Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pflugers Arch ; 467(7): 1495-1508, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25163766

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) has one of the worst survival rates of all cancers. ANO1 (TMEM16A) is a recently identified Ca(2+)-activated Cl(-) channel (CaCC) that is upregulated in several tumors. Although ANO1 was subject to extensive studies in the recent years, its pathophysiological function has only been poorly understood. The aim of the present study is to establish the significance of ANO1 in PDAC behavior and demarcate its roles in PDAC from those of the volume-regulated anion channel (VRAC). We performed qPCR and Western blot measurements on different PDAC cell lines (Panc-1, Mia PaCa 2, Capan-1, AsPC-1, BxPC-3) and compared the results to those obtained in a human pancreatic ductal epithelium (HPDE) cell line. All cancer cell lines showed an upregulation of ANO1 on mRNA and protein levels. Whole-cell patch-clamp recordings identified large Ca(2+) and voltage-dependent Cl(-) currents in PDAC cells. Using siRNA knockdown of ANO1 and three ANO1 inhibitors (T16Ainh-A01, CaCCinh-A01, and NS3728), we found that ANO1 is the main constituent of CaCC current in PDAC cells. We further characterized these three inhibitors and found that they had unspecific effects on the free intracellular calcium concentration. Functional studies on PDAC behavior showed that surprisingly inhibition of ANO1 did not influence cellular proliferation. On the other hand, we found ANO1 channel to be pivotal in PDAC cell migration as assessed in wound healing experiments.


Assuntos
Adenocarcinoma/metabolismo , Canais de Cloreto/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/metabolismo , Anoctamina-1 , Cálcio/metabolismo , Estudos de Casos e Controles , Linhagem Celular Tumoral , Movimento Celular , Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/genética , Cloretos/metabolismo , Humanos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Regulação para Cima
2.
Pflugers Arch ; 466(10): 1899-910, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24419539

RESUMO

Anoctamin 6 (ANO6), also known as TMEM16F, has been shown to be a calcium-activated anion channel with delayed calcium activation. The cellular function of ANO6 is under debate, and different groups have come to different conclusions about ANO6's physiological role. Although it is now quite well established that ANO6 is distinct from the volume-regulated anion channel, it is still unclear whether ANO6 or other anoctamins can be activated by cell swelling. In this study, we suggest that ANO1, ANO6, and ANO10 do not contribute to the volume-activated current in ANO-overexpressing HEK293 cells. Furthermore, knock-down of ANO6 in Ehrlich ascites tumor cells (EATC) and Ehrlich-Lettre ascites (ELA) did not decrease but instead significantly increased swelling-activated membrane currents. Knock-down of ANO6 in EATC did not reduce regulatory volume decrease (RVD) in the absence of extracellular calcium, whereas it significantly reduced RVD in the presence of calcium. Interestingly, we found that knock-down of ANO6 in ELA cells resulted in a decrease in cisplatin-induced caspase-3 activity, confirming earlier findings that ANO6 is involved in apoptosis. Finally, knock-down of ANO1 and ANO6 did not affect the volume-sensitive release of taurine in ELA cells. Thus, our data provide evidence that ANO6 cannot be activated directly by cell swelling unless Ca(2+) is present. We also conclude that ANO6 carries a current during RVD, provided extracellular calcium is present. Thus, swelling activation of ANO6 requires the presence of free calcium.


Assuntos
Apoptose , Cálcio/metabolismo , Tamanho Celular , Proteínas de Transferência de Fosfolipídeos/metabolismo , Animais , Anoctamina-1 , Anoctaminas , Caspase 3/metabolismo , Linhagem Celular Tumoral , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Células HEK293 , Humanos , Camundongos , Proteínas de Transferência de Fosfolipídeos/genética , Taurina/metabolismo
3.
Pflugers Arch ; 465(12): 1753-62, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23832500

RESUMO

Members of the TMEM16 family have recently been described as Ca(2+)-activated Cl(-) channels. They have been implicated in cancer and appear to be associated with poor patient prognosis. Here, we investigate the role of TMEM16 channels in cell migration, which is largely unknown. We focused on TMEM16A and TMEM16F channels that have the highest expression of TMEM16 channels in Ehrlich Lettre ascites (ELA) cells. Due to the lack of specific pharmacological modulators, we employed a miRNA approach and stably knocked down the expression of TMEM16A and TMEM16F channels, respectively. Migration analysis shows that TMEM16A KD clones are affected in their directional migration, whereas TMEM16F KD clones show a 40 % reduced rate of cell migration. Moreover, TMEM16A KD clones have a smaller projected cell area, and they are rounder than TMEM16F KD clones. The morphological changes are linearly correlated with the directionality of cells. TMEM16A and TMEM16F, thus, have an important function in cell migration-TMEM16A in directional migration, TMEM16F in determination of the speed of migration. We conclude that TMEM16A and TMEM16F channels have a distinct impact on the steering and motor mechanisms of migrating ELA cells.


Assuntos
Movimento Celular/fisiologia , Canais de Cloreto/fisiologia , Proteínas de Transferência de Fosfolipídeos/fisiologia , Animais , Anoctamina-1 , Anoctaminas , Carcinoma de Ehrlich , Técnicas de Silenciamento de Genes , Camundongos
4.
Am J Physiol Cell Physiol ; 303(6): C625-34, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22744003

RESUMO

Ca(+) signaling plays a crucial role in control of cell cycle progression, but the understanding of the dynamics of Ca(2+) influx and release of Ca(2+) from intracellular stores during the cell cycle is far from complete. The aim of the present study was to investigate the role of the free extracellular Ca(2+) concentration ([Ca(2+)](o)) in cell proliferation, the pattern of changes in the free intracellular Ca(2+) concentration ([Ca(2+)](i)) during cell cycle progression, and the role of the transient receptor potential (TRP)C1 in these changes as well as in cell cycle progression and cell volume regulation. In Ehrlich Lettré Ascites (ELA) cells, [Ca(2+)](i) decreased significantly, and the thapsigargin-releasable Ca(2+) pool in the intracellular stores increased in G(1) as compared with G(0). Store-depletion-operated Ca(2+) entry (SOCE) and TRPC1 protein expression level were both higher in G(1) than in G(0) and S phase, in parallel with a more effective volume regulation after swelling [regulatory volume decrease (RVD)] in G(1) as compared with S phase. Furthermore, reduction of [Ca(2+)](o), as well as two unspecific SOCE inhibitors, 2-APB (2-aminoethyldiphenyl borinate) and SKF96365 (1-(ß-[3-(4-methoxy-phenyl)propoxyl-4-methoxyphenethyl)1H-imidazole-hydrochloride), inhibited ELA cell proliferation. Finally, Madin-Darby canine kidney cells in which TRPC1 was stably silenced [TRPC1 knockdown (TRPC1-KD) MDCK] exhibited reduced SOCE, slower RVD, and reduced cell proliferation compared with mock controls. In conclusion, in ELA cells, SOCE and TRPC1 both seem to be upregulated in G(1) as compared with S phase, concomitant with an increased rate of RVD. Furthermore, TRPC1-KD MDCK cells exhibit decreased SOCE, decreased RVD, and decreased proliferation, suggesting that, at least in certain cell types, TRPC1 is regulated during cell cycle progression and is involved in SOCE, RVD, and cell proliferation.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Ciclo Celular/fisiologia , Tamanho Celular , Canais de Cátion TRPC/fisiologia , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Transformada , Tamanho Celular/efeitos dos fármacos , Cães , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Fase G1/efeitos dos fármacos , Fase G1/fisiologia , Técnicas de Silenciamento de Genes , Humanos , Células Madin Darby de Rim Canino , Fase S/efeitos dos fármacos , Fase S/fisiologia , Canais de Cátion TRPC/biossíntese , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
5.
Acta Physiol (Oxf) ; 202(3): 465-85, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20874806

RESUMO

Cell volume homeostasis and its fine-tuning to the specific physiological context at any given moment are processes fundamental to normal cell function. The understanding of cell volume regulation owes much to August Krogh, yet has advanced greatly over the last decades. In this review, we outline the historical context of studies of cell volume regulation, focusing on the lineage started by Krogh, Bodil Schmidt-Nielsen, Hans-Henrik Ussing, and their students. The early work was focused on understanding the functional behaviour, kinetics and thermodynamics of the volume-regulatory ion transport mechanisms. Later work addressed the mechanisms through which cellular signalling pathways regulate the volume regulatory effectors or flux pathways. These studies were facilitated by the molecular identification of most of the relevant channels and transporters, and more recently also by the increased understanding of their structures. Finally, much current research in the field focuses on the most up- and downstream components of these paths: how cells sense changes in cell volume, and how cell volume changes in turn regulate cell function under physiological and pathophysiological conditions.


Assuntos
Fenômenos Fisiológicos Celulares/fisiologia , Tamanho Celular , Homeostase , Transdução de Sinais/fisiologia , Equilíbrio Hidroeletrolítico/fisiologia , Morte Celular/fisiologia , Movimento Celular , Canais de Cloreto/metabolismo , Cloretos/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Potássio/metabolismo , Canais de Potássio/metabolismo , Prótons , Sódio/metabolismo
6.
Am J Physiol Cell Physiol ; 298(1): C14-25, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19846756

RESUMO

Changes in cell volume and ion gradients across the plasma membrane play a pivotal role in the initiation of apoptosis. Here we explore the kinetics of apoptotic volume decrease (AVD) and ion content dynamics in wild-type (WT) and multidrug-resistant (MDR) Ehrlich ascites tumor cells (EATC). In WT EATC, induction of apoptosis with cisplatin (5 muM) leads to three distinctive AVD stages: an early AVD(1) (4-12 h), associated with a 30% cell water loss; a transition stage AVD(T) ( approximately 12 to 32 h), where cell volume is partly recovered; and a secondary AVD(2) (past 32 h), where cell volume was further reduced. AVD(1) and AVD(2) were coupled to net loss of Cl(-), K(+), Na(+), and amino acids (ninhydrin-positive substances), whereas during AVD(T), Na(+) and Cl(-) were accumulated. MDR EATC was resistant to cisplatin, showing increased viability and less caspase 3 activation. Compared with WT EATC, MDR EATC underwent a less pronounced AVD(1,) an augmented AVD(T), and a delay in induction of AVD(2). Changes in AVD were associated with inhibition of Cl(-) loss during AVD(1), augmented NaCl uptake during AVD(T), and a delay of Cl(-) loss during AVD(2). Application of the anion channel inhibitor NS3728 inhibited AVD and completely abolished the differences in AVD, ionic movements, and caspase 3 activation between WT and MDR EATC. Finally, the maximal capacity of volume-regulated anion channel was found to be strongly repressed in MDR EATC. Together, these data suggest that impairment of AVD, primarily via modulation of NaCl movements, contribute to protection against apoptosis in MDR EATC.


Assuntos
Apoptose/fisiologia , Canais de Cloreto/fisiologia , Resistência a Múltiplos Medicamentos/fisiologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Proteínas de Transporte/genética , Ciclo Celular , Tamanho Celular , Cisplatino/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Proteínas da Membrana Plasmática de Transporte de GABA , Homeostase/fisiologia , Humanos , Neoplasias/tratamento farmacológico
7.
Acta Physiol (Oxf) ; 194(4): 255-82, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18945273

RESUMO

Cell volume perturbation initiates a wide array of intracellular signalling cascades, leading to protective and adaptive events and, in most cases, activation of volume-regulatory osmolyte transport, water loss, and hence restoration of cell volume and cellular function. Cell volume is challenged not only under physiological conditions, e.g. following accumulation of nutrients, during epithelial absorption/secretion processes, following hormonal/autocrine stimulation, and during induction of apoptosis, but also under pathophysiological conditions, e.g. hypoxia, ischaemia and hyponatremia/hypernatremia. On the other hand, it has recently become clear that an increase or reduction in cell volume can also serve as a specific signal in the regulation of physiological processes such as transepithelial transport, cell migration, proliferation and death. Although the mechanisms by which cell volume perturbations are sensed are still far from clear, significant progress has been made with respect to the nature of the sensors, transducers and effectors that convert a change in cell volume into a physiological response. In the present review, we summarize recent major developments in the field, and emphasize the relationship between cell volume regulation and organism physiology/pathophysiology.


Assuntos
Proteínas de Transporte/fisiologia , Tamanho Celular , Citoesqueleto/fisiologia , Proteínas Quinases/fisiologia , Taurina/fisiologia , Actinas/fisiologia , Animais , Apoptose/fisiologia , Transporte Biológico/fisiologia , Movimento Celular/fisiologia , Substâncias de Crescimento/fisiologia , Humanos , Integrinas/fisiologia , Canais Iônicos/fisiologia , Mamíferos
8.
Am J Physiol Cell Physiol ; 294(4): C1046-55, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18272822

RESUMO

Signaling in cell proliferation, cell migration, and apoptosis is highly affected by osmotic stress and changes in cell volume, although the mechanisms underlying the significance of cell volume as a signal in cell growth and death are poorly understood. In this study, we used NIH-3T3 fibroblasts in a serum- and nutrient-free inorganic medium (300 mosM) to analyze the effects of osmotic stress on MAPK activity and PDGF receptor (PDGFR)-beta-mediated signal transduction. We found that hypoosmolarity (cell swelling at 211 mosM) induced the phosphorylation and nuclear translocation of ERK1/2, most likely via a pathway independent of PDGFR-beta and MEK1/2. Conversely, hyperosmolarity (cell shrinkage at 582 mosM) moved nuclear and phosphorylated ERK1/2 to the cytoplasm and induced the phosphorylation and nuclear translocation of p38 and phosphorylation of JNK1/2. In a series of parallel experiments, hypoosmolarity did not affect PDGF-BB-induced activation of PDGFR-beta, whereas hyperosmolarity strongly inhibited ligand-dependent PDGFR-beta activation as well as downstream mitogenic signal components of the receptor, including Akt and the MEK1/2-ERK1/2 pathway. Based on these results, we conclude that ligand-dependent activation of PDGFR-beta and its downstream effectors Akt, MEK1/2, and ERK1/2 is strongly modulated (inhibited) by hyperosmotic cell shrinkage, whereas cell swelling does not seem to affect the activation of the receptor but rather to activate ERK1/2 via a different mechanism. It is thus likely that cell swelling via activation of ERK1/2 and cell shrinkage via activation of the p38 and JNK pathway and inhibition of the PDGFR signaling pathway may act as key players in the regulation of tissue homeostasis.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/fisiologia , Animais , Camundongos , Células NIH 3T3 , Pressão Osmótica
9.
Artigo em Inglês | MEDLINE | ID: mdl-17289411

RESUMO

This review focuses on using the knowledge on volume-sensitive transport systems in Ehrlich ascites tumour cells and NIH-3T3 cells to elucidate osmotic regulation of salt transport in epithelia. Using the intestine of the European eel (Anguilla anguilla) (an absorptive epithelium of the type described in the renal cortex thick ascending limb (cTAL)) we have focused on the role of swelling-activated K+- and anion-conductive pathways in response to hypotonicity, and on the role of the apical (luminal) Na+-K+-2Cl- cotransporter (NKCC2) in the response to hypertonicity. The shrinkage-induced activation of NKCC2 involves an interaction between the cytoskeleton and protein phosphorylation events via PKC and myosin light chain kinase (MLCK). Killifish (Fundulus heteroclitus) opercular epithelium is a Cl(-)-secreting epithelium of the type described in exocrine glands, having a CFTR channel on the apical side and the Na+/K+ ATPase, NKCC1 and a K+ channel on the basolateral side. Osmotic control of Cl- secretion across the operculum epithelium includes: (i) hyperosmotic shrinkage activation of NKCC1 via PKC, MLCK, p38, OSR1 and SPAK; (ii) deactivation of NKCC by hypotonic cell swelling and a protein phosphatase, and (iii) a protein tyrosine kinase acting on the focal adhesion kinase (FAK) to set levels of NKCC activity.


Assuntos
Enguias/fisiologia , Epitélio/metabolismo , Transporte de Íons , Animais
10.
Acta Physiol (Oxf) ; 187(1-2): 27-42, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16734740

RESUMO

Cell volume regulation is one of the most fundamental homeostatic mechanisms and essential for normal cellular function. At the same time, however, many physiological mechanisms are associated with regulatory changes in cell size meaning that the set point for cell volume regulation is under physiological control. Thus, cell volume is under a tight and dynamic control and abnormal cell volume regulation will ultimately lead to severe cellular dysfunction, including alterations in cell proliferation and cell death. This review describes the different swelling-activated ion channels that participate as key players in the maintenance of normal steady-state cell volume, with particular emphasis on the intracellular signalling pathways responsible for their regulation during hypotonic stress, cell proliferation and apoptosis.


Assuntos
Fenômenos Fisiológicos Celulares , Ativação do Canal Iônico/fisiologia , Canais Iônicos/metabolismo , Transdução de Sinais/fisiologia , Animais , Apoptose/fisiologia , Membrana Celular/metabolismo , Proliferação de Células , Tamanho Celular , Humanos , Concentração Osmolar
12.
J Exp Biol ; 208(Pt 6): 1063-77, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15767308

RESUMO

Hypotonic shock rapidly inhibits Cl(-) secretion by chloride cells, an effect that is osmotic and not produced by NaCl-depleted isosmotic solutions, yet the mechanism for the inhibition and its recovery are not known. We exposed isolated opercular epithelia, mounted in Ussing chambers, to hypotonic shock in the presence of a variety of chemicals: a general protein kinase C (PKC) inhibitor chelerythrine, Gö6976 that selectively blocks PKC alpha and beta subtypes, H-89 that blocks PKA, SB203580 that blocks p38 mitogen-activated protein kinase (MAPK), as well as serine/threonine protein phosphatase (PP1 and 2A) inhibitor okadaic acid, and finally tamoxifen, a blocker of volume-activated anion channels (VSOAC). Chelerythrine has no effect on hypotonic inhibition but blocked the recovery, indicating PKC involvement in stimulation. Gö6976 had little effect, suggesting that PKC alpha and PKC beta subtypes are not involved. H-89 did not block hypotonic inhibition but decreased the recovery, indicating PKA may be involved in the recovery and overshoot (after restoration of isotonic conditions). SB203580 significantly enhanced the decrease in current by hypotonic shock, suggesting an inhibitory role of p38 MAPK in the hypotonic inhibition. Okadaic acid increased the steady state current, slowed the hypotonic inhibition but made the decrease in current larger; also the recovery and overshoot were completely blocked. Hypotonic stress rapidly and transiently increased phosphorylated p38 MAPK (pp38) MAPK (measured by western analysis) by eightfold at 5 min, then more slowly again to sevenfold at 60 min. Hypertonic shock slowly increased p38 by sevenfold at 60 min. Phosphorylated JNK kinase was increased by 40-50% by both hypotonic and hypertonic shock and was still elevated at 30 min in hypertonic medium. By immunoblot analysis it was found that the stress protein kinase (SPAK) and oxidation stress response kinase 1 (OSR1) were present in salt and freshwater acclimated fish with higher expression in freshwater. By immunocytochemistry, SPAK, OSR1 and phosphorylated focal adhesion kinase (pFAK) were colocalized with NKCC at the basolateral membrane. The protein tyrosine kinase inhibitor genistein (100 micromol l(-1)) inhibited Cl(-) secretion that was high, increased Cl(-) secretion that was low and reduced immunocytochemical staining for phosphorylated FAK. We present a model for rapid control of CFTR and NKCC in chloride cells that includes: (1) activation of NKCC and CFTR via cAMP/PKA, (2) activation of NKCC by PKC, myosin light chain kinase (MLCK), p38, OSR1 and SPAK, (3) deactivation of NKCC by hypotonic cell swelling, Ca(2+) and an as yet unidentified protein phosphatase and (4) involvement of protein tyrosine kinase (PTK) acting on FAK to set levels of NKCC activity.


Assuntos
Cloretos/metabolismo , Fundulidae/fisiologia , Modelos Biológicos , Solução Salina Hipertônica/metabolismo , Equilíbrio Hidroeletrolítico/fisiologia , Alcaloides , Animais , Benzofenantridinas , Western Blotting , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Eletroforese em Gel de Poliacrilamida , Eletrofisiologia , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Proteína-Tirosina Quinases de Adesão Focal , Imidazóis/farmacologia , Imuno-Histoquímica , Canais Iônicos/antagonistas & inibidores , Isoquinolinas/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase 4 , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Nova Escócia , Ácido Okadáico/farmacologia , Fenantridinas/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Piridinas/farmacologia , Solução Salina Hipertônica/toxicidade , Sulfonamidas/farmacologia , Tamoxifeno/farmacologia , Equilíbrio Hidroeletrolítico/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
J Exp Biol ; 208(Pt 4): 749-60, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15695766

RESUMO

Control of cell volume is a fundamental and highly conserved physiological mechanism, essential for survival under varying environmental and metabolic conditions. Epithelia (such as intestine, renal tubule, gallbladder and gills) are tissues physiologically exposed to osmotic stress. Therefore, the activation of 'emergency' systems of rapid cell volume regulation is fundamental in their physiology. The aim of the present work was to study the physiological response to hypotonic stress in a salt-transporting epithelium, the intestine of the euryhaline teleost Anguilla anguilla. Eel intestinal epithelium, when symmetrically bathed with Ringer solution, develops a net Cl- current giving rise to a negative transepithelial potential at the basolateral side of the epithelium. The eel intestinal epithelium responded to a hypotonic challenge with a biphasic decrease in the transepithelial voltage (V(te)) and the short circuit current (I(sc)). This electrophysiological response correlated with a regulatory volume decrease (RVD) response, recorded by morphometrical measurement of the epithelium height. Changes in the transepithelial resistance were also observed following the hypotonicity exposure. The electrogenic V(te) and I(sc) responses to hypotonicity resulted from the activation of different K+ and anion conductive pathways on the apical and basolateral membranes of the epithelium: (a) iberiotoxin-sensitive K+ channels on the apical and basolateral membrane, (b) apamin-sensitive K+ channels mainly on the basolateral membrane, (c) DIDS-sensitive anion channels on the apical membrane. The functional integrity of the basal Cl- conductive pathway on the basolateral membrane is also required. The electrophysiological response to hypotonic stress was completely abolished by Ca2+ removal from the Ringer perfusing solution, but was not affected by depletion of intracellular Ca2+ stores by thapsigargin.


Assuntos
Anguilla/metabolismo , Tamanho Celular/efeitos dos fármacos , Soluções Hipotônicas/farmacologia , Mucosa Intestinal/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Canais de Potássio/metabolismo , Análise de Variância , Animais , Eletrofisiologia , Mucosa Intestinal/citologia , Mucosa Intestinal/fisiologia , Soluções Isotônicas , Concentração Osmolar , Canais de Potássio/efeitos dos fármacos , Solução de Ringer , Tapsigargina
14.
Pflugers Arch ; 449(2): 175-85, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15340851

RESUMO

Cell shrinkage is a ubiquitous feature of programmed cell death (PCD), but whether it is an obligatory signalling event in PCD is unclear. Heat shock protein 70 (Hsp70) potently counteracts PCD in many cells, by mechanisms that are incompletely understood. In the present investigation, we found that severe hypertonic stress greatly diminished the viability of murine fibrosarcoma cells (WEHI-902) and immortalized murine embryonic fibroblasts (iMEFs). This effect was attenuated markedly by Hsp70 over-expression. To determine whether the protective effect of Hsp70 was mediated via an effect on volume regulatory ion transport, we compared regulatory volume decrease (RVD) and increase (RVI) in control WEHI-902 cells and after increasing Hsp70 levels by heat shock or over-expression (WEHI-912). Hsp70 levels affected neither RVD, RVI nor the relative contributions of the Na(+)/H(+)-exchanger (NHE1) and Na(+),K(+),2Cl(-)-cotransporter (NKCC1) to RVI. Hypertonic stress induced caspase-3 activity in WEHI cells and iMEFs, an effect potentiated by Hsp70 in WEHI cells but inhibited by Hsp70 in iMEFs. Osmotic shrinkage-induced PCD was associated with Hsp70-inhibitable cysteine cathepsin release in iMEFs and attenuated by caspase and cathepsin inhibitors in WEHI cells. Treatment with TNF-alpha or the NHE1 inhibitor 5'-(N-ethyl-N-isopropyl)amiloride (EIPA) reduced the viability of WEHI cells further under isotonic and mildly, but not severely, hypertonic conditions. Thus, it is concluded that shrinkage-induced PCD involves both caspase- and cathepsin-dependent death mechanisms and is potently counteracted by Hsp70.


Assuntos
Apoptose/fisiologia , Fibrossarcoma , Proteínas de Choque Térmico HSP70/metabolismo , Animais , Caspases/metabolismo , Catepsinas/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular Tumoral , Tamanho Celular , Homeostase/fisiologia , Soluções Hipertônicas/farmacologia , Proteínas de Membrana/metabolismo , Camundongos , Pressão Osmótica , Transdução de Sinais/fisiologia , Trocador 1 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
15.
Biochim Biophys Acta ; 1566(1-2): 129-39, 2002 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-12421544

RESUMO

The eurohaline fish, Fundulus heteroclitus, adapts rapidly to enhanced salinity by increasing the ion secretion by gill chloride cells. An increase of approximately 70 mOsm in plasma osmolarity was previously found during the transition. To mimic this in vitro, isolated opercular epithelia of seawater-adapted Fundulus mounted in a modified Ussing chamber were exposed to an increase in NaCl and/or osmolarity on the basolateral side, which immediately increased I(SC). Various Cl(-) channel blockers as well as the K(+) channel blocker Ba(2+) added to the basolateral side all inhibited the steady-state as well as the hypertonic stimulation of I(SC). The exists -agonist isoproterenol stimulates I(SC) in standard Ringer solutions. In contrast, when cell volume was kept at the larger value by simultaneous addition of water, the stimulation with isoproterenol was abolished, suggesting that the key process for activation of the Na(+), K(+), 2Cl(-) cotransporter is cell shrinkage. The protein kinase C (PKC) inhibitor chelerythrine and the myosin light chain kinase (MLCK) inhibitor ML-7 had strong inhibitory effects on the mannitol activation of I(SC), thus both MLCK and PKC are involved. The two specific protein kinase A (PKA) inhibitors H-89 and KT 5720 had no effect after mannitol addition whereas isoproterenol stimulation was completely blocked by H-89. This indicates that PKA is involved in the activation of the apical Cl(-) channel via c-AMP whereas the shrinkage activation of the Na(+), K(+), 2Cl(-) cotransporter is independent of PKA activation. The steady-state Cl(-) secretion was stimulated by an inhibitor of serine/threonine phosphatases of the PP-1 and PP-2A type and inhibited by a PKC inhibitor but not by a PKA inhibitor. Thus, it seems to be determined by continuous phosphorylation and dephosphorylation involving PKC but not PKA. The steady-state Cl(-) secretion and the maximal obtainable Cl(-) secretion were measured in freshwater-adapted fish and in fish retransferred to saltwater. No I(SC) could be measured in freshwater-adapted fish or in the fish within the first 18 h after transfer to saltwater. As evidenced from Western blot analysis using antiserine-antibodies, a heavily serine phosphorylated protein of about 190 kDa was consistently observed in the saltwater-acclimated fish, but was only weakly present in freshwater-acclimated fish. This observation indicates that acclimatization to saltwater stimulates the expression of this 190-kDa protein and/or a serine/threonine kinase, which subsequently phosphorylates the protein.


Assuntos
Cloretos/metabolismo , Epitélio/metabolismo , Fundulidae/metabolismo , Brânquias/metabolismo , Proteínas de Protozoários , Aclimatação , Animais , Tamanho Celular/efeitos dos fármacos , Canais de Cloreto/antagonistas & inibidores , Cultura em Câmaras de Difusão , Condutividade Elétrica , Glibureto/farmacologia , Transporte de Íons , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , Quinase de Cadeia Leve de Miosina/metabolismo , Nitrobenzoatos/farmacologia , Concentração Osmolar , Fosforilação , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Água do Mar , Cloreto de Sódio , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Membro 2 da Família 12 de Carreador de Soluto , Fatores de Tempo
16.
J Membr Biol ; 189(1): 67-81, 2002 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12202953

RESUMO

The Na+/H+ exchanger isoforms NHE1, NHE2, and NHE3 were all found to be expressed in Ehrlich ascites tumor cells, as evaluated by Western blotting and confocal microscopy. Under unstimulated conditions, NHE1 was found predominantly in the plasma membrane, NHE3 intracellularly, and NHE2 in both compartments. Osmotic cell shrinkage elicited a rapid intracellular alkalinization, the sensitivity of which to EIPA (IC50 0.19 microM) and HOE 642 (IC50 0.85 microM) indicated that it predominantly reflected activation of NHE1. NHE activation by osmotic shrinkage was inhibited by the protein kinase C inhibitors chelerythrine (IC50 12.5 microM), Gö 6850 (5 microM), and Gö 6976 (1 microM), and by the p38 MAPK inhibitor SB 203580 (10 microM). Furthermore, hypertonic cell shrinkage elicited a biphasic increase in p38 MAPK phosphorylation, with the first significant increase detectable 2 minutes after the hypertonic challenge. Neither myosin light chain kinase-specific concentrations of ML-7 (IC50 40 microM) nor ERK1/2 inhibition by PD 98059 (50 microM) had any effect on NHE activation. Under isotonic conditions, the serine/threonine protein phosphatase inhibitor calyculin A elicited an EIPA- and HOE 642-inhibitable intracellular alkalinization, indicating NHE1 activation. Similarly, shrinkage-induced NHE activation was potentiated by calyculin A. The calyculin A-induced alkalinization was not associated with an increase in the free, intracellular calcium concentration, but was abolished by chelerythrine. It is concluded that shrinkage-induced NHE activation is dependent on PKC and p38 MAPK, but not on MLCK or ERK1/2. NHE activity under both iso- and hypertonic conditions is increased by inhibition of serine/threonine phosphatases, and this effect appears to be PKC-dependent.


Assuntos
Carcinoma de Ehrlich/metabolismo , Carcinoma de Ehrlich/patologia , Mecanotransdução Celular , Oxazóis/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Tamanho Celular , Meios de Cultura/farmacologia , Concentração de Íons de Hidrogênio , Soluções Hipertônicas/metabolismo , Soluções Hipertônicas/farmacologia , Toxinas Marinhas , Concentração Osmolar , Pressão Osmótica , Oxazóis/farmacologia , Isoformas de Proteínas/metabolismo , Proteína Quinase C/metabolismo , Sensibilidade e Especificidade , Trocador 3 de Sódio-Hidrogênio , Células Tumorais Cultivadas
17.
Exp Cell Res ; 277(1): 57-73, 2002 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12061817

RESUMO

Osmotic shrinkage of Ehrlich ascites tumor cells (EATC) elicited translocation of myosin II from the cytosol to the cortical region, and swelling elicits concentration of myosin II in the Golgi region. Rho kinase and p38 both appeared to be involved in shrinkage-induced myosin II reorganization. In contrast, the previously reported shrinkage-induced actin polymerization [Pedersen et al. (1999) Exp. Cell Res. 252, 63-74] was independent of Rho kinase, p38, myosin light chain kinase (MLCK), and protein kinase C (PKC), which thus do not exert their effects on the shrinkage-activated transporters via effects on F-actin. The subsequent F-actin depolymerization, however, appeared MLCK- and PKC-dependent, and the initial swelling-induced F-actin depolymerization was MLCK-dependent; both effects were apparently secondary to kinase-mediated effects on cell volume changes. NHE1 in EATC is activated both by osmotic shrinkage and by the serine/threonine phosphatase inhibitor Calyculin A (CL-A). Both stimuli caused Rho kinase-dependent myosin II relocation to the cortical cytoplasm, but in contrast to the shrinkage-induced F-actin polymerization, CL-A treatment elicited a slight F-actin depolymerization. Moreover, Rho kinase inhibition did not significantly affect NHE1 activation, neither by shrinkage nor by CL-A. Implications for the possible interrelationship between changes in F-actin and myosin II, protein phosphorylation, and cell volume regulation are discussed.


Assuntos
Actinas/metabolismo , Miosina Tipo II/metabolismo , Transdução de Sinais , Amidas/farmacologia , Animais , Anticorpos/imunologia , Azepinas/farmacologia , Carcinoma de Ehrlich , Tamanho Celular , Ativação Enzimática , Soluções Isotônicas , Toxinas Marinhas , Camundongos , Miosina Tipo II/imunologia , Naftalenos/farmacologia , Osmose , Oxazóis/farmacologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosforilação , Isoformas de Proteínas/imunologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Piridinas/farmacologia , Trocadores de Sódio-Hidrogênio/metabolismo , Células Tumorais Cultivadas
18.
Pflugers Arch ; 442(4): 622-33, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11510896

RESUMO

The effects of extracellular and intracellular pH (pHo and pHi respectively) on the regulatory volume decrease (RVD) response and on the volume-sensitive K+ and Cl- currents (IK,vol and ICl,vol respectively) were studied in Ehrlich ascites tumour cells. Alkaline pHo accelerated and acidic pHo decelerated the RVD response significantly. Intra- and extracellular alkalinisation increased the amplitude of IK,vol whereas acidification had an inhibitory effect. The magnitude of ICl,vol was not affected by changes in pHi or pHo. A significant reduction in the activation time for IK,vol after hypotonic cell swelling was observed upon moderate intracellular alkalinisation (to pHi 7.9). A further increase in pHi to 8.4 resulted in the spontaneous activation of an IK under isotonic conditions which resembled IK,vol with respect to its pharmacological profile and current/voltage (I/V) relation. Noise analysis demonstrated that the increased amplitude of IK,vol at alkaline pH resulted mainly from an increase in the number of channels (N) contributing to the current. The channel open probability, Po, was largely unaffected by pH. The pH dependence and the biophysical and pharmacological properties of IK,vol are similar to those of the cloned tandem pore-domain acid-sensitive K+ (TASK) channels, and in the current study the presence of TASK-1 was confirmed in Ehrlich cells.


Assuntos
Carcinoma de Ehrlich , Concentração de Íons de Hidrogênio , Proteínas do Tecido Nervoso , Canais de Potássio de Domínios Poros em Tandem , Canais de Potássio/metabolismo , Potássio/metabolismo , Álcalis , Animais , Artefatos , Soluções Tampão , Tamanho Celular , Cloretos/metabolismo , Ativação do Canal Iônico/fisiologia , Soluções Isotônicas , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos , Técnicas de Patch-Clamp , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/metabolismo
19.
Biochim Biophys Acta ; 1517(3): 455-9, 2001 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-11342226

RESUMO

We report the identification of an EST encoding a murine cysteinyl leukotriene (mCysLT) receptor. LTD4, LTC4 and LTE4 but not LTB4 or various nucleotides activated Ca2+-evoked Cl- currents in mCysLT1 expressing Xenopus laevis oocytes. The response to LTD4 was blocked by MK-571, reduced by pretreatment with pertussis toxin (PTX), and was partly dependent on extracellular Ca2+. The identified murine CysLT1 receptor differs from the hCysLT1 receptor with regard to PTX sensitivity, receptor-mediated Ca2+ influx, and antagonist sensitivity.


Assuntos
Proteínas de Membrana , Receptores de Leucotrienos/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Cálcio/metabolismo , Cálcio/farmacologia , Cloretos/metabolismo , Etiquetas de Sequências Expressas , Feminino , Humanos , Técnicas In Vitro , Antagonistas de Leucotrienos , Leucotrienos/farmacologia , Camundongos , Dados de Sequência Molecular , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Toxina Pertussis , Propionatos/farmacologia , Quinolinas/farmacologia , Receptores de Leucotrienos/metabolismo , Homologia de Sequência de Aminoácidos , Fatores de Virulência de Bordetella/farmacologia , Xenopus laevis
20.
Artigo em Inglês | MEDLINE | ID: mdl-11913449

RESUMO

The cell swelling-activated K+ channel in Ehrlich ascites tumour cells has a conductance of 5 pS estimated from noise analysis of the volume-sensitive whole-cell K+ current (I(K,vol)). I(K,vol) exhibits Goldman-Hodgkin-Katz type behaviour and is insensitive to clotrimazole, apamin and charybdotoxin (ChTX), but inhibited by clofilium. Its small conductance, lack of intrinsic voltage-dependence and peculiar pharmacological profile are similar to properties described for the two-pore domain background K+ TASK channels. Neither Ca2+ nor ATP work as initiators in the activation of I(K,vol). In contrast, several investigations in Ehrlich cells suggest an important role for leukotriene D4 (LTD4) in the activation of I(K,vol). Under isotonic conditions, LTD4 activates Ca2+-dependent, ChTX-sensitive K+ channels as well as Ca2+-independent. ChTX-insensitive K+ channels. The LTD4-activated, ChTX-insensitive K+ current exhibits a current-voltage relation, pharmacological profile and single channel conductance similar to that of I(K,vol), indicating that LTD4 is the signalling molecule responsible for activation of the volume-sensitive K+ channels in Ehrlich cells. Hypotonic swelling of Ehrlich cells results in translocation of the 85-kDa cytosolic (c) PLA2alpha to the nucleus where it is activated. This activation leads to an increase in arachidonic acid release followed by an increased release of leukotrienes, and is essential in cell swelling-induced activation of I(K,vol) and of the organic osmolyte channels.


Assuntos
Carcinoma de Ehrlich/metabolismo , Canais de Potássio/metabolismo , Transdução de Sinais , Animais , Carcinoma de Ehrlich/patologia , Canais de Potássio/efeitos dos fármacos , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...