Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Br J Cancer ; 122(2): 266-278, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31819190

RESUMO

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) has poor survival and treatment options. PDAC cells shift their metabolism towards glycolysis, which fuels the plasma membrane calcium pump (PMCA), thereby preventing Ca2+-dependent cell death. The ATP-generating pyruvate kinase-M2 (PKM2) is oncogenic and overexpressed in PDAC. This study investigated the PKM2-derived ATP supply to the PMCA as a potential therapeutic locus. METHODS: PDAC cell growth, migration and death were assessed by using sulforhodamine-B/tetrazolium-based assays, gap closure assay and poly-ADP ribose polymerase (PARP1) cleavage, respectively. Cellular ATP and metabolism were assessed using luciferase/fluorescent-based assays and the Seahorse XFe96 analyzer, respectively. Cell surface biotinylation identified membrane-associated proteins. Fura-2 imaging was used to assess cytosolic Ca2+ overload and in situ Ca2+ clearance. PKM2 knockdown was achieved using siRNA. RESULTS: The PKM2 inhibitor (shikonin) reduced PDAC cell proliferation, cell migration and induced cell death. This was due to inhibition of glycolysis, ATP depletion, inhibition of PMCA and cytotoxic Ca2+ overload. PKM2 associates with plasma membrane proteins providing a privileged ATP supply to the PMCA. PKM2 knockdown reduced PMCA activity and reduced the sensitivity of shikonin-induced cell death. CONCLUSIONS: Cutting off the PKM2-derived ATP supply to the PMCA represents a novel therapeutic strategy for the treatment of PDAC.


Assuntos
Carcinoma Ductal Pancreático/tratamento farmacológico , Proteínas de Transporte/genética , Proliferação de Células/genética , Proteínas de Membrana/genética , Neoplasias Pancreáticas/tratamento farmacológico , Hormônios Tireóideos/genética , Trifosfato de Adenosina/metabolismo , Cálcio/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Proteínas de Transporte/antagonistas & inibidores , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citosol/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Humanos , Proteínas de Membrana/antagonistas & inibidores , Naftoquinonas/farmacologia , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Pâncreas/patologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteínas de Ligação a Hormônio da Tireoide
3.
J Biol Chem ; 290(41): 24760-71, 2015 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-26294767

RESUMO

Evidence suggests that the plasma membrane Ca(2+)-ATPase (PMCA), which is critical for maintaining a low intracellular Ca(2+) concentration ([Ca(2+)]i), utilizes glycolytically derived ATP in pancreatic ductal adenocarcinoma (PDAC) and that inhibition of glycolysis in PDAC cell lines results in ATP depletion, PMCA inhibition, and an irreversible [Ca(2+)]i overload. We explored whether this is a specific weakness of highly glycolytic PDAC by shifting PDAC cell (MIA PaCa-2 and PANC-1) metabolism from a highly glycolytic phenotype toward mitochondrial metabolism and assessing the effects of mitochondrial versus glycolytic inhibitors on ATP depletion, PMCA inhibition, and [Ca(2+)]i overload. The highly glycolytic phenotype of these cells was first reversed by depriving MIA PaCa-2 and PANC-1 cells of glucose and supplementing with α-ketoisocaproate or galactose. These culture conditions resulted in a significant decrease in both glycolytic flux and proliferation rate, and conferred resistance to ATP depletion by glycolytic inhibition while sensitizing cells to mitochondrial inhibition. Moreover, in direct contrast to cells exhibiting a high glycolytic rate, glycolytic inhibition had no effect on PMCA activity and resting [Ca(2+)]i in α-ketoisocaproate- and galactose-cultured cells, suggesting that the glycolytic dependence of the PMCA is a specific vulnerability of PDAC cells exhibiting the Warburg phenotype.


Assuntos
Trifosfato de Adenosina/metabolismo , Membrana Celular/enzimologia , Glicólise , Neoplasias Pancreáticas/patologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Adenocarcinoma/patologia , Cálcio/metabolismo , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Citosol/efeitos dos fármacos , Citosol/metabolismo , Inibidores Enzimáticos/farmacologia , Galactose/farmacologia , Glicólise/efeitos dos fármacos , Humanos , Ácido Iodoacético/farmacologia , Cetoácidos/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/antagonistas & inibidores
4.
J Physiol ; 598(6): 1119-1120, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32053213
5.
J Biol Chem ; 289(34): 23582-95, 2014 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-24993827

RESUMO

Acute pancreatitis is a serious and sometimes fatal inflammatory disease where the pancreas digests itself. The non-oxidative ethanol metabolites palmitoleic acid (POA) and POA-ethylester (POAEE) are reported to induce pancreatitis caused by impaired mitochondrial metabolism, cytosolic Ca(2+) ([Ca(2+)]i) overload and necrosis of pancreatic acinar cells. Metabolism and [Ca(2+)]i are linked critically by the ATP-driven plasma membrane Ca(2+)-ATPase (PMCA) important for maintaining low resting [Ca(2+)]i. The aim of the current study was to test the protective effects of insulin on cellular injury induced by the pancreatitis-inducing agents, ethanol, POA, and POAEE. Rat pancreatic acinar cells were isolated by collagenase digestion and [Ca(2+)]i was measured by fura-2 imaging. An in situ [Ca(2+)]i clearance assay was used to assess PMCA activity. Magnesium green (MgGreen) and a luciferase-based ATP kit were used to assess cellular ATP depletion. Ethanol (100 mM) and POAEE (100 µM) induced a small but irreversible Ca(2+) overload response but had no significant effect on PMCA activity. POA (50-100 µM) induced a robust Ca(2+) overload, ATP depletion, inhibited PMCA activity, and consequently induced necrosis. Insulin pretreatment (100 nm for 30 min) prevented the POA-induced Ca(2+) overload, ATP depletion, inhibition of the PMCA, and necrosis. Moreover, the insulin-mediated protection of the POA-induced Ca(2+) overload was partially prevented by the phosphoinositide-3-kinase (PI3K) inhibitor, LY294002. These data provide the first evidence that insulin directly protects pancreatic acinar cell injury induced by bona fide pancreatitis-inducing agents, such as POA. This may have important therapeutic implications for the treatment of pancreatitis.


Assuntos
Ácidos Graxos Monoinsaturados/farmacologia , Insulina/fisiologia , Pâncreas/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Animais , Cálcio/metabolismo , Morte Celular , Cromonas/farmacologia , Etanol/administração & dosagem , Etanol/metabolismo , Ácidos Graxos/metabolismo , Fluorescência , Morfolinas/farmacologia , Pâncreas/citologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Ratos , Ratos Sprague-Dawley
6.
J Biol Chem ; 288(50): 36007-19, 2013 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-24158437

RESUMO

Pancreatic cancer is an aggressive cancer with poor prognosis and limited treatment options. Cancer cells rapidly proliferate and are resistant to cell death due, in part, to a shift from mitochondrial metabolism to glycolysis. We hypothesized that this shift is important in regulating cytosolic Ca(2+) ([Ca(2+)]i), as the ATP-dependent plasma membrane Ca(2+) ATPase (PMCA) is critical for maintaining low [Ca(2+)]i and thus cell survival. The present study aimed to determine the relative contribution of mitochondrial versus glycolytic ATP in fuelling the PMCA in human pancreatic cancer cells. We report that glycolytic inhibition induced profound ATP depletion, PMCA inhibition, [Ca(2+)]i overload, and cell death in PANC1 and MIA PaCa-2 cells. Conversely, inhibition of mitochondrial metabolism had no effect, suggesting that glycolytic ATP is critical for [Ca(2+)]i homeostasis and thus survival. Targeting the glycolytic regulation of the PMCA may, therefore, be an effective strategy for selectively killing pancreatic cancer while sparing healthy cells.


Assuntos
Trifosfato de Adenosina/metabolismo , Cálcio/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Membrana Celular/metabolismo , Glicólise , Neoplasias Pancreáticas/patologia , Adenocarcinoma/patologia , Trifosfato de Adenosina/deficiência , Morte Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Citosol/metabolismo , Humanos , Mitocôndrias/metabolismo
7.
J Biol Chem ; 287(3): 1823-36, 2012 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-22128146

RESUMO

Acute pancreatitis is a serious and sometimes fatal inflammatory disease of the pancreas without any reliable treatment or imminent cure. In recent years, impaired metabolism and cytosolic Ca(2+) ([Ca(2+)](i)) overload in pancreatic acinar cells have been implicated as the cardinal pathological events common to most forms of pancreatitis, regardless of the precise causative factor. Therefore, restoration of metabolism and protection against cytosolic Ca(2+) overload likely represent key therapeutic untapped strategies for the treatment of this disease. The plasma membrane Ca(2+)-ATPase (PMCA) provides a final common path for cells to "defend" [Ca(2+)](i) during cellular injury. In this paper, we use fluorescence imaging to show for the first time that insulin treatment, which is protective in animal models and clinical studies of human pancreatitis, directly protects pancreatic acinar cells from oxidant-induced cytosolic Ca(2+) overload and inhibition of the PMCA. This protection was independent of oxidative stress or mitochondrial membrane potential but appeared to involve the activation of Akt and an acute metabolic switch from mitochondrial to predominantly glycolytic metabolism. This switch to glycolysis appeared to be sufficient to maintain cellular ATP and thus PMCA activity, thereby preventing Ca(2+) overload, even in the face of impaired mitochondrial function.


Assuntos
Cálcio/metabolismo , Citosol/metabolismo , Insulina/metabolismo , Pâncreas Exócrino/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/antagonistas & inibidores , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Animais , Células Cultivadas , Glicólise/fisiologia , Humanos , Potencial da Membrana Mitocondrial/fisiologia , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Estresse Oxidativo/fisiologia , Pâncreas Exócrino/citologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley
8.
Am J Physiol Cell Physiol ; 300(4): C792-802, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21123738

RESUMO

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


Assuntos
Proliferação de Células , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Potássio/metabolismo , Cálcio/metabolismo , Membrana Celular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HEK293 , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5 , Técnicas de Patch-Clamp , Pirazóis/metabolismo , Transdução de Sinais/fisiologia , Canais de Sódio/genética , Canais de Sódio/metabolismo
9.
Nat Commun ; 12(1): 4386, 2021 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-34282152

RESUMO

Acute pancreatitis (AP) is serious inflammatory disease of the pancreas. Accumulating evidence links diabetes with severity of AP, suggesting that endogenous insulin may be protective. We investigated this putative protective effect of insulin during cellular and in vivo models of AP in diabetic mice (Ins2Akita) and Pancreatic Acinar cell-specific Conditional Insulin Receptor Knock Out mice (PACIRKO). Caerulein and palmitoleic acid (POA)/ethanol-induced pancreatitis was more severe in both Ins2Akita and PACIRKO vs control mice, suggesting that endogenous insulin directly protects acinar cells in vivo. In isolated pancreatic acinar cells, insulin induced Akt-mediated phosphorylation of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 2 (PFKFB2) which upregulated glycolysis thereby preventing POA-induced ATP depletion, inhibition of the ATP-dependent plasma membrane Ca2+ ATPase (PMCA) and cytotoxic Ca2+ overload. These data provide the first mechanistic link between diabetes and severity of AP and suggest that phosphorylation of PFKFB2 may represent a potential therapeutic strategy for treatment of AP.


Assuntos
Células Acinares/metabolismo , Trifosfato de Adenosina/metabolismo , Cálcio/metabolismo , Glicólise/efeitos dos fármacos , Insulina/metabolismo , Insulina/farmacologia , Pancreatite/metabolismo , Substâncias Protetoras/farmacologia , Células Acinares/efeitos dos fármacos , Doença Aguda , Animais , ATPases Transportadoras de Cálcio/metabolismo , Ceruletídeo , Diabetes Mellitus Experimental/metabolismo , Ácidos Graxos Monoinsaturados , Masculino , Camundongos , Camundongos Knockout , Pâncreas/metabolismo , Pancreatite/tratamento farmacológico , Pancreatite/patologia
10.
J Membr Biol ; 235(3): 191-210, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20544344

RESUMO

Calcium (Ca(2+))-activated K(+) (K(Ca)) channels regulate membrane excitability and are activated by an increase in cytosolic Ca(2+) concentration ([Ca(2+)](i)), leading to membrane hyperpolarization. Most patch clamp experiments that measure K(Ca) currents use steady-state [Ca(2+)] buffered within the patch pipette. However, when cells are stimulated physiologically, [Ca(2+)](i) changes dynamically, for example during [Ca(2+)](i) oscillations. Therefore, the aim of the present study was to examine the effect of dynamic changes in [Ca(2+)](i) on small (SK3), intermediate (hIK1), and large conductance (BK) channels. HEK293 cells stably expressing each K(Ca) subtype in isolation were used to simultaneously measure agonist-evoked [Ca(2+)](i) signals, using indo-1 fluorescence, and current/voltage, using perforated patch clamp. Agonist-evoked [Ca(2+)](i) oscillations induced a corresponding K(Ca) current that faithfully followed the [Ca(2+)](i) in 13-50% of cells, suggesting a good synchronization. However, [Ca(2+)](i) and K(Ca) current was much less synchronized in 50-76% of cells that exhibited Ca(2+)-independent current events (55% of SK3-, 50% of hIK1-, and 53% of BK-expressing cells) and current-independent [Ca(2+)](i) events (18% SK3- and 33% of BK-expressing cells). Moreover, in BK-expressing cells, where [Ca(2+)](i) and K(Ca) current was least synchronized, 36% of total [Ca(2+)](i) spikes occurred without activating a corresponding K(Ca) current spike, suggesting that BK(Ca) channels were either inhibited or had become desensitized. This desynchronization between dynamic [Ca(2+)](i) and K(Ca) current suggests that this relationship is more complex than could be predicted from steady-state [Ca(2+)](i) and K(Ca) current. These phenomena may be important for encoding stimulus-response coupling in various cell types.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/fisiologia , Carbacol/farmacologia , Células Cultivadas , Fura-2 , Humanos , Indóis/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Técnicas de Patch-Clamp , Canais de Potássio Cálcio-Ativados/agonistas
11.
Cancers (Basel) ; 12(9)2020 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-32825277

RESUMO

Cancer is caused by excessive cell proliferation and a propensity to avoid cell death, while the spread of cancer is facilitated by enhanced cellular migration, invasion, and vascularization. Cytosolic Ca2+ is central to each of these important processes, yet to date, there are no cancer drugs currently being used clinically, and very few undergoing clinical trials, that target the Ca2+ signalling machinery. The aim of this review is to highlight some of the emerging evidence that targeting key components of the Ca2+ signalling machinery represents a novel and relatively untapped therapeutic strategy for the treatment of cancer.

12.
Cancers (Basel) ; 12(1)2020 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-31963119

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is largely resistant to standard treatments leading to poor patient survival. The expression of plasma membrane calcium ATPase-4 (PMCA4) is reported to modulate key cancer hallmarks including cell migration, growth, and apoptotic resistance. Data-mining revealed that PMCA4 was over-expressed in pancreatic ductal adenocarcinoma (PDAC) tumors which correlated with poor patient survival. Western blot and RT-qPCR revealed that MIA PaCa-2 cells almost exclusively express PMCA4 making these a suitable cellular model of PDAC with poor patient survival. Knockdown of PMCA4 in MIA PaCa-2 cells (using siRNA) reduced cytosolic Ca2+ ([Ca2+]i) clearance, cell migration, and sensitized cells to apoptosis, without affecting cell growth. Knocking down PMCA4 had minimal effects on numerous metabolic parameters (as assessed using the Seahorse XF analyzer). In summary, this study provides the first evidence that PMCA4 is over-expressed in PDAC and plays a role in cell migration and apoptotic resistance in MIA PaCa-2 cells. This suggests that PMCA4 may offer an attractive novel therapeutic target in PDAC.

13.
Cell Calcium ; 69: 28-36, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28625348

RESUMO

The plasma membrane Ca2+-ATPase (PMCA) is a ubiquitously expressed, ATP-driven Ca2+ pump that is critical for maintaining low resting cytosolic Ca2+ ([Ca2+]i) in all eukaryotic cells. Since cytotoxic Ca2+ overload has such a central role in cell death, the PMCA represents an essential "linchpin" for the delicate balance between cell survival and cell death. In general, impaired PMCA activity and reduced PMCA expression leads to cytotoxic Ca2+ overload and Ca2+ dependent cell death, both apoptosis and necrosis, whereas maintenance of PMCA activity or PMCA overexpression is generally accepted as being cytoprotective. However, the PMCA has a paradoxical role in cell death depending on the cell type and cellular context. The PMCA can be differentially regulated by Ca2+-dependent proteolysis, can be maintained by a localised glycolytic ATP supply, even in the face of global ATP depletion, and can be profoundly affected by the specific phospholipid environment that it sits within the membrane. The major focus of this review is to highlight some of the controversies surrounding the paradoxical role of the PMCA in cell death and survival, challenging the conventional view of ATP-dependent regulation of the PMCA and how this might influence cell fate.


Assuntos
ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Apoptose , Sobrevivência Celular , Humanos , Mitocôndrias/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/química , Espécies Reativas de Oxigênio/metabolismo
14.
World J Gastroenterol ; 22(12): 3471-85, 2016 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-27022229

RESUMO

AIM: To construct a global "metabolic phenotype" of pancreatic ductal adenocarcinoma (PDAC) reflecting tumour-related metabolic enzyme expression. METHODS: A systematic review of the literature was performed using OvidSP and PubMed databases using keywords "pancreatic cancer" and individual glycolytic and mitochondrial oxidative phosphorylation (MOP) enzymes. Both human and animal studies investigating the oncological effect of enzyme expression changes and inhibitors in both an in vitro and in vivo setting were included in the review. Data reporting changes in enzyme expression and the effects on PDAC cells, such as survival and metastatic potential, were extracted to construct a metabolic phenotype. RESULTS: Seven hundred and ten papers were initially retrieved, and were screened to meet the review inclusion criteria. 107 unique articles were identified as reporting data involving glycolytic enzymes, and 28 articles involving MOP enzymes in PDAC. Data extraction followed a pre-defined protocol. There is consistent over-expression of glycolytic enzymes and lactate dehydrogenase in keeping with the Warburg effect to facilitate rapid adenosine-triphosphate production from glycolysis. Certain isoforms of these enzymes were over-expressed specifically in PDAC. Altering expression levels of HK, PGI, FBA, enolase, PK-M2 and LDA-A with metabolic inhibitors have shown a favourable effect on PDAC, thus identifying these as potential therapeutic targets. However, the Warburg effect on MOP enzymes is less clear, with different expression levels at different points in the Krebs cycle resulting in a fundamental change of metabolite levels, suggesting that other essential anabolic pathways are being stimulated. CONCLUSION: Further characterisation of the PDAC metabolic phenotype is necessary as currently there are few clinical studies and no successful clinical trials targeting metabolic enzymes.


Assuntos
Carcinoma Ductal Pancreático/enzimologia , Metabolismo Energético , Glucose/metabolismo , Neoplasias Pancreáticas/enzimologia , Animais , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Metabolismo Energético/genética , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Fenótipo
15.
Cell Calcium ; 34(6): 431-44, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14572802

RESUMO

An impressive array of cytosolic calcium ([Ca2+](i)) signals exert control over a broad range of physiological processes. The specificity and fidelity of these [Ca2+](i) signals is encoded by the frequency, amplitude, and sub-cellular localization of the response. It is believed that the distinct characteristics of [Ca2+](i) signals underlies the differential activation of effectors and ultimately cellular events. This "shaping" of [Ca2+](i) signals can be achieved by the influence of additional signaling pathways modulating the molecular machinery responsible for generating [Ca2+](i) signals. There is a particularly rich source of potential sites of crosstalk between the cAMP and the [Ca2+](i) signaling pathways. This review will focus on the predominant molecular loci at which these classical signaling systems interact to impact the spatio-temporal pattern of [Ca2+](i) signaling in non-excitable cells.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , AMP Cíclico/metabolismo , Pâncreas/metabolismo , Sistemas do Segundo Mensageiro , Animais , Canais de Cálcio/metabolismo , Citosol/metabolismo , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Modelos Biológicos , Pâncreas/citologia , Receptores de Superfície Celular/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
16.
Am J Physiol Cell Physiol ; 295(5): C1247-60, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18787078

RESUMO

Impairment of the normal spatiotemporal pattern of intracellular Ca(2+) ([Ca(2+)](i)) signaling, and in particular, the transition to an irreversible "Ca(2+) overload" response, has been implicated in various pathophysiological states. In some diseases, including pancreatitis, oxidative stress has been suggested to mediate this Ca(2+) overload and the associated cell injury. We have previously demonstrated that oxidative stress with hydrogen peroxide (H(2)O(2)) evokes a Ca(2+) overload response and inhibition of plasma membrane Ca(2+)-ATPase (PMCA) in rat pancreatic acinar cells (Bruce JI and Elliott AC. Am J Physiol Cell Physiol 293: C938-C950, 2007). The aim of the present study was to further examine this oxidant-impaired inhibition of the PMCA, focusing on the role of the mitochondria. Using a [Ca(2+)](i) clearance assay in which mitochondrial Ca(2+) uptake was blocked with Ru-360, H(2)O(2) (50 microM-1 mM) markedly inhibited the PMCA activity. This H(2)O(2)-induced inhibition of the PMCA correlated with mitochondrial depolarization (assessed using tetramethylrhodamine methylester fluorescence) but could occur without significant ATP depletion (assessed using Magnesium Green fluorescence). The H(2)O(2)-induced PMCA inhibition was sensitive to the mitochondrial permeability transition pore (mPTP) inhibitors, cyclosporin-A and bongkrekic acid. These data suggest that oxidant-induced opening of the mPTP and mitochondrial depolarization may lead to an inhibition of the PMCA that is independent of mitochondrial Ca(2+) handling and ATP depletion, and we speculate that this may involve the release of a mitochondrial factor. Such a phenomenon may be responsible for the Ca(2+) overload response, and for the transition between apoptotic and necrotic cell death thought to be important in many disease states.


Assuntos
Sinalização do Cálcio , Mitocôndrias/metabolismo , Estresse Oxidativo , Pâncreas Exócrino/enzimologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Antimicina A/farmacologia , Bioensaio , Ácido Bongcréquico/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Ciclosporina/farmacologia , Peróxido de Hidrogênio/metabolismo , Potencial da Membrana Mitocondrial , Mitocôndrias/efeitos dos fármacos , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Oligomicinas/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Pâncreas Exócrino/efeitos dos fármacos , ATPases Transportadoras de Cálcio da Membrana Plasmática/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Compostos de Rutênio/farmacologia , Fatores de Tempo , Desacopladores/farmacologia
17.
Am J Physiol Cell Physiol ; 293(3): C938-50, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17494627

RESUMO

Pancreatitis is an inflammatory disease of pancreatic acinar cells whereby intracellular calcium concentration ([Ca(2+)](i)) signaling and enzyme secretion are impaired. Increased oxidative stress has been suggested to mediate the associated cell injury. The present study tested the effects of the oxidant, hydrogen peroxide, on [Ca(2+)](i) signaling in rat pancreatic acinar cells by simultaneously imaging fura-2, to measure [Ca(2+)](i), and dichlorofluorescein, to measure oxidative stress. Millimolar concentrations of hydrogen peroxide increased cellular oxidative stress and irreversibly increased [Ca(2+)](i), which was sensitive to antioxidants and removal of external Ca(2+), and ultimately led to cell lysis. Responses were also abolished by pretreatment with (sarco)endoplasmic reticulum Ca(2+)-ATPase inhibitors, unless cells were prestimulated with cholecystokinin to promote mitochondrial Ca(2+) uptake. This suggests that hydrogen peroxide promotes Ca(2+) release from the endoplasmic reticulum and the mitochondria and that it promotes Ca(2+) influx. Lower concentrations of hydrogen peroxide (10-100 muM) increased [Ca(2+)](i) and altered cholecystokinin-evoked [Ca(2+)](i) oscillations with marked heterogeneity, the severity of which was directly related to oxidative stress, suggesting differences in cellular antioxidant capacity. These changes in [Ca(2+)](i) also upregulated the activity of the plasma membrane Ca(2+)-ATPase in a Ca(2+)-dependent manner, whereas higher concentrations (0.1-1 mM) inactivated the plasma membrane Ca(2+)-ATPase. This may be important in facilitating "Ca(2+) overload," resulting in cell injury associated with pancreatitis.


Assuntos
Sinalização do Cálcio/fisiologia , Peróxido de Hidrogênio/farmacologia , Oxidantes/farmacologia , Pâncreas Exócrino/enzimologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Animais , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Membrana Celular/enzimologia , Colagogos e Coleréticos/farmacologia , Colecistocinina/farmacologia , Fluoresceínas , Técnicas In Vitro , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Pâncreas Exócrino/citologia , Pancreatite/metabolismo , Ratos
18.
J Biol Chem ; 282(52): 37678-93, 2007 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-17938178

RESUMO

Cross-talk between intracellular calcium ([Ca(2+)](i)) signaling and cAMP defines the specificity of stimulus-response coupling in a variety of cells. Previous studies showed that protein kinase A (PKA) potentiates and phosphorylates the plasma membrane Ca(2+)-ATPase (PMCA) in a Ca(2+)-dependent manner in parotid acinar cells (Bruce, J. I. E., Yule, D. I., and Shuttleworth, T. J. (2002) J. Biol. Chem. 277, 48172-48181). The aim of this study was to further investigate the spatial regulation of [Ca(2+)](i) clearance in parotid acinar cells. Par-C10 cells were used to functionally isolate the apical and basolateral PMCA activity by applying La(3+) to the opposite side to inhibit the PMCA. Activation of PKA (using forskolin) differentially potentiated apical [Ca(2+)](i) clearance in mouse parotid acinar cells and apical PMCA activity in Par-C10 cells. Immunofluorescence of parotid tissue slices revealed that PMCA1 was distributed throughout the plasma membrane, PMCA2 was localized to the basolateral membrane, and PMCA4 was localized to the apical membrane of parotid acinar cells. However, in situ phosphorylation assays demonstrated that PMCA1 was the only isoform phosphorylated by PKA following stimulation. Similarly, immunofluorescence of acutely isolated parotid acinar cells showed that the regulatory subunit of PKA (RIIbeta) translocated to the apical region following stimulation. These data suggest that PKA-mediated phosphorylation of PMCA1 differentially regulates [Ca(2+)](i) clearance in the apical region of parotid acinar cells because of a dynamic translocation of PKA. Such tight spatial regulation of Ca(2+) efflux is likely important for the fine-tuning of Ca(2+)-dependent effectors close to the apical membrane important for the regulation of fluid secretion and exocytosis.


Assuntos
ATPases Transportadoras de Cálcio/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulação da Expressão Gênica , Glândula Parótida/citologia , Glândula Parótida/metabolismo , Animais , Cálcio/metabolismo , Membrana Celular/metabolismo , Colforsina/metabolismo , Cinética , Camundongos , Modelos Biológicos , Fosforilação , Transporte Proteico , Ratos , Transdução de Sinais
19.
J Biol Chem ; 277(50): 48172-81, 2002 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-12368283

RESUMO

Cross-talk between cAMP and [Ca(2+)](i) signaling pathways represents a general feature that defines the specificity of stimulus-response coupling in a variety of cell types including parotid acinar cells. We have reported recently that cAMP potentiates Ca(2+) release from intracellular stores, primarily because of a protein kinase A-mediated phosphorylation of type II inositol 1,4,5-trisphosphate receptors (Bruce, J. I. E., Shuttleworth, T. J. S., Giovannucci, D. R., and Yule, D. I. (2002) J. Biol. Chem. 277, 1340-1348). The aim of the present study was to evaluate the functional and molecular mechanism whereby cAMP regulates Ca(2+) clearance pathways in parotid acinar cells. Following an agonist-induced increase in [Ca(2+)](i) the rate of Ca(2+) clearance, after the removal of the stimulus, was potentiated substantially ( approximately 2-fold) by treatment with forskolin. This effect was prevented completely by inhibition of the plasma membrane Ca(2+)-ATPase (PMCA) with La(3+). PMCA activity, when isolated pharmacologically, was also potentiated ( approximately 2-fold) by forskolin. Ca(2+) uptake into the endoplasmic reticulum of streptolysin-O-permeabilized cells by sarco/endoplasmic reticulum Ca(2+)-ATPase was largely unaffected by treatment with dibutyryl cAMP. Finally, in situ phosphorylation assays demonstrated that PMCA was phosphorylated by treatment with forskolin but only in the presence of carbamylcholine (carbachol). This effect of forskolin was Ca(2+)-dependent, and protein kinase C-independent, as potentiation of PMCA activity and phosphorylation of PMCA by forskolin also occurred when [Ca(2+)](i) was elevated by the sarco/endoplasmic reticulum Ca(2+)-ATPase inhibitor cyclopiazonic acid and was attenuated by pre-incubation with the Ca(2+) chelator, 1,2-bis(o-aminophenoxy) ethane-N,N,N',N'-tetraacetic acid (BAPTA). The present study demonstrates that elevated cAMP enhances the rate of Ca(2+) clearance because of a complex modulation of PMCA activity that involves a Ca(2+)-dependent step. Tight regulation of both Ca(2+) release and Ca(2+) efflux may represent a general feature of the mechanism whereby cAMP improves the fidelity and specificity of Ca(2+) signaling.


Assuntos
ATPases Transportadoras de Cálcio/metabolismo , Cálcio/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Glândula Parótida/enzimologia , Bucladesina/farmacologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/enzimologia , Colforsina/farmacologia , Proteína Quinase Tipo II Dependente de AMP Cíclico , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Glândula Parótida/citologia , Glândula Parótida/efeitos dos fármacos , Fosforilação
20.
J Biol Chem ; 277(35): 31949-56, 2002 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-12065595

RESUMO

Stimulation of pancreatic acinar cells with acetylcholine (ACh) and cholecystokinin (CCK) results in an elevation of cytosolic calcium ([Ca(2+)](c)) through activation of inositol 1,4,5-trisphosphate receptors (InsP(3)R). The global temporal pattern of the [Ca(2+)](c) changes produced by ACh or CCK stimulation differs significantly. The hypothesis was tested that CCK stimulation results in a protein kinase A (PKA)-mediated phosphorylation of InsP(3)R and this event contributes to the generation of agonist-specific [Ca(2+)](c) signals. Physiological concentrations of CCK evoked phosphorylation of the type III InsP(3)R, which was blocked by pharmacological inhibition of PKA. Imaging of fura-2-loaded acinar cells revealed that the rate of [Ca(2+)](c) rise during CCK-evoked oscillations slows with each subsequent oscillation, consistent with a developing modulation of release, whereas the kinetics of ACh-evoked oscillations remain constant. Stimulation of cells with ACh following activation of PKA resulted in a slowing of the ACh-evoked [Ca(2+)](c) rise, which now resembled a time-matched CCK response. PKA activation also resulted in a slowing of [Ca(2+)](c) increases elicited by photolysis of caged InsP(3). Targeted, PKA-mediated phosphorylation of type III InsP(3)R is involved in a physiological CCK response, as disruption of the targeting of PKA with the peptide HT31 resulted in marked changes in the CCK-evoked [Ca(2+)](c) signal but had no effect on ACh-evoked responses. Stimulation of cells with bombesin, which evokes [Ca(2+)](c) oscillations indistinguishable from those produced by CCK, also results in PKA-mediated phosphorylation of type III InsP(3)R. Thus, we conclude that PKA-mediated phosphorylation of type III InsP(3)R is a general mechanism by which the patterns of [Ca(2+)](c) oscillations are shaped in pancreatic acinar cells.


Assuntos
Acetilcolina/farmacologia , Canais de Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Colecistocinina/farmacologia , Pâncreas/fisiologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Bucladesina/farmacologia , Canais de Cálcio/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Colforsina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Citosol/fisiologia , Técnicas In Vitro , Receptores de Inositol 1,4,5-Trifosfato , Cinética , Masculino , Camundongos , Oscilometria , Pâncreas/efeitos dos fármacos , Técnicas de Patch-Clamp , Fosforilação , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa