Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Biochim Biophys Acta Bioenerg ; 1858(7): 519-528, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28442264

ABSTRACT

The mitochondrial ATP dependent matrix protease, Lon, is involved in the maintenance of mitochondrial DNA nucleoids and degradation of abnormal or misfolded proteins. The Lon protease regulates mitochondrial Tfam (mitochondrial transcription factor A) level and thus modulates mitochondrial DNA (mtDNA) content. We have previously shown that hypoxic stress induces the PKA-dependent phosphorylation of cytochrome c oxidase (CcO) subunits I, IVi1, and Vb and a time-dependent reduction of these subunits in RAW 264.7 murine macrophages subjected to hypoxia and rabbit hearts subjected to ischemia/reperfusion. Here, we show that Lon is involved in the preferential turnover of phosphorylated CcO subunits under hypoxic/ischemic stress. Induction of Lon protease occurs at 6 to 12 h of hypoxia and this increase coincides with lower CcO subunit contents. Over-expression of flag-tagged wild type and phosphorylation site mutant Vb and IVi1 subunits (S40A and T52A, respectively) caused marked degradation of wild type protein under hypoxia while the mutant proteins were relatively resistant. Furthermore, the recombinant purified Lon protease degraded the phosphorylated IVi1 and Vb subunits, while the phosphorylation-site mutant proteins were resistant to degradation. 3D structural modeling shows that the phosphorylation sites are exposed to the matrix compartment, accessible to matrix PKA and Lon protease. Hypoxic stress did not alter CcO subunit levels in Lon depleted cells, confirming its role in CcO turnover. Our results therefore suggest that Lon preferentially degrades the phosphorylated subunits of CcO and plays a role in the regulation of CcO activity in hypoxia and ischemia/reperfusion injury.


Subject(s)
ATP-Dependent Proteases/metabolism , Cell Hypoxia/physiology , Electron Transport Complex IV/metabolism , Mitochondria, Heart/enzymology , Mitochondrial Proteins/metabolism , Myocardial Ischemia/enzymology , ATP-Dependent Proteases/chemistry , ATP-Dependent Proteases/genetics , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Male , Mice , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Models, Molecular , Phosphorylation , Protein Conformation , Protein Processing, Post-Translational , Protein Subunits , RAW 264.7 Cells , RNA Interference , RNA, Small Interfering/genetics , Rabbits , Recombinant Proteins/metabolism
2.
J Biol Chem ; 289(14): 9936-51, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24497629

ABSTRACT

We report that polycyclic aromatic hydrocarbon (PAH)-inducible CYP1B1 is targeted to mitochondria by sequence-specific cleavage at the N terminus by a cytosolic Ser protease (polyserase 1) to activate the cryptic internal signal. Site-directed mutagenesis, COS-7 cell transfection, and in vitro import studies in isolated mitochondria showed that a positively charged domain at residues 41-48 of human CYP1B1 is part of the mitochondrial (mt) import signal. Ala scanning mutations showed that the Ser protease cleavage site resides between residues 37 and 41 of human CYP1B1. Benzo[a]pyrene (BaP) treatment induced oxidative stress, mitochondrial respiratory defects, and mtDNA damage that was attenuated by a CYP1B1-specific inhibitor, 2,3,4,5-tetramethoxystilbene. In support, the mitochondrial CYP1B1 supported by mitochondrial ferredoxin (adrenodoxin) and ferredoxin reductase showed high aryl hydrocarbon hydroxylase activity. Administration of benzo[a]pyrene or 2,3,7,8-tetrachlorodibenzodioxin induced similar mitochondrial functional abnormalities and oxidative stress in the lungs of wild-type mice and Cyp1a1/1a2-null mice, but the effects were markedly blunted in Cyp1b1-null mice. These results confirm a role for CYP1B1 in inducing PAH-mediated mitochondrial dysfunction. The role of mitochondrial CYP1B1 was assessed using A549 lung epithelial cells stably expressing shRNA against NADPH-cytochrome P450 oxidoreductase or mitochondrial adrenodoxin. Our results not only show conservation of the endoprotease cleavage mechanism for mitochondrial import of family 1 CYPs but also reveal a direct role for mitochondrial CYP1B1 in PAH-mediated oxidative and chemical damage to mitochondria.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Benzo(a)pyrene/adverse effects , Mitochondria/enzymology , Oxygen Consumption/drug effects , Polychlorinated Dibenzodioxins/adverse effects , Protein Sorting Signals , Teratogens , Adrenodoxin/genetics , Adrenodoxin/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Benzo(a)pyrene/pharmacology , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Cytochrome P-450 CYP1B1 , Female , Humans , Male , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/pathology , Mutagenesis , Oxidation-Reduction/drug effects , Polychlorinated Dibenzodioxins/pharmacology , Protein Transport/drug effects
3.
J Biol Chem ; 288(18): 12627-44, 2013 May 03.
Article in English | MEDLINE | ID: mdl-23471973

ABSTRACT

Human polymorphisms in the 5'-upstream regulatory regions and also protein coding regions of cytochrome P450 2E1 (CYP2E1) are known to be associated with several diseases, including cancer and alcohol liver toxicity. In this study, we report novel mutations in the N-terminal protein targeting regions of CYP2E1 that markedly affect subcellular localization of the protein. Variant W23R/W30R protein (termed W23/30R) is preferentially targeted to mitochondria but very poorly to the endoplasmic reticulum, whereas the L32N protein is preferentially targeted to the endoplasmic reticulum and poorly to mitochondria. These results explain the physiological significance of bimodal CYP targeting to the endoplasmic reticulum and mitochondria previously described. COS-7 cells and HepG2 cells stably expressing W23/30R mutations showed markedly increased alcohol toxicity in terms of increased production of reactive oxygen species, respiratory dysfunction, and loss of cytochrome c oxidase subunits and activity. Stable cells expressing the L32N variant, on the other hand, were relatively less responsive to alcohol-induced toxicity and mitochondrial dysfunction. These results further support our previous data, based on mutational studies involving altered targeting, indicating that mitochondria-targeted CYP2E1 plays an important role in alcohol liver toxicity. The results also provide an interesting new link to genetic variations affecting subcellular distribution of CYP2E1 with alcohol-induced toxicity.


Subject(s)
Central Nervous System Depressants/adverse effects , Cytochrome P-450 CYP2E1/metabolism , Ethanol/adverse effects , Liver/enzymology , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Models, Biological , Mutation, Missense , Amino Acid Substitution , Animals , COS Cells , Central Nervous System Depressants/pharmacology , Chlorocebus aethiops , Cytochrome P-450 CYP2E1/genetics , Ethanol/pharmacology , Hep G2 Cells , Humans , Liver/pathology , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/genetics , Oxygen Consumption/drug effects , Oxygen Consumption/genetics , Protein Transport/drug effects , Protein Transport/genetics
4.
J Biol Chem ; 285(32): 24609-19, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20529841

ABSTRACT

The ethanol-inducible cytochrome P450 2E1 (CYP2E1) is also induced under different pathological and physiological conditions. Studies including ours have shown that CYP2E1 is bimodally targeted to both the endoplasmic reticulum (microsomes) (mc CYP2E1) and mitochondria (mt CYP2E1). In this study we investigated the role of mtCYP2E1 in ethanol-mediated oxidative stress in stable cell lines expressing predominantly mt CYP2E1 or mc CYP2E1. The ER+ mutation (A2L, A9L), which increases the affinity of the nascent protein for binding to the signal recognition particle, preferentially targets CYP2E1 to the endoplasmic reticulum. The Mt+ (L17G) and Mt++ (I8R, L11R, L17R) mutant proteins, showing progressively lower affinity for signal recognition particle binding, were targeted to mitochondria at correspondingly higher levels. The rate of GSH depletion, used as a measure of oxidative stress, was higher in cells expressing Mt++ and Mt+ proteins as compared with cells expressing ER+ protein. In addition, the cellular level of F(2)-isoprostanes, a direct indicator of oxidative stress, was increased markedly in Mt++ cells after ethanol treatment. Notably, expression of Mt++ CYP2E1 protein in yeast cells caused more severe mitochondrial DNA damage and respiratory deficiency than the wild type or ER+ proteins as tested by the inability of cells to grow on glycerol or ethanol. Additionally, liver mitochondria from ethanol-fed rats containing high mt CYP2E1 showed higher levels of F(2)-isoprostane production. These results strongly suggest that mt CYP2E1 induces oxidative stress and augments alcohol-mediated cell/tissue injury.


Subject(s)
Cytochrome P-450 CYP2E1/metabolism , Mitochondria/metabolism , Animals , COS Cells , Chlorocebus aethiops , DNA, Mitochondrial/metabolism , Endoplasmic Reticulum/metabolism , Ethanol/chemistry , Glutathione/metabolism , Humans , Mitochondria, Liver/metabolism , Oxidative Stress , Rats , Rats, Sprague-Dawley , Subcellular Fractions/metabolism
5.
Biochim Biophys Acta ; 1802(1): 11-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19619643

ABSTRACT

Mitochondrial dysfunction is an important intracellular lesion associated with a wide variety of diseases including neurodegenerative disorders. In addition to aging, oxidative stress and mitochondrial DNA mutations, recent studies have implicated a role for the mitochondrial accumulation of proteins such as plasma membrane associated amyloid precursor protein (APP) and cytosolic alpha synuclein in the pathogenesis of mitochondrial dysfunction in Alzheimer's disease (AD) and Parkinson's disease (PD), respectively. Both of these proteins contain cryptic mitochondrial targeting signals, which drive their transport across mitochondria. In general, mitochondrial entry of nuclear coded proteins is assisted by import receptors situated in both outer and inner mitochondrial membranes. A growing number of evidence suggests that APP and alpha synclein interact with import receptors to gain entry into mitochondrial compartment. Additionally, carboxy terminal cleaved product of APP, approximately 4 kDa Abeta, is also transported into mitochondria with the help of mitochondrial outer membrane import receptors. This review focuses on the mitochondrial targeting and accumulation of these two structurally different proteins and the mode of mechanism by which they affect the physiological functions of mitochondria.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Mitochondria/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Alzheimer Disease/physiopathology , Amino Acid Sequence , Animals , Humans , Mice , Mitochondria/physiology , Mitochondrial Membranes/metabolism , Models, Biological , Molecular Sequence Data , Parkinson Disease/physiopathology , Protein Transport/physiology
6.
Proc Natl Acad Sci U S A ; 105(1): 186-91, 2008 Jan 08.
Article in English | MEDLINE | ID: mdl-18172213

ABSTRACT

The environmental toxin 2,3,7,8-tetrachlorodibenzodioxin (TCDD) is a known human carcinogen; however, its precise mechanism of action remains unclear. Here we show that TCDD induces mitochondrial dysfunction, stress signaling, and tumor invasion by a mechanism similar to that described for mtDNA-depleted cells. Treatment of C2C12 cells with TCDD disrupted mitochondrial transmembrane potential in a time-dependent fashion and inhibited mitochondrial transcription and translation. TCDD also increased cytosolic [Ca(2+)](c) and RyR1-specific Ca(2+) release. These changes were associated with increased calcineurin (CnA) levels and activation of CnA-sensitive NF-kappaB/Rel (IkappaBbeta-dependent) factors. Cells treated with TCDD displayed resistance to apoptosis, increased expression of the tumor marker cathepsin L, and a high degree of invasiveness as tested by the Matrigel membrane invasion assay. These effects were reversed by the CnA inhibitor FK506, and CnA mRNA silencing suggesting that TCDD triggers a signaling pathway similar to mtDNA depletion. Taken together, these results reveal that TCDD may promote tumor progression in vivo by directly targeting mitochondrial transcription and induction of mitochondrial stress signaling.


Subject(s)
Cell Nucleus/metabolism , Dioxins/toxicity , Mitochondria/pathology , Neoplasms/chemically induced , Neoplasms/pathology , Animals , Apoptosis , Calcium/metabolism , Carcinogens/toxicity , Cell Line , Cell Line, Tumor , Disease Progression , Membrane Potentials , Mice , Models, Biological , Neoplasm Invasiveness , Polychlorinated Dibenzodioxins/toxicity , Signal Transduction , Transcription, Genetic
7.
J Biol Chem ; 284(25): 17352-17363, 2009 Jun 19.
Article in English | MEDLINE | ID: mdl-19401463

ABSTRACT

Previously we showed that xenobiotic inducible cytochrome P450 (CYP) proteins are bimodally targeted to the endoplasmic reticulum and mitochondria. In this study, we investigated the mechanism of delivery of chimeric signal containing CYP proteins to the peripheral and channel-forming mitochondrial outer membrane translocases (TOMs). CYP+33/1A1 and CYP2B1 did not require peripheral TOM70, TOM20, or TOM22 for translocation through the channel-forming TOM40 protein. In contrast, CYP+5/1A1 and CYP2E1 were able to bypass TOM20 and TOM22 but required TOM70. CYP27, which contains a canonical cleavable mitochondrial signal, required all of the peripheral TOMs for its mitochondrial translocation. We investigated the underlying mechanisms of bypass of peripheral TOMs by CYPs with chimeric signals. The results suggested that interaction of CYPs with Hsp70, a cytosolic chaperone involved in the mitochondrial import, alone was sufficient for the recognition of chimeric signals by peripheral TOMs. However, sequential interaction of chimeric signal containing CYPs with Hsp70 and Hsp90 resulted in the bypass of peripheral TOMs, whereas CYP27A1 interacted only with Hsp70 and was not able to bypass peripheral TOMs. Our results also show that delivery of a chimeric signal containing client protein by Hsp90 required the cytosol-exposed NH(2)-terminal 143 amino acids of TOM40. TOM40 devoid of this domain was unable to import CYP proteins. These results suggest that compared with the unimodal mitochondrial targeting signals, the chimeric mitochondrial targeting signals are highly evolved and dynamic in nature.


Subject(s)
Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/metabolism , Membrane Transport Proteins/metabolism , Mitochondria, Liver/metabolism , Mitochondrial Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Biological Transport, Active , Cytochrome P-450 Enzyme System/genetics , Genes, Fungal , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , In Vitro Techniques , Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Proteins/genetics , Models, Biological , Mutation , Protein Sorting Signals/genetics , Protein Sorting Signals/physiology , Rabbits , Rats , Receptors, Cell Surface , Receptors, Cytoplasmic and Nuclear/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
8.
Mol Genet Metab ; 99(1): 90-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19781968

ABSTRACT

Human cytochrome P450 2D6 (CYP2D6) is responsible for the metabolism of approximately 20% of drugs in common clinical use. The CYP2D6 gene locus is highly polymorphic. Many of the polymorphisms have been shown to be clinically relevant and can account for inter-individual differences in the metabolism of specific drugs. In addition to the established sources of variability in CYP2D6-dependent drug metabolism, a recent study in our laboratory identified CYP2D6 in the mitochondria of human liver samples and found that it is metabolically active in this novel location. In the present study we show that mutations are present in the targeting signal region of CYP2D6 that may help to account for the inter-individual variability that was observed previously in the level of the mitochondrial enzyme in human liver samples. These mutations were identified within the ER targeting domain, the proline-rich domain as well as the putative protein kinase A (PKA) and protein kinase C (PKC)-specific phosphorylation sites. In vitro studies demonstrate that the mutations identified in the targeting signals affect the efficiency of mitochondrial targeting of CYP2D6. Since the mitochondrial enzyme has been shown to be active in drug metabolism, this pharmacogenetic variation could play a role in modulating the response of an individual to drug therapy.


Subject(s)
Cytochrome P-450 CYP2D6/genetics , Mitochondria, Liver/metabolism , Mitochondrial Proteins/genetics , Mutation , Alternative Splicing , Amino Acid Sequence , Animals , Cytochrome P-450 CYP2D6/metabolism , Exons/genetics , Genetic Variation , Humans , Liver/metabolism , Microsomes/metabolism , Mitochondrial Proteins/metabolism , Molecular Sequence Data , Protein Sorting Signals/genetics , Protein Transport , Rats , Reverse Transcriptase Polymerase Chain Reaction
9.
J Cell Biol ; 161(3): 507-19, 2003 May 12.
Article in English | MEDLINE | ID: mdl-12732617

ABSTRACT

Mitochondrial genetic and metabolic stress causes activation of calcineurin (Cn), NFAT, ATF2, and NFkappaB/Rel factors, which collectively alter the expression of an array of nuclear genes. We demonstrate here that mitochondrial stress-induced activation of NFkappaB/Rel factors involves inactivation of IkappaBbeta through Cn-mediated dephosphorylation. Phosphorylated IkappaBbeta is a substrate for Cn phosphatase, which was inhibited by FK506 and RII peptide. Chemical cross-linking and coimmunoprecipitation show that NFkappaB/Rel factor-bound IkappaBbeta forms a ternary complex with Cn under in vitro and in vivo conditions that was sensitive to FK506. Results show that phosphorylation at S313 and S315 from the COOH-terminal PEST domain of IkappaBbeta is critical for binding to Cn. Mutations at S313/S315 of IkappaBbeta abolished Cn binding, inhibited Cn-mediated increase of Rel proteins in the nucleus, and had a dominant-negative effect on the mitochondrial stress-induced expression of RyR1 and cathepsin L genes. Our results show the distinctive nature of mitochondrial stress-induced NFkappaB/Rel activation, which is independent of IKKalpha and IKKbeta kinases and affects gene target(s) that are different from cytokine and TNFalpha-induced stress signaling. The results provide new insights into the role of Cn as a critical link between Ca2+ signaling and NFkappaB/Rel activation.


Subject(s)
Calcineurin/metabolism , Calcium Signaling/genetics , Cell Nucleus/metabolism , I-kappa B Proteins/metabolism , Mitochondria/metabolism , NF-kappa B/metabolism , Oncogene Proteins v-rel/metabolism , Stress, Physiological/metabolism , Animals , Binding Sites/drug effects , Binding Sites/genetics , Calcineurin/genetics , Calcium Signaling/drug effects , Cell Nucleus/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Eukaryotic Cells/drug effects , Eukaryotic Cells/metabolism , I-kappa B Proteins/genetics , Mice , Mitochondria/drug effects , Mutation/genetics , NF-kappa B/genetics , Oncogene Proteins v-rel/genetics , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/metabolism , Phosphorylation/drug effects , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/genetics , Serine/metabolism , Stress, Physiological/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics
10.
J Cell Biol ; 161(1): 41-54, 2003 Apr 14.
Article in English | MEDLINE | ID: mdl-12695498

ABSTRACT

Alzheimer's amyloid precursor protein 695 (APP) is a plasma membrane protein, which is known to be the source of the toxic amyloid beta (Abeta) peptide associated with the pathogenesis of Alzheimer's disease (AD). Here we demonstrate that by virtue of its chimeric NH2-terminal signal, APP is also targeted to mitochondria of cortical neuronal cells and select regions of the brain of a transgenic mouse model for AD. The positively charged residues at 40, 44, and 51 of APP are critical components of the mitochondrial-targeting signal. Chemical cross-linking together with immunoelectron microscopy show that the mitochondrial APP exists in NH2-terminal inside transmembrane orientation and in contact with mitochondrial translocase proteins. Mutational studies show that the acidic domain, which spans sequence 220-290 of APP, causes the transmembrane arrest with the COOH-terminal 73-kD portion of the protein facing the cytoplasmic side. Accumulation of full-length APP in the mitochondrial compartment in a transmembrane-arrested form, but not lacking the acidic domain, caused mitochondrial dysfunction and impaired energy metabolism. These results show, for the first time, that APP is targeted to neuronal mitochondria under some physiological and pathological conditions.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Intracellular Membranes/metabolism , Mitochondria/metabolism , Neurons/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amino Acid Sequence/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Brain/physiopathology , Brain/ultrastructure , Cell Compartmentation/genetics , Cells, Cultured , Cytochrome-c Oxidase Deficiency/genetics , Disease Models, Animal , Energy Metabolism/genetics , Humans , Immunohistochemistry , Intracellular Membranes/ultrastructure , Mice , Mice, Transgenic , Microscopy, Electron , Mitochondria/genetics , Mitochondria/ultrastructure , Molecular Conformation , Neurons/ultrastructure , Protein Structure, Tertiary/genetics , Protein Transport/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
11.
J Neurosci ; 26(35): 9057-68, 2006 Aug 30.
Article in English | MEDLINE | ID: mdl-16943564

ABSTRACT

Mitochondrial dysfunction is one of the major intracellular lesions of Alzheimer's disease (AD). However, the causative factors involved in the mitochondrial dysfunction in human AD are not well understood. Here we report that nonglycosylated full-length and C-terminal truncated amyloid precursor protein (APP) accumulates exclusively in the protein import channels of mitochondria of human AD brains but not in age-matched controls. Furthermore, in AD brains, mitochondrially associated APP formed stable approximately 480 kDa complexes with the translocase of the outer mitochondrial membrane 40 (TOM40) import channel and a super complex of approximately 620 kDa with both mitochondrial TOM40 and the translocase of the inner mitochondrial membrane 23 (TIM23) import channel TIM23 in an "N(in mitochondria)-C(out cytoplasm)" orientation. Accumulation of APP across mitochondrial import channels, which varied with the severity of AD, inhibited the entry of nuclear-encoded cytochrome c oxidase subunits IV and Vb proteins, which was associated with decreased cytochrome c oxidase activity and increased levels of H2O2. Regional distribution of mitochondrial APP showed higher levels in AD-vulnerable brain regions, such as the frontal cortex, hippocampus, and amygdala. Mitochondrial accumulation of APP was also observed in the cholinergic, dopaminergic, GABAergic, and glutamatergic neuronal types in the category III AD brains. The levels of translocationally arrested mitochondrial APP directly correlated with mitochondrial dysfunction. Moreover, apolipoprotein genotype analysis revealed that AD subjects with the E3/E4 alleles had the highest content of mitochondrial APP. Collectively, these results suggest that abnormal accumulation of APP across mitochondrial import channels, causing mitochondrial dysfunction, is a hallmark of human AD pathology.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/metabolism , Brain/physiopathology , Mitochondria , Alzheimer Disease/metabolism , Apolipoprotein E3 , Apolipoprotein E4 , Apolipoproteins E/metabolism , Biological Transport , Brain/metabolism , Cadaver , Case-Control Studies , Electron Transport Complex IV/metabolism , Glycosylation , Humans , Hydrogen Peroxide/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/enzymology , Mitochondrial Proteins/metabolism , Neurons/metabolism , Tissue Distribution
12.
Neuroscientist ; 13(6): 626-38, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17911214

ABSTRACT

Growing evidence suggests that mitochondrial dysfunction is one of the key intracellular lesions associated with the pathogenesis of Alzheimer's disease (AD). Mitochondria, the powerhouses of the cell, participate in a number of physiological functions that include calcium homeostasis, signal transduction, and apoptosis. However, the pathophysiological mechanisms underlying the decline of mitochondrial vital functions leading to the dysfunction of mitochondria during AD are not well understood. Recent literature has observed the accumulation of Alzheimer's amyloid precursor protein (APP) and its C-terminal-cleaved product beta-amyloid (Abeta) in the mitochondrial compartment. Furthermore, evidence also implicates that the accumulation of full-length APP and Abeta in the mitochondrial compartment has a causative role in impairing mitochondrial physiological functions. Here, we review the mode of mitochondrial transport of full-length APP and Abeta and its pathological implications in bringing about mitochondrial dysfunction as seen in AD.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Mitochondrial Diseases/complications , Animals , Humans , Mitochondria/pathology , Mitochondria/physiology , Models, Biological
13.
FEBS Lett ; 581(7): 1302-10, 2007 Apr 03.
Article in English | MEDLINE | ID: mdl-17349628

ABSTRACT

We have mapped the sites of ischemia/reperfusion-induced phosphorylation of cytochrome c oxidase (CcO) subunits in rabbit hearts by using a combination of Blue Native gel/Tricine gel electrophoresis and nano-LC-MS/MS approaches. We used precursor ion scanning combined with neutral loss scanning and found that mature CcO subunit I was phosphorylated at tandem Ser115/Ser116 positions, subunit IVi1 at Thr52 and subunit Vb at Ser40. These sites are highly conserved in mammalian species. Molecular modeling suggests that phosphorylation sites of subunit I face the inter membrane space while those of subunits IVi1 and Vb face the matrix side.


Subject(s)
Electron Transport Complex IV/chemistry , Myocardial Reperfusion Injury/enzymology , Myocardium/enzymology , Amino Acid Sequence , Animals , Chromatography, Liquid , Electron Transport Complex IV/metabolism , Electrophoresis, Polyacrylamide Gel , Glycine/analogs & derivatives , Glycine/chemistry , Heart , Molecular Sequence Data , Phosphorylation , Protein Conformation , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/metabolism , Rabbits , Serine/metabolism , Spectrometry, Mass, Electrospray Ionization , Threonine/metabolism
14.
Rev Neurosci ; 18(5): 343-54, 2007.
Article in English | MEDLINE | ID: mdl-19544621

ABSTRACT

Alzheimer's disease (AD) is a multifactorial disorder. Mitochondrial dysfunction is one of the key characteristics of AD pathogenesis. However, the mechanisms underlying the progression of mitochondrial impairment during AD are not clear. Growing evidence suggests a causative role for intracellular accumulation of amyloid precursor protein (APP) and its proteolytic products in the pathogenesis of AD. Furthermore, APP possesses several domains including a mitochondrial targeting sequence. Recent literature suggests that mitochondrial localization of full length APP and its C-terminal proteolytically cleaved derivative beta amyloid (Abeta) are associated with the mitochondrial dysfunction. Here, we review the nature of mitochondrial localization of APP and Abeta and their pathological implications in AD mitochondrial dysfunction.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/chemistry , Animals , Apolipoproteins E/metabolism , Brain/physiopathology , Humans , Mitochondrial Diseases/physiopathology , Protein Structure, Tertiary/physiology , Protein Transport/physiology
15.
FEBS J ; 274(17): 4615-30, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17697118

ABSTRACT

Previously we showed that intact rat cytochrome P450 2E1, cytochrome P450 2B1 and truncated cytochrome P450 1A1 are targeted to mitochondria in rat tissues and COS cells. However, some reports suggest that truncated cytochrome P450 2E1 is targeted to mitochondria. In this study, we used a heterologous yeast system to ascertain the conservation of targeting mechanisms and the nature of mitochondria-targeted proteins. Mitochondrial integrity and purity were established using electron microscopy, and treatment with digitonin and protease. Full-length cytochrome P450 2E1 and cytochrome P450 2B1 were targeted both to microsomes and mitochondria, whereas truncated cytochrome P450 1A1 (+ 5 and + 33/cytochrome P450 1A1) were targeted to mitochondria. Inability to target intact cytochrome P450 1A1 was probably due to lack of cytosolic endoprotease activity in yeast cells. Mitochondrial targeting of cytochrome P450 2E1 was severely impaired in protein kinase A-deficient cells. Similarly, a phosphorylation site mutant cytochrome P450 2E1 (Ser129A) was poorly targeted to the mitochondria, thus confirming the importance of protein kinase A-mediated protein phosphorylation in mitochondrial targeting. Mitochondria-targeted proteins were localized in the matrix compartment peripherally associated with the inner membrane and their ethoxyresorufin O-dealkylation, erythromycin N-demethylase, benzoxyresorufin O-dealkylation and nitrosodimethylamine N-demethylase activities were fully supported by yeast mitochondrial ferredoxin and ferredoxin reductase.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP2B1/metabolism , Cytochrome P-450 CYP2E1/metabolism , Saccharomyces cerevisiae/enzymology , Animals , Binding Sites , Catalysis , Microscopy, Electron, Scanning , Microsomes/enzymology , Rats
16.
FASEB J ; 16(3): 302-14, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11874980

ABSTRACT

CD38 is an ectocyclase that converts NAD+ to the Ca2+-releasing second messenger cyclic ADP-ribose (cADPr). Here we report that in addition to CD38 ecto-catalysis, intracellularly expressed CD38 may catalyze NAD+-->cADPr conversion to cause cytosolic Ca2+ release. High levels of CD38 were found in the plasma membranes, endoplasmic reticulum, and nuclear membranes of osteoblastic MC3T3-E1 cells. More important, intracellular CD38 was colocalized with target ryanodine receptors. The cyclase also converted a NAD+ surrogate, NGD+, to its fluorescent product, cGDPr (Km approximately 5.13 microM). NAD+ also triggered a cytosolic Ca2+ signal. Similar results were obtained with NIH3T3 cells, which overexpressed a CD38-EGFP fusion protein. The Delta(-49)-CD38-EGFP mutant with a deleted amino-terminal tail and transmembrane domain appeared mainly in the mitochondria with an expected loss of its membrane localization, but the NAD+-induced cytosolic Ca2+ signal was preserved. Likewise, Ca2+ release persisted in cells transfected with the Myr-Delta(-49)-CD38-EGFP or Delta(-49)-CD38-EGFP-Fan mutants, both directed to the plasma membrane but in an opposite topology to the full-length CD38-EGFP. Finally, ryanodine inhibited Ca2+ signaling, indicating the downstream activation of ryanodine receptors by cADPr. We conclude that intracellularly expressed CD38 might link cellular NAD+ production to cytosolic Ca2+ signaling.


Subject(s)
Antigens, CD , Antigens, Differentiation/physiology , Calcium Signaling , NAD+ Nucleosidase/physiology , NAD/pharmacology , 3T3 Cells , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Animals , Antigens, Differentiation/analysis , Antigens, Differentiation/genetics , Cell Line , Cell Membrane/enzymology , Cell Nucleus/enzymology , Cytosol/metabolism , Endoplasmic Reticulum/enzymology , Green Fluorescent Proteins , Indicators and Reagents/analysis , Intracellular Membranes/enzymology , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Membrane Glycoproteins , Mice , Microscopy, Confocal , Models, Biological , Mutation , NAD+ Nucleosidase/analysis , NAD+ Nucleosidase/genetics , Osteoblasts/enzymology , Recombinant Fusion Proteins/analysis , Ryanodine Receptor Calcium Release Channel/analysis
17.
FEBS J ; 276(13): 3440-53, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19438707

ABSTRACT

Constitutively expressed human cytochrome P450 2D6 (CYP2D6; EC 1.14.14.1) is responsible for the metabolism of approximately 25% of drugs in common clinical use. It is widely accepted that CYP2D6 is localized in the endoplasmic reticulum of cells; however, we have identified this enzyme in the mitochondria of human liver samples and found that extensive inter-individual variability exists with respect to the level of the mitochondrial enzyme. Metabolic assays using 7-methoxy-4-aminomethylcoumarin as a substrate show that the human liver mitochondrial enzyme is capable of oxidizing this substrate and that the catalytic activity is supported by mitochondrial electron transfer proteins. In the present study, we show that CYP2D6 contains an N-terminal chimeric signal that mediates its bimodal targeting to the endoplasmic reticulum and mitochondria. In vitro mitochondrial import studies using both N-terminal deletions and point mutations suggest that the mitochondrial targeting signal is localized between residues 23-33 and that the positively-charged residues at positions 24, 25, 26, 28 and 32 are required for mitochondrial targeting. The importance of the positively-charged residues was confirmed by transient transfection of a CYP2D6 mitochondrial targeting signal mutant in COS-7 cells. Both the mitochondria and the microsomes from a CYP2D6 stable expression cell line contain the enzyme and both fractions exhibit bufuralol 1'-hydroxylation activity, which is completely inhibited by CYP2D6 inhibitory antibody. Overall, these results suggest that the targeting of CYP2D6 to mitochondria could be an important physiological process that has significance in xenobiotic metabolism.


Subject(s)
Cytochrome P-450 CYP2D6 , Isoenzymes , Microsomes, Liver/enzymology , Pharmaceutical Preparations/metabolism , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Coumarins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/metabolism , Ethanolamines/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Microsomes, Liver/metabolism , Molecular Sequence Data , Protein Sorting Signals/genetics
18.
J Biol Chem ; 283(28): 19769-80, 2008 Jul 11.
Article in English | MEDLINE | ID: mdl-18480056

ABSTRACT

Previously we showed that xenobiotic-inducible cytochrome P450 (CYP) proteins are bimodally targeted to the endoplasmic reticulum and mitochondria. In the present study, we investigated the mechanism of delivery of chimeric signal-containing CYP proteins to the peripheral and channel-forming mitochondrial outer membrane translocases (TOMs). CYP+33/1A1 and CYP2B1 did not require peripheral TOM70, TOM20, or TOM22 for translocation through the channel-forming TOM40 protein. In contrast, CYP+5/1A1 and CYP2E1 were able to bypass TOM20 and TOM22 but required TOM70. CYP27, which contains a canonical cleavable mitochondrial signal, required all of the peripheral TOMs for its mitochondrial translocation. We investigated the underlying mechanisms of bypass of peripheral TOMs by CYPs with chimeric signals. The results suggested that interaction of CYPs with Hsp70, a cytosolic chaperone involved in the mitochondrial import, alone was sufficient for the recognition of chimeric signals by peripheral TOMs. However, sequential interaction of chimeric signal-containing CYPs with Hsp70 and Hsp90 resulted in the bypass of peripheral TOMs, whereas CYP27 interacted only with Hsp70 and was not able to bypass peripheral TOMs. Our results also show that delivery of chimeric signal-containing client proteins by Hsp90 required the cytosol-exposed N-terminal 143 amino acids of TOM40. TOM40 devoid of this domain was unable to bind CYP proteins. These results suggest that, compared with the unimodal mitochondria-targeting signals, the chimeric mitochondria-targeting signals are highly evolved and dynamic in nature.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Membrane Transport Proteins/metabolism , Mitochondria, Liver/metabolism , Mitochondrial Proteins/metabolism , Protein Sorting Signals/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cytochrome P-450 Enzyme System/genetics , Cytoplasm/genetics , Cytoplasm/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Membrane Transport Proteins/genetics , Mitochondria, Liver/genetics , Mitochondrial Membrane Transport Proteins , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Proteins/genetics , Protein Transport/physiology , Rabbits , Rats , Receptors, Cell Surface , Receptors, Cytoplasmic and Nuclear/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
19.
J Biol Chem ; 283(14): 9089-100, 2008 Apr 04.
Article in English | MEDLINE | ID: mdl-18245082

ABSTRACT

Alpha-synuclein, a protein implicated in the pathogenesis of Parkinson disease (PD), is thought to affect mitochondrial functions, although the mechanisms of its action remain unclear. In this study we show that the N-terminal 32 amino acids of human alpha-synuclein contain cryptic mitochondrial targeting signal, which is important for mitochondrial targeting of alpha-synuclein. Mitochondrial imported alpha-synuclein is predominantly associated with the inner membrane. Accumulation of wild-type alpha-synuclein in the mitochondria of human dopaminergic neurons caused reduced mitochondrial complex I activity and increased production of reactive oxygen species. However, these defects occurred at an early time point in dopaminergic neurons expressing familial alpha-synuclein with A53T mutation as compared with wild-type alpha-synuclein. Importantly, alpha-synuclein that lacks mitochondrial targeting signal failed to target to the mitochondria and showed no detectable effect on complex I function. The PD relevance of these results was investigated using mitochondria of substantia nigra, striatum, and cerebellum of postmortem late-onset PD and normal human brains. Results showed the constitutive presence of approximately 14-kDa alpha-synuclein in the mitochondria of all three brain regions of normal subjects. Mitochondria of PD-vulnerable substantia nigra and striatum but not cerebellum from PD subjects showed significant accumulation of alpha-synuclein and decreased complex I activity. Analysis of mitochondria from PD brain and alpha-synuclein expressing dopaminergic neuronal cultures using blue native gel electrophoresis and immunocapture technique showed the association of alpha-synuclein with complex I. These results provide evidence that mitochondrial accumulated alpha-synuclein may interact with complex I and interfere with its functions.


Subject(s)
Electron Transport Complex I/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Aged , Aged, 80 and over , Amino Acid Substitution , Dopamine/metabolism , Electron Transport Complex I/genetics , Female , Humans , Male , Middle Aged , Mitochondria/ultrastructure , Mitochondrial Membranes/ultrastructure , Mutation, Missense , Neurons/pathology , Organ Specificity/genetics , Parkinson Disease/genetics , Parkinson Disease/pathology , Protein Sorting Signals/genetics , Protein Transport/genetics , Reactive Oxygen Species/metabolism , alpha-Synuclein/genetics
20.
Am J Physiol Heart Circ Physiol ; 292(5): H2459-66, 2007 May.
Article in English | MEDLINE | ID: mdl-17237252

ABSTRACT

Protein kinase A (PKA) activation has been implicated in early-phase ischemic preconditioning. We recently found that during ischemia PKA activation causes inactivation of cytochrome-c oxidase (CcO) and contributes to myocardial damage due to ischemia-reperfusion. It may be that beta-adrenergic stimulation during ischemia via endogenous catecholamine release activates PKA. Thus beta-adrenergic stimulation may mediate both myocardial protection and damage during ischemia. The present studies were designed to determine the role of the beta(1)-adrenergic receptor (beta(1)-AR) in myocardial ischemic damage and ischemic preconditioning. Langendorff-perfused rabbit hearts underwent 30-min ischemia by anterior coronary artery ligation followed by 2-h reperfusion. Occlusion-reperfusion damage was evaluated by delineating the nonperfused volume of myocardium at risk and volume of myocardial necrosis after 2-h reperfusion. In some hearts ischemic preconditioning was accomplished by two 5-min episodes of global low-flow ischemia separated by 10 min before coronary occlusion-reperfusion. Orthogonal electrocardiograms were recorded, and coronary flow was monitored by a drip count. Three hearts from each experimental group were used to determine mitochondrial CcO and aconitase activities. Two-hour reperfusion after occlusion caused an additional decrease in CcO activity vs. that after 30-min occlusion alone. Blocking the beta(1)-AR during occlusion-reperfusion reversed CcO activity depression and preserved myocardium at risk for necrosis. Similarly, mitochondrial aconitase activity exhibited a parallel response after occlusion-reperfusion as well as for the other interventions. Furthermore, classic ischemic preconditioning had no effect on CcO depression. However, blocking the beta(1)-AR during preconditioning eliminated the cardioprotection. If the beta(1)-AR was blocked after preconditioning, the myocardium was preserved. Interestingly, in both of the latter cases the depression in CcO activity was reversed. Thus the beta(1)-AR plays a dual role in myocardial ischemic damage. Our findings may lead to therapeutic strategies for preserving myocardium at risk for infarction, especially in coronary reperfusion intervention.


Subject(s)
Adrenergic beta-1 Receptor Agonists , Ischemic Preconditioning, Myocardial/methods , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Receptors, Adrenergic, beta-1/metabolism , Animals , Male , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL