Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Physiol ; 593(16): 3447-62, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25809592

ABSTRACT

Calcium signalling is fundamental to the function of the nervous system, in association with changes in ionic gradients across the membrane. Although restoring ionic gradients is energetically costly, a rise in intracellular Ca(2+) acts through multiple pathways to increase ATP synthesis, matching energy supply to demand. Increasing cytosolic Ca(2+) stimulates metabolite transfer across the inner mitochondrial membrane through activation of Ca(2+) -regulated mitochondrial carriers, whereas an increase in matrix Ca(2+) stimulates the citric acid cycle and ATP synthase. The aspartate-glutamate exchanger Aralar/AGC1 (Slc25a12), a component of the malate-aspartate shuttle (MAS), is stimulated by modest increases in cytosolic Ca(2+) and upregulates respiration in cortical neurons by enhancing pyruvate supply into mitochondria. Failure to increase respiration in response to small (carbachol) and moderate (K(+) -depolarization) workloads and blunted stimulation of respiration in response to high workloads (veratridine) in Aralar/AGC1 knockout neurons reflect impaired MAS activity and limited mitochondrial pyruvate supply. In response to large workloads (veratridine), acute stimulation of respiration occurs in the absence of MAS through Ca(2+) influx through the mitochondrial calcium uniporter (MCU) and a rise in matrix [Ca(2+) ]. Although the physiological importance of the MCU complex in work-induced stimulation of respiration of CNS neurons is not yet clarified, abnormal mitochondrial Ca(2+) signalling causes pathology. Indeed, loss of function mutations in MICU1, a regulator of MCU complex, are associated with neuromuscular disease. In patient-derived MICU1 deficient fibroblasts, resting matrix Ca(2+) is increased and mitochondria fragmented. Thus, the fine tuning of Ca(2+) signals plays a key role in shaping mitochondrial bioenergetics.


Subject(s)
Calcium Signaling , Mitochondria/metabolism , Neurons/metabolism , Adenosine Triphosphate/metabolism , Animals , Calcium Channels/metabolism , Humans
2.
Cell Death Dis ; 5: e1257, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24874729

ABSTRACT

The platinum derivative cis-diamminedichloroplatinum(II), best known as cisplatin, is currently employed for the clinical management of patients affected by testicular, ovarian, head and neck, colorectal, bladder and lung cancers. For a long time, the antineoplastic effects of cisplatin have been fully ascribed to its ability to generate unrepairable DNA lesions, hence inducing either a permanent proliferative arrest known as cellular senescence or the mitochondrial pathway of apoptosis. Accumulating evidence now suggests that the cytostatic and cytotoxic activity of cisplatin involves both a nuclear and a cytoplasmic component. Despite the unresolved issues regarding its mechanism of action, the administration of cisplatin is generally associated with high rates of clinical responses. However, in the vast majority of cases, malignant cells exposed to cisplatin activate a multipronged adaptive response that renders them less susceptible to the antiproliferative and cytotoxic effects of the drug, and eventually resume proliferation. Thus, a large fraction of cisplatin-treated patients is destined to experience therapeutic failure and tumor recurrence. Throughout the last four decades great efforts have been devoted to the characterization of the molecular mechanisms whereby neoplastic cells progressively lose their sensitivity to cisplatin. The advent of high-content and high-throughput screening technologies has accelerated the discovery of cell-intrinsic and cell-extrinsic pathways that may be targeted to prevent or reverse cisplatin resistance in cancer patients. Still, the multifactorial and redundant nature of this phenomenon poses a significant barrier against the identification of effective chemosensitization strategies. Here, we discuss recent systems biology studies aimed at deconvoluting the complex circuitries that underpin cisplatin resistance, and how their findings might drive the development of rational approaches to tackle this clinically relevant problem.


Subject(s)
Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Systems Biology , Animals , Humans , Systems Biology/methods , Systems Biology/trends
3.
Cell Death Dis ; 5: e1082, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24577080

ABSTRACT

Novel therapeutic targets are required to protect the heart against cell death from acute ischemia-reperfusion injury (IRI). Mutations in the DJ-1 (PARK7) gene in dopaminergic neurons induce mitochondrial dysfunction and a genetic form of Parkinson's disease. Genetic ablation of DJ-1 renders the brain more susceptible to cell death following ischemia-reperfusion in a model of stroke. Although DJ-1 is present in the heart, its role there is currently unclear. We sought to investigate whether mitochondrial DJ-1 may protect the heart against cell death from acute IRI by preventing mitochondrial dysfunction. Overexpression of DJ-1 in HL-1 cardiac cells conferred the following beneficial effects: reduced cell death following simulated IRI (30.4±4.7% with DJ-1 versus 52.9±4.7% in control; n=5, P<0.05); delayed mitochondrial permeability transition pore (MPTP) opening (a critical mediator of cell death) (260±33 s with DJ-1 versus 121±12 s in control; n=6, P<0.05); and induction of mitochondrial elongation (81.3±2.5% with DJ-1 versus 62.0±2.8% in control; n=6 cells, P<0.05). These beneficial effects of DJ-1 were absent in cells expressing the non-functional DJ-1(L166P) and DJ-1(Cys106A) mutants. Adult mice devoid of DJ-1 (KO) were found to be more susceptible to cell death from in vivo IRI with larger myocardial infarct sizes (50.9±3.5% DJ-1 KO versus 41.1±2.5% in DJ-1 WT; n≥7, P<0.05) and resistant to cardioprotection by ischemic preconditioning. DJ-1 KO hearts showed increased mitochondrial fragmentation on electron microscopy, although there were no differences in calcium-induced MPTP opening, mitochondrial respiratory function or myocardial ATP levels. We demonstrate that loss of DJ-1 protects the heart from acute IRI cell death by preventing mitochondrial dysfunction. We propose that DJ-1 may represent a novel therapeutic target for cardioprotection.


Subject(s)
Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Oncogene Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Cell Death , Cell Line , Disease Models, Animal , Ischemic Preconditioning, Myocardial , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/ultrastructure , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Peroxiredoxins , Protein Deglycase DJ-1 , Transfection
4.
Cell Death Differ ; 21(5): 685-95, 2014 May.
Article in English | MEDLINE | ID: mdl-24464223

ABSTRACT

Intrinsic and acquired chemoresistance are frequent causes of cancer eradication failure. Thus, long-term cis-diaminedichloroplatine(II) (CDDP) or cisplatin treatment is known to promote tumor cell resistance to apoptosis induction via multiple mechanisms involving gene expression modulation of oncogenes, tumor suppressors and blockade of pro-apoptotic mitochondrial membrane permeabilization. Here, we demonstrate that CDDP-resistant non-small lung cancer cells undergo profound remodeling of their endoplasmic reticulum (ER) proteome (>80 proteins identified by proteomics) and exhibit a dramatic overexpression of two protein disulfide isomerases, PDIA4 and PDIA6, without any alteration in ER-cytosol Ca(2+) fluxes. Using pharmacological and genetic inhibition, we show that inactivation of both proteins directly stimulates CDDP-induced cell death by different cellular signaling pathways. PDIA4 inactivation restores a classical mitochondrial apoptosis pathway, while knockdown of PDIA6 favors a non-canonical cell death pathway sharing some necroptosis features. Overexpression of both proteins has also been found in lung adenocarcinoma patients, suggesting a clinical importance of these proteins in chemoresistance.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Protein Disulfide-Isomerases/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Apoptosis/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Lung Neoplasms/pathology
5.
Oncogene ; 32(20): 2592-600, 2013 May 16.
Article in English | MEDLINE | ID: mdl-22777349

ABSTRACT

Primary mitochondrial dysfunction commonly leads to failure in cellular adaptation to stress. Paradoxically, however, nonsynonymous mutations of mitochondrial DNA (mtDNA) are frequently found in cancer cells and may have a causal role in the development of resistance to genotoxic stress induced by common chemotherapeutic agents, such as cis-diammine-dichloroplatinum(II) (cisplatin, CDDP). Little is known about how these mutations arise and the associated mechanisms leading to chemoresistance. Here, we show that the development of adaptive chemoresistance in the A549 non-small-cell lung cancer cell line to CDDP is associated with the hetero- to homoplasmic shift of a nonsynonymous mutation in MT-ND2, encoding the mitochondrial Complex-I subunit ND2. The mutation resulted in a 50% reduction of the NADH:ubiquinone oxidoreductase activity of the complex, which was compensated by increased biogenesis of respiratory chain complexes. The compensatory mitochondrial biogenesis was most likely mediated by the nuclear co-activators peroxisome proliferator-activated receptor gamma co-activator-1α (PGC-1α) and PGC-1ß, both of which were significantly upregulated in the CDDP-resistant cells. Importantly, both transient and stable silencing of PGC-1ß re-established the sensitivity of these cells to CDDP-induced apoptosis. Remarkably, the PGC-1ß-mediated CDDP resistance was independent of the mitochondrial effects of the co-activator. Altogether, our results suggest that partial respiratory chain defects because of mtDNA mutations can lead to compensatory upregulation of nuclear transcriptional co-regulators, in turn mediating resistance to genotoxic stress.


Subject(s)
Carrier Proteins/metabolism , DNA, Mitochondrial , Drug Resistance, Neoplasm/genetics , Mutation , Adaptation, Physiological , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , NADH Dehydrogenase/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA-Binding Proteins , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Cell Death Differ ; 19(4): 650-60, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22015608

ABSTRACT

Ca(2+)-mediated mitochondrial permeability transition (mPT) is the final common pathway of stress-induced cell death in many major pathologies, but its regulation in intact cells is poorly understood. Here we report that the mitochondrial carrier SCaMC-1/SLC25A24 mediates ATP-Mg(2-)/Pi(2-) and/or HADP(2-)/Pi(2-) uptake into the mitochondria after an increase in cytosolic [Ca(2+)]. ATP and ADP contribute to Ca(2+) buffering in the mitochondrial matrix, resulting in desensitization of the mPT. Comprehensive gene expression analysis showed that SCaMC-1 overexpression is a general feature of transformed and cancer cells. Knockdown of the transporter led to vast reduction of mitochondrial Ca(2+) buffering capacity and sensitized cells to mPT-mediated necrotic death triggered by oxidative stress and Ca(2+) overload. These findings revealed that SCaMC-1 exerts a negative feedback control between cellular Ca(2+) overload and mPT-dependent cell death, suggesting that the carrier might represent a novel target for cancer therapy.


Subject(s)
Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Calcium-Binding Proteins/metabolism , Calcium/metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Adenosine Diphosphate/genetics , Adenosine Triphosphate/genetics , Animals , COS Cells , Calcium-Binding Proteins/genetics , Cell Line, Tumor , Cell Survival , Chlorocebus aethiops , Mice , Mitochondria/genetics , Mitochondria/pathology , Necrosis , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Oxidative Stress/genetics , Permeability
7.
Aliment Pharmacol Ther ; 34(8): 840-52, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21851372

ABSTRACT

BACKGROUND: In addition to the circulating (endocrine) renin-angiotensin system (RAS), local renin-angiotensin systems are now known to exist in diverse cells and tissues. Amongst these, pancreatic renin-angiotensin systems have recently been identified and may play roles in the physiological regulation of pancreatic function, as well as being implicated in the pathogenesis of pancreatic diseases including diabetes, pancreatitis and pancreatic cancer. AIM: To review and summarise current knowledge of pancreatic renin-angiotensin systems. METHODS: We performed an extensive PubMed, Medline and online review of all relevant literature. RESULTS: Pancreatic RAS appear to play various roles in the regulation of pancreatic physiology and pathophysiology. Ang II may play a role in the development of pancreatic ductal adenocarcinoma, via stimulation of angiogenesis and prevention of chemotherapy toxicity, as well as in the initiation and propagation of acute pancreatitis (AP); whereas, RAS antagonism is capable of preventing new-onset diabetes and improving glycaemic control in diabetic patients. Current evidence for the roles of pancreatic RAS is largely based upon cell and animal models, whilst definitive evidence from human studies remains lacking. CONCLUSIONS: The therapeutic potential for RAS antagonism, using cheap and widely available agents, and may be untapped and such roles are worthy of active investigation in diverse pancreatic disease states.


Subject(s)
Carcinoma, Pancreatic Ductal/physiopathology , Diabetes Mellitus/physiopathology , Pancreatic Neoplasms/physiopathology , Pancreatitis/physiopathology , Renin-Angiotensin System/physiology , Angiotensins/physiology , Humans , Receptors, Angiotensin/metabolism
8.
Mini Rev Med Chem ; 11(5): 390-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21443511

ABSTRACT

Increasing evidence suggests that nongenomic effects of testosterone and anabolic androgenic steroids (AAS) operate concertedly with genomic effects. Classically, these responses have been viewed as separate and independent processes, primarily because nongenomic responses are faster and appear to be mediated by membrane androgen receptors, whereas long-term genomic effects are mediated through cytosolic androgen receptors regulating transcriptional activity. Numerous studies have demonstrated increases in intracellular Ca2+ in response to AAS. These Ca2+ mediated responses have been seen in a diversity of cell types, including osteoblasts, platelets, skeletal muscle cells, cardiac myocytes and neurons. The versatility of Ca2+ as a second messenger provides these responses with a vast number of pathophysiological implications. In cardiac cells, testosterone elicits voltage-dependent Ca2+ oscillations and IP3R-mediated Ca2+ release from internal stores, leading to activation of MAPK and mTOR signaling that promotes cardiac hypertrophy. In neurons, depending upon concentration, testosterone can provoke either physiological Ca2+ oscillations, essential for synaptic plasticity, or sustained, pathological Ca2+ transients that lead to neuronal apoptosis. We propose therefore, that Ca2+ acts as an important point of crosstalk between nongenomic and genomic AAS signaling, representing a central regulator that bridges these previously thought to be divergent responses.


Subject(s)
Anabolic Agents/pharmacology , Androgens/pharmacology , Calcium Signaling/drug effects , Cardiomegaly , Signal Transduction/drug effects , Steroids/pharmacology , Anabolic Agents/adverse effects , Androgens/adverse effects , Cardiomegaly/chemically induced , Humans , Steroids/adverse effects
9.
Cell Death Differ ; 16(7): 1006-17, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19325567

ABSTRACT

The inositol 1,4,5-trisphosphate receptor (IP(3)R) is a major regulator of apoptotic signaling. Through interactions with members of the Bcl-2 family of proteins, it drives calcium (Ca(2+)) transients from the endoplasmic reticulum (ER) to mitochondria, thereby establishing a functional and physical link between these organelles. Importantly, the IP(3)R also regulates autophagy, and in particular, its inhibition/depletion strongly induces macroautophagy. Here, we show that the IP(3)R antagonist xestospongin B induces autophagy by disrupting a molecular complex formed by the IP(3)R and Beclin 1, an interaction that is increased or inhibited by overexpression or knockdown of Bcl-2, respectively. An effect of Beclin 1 on Ca(2+) homeostasis was discarded as siRNA-mediated knockdown of Beclin 1 did not affect cytosolic or luminal ER Ca(2+) levels. Xestospongin B- or starvation-induced autophagy was inhibited by overexpression of the IP(3)R ligand-binding domain, which coimmunoprecipitated with Beclin 1. These results identify IP(3)R as a new regulator of the Beclin 1 complex that may bridge signals converging on the ER and initial phagophore formation.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy/physiology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Autophagy/drug effects , Beclin-1 , Calcium/metabolism , Cell Line , Cell Line, Tumor , Gene Knockdown Techniques , HeLa Cells , Humans , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Macrocyclic Compounds/pharmacology , Membrane Proteins/genetics , Oxazoles/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Small Interfering/metabolism , Rats
10.
Cell Prolif ; 41(3): 554-73, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18422703

ABSTRACT

OBJECTIVES: Polycystin-1 (PC1), a signalling receptor regulating Ca(2+)-permeable cation channels, is mutated in autosomal dominant polycystic kidney disease, which is typically characterized by increased cell proliferation. However, the precise mechanisms by which PC1 functions on Ca(2+) homeostasis, signalling and cell proliferation remain unclear. Here, we investigated the possible role of PC1 as a modulator of non-capacitative Ca(2+) entry (NCCE) and Ca(2+) oscillations, with downstream effects on cell proliferation. RESULTS AND DISCUSSION: By employing RNA interference, we show that depletion of endogenous PC1 in HEK293 cells leads to an increase in serum-induced Ca(2+) oscillations, triggering nuclear factor of activated T cell activation and leading to cell cycle progression. Consistently, Ca(2+) oscillations and cell proliferation are increased in PC1-mutated kidney cystic cell lines, but both abnormal features are reduced in cells that exogenously express PC1. Notably, blockers of the NCCE pathway, but not of the CCE, blunt abnormal oscillation and cell proliferation. Our study therefore provides the first demonstration that PC1 modulates Ca(2+) oscillations and a molecular mechanism to explain the association between abnormal Ca(2+) homeostasis and cell proliferation in autosomal dominant polycystic kidney disease.


Subject(s)
Calcium Signaling , Kidney/pathology , TRPP Cation Channels/metabolism , Cell Line , Cell Line, Transformed , Cell Proliferation , Codon, Nonsense/genetics , Cytoplasm/metabolism , Enzyme Activation , Humans , Kidney/enzymology , Models, Biological , NFATC Transcription Factors/metabolism , Polycystic Kidney, Autosomal Dominant/enzymology , Polycystic Kidney, Autosomal Dominant/pathology , Protein Kinase C-alpha/metabolism , RNA Interference
11.
Cell Death Differ ; 15(2): 274-82, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18034188

ABSTRACT

Some chemotherapeutic agents can elicit apoptotic cancer cell death, thereby activating an anticancer immune response that influences therapeutic outcome. We previously reported that anthracyclins are particularly efficient in inducing immunogenic cell death, correlating with the pre-apoptotic exposure of calreticulin (CRT) on the plasma membrane surface of anthracyclin-treated tumor cells. Here, we investigated the role of cellular Ca(2+) homeostasis on CRT exposure. A neuroblastoma cell line (SH-SY5Y) failed to expose CRT in response to anthracyclin treatment. This defect in CRT exposure could be overcome by the overexpression of Reticulon-1C, a manipulation that led to a decrease in the Ca(2+) concentration within the endoplasmic reticulum lumen. The combination of Reticulon-1C expression and anthracyclin treatment yielded more pronounced endoplasmic reticulum Ca(2+) depletion than either of the two manipulations alone. Chelation of intracellular (and endoplasmic reticulum) Ca(2+), targeted expression of the ligand-binding domain of the IP(3) receptor and inhibition of the sarco-endoplasmic reticulum Ca(2+)-ATPase pump reduced endoplasmic reticulum Ca(2+) load and promoted pre-apoptotic CRT exposure on the cell surface, in SH-SY5Y and HeLa cells. These results provide evidence that endoplasmic reticulum Ca(2+) levels control the exposure of CRT.


Subject(s)
Anthracyclines/pharmacology , Calcium/metabolism , Calreticulin/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Nerve Tissue Proteins/metabolism , Apoptosis , Brefeldin A/pharmacology , Cell Line, Tumor , HeLa Cells , Homeostasis , Humans , Protein Synthesis Inhibitors/pharmacology
12.
Cell Death Differ ; 14(5): 1029-39, 2007 May.
Article in English | MEDLINE | ID: mdl-17256008

ABSTRACT

The reduction of intracellular 1,4,5-inositol trisphosphate (IP(3)) levels stimulates autophagy, whereas the enhancement of IP(3) levels inhibits autophagy induced by nutrient depletion. Here, we show that knockdown of the IP(3) receptor (IP(3)R) with small interfering RNAs and pharmacological IP(3)R blockade is a strong stimulus for the induction of autophagy. The IP(3)R is known to reside in the membranes of the endoplasmic reticulum (ER) as well as within ER-mitochondrial contact sites, and IP(3)R blockade triggered the autophagy of both ER and mitochondria, as exactly observed in starvation-induced autophagy. ER stressors such as tunicamycin and thapsigargin also induced autophagy of ER and, to less extent, of mitochondria. Autophagy triggered by starvation or IP(3)R blockade was inhibited by Bcl-2 and Bcl-X(L) specifically targeted to ER but not Bcl-2 or Bcl-X(L) proteins targeted to mitochondria. In contrast, ER stress-induced autophagy was not inhibited by Bcl-2 and Bcl-X(L). Autophagy promoted by IP(3)R inhibition could not be attributed to a modulation of steady-state Ca(2+) levels in the ER or in the cytosol, yet involved the obligate contribution of Beclin-1, autophagy-related gene (Atg)5, Atg10, Atg12 and hVps34. Altogether, these results strongly suggest that IP(3)R exerts a major role in the physiological control of autophagy.


Subject(s)
Autophagy , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Animals , Autophagy/genetics , Calcium/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Food Deprivation , HeLa Cells , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Macrocyclic Compounds/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Oxazoles/pharmacology , Protein Isoforms/metabolism , Rats , bcl-X Protein/metabolism
13.
Biochim Biophys Acta ; 1763(5-6): 442-9, 2006.
Article in English | MEDLINE | ID: mdl-16750865

ABSTRACT

Recent data shed light on two novel aspects of the mitochondria-Ca2+ liaison. First, it was extensively investigated how Ca2+ handling is controlled by mitochondrial shape, and positioning; a playground also of cell death and survival regulation. On the other hand, significant progress has been made to explore how intra- and near-mitochondrial Ca2+ signals modify mitochondrial morphology and cellular distribution. Here, we shortly summarize these advances and provide a model of Ca2+-mitochondria interactions.


Subject(s)
Calcium Signaling , Mitochondria/metabolism , Animals , Biological Evolution , Endoplasmic Reticulum/metabolism , Humans
15.
Cell Death Differ ; 13(4): 586-96, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16239931

ABSTRACT

Mitochondrial Ca2+ uptake controls cellular functions as diverse as aerobic metabolism, cytosolic Ca2+signalling and mitochondrial participation in apoptosis. Modulatory inputs converging on the organelle can regulate this process, determining the final outcome of Ca2+-mediated cell stimulation. We investigated in HeLa cells and primary skeletal myotubes the effect on Ca2+ signalling of the transcriptional peroxisome-proliferator-activated-receptor-gamma-coactivator-1alpha (PGC-1alpha), which triggers organelle biogenesis and modifies the mitochondrial proteome. PGC-1alpha selectively reduced mitochondrial Ca2+ responses to cell stimulation by reducing the efficacy of mitochondrial Ca2+ uptake sites and increasing organelle volume. In turn, this affected ER Ca2+ release and cytosolic responses in HeLa cells. Most importantly, the modulation of mitochondrial Ca2+ uptake significantly reduced cellular sensitivity to the Ca2+-mediated proapoptotic effect of C2 ceramide. These results reveal a primary role of PGC-1alpha in shaping mitochondrial participation in calcium signalling, that underlies its protective role against stress and proapoptotic stimuli in pathophysiological conditions.


Subject(s)
Apoptosis , Calcium Signaling , Calcium/metabolism , Heat-Shock Proteins/metabolism , Mitochondria, Muscle/metabolism , Muscle Fibers, Skeletal/metabolism , Transcription Factors/metabolism , Adrenergic beta-Agonists/pharmacology , Albuterol/pharmacology , Animals , Calcium Channels/metabolism , Enzyme Inhibitors/pharmacology , HeLa Cells , Heat-Shock Proteins/genetics , Histamine/pharmacology , Homeostasis , Humans , Inositol 1,4,5-Trisphosphate Receptors , Ion Channels , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mitochondria, Muscle/drug effects , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Swelling , Muscle Fibers, Skeletal/pathology , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Receptors, Cytoplasmic and Nuclear/metabolism , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Transcription Factors/genetics , Transfection , Uncoupling Protein 2
16.
Cell Calcium ; 32(5-6): 413-20, 2002.
Article in English | MEDLINE | ID: mdl-12543100

ABSTRACT

In the complex signalling interplay that allows extracellular signals to be decoded into activation of apoptotic cell death, Ca(2+) plays a significant role. This is supported not only by evidence linking alterations in Ca(2+) homeostasis to the triggering of apoptotic (and in some cases necrotic) cell death, but also by recent data indicating that a key anti-apoptotic protein, Bcl-2, has a direct effect on ER Ca(2+) handling. We will briefly summarise the first aspect, and describe in more detail these new data, demonstrating that (i) Bcl-2 reduces the state of filling of the ER Ca(2+) store and (ii) this Ca(2+) signalling alteration renders the cells less sensitive to apoptotic stimuli. Overall, these results suggest that calcium homeostasis may represent a pharmacological target in the fundamental pathological process of apoptosis.


Subject(s)
Apoptosis/physiology , Calcium/physiology , Endoplasmic Reticulum/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Animals , Apoptosis/drug effects , Endoplasmic Reticulum/drug effects , Humans
17.
News Physiol Sci ; 16: 197-200, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11572920

ABSTRACT

Ca(2+) signal in high-Ca(2+) perimitochondrial microdomains is amplified within the mitochondrial matrix and activates Ca(2+)-dependent dehydrogenases. In steroid-secreting cells, small cytoplasmic Ca(2+) signals may also augment mitochondrial Ca(2+) concentration. The ensuing formation of NADH and NADPH may have an essential role in supporting the increased steroid secretion.


Subject(s)
Calcium Signaling/physiology , Mitochondria/metabolism , Steroids/metabolism , Animals
18.
Pflugers Arch ; 441(5): 678-85, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11294250

ABSTRACT

The cytoplasmic Ca2+ signal is transferred to the mitochondrial matrix and activates mitochondrial dehydrogenases. The requirement for supramicromolar cytoplasmic [Ca2+] ([Ca2+]i) in perimitochondrial microdomains in this response has been suggested. We studied the correlation between [Ca2+]i, mitochondrial [Ca2+] ([Ca2+]m) and mitochondrial formation of reduced nicotinamide adenine dinucleotide (phosphate) [NAD(P)H] in the presence of submicromolar [Ca2+]i in cultured rat "large" luteal cells. [Ca2+]i was monitored fluorimetrically with fura-PE3, [Ca2+]m with rhod-2 and NAD(P)H with autofluorescence. In intact cells, prostaglandin F2alpha, which induces both intracellular Ca2+ release and Ca2+ entry, stimulated mitochondrial NAD(P)H formation. Thapsigargin-induced Ca2+ release and subsequent capacitative Ca2+ entry, both resulting in Ca2+ responses not exceeding 150-200 nM, also enhanced the reduction of pyridine nucleotides. As shown in inhibitor studies, the increased steady-state NAD(P)H level was due to activation of Ca2+-dependent dehydrogenases. [Ca2+]m, measured in permeabilized cells, increased moderately, but significantly, following elevation of [Ca2+]i from 50 to 180 nM, showed a further gradual increase at higher submicromolar [Ca2+]i values and rose steeply at supramicromolar [Ca2+]i. In summary, our results demonstrate that, in a steroid-producing cell type, net mitochondrial Ca2+ uptake and mitochondrial dehydrogenation can be activated even by low submicromolar increases of [Ca2+]i.


Subject(s)
Calcium/metabolism , Corpus Luteum/metabolism , Cytoplasm/metabolism , Mitochondria/metabolism , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cells, Cultured , Corpus Luteum/cytology , Dinoprost/pharmacology , Enzyme Inhibitors/pharmacology , Female , NADP/metabolism , Oxytocics/pharmacology , Pseudopregnancy/metabolism , Rats , Rats, Wistar , Rotenone/pharmacology , Stimulation, Chemical , Thapsigargin/pharmacology , Uncoupling Agents/pharmacology
19.
IUBMB Life ; 52(3-5): 213-9, 2001.
Article in English | MEDLINE | ID: mdl-11798035

ABSTRACT

Despite the early characterisation of Ca2+ fluxes in isolated mitochondria and the ability of this ion to up-regulate dehydrogenases of the Krebs cycle, the low affinity of the organelle uptake pathways was a long-standing obstacle to the recognition of a physiological role for mitochondrial Ca2+ uptake. This review begins with a historical account of the main results that proved the contrary and provides a brief description of mitochondrial Ca2+ signals. Then, it discusses the characteristics of Ca2+ regulation of mitochondrial function. Finally, it summarizes recent discoveries on structural aspects of mitochondrial reticulum and its connections to signalling partners such as the endoplasmic reticulum or the plasma membrane.


Subject(s)
Calcium/metabolism , Mitochondria/metabolism , Animals , Calcium Signaling , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Homeostasis , Intracellular Membranes/metabolism , Ion Transport
SELECTION OF CITATIONS
SEARCH DETAIL
...