Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
PLoS One ; 15(11): e0242443, 2020.
Article in English | MEDLINE | ID: mdl-33216776

ABSTRACT

Idiopathic Inflammatory Myopathies (IIMs) have been studied within the framework of autoimmune diseases where skeletal muscle appears to have a passive role in the illness. However, persiting weakness even after resolving inflammation raises questions about the role that skeletal muscle plays by itself in these diseases. "Non-immune mediated" hypotheses have arisen to consider inner skeletal muscle cell processes as trigger factors in the clinical manifestations of IIMs. Alterations in oxidative phosphorylation, ATP production, calcium handling, autophagy, endoplasmic reticulum stress, among others, have been proposed as alternative cellular pathophysiological mechanisms. In this study, we used skeletal muscle-derived cells, from healthy controls and IIM patients to determine mitochondrial function and mitochondrial ability to adapt to a metabolic stress when deprived of glucose. We hypothesized that mitochondria would be dysfunctional in IIM samples, which was partially true in normal glucose rich growing medium as determined by oxygen consumption rate. However, in the glucose-free and galactose supplemented condition, a medium that forced mitochondria to function, IIM cells increased their respiration, reaching values matching normal derived cells. Unexpectedly, cell death significantly increased in IIM cells under this condition. Our findings show that mitochondria in IIM is functional and the decrease respiration observed is part of an adaptative response to improve survival. The increased metabolic function obtained after forcing IIM cells to rely on mitochondrial synthesized ATP is detrimental to the cell's viability. Thus, therapeutic interventions that activate mitochondria, could be detrimental in IIM cell physiology, and must be avoided in patients with IIM.


Subject(s)
Apoptosis/physiology , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Myoblasts/pathology , Myositis/pathology , Adenosine Triphosphate/analysis , Aged , Autoantibodies/immunology , Autoimmune Diseases/pathology , Cells, Cultured , Endoplasmic Reticulum Stress/physiology , Female , Humans , Male , Membrane Potential, Mitochondrial/physiology , Middle Aged , Oxidative Phosphorylation , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism
2.
Sci Signal ; 13(640)2020 07 14.
Article in English | MEDLINE | ID: mdl-32665411

ABSTRACT

Spontaneous Ca2+ signaling from the InsP3R intracellular Ca2+ release channel to mitochondria is essential for optimal oxidative phosphorylation (OXPHOS) and ATP production. In cells with defective OXPHOS, reductive carboxylation replaces oxidative metabolism to maintain amounts of reducing equivalents and metabolic precursors. To investigate the role of mitochondrial Ca2+ uptake in regulating bioenergetics in these cells, we used OXPHOS-competent and OXPHOS-defective cells. Inhibition of InsP3R activity or mitochondrial Ca2+ uptake increased α-ketoglutarate (αKG) abundance and the NAD+/NADH ratio, indicating that constitutive endoplasmic reticulum (ER)-to-mitochondria Ca2+ transfer promoted optimal αKG dehydrogenase (αKGDH) activity. Reducing mitochondrial Ca2+ inhibited αKGDH activity and increased NAD+, which induced SIRT1-dependent autophagy in both OXPHOS-competent and OXPHOS-defective cells. Whereas autophagic flux in OXPHOS-competent cells promoted cell survival, it was impaired in OXPHOS-defective cells because of inhibition of autophagosome-lysosome fusion. Inhibition of αKGDH and impaired autophagic flux in OXPHOS-defective cells resulted in pronounced cell death in response to interruption of constitutive flux of Ca2+ from ER to mitochondria. These results demonstrate that mitochondria play a fundamental role in maintaining bioenergetic homeostasis of both OXPHOS-competent and OXPHOS-defective cells, with Ca2+ regulation of αKGDH activity playing a pivotal role. Inhibition of ER-to-mitochondria Ca2+ transfer may represent a general therapeutic strategy against cancer cells regardless of their OXPHOS status.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Oxidative Phosphorylation , Cell Line, Tumor , Cell Survival , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Humans , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/pathology
3.
FASEB J ; 34(6): 7847-7865, 2020 06.
Article in English | MEDLINE | ID: mdl-32301552

ABSTRACT

Transient receptor potential melastatin 4 (TRPM4) is a Ca2+ -activated nonselective cationic channel that regulates cell migration and contractility. Increased TRPM4 expression has been related to pathologies, in which cytoskeletal rearrangement and cell migration are altered, such as metastatic cancer. Here, we identify the K+ channel tetramerization domain 5 (KCTD5) protein, a putative adaptor of cullin3 E3 ubiquitin ligase, as a novel TRPM4-interacting protein. We demonstrate that KCTD5 is a positive regulator of TRPM4 activity by enhancing its Ca2+ sensitivity. We show that through its effects on TRPM4 that KCTD5 promotes cell migration and contractility. Finally, we observed that both TRPM4 and KCTD5 expression are increased in distinct patterns in different classes of breast cancer tumor samples. Together, these data support that TRPM4 activity can be regulated through expression levels of either TRPM4 or KCTD5, not only contributing to increased understanding of the molecular mechanisms involved on the regulation of these important ion channels, but also providing information that could inform treatments based on targeting these distinct molecules that define TRPM4 activity.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement/physiology , Potassium Channels/metabolism , TRPM Cation Channels/metabolism , Animals , Breast/metabolism , Breast/pathology , COS Cells , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Female , HEK293 Cells , Humans , MCF-7 Cells , Prognosis , Ubiquitin-Protein Ligases/metabolism
4.
Mitochondrion ; 49: 73-82, 2019 11.
Article in English | MEDLINE | ID: mdl-31310854

ABSTRACT

Cytosolic calcium (cCa2+) entry into mitochondria is facilitated by the mitochondrial membrane potential (ΔΨm), an electrochemical gradient generated by the electron transport chain (ETC). Is has been assumed that as long as mutations that affect the ETC do not affect the ΔΨm, the mitochondrial Ca2+ (mCa2+) homeostasis remains normal. We show that knockdown of NDUFAF3 and SDHB reduce ETC activity altering mCa2+ efflux and influx rates while ΔΨm remains intact. Shifting the equilibrium toward lower [Ca2+]m accumulation renders cells resistant to death. Our findings reveal an unexpected relationship between complex I and II with the mCa2+ homeostasis independent of ΔΨm.


Subject(s)
Calcium/metabolism , Electron Transport Complex I/metabolism , Homeostasis , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Succinate Dehydrogenase/metabolism , Electron Transport Complex I/genetics , Humans , MCF-7 Cells , Membrane Potential, Mitochondrial/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Succinate Dehydrogenase/genetics
6.
Nat Cell Biol ; 21(6): 755-767, 2019 06.
Article in English | MEDLINE | ID: mdl-31110288

ABSTRACT

Mitochondria-associated membranes (MAMs) are central microdomains that fine-tune bioenergetics by the local transfer of calcium from the endoplasmic reticulum to the mitochondrial matrix. Here, we report an unexpected function of the endoplasmic reticulum stress transducer IRE1α as a structural determinant of MAMs that controls mitochondrial calcium uptake. IRE1α deficiency resulted in marked alterations in mitochondrial physiology and energy metabolism under resting conditions. IRE1α determined the distribution of inositol-1,4,5-trisphosphate receptors at MAMs by operating as a scaffold. Using mutagenesis analysis, we separated the housekeeping activity of IRE1α at MAMs from its canonical role in the unfolded protein response. These observations were validated in vivo in the liver of IRE1α conditional knockout mice, revealing broad implications for cellular metabolism. Our results support an alternative function of IRE1α in orchestrating the communication between the endoplasmic reticulum and mitochondria to sustain bioenergetics.


Subject(s)
Endoplasmic Reticulum/metabolism , Endoribonucleases/genetics , Energy Metabolism , Mitochondria/metabolism , Protein Serine-Threonine Kinases/genetics , Animals , Calcium/metabolism , Calcium Signaling/genetics , Endoplasmic Reticulum/genetics , Inositol 1,4,5-Trisphosphate Receptors/genetics , Mice , Mice, Knockout , Mitochondria/genetics
7.
Cell Rep ; 26(13): 3709-3725.e7, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30917323

ABSTRACT

Mitochondrial Ca2+ uniporter (MCU)-mediated Ca2+ uptake promotes the buildup of reducing equivalents that fuel oxidative phosphorylation for cellular metabolism. Although MCU modulates mitochondrial bioenergetics, its function in energy homeostasis in vivo remains elusive. Here we demonstrate that deletion of the Mcu gene in mouse liver (MCUΔhep) and in Danio rerio by CRISPR/Cas9 inhibits mitochondrial Ca2+ (mCa2+) uptake, delays cytosolic Ca2+ (cCa2+) clearance, reduces oxidative phosphorylation, and leads to increased lipid accumulation. Elevated hepatic lipids in MCUΔhep were a direct result of extramitochondrial Ca2+-dependent protein phosphatase-4 (PP4) activity, which dephosphorylates AMPK. Loss of AMPK recapitulates hepatic lipid accumulation without changes in MCU-mediated Ca2+ uptake. Furthermore, reconstitution of active AMPK, or PP4 knockdown, enhances lipid clearance in MCUΔhep hepatocytes. Conversely, gain-of-function MCU promotes rapid mCa2+ uptake, decreases PP4 levels, and reduces hepatic lipid accumulation. Thus, our work uncovers an MCU/PP4/AMPK molecular cascade that links Ca2+ dynamics to hepatic lipid metabolism.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Hepatocytes/metabolism , Lipid Metabolism , Mitochondrial Proteins/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Calcium Channels/genetics , Cells, Cultured , Female , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mitochondria, Liver/metabolism , Mitochondrial Proteins/genetics , Phosphoprotein Phosphatases/metabolism , Protein Kinases/metabolism , Zebrafish
8.
Cell Rep ; 23(4): 1005-1019, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29694881

ABSTRACT

Mitochondria shape cytosolic calcium ([Ca2+]c) transients and utilize the mitochondrial Ca2+ ([Ca2+]m) in exchange for bioenergetics output. Conversely, dysregulated [Ca2+]c causes [Ca2+]m overload and induces permeability transition pore and cell death. Ablation of MCU-mediated Ca2+ uptake exhibited elevated [Ca2+]c and failed to prevent stress-induced cell death. The mechanisms for these effects remain elusive. Here, we report that mitochondria undergo a cytosolic Ca2+-induced shape change that is distinct from mitochondrial fission and swelling. [Ca2+]c elevation, but not MCU-mediated Ca2+ uptake, appears to be essential for the process we term mitochondrial shape transition (MiST). MiST is mediated by the mitochondrial protein Miro1 through its EF-hand domain 1 in multiple cell types. Moreover, Ca2+-dependent disruption of Miro1/KIF5B/tubulin complex is determined by Miro1 EF1 domain. Functionally, Miro1-dependent MiST is essential for autophagy/mitophagy that is attenuated in Miro1 EF1 mutants. Thus, Miro1 is a cytosolic Ca2+ sensor that decodes metazoan Ca2+ signals as MiST.


Subject(s)
Calcium/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics , Receptors, G-Protein-Coupled/metabolism , Stress, Physiological , rho GTP-Binding Proteins/metabolism , Animals , HeLa Cells , Humans , Mice , Mice, Mutant Strains , Mitochondria/genetics , Receptors, G-Protein-Coupled/genetics , rho GTP-Binding Proteins/genetics
9.
PLoS One ; 12(9): e0184189, 2017.
Article in English | MEDLINE | ID: mdl-28898261

ABSTRACT

Aging is a gradual biological process characterized by a decrease in cell and organism functions. Gingival wound healing is one of the impaired processes found in old rats. Here, we studied the in vivo wound healing process using a gingival repair rat model and an in vitro model using human gingival fibroblast for cellular responses associated to wound healing. To do that, we evaluated cell proliferation of both epithelial and connective tissue cells in gingival wounds and found decreased of Ki67 nuclear staining in old rats when compared to their young counterparts. We next evaluated cellular responses of primary gingival fibroblast obtained from young subjects in the presence human blood serum of individuals of different ages. Eighteen to sixty five years old masculine donors were classified into 3 groups: "young" from 18 to 22 years old, "middle-aged" from 30 to 48 years old and "aged" over 50 years old. Cell proliferation, measured through immunofluorescence for Ki67 and flow cytometry for DNA content, was decreased when middle-aged and aged serum was added to gingival fibroblast compared to young serum. Myofibroblastic differentiation, measured through alpha-smooth muscle actin (α-SMA), was stimulated with young but not middle-aged or aged serum both the protein levels and incorporation of α-SMA into actin stress fibers. High levels of PDGF, VEGF, IL-6R were detected in blood serum from young subjects when compared to middle-aged and aged donors. In addition, the pro-inflammatory cytokines MCP-1 and TNF were increased in the serum of aged donors. In old rat wound there is an increased of staining for TNF compared to young wound. Moreover, healthy gingiva (non injury) shows less staining compared to a wound site, suggesting a role in wound healing. Moreover, serum from middle-aged and aged donors was able to stimulate cellular senescence in young cells as determined by the expression of senescence associated beta-galactosidase and histone H2A.X phosphorylated at Ser139. Moreover, we detected an increased frequency of γ-H2A.X-positive cells in aged rat gingival tissues. The present study suggests that serum factors present in middle-aged and aged individuals may be responsible, at least in part, for the altered responses observed during wound healing in aging.


Subject(s)
Aging/blood , Gingiva/metabolism , Gingiva/pathology , Wound Healing , Age Factors , Aged , Aged, 80 and over , Animals , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Proliferation , Cytokines/blood , Cytokines/pharmacology , Disease Models, Animal , Fibroblasts/metabolism , Gingiva/drug effects , Humans , Inflammation Mediators/blood , Inflammation Mediators/pharmacology , Male , Middle Aged , Myofibroblasts/cytology , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Rats , Serum , Wound Healing/drug effects , beta-Galactosidase/metabolism
10.
Mol Cell ; 65(6): 1014-1028.e7, 2017 Mar 16.
Article in English | MEDLINE | ID: mdl-28262504

ABSTRACT

Ca2+ dynamics and oxidative signaling are fundamental mechanisms for mitochondrial bioenergetics and cell function. The MCU complex is the major pathway by which these signals are integrated in mitochondria. Whether and how these coactive elements interact with MCU have not been established. As an approach toward understanding the regulation of MCU channel by oxidative milieu, we adapted inflammatory and hypoxia models. We identified the conserved cysteine 97 (Cys-97) to be the only reactive thiol in human MCU that undergoes S-glutathionylation. Furthermore, biochemical, structural, and superresolution imaging analysis revealed that MCU oxidation promotes MCU higher order oligomer formation. Both oxidation and mutation of MCU Cys-97 exhibited persistent MCU channel activity with higher [Ca2+]m uptake rate, elevated mROS, and enhanced [Ca2+]m overload-induced cell death. In contrast, these effects were largely independent of MCU interaction with its regulators. These findings reveal a distinct functional role for Cys-97 in ROS sensing and regulation of MCU activity.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling , Calcium/metabolism , Endothelial Cells/metabolism , Ion Channel Gating , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Reactive Oxygen Species/metabolism , Animals , COS Cells , Calcium Channels/chemistry , Calcium Channels/genetics , Calcium Signaling/drug effects , Cell Death , Cell Hypoxia , Chlorocebus aethiops , Cysteine , Endothelial Cells/drug effects , Endothelial Cells/pathology , Energy Metabolism , Glutathione/metabolism , HEK293 Cells , HeLa Cells , Humans , Ion Channel Gating/drug effects , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/pathology , Mutation , Oxidation-Reduction , Protein Multimerization , Protein Processing, Post-Translational , Protein Structure, Quaternary , Structure-Activity Relationship , Thrombin/pharmacology , Time Factors , Transfection
11.
PLoS One ; 12(1): e0170213, 2017.
Article in English | MEDLINE | ID: mdl-28129327

ABSTRACT

AIMS: Pancreatic ß-cells synthesize and release serotonin (5 hydroxytryptamine, 5HT); however, the role of 5HT receptors on glucose stimulated insulin secretion (GSIS) and the mechanisms mediating this function is not fully understood. The aims of this study were to determine the expression profile of 5HT receptors in murine MIN6 ß-cells and to examine the effects of pharmacological activation of 5HT receptor Htr2b on GSIS and mitochondrial function. MATERIALS AND METHODS: mRNA levels of 5HT receptors in MIN6 cells were quantified by RT qPCR. GSIS was assessed in MIN6 cells in response to global serotonergic activation with 5HT and pharmacological Htr2b activation or inhibition with BW723C86 or SB204741, respectively. In response to Htr2b activation also was evaluated the mRNA and protein levels of PGC1α and PPARy by RT-qPCR and western blotting and mitochondrial function by oxygen consumption rate (OCR) and ATP cellular content. RESULTS: We found that mRNA levels of most 5HT receptors were either very low or undetectable in MIN6 cells. By contrast, Htr2b mRNA was present at moderate levels in these cells. Preincubation (6 h) of MIN6 cells with 5HT or BW723C86 reduced GSIS and the effect of 5HT was prevented by SB204741. Preincubation with BW723C86 increased PGC1α and PPARy mRNA and protein levels and decreased mitochondrial respiration and ATP content in MIN6 cells. CONCLUSIONS: Our results indicate that prolonged Htr2b activation in murine ß-cells decreases glucose-stimulated insulin secretion and mitochondrial activity by mechanisms likely dependent on enhanced PGC1α/PPARy expression.


Subject(s)
Insulin/metabolism , PPAR gamma/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Receptors, Serotonin/genetics , Serotonin/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Gene Expression Regulation/drug effects , Glucose/metabolism , Humans , Indoles/pharmacology , Insulin/genetics , Insulin-Secreting Cells/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Mice , Mitochondria/genetics , Mitochondria/metabolism , Oxygen Consumption/genetics , PPAR gamma/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Receptors, Serotonin/biosynthesis , Serotonin/genetics , Serotonin/pharmacology , Thiophenes/pharmacology , Urea/analogs & derivatives , Urea/pharmacology
12.
Cell Rep ; 15(8): 1673-85, 2016 05 24.
Article in English | MEDLINE | ID: mdl-27184846

ABSTRACT

Mitochondrial Ca(2+) Uniporter (MCU)-dependent mitochondrial Ca(2+) uptake is the primary mechanism for increasing matrix Ca(2+) in most cell types. However, a limited understanding of the MCU complex assembly impedes the comprehension of the precise mechanisms underlying MCU activity. Here, we report that mouse cardiomyocytes and endothelial cells lacking MCU regulator 1 (MCUR1) have severely impaired [Ca(2+)]m uptake and IMCU current. MCUR1 binds to MCU and EMRE and function as a scaffold factor. Our protein binding analyses identified the minimal, highly conserved regions of coiled-coil domain of both MCU and MCUR1 that are necessary for heterooligomeric complex formation. Loss of MCUR1 perturbed MCU heterooligomeric complex and functions as a scaffold factor for the assembly of MCU complex. Vascular endothelial deletion of MCU and MCUR1 impaired mitochondrial bioenergetics, cell proliferation, and migration but elicited autophagy. These studies establish the existence of a MCU complex that assembles at the mitochondrial integral membrane and regulates Ca(2+)-dependent mitochondrial metabolism.


Subject(s)
Calcium Channels/metabolism , Energy Metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Animals , Autophagy , Calcium/metabolism , Calcium Channels/chemistry , Cell Movement , Endothelial Cells/metabolism , Gene Deletion , HEK293 Cells , HeLa Cells , Heart/physiology , Humans , Mice, Knockout , Mitochondrial Proteins/chemistry , Neovascularization, Physiologic , Protein Binding , Protein Domains
14.
Cell Mol Life Sci ; 73(18): 3583-97, 2016 09.
Article in English | MEDLINE | ID: mdl-27001668

ABSTRACT

L-Dopa continues to be the gold drug in Parkinson's disease (PD) treatment from 1967. The failure to translate successful results from preclinical to clinical studies can be explained by the use of preclinical models which do not reflect what happens in the disease since these induce a rapid and extensive degeneration; for example, MPTP induces a severe Parkinsonism in only 3 days in humans contrasting with the slow degeneration and progression of PD. This study presents a new anatomy and develops preclinical model based on aminochrome which induces a slow and progressive dysfunction of dopaminergic neurons. The unilateral injection of aminochrome into rat striatum resulted in (1) contralateral rotation when the animals are stimulated with apomorphine; (2) absence of significant loss of tyrosine hydroxylase-positive neuronal elements both in substantia nigra and striatum; (3) cell shrinkage; (4) significant reduction of dopamine release; (5) significant increase in GABA release; (6) significant decrease in the number of monoaminergic presynaptic vesicles; (7) significant increase of dopamine concentration inside of monoaminergic vesicles; (8) significant increase of damaged mitochondria; (9) significant decrease of ATP level in the striatum (10) significant decrease in basal and maximal mitochondrial respiration. These results suggest that aminochrome induces dysfunction of dopaminergic neurons where the contralateral behavior can be explained by aminochrome-induced ATP decrease required both for anterograde transport of synaptic vesicles and dopamine release. Aminochrome could be implemented as a new model neurotoxin to study Parkinson's disease.


Subject(s)
Dopaminergic Neurons/drug effects , Indolequinones/pharmacology , Parkinson Disease/pathology , Adenosine Triphosphate/metabolism , Animals , Behavior, Animal/drug effects , Chromatography, High Pressure Liquid , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/analysis , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Humans , Indolequinones/chemical synthesis , Indolequinones/chemistry , Male , Mitochondria/drug effects , Mitochondria/metabolism , Oxygen Consumption/drug effects , Oxygen Consumption/physiology , Parkinson Disease/metabolism , Parkinson Disease/veterinary , Rats , Rats, Sprague-Dawley , Substantia Nigra/metabolism , Synaptic Vesicles/metabolism , Tyrosine 3-Monooxygenase/metabolism , gamma-Aminobutyric Acid/analysis
15.
Cell Rep ; 14(10): 2313-24, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26947070

ABSTRACT

In the absence of low-level ER-to-mitochondrial Ca(2+) transfer, ATP levels fall, and AMPK-dependent, mTOR-independent autophagy is induced as an essential survival mechanism in many cell types. Here, we demonstrate that tumorigenic cancer cell lines, transformed primary human fibroblasts, and tumors in vivo respond similarly but that autophagy is insufficient for survival, and cancer cells die while their normal counterparts are spared. Cancer cell death is due to compromised bioenergetics that can be rescued with metabolic substrates or nucleotides and caused by necrosis associated with mitotic catastrophe during their proliferation. Our findings reveal an unexpected dependency on constitutive Ca(2+) transfer to mitochondria for viability of tumorigenic cells and suggest that mitochondrial Ca(2+) addiction is a feature of cancer cells.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , AMP-Activated Protein Kinases/metabolism , Acetylcysteine/pharmacology , Adenosine Triphosphate/metabolism , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Blotting, Western , Cell Line, Tumor , Humans , Inositol 1,4,5-Trisphosphate Receptors/antagonists & inhibitors , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Macrocyclic Compounds/pharmacology , Microscopy, Video , Oxazoles/pharmacology , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
16.
J Med Chem ; 57(6): 2440-54, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24568614

ABSTRACT

Tumor cells principally exhibit increased mitochondrial transmembrane potential (ΔΨ(m)) and altered metabolic pathways. The therapeutic targeting and delivery of anticancer drugs to the mitochondria might improve treatment efficacy. Gallic acid exhibits a variety of biological activities, and its ester derivatives can induce mitochondrial dysfunction. Four alkyl gallate triphenylphosphonium lipophilic cations were synthesized, each differing in the size of the linker chain at the cationic moiety. These derivatives were selectively cytotoxic toward tumor cells. The better compound (TPP(+)C10) contained 10 carbon atoms within the linker chain and exhibited an IC50 value of approximately 0.4-1.6 µM for tumor cells and a selectivity index of approximately 17-fold for tumor compared with normal cells. Consequently, its antiproliferative effect was also assessed in vivo. The oxygen consumption rate and NAD(P)H oxidation levels increased in the tumor cell lines (uncoupling effect), resulting in a ΔΨ(m) decrease and a consequent decrease in intracellular ATP levels. Moreover, TPP(+)C10 significantly inhibited the growth of TA3/Ha tumors in mice. According to these results, the antineoplastic activity and safety of TPP(+)C10 warrant further comprehensive evaluation.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Gallic Acid/analogs & derivatives , Gallic Acid/chemical synthesis , Adenosine Triphosphate/metabolism , Analysis of Variance , Animals , Apoptosis/drug effects , Caspase 3/drug effects , Caspase Inhibitors/chemical synthesis , Caspase Inhibitors/pharmacology , Cations/chemistry , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Gallic Acid/pharmacology , Humans , L-Lactate Dehydrogenase/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondrial Swelling/drug effects , NADP/metabolism , Oxidation-Reduction , Oxygen Consumption/drug effects , Reproducibility of Results , Uncoupling Agents/chemical synthesis , Uncoupling Agents/pharmacology
17.
Diabetes ; 63(1): 75-88, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24009260

ABSTRACT

Insulin regulates heart metabolism through the regulation of insulin-stimulated glucose uptake. Studies have indicated that insulin can also regulate mitochondrial function. Relevant to this idea, mitochondrial function is impaired in diabetic individuals. Furthermore, the expression of Opa-1 and mitofusins, proteins of the mitochondrial fusion machinery, is dramatically altered in obese and insulin-resistant patients. Given the role of insulin in the control of cardiac energetics, the goal of this study was to investigate whether insulin affects mitochondrial dynamics in cardiomyocytes. Confocal microscopy and the mitochondrial dye MitoTracker Green were used to obtain three-dimensional images of the mitochondrial network in cardiomyocytes and L6 skeletal muscle cells in culture. Three hours of insulin treatment increased Opa-1 protein levels, promoted mitochondrial fusion, increased mitochondrial membrane potential, and elevated both intracellular ATP levels and oxygen consumption in cardiomyocytes in vitro and in vivo. Consequently, the silencing of Opa-1 or Mfn2 prevented all the metabolic effects triggered by insulin. We also provide evidence indicating that insulin increases mitochondrial function in cardiomyocytes through the Akt-mTOR-NFκB signaling pathway. These data demonstrate for the first time in our knowledge that insulin acutely regulates mitochondrial metabolism in cardiomyocytes through a mechanism that depends on increased mitochondrial fusion, Opa-1, and the Akt-mTOR-NFκB pathway.


Subject(s)
Insulin/pharmacology , Mitochondria/metabolism , Mitochondrial Dynamics/physiology , Myocytes, Cardiac/metabolism , Signal Transduction/physiology , Animals , Cell Line , Cells, Cultured , GTP Phosphohydrolases/metabolism , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
18.
Toxicol Appl Pharmacol ; 272(2): 356-64, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23777606

ABSTRACT

Changes in mitochondrial ATP synthesis can affect the function of tumor cells due to the dependence of the first step of glycolysis on mitochondrial ATP. The oxidative phosphorylation (OXPHOS) system is responsible for the synthesis of approximately 90% of the ATP in normal cells and up to 50% in most glycolytic cancers; therefore, inhibition of the electron transport chain (ETC) emerges as an attractive therapeutic target. We studied the effect of a lipophilic isoprenylated catechol, 3-hydroxybakuchiol (3-OHbk), a putative ETC inhibitor isolated from Psoralea glandulosa. 3-OHbk exerted cytotoxic and anti-proliferative effects on the TA3/Ha mouse mammary adenocarcinoma cell line and induced a decrease in the mitochondrial transmembrane potential, the activation of caspase-3, the opening of the mitochondrial permeability transport pore (MPTP) and nuclear DNA fragmentation. Additionally, 3-OHbk inhibited oxygen consumption, an effect that was completely reversed by succinate (an electron donor for Complex II) and duroquinol (electron donor for Complex III), suggesting that 3-OHbk disrupted the electron flow at the level of Complex I. The inhibition of OXPHOS did not increase the level of reactive oxygen species (ROS) but caused a large decrease in the intracellular ATP level. ETC inhibitors have been shown to induce cell death through necrosis and apoptosis by increasing ROS generation. Nevertheless, we demonstrated that 3-OHbk inhibited the ETC and induced apoptosis through an interaction with Complex I. By delivering electrons directly to Complex III with duroquinol, cell death was almost completely abrogated. These results suggest that 3-OHbk has antitumor activity resulting from interactions with the ETC, a system that is already deficient in cancer cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Catechols/pharmacology , Electron Transport/drug effects , Mitochondria/drug effects , Phenols/pharmacology , Animals , Antineoplastic Agents, Phytogenic/chemistry , Catechols/chemistry , Cell Culture Techniques , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Swelling/drug effects , Molecular Structure , Necrosis , Phenols/chemistry , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...