Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Am J Physiol Cell Physiol ; 324(6): C1249-C1262, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37125772

RESUMO

Control of the movement of ions and water across epithelia is essential for homeostasis. Changing the number or activity of ion channels at the plasma membrane is a significant regulator of epithelial transport. In polarized epithelia, the intermediate-conductance calcium-activated potassium channel, KCa3.1 is delivered to the basolateral membrane where it generates and maintains the electrochemical gradients required for epithelial transport. The mechanisms that control the delivery of KCa3.1 to the basolateral membrane are still emerging. Herein, we investigated the role of the highly conserved tethering complex exocyst. In epithelia, exocyst is involved in the tethering of post-Golgi secretory vesicles with the basolateral membrane, which is required before membrane fusion. In our Fisher rat thyroid cell line that stably expresses KCa3.1, siRNA knockdown of either of the exocyst subunits Sec3, Sec6, or Sec8 significantly decreased KCa3.1-specific current. In addition, knockdown of exocyst complex subunits significantly reduced the basolateral membrane protein level of KCa3.1. Finally, co-immunoprecipitation experiments suggest associations between Sec6 and KCa3.1, but not between Sec8 and KCa3.1. Collectively, based on these data and our previous studies, we suggest that components of exocyst complex are crucially important in the tethering of KCa3.1 to the basolateral membrane. After which, Soluble N-ethylmaleimide-sensitive factor (SNF) Attachment Receptors (SNARE) proteins aid in the insertion of KCa3.1-containing vesicles into the basolateral membrane of polarized epithelia.NEW & NOTEWORTHY Our Ussing chamber and immunoblot experiments demonstrate that when subunits of the exocyst complex were transiently knocked down, this significantly reduced the basolateral population and functional expression of KCa3.1. These data suggest, combined with our protein association experiments, that the exocyst complex regulates the tethering of KCa3.1-containing vesicles to the basolateral membrane prior to the SNARE-dependent insertion of channels into the basolateral membrane of epithelial cells.


Assuntos
Células Epiteliais , Fusão de Membrana , Ratos , Animais , Membrana Celular/metabolismo , Epitélio , Células Epiteliais/metabolismo , Proteínas SNARE/genética , Proteínas SNARE/metabolismo
2.
Am J Physiol Renal Physiol ; 319(5): F895-F907, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-33017185

RESUMO

The epithelial Na+ channel (ENaC) located at the apical membrane in many epithelia is the rate-limiting step for Na+ reabsorption. Tight regulation of the plasma membrane population of ENaC is required, as hypertension or hypotension may result if too many or too few ENaCs are present. Endocytosed ENaC travels to the early endosome and is then either trafficked to the lysosome for degradation or recycled back to the plasma membrane. Recently, the retromer recycling complex, located at the early endosome, has been implicated in plasma membrane protein recycling pathways. We hypothesized that the retromer is required for recycling of ENaC. Stabilization of retromer function with the retromer stabilizing chaperone R55 increased ENaC current, whereas knockdown or overexpression of individual retromer and associated proteins altered ENaC current and cell surface population of ENaC. KIBRA was identified as an ENaC-binding protein allowing ENaC to link to sorting nexin 4 to alter ENaC trafficking. Knockdown of the retromer-associated cargo-binding sorting nexin 27 protein did not alter ENaC current, whereas CCDC22, a CCC-complex protein, coimmunoprecipitated with ENaC, and CCDC22 knockdown decreased ENaC current and population at the cell surface. Together, our results confirm that retromer and the CCC complex play a role in recycling of ENaC to the plasma membrane.


Assuntos
Endossomos/metabolismo , Células Epiteliais/metabolismo , Canais Epiteliais de Sódio/metabolismo , Transporte Proteico/fisiologia , Membrana Celular/metabolismo , Movimento Celular/fisiologia , Endocitose/fisiologia , Células Epiteliais/fisiologia , Humanos , Sódio/metabolismo
3.
Int J Mol Sci ; 20(5)2019 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-30857243

RESUMO

Abstract: The epithelial intermediate-conductance calcium/calmodulin-regulated KCa3.1 channel is considered to be a regulator of intestine function by controlling chloride secretion and water/salt balance. Yet, little is known about the functional importance of KCa3.1 in the intestinal epithelium in vivo. Our objective was to determine the impact of epithelial-specific inducible overexpression of a KCa3.1 transgene (KCa3.1+) and of inducible suppression (KCa3.1-) on intestinal homeostasis and function in mice. KCa3.1 overexpression in the duodenal epithelium of doxycycline (DOX)-treated KCa3.1+ mice was 40-fold above the control levels. Overexpression caused an inflated duodenum and doubling of the chyme content. Histology showed conserved architecture of crypts, villi, and smooth muscle. Unaltered proliferating cell nuclear antigen (PCNA) immune reactivity and reduced amounts of terminal deoxynucleotide transferase mediated X-dUTP nick end labeling (TUNEL)-positive apoptotic cells in villi indicated lower epithelial turnover. Myography showed a reduction in the frequency of spontaneous propulsive muscle contractions with no change in amplitude. The amount of stool in the colon was increased and the frequency of colonic contractions was reduced in KCa3.1+ animals. Senicapoc treatment prevented the phenotype. Suppression of KCa3.1 in DOX-treated KCa3.1- mice caused no overt intestinal phenotype. In conclusion, inducible KCa3.1 overexpression alters intestinal functions by increasing the chyme content and reducing spontaneous contractions and epithelial apoptosis. Induction of epithelial KCa3.1 can play a mechanistic role in the process of adaptation of the intestine.


Assuntos
Duodeno/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Mucosa Intestinal/fisiologia , Animais , Digestão , Duodeno/ultraestrutura , Deleção de Genes , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Mucosa Intestinal/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Contração Muscular , Transgenes , Regulação para Cima
4.
Am J Physiol Renal Physiol ; 311(2): F291-304, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27122544

RESUMO

Technical advancements in research techniques in science are made in slow increments. Even so, large advances from insight and hard work of an individual with a single technique can have astonishing ramifications. Here, we examine the impact of Dr. Maurice B. Burg and the isolated perfused renal tubule technique and celebrate the 50th anniversary of the publication by Dr. Burg and his colleagues of their landmark paper in the American Journal of Physiology in 1966. In this study, we have taken a scientific visualization approach to study the scientific contributions of Dr. Burg and the isolated perfused tubule preparation as determining research impact by the number of research students, postdoctoral fellows, visiting scientists, and national and international collaborators. Additionally, we have examined the research collaborations (first and second generation scientists), established the migrational visualization of the first generation scientists who worked directly with Dr. Burg, quantified the metrics indices, identified and quantified the network of coauthorship of the first generation scientists with their second generation links, and determined the citations analyses of outputs of Dr. Burg and/or his first generation collaborators as coauthors. We also review the major advances in kidney physiology that have been made with the isolated perfused tubule technique. Finally, we are all waiting for the discoveries that the isolated perfused preparation technique will bring during the next 50 years.


Assuntos
Túbulos Renais/fisiologia , Rim/fisiologia , Fisiologia/história , Animais , História do Século XX , Técnicas In Vitro , Rim/metabolismo , Túbulos Renais/metabolismo , Modelos Biológicos , Néfrons/fisiologia , Perfusão , Estados Unidos
6.
Am J Physiol Cell Physiol ; 306(8): C715-20, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24573083

RESUMO

An old proverb states "necessity is the mother of invention." This certainly holds true in science as one pursues research questions. Experimental techniques have evolved as scientists have asked more specific research questions. Indeed, techniques such as the Ussing chamber, the perfused renal tubule preparation, patch-clamp, polymerase chain reaction, and site-directed mutagenesis have been developed over the past 60 years. However, sometimes, simple techniques may be useful and still very informative, and this certainly applies to intestinal physiology. Indeed, Gerald Wiseman and Thomas Hastings Wilson described the intestinal everted sac preparation some 60 years ago. Since then, this technique has been used for numerous research purposes including determining ion, amino acid, water and solute transport across the intestinal epithelium; and drug metabolism, absorption, and interactions in pharmaceutical/pharmacological research and even in education. This article provides a historical review of the development of the in vitro intestinal preparation that led to the everted sac preparation and its use in science.


Assuntos
Pesquisa Biomédica/história , Absorção Intestinal/fisiologia , Mucosa Intestinal/fisiologia , Animais , Pesquisa Biomédica/métodos , História do Século XIX , História do Século XX , História do Século XXI
8.
Adv Physiol Educ ; 37(4): 415-26, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24292921

RESUMO

The Na(+)-glucose cotransporter is a key transport protein that is responsible for absorbing Na(+) and glucose from the luminal contents of the small intestine and reabsorption by the proximal straight tubule of the nephron. Robert K. Crane originally described the cellular model of absorption of Na(+) and glucose by a "cotransport process" in 1960. Over the past 50+ yr, numerous groups have tested and verified Crane's hypothesis. Eventually, Wright and colleagues cloned the Na(+)-glucose cotransporter (SGLT1; the product of the SLC5A1 gene) in 1987. This article provides a "hands-on" laboratory exercise using the everted mouse jejunal preparation (everted sac) that allows students to investigate various components of the Na(+)-glucose cotransport absorptive cell model (e.g., Na(+) dependence of SGLT1, inhibition of SGLT1, and inhibition of Na(+)-K(+)-ATPase). Additionally, the laboratory exercise includes a case-based study of glucose-galactose malabsorption in which the students conduct an internet search and participate in a small-group discussion during the laboratory period to better understand the basic principles and functions of the Na(+)-glucose absorptive process of the small intestine. This laboratory exercise was introduced into the second-year undergraduate physiology curriculum in 2008, and >850 physiology students have participated in this laboratory exercise. The students have produced very robust and reproducible data that clearly illustrate the theory of the cellular model for Na(+)-glucose absorption by the jejunum.


Assuntos
Glucose/metabolismo , Intestino Delgado/metabolismo , Animais , Transporte Biológico , Camundongos , Sódio/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo
9.
Am J Physiol Renal Physiol ; 302(9): F1069-81, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22338089

RESUMO

The major function of epithelial tissues is to maintain proper ion, solute, and water homeostasis. The tubule of the renal nephron has an amazingly simple structure, lined by epithelial cells, yet the segments (i.e., proximal tubule vs. collecting duct) of the nephron have unique transport functions. The functional differences are because epithelial cells are polarized and thus possess different patterns (distributions) of membrane transport proteins in the apical and basolateral membranes of the cell. K(+) channels play critical roles in normal physiology. Over 90 different genes for K(+) channels have been identified in the human genome. Epithelial K(+) channels can be located within either or both the apical and basolateral membranes of the cell. One of the primary functions of basolateral K(+) channels is to recycle K(+) across the basolateral membrane for proper function of the Na(+)-K(+)-ATPase, among other functions. Mutations of these channels can cause significant disease. The focus of this review is to provide an overview of the basolateral K(+) channels of the nephron, providing potential physiological functions and pathophysiology of these channels, where appropriate. We have taken a "K(+) channel gene family" approach in presenting the representative basolateral K(+) channels of the nephron. The basolateral K(+) channels of the renal epithelia are represented by members of the KCNK, KCNJ, KCNQ, KCNE, and SLO gene families.


Assuntos
Membrana Celular/fisiologia , Células Epiteliais/fisiologia , Rim/fisiologia , Canais de Potássio/genética , Canais de Potássio/fisiologia , Homeostase/fisiologia , Humanos , Rim/citologia , Potássio/metabolismo , ATPase Trocadora de Sódio-Potássio/fisiologia
10.
Front Physiol ; 13: 905834, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35832483

RESUMO

Targeting proteins to a specific membrane is crucial for proper epithelial cell function. KCa3.1, a calcium-activated, intermediate-conductance potassium channel, is targeted to the basolateral membrane (BLM) in epithelial cells. Surprisingly, the mechanism of KCa3.1 membrane targeting is poorly understood. We previously reported that targeting of KCa3.1 to the BLM of epithelial cells is Myosin-Vc-, Rab1-and Rab8-dependent. Here, we examine the role of the SNARE proteins VAMP3, SNAP-23 and syntaxin 4 (STX-4) in the targeting of KCa3.1 to the BLM of Fischer rat thyroid (FRT) epithelial cells. We carried out immunoblot, siRNA and Ussing chamber experiments on FRT cells, stably expressing KCa3.1-BLAP/Bir-A-KDEL, grown as high-resistance monolayers. siRNA-mediated knockdown of VAMP3 reduced BLM expression of KCa3.1 by 57 ± 5% (p ≤ 0.05, n = 5). Measurements of BLM-localized KCa3.1 currents, in Ussing chambers, demonstrated knockdown of VAMP3 reduced KCa3.1 current by 70 ± 4% (p ≤ 0.05, n = 5). Similarly, siRNA knockdown of SNAP-23 reduced the expression of KCa3.1 at the BLM by 56 ± 7% (p ≤ 0.01, n = 6) and reduced KCa3.1 current by 80 ± 11% (p ≤ 0.05, n = 6). Also, knockdown of STX-4 lowered the BLM expression of KCa3.1 by 54 ± 6% (p ≤ 0.05, n = 5) and reduced KCa3.1 current by 78 ± 11% (p ≤ 0.05, n = 5). Finally, co-immunoprecipitation experiments demonstrated associations between KCa3.1, VAMP3, SNAP-23 and STX-4. These data indicate that VAMP3, SNAP-23 and STX-4 are critical for the targeting KCa3.1 to BLM of polarized epithelial cells.

12.
Am J Physiol Cell Physiol ; 300(4): C792-802, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21123738

RESUMO

The intermediate-conductance calcium-activated potassium channel (IK1) promotes cell proliferation of numerous cell types including endothelial cells, T lymphocytes, and several cancer cell lines. The mechanism underlying IK1-mediated cell proliferation was examined in human embryonic kidney 293 (HEK293) cells expressing recombinant human IK1 (hIK1) channels. Inhibition of hIK1 with TRAM-34 reduced cell proliferation, while expression of hIK1 in HEK293 cells increased proliferation. When HEK293 cells were transfected with a mutant (GYG/AAA) hIK1 channel, which neither conducts K(+) ions nor promotes Ca(2+) entry, proliferation was increased relative to mock-transfected cells. Furthermore, when HEK293 cells were transfected with a trafficking mutant (L18A/L25A) hIK1 channel, proliferation was also increased relative to control cells. The lack of functional activity of hIK1 mutants at the cell membrane was confirmed by a combination of whole cell patch-clamp electrophysiology and fura-2 imaging to assess store-operated Ca(2+) entry and cell surface immunoprecipitation assays. Moreover, in cells expressing hIK1, inhibition of ERK1/2 and JNK kinases, but not of p38 MAP kinase, reduced cell proliferation. We conclude that functional K(+) efflux at the plasma membrane and the consequent hyperpolarization and enhanced Ca(2+) entry are not necessary for hIK1-induced HEK293 cell proliferation. Rather, our data suggest that hIK1-induced proliferation occurs by a direct interaction with ERK1/2 and JNK signaling pathways.


Assuntos
Proliferação de Células , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Potássio/metabolismo , Cálcio/metabolismo , Membrana Celular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HEK293 , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5 , Técnicas de Patch-Clamp , Pirazóis/metabolismo , Transdução de Sinais/fisiologia , Canais de Sódio/genética , Canais de Sódio/metabolismo
13.
PLoS One ; 15(3): e0222619, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32150577

RESUMO

Ion channels have recently attracted attention as potential mediators of skin disease. Here, we explored the consequences of genetically encoded induction of the cell volume-regulating Ca2+-activated KCa3.1 channel (Kcnn4) for murine epidermal homeostasis. Doxycycline-treated mice harboring the KCa3.1+-transgene under the control of the reverse tetracycline-sensitive transactivator (rtTA) showed 800-fold channel overexpression above basal levels in the skin and solid KCa3.1-currents in keratinocytes. This overexpression resulted in epidermal spongiosis, progressive epidermal hyperplasia and hyperkeratosis, itch and ulcers. The condition was accompanied by production of the pro-proliferative and pro-inflammatory cytokines, IL-ß1 (60-fold), IL-6 (33-fold), and TNFα (26-fold) in the skin. Treatment of mice with the KCa3.1-selective blocker, Senicapoc, significantly suppressed spongiosis and hyperplasia, as well as induction of IL-ß1 (-88%) and IL-6 (-90%). In conclusion, KCa3.1-induction in the epidermis caused expression of pro-proliferative cytokines leading to spongiosis, hyperplasia and hyperkeratosis. This skin condition resembles pathological features of eczematous dermatitis and identifies KCa3.1 as a regulator of epidermal homeostasis and spongiosis, and as a potential therapeutic target.


Assuntos
Eczema/genética , Epiderme/patologia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Ceratose/genética , Pele/metabolismo , Transgenes , Acetamidas/farmacologia , Animais , Citocinas/metabolismo , Doxiciclina/farmacologia , Eczema/tratamento farmacológico , Feminino , Homeostase/genética , Hiperplasia/tratamento farmacológico , Hiperplasia/genética , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Queratinócitos/metabolismo , Ceratose/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transativadores/metabolismo , Compostos de Tritil/farmacologia
14.
Am J Physiol Cell Physiol ; 297(3): C688-98, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19535515

RESUMO

Previously, we demonstrated that genistein stimulated Cl(-) secretion in the mouse jejunum (Baker MJ and Hamilton KL, Am J Physiol Cell Physiol 287: C1636-C1645, 2004); however, the mode of action of genistein still remains unclear. Here, we examined the activation of Cl(-) secretion by the modulation of phosphodiesterases (PDEs) by genistein (75 microM) in the mouse jejunum with the Ussing short-circuit current (I(sc)) technique. Drugs tested included theophylline (10 mM), a nonspecific PDE inhibitor; 8-methoxymethyl-3-isobutyl-1-methylxanthine (8-MM-IBMX; 100 microM), erythro-9-(2-hydroxyl-3-nonyl)-adenine (EHNA; 40 microM), milrinone (100 microM), and rolipram (40 and 100 microM), which are specific inhibitors of PDE1-PDE4, respectively. Theophylline stimulated a bumetanide-sensitive I(sc), indicative of Cl(-) secretion, and abolished genistein's stimulatory action on I(sc). Neither 8-MM-IBMX nor EHNA altered the basal I(sc) nor did these PDE inhibitors affect the stimulatory action of genistein on the I(sc) of the mouse jejunum. Rolipram had no effect on basal I(sc), but it reduced the genistein-stimulated I(sc) compared with time-matched control tissues. Milrinone stimulated a concentration-dependent increase in I(sc). Bumetanide (10 microM) inhibited 60 +/- 4% of milrinone-induced I(sc). Pretreating tissues with milrinone prevented genistein from stimulating I(sc), and pretreatment with genistein reduced the effect of milrinone on I(sc). H89 (50 microM), a PKA inhibitor, reduced the milrinone-stimulated I(sc). Likewise, H89 reduced the genistein-stimulated I(sc). Here, we demonstrate, for the first time, that genistein activates Cl(-) secretion of the mouse jejunum via inhibition of a PDE3-dependent pathway.


Assuntos
Antiportadores de Cloreto-Bicarbonato/metabolismo , Genisteína/farmacologia , Jejuno/enzimologia , Diester Fosfórico Hidrolases/metabolismo , Animais , Bumetanida/farmacologia , Eletrofisiologia , Masculino , Camundongos , Milrinona/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Teofilina/farmacologia
17.
PLoS One ; 13(5): e0196717, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29768434

RESUMO

We previously demonstrated endocytosis of KCa2.3 is caveolin-1-, dynamin II- and Rab5-dependent. KCa2.3 then enters Rab35/EPI64C- and RME-1-containing recycling endosomes and is returned to the plasma membrane (PM). Herein, we report on the mechanism by which KCa2.3 is inserted into the PM during recycling and following exit from the Golgi. We demonstrate KCa2.3 colocalizes with SNAP-23 and Syntaxin-4 in the PM of HEK and endothelial cells by confocal immunofluorescence microscopy. We further show KCa2.3 can be co-immunoprecipitated with SNAP-23 and Syntaxin-4. Overexpression of either Syntaxin-4 or SNAP-23 increased PM expression of KCa2.3, whereas shRNA-mediated knockdown of these SNARE proteins significantly decreased PM KCa2.3 expression, as assessed by cell surface biotinylation. Whole-cell patch clamp studies confirmed knockdown of SNAP-23 significantly decreased the apamin sensitive, KCa2.3 current. Using standard biotinylation/stripping methods, we demonstrate shRNA mediated knockdown of SNAP-23 inhibits recycling of KCa2.3 following endocytosis, whereas scrambled shRNA had no effect. Finally, using biotin ligase acceptor peptide (BLAP)-tagged KCa2.3, coupled with ER-resident biotin ligase (BirA), channels could be biotinylated in the ER after which we evaluated their rate of insertion into the PM following Golgi exit. We demonstrate knockdown of SNAP-23 significantly slows the rate of Golgi to PM delivery of KCa2.3. The inhibition of both recycling and PM delivery of newly synthesized KCa2.3 channels likely accounts for the decreased PM expression observed following knockdown of these SNARE proteins. In total, our results suggest insertion of KCa2.3 into the PM depends upon the SNARE proteins, Syntaxin-4 and SNAP-23.


Assuntos
Membrana Celular/metabolismo , Proteínas Qa-SNARE/metabolismo , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Complexo de Golgi/metabolismo , Células HEK293 , Humanos , Microscopia Confocal , RNA Interferente Pequeno/metabolismo
18.
PLoS One ; 13(1): e0190307, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29293584

RESUMO

BACKGROUND: TRPV4 channels are calcium-permeable cation channels that are activated by several physicochemical stimuli. Accordingly, TRPV4 channels have been implicated in the regulation of osmosensing, mechanotransduction, thermosensation, and epithelial/endothelial barrier functions. Whether TRPV4 is also mechanistically implicated in melanoma cell proliferation is not clear. Here, we hypothesized that TRPV4 is expressed in human melanoma and that pharmacological activation interferes with cell proliferation. METHODOLOGY/PRINCIPAL FINDINGS: TRPV4 functions were studied in melanoma cell lines (A375, SK-MEL-28, MKTBR), immortalized non-cancer keratinocytes (HaCaT), and murine 3T3 fibroblasts by patch-clamp, qRT-PCR, intracellular calcium measurements, cell proliferation, and flow cytometric assays of apoptosis and cell cycle. The selective TRPV4-activator, GSK1016790A, elicited non-selective cation currents with TRPV4-typical current-voltage-relationship in all cell lines. GSK1016790A-induced currents were blocked by the TRPV4-blocker, HC067047. TRPV4 mRNA expression was demonstrated by qRT-PCR. In A375 cells, TRPV4 activation was frequently paralleled by co-activation of calcium/calmodulin-regulated KCa3.1 channels. Light microscopy showed that TRPV4-activation produced rapid cellular disarrangement, nuclear densification, and detachment of a large fraction of all melanoma cell lines and HaCaT cells. TRPV4-activation induced apoptosis and drastically inhibited A375 and HaCaT proliferation that could be partially prevented by HC067047. CONCLUSIONS/SIGNIFICANCE: Our study showed that TRPV4 channels were functionally expressed in human melanoma cell lines and in human keratinocytes. Pharmacological TRPV4 activation in human melanoma cells and keratinocytes caused severe cellular disarrangement, necrosis and apoptosis. Pharmacological targeting of TRPV4 could be an alternative or adjuvant therapeutic strategy to treat melanoma progression and other proliferative skin disorders.


Assuntos
Apoptose/efeitos dos fármacos , Queratinócitos/patologia , Melanoma/patologia , Canais de Cátion TRPV/agonistas , Células 3T3 , Animais , Cálcio/metabolismo , Ciclo Celular , Linhagem Celular , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Queratinócitos/metabolismo , Leucina/análogos & derivados , Leucina/farmacologia , Melanoma/metabolismo , Camundongos , Técnicas de Patch-Clamp , Sulfonamidas/farmacologia
19.
Front Physiol ; 8: 489, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28769813

RESUMO

In epithelia, the intermediate conductance, Ca2+-activated K+ channel (KCa3.1) is targeted to the basolateral membrane (BLM) where this channel plays numerous roles in absorption and secretion. A growing body of research suggests that the membrane resident population of KCa3.1 may be critical in clinical manifestation of diseases. In this study, we investigated the key molecular components that regulate the degradation of KCa3.1 using a Fisher rat thyroid cell line stably expressing KCa3.1. Using immunoblot, Ussing chamber, and pharmacological approaches, we demonstrated that KCa3.1 is targeted exclusively to the BLM, provided a complete time course of degradation of KCa3.1 and degradation time courses of the channel in the presence of pharmacological inhibitors of ubiquitylation and deubiquitylation to advance our understanding of the retrograde trafficking of KCa3.1. We provide a complete degradation profile of KCa3.1 and that the degradation is via an ubiquitin-dependent pathway. Inhibition of E1 ubiquitin activating enzyme by UBEI-41 crippled the ability of the cells to internalize the channel, shown by the increased BLM surface expression resulting in an increased function of the channel as measured by a DCEBIO sensitive K+ current. Additionally, the involvement of deubiquitylases and degradation by the lysosome were also confirmed by treating the cells with PR-619 or leupeptin/pepstatin, respectively; which significantly decreased the degradation rate of membrane KCa3.1. Additionally, we provided the first evidence that KCa3.1 channels were not deubiquitylated at the BLM. These data further define the retrograde trafficking of KCa3.1, and may provide an avenue for therapeutic approach for treatment of disease.

20.
Front Physiol ; 7: 639, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28101059

RESUMO

Understanding the targeting of KCa3.1 to the basolateral membrane (BLM) of polarized epithelial cells is still emerging. Here, we examined the role of the cytoskeleton (microtubules and microfilaments) and Myosin-Vc (Myo-Vc) in the targeting of KCa3.1 in Fischer rat thyroid epithelial cells. We used a pharmacological approach with immunoblot (for the BLM expression of KCa3.1), Ussing chamber (functional BLM expression of KCa3.1) and siRNA experiments. The actin cytoskeleton inhibitors cytochalasin D (10 µM, 5 h) and latrunculin A (10 µM, 5 h) reduced the targeting of KCa3.1 to the BLM by 88 ± 4 and 70 ± 5%, respectively. Colchicine (10 µM, 5 h) a microtubule inhibitor reduced targeting of KCa3.1 to the BLM by 63 ± 7% and decreased 1-EBIO-stimulated KCa3.1 K+ current by 46 ± 18%, compared with control cells. ML9 (10 µM, 5 h), an inhibitor of myosin light chain kinase, decreased targeting of the channel by 83 ± 2% and reduced K+ current by 54 ± 8% compared to control cells. Inhibiting Myo-V with 2,3-butanedione monoxime (10 mM, 5 h) reduced targeting of the channel to the BLM by 58 ± 5% and decreased the stimulated current of KCa3.1 by 48 ± 12% compared with control cells. Finally, using siRNA for Myo-Vc, we demonstrated that knockdown of Myo-Vc reduced the BLM expression of KCa3.1 by 44 ± 7% and KCa3.1 K+ current by 1.04 ± 0.14 µA compared with control cells. These data suggest that the microtubule and microfilament cytoskeleton and Myo-Vc are critical for the targeting of KCa3.1.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA