Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Dev Cell ; 40(6): 583-594.e6, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28350990

ABSTRACT

Mitochondrial fission mediated by the GTPase dynamin-related protein 1 (Drp1) is an attractive drug target in numerous maladies that range from heart disease to neurodegenerative disorders. The compound mdivi-1 is widely reported to inhibit Drp1-dependent fission, elongate mitochondria, and mitigate brain injury. Here, we show that mdivi-1 reversibly inhibits mitochondrial complex I-dependent O2 consumption and reverse electron transfer-mediated reactive oxygen species (ROS) production at concentrations (e.g., 50 µM) used to target mitochondrial fission. Respiratory inhibition is rescued by bypassing complex I using yeast NADH dehydrogenase Ndi1. Unexpectedly, respiratory impairment by mdivi-1 occurs without mitochondrial elongation, is not mimicked by Drp1 deletion, and is observed in Drp1-deficient fibroblasts. In addition, mdivi-1 poorly inhibits recombinant Drp1 GTPase activity (Ki > 1.2 mM). Overall, these results suggest that mdivi-1 is not a specific Drp1 inhibitor. The ability of mdivi-1 to reversibly inhibit complex I and modify mitochondrial ROS production may contribute to effects observed in disease models.


Subject(s)
Dynamins/antagonists & inhibitors , Electron Transport Complex I/antagonists & inhibitors , GTP Phosphohydrolases/antagonists & inhibitors , Microtubule-Associated Proteins/antagonists & inhibitors , Mitochondria/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Quinazolinones/pharmacology , Reactive Oxygen Species/metabolism , Animals , COS Cells , Cell Respiration/drug effects , Chlorocebus aethiops , Dynamins/metabolism , Electron Transport Complex I/metabolism , Fibroblasts/metabolism , Fibroblasts/ultrastructure , GTP Phosphohydrolases/metabolism , Humans , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , NAD/metabolism , Neurons/metabolism , Oxidation-Reduction/drug effects , Oxygen Consumption/drug effects , Rats, Sprague-Dawley , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
2.
Drug Discov Today ; 21(6): 939-49, 2016 06.
Article in English | MEDLINE | ID: mdl-26861067

ABSTRACT

Although amyotrophic lateral sclerosis (ALS), also referred as 'Lou Gehrig's Disease,' was first described in 1869 and the first disease-associated gene was discovered almost 20 years ago, the disease etiology is still not fully understood and treatment options are limited to one drug approved by the US Food and Drug Administration (FDA). The slow translational progress suggests that current research models are not ideal to study such a complicated disease and need to be re-examined. Progress will require greater insight into human genes and biology involved in ALS susceptibility, as well as a deeper understanding of disease phenotype at the histological and molecular levels. Improving human disease outcome will require directing focus toward improved assessment technologies and innovative approaches.


Subject(s)
Amyotrophic Lateral Sclerosis , Animals , Biomedical Research , Disease Models, Animal , Humans
3.
Transl Stroke Res ; 4(6): 643-51, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24323418

ABSTRACT

Mitochondrial respiratory capacity is critical for responding to changes in neuronal energy demand. One approach toward neuroprotection is the administration of alternative energy substrates ("biofuels") to overcome brain injury-induced inhibition of glucose-based aerobic energy metabolism. This study tested the hypothesis that exogenous pyruvate, lactate, ß-hydroxybutyrate, and acetyl-L-carnitine each increase neuronal respiratory capacity in vitro either in the absence of or following transient excitotoxic glutamate receptor stimulation. Compared to the presence of 5 mM glucose alone, the addition of pyruvate, lactate, or ß-hydroxybutyrate (1.0-10.0 mM) to either day in vitro (DIV) 14 or 7 rat cortical neurons resulted in significant, dose-dependent stimulation of respiratory capacity, measured by cell respirometry as the maximal O2 consumption rate in the presence of the respiratory uncoupler carbonyl cyanide-p-trifluoromethoxyphenylhydrazone. A 30-min exposure to 100 µM glutamate impaired respiratory capacity for DIV 14, but not DIV 7, neurons. Glutamate reduced the respiratory capacity for DIV 14 neurons with glucose alone by 25 % and also reduced respiratory capacity with glucose plus pyruvate, lactate, or ß-hydroxybutyrate. However, respiratory capacity in glutamate-exposed neurons following pyruvate or ß-hydroxybutyrate addition was still, at least, as high as that obtained with glucose alone in the absence of glutamate exposure. These results support the interpretation that previously observed neuroprotection by exogenous pyruvate, lactate, or ß-hydroxybutyrate is at least partially mediated by their preservation of neuronal respiratory capacity.


Subject(s)
Cerebral Cortex/drug effects , Glutamic Acid/pharmacology , Mitochondria/drug effects , Neurons/drug effects , Oxygen Consumption/drug effects , 3-Hydroxybutyric Acid/pharmacology , Acetylcarnitine/pharmacology , Animals , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cell Respiration/drug effects , Cerebral Cortex/metabolism , Dose-Response Relationship, Drug , Energy Metabolism/drug effects , Glutamic Acid/metabolism , In Vitro Techniques , Lactic Acid/pharmacology , Mitochondria/metabolism , Neurons/metabolism , Nootropic Agents/pharmacology , Proton Ionophores/pharmacology , Pyruvic Acid/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Glutamate/metabolism
4.
PLoS One ; 8(11): e79982, 2013.
Article in English | MEDLINE | ID: mdl-24236167

ABSTRACT

Extracellular glutamate is elevated following brain ischemia or trauma and contributes to neuronal injury. We tested the hypothesis that magnesium sulfate (MgSO4, 3 mM) protects against metabolic failure caused by excitotoxic glutamate exposure. Rat cortical neuron preparations treated in medium already containing a physiological concentration of Mg(2+) (1 mM) could be segregated based on their response to glutamate (100 µM). Type I preparations responded with a decrease or small transient increase in oxygen consumption rate (OCR). Type II neurons responded with >50% stimulation in OCR, indicating a robust response to increased energy demand without immediate toxicity. Pre-treatment with MgSO4 improved the initial bioenergetic response to glutamate and ameliorated subsequent loss of spare respiratory capacity, measured following addition of the uncoupler FCCP, in Type I but not Type II neurons. Spare respiratory capacity in Type I neurons was also improved by incubation with MgSO4 or NMDA receptor antagonist MK801 in the absence of glutamate treatment. This finding indicates that the major difference between Type I and Type II preparations is the amount of endogenous glutamate receptor activity. Incubation of Type II neurons with 5 µM glutamate prior to excitotoxic (100 µM) glutamate exposure recapitulated a Type I phenotype. MgSO4 protected against an excitotoxic glutamate-induced drop in neuronal ATP both with and without prior 5 µM glutamate exposure. Results indicate that MgSO4 protects against chronic moderate glutamate receptor stimulation and preserves cellular ATP following treatment with excitotoxic glutamate.


Subject(s)
Magnesium Sulfate/pharmacology , Receptors, Glutamate/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Respiration/drug effects , Cells, Cultured , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Glycolysis/drug effects , Mitochondria/metabolism , Neurons/drug effects , Neurons/metabolism , Oxygen Consumption , Phenotype , Pyruvic Acid/metabolism , Rats
5.
PLoS One ; 7(8): e42487, 2012.
Article in English | MEDLINE | ID: mdl-22880001

ABSTRACT

Cells that exhibit an absolute dependence on the anti-apoptotic BCL-2 protein for survival are termed "primed for death" and are killed by the BCL-2 antagonist ABT-737. Many cancers exhibit a primed phenotype, including some that are resistant to conventional chemotherapy due to high BCL-2 expression. We show here that 1) stable BCL-2 overexpression alone can induce a primed for death state and 2) that an ABT-737-induced loss of functional cytochrome c from the electron transport chain causes a reduction in maximal respiration that is readily detectable by microplate-based respirometry. Stable BCL-2 overexpression sensitized non-tumorigenic MCF10A mammary epithelial cells to ABT-737-induced caspase-dependent apoptosis. Mitochondria within permeabilized BCL-2 overexpressing cells were selectively vulnerable to ABT-737-induced cytochrome c release compared to those from control-transfected cells, consistent with a primed state. ABT-737 treatment caused a dose-dependent impairment of maximal O(2) consumption in MCF10A BCL-2 overexpressing cells but not in control-transfected cells or in immortalized mouse embryonic fibroblasts lacking both BAX and BAK. This impairment was rescued by delivering exogenous cytochrome c to mitochondria via saponin-mediated plasma membrane permeabilization. An ABT-737-induced reduction in maximal O(2) consumption was also detectable in SP53, JeKo-1, and WEHI-231 B-cell lymphoma cell lines, with sensitivity correlating with BCL-2:MCL-1 ratio and with susceptibility (SP53 and JeKo-1) or resistance (WEHI-231) to ABT-737-induced apoptosis. Multiplexing respirometry assays to ELISA-based determination of cytochrome c redistribution confirmed that respiratory inhibition was associated with cytochrome c release. In summary, cell-based respiration assays were able to rapidly identify a primed for death state in cells with either artificially overexpressed or high endogenous BCL-2. Rapid detection of a primed for death state in individual cancers by "bioenergetics-based profiling" may eventually help identify the subset of patients with chemoresistant but primed tumors who can benefit from treatment that incorporates a BCL-2 antagonist.


Subject(s)
Biphenyl Compounds/pharmacology , Cytological Techniques/instrumentation , Cytological Techniques/methods , Nitrophenols/pharmacology , Sulfonamides/pharmacology , Aniline Compounds/pharmacology , Animals , Cell Death/drug effects , Cell Line , Cell Line, Tumor , Cell Respiration/drug effects , Cytochromes c/metabolism , Energy Metabolism/drug effects , Humans , Lymphoma, B-Cell/pathology , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Oxygen Consumption/drug effects , Phenotype , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
6.
PLoS One ; 7(4): e34465, 2012.
Article in English | MEDLINE | ID: mdl-22496810

ABSTRACT

Mitochondrial dysfunction is a component of many neurodegenerative conditions. Measurement of oxygen consumption from intact neurons enables evaluation of mitochondrial bioenergetics under conditions that are more physiologically realistic compared to isolated mitochondria. However, mechanistic analysis of mitochondrial function in cells is complicated by changing energy demands and lack of substrate control. Here we describe a technique for sequentially measuring respiration from intact and saponin-permeabilized cortical neurons on single microplates. This technique allows control of substrates to individual electron transport chain complexes following permeabilization, as well as side-by-side comparisons to intact cells. To illustrate the utility of the technique, we demonstrate that inhibition of respiration by the drug KB-R7943 in intact neurons is relieved by delivery of the complex II substrate succinate, but not by complex I substrates, via acute saponin permeabilization. In contrast, methyl succinate, a putative cell permeable complex II substrate, failed to rescue respiration in intact neurons and was a poor complex II substrate in permeabilized cells. Sequential measurements of intact and permeabilized cell respiration should be particularly useful for evaluating indirect mitochondrial toxicity due to drugs or cellular signaling events which cannot be readily studied using isolated mitochondria.


Subject(s)
Cell Membrane Permeability/drug effects , Cell Respiration/physiology , Energy Metabolism , Microtechnology/methods , Mitochondria/pathology , Neurons/metabolism , Oxygen Consumption/drug effects , Animals , Anti-Arrhythmia Agents/pharmacology , Cell Respiration/drug effects , Electron Transport , Electron Transport Complex I/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Mitochondria/drug effects , Neurons/cytology , Neurons/drug effects , Oxidation-Reduction , Rats , Saponins/pharmacology , Signal Transduction , Succinic Acid/pharmacology , Thiourea/analogs & derivatives , Thiourea/pharmacology
7.
Mol Biol Cell ; 22(13): 2235-45, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21551073

ABSTRACT

Previous studies using in vitro cell culture systems have shown the role of the dynamin-related GTPase Opa1 in apoptosis prevention and mitochondrial DNA (mtDNA) maintenance. However, it remains to be tested whether these functions of Opa1 are physiologically important in vivo in mammals. Here, using the Cre-loxP system, we deleted mouse Opa1 in pancreatic beta cells, in which glucose-stimulated ATP production in mitochondria plays a key role in insulin secretion. Beta cells lacking Opa1 maintained normal copy numbers of mtDNA; however, the amount and activity of electron transport chain complex IV were significantly decreased, leading to impaired glucose-stimulated ATP production and insulin secretion. In addition, in Opa1-null beta cells, cell proliferation was impaired, whereas apoptosis was not promoted. Consequently, mice lacking Opa1 in beta cells develop hyperglycemia. The data suggest that the function of Opa1 in the maintenance of the electron transport chain is physiologically relevant in beta cells.


Subject(s)
Adenosine Triphosphate/biosynthesis , Dynamins/metabolism , GTP Phosphohydrolases/metabolism , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis/physiology , Cell Proliferation , DNA, Mitochondrial/metabolism , Electron Transport Complex IV/metabolism , GTP Phosphohydrolases/deficiency , GTP Phosphohydrolases/genetics , Glucose Intolerance/metabolism , Hyperglycemia/metabolism , Hyperglycemia/pathology , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/enzymology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
8.
J Neurosci Res ; 89(12): 1979-88, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21520220

ABSTRACT

Multiple neurodegenerative disorders are associated with altered mitochondrial bioenergetics. Although mitochondrial O(2) consumption is frequently measured in isolated mitochondria, isolated synaptic nerve terminals (synaptosomes), or cultured cells, the absence of mature brain circuitry is a remaining limitation. Here we describe the development of a method that adapts the Seahorse Extracellular Flux Analyzer (XF24) for the microplate-based measurement of hippocampal slice O(2) consumption. As a first evaluation of the technique, we compared whole-slice bioenergetics with previous measurements made with synaptosomes or cultured neurons. We found that mitochondrial respiratory capacity and O(2) consumption coupled to ATP synthesis could be estimated in cultured or acute hippocampal slices with preserved neural architecture. Mouse organotypic hippocampal slices oxidizing glucose displayed mitochondrial O(2) consumption that was well coupled, as determined by the sensitivity to the ATP synthase inhibitor oligomycin. However, stimulation of respiration by uncoupler was modest (<120% of basal respiration) compared with previous measurements in cells or synaptosomes, though enhanced slightly (to ∼150% of basal respiration) by acute addition of the mitochondrial complex I-linked substrate pyruvate. These findings suggest a high basal utilization of respiratory capacity in slices and a limitation of glucose-derived substrate for maximal respiration. The improved throughput of microplate-based hippocampal respirometry over traditional O(2) electrode-based methods is conducive to neuroprotective drug screening. When coupled with cell type-specific pharmacology or genetic manipulations, the ability to measure O(2) consumption efficiently from whole slices should advance our understanding of mitochondrial roles in physiology and neuropathology.


Subject(s)
Cell Respiration/physiology , Hippocampus/physiology , Organ Culture Techniques/methods , Oxygen/analysis , Animals , Energy Metabolism/physiology , Mice , Mice, Inbred C57BL , Oxygen Consumption/physiology , Rats , Rats, Sprague-Dawley
9.
EMBO J ; 28(11): 1589-600, 2009 Jun 03.
Article in English | MEDLINE | ID: mdl-19360003

ABSTRACT

Mitochondria are dynamic organelles, the morphology of which results from an equilibrium between two opposing processes, fusion and fission. Mitochondrial fusion relies on dynamin-related GTPases, the mitofusins (MFN1 and 2) in the outer mitochondrial membrane and OPA1 (optic atrophy 1) in the inner mitochondrial membrane. Apart from a role in the maintenance of mitochondrial DNA, little is known about the physiological role of mitochondrial fusion. Here we report that mitochondria hyperfuse and form a highly interconnected network in cells exposed to selective stresses. This process precedes mitochondrial fission when it is triggered by apoptotic stimuli such as UV irradiation or actinomycin D. Stress-induced mitochondrial hyperfusion (SIMH) is independent of MFN2, BAX/BAK, and prohibitins, but requires L-OPA1, MFN1, and the mitochondrial inner membrane protein SLP-2. In the absence of SLP-2, L-OPA1 is lost and SIMH is prevented. SIMH is accompanied by increased mitochondrial ATP production and represents a novel adaptive pro-survival response against stress.


Subject(s)
Fibroblasts/physiology , Membrane Proteins/physiology , Mitochondria/physiology , Stress, Physiological , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Dactinomycin/toxicity , Fibroblasts/drug effects , Fibroblasts/radiation effects , GTP Phosphohydrolases/physiology , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/radiation effects , Ultraviolet Rays
10.
J Bioenerg Biomembr ; 39(2): 158-66, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17447126

ABSTRACT

We have studied the effect of nitric oxide (NO) and potassium cyanide (KCN) on oxidative phosphorylation efficiency. Concentrations of NO or KCN that decrease resting oxygen consumption by 10-20% increased oxidative phosphorylation efficiency in mitochondria oxidizing succinate or palmitoyl-L-carnitine, but not in mitochondria oxidizing malate plus glutamate. When compared to malate plus glutamate, succinate or palmitoyl-L-carnitine reduced the redox state of cytochrome oxidase. The relationship between membrane potential and oxygen consumption rates was measured at different degrees of ATP synthesis. The use of malate plus glutamate instead of succinate (that changes the H(+)/2e(-) stoichiometry of the respiratory chain) affected the relationship, whereas a change in membrane permeability did not affect it. NO or KCN also affected the relationship, suggesting that they change the H(+)/2e(-) stoichiometry of the respiratory chain. We propose that NO may be a natural short-term regulator of mitochondrial physiology that increases oxidative phosphorylation efficiency in a redox-sensitive manner by decreasing the slipping in the proton pumps.


Subject(s)
Mitochondria, Liver/metabolism , Nitric Oxide/physiology , Oxygen Consumption/physiology , Animals , Cell Respiration/drug effects , Cell Respiration/physiology , Glutamic Acid/metabolism , In Vitro Techniques , Malates/metabolism , Membrane Potential, Mitochondrial , Mitochondria, Liver/drug effects , Nitric Oxide/pharmacology , Oxidative Phosphorylation , Oxygen Consumption/drug effects , Palmitoylcarnitine/metabolism , Potassium Cyanide/pharmacology , Rats , Succinic Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...