canales de Potasio con Entrada de Voltaje/análisis , Canales de Potasio con Entrada de Voltaje/genética , Subunidades de Proteína , ARN Mensajero/análisis , Canales de Potasio de la Superfamilia Shaker/efectos de los fármacos , Canales de Potasio de la Superfamilia Shaker/fisiología , Canales de Potasio Shal/análisis , Canales de Potasio Shal/genética , Canales de Potasio Shaw/efectos de los fármacos , Canales de Potasio Shaw/genética , Canales de Potasio Shaw/fisiología , Compuestos de Tetraetilamonio/farmacología , Triterpenos/farmacología
20.
Respir Physiol Neurobiol ; 157(1): 65-74, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17442633

RESUMEN

Hypoxic inhibition of K(+) channels has been documented in many native chemoreceptor cells, and is crucial to initiate reflexes directed to improve tissue O(2) supply. In the carotid body (CB) chemoreceptors, there is a general consensus regarding the facts that a decrease in P(O2) leads to membrane depolarization, increase of Ca(2+) entry trough voltage-dependent Ca(2+) channels and Ca(2+)-dependent release of neurotransmitters. Central to this pathway is the modulation by hypoxia of K(+) channels that triggers depolarization. However, the details of this process are still controversial, and even the molecular nature of these oxygen-sensitive K(+) (K(O2)) channels in the CB is hotly debated. Clearly there are inter-species differences, and even in the same preparation more that one K(O2) may be present. Here we recapitulate our present knowledge of the role of voltage dependent K(+) channels as K(O2) in the CB from different species, and their functional contribution to cell excitability in response to acute and chronic exposure to hypoxia.


Asunto(s)
Cuerpo Carotídeo/fisiología , Células Quimiorreceptoras/fisiología , Mecanotransducción Celular/fisiología , Canales de Potasio/fisiología , Animales , Humanos , Hipoxia/metabolismo , Hipoxia/fisiopatología , Filogenia , Canales de Potasio/química , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA