Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Exp Cell Res ; 356(2): 217-222, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28327410

ABSTRACT

There are numerous mechanisms by which mammals respond to hypoxia. These include acute changes in pulmonary arterial tone due to smooth muscle cell contraction, acute increases in respiration triggered by the carotid body chemosensory cells, and chronic changes such as induction of red blood cell proliferation and angiogenesis by hypoxia inducible factor targets erythropoietin and vascular endothelial growth factor, respectively. Mitochondria account for the majority of oxygen consumption in the cell and have recently been appreciated to serve as signaling organelles required for the initiation or propagation of numerous homeostatic mechanisms. Mitochondria can influence cell signaling by production of reactive oxygen species and metabolites. Here we review recent evidence that mitochondrial signals can imitate acute and chronic hypoxia responses.


Subject(s)
Cell Hypoxia/physiology , Homeostasis/physiology , Hypoxia/metabolism , Mitochondria/metabolism , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Animals , Humans
2.
Cell Death Differ ; 22(1): 58-73, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25236395

ABSTRACT

Cells exposed to extreme physicochemical or mechanical stimuli die in an uncontrollable manner, as a result of their immediate structural breakdown. Such an unavoidable variant of cellular demise is generally referred to as 'accidental cell death' (ACD). In most settings, however, cell death is initiated by a genetically encoded apparatus, correlating with the fact that its course can be altered by pharmacologic or genetic interventions. 'Regulated cell death' (RCD) can occur as part of physiologic programs or can be activated once adaptive responses to perturbations of the extracellular or intracellular microenvironment fail. The biochemical phenomena that accompany RCD may be harnessed to classify it into a few subtypes, which often (but not always) exhibit stereotyped morphologic features. Nonetheless, efficiently inhibiting the processes that are commonly thought to cause RCD, such as the activation of executioner caspases in the course of apoptosis, does not exert true cytoprotective effects in the mammalian system, but simply alters the kinetics of cellular demise as it shifts its morphologic and biochemical correlates. Conversely, bona fide cytoprotection can be achieved by inhibiting the transduction of lethal signals in the early phases of the process, when adaptive responses are still operational. Thus, the mechanisms that truly execute RCD may be less understood, less inhibitable and perhaps more homogeneous than previously thought. Here, the Nomenclature Committee on Cell Death formulates a set of recommendations to help scientists and researchers to discriminate between essential and accessory aspects of cell death.


Subject(s)
Apoptosis , Signal Transduction , Animals , Humans , Terminology as Topic
4.
Cell Death Differ ; 15(4): 660-6, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18219320

ABSTRACT

Decreases in oxygen levels are observed in physiological processes, such as development, and pathological situations, such as tumorigenesis and ischemia. In the complete absence of oxygen (anoxia), mammalian cells are unable to generate sufficient energy for survival, so a mechanism for sensing a decrease in the oxygen level (hypoxia) before it reaches a critical point is crucial for the survival of the organism. In response to decreased oxygen levels, cells activate the transcription factors hypoxia-inducible factors (HIFs), which lead to metabolic adaptation to hypoxia, as well as to generate new vasculature to increase oxygen supply. How cells sense decreases in oxygen levels to regulate HIF activation has been hotly debated. Emerging evidence indicates that reactive oxygen species (ROS) generated by mitochondrial complex III are required for hypoxic activation of HIF. This review examines the current knowledge about the role of mitochondrial ROS in HIF activation, as well as implications of ROS-level regulation in pathological processes such as cancer.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Electron Transport Complex III/metabolism , Hypoxia/metabolism , Mitochondria/metabolism , Protein Processing, Post-Translational , Adaptation, Physiological , Animals , Cell Hypoxia , Cell Transformation, Neoplastic/metabolism , Humans , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mitochondria/enzymology , Neoplasms/metabolism , Reactive Oxygen Species/metabolism
5.
Exp Cell Res ; 294(1): 281-9, 2004 Mar 10.
Article in English | MEDLINE | ID: mdl-14980521

ABSTRACT

The alpha4 laminin subunit is a major structural component of assembling basement membranes of endothelial cells. We have been investigating its functions with regard to endothelial cell survival. An anti-laminin alpha4 antibody (2A3), directed against the G domain of the alpha4 laminin subunit of laminins-8 and -9, inhibits proliferation and enhances apoptosis of endothelial cells when cells are maintained in vitro. Activation of caspases-9 and -3 plays a role in 2A3 antibody-induced apoptosis, since inhibitors specific for these caspases and overexpression of the anti-apoptotic protein Bcl-X(L), but not c-FLIP, inhibit 2A3 antibody-triggered endothelial cell death. Extracellular matrix is known to play a role in regulating programmed cell death in an integrin-dependent fashion. The alpha4 laminin subunit conforms to this idea since activation of beta1 integrin subunits on endothelial cells blocks the ability of 2A3 antibody to induce endothelial cell death. In summary, our data indicate that complexes composed of alpha4 laminin/beta1 subunit-containing integrins at the cell surface support endothelial cell survival. Furthermore, we propose that antagonists of alpha4 laminin function, including antibody 2A3, have value as angiogenesis inhibitors in a clinical setting where blocking aberrant growth of blood vessel by triggering apoptosis of endothelial cells may be therapeutic.


Subject(s)
Endothelium, Vascular/cytology , Laminin/physiology , Antibodies/pharmacology , Apoptosis , Caspases/physiology , Cell Line , Cell Survival , Endothelium, Vascular/drug effects , Humans , Integrins/physiology , Laminin/antagonists & inhibitors , Laminin/immunology , Protein Subunits/antagonists & inhibitors , Protein Subunits/physiology
6.
Apoptosis ; 7(6): 475-82, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12370489

ABSTRACT

Mammalian cells have multiple responses to low or zero oxygen concentrations. In the complete absence of oxygen, cells undergo cell death through apoptosis, and not necrosis. Apoptotic signaling during oxygen deprivation occurs through the release of cytochrome c and apaf-1 mediated caspase-9 activation. The upstream regulators of cytochrome c release are the Bcl-2 family members. Pro-apoptotic Bcl-2 family members such as bax or bak are clearly required to initiate cytochrome c/apaf-1/caspase-9 mediated cell death during oxygen deprivation. Here we review what is currently known oxygen deprivation induced cell death and speculate about initiating mechanisms resulting in the activation of pro-apoptotic Bcl-2 family members.


Subject(s)
Apoptosis/physiology , Cell Hypoxia/physiology , Proto-Oncogene Proteins c-bcl-2 , Animals , Apoptosis/genetics , Caspase 9 , Caspases/metabolism , Cell Hypoxia/genetics , Cytochrome c Group/metabolism , Electron Transport , Humans , Membrane Proteins/metabolism , Mitochondria/metabolism , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Transcription, Genetic , bcl-2 Homologous Antagonist-Killer Protein , bcl-2-Associated X Protein
7.
J Biol Chem ; 276(46): 42728-36, 2001 Nov 16.
Article in English | MEDLINE | ID: mdl-11559697

ABSTRACT

Members of the TNFR (tumor necrosis factor receptor) superfamily are involved in regulating activation and differentiation of cells as well as cell survival and programmed cell death/apoptosis. Multimerization of TNFRs can lead to recruitment of TRAFs (TNFR-associated factors) by the receptors resulting in activation of kinases and transcription factors, such as c-Jun N-terminal kinase and nuclear factor kappaB (NF-kappaB). Signal transduction triggered by TNF-alpha also induces an increase in intracellular reactive oxygen species (ROS). ROS have been suggested to play a role in NF-kappaB activation, which is thought to promote cell survival. However, oxidation of proteins and lipids by ROS can also result in apoptosis. The processes generating intracellular ROS and the mechanism(s) regulating the cellular redox status have not been fully elucidated. We investigated whether TRAFs play a role in controlling intracellular ROS levels. Our results indicate that recruitment of TRAFs to the plasma membrane of human embryonic kidney (HEK) 293 cells is crucial for activation of signaling pathways, which regulate ROS production in mitochondria. TRAF-mediated changes in ROS levels enhanced NF-kappaB activation but were not dependent on NF-kappaB-inducing kinase. Consistent with its anti-apoptotic function, Bcl-x(L) interfered with TRAF-mediated ROS generation but not NF-kappaB activation. Taken together, our results suggest a novel role of TRAFs in signal transduction pathways triggered by TNFR-related proteins, which balance cell survival and apoptosis by regulating the electron transport in mitochondria.


Subject(s)
Bacterial Proteins/metabolism , Reactive Oxygen Species , Signal Transduction , Apoptosis , Blotting, Western , CD28 Antigens/metabolism , Cell Line , Cell Membrane , Cell Survival , Cytoplasm/metabolism , Electron Transport , Enzyme Activation , Humans , JNK Mitogen-Activated Protein Kinases , Ki-1 Antigen/metabolism , Luciferases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , NF-kappa B/metabolism , Oxidation-Reduction , Oxygen/metabolism , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Subcellular Fractions , Transfection , bcl-X Protein
9.
Circ Res ; 88(12): 1259-66, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11420302

ABSTRACT

We tested whether mitochondria function as the O(2) sensor underlying hypoxic pulmonary vasoconstriction (HPV). In buffer-perfused rat lungs, rotenone, myxothiazol, and diphenyleneiodonium, which inhibit mitochondria in the proximal region of the electron transport chain (ETC), abolished HPV without attenuating the response to U46619. Cyanide and antimycin A inhibit electron transfer in the distal region of the ETC, but they did not abolish HPV. Cultured pulmonary artery (PA) myocytes contract in response to hypoxia or to U46619. The hypoxic response was abolished while the response to U46619 was maintained in mutant (rho(0)) PA myocytes lacking a mitochondrial ETC. To test whether reactive oxygen species (ROS) derived from mitochondria act as signaling agents in HPV, the antioxidants pyrrolidinedithiocarbamate and ebselen and the Cu,Zn superoxide dismutase inhibitor diethyldithiocarbamate were used. These abolished HPV without affecting contraction to U46619, suggesting that ROS act as second messengers. In cultured PA myocytes, oxidation of intracellular 2',7'-dichlorofluorescin diacetate (DCFH) dye increased under 2% O(2), indicating that myocytes increase their generation of H(2)O(2) during hypoxia. This was attenuated by myxothiazol, implicating mitochondria as the source of increased ROS during HPV. These results indicate that mitochondrial ATP is not required for HPV, that mitochondria function as O(2) sensors during hypoxia, and that ROS generated in the proximal region of the ETC act as second messengers in the response.


Subject(s)
Hypoxia/metabolism , Lung/blood supply , Mitochondria/metabolism , Oxygen/metabolism , Vasoconstriction/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Antimycin A/pharmacology , Antioxidants/pharmacology , Cells, Cultured , Electron Transport/drug effects , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Ion Channels/drug effects , Methacrylates , Mitochondria/drug effects , Models, Biological , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Onium Compounds/pharmacology , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Rotenone/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Thiazoles/pharmacology , Uncoupling Agents/pharmacology , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology
11.
Oncogene ; 19(34): 3840-8, 2000 Aug 10.
Article in English | MEDLINE | ID: mdl-10951577

ABSTRACT

The transcription factor p53 can induce growth arrest or death in cells. Tumor cells that develop mutations in p53 demonstrate a diminished apoptotic potential, which may contribute to growth and tumor metastasis. Cellular levels of p53 are stabilized during hypoxia. The present study tested the hypothesis that reactive oxygen species (ROS) released from mitochondria regulate the cytosolic redox state and are required for the stabilization of p53 protein levels in response to hypoxia. Our results indicate that hypoxia (1.5% O2) increases mitochondrial ROS generation and increases p53 protein levels in human breast carcinoma MCF-7 cells and in normal human diploid fibroblast IMR-90 cells. MCF-7 cells depleted of their mitochondrial DNA (rho(o) cells) failed to stabilize p53 protein levels during hypoxia. The antioxidant N-acetylcysteine and the Cu/Zn superoxide dismutase inhibitor diethyldithiocarbamic acid abolished the hypoxia-induced increases in ROS and p53 levels. Rotenone, an inhibitor of mitochondrial complex I, and 4,4'-diisothiocyanato-stilbene-2,2'-disulfonate, a mitochondrial anion channel inhibitor, also abolished the increase in ROS signal and p53 levels during hypoxia. The p53-dependent gene p21WAF1/CIP1 was also induced by hypoxia in both MCF-7 and IMR-90 cells without affecting the growth rate of either cell line. In contrast, both cell lines exhibited increases in p21WAF1/CIP1 expression and growth arrest after gamma irradiation. Primary chick cardiac myocytes and murine embryonic fibroblasts also showed an increase in p53 protein levels in response to hypoxia without cell death or growth arrest. These results indicate that mitochondria regulate p53 protein levels during hypoxia through a redox-dependent mechanism involving ROS. Despite p53-induction, hypoxia alone does not cause either growth arrest or cell death.


Subject(s)
Cell Hypoxia , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Breast Neoplasms/metabolism , Carcinoma/metabolism , Cell Cycle/radiation effects , Cell Death/drug effects , Cell Death/radiation effects , Cell Division/drug effects , Cell Division/radiation effects , Cells, Cultured , Chick Embryo , Cobalt/pharmacology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/metabolism , Ditiocarb/pharmacology , Fibroblasts , Fluoresceins/analysis , Fluoresceins/metabolism , Humans , Hydrogen Peroxide/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Myocardium/cytology , Rotenone/pharmacology , Superoxide Dismutase/antagonists & inhibitors , Tumor Cells, Cultured , Uncoupling Agents/pharmacology
12.
J Immunol ; 165(2): 1013-21, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10878378

ABSTRACT

The transcription factor NF-kappa B stimulates the transcription of proinflammatory cytokines including TNF-alpha. LPS (endotoxin) and hypoxia both induce NF-kappa B activation and TNF-alpha gene transcription. Furthermore, hypoxia augments LPS induction of TNF-alpha mRNA. Previous reports have indicated that antioxidants abolish NF-kappa B activation in response to LPS or hypoxia, which suggests that reactive oxygen species (ROS) are involved in NF-kappa B activation. This study tested whether mitochondrial ROS are required for both NF-kappaB activation and the increase in TNF-alpha mRNA levels during hypoxia and LPS. Our results indicate that hypoxia (1.5% O2) stimulates NF-kappa B and TNF-alpha gene transcription and increases ROS generation as measured by the oxidant sensitive dye 2',7'-dichlorofluorescein diacetate in murine macrophage J774.1 cells. The antioxidants N-acetylcysteine and pyrrolidinedithiocarbamic acid abolished the hypoxic activation of NF-kappa B, TNF-alpha gene transcription, and increases in ROS levels. Rotenone, an inhibitor of mitochondrial complex I, abolished the increase in ROS signal, the activation of NF-kappa B, and TNF-alpha gene transcription during hypoxia. LPS stimulated NF-kappa B and TNF-alpha gene transcription but not ROS generation in J774.1 cells. Rotenone, pyrrolidinedithiocarbamic acid, and N-acetylcysteine had no effect on the LPS stimulation of NF-kappa B and TNF-alpha gene transcription, indicating that LPS activates NF-kappa B and TNF-alpha gene transcription through a ROS-independent mechanism. These results indicate that mitochondrial ROS are required for the hypoxic activation of NF-kappa B and TNF-alpha gene transcription, but not for the LPS activation of NF-kappa B.


Subject(s)
Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Oxidants/physiology , Transcription, Genetic/immunology , Tumor Necrosis Factor-alpha/genetics , Animals , Cell Hypoxia/genetics , Cell Hypoxia/immunology , Cell Line , DNA/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mitochondria/metabolism , Protein Binding/immunology , Protein Biosynthesis , RNA, Messenger/biosynthesis , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolism
13.
J Biol Chem ; 275(33): 25130-8, 2000 Aug 18.
Article in English | MEDLINE | ID: mdl-10833514

ABSTRACT

During hypoxia, hypoxia-inducible factor-1alpha (HIF-1alpha) is required for induction of a variety of genes including erythropoietin and vascular endothelial growth factor. Hypoxia increases mitochondrial reactive oxygen species (ROS) generation at Complex III, which causes accumulation of HIF-1alpha protein responsible for initiating expression of a luciferase reporter construct under the control of a hypoxic response element. This response is lost in cells depleted of mitochondrial DNA (rho(0) cells). Overexpression of catalase abolishes hypoxic response element-luciferase expression during hypoxia. Exogenous H(2)O(2) stabilizes HIF-1alpha protein during normoxia and activates luciferase expression in wild-type and rho(0) cells. Isolated mitochondria increase ROS generation during hypoxia, as does the bacterium Paracoccus denitrificans. These findings reveal that mitochondria-derived ROS are both required and sufficient to initiate HIF-1alpha stabilization during hypoxia.


Subject(s)
DNA-Binding Proteins/metabolism , Electron Transport Complex III/chemistry , Electron Transport Complex III/metabolism , Hypoxia , Mitochondria/metabolism , Nuclear Proteins/metabolism , Reactive Oxygen Species/metabolism , Transcription Factors , Androstadienes/pharmacology , Animals , Cell Line , Cell Nucleus/metabolism , Chelating Agents/pharmacology , Cobalt/pharmacology , Cytosol/chemistry , Deferoxamine/pharmacology , Dose-Response Relationship, Drug , Electron Transport Complex IV/metabolism , Enzyme Inhibitors/pharmacology , Genes, Reporter , Humans , Hydrogen Peroxide/metabolism , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Immunoblotting , Marine Toxins , Mitochondria/enzymology , Mitochondria, Liver/metabolism , Models, Biological , Oxazoles/pharmacology , Oxidation-Reduction , Oxygen/metabolism , Paracoccus denitrificans/metabolism , Rats , Time Factors , Transfection , Tumor Cells, Cultured , Wortmannin
14.
J Appl Physiol (1985) ; 88(5): 1880-9, 2000 May.
Article in English | MEDLINE | ID: mdl-10797153

ABSTRACT

Hypoxia elicits a variety of adaptive responses at the tissue level, at the cellular level, and at the molecular level. A physiological response to hypoxia requires the existence of an O(2) sensor coupled to a signal transduction system, which in turn activates the functional response. Although much has been learned about the signaling systems activated by hypoxia, no consensus exists regarding the nature of the underlying O(2) sensor or whether multiple sensors exist. Among previously considered mechanisms, heme proteins have been suggested to undergo allosteric modification in response to O(2) binding or release at different PO(2) levels. Other studies suggest that ion channels may change conductance as a function of PO(2), allowing them to signal the onset of hypoxia. Still other studies suggest that NADPH oxidase may decrease its generation of reactive O(2) species (ROS) during hypoxia. Recent data suggest that mitochondria may function as O(2) sensors by increasing their generation of ROS during hypoxia. These oxidant signals appear to act as second messengers in the adaptive responses to hypoxia in a variety of cell types. Such observations contribute to a growing awareness that mitochondria do more than just generate ATP, in that they initiate signaling cascades involved in adaptive responses to hypoxia and that they participate in the control of cell death pathways.


Subject(s)
Mitochondria/physiology , Oxygen/metabolism , Animals , Hemeproteins/physiology , Humans , Ion Channels/physiology , Mitochondria/metabolism , Models, Biological , NADP/physiology , Reactive Oxygen Species/physiology
15.
Proc Natl Acad Sci U S A ; 97(9): 4666-71, 2000 Apr 25.
Article in English | MEDLINE | ID: mdl-10781072

ABSTRACT

Coupled cellular respiration requires that ATP and ADP be efficiently exchanged between the cytosol and the mitochondrial matrix. When growth factors are withdrawn from dependent cells, metabolism is disrupted by a defect in ATP/ADP exchange across the mitochondrial membranes. Unexpectedly, we find that this defect results from loss of outer mitochondrial membrane permeability to metabolic anions. This decrease in anion permeability correlates with the changes in conductance properties that accompany closure of the voltage-dependent anion channel (also known as mitochondrial porin). Loss of outer membrane permeability (i) results in the accumulation of stored metabolic energy within the intermembrane space in the form of creatine phosphate, (ii) is prevented by the outer mitochondrial membrane proteins Bcl-x(L) and Bcl-2, and (iii) can be reversed by growth factor readdition. If outer membrane impermeability persists, the disruption of mitochondrial homeostasis culminates in loss of outer mitochondrial membrane integrity, cytochrome c redistribution, and apoptosis. The recognition that outer membrane permeability is regulated under physiological conditions has important implications for the understanding of bioenergetics and cell survival.


Subject(s)
Cell Survival , Intracellular Membranes/physiology , Mitochondria/metabolism , Oxygen Consumption , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis , Cell Division , Cell Line , Creatine Kinase/metabolism , Kinetics , Mice , Permeability , Phosphocreatine/metabolism , Porins/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Voltage-Dependent Anion Channels , bcl-X Protein
16.
Am J Physiol ; 277(5): L1057-65, 1999 11.
Article in English | MEDLINE | ID: mdl-10564193

ABSTRACT

Prolonged hypoxia produces reversible changes in endothelial permeability, but the mechanisms involved are not fully known. Previous studies have implicated reactive oxygen species (ROS) and cytokines in the regulation of permeability. We tested whether prolonged hypoxia alters permeability to increasing ROS generation, which amplifies cytokine production. Human umbilical vein endothelial cell (HUVEC) monolayers were exposed to hypoxia while secretion of tumor necrosis factor-alpha (TNF-alpha), interleukin (IL)-1alpha, IL-6, and IL-8 was measured. IL-6 and IL-8 secretion increased fourfold over 24 h in a pattern corresponding to changes in HUVEC permeability measured by transendothelial electrical resistance (TEER). Addition of exogenous IL-6 to normoxic HUVEC monolayers caused time-dependent changes in TEER that mimicked the hypoxic response. An antibody to IL-6 significantly attenuated the hypoxia-induced changes in TEER (86 +/- 4 vs. 63 +/- 3% with hypoxia alone at 18 h), whereas treatment with anti-IL-8 had no effect. To determine the role of hypoxia-induced ROS on this response, HUVEC monolayers were incubated with the antioxidants ebselen (50 microM) and N-acetyl-L-cysteine (NAC, 1 mM) before hypoxia. Antioxidants attenuated hypoxia-induced IL-6 secretion (13 +/- 2 pg/ml with ebselen and 19 +/- 3 pg/ml with NAC vs. 140 +/- 15 pg/ml with hypoxia). Ebselen and NAC prevented changes in TEER during hypoxia (94 +/- 2% with ebselen and 90 +/- 6% with NAC vs. 63 +/- 3% with hypoxia at 18 h). N-nitro-L-arginine (500 microM) did not decrease hypoxia-induced changes in dichlorofluorescin fluorescence, IL-6 secretion, or TEER. Thus ROS generated during hypoxia act as signaling elements, regulating secretion of the proinflammatory cytokines that lead to alterations of endothelial permeability.


Subject(s)
Endothelium, Vascular/metabolism , Hypoxia/metabolism , Interleukin-6/biosynthesis , Reactive Oxygen Species/metabolism , Signal Transduction/immunology , Antibodies/pharmacology , Antioxidants/pharmacology , Azoles/pharmacology , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/immunology , Cells, Cultured , Electric Impedance , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Humans , Hydrogen Peroxide/pharmacology , Hypoxia/immunology , Interleukin-6/immunology , Isoindoles , Organoselenium Compounds/pharmacology , Oxidants/pharmacology , Oxygen/pharmacology , Umbilical Veins/cytology
17.
FEBS Lett ; 454(3): 173-6, 1999 Jul 09.
Article in English | MEDLINE | ID: mdl-10431801

ABSTRACT

A resurgence of interest in mitochondrial physiology has recently developed as a result of new experimental data demonstrating that mitochondria function as important participants in a diverse collection of novel intracellular signaling pathways. Cells depleted of mitochondrial DNA, or rho0 cells, lack critical respiratory chain catalytic subunits that are encoded in the mitochondrial genome. Although rho0 cells contain petit mitochondria, they cannot support normal oxidative phosphorylation and must survive and replicate using ATP derived solely from glycolysis. Without a functional electron transport chain, rho0 cells cannot normally regulate redox potential and their mitochondria appear to be incapable of generating reactive oxygen species. Emerging evidence suggests that these signals are important components in a number of mitochondria-initiated signaling pathways. The present article focuses on how rho0 cells have contributed to an understanding of the role that mitochondria play in distinct physiological pathways involved with apoptosis, glucose-induced insulin secretion, and oxygen sensing.


Subject(s)
DNA, Mitochondrial/physiology , Mitochondria/physiology , Animals , Apoptosis , Humans , Reactive Oxygen Species/physiology , Signal Transduction/physiology
18.
Mol Cell ; 3(2): 159-67, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10078199

ABSTRACT

Growth factor withdrawal is associated with a metabolic arrest that can result in apoptosis. Cell death is preceded by loss of outer mitochondrial membrane integrity and cytochrome c release. These mitochondrial events appear to follow a relative increase in mitochondrial membrane potential. This change in membrane potential results from the failure of the adenine nucleotide translocator (ANT)/voltage-dependent anion channel (VDAC) complex to maintain ATP/ADP exchange. Bcl-xL expression allows growth factor-deprived cells to maintain sufficient mitochondrial ATP/ADP exchange to sustain coupled respiration. These data demonstrate that mitochondrial adenylate transport is under active regulation. Efficient exchange of ADP for ATP is promoted by Bcl-xL expression permitting oxidative phosphorylation to be regulated by cellular ATP/ADP levels and allowing mitochondria to adapt to changes in metabolic demand.


Subject(s)
Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Apoptosis/physiology , Interleukin-3/pharmacology , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/physiology , Animals , Biological Transport, Active , Caspase Inhibitors , Fluorescent Dyes , Gramicidin/pharmacology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Membrane Potentials , Mice , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Porins/physiology , Proto-Oncogene Proteins c-bcl-2/genetics , Recombinant Fusion Proteins/physiology , Rhodamine 123 , Sodium-Potassium-Exchanging ATPase/metabolism , Transfection , Tumor Cells, Cultured/drug effects , Voltage-Dependent Anion Channels , bcl-X Protein
20.
Proc Natl Acad Sci U S A ; 95(20): 11715-20, 1998 Sep 29.
Article in English | MEDLINE | ID: mdl-9751731

ABSTRACT

Transcriptional activation of erythropoietin, glycolytic enzymes, and vascular endothelial growth factor occurs during hypoxia or in response to cobalt chloride (CoCl2) in Hep3B cells. However, neither the mechanism of cellular O2 sensing nor that of cobalt is fully understood. We tested whether mitochondria act as O2 sensors during hypoxia and whether hypoxia and cobalt activate transcription by increasing generation of reactive oxygen species (ROS). Results show (i) wild-type Hep3B cells increase ROS generation during hypoxia (1. 5% O2) or CoCl2 incubation, (ii) Hep3B cells depleted of mitochondrial DNA (rho0 cells) fail to respire, fail to activate mRNA for erythropoietin, glycolytic enzymes, or vascular endothelial growth factor during hypoxia, and fail to increase ROS generation during hypoxia; (iii) rho0 cells increase ROS generation in response to CoCl2 and retain the ability to induce expression of these genes; and (iv) the antioxidants pyrrolidine dithiocarbamate and ebselen abolish transcriptional activation of these genes during hypoxia or CoCl2 in wild-type cells, and abolish the response to CoCl2 in rho degrees cells. Thus, hypoxia activates transcription via a mitochondria-dependent signaling process involving increased ROS, whereas CoCl2 activates transcription by stimulating ROS generation via a mitochondria-independent mechanism.


Subject(s)
Cell Hypoxia/genetics , Cell Hypoxia/physiology , Mitochondria, Liver/metabolism , Reactive Oxygen Species/metabolism , Transcription Factors , Transcriptional Activation , Adenosine Triphosphate/metabolism , Antioxidants/pharmacology , Azoles/pharmacology , Cell Line , Cobalt/pharmacology , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/metabolism , Endothelial Growth Factors/genetics , Erythropoietin/genetics , Glycolysis/genetics , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Isoindoles , Lymphokines/genetics , Mitochondria, Liver/drug effects , Nuclear Proteins/metabolism , Organoselenium Compounds/pharmacology , Pyrrolidines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thiocarbamates/pharmacology , Transcriptional Activation/drug effects , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...