Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nutrients ; 16(5)2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38474717

RESUMO

(Poly)phenolic-rich Mediterranean plants such as Thymbra spicata have been associated with several health-promoting effects. The nutritional value, as well as physiological interaction of T. spicata with the gastrointestinal tract, has not been investigated before. The nutritional composition of T. spicata leaves was here characterized by standard analytical methods. T. spicata leaves were subjected to ethanolic extraction, simulated gastrointestinal digestion, and anaerobic microbial gut fermentation. Phenols/flavonoid contents and radical scavenging activity were assessed by colorimetric methods. The volatile organic compounds (VOCs) were detected by gas chromatography coupled with mass spectrometry. The effect on intestinal integrity was evaluated using a Caco-2 monolayers mounted in a Ussing chamber. T. spicata contains a high amount of fiber (12.3%) and unsaturated fatty acids (76% of total fat). A positive change in VOCs including short-chain fatty acids was observed without significant change in viable microbe. T. spicata and carvacrol (main phenolic compound) enhanced ionic currents in a concentration-dependent manner without compromising the Caco-2 monolayer's integrity. These effects were partially lost upon simulated digestion and completely abolished after colonic fermentation in line with polyphenols and carvacrol content. Conclusion: T. spicata represents a promising nutrient for the modulation of gut microbiota and the gut barrier. Further studies must better define its mechanisms of action.


Assuntos
Cimenos , Lamiaceae , Humanos , Fermentação , Células CACO-2 , Cromatografia Gasosa-Espectrometria de Massas , Lamiaceae/química , Fenóis , Digestão , Ácidos Graxos Voláteis
2.
Int J Mol Sci ; 24(13)2023 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-37445810

RESUMO

Intracellular pH (pHi) regulation is a challenge for the exocrine pancreas, where the luminal secretion of bicarbonate-rich fluid is accompanied by interstitial flows of acid. This acid-base transport requires a plethora of ion transporters, including bicarbonate transporters and the Na+/H+ exchanger isoform 1 (NHE1), which are dysregulated in Pancreatic Ductal Adenocarcinoma (PDAC). PDAC progression is favored by a Collagen-I rich extracellular matrix (ECM) which exacerbates the physiological interstitial acidosis. In organotypic cultures of normal human pancreatic cells (HPDE), parenchymal cancer cells (CPCs) and cancer stem cells (CSCs) growing on matrices reproducing ECM changes during progression, we studied resting pHi, the pHi response to fluxes of NaHCO3 and acidosis and the role of NHE1 in pHi regulation. Our findings show that: (i) on the physiological ECM, HPDE cells have the most alkaline pHi, followed by CSCs and CPCs, while a Collagen I-rich ECM reverses the acid-base balance in cancer cells compared to normal cells; (ii) both resting pHi and pHi recovery from an acid load are reduced by extracellular NaHCO3, especially in HPDE cells on a normal ECM; (iii) cancer cell NHE1 activity is less affected by NaHCO3. We conclude that ECM composition and the fluctuations of pHe cooperate to predispose pHi homeostasis towards the presence of NaHCO3 gradients similar to that expected in the tumor.


Assuntos
Acidose , Neoplasias , Humanos , Concentração de Íons de Hidrogênio , Bicarbonatos/metabolismo , Matriz Extracelular/metabolismo , Colágeno Tipo I , Ductos Pancreáticos/metabolismo , Células Epiteliais/metabolismo , Trocadores de Sódio-Hidrogênio
3.
Nat Cancer ; 3(12): 1464-1483, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36522548

RESUMO

Solid tumors are generally characterized by an acidic tumor microenvironment (TME) that favors cancer progression, therapy resistance and immune evasion. By single-cell RNA-sequencing analysis in individuals with pancreatic ductal adenocarcinoma (PDAC), we reveal solute carrier family 4 member 4 (SLC4A4) as the most abundant bicarbonate transporter, predominantly expressed by epithelial ductal cells. Functionally, SLC4A4 inhibition in PDAC cancer cells mitigates the acidosis of the TME due to bicarbonate accumulation in the extracellular space and a decrease in lactate production by cancer cells as the result of reduced glycolysis. In PDAC-bearing mice, genetic or pharmacological SLC4A4 targeting improves T cell-mediated immune response and breaches macrophage-mediated immunosuppression, thus inhibiting tumor growth and metastases. In addition, Slc4a4 targeting in combination with immune checkpoint blockade is able to overcome immunotherapy resistance and prolong survival. Overall, our data propose SLC4A4 as a therapeutic target to unleash an antitumor immune response in PDAC.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Simportadores de Sódio-Bicarbonato , Animais , Camundongos , Bicarbonatos/metabolismo , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Imunoterapia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Simportadores de Sódio-Bicarbonato/genética , Microambiente Tumoral , Tolerância Imunológica , Neoplasias Pancreáticas
4.
Int J Mol Sci ; 22(22)2021 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-34830416

RESUMO

We previously showed that mesothelial cells in human peritoneum express the water channel aquaporin 1 (AQP1) at the plasma membrane, suggesting that, although in a non-physiological context, it may facilitate osmotic water exchange during peritoneal dialysis (PD). According to the three-pore model that predicts the transport of water during PD, the endothelium of peritoneal capillaries is the major limiting barrier to water transport across peritoneum, assuming the functional role of the mesothelium, as a semipermeable barrier, to be negligible. We hypothesized that an intact mesothelial layer is poorly permeable to water unless AQP1 is expressed at the plasma membrane. To demonstrate that, we characterized an immortalized cell line of human mesothelium (HMC) and measured the osmotically-driven transmesothelial water flux in the absence or in the presence of AQP1. The presence of tight junctions between HMC was investigated by immunofluorescence. Bioelectrical parameters of HMC monolayers were studied by Ussing Chambers and transepithelial water transport was investigated by an electrophysiological approach based on measurements of TEA+ dilution in the apical bathing solution, through TEA+-sensitive microelectrodes. HMCs express Zo-1 and occludin at the tight junctions and a transepithelial vectorial Na+ transport. Real-time transmesothelial water flux, in response to an increase of osmolarity in the apical solution, indicated that, in the presence of AQP1, the rate of TEA+ dilution was up to four-fold higher than in its absence. Of note, we confirmed our data in isolated mouse mesentery patches, where we measured an AQP1-dependent transmesothelial osmotic water transport. These results suggest that the mesothelium may represent an additional selective barrier regulating water transport in PD through functional expression of the water channel AQP1.


Assuntos
Aquaporina 1/genética , Transporte Biológico/genética , Epitélio/metabolismo , Peritônio/metabolismo , Aquaporinas/genética , Linhagem Celular , Regulação da Expressão Gênica/genética , Humanos , Diálise Peritoneal/normas , Peritônio/patologia , Sódio/metabolismo
5.
Cell Calcium ; 51(5): 393-401, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22361140

RESUMO

Secretory granules of pancreatic ß-cells contain high concentrations of Ca2+ ions that are co-released with insulin in the extracellular milieu upon activation of exocytosis. As a consequence, an increase in the extracellular Ca2+ concentration ([Ca2+]ext) in the microenvironment immediately surrounding ß-cells should be expected following the exocytotic event. Using Ca2+-selective microelectrodes we show here that both high glucose and non-nutrient insulinotropic agents elicit a reversible increase of [Ca2+]ext within rat insulinoma (INS-1E) ß-cells pseudoislets. The glucose-induced increases in [Ca2+]ext are blocked by pretreatment with different Ca2+ channel blockers. Physiological agonists acting as positive or negative modulators of the insulin secretion and drugs known to intersect the secretory machinery at different levels also induce [Ca2+]ext changes as predicted on the basis of their described action on insulin secretion. Finally, the glucose-induced [Ca2+]ext increase is strongly inhibited after disruption of the actin web, indicating that the dynamic [Ca2+]ext changes recorded in INS-1E pseudoislets by Ca2+-selective microelectrodes occur mainly as a consequence of exocytosis of Ca2+-rich granules. In conclusion, our data directly demonstrate that the extracellular spaces surrounding ß-cells constitute a restricted domain where Ca2+ is co-released during insulin exocytosis, creating the basis for an autocrine/paracrine cell-to-cell communication system via extracellular Ca2+ sensors.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Glucose/administração & dosagem , Insulina/metabolismo , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Comunicação Celular/efeitos dos fármacos , Microambiente Celular/efeitos dos fármacos , Exocitose , Glibureto/administração & dosagem , Concentração de Íons de Hidrogênio , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Microeletrodos , Ratos , Vesículas Secretórias/efeitos dos fármacos
6.
Toxicol Appl Pharmacol ; 245(2): 264-71, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20307561

RESUMO

Cadmium, a toxic environmental pollutant, affects the function of different organs such as lungs, liver and kidney. Less is known about its toxic effects on the gastric mucosa. The aim of this study was to investigate the mechanisms by which cadmium impacts on the physiology of gastric mucosa. To this end, intact amphibian mucosae were mounted in Ussing chambers and the rate of acid secretion, short circuit current (I(sc)), transepithelial potential (V(t)) and resistance (R(t)) were recorded in the continuous presence of cadmium. Addition of cadmium (20 microM to 1mM) on the serosal but not luminal side of the mucosae resulted in inhibition of acid secretion and increase in NPPB-sensitive, chloride-dependent short circuit current. Remarkably, cadmium exerted its effects only on histamine-stimulated tissues. Experiments with TPEN, a cell-permeant chelator for heavy metals, showed that cadmium acts from the intracellular side of the acid secreting cells. Furthermore, cadmium-induced inhibition of acid secretion and increase in I(sc) cannot be explained by an action on: 1) H(2) histamine receptor, 2) Ca(2+) signalling 3) adenylyl cyclase or 4) carbonic anhydrase. Conversely, cadmium was ineffective in the presence of the H(+)/K(+)-ATPase blocker omeprazole suggesting that the two compounds likely act on the same target. Our findings suggest that cadmium affects the functionality of histamine-stimulated gastric mucosa by inhibiting the H(+)/K(+)-ATPase from the intracellular side. These data shed new light on the toxic effect of this dangerous environmental pollutant and may result in new avenues for therapeutic intervention in acute and chronic intoxication.


Assuntos
Cádmio/toxicidade , Poluentes Ambientais/toxicidade , Ácido Gástrico/metabolismo , Mucosa Gástrica/efeitos dos fármacos , Adenilil Ciclases/metabolismo , Animais , Anidrases Carbônicas/metabolismo , Quelantes/farmacologia , Relação Dose-Resposta a Droga , Etilenodiaminas/farmacologia , Mucosa Gástrica/metabolismo , Histamina/farmacologia , Técnicas In Vitro , Omeprazol/farmacologia , Inibidores da Bomba de Prótons , Rana esculenta
7.
Gastroenterology ; 137(3): 955-64, 964.e1-2, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19328800

RESUMO

BACKGROUND & AIMS: Deoxycholate (DC) has proapoptotic and tumorigenic effects in different cell types of the gastrointestinal tract. Exposure of BHK-21 (stromal) cells to DC induces Ca(2+) entry at the plasma membrane, which affects intracellular Ca(2+) signaling. We assessed whether DC-induced increases in [Ca(2+)] can impinge on plasma membrane properties (eg, ionic conductances) involved in cell apoptosis. METHODS: Single- and double-barreled microelectrodes were used to measure membrane potential (V(m)) and extracellular [K(+)] in BHK-21 fibroblasts and Caco-2 colon carcinoma cells. Apoptosis was assessed by Hoechst labeling, propidium iodide staining, and caspase-3 and caspase-7 assays. RESULTS: DC-induced cell membrane hyperpolarization was directly measured with intracellular microelectrodes in both cell lines. Diverse Ca(2+) mobilizing agents, such as membrane receptor agonists, an inhibitor of the sarco/endoplasmic reticulum Ca(2+) adenosine triphosphatase and a Ca(2+) ionophore, also induced increases in V(m). Removal of extracellular Ca(2+) reduced the agonist- and DC-induced membrane hyperpolarization by approximately 15% and 60%, respectively. These findings indicate a prominent role for Ca(2+) entry at the plasma membrane in the action of this bile salt. Blockade of Ca(2+)-activated K(+) conductances by charybdotoxin and apamin reduced DC-induced hyperpolarization by 75% and 64% in BHK-21 and Caco-2 cells, respectively. These inhibitors also reduced the DC-induced increase in extracellular [K(+)] by 75% and cell apoptosis by approximately 50% in both cell lines. CONCLUSIONS: Ca(2+)-dependent K(+) conductance is an important regulator of DC-induced apoptosis in stromal and colon cancer cells.


Assuntos
Apoptose/efeitos dos fármacos , Ácido Desoxicólico/farmacologia , Canais de Potássio Cálcio-Ativados/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Apamina/farmacologia , Bradicinina/farmacologia , Células CACO-2 , Cálcio/metabolismo , ATPases Transportadoras de Cálcio/antagonistas & inibidores , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular , Charibdotoxina/farmacologia , Quelantes/farmacologia , Cricetinae , Citosol/metabolismo , Relação Dose-Resposta a Droga , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Espaço Extracelular/metabolismo , Fibroblastos/fisiologia , Humanos , Indóis/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Microeletrodos , Potássio/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo
8.
Front Biosci ; 13: 2989-99, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17981772

RESUMO

Different gene targeting approaches have been developed to modify endogenous genomic DNA in both human and mouse cells. Briefly, the process involves the targeting of a specific mutation in situ leading to the gene correction and the restoration of a normal gene function. Most of these protocols with therapeutic potential are oligonucleotide based, and rely on endogenous enzymatic pathways. One gene targeting approach, "Small Fragment Homologous Replacement (SFHR)", has been found to be effective in modifying genomic DNA. This approach uses small DNA fragments (SDF) to target specific genomic loci and induce sequence and subsequent phenotypic alterations. This study shows that SFHR can stably introduce a 3-bp deletion (deltaF508, the most frequent cystic fibrosis (CF) mutation) into the Cftr (CF Transmembrane Conductance Regulator) locus in the mouse embryonic stem (ES) cell genome. After transfection of deltaF508-SDF into murine ES cells, SFHR-mediated modification was evaluated at the molecular levels on DNA and mRNA obtained from transfected ES cells. About 12% of transcript corresponding to deleted allele was detected, while 60% of the electroporated cells completely lost any measurable CFTR-dependent chloride efflux. The data indicate that the SFHR technique can be used to effectively target and modify genomic sequences in ES cells. Once the SFHR-modified ES cells differentiate into different cell lineages they can be useful for elucidating tissue-specific gene function and for the development of transplantation-based cellular and therapeutic protocols.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Células-Tronco Embrionárias/metabolismo , Animais , Clonagem Molecular , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , DNA/metabolismo , Marcação de Genes , Técnicas Genéticas , Camundongos , Microscopia de Fluorescência , Microscopia de Vídeo , Neurônios Motores/metabolismo , Mutação , RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco
9.
J Biol Chem ; 282(18): 13477-86, 2007 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-17363364

RESUMO

The mechanisms for the formation of the osmotic gradient driving water movements in the gastric gland and its modulation via the extracellular Ca(2+)-sensing receptor (CaR) were investigated. Real time measurements of net water flux in the lumen of single gastric glands of the intact amphibian stomach were performed using ion-selective double-barreled microelectrodes. Water movement was measured by recording changes in the concentration of impermeant TEA(+) ions ([TEA(+)](gl)) with TEA(+)-sensitive microelectrodes inserted in the lumen of individual gastric glands. Glandular K(+) (K(+)(gl)) and H(+) (pH(gl)) were also measured by using K(+)- and H(+)-sensitive microelectrodes, respectively. Stimulation with histamine significantly decreased [TEA](gl), indicating net water flow toward the gland lumen. This response was inhibited by the H(+)/K(+)-ATPase inhibitor, SCH 28080. Histamine also elicited a significant and reversible increase in [K(+)](gl) that was blocked by chromanol 293B, a blocker of KCQN1 K(+) channels. Histamine failed to induce net water flow in the presence of chromanol 293B. In the "resting state," stimulation of CaR with diverse agonists resulted in significant increase in [TEA](gl). CaR activation also significantly reduced histamine-induced water secretion and apical K(+) transport. Our data validate the strong link between histamine-stimulated acid secretion and water transport. We also show that cAMP-dependent [K(+)](gl) elevation prior to the onset of acid secretion generates the osmotic gradient initially driving water into the gastric glands and that CaR activation inhibits this process, probably through reduction of intracellular cAMP levels.


Assuntos
Proteínas de Anfíbios/metabolismo , Ácido Gástrico/metabolismo , Mucosa Gástrica/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Água/metabolismo , Proteínas de Anfíbios/antagonistas & inibidores , Animais , Cromanos/farmacologia , AMP Cíclico/metabolismo , Inibidores Enzimáticos/farmacologia , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Histamina/farmacologia , Imidazóis/farmacologia , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/fisiologia , Microeletrodos , Osmose/efeitos dos fármacos , Osmose/fisiologia , Bloqueadores dos Canais de Potássio/farmacologia , Inibidores da Bomba de Prótons , Rana esculenta , Receptores de Detecção de Cálcio/antagonistas & inibidores , Sulfonamidas/farmacologia
10.
J Cell Biol ; 166(1): 111-9, 2004 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-15240573

RESUMO

It is generally assumed that the functional consequences of stimulation with Ca2+ -mobilizing agonists are derived exclusively from the second messenger action of intracellular Ca2+, acting on targets inside the cells. However, during Ca2+ signaling events, Ca2+ moves in and out of the cell, causing changes not only in intracellular Ca2+, but also in local extracellular Ca2+. The fact that numerous cell types possess an extracellular Ca2+ "sensor" raises the question of whether these dynamic changes in external [Ca2+] may serve some sort of messenger function. We found that in intact gastric mucosa, the changes in extracellular [Ca2+] secondary to carbachol-induced increases in intracellular [Ca2+] were sufficient and necessary to elicit alkaline secretion and pepsinogen secretion, independent of intracellular [Ca2+] changes. These findings suggest that extracellular Ca2+ can act as a "third messenger" via Ca2+ sensor(s) to regulate specific subsets of tissue function previously assumed to be under the direct control of intracellular Ca2+.


Assuntos
Adenina/análogos & derivados , Cálcio/metabolismo , Carbacol/farmacologia , Mucosa Gástrica/patologia , Ácido 4,4'-Di-Isotiocianoestilbeno-2,2'-Dissulfônico/farmacologia , Adenina/farmacologia , Animais , Eletrodos , Eletrofisiologia , Inibidores Enzimáticos/farmacologia , Mucosa Gástrica/metabolismo , Concentração de Íons de Hidrogênio , Microscopia de Fluorescência , Pepsinogênio A/química , Pepsinogênio A/metabolismo , Ranidae , Transdução de Sinais , Espermina/metabolismo
11.
Cell Calcium ; 35(3): 297-306, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15200154

RESUMO

In multicellular organisms, cells are crowded together in organized communities, surrounded by an interstitial fluid of extremely limited volume. Local communication between adjacent cells is known to occur through gap junctions in cells that are physically connected, or through the release of paracrine signaling molecules (e.g. ATP, glutamate, nitric oxide) that diffuse to their target receptors through the extracellular microenvironment. Recent evidence hints that calcium ions may possibly be added to the list of paracrine messengers that allow cells to communicate with one another. Local fluctuations in extracellular [Ca2+] can be generated as a consequence of intracellular Ca2+ signaling events, owing to the activation of Ca2+ influx and efflux pathways at the plasma membrane. In intact tissues, where the interstitial volumes between cells are much smaller than the cells themselves, this can result in significant alterations in external [Ca2+]. This article will explore emerging evidence that these extracellular [Ca2+] changes can be detected by the extracellular calcium-sensing receptor (CaR) on adjacent cells, forming the basis for a paracrine signaling system. Such a mechanism could potentially provide CaR-expressing cells with the means to sense the Ca2+ signaling status of their neighbors, and expand the utility of the intracellular Ca2+ signal to a domain outside the cell.


Assuntos
Comunicação Celular/fisiologia , Epitélio/fisiologia , Receptores de Detecção de Cálcio/fisiologia , Animais , Comunicação Autócrina/fisiologia , Cálcio/análise , Cálcio/fisiologia , Espaço Extracelular/química , Espaço Extracelular/metabolismo , Mucosa Gástrica/metabolismo , Humanos , Espaço Intracelular/química , Espaço Intracelular/metabolismo , Modelos Biológicos
12.
Acta Histochem ; 106(1): 21-7, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15032325

RESUMO

Caveolin-1, an integral membrane protein, is the principal component of caveolae, which are specialised vesicular microdomains of the plasma membrane. Caveolae are found in most cell types, but they are most abundant in adipocytes, endothelial cells, fibroblasts, and muscle cells. Functionally, they have been implicated in endothelial transcytosis, potocytosis, and signal transduction. Recently, caveolin-1 has been found unexpectedly in the cytoplasm, mitochondria and elements of the secretory pathways of exocrine secretory cells. We have co-localised caveolin-1 and pepsinogen immunohistochemically in serous cells of oesophageal glands of the red-legged frog, Rana aurora aurora. Thus, according to its intracellular localisation pattern, caveolin-1 may be either a soluble protein, located in secretory droplets, or a protein that is inserted in caveolar membranes. Soluble caveolin-1, which is probably embedded in a lipid particle surrounded by a phospholipid shell, may be involved in intracellular and extracellular lipid transport. In the gut, caveolin-1-rich lipid particles can act as donor particles to facilitate (protein-mediated) intestinal uptake of cholesterol and phospholipids. Our findings strengthen the hypothesis that caveolin-1 has a physiological autocrine/paracrine function and demonstrate that secretion of this protein also occurs in vertebrates other than mammals, such as amphibians, which may be a useful alternative animal model to study caveolin-1.


Assuntos
Caveolinas/metabolismo , Esôfago/metabolismo , Pepsinogênio A/metabolismo , Ranidae/metabolismo , Animais , Caveolina 1 , Caveolinas/análise , Glândulas Exócrinas/química , Glândulas Exócrinas/citologia , Glândulas Exócrinas/ultraestrutura , Mucosa Gástrica/química , Mucosa Gástrica/citologia , Células Caliciformes/citologia , Imuno-Histoquímica , Microscopia Imunoeletrônica , Células Parietais Gástricas/química , Células Parietais Gástricas/citologia , Células Parietais Gástricas/ultraestrutura , Pepsinogênio A/análise , Vesículas Secretórias/química , Vesículas Secretórias/ultraestrutura
13.
J Biol Chem ; 278(41): 39503-8, 2003 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-12888563

RESUMO

Inositol 1,4,5-trisphosphate (InsP3)-induced Ca2+ release from intracellular stores displays complex kinetic behavior. While it well established that cytosolic [Ca2+] can modulate release by acting on the InsP3 receptor directly, the role of the filling state of internal Ca2+stores in modulating Ca2+ release remains unclear. Here we have reevaluated this topic using a technique that permits rapid and reversible changes in free [Ca2+] in internal stores of living intact cells without altering cytoplasmic [Ca2+], InsP3 receptors, or sarcoendoplasmic reticulum Ca2+ ATPases (SERCAs). N,N,N',N'-Tetrakis(2-pyridylmethyl)ethylene diamine (TPEN), a membrane-permeant, low affinity Ca2+ chelator was used to manipulate [Ca2+] in intracellular stores, while [Ca2+] changes within the store were monitored directly with the low-affinity Ca2+ indicator, mag-fura-2, in intact BHK-21 cells. 200 microM TPEN caused a rapid drop in luminal free [Ca2+] and significantly reduced the extent of the response to stimulation with 100 nm bradykinin, a calcium-mobilizing agonist. The same effect was observed when intact cells were pretreated with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid(acetoxymethyl ester) (BAPTA-AM) to buffer cytoplasmic [Ca2+] changes. Although inhibition of Ca2+ uptake using the SERCA inhibitor tBHQ permitted significantly larger release of Ca2+ from stores, TPEN still attenuated the release in the presence of tBHQ in BAPTA-AM-loaded cells. These results demonstrate that the filling state of stores modulates the magnitude of InsP3-induced Ca2+release by additional mechanism(s) that are independent of regulation by cytoplasmic [Ca2+] or effects on SERCA pumps.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Ácido Egtázico/análogos & derivados , Inositol 1,4,5-Trifosfato/farmacologia , Animais , Canais de Cálcio/metabolismo , ATPases Transportadoras de Cálcio/antagonistas & inibidores , ATPases Transportadoras de Cálcio/metabolismo , Linhagem Celular , Quelantes/farmacologia , Cricetinae , Ácido Egtázico/farmacologia , Inibidores Enzimáticos/farmacologia , Etilenodiaminas/farmacologia , Hidroquinonas/farmacologia , Receptores de Inositol 1,4,5-Trifosfato , Metais Pesados/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...