Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Physiol Rev ; 99(4): 2015-2113, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31507243

RESUMO

Na+/H+ exchangers play pivotal roles in the control of cell and tissue pH by mediating the electroneutral exchange of Na+ and H+ across cellular membranes. They belong to an ancient family of highly evolutionarily conserved proteins, and they play essential physiological roles in all phyla. In this review, we focus on the mammalian Na+/H+ exchangers (NHEs), the solute carrier (SLC) 9 family. This family of electroneutral transporters constitutes three branches: SLC9A, -B, and -C. Within these, each isoform exhibits distinct tissue expression profiles, regulation, and physiological roles. Some of these transporters are highly studied, with hundreds of original articles, and some are still only rudimentarily understood. In this review, we present and discuss the pioneering original work as well as the current state-of-the-art research on mammalian NHEs. We aim to provide the reader with a comprehensive view of core knowledge and recent insights into each family member, from gene organization over protein structure and regulation to physiological and pathophysiological roles. Particular attention is given to the integrated physiology of NHEs in the main organ systems. We provide several novel analyses and useful overviews, and we pinpoint main remaining enigmas, which we hope will inspire novel research on these highly versatile proteins.


Assuntos
Equilíbrio Ácido-Base , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Evolução Molecular , Regulação da Expressão Gênica , Humanos , Conformação Proteica , Trocadores de Sódio-Hidrogênio/química , Trocadores de Sódio-Hidrogênio/genética , Relação Estrutura-Atividade , Distribuição Tecidual
2.
BMC Cancer ; 17(1): 542, 2017 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-28806945

RESUMO

BACKGROUND: Chronic angiogenesis is a hallmark of most tumors and takes place in a hostile tumor microenvironment (TME) characterized by hypoxia, low nutrient and glucose levels, elevated lactate and low pH. Despite this, most studies addressing angiogenic signaling use hypoxia as a proxy for tumor conditions. Here, we compared the effects of hypoxia and TME conditions on regulation of the Na+/H+ exchanger NHE1, Ser/Thr kinases Akt1-3, and downstream effectors in endothelial cells. METHODS: Human umbilical vein endothelial cells (HUVEC) and Ea.hy926 endothelial cells were exposed to simulated TME (1% hypoxia, low serum, glucose, pH, high lactate) or 1% hypoxia for 24 or 48 h, with or without NHE1 inhibition or siRNA-mediated knockdown. mRNA and protein levels of NHE1, Akt1-3, and downstream effectors were assessed by qPCR and Western blotting, vascular endothelial growth factor (VEGF) release by ELISA, and motility by scratch assay. RESULTS: Within 24 h, HIF-1α level and VEGF mRNA level were increased robustly by TME and modestly by hypoxia alone. The NHE1 mRNA level was decreased by both hypoxia and TME, and NHE1 protein was reduced by TME in Ea.hy926 cells. Akt1-3 mRNA was detected in HUVEC and Ea.hy926 cells, Akt1 most abundantly. Akt1 protein expression was reduced by TME yet unaffected by hypoxia, while Akt phosphorylation was increased by TME. The Akt loss was partly reversed by MCF-7 human breast cancer cell conditioned medium, suggesting that in vivo, the cancer cell secretome may compensate for adverse effects of TME on endothelial cells. TME, yet not hypoxia, reduced p70S6 kinase activity and ribosomal protein S6 phosphorylation and increased eIF2α phosphorylation, consistent with inhibition of protein translation. Finally, TME reduced Retinoblastoma protein phosphorylation and induced poly-ADP-ribose polymerase (PARP) cleavage consistent with inhibition of proliferation and induction of apoptosis. NHE1 knockdown, mimicking the effect of TME on NHE1 expression, reduced Ea.hy926 migration. TME effects on HIF-1α, VEGF, Akt, translation, proliferation or apoptosis markers were unaffected by NHE1 knockdown/inhibition. CONCLUSIONS: NHE1 and Akt are downregulated by TME conditions, more potently than by hypoxia alone. This inhibits endothelial cell migration and growth in a manner likely modulated by the cancer cell secretome.


Assuntos
Células Endoteliais da Veia Umbilical Humana/metabolismo , Hipóxia/metabolismo , Neoplasias/fisiopatologia , Proteínas Proto-Oncogênicas c-akt/genética , Trocador 1 de Sódio-Hidrogênio/genética , Células A549 , Feminino , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Células MCF-7 , Masculino , Neoplasias/metabolismo , Transdução de Sinais , Microambiente Tumoral
3.
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
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.
Exp Cell Res ; 316(15): 2538-53, 2010 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-20542029

RESUMO

Altered pH-regulatory ion transport is characteristic of many cancers; however, the mechanisms and consequences are poorly understood. Here, we investigate how a truncated, constitutively active ErbB2 receptor (DeltaNErbB2) common in breast cancer impacts on the Na(+)/H(+)-exchanger NHE1 and the Na(+),HCO(3)(-)-cotransporter NBCn1 in MCF-7 human breast cancer cells and address the roles of these transporters in chemotherapy resistance. Upon DeltaNErbB2 expression, mRNA and protein levels of NBCn1, yet not of NHE1, increased several-fold, and the localization of both transporters was altered paralleling extensive morphological changes. The rate of pH(i) recovery after acid loading increased by 50% upon DeltaNErbB2 expression. Knockdown and pharmacological inhibition confirmed the involvement of both NHE1 and NBCn1 in acid extrusion. NHE1 inhibition or knockdown sensitized DeltaNErbB2-expressing cells to cisplatin-induced programmed cell death (PCD) in a caspase-, cathepsin-, and reactive oxygen species-dependent manner. NHE1 inhibition augmented cisplatin-induced caspase activity and lysosomal membrane permeability followed by cysteine cathepsin release. In contrast, NBCn1 inhibition attenuated cathepsin release and had no net effect on viability. These findings warrant studies of NHE1 as a potential target in breast cancer and demonstrate that in spite of their similar transport functions, NHE1 and NBCn1 serve different functions in MCF-7 cells.


Assuntos
Equilíbrio Ácido-Base/genética , Neoplasias da Mama/genética , Proteínas de Transporte de Cátions/fisiologia , Resistencia a Medicamentos Antineoplásicos/genética , Receptor ErbB-2/genética , Simportadores de Sódio-Bicarbonato/fisiologia , Trocadores de Sódio-Hidrogênio/fisiologia , Antineoplásicos/uso terapêutico , Transporte Biológico/genética , Transporte Biológico/fisiologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Catepsinas/metabolismo , Proteínas de Transporte de Cátions/antagonistas & inibidores , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Concentração de Íons de Hidrogênio , Membranas Intracelulares/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Estrutura Terciária de Proteína/genética , RNA Interferente Pequeno/farmacologia , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Simportadores de Sódio-Bicarbonato/antagonistas & inibidores , Simportadores de Sódio-Bicarbonato/genética , Simportadores de Sódio-Bicarbonato/metabolismo , Trocador 1 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/metabolismo
6.
Magn Reson Med ; 62(2): 341-7, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19526499

RESUMO

A crucial requirement in MR-guided interventions is the visualization of catheter devices in real time. However, true 3D visualization of the full length of catheters has hitherto been impossible given scan time constraints. Compressed sensing (CS) has recently been proposed as a method to accelerate MR imaging of sparse objects. Images acquired with active interventional devices exhibit a high CNR and are inherently sparse, therefore rendering CS ideally suited for accelerating data acquisition. A framework for true visualization of active catheters in 3D is proposed employing CS to gain high undersampling factors making real-time applications feasible. Constraints are introduced taking into account prior knowledge of catheter geometry and catheter motion over time to improve and accelerate image reconstruction. The potential of the method is demonstrated using computer simulations and phantom experiments and in vivo feasibility is demonstrated in a pig experiment.


Assuntos
Cateterismo Cardíaco/métodos , Vasos Coronários/anatomia & histologia , Aumento da Imagem/métodos , Imageamento Tridimensional/métodos , Angiografia por Ressonância Magnética/métodos , Radiografia Intervencionista/métodos , Animais , Compressão de Dados/métodos , Angiografia por Ressonância Magnética/instrumentação , Imagens de Fantasmas , Suínos
7.
Eur J Vasc Endovasc Surg ; 37(6): 714-21, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19112034

RESUMO

OBJECTIVES: Atherosclerosis is recognised as an inflammatory disease, and new diagnostic tools are warranted to evaluate plaque inflammatory activity and risk of cardiovascular events. We investigated [18]-fluorodeoxyglucose (FDG) uptake in vulnerable carotid plaques visualised by positron emission tomography (PET). Uptake was correlated to quantitative gene expression of known markers of inflammation and plaque vulnerability. METHODS: Ten patients with recent transient ischaemic attack and carotid artery stenosis (>50%) underwent combined FDG-PET and computed tomography angiography (CTA) the day before carotid endarterectomy. Plaque mRNA expression of the inflammatory cytokine interleukin 18 (IL-18), the macrophage-specific marker CD68 and the two proteinases, Cathepsin K and matrix metalloproteinase 9 (MMP-9), were quantified using real-time quantitative polymerase chain reaction. RESULTS: Consistent up-regulation of CD68 (3.8-fold+/-0.9; mean+/-standard error), Cathepsin K (2.1-fold+/-0.5), MMP-9 (122-fold+/-65) and IL-18 (3.4-fold+/-0.7) were found in the plaques, compared to reference-artery specimens. The FDG uptake by plaques was strongly correlated with CD68 gene expression (r=0.71, P=0.02). Any correlations with Cathepsin K, MMP-9 or IL-18 gene expression were weaker. CONCLUSIONS: FDG-PET uptake in carotid plaques is correlated to gene expression of CD68 and other molecular markers of inflammation and vulnerability.


Assuntos
Estenose das Carótidas/diagnóstico por imagem , Fluordesoxiglucose F18 , Ataque Isquêmico Transitório/etiologia , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos , Idoso , Idoso de 80 Anos ou mais , Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/genética , Estenose das Carótidas/complicações , Estenose das Carótidas/genética , Estenose das Carótidas/cirurgia , Catepsina K , Catepsinas/genética , Endarterectomia das Carótidas , Feminino , Regulação da Expressão Gênica , Humanos , Mediadores da Inflamação/análise , Interleucina-18/genética , Ataque Isquêmico Transitório/diagnóstico por imagem , Ataque Isquêmico Transitório/genética , Ataque Isquêmico Transitório/cirurgia , Masculino , Metaloproteinase 9 da Matriz/genética , Pessoa de Meia-Idade , Valor Preditivo dos Testes , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medição de Risco , Índice de Gravidade de Doença , Tomografia Computadorizada por Raios X
8.
Acta Physiol (Oxf) ; 223(3): e13068, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29575508

RESUMO

Precise spatiotemporal regulation of intracellular pH (pHi ) is a prerequisite for normal cell function, and changes in pHi or pericellular pH (pHe ) exert important signalling functions. It is well established that proliferation of mammalian cells is dependent on a permissive pHi in the slightly alkaline range (7.0-7.2). It is also clear that mitogen signalling in nominal absence of HCO3- is associated with an intracellular alkalinization (~0.3 pH unit above steady-state pHi ), which is secondary to activation of Na+ /H+ exchange. However, it remains controversial whether this increase in pHi is part of the mitogenic signal cascade leading to cell cycle entry and progression, and whether it is relevant under physiological conditions. Furthermore, essentially all studies of pHi in mammalian cell proliferation have focused on the mitogen-induced G0-G1 transition, and the regulation and roles of pHi during the cell cycle remain poorly understood. The aim of this review is to summarize and critically discuss the possible roles of pHi and pHe in cell cycle progression. While the focus is on the mammalian cell cycle, important insights from studies in lower eukaryotes are also discussed. We summarize current evidence of links between cell cycle progression and pHi and discuss possible pHi - and pHe sensors and signalling pathways relevant to mammalian proliferation control. The possibility that changes in pHi during cell cycle progression may be an integral part of the checkpoint control machinery is explored. Finally, we discuss the relevance of links between pH and proliferation in the context of the perturbed pH homoeostasis and acidic microenvironment of solid tumours.


Assuntos
Ciclo Celular , Proliferação de Células , Animais , Homeostase , Humanos , Concentração de Íons de Hidrogênio , Biossíntese de Proteínas , Transdução de Sinais
9.
Oncogene ; 35(16): 2112-22, 2016 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-26212013

RESUMO

Increased metabolism and insufficient blood supply cause acidic waste product accumulation in solid cancers. During carcinogenesis, cellular acid extrusion is upregulated but the underlying molecular mechanisms and their consequences for cancer growth and progression have not been established. Genome-wide association studies have indicated a possible link between the Na⁺, HCO3⁻-cotransporter NBCn1 (SLC4A7) and breast cancer. We tested the functional consequences of NBCn1 knockout (KO) for breast cancer development. NBCn1 protein expression increased 2.5-fold during breast carcinogenesis and was responsible for the increased net acid extrusion and alkaline intracellular pH of breast cancer compared with normal breast tissue. Genetic disruption of NBCn1 delayed breast cancer development: tumor latency was ~50% increased while tumor growth rate was ~65% reduced in NBCn1 KO compared with wild-type (WT) mice. Breast cancer histopathology in NBCn1 KO mice differed from that in WT mice and included less aggressive tumor types. The extracellular tumor microenvironment in NBCn1 KO mice contained higher concentrations of glucose and lower concentrations of lactate than that in WT mice. Independently of NBCn1 genotype, the cleaved fraction of poly(ADP-ribose) polymerase (PARP)-1 and expression of monocarboxylate transporter (MCT)1 increased while phosphorylation of Akt and ERK1 decreased as functions of tumor volume. Cell proliferation, evaluated from Ki-67 and phospho-histone H3staining, was ~60% lower in breast cancer of NBCn1 KO than that of WT mice when corrected for variations in tumor size. We conclude that NBCn1 facilitates acid extrusion from breast cancer tissue, maintains the alkaline intracellular environment and promotes aggressive cancer development and growth.


Assuntos
Neoplasias Mamárias Experimentais/patologia , Simportadores de Sódio-Bicarbonato/metabolismo , Animais , Proliferação de Células , Feminino , Glicólise , Concentração de Íons de Hidrogênio , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Knockout , Simportadores de Sódio-Bicarbonato/genética
10.
Biochim Biophys Acta ; 1374(1-2): 94-106, 1998 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-9814856

RESUMO

The mechanisms, by which the P2 receptor agonists adenosine 5'-triphosphate (ATP) and uridine 5'-triphosphate (UTP) evoke an increase in the free cytosolic calcium concentration ([Ca2+]i) and in intracellular pH (pHi), have been investigated in Ehrlich ascites tumor cells. The increase in [Ca2+]i evoked by ATP or UTP is abolished after depletion of intracellular Ca2+ stores with thapsigargin in Ca2+-free medium, and is inhibited by U73122, an inhibitor of phospholipase C (PLC), indicating that the increase in [Ca2+]i is primarily due to release from intracellular, Ins(1,4,5)P3-sensitive Ca2+ stores. ATP also activates a capacitative Ca2+-entry pathway. ATP as well as UTP evokes a biphasic change in pHi, consisting of an initial acidification followed by alkalinization. Suramin and 4,4'-diisothiocyano-2,2'-stilbene-disulfonic acid (DIDS) inhibit the biphasic change in pHi, apparently by acting as antagonists at P2 receptors. The alkalinization evoked by the P2 receptor agonists is found to be due to activation of a 5'-(N-ethyl-N-isopropyl)amiloride (EIPA)-sensitive Na+/H+ exchanger. ATP and UTP elicit rapid cell shrinkage, presumably due to activation of Ca2+ sensitive K+ and Cl- efflux pathways. Preventing cell shrinkage, either by incubating the cells at high extracellular K+ concentration, or by adding the K+-channel blocker, charybdotoxin, does not affect the increase in [Ca2+]i, but abolishes the activation of the Na+/H+ exchanger, indicating that activation of the Na+/H+ exchanger is secondary to the Ca2+-induced cell shrinkage.


Assuntos
Carcinoma de Ehrlich/metabolismo , Receptores Purinérgicos P2/metabolismo , Transdução de Sinais/fisiologia , Ácido 4,4'-Di-Isotiocianoestilbeno-2,2'-Dissulfônico/farmacologia , Trifosfato de Adenosina/farmacologia , Amilorida/análogos & derivados , Amilorida/farmacologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Carcinoma de Ehrlich/patologia , Tamanho Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Estrenos/farmacologia , Concentração de Íons de Hidrogênio , Camundongos , Agonistas do Receptor Purinérgico P2 , Antagonistas do Receptor Purinérgico P2 , Pirrolidinonas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Trocadores de Sódio-Hidrogênio/metabolismo , Suramina/farmacologia , Tapsigargina/farmacologia , Fosfolipases Tipo C/antagonistas & inibidores , Uridina Trifosfato/farmacologia
11.
Biochim Biophys Acta ; 1416(1-2): 271-84, 1999 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-9889382

RESUMO

The supernatant from a suspension of Ehrlich cells exposed to centrifugation at 700xg for 45 s induced a transient increase in the intracellular concentration of free, cytosolic Ca2+, [Ca2+]i, as well as activation of an outwardly rectifying whole-cell current when added to a suspension of non-stimulated cells. These effects were inhibited by suramin, a non-specific P2 receptor antagonist, and mimicked by ATP. Reversed phase HPLC analysis revealed that the supernatant from Ehrlich cells exposed to centrifugation contained 2. 6+/-0.2 microM ATP, and that the mechanical stress-induced release of ATP was inhibited by glibenclamide and verapamil, non-specific inhibitors of the cystic fibrosis transmembrane conductance regulator and P-glycoprotein, respectively. After trypan blue staining, less than 0.5% of the cells were unable to extrude the dye. Addition of extracellular ATP induced a suramin-sensitive, transient, concentration-dependent increase in [Ca2+]i, activation of an outwardly rectifying whole-cell current and a hyperpolarization of the plasma membrane. The ATP-induced hyperpolarization of the plasma membrane was strongly inhibited in the presence of charybdotoxin (ChTX), an inhibitor of several Ca2+-activated K+ channels, suggesting that stimulation of P2 receptors in Ehrlich cells evokes a Ca2+-activated K+ current. The relative potencies of several nucleotides (ATP, UTP, ADP, 2-MeSATP, alpha,beta-MeATP, bzATP) in eliciting an increase in [Ca2+]i, as well as the effect of repetitive addition of nucleotides were investigated. The results lead us to conclude that mechanical stimulation of Ehrlich cells leads to release of ATP, which in turn stimulates both P2Y1 and P2Y2 receptors, resulting in Ca2+ influx as well as release and activation of an outwardly rectifying whole-cell current.


Assuntos
Trifosfato de Adenosina/metabolismo , Carcinoma de Ehrlich/metabolismo , Estresse Mecânico , Trifosfato de Adenosina/farmacologia , Animais , Cálcio/metabolismo , Charibdotoxina/farmacologia , Agonistas do Receptor Purinérgico P2 , Antagonistas do Receptor Purinérgico P2 , Receptores Purinérgicos P2/análise , Receptores Purinérgicos P2Y1 , Receptores Purinérgicos P2Y2 , Suramina/farmacologia , Células Tumorais Cultivadas
12.
Acta Physiol (Oxf) ; 213(4): 868-81, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25565132

RESUMO

The volume-regulated anion channel (VRAC) plays a pivotal role in cell volume regulation in essentially all cell types studied. Additionally, VRAC appears to contribute importantly to a wide range of other cellular functions and pathological events, including cell motility, cell proliferation, apoptosis and excitotoxic glutamate release in stroke. Although biophysically, pharmacologically and functionally thoroughly described, VRAC has until very recently remained a genetic orphan. The search for the molecular identity of VRAC has been long and has yielded multiple potential candidates, all of which eventually turned out to have properties not fully compatible with those of VRAC. Recently, two groups have independently identified the protein leucine-rich repeats containing 8A (LRRC8A), belonging to family of proteins (LRRC8A-E) distantly related to pannexins, as the likely pore-forming subunit of VRAC. In this brief review, we summarize the history of the discovery of VRAC, outline its basic biophysical and pharmacological properties, link these to several cellular functions in which VRAC appears to play important roles, and sketch the amazing search for the molecular identity of this channel. Finally, we describe properties of the LRRC8 proteins, highlight some features of the LRRC8A knockout mouse and discuss the impact of the discovery of LRRC8 as VRAC on future research.


Assuntos
Regulação da Expressão Gênica/fisiologia , Canais Iônicos/fisiologia , Proteínas de Membrana/metabolismo , Animais , Humanos , Ativação do Canal Iônico , Proteínas de Membrana/genética , Família Multigênica
13.
Arch Neurol ; 53(7): 617-24, 1996 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8929169

RESUMO

OBJECTIVE: To determine the dosing, response expectation, efficacy, and most rational strategy for using intramuscular injections of botulinum toxin A (BTX) for limb disorders. DESIGN: Open-label prospective analysis of outcome after BTX treatment in patients with limb disorders. PROCEDURE: Botulinum toxin A prepared from lyophilized botulinum toxin was injected into selected upper and lower limb muscles under electromyographic guidance. Booster injections were given every 10 to 14 days during the first month (if needed) until optimal effects were achieved. Clinical data and muscle strength testing were obtained before the first injections and repeated at each visit. Level of disability, global functional improvement, and relief of pain were evaluated 6 to 8 weeks after the first set of injections. Practical and meaningful BTX doses by muscle, limb, or condition according to specified levels of efficacy were developed. MAIN OUTCOME MEASURES: Botulinum toxin A efficacy was calculated as an arithmetic combination of changes in the 3 clinical ratings before and after administration of BTX. RESULTS: Botulinum toxin A injections were given to 187 patients with limb disorders during an 8-year period (136 with dystonia, 37 with parkinsonian, essential, and cerebellar tremors, and 14 with spasticity). Four overall outcomes from no effect to almost complete improvement in the use of the limb or relief of pain were found, and determined the strategy for follow-up injections. Average BTX efficacy for all patients was 65% and ranged from 83.5% for focal hand dystonia to 35.7% for parkinsonian tremor. Botulinum toxin A injections relieved pain, independent of motor function, in 82.7% of patients with painful muscle spasms. CONCLUSIONS: Botulinum toxin A was found to be a safe and useful treatment of various limb conditions. Botulinum toxin A was significantly more effective when only a few muscles needing low doses were injected, and tended to be more useful in dystonia and spasticity than tremor. Candidates for BTX injection could be categorized functionally into 3 groups independent of the underlying disorders. The only significant adverse effect of BTX injection in limbs was transient weakness in injected or neighboring muscles.


Assuntos
Toxinas Botulínicas/uso terapêutico , Extremidades/fisiopatologia , Doenças Neuromusculares/tratamento farmacológico , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Doenças Cerebelares/tratamento farmacológico , Doenças Cerebelares/fisiopatologia , Criança , Pré-Escolar , Avaliação da Deficiência , Relação Dose-Resposta a Droga , Humanos , Injeções Intramusculares , Pessoa de Meia-Idade , Espasticidade Muscular/tratamento farmacológico , Espasticidade Muscular/fisiopatologia , Doenças Neuromusculares/fisiopatologia , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/fisiopatologia , Estudos Prospectivos , Resultado do Tratamento , Tremor/tratamento farmacológico , Tremor/fisiopatologia
14.
J Am Soc Mass Spectrom ; 5(5): 452-9, 1994 May.
Artigo em Inglês | MEDLINE | ID: mdl-24222600

RESUMO

Results of mass analyzed ion kinetic energy (MIKE) spectra and kinetic energy release (KER) measurements of diastereomeric octahedral cobalt complexes indicate that these diastereomers can be distinguished in the gas phase. Four alkyl tartrate esters were complexed to cobalt trisacetylacetonate (Co(acac)3) in the presence of a chiral auxiliary, RR- and SS-threohydrobenzoin. Different KER values of the product ion generated from [Co(acac)2/D- or L-diisopropyl tartrate](+) reflect differences in the precursor ion structure. The dissociation pathway resulting in this product ion is believed to arise via a hydride transfer from the acetylacetonate ligand to the metal center with subsequent loss of neutral organic species. It has been established that two conditions are necessary for observation of chiral recognition in this system; (1) the cobalt complex must be octahedral and (2) a chemical kinetic resolving agent must be present during formation of the complex.

15.
Curr Pharm Des ; 18(10): 1345-71, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22360557

RESUMO

The Na⁺/H⁺-exchanger 1, NHE1 (SLC9A1) and the electroneutral Na⁺,HCO3⁻ cotransporter NBCn1 (SLC4A7) are coexpressed in a wide range of tissues. Under normal physiological conditions these transporters play an ostensibly similar role, namely that of net acid extrusion after cellular acidification. In addition, they have been implicated in multiple other cellular processes, including regulation of transepithelial transport, cell volume, cell death/survival balance, and cell motility. In spite of their apparent functional similarity, the two transporters also serve distinctly different functions and are differentially regulated. Here, we provide an update on the basic structure, function, regulation, physiology and pharmacology of NHE1 and NBCn1, with particular focus on the factors responsible for their functional similarities and differences. Finally, we highlight recent findings implicating these transporters in cancer development, and discuss issues relating to NHE1 and NBCn1 as potential targets in cancer treatment.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias/metabolismo , Simportadores de Sódio-Bicarbonato/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Antineoplásicos/farmacologia , Proteínas de Transporte de Cátions/antagonistas & inibidores , Proteínas de Transporte de Cátions/genética , Humanos , Simportadores de Sódio-Bicarbonato/antagonistas & inibidores , Simportadores de Sódio-Bicarbonato/genética , Trocador 1 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Trocadores de Sódio-Hidrogênio/genética
16.
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
17.
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
18.
Am J Physiol Cell Physiol ; 293(5): C1605-15, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17804611

RESUMO

The involvement of group VI Ca(2+)-independent PLA(2)s (iPLA(2)-VI) in in vitro ischemia [oxygen and glucose deprivation (OGD)] in mouse C2C12 myotubes was investigated. OGD induced a time-dependent (0-6 h) increase in bromoenol lactone (BEL)-sensitive iPLA(2) activity, which was suppressed by specific short interfering (si)RNA knockdown of iPLA(2)-VIA. OGD was associated with an increase in iPLA(2)-VIA protein levels, whereas mRNA levels were unchanged. The levels of iPLA(2)-VIB mRNA and protein were not increased by OGD. RT-PCR and Western blot analysis identified a mouse iPLA(2)-VIA homolog to catalytically inactive 50-kDa iPLA(2)-VIA-ankyrin variants previously identified in humans. Both the mRNA and protein levels of this approximately 50-kDa variant were reduced significantly within 1 h following OGD. In C2C12 myoblasts, iPLA(2)-VIA seemed to predominantly reside at the endoplasmatic reticulum, where it accumulated further during OGD. A time-dependent reduction in cell viability during the early OGD period (3 h) was partially prevented by iPLA(2)-VIA knockdown or pharmacological inhibition (10 microM BEL), whereas iPLA(2)-VIA overexpression had no effect on cell viability. Taken together, these data demonstrate that OGD in C2C12 myotubes is associated with an increase in iPLA(2)-VIA activity that decreases cell viability. iPLA(2)-VIA activation may be modulated by changes in the levels of active and inactive iPLA(2)-VIA isoforms.


Assuntos
Fosfolipases A2 do Grupo IV/biossíntese , Isquemia/enzimologia , Fibras Musculares Esqueléticas/enzimologia , Músculo Esquelético/irrigação sanguínea , Animais , Ácidos Araquidônicos/metabolismo , Hipóxia Celular , Linhagem Celular , Sobrevivência Celular , Retículo Endoplasmático/enzimologia , Indução Enzimática , Glucose/deficiência , Glucose/metabolismo , Fosfolipases A2 do Grupo IV/química , Fosfolipases A2 do Grupo IV/genética , Isquemia/genética , Camundongos , Peso Molecular , Fibras Musculares Esqueléticas/efeitos dos fármacos , Naftalenos/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Isoformas de Proteínas , Pironas/farmacologia , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Fatores de Tempo
19.
Acta Physiol (Oxf) ; 187(1-2): 75-85, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16734744

RESUMO

Phospholipase A2 (PLA2) activity is increased in mammalian cells in response to numerous stimuli such as osmotic challenge, oxidative stress and exposure to allergens. The increased PLA2 activity is seen as an increased release of free, polyunsaturated fatty acids, e.g. arachidonic acid and membrane-bound lysophospholipids. Even though arachidonic acid acts as a second messenger in its own most mammalian cells seem to rely on oxidation of the fatty acid into highly potent second messengers via, e.g. cytochrome P450, the cyclo-oxygenase, or the lipoxygenase systems for downstream signalling. Here, we review data that illustrates that stress-induced PLA2 activity involves various PLA2 subtypes and that the PLA2 in question is determined by the cell type and the physiological stress condition.


Assuntos
Isoenzimas/metabolismo , Células Musculares/metabolismo , Fosfolipases A/metabolismo , Sistemas do Segundo Mensageiro/fisiologia , Estresse Fisiológico/metabolismo , Animais , Hipóxia Celular , Tamanho Celular , Ativação Enzimática/fisiologia , Humanos , Concentração Osmolar , Fosfolipases A2 , Espécies Reativas de Oxigênio/metabolismo
20.
Am J Physiol Regul Integr Comp Physiol ; 291(1): R1-25, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16484438

RESUMO

Maintenance of a stable cell volume and intracellular pH is critical for normal cell function. Arguably, two of the most important ion transporters involved in these processes are the Na+/H+ exchanger isoform 1 (NHE1) and Na+ -K+ -2Cl- cotransporter isoform 1 (NKCC1). Both NHE1 and NKCC1 are stimulated by cell shrinkage and by numerous other stimuli, including a wide range of hormones and growth factors, and for NHE1, intracellular acidification. Both transporters can be important regulators of cell volume, yet their activity also, directly or indirectly, affects the intracellular concentrations of Na+, Ca2+, Cl-, K+, and H+. Conversely, when either transporter responds to a stimulus other than cell shrinkage and when the driving force is directed to promote Na+ entry, one consequence may be cell swelling. Thus stimulation of NHE1 and/or NKCC1 by a deviation from homeostasis of a given parameter may regulate that parameter at the expense of compromising others, a coupling that may contribute to irreversible cell damage in a number of pathophysiological conditions. This review addresses the roles of NHE1 and NKCC1 in the cellular responses to physiological and pathophysiological stress. The aim is to provide a comprehensive overview of the mechanisms and consequences of stress-induced stimulation of these transporters with focus on the heart, brain, and blood. The physiological stressors reviewed are metabolic/exercise stress, osmotic stress, and mechanical stress, conditions in which NHE1 and NKCC1 play important physiological roles. With respect to pathophysiology, the focus is on ischemia and severe hypoxia where the roles of NHE1 and NKCC1 have been widely studied yet remain controversial and incompletely elucidated.


Assuntos
Encéfalo/metabolismo , Miocárdio/metabolismo , Trocadores de Sódio-Hidrogênio/sangue , Trocadores de Sódio-Hidrogênio/metabolismo , Simportadores de Cloreto de Sódio-Potássio/sangue , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Animais , Humanos , Membro 2 da Família 12 de Carreador de Soluto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA