Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Biochemistry ; 54(3): 753-64, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25545070

ABSTRACT

The NuoD segment (homologue of mitochondrial 49 kDa subunit) of the proton-translocating NADH:quinone oxidoreductase (complex I/NDH-1) from Escherichia coli is in the hydrophilic domain and bears many highly conserved amino acid residues. The three-dimensional structural model of NDH-1 suggests that the NuoD segment, together with the neighboring subunits, constitutes a putative quinone binding cavity. We used the homologous DNA recombination technique to clarify the role of selected key amino acid residues of the NuoD segment. Among them, residues Tyr273 and His224 were considered candidates for having important interactions with the quinone headgroup. Mutant Y273F retained partial activity but lost sensitivity to capsaicin-40. Mutant H224R scarcely affected the activity, suggesting that this residue may not be essential. His224 is located in a loop near the N-terminus of the NuoD segment (Gly217-Phe227) which is considered to form part of the quinone binding cavity. In contrast to the His224 mutation, mutants G217V, P218A, and G225V almost completely lost the activity. One region of this loop is positioned close to a cytosolic loop of the NuoA subunit in the membrane domain, and together they seem to be important in keeping the quinone binding cavity intact. The structural role of the longest helix in the NuoD segment located behind the quinone binding cavity was also investigated. Possible roles of other highly conserved residues of the NuoD segment are discussed.


Subject(s)
Amino Acids/metabolism , Conserved Sequence , Electron Transport Complex I/chemistry , Electron Transport Complex I/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Amino Acid Sequence , Enzyme Inhibitors/pharmacology , Immunoblotting , Inhibitory Concentration 50 , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation , Native Polyacrylamide Gel Electrophoresis , Oxidoreductases/metabolism , Protein Subunits/chemistry , Protein Subunits/metabolism , Protons , Sequence Alignment , Sequence Analysis, Protein , Structure-Activity Relationship
2.
Neurosci Lett ; 585: 171-6, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25481764

ABSTRACT

Leber's hereditary optic neuropathy (LHON) is an inherited disorder affecting the retinal ganglion cells (RGCs) and their axons that lead to the loss of central vision. This study is aimed at evaluating the LHON symptoms in rats administered with rotenone microspheres into the superior colliculus (SC). Optical coherence tomography (OCT) analysis showed substantial loss of retinal nerve fiber layer (RNFL) thickness in rotenone injected rats. Optokinetic testing in rotenone treated rats showed decrease in head-tracking response. Electrophysiological mapping of the SC surface demonstrated attenuation of visually evoked responses; however, no changes were observed in the ERG data. The progressive pattern of disease manifestation in rotenone administered rats demonstrated several similarities with human disease symptoms. These rats with LHON-like symptoms can serve as a model for future investigators to design and implement reliable tests to assess the beneficial effects of therapeutic interventions for LHON disease.


Subject(s)
Disease Models, Animal , Optic Atrophy, Hereditary, Leber/physiopathology , Rotenone , Animals , Electroretinography , Evoked Potentials, Visual , Humans , Microspheres , Optic Atrophy, Hereditary, Leber/chemically induced , Rats , Superior Colliculi/physiopathology , Tomography, Optical Coherence
4.
J Bioenerg Biomembr ; 46(4): 279-87, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24973951

ABSTRACT

The proton-translocating NADH-quinone oxidoreductase (complex I/NDH-1) is the first and largest enzyme of the respiratory chain which has a central role in cellular energy production and is implicated in many human neurodegenerative diseases and aging. It is believed that the peripheral domain of complex I/NDH-1 transfers the electron from NADH to Quinone (Q) and the redox energy couples the proton translocation in the membrane domain. To investigate the mechanism of the proton translocation, in a series of works we have systematically studied all membrane subunits in the Escherichia coli NDH-1 by site-directed mutagenesis. In this mini-review, we have summarized our strategy and results of the mutagenesis by depicting residues essential for proton translocation, along with those for subunit connection. It is suggested that clues to understanding the driving forces of proton translocation lie in the similarities and differences of the membrane subunits, highlighting the communication of essential charged residues among the subunits. A possible proton translocation mechanism with all membrane subunits operating in unison is described.


Subject(s)
Cell Membrane/chemistry , Electron Transport Complex I/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Membrane Proteins/chemistry , Protein Subunits/chemistry , Protons , Benzoquinones/chemistry , Benzoquinones/metabolism , Cell Membrane/genetics , Cell Membrane/metabolism , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Humans , Ion Transport/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutagenesis, Site-Directed , NAD/chemistry , NAD/genetics , NAD/metabolism , Protein Structure, Tertiary , Protein Subunits/genetics , Protein Subunits/metabolism
5.
Neurobiol Dis ; 58: 281-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23816754

ABSTRACT

Mitochondrial defects can have significant consequences on many aspects of neuronal physiology. In particular, deficiencies in the first enzyme complex of the mitochondrial respiratory chain (complex I) are considered to be involved in a number of human neurodegenerative diseases. The current work highlights a tight correlation between the inhibition of complex I and the state of axonal myelination of the optic nerve. Exposing the visual pathway of rats to rotenone, a complex I inhibitor, resulted in disorganization of the node of Ranvier. The structure and function of the node depend on specific cell adhesion molecules, among others, CASPR (contactin associated protein) and contactin. CASPR and contactin are both on the axonal surfaces and need to be associated to be able to anchor their myelin counterpart. Here we show that inhibition of mitochondrial complex I by rotenone in rats induces reactive oxygen species, disrupts the interaction of CASPR and contactin couple, and thus damages the organization and function of the node of Ranvier. Demyelination of the optic nerve occurs as a consequence which is accompanied by a loss of vision. The physiological impairment could be reversed by introducing an alternative NADH dehydrogenase to the mitochondria of the visual system. The restoration of the nodal structure was specifically correlated with visual recovery in the treated animal.


Subject(s)
Demyelinating Diseases/pathology , Electron Transport Complex I/metabolism , Optic Nerve/pathology , Ranvier's Nodes/pathology , Animals , Cell Adhesion Molecules , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Count , Contactins/genetics , Contactins/metabolism , Demyelinating Diseases/chemically induced , Disease Models, Animal , Insecticides/pharmacology , Male , Myelin Proteins/genetics , Myelin Proteins/metabolism , Nerve Tissue Proteins/metabolism , Optic Nerve/drug effects , Ranvier's Nodes/drug effects , Ranvier's Nodes/ultrastructure , Rats , Rats, Long-Evans , Reactive Oxygen Species/metabolism , Rotenone/pharmacology , Time Factors , Visual Pathways/drug effects , Visual Pathways/metabolism , Visual Pathways/ultrastructure
6.
J Biol Chem ; 288(34): 24705-16, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23864658

ABSTRACT

The proton-translocating NADH-quinone oxidoreductase (complex I/NDH-1) contains a peripheral and a membrane domain. Three antiporter-like subunits in the membrane domain, NuoL, NuoM, and NuoN (ND5, ND4 and ND2, respectively), are structurally similar. We analyzed the role of NuoN in Escherichia coli NDH-1. The lysine residue at position 395 in NuoN (NLys(395)) is conserved in NuoL (LLys(399)) but is replaced by glutamic acid (MGlu(407)) in NuoM. Our mutation study on NLys(395) suggests that this residue participates in the proton translocation. Furthermore, we found that MGlu(407) is also essential and most likely interacts with conserved LArg(175). Glutamic acids, NGlu(133), MGlu(144), and LGlu(144), are corresponding residues. Unlike mutants of MGlu(144) and LGlu(144), mutation of NGlu(133) scarcely affected the energy-transducing activities. However, a double mutant of NGlu(133) and nearby KGlu(72) showed significant inhibition of these activities. This suggests that NGlu(133) bears a functional role similar to LGlu(144) and MGlu(144) but its mutation can be partially compensated by the nearby carboxyl residue. Conserved prolines located at loops of discontinuous transmembrane helices of NuoL, NuoM, and NuoN were shown to play a similar role in the energy-transducing activity. It seems likely that NuoL, NuoM, and NuoN pump protons by a similar mechanism. Our data also revealed that NLys(158) is one of the key interaction points with helix HL in NuoL. A truncation study indicated that the C-terminal amphipathic segments of NTM14 interacts with the Mß sheet located on the opposite side of helix HL. Taken together, the mechanism of H(+) translocation in NDH-1 is discussed.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Membrane Proteins/metabolism , Protein Subunits/metabolism , Amino Acid Substitution , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Ion Transport/physiology , Membrane Proteins/genetics , Mutation, Missense , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits/genetics , Protons
7.
J Clin Invest ; 123(3): 1068-81, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23426180

ABSTRACT

Despite advances in clinical therapy, metastasis remains the leading cause of death in breast cancer patients. Mutations in mitochondrial DNA, including those affecting complex I and oxidative phosphorylation, are found in breast tumors and could facilitate metastasis. This study identifies mitochondrial complex I as critical for defining an aggressive phenotype in breast cancer cells. Specific enhancement of mitochondrial complex I activity inhibited tumor growth and metastasis through regulation of the tumor cell NAD+/NADH redox balance, mTORC1 activity, and autophagy. Conversely, nonlethal reduction of NAD+ levels by interfering with nicotinamide phosphoribosyltransferase expression rendered tumor cells more aggressive and increased metastasis. The results translate into a new therapeutic strategy: enhancement of the NAD+/NADH balance through treatment with NAD+ precursors inhibited metastasis in xenograft models, increased animal survival, and strongly interfered with oncogene-driven breast cancer progression in the MMTV-PyMT mouse model. Thus, aberration in mitochondrial complex I NADH dehydrogenase activity can profoundly enhance the aggressiveness of human breast cancer cells, while therapeutic normalization of the NAD+/NADH balance can inhibit metastasis and prevent disease progression.


Subject(s)
Brain Neoplasms/metabolism , Electron Transport Complex I/physiology , Lung Neoplasms/metabolism , Mammary Neoplasms, Experimental/metabolism , NAD/metabolism , Saccharomyces cerevisiae Proteins/physiology , Acrylamides/pharmacology , Animals , Autophagy , Autophagy-Related Protein 5 , Brain Neoplasms/secondary , Cell Line, Tumor , Cell Proliferation , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Disease Progression , Electron Transport Complex I/biosynthesis , Female , Gene Knockdown Techniques , Humans , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred BALB C , Mice, SCID , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Multiprotein Complexes , NAD/physiology , Neoplasm Transplantation , Niacin/pharmacology , Niacinamide/pharmacology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/metabolism , Piperidines/pharmacology , Protein Transport , Proteins/metabolism , Recombinant Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/biosynthesis , TOR Serine-Threonine Kinases
8.
Brain Behav ; 3(4): 351-66, 2013 Jul.
Article in English | MEDLINE | ID: mdl-24381808

ABSTRACT

This study was undertaken to develop a phenotypic model recapitulating the neuropathology of Parkinson's disease (PD). Such a model would show loss of dopamine in the basal ganglia, appearance of Lewy bodies, and the early stages of motor dysfunction. The model was developed by subcutaneously injecting biodegradable microspheres of rotenone, a complex I inhibitor in 8-9 month old, ovariectomized Long-Evans rats. Animals were observed for changes in body weight and motor activity. At the end of 11-12 weeks animals were euthanized and the brains examined for histopathological changes. Rotenone treated animals gain weight and appear normal and healthy as compared to controls but showed modest hypokinesia around 5-6 weeks posttreatment. Animals showed loss of dopaminergic (DA) neurons and the appearance of putative Lewy bodies in the substantia nigra. Neuroinflammation and oxidative stress were evidenced by the appearance of activated microglia, iron precipitates, and 8-oxo-2'-deoxyguanosine a major product of DNA oxidation. The dorsal striatum, the projection site of midbrain DA neurons, showed a significant reduction in tyrosine hydroxylase immunostaining, together with an increase in reactive astrocytes, an early sign of DA nerve terminal damage. Levels of vesicular monoamine transporter 2 (VMAT2) were significantly reduced in the dorsal striatum; however, there was an unexpected increase in dopamine transporter (DAT) levels. Old, ovariectomized females treated with rotenone microspheres present with normal weight gain and good health but a modest hypokinesia. Accompanying this behavioral phenotype are a constellation of neuropathologies characteristic of PD that include loss of DA neurons, microglia activation, oxidative damage to nuclear DNA, iron deposition, and appearance of putative Lewy bodies. This phenotypic model recapitulating the neuropathology of Parkinson's disease could provide insight into early mechanisms of pathogenesis and could aid in the identification of biomarkers to identify patients in early stage, PD.

9.
J Biol Chem ; 287(51): 42763-72, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23105119

ABSTRACT

The bacterial H(+)-translocating NADH:quinone oxidoreductase (NDH-1) catalyzes electron transfer from NADH to quinone coupled with proton pumping across the cytoplasmic membrane. The NuoK subunit (counterpart of the mitochondrial ND4L subunit) is one of the seven hydrophobic subunits in the membrane domain and bears three transmembrane segments (TM1-3). Two glutamic residues located in the adjacent transmembrane helices of NuoK are important for the energy coupled activity of NDH-1. In particular, mutation of the highly conserved carboxyl residue ((K)Glu-36 in TM2) to Ala led to a complete loss of the NDH-1 activities. Mutation of the second conserved carboxyl residue ((K)Glu-72 in TM3) moderately reduced the activities. To clarify the contribution of NuoK to the mechanism of proton translocation, we relocated these two conserved residues. When we shifted (K)Glu-36 along TM2 to positions 32, 38, 39, and 40, the mutants largely retained energy transducing NDH-1 activities. According to the recent structural information, these positions are located in the vicinity of (K)Glu-36, present in the same helix phase, in an immediately before and after helix turn. In an earlier study, a double mutation of two arginine residues located in a short cytoplasmic loop between TM1 and TM2 (loop-1) showed a drastic effect on energy transducing activities. Therefore, the importance of this cytosolic loop of NuoK ((K)Arg-25, (K)Arg-26, and (K)Asn-27) for the energy transducing activities was extensively studied. The probable roles of subunit NuoK in the energy transducing mechanism of NDH-1 are discussed.


Subject(s)
Energy Metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Membrane Proteins/metabolism , NADH Dehydrogenase/metabolism , Protein Subunits/metabolism , Amino Acid Sequence , Glutamic Acid/metabolism , Hydrogen-Ion Concentration , Immunoblotting , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation/genetics , NAD/metabolism , NADH Dehydrogenase/chemistry , Native Polyacrylamide Gel Electrophoresis , Oxidation-Reduction , Protein Structure, Secondary , Protein Subunits/chemistry , Protons
10.
J Biol Chem ; 287(21): 17363-17373, 2012 May 18.
Article in English | MEDLINE | ID: mdl-22474289

ABSTRACT

Bacterial proton-translocating NADH:quinone oxidoreductase (NDH-1) consists of a peripheral and a membrane domain. The peripheral domain catalyzes the electron transfer from NADH to quinone through a chain of seven iron-sulfur (Fe/S) clusters. Subunit NuoI in the peripheral domain contains two [4Fe-4S] clusters (N6a and N6b) and plays a role in bridging the electron transfer from cluster N5 to the terminal cluster N2. We constructed mutants for eight individual Cys-coordinating Fe/S clusters. With the exception of C63S, all mutants had damaged architecture of NDH-1, suggesting that Cys-coordinating Fe/S clusters help maintain the NDH-1 structure. Studies of three mutants (C63S-coordinating N6a, P110A located near N6a, and P71A in the vicinity of N6b) were carried out using EPR measurement. These three mutations did not affect the EPR signals from [2Fe-2S] clusters and retained electron transfer activities. Signals at g(z) = 2.09 disappeared in C63S and P110A but not in P71A. Considering our data together with the available information, g(z,x) = 2.09, 1.88 signals are assigned to cluster N6a. It is of interest that, in terms of g(z,x) values, cluster N6a is similar to cluster N4. In addition, we investigated the residues (Ile-94 and Ile-100) that are predicted to serve as electron wires between N6a and N6b and between N6b and N2, respectively. Replacement of Ile-100 and Ile-94 with Ala/Gly did not affect the electron transfer activity significantly. It is concluded that conserved Ile-100 and Ile-94 are not essential for the electron transfer.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Membrane Proteins/metabolism , NADH Dehydrogenase/metabolism , NAD/metabolism , Amino Acid Substitution , Electron Spin Resonance Spectroscopy , Electron Transport/physiology , Escherichia coli Proteins/genetics , Membrane Proteins/genetics , Mutation, Missense , NAD/genetics , NADH Dehydrogenase/genetics , Protein Structure, Tertiary
11.
PLoS One ; 6(10): e25910, 2011.
Article in English | MEDLINE | ID: mdl-21991386

ABSTRACT

BACKGROUND: The rotenone-insensitive internal NADH-quinone oxidoreductase from yeast, Ndi1, has been shown to work as a replacement molecule for complex I in the respiratory chain of mammalian mitochondria. In the so-called transkingdom gene therapy, one major concern is the fact that the yeast protein is foreign in mammals. Long term expression of Ndi1 observed in rodents with no apparent damage to the target tissue was indicative of no action by the host's immune system. METHODOLOGY/PRINCIPAL FINDINGS: In the present study, we examined rat skeletal muscles expressing Ndi1 for possible signs of inflammatory or immune response. In parallel, we carried out delivery of the GFP gene using the same viral vector that was used for the NDI1 gene. The tissues were subjected to H&E staining and immunohistochemical analyses using antibodies specific for markers, CD11b, CD3, CD4, and CD8. The data showed no detectable signs of an immune response with the tissues expressing Ndi1. In contrast, mild but distinctive positive reactions were observed in the tissues expressing GFP. This clear difference most likely comes from the difference in the location of the expressed protein. Ndi1 was localized to the mitochondria whereas GFP was in the cytosol. CONCLUSIONS/SIGNIFICANCE: We demonstrated that Ndi1 expression did not trigger any inflammatory or immune response in rats. These results push forward the Ndi1-based molecular therapy and also expand the possibility of using foreign proteins that are directed to subcellular organelle such as mitochondria.


Subject(s)
Electron Transport Complex I/immunology , Immunity/immunology , Saccharomyces cerevisiae Proteins/immunology , Saccharomyces cerevisiae/immunology , Animals , Antibodies, Fungal/blood , Brain/immunology , Brain/pathology , Cell Death , Immunity, Humoral/immunology , Inflammation/blood , Inflammation/immunology , Inflammation/pathology , Macrophages/pathology , Male , Muscles/immunology , Muscles/pathology , Rats , Rats, Sprague-Dawley , Staining and Labeling
12.
J Biol Chem ; 286(39): 34007-14, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21835926

ABSTRACT

The proton-translocating NADH-quinone oxidoreductase (complex I/NDH-1) is a multisubunit enzymatic complex. It has a characteristic L-shaped form with two domains, a hydrophilic peripheral domain and a hydrophobic membrane domain. The membrane domain contains three antiporter-like subunits (NuoL, NuoM, and NuoN, Escherichia coli naming) that are considered to be involved in the proton translocation. Deletion of either NuoL or NuoM resulted in an incomplete assembly of NDH-1 and a total loss of the NADH-quinone oxidoreductase activity. We have truncated the C terminus segments of NuoM and NuoL by introducing STOP codons at different locations using site-directed mutagenesis of chromosomal DNA. Our results suggest an important structural role for the C-terminal segments of both subunits. The data further advocate that the elimination of the last transmembrane helix (TM14) of NuoM and the TM16 (at least C-terminal seven residues) or together with the HL helix and the TM15 of the NuoL subunit lead to reduced stability of the membrane arm and therefore of the whole NDH-1 complex. A region of NuoL critical for stability of NDH-1 architecture has been discussed.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , NADH Dehydrogenase/metabolism , Chromosomes, Bacterial/genetics , DNA, Bacterial/genetics , Enzyme Stability/physiology , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Gene Deletion , Mutagenesis, Site-Directed , NADH Dehydrogenase/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Protons
13.
J Biol Chem ; 286(11): 9287-97, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21220430

ABSTRACT

The flavoprotein rotenone-insensitive internal NADH-ubiquinone (UQ) oxidoreductase (Ndi1) is a member of the respiratory chain in Saccharomyces cerevisiae. We reported previously that bound UQ in Ndi1 plays a key role in preventing the generation of reactive oxygen species. Here, to elucidate this mechanism, we investigated biochemical properties of Ndi1 and its mutants in which highly conserved amino acid residues (presumably involved in NADH and/or UQ binding sites) were replaced. We found that wild-type Ndi1 formed a stable charge transfer (CT) complex (around 740 nm) with NADH, but not with NADPH, under anaerobic conditions. The intensity of the CT absorption band was significantly increased by the presence of bound UQ or externally added n-decylbenzoquinone. Interestingly, however, when Ndi1 was exposed to air, the CT band transiently reached the same maximum level regardless of the presence of UQ. This suggests that Ndi1 forms a ternary complex with NADH and UQ, but the role of UQ in withdrawing an electron can be substitutable with oxygen. Proteinase K digestion analysis showed that NADH (but not NADPH) binding induces conformational changes in Ndi1. The kinetic study of wild-type and mutant Ndi1 indicated that there is no overlap between NADH and UQ binding sites. Moreover, we found that the bound UQ can reversibly dissociate from Ndi1 and is thus replaceable with other quinones in the membrane. Taken together, unlike other NAD(P)H-UQ oxidoreductases, the Ndi1 reaction proceeds through a ternary complex (not a ping-pong) mechanism. The bound UQ keeps oxygen away from the reduced flavin.


Subject(s)
Electron Transport Complex I/chemistry , NAD/chemistry , Oxygen/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , Ubiquinone/chemistry , Anaerobiosis/physiology , Binding Sites , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Kinetics , Mutation , NAD/genetics , NAD/metabolism , Oxidation-Reduction , Oxygen/metabolism , Protein Structure, Quaternary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Ubiquinone/genetics , Ubiquinone/metabolism
14.
Parkinsons Dis ; 2011: 438370, 2010 Dec 06.
Article in English | MEDLINE | ID: mdl-21188192

ABSTRACT

Defects in mitochondrial proton-translocating NADH-quinone oxidoreductase (complex I) have been implicated in a number of acquired and hereditary diseases including Leigh's syndrome and more recently Parkinson's disease. A limited number of strategies have been attempted to repair the damaged complex I with little or no success. We have recently shown that the non-proton-pumping, internal NADH-ubiquinone oxidoreductase (Ndi1) from Saccharomyces cerevisiae (baker's yeast) can be successfully inserted into the mitochondria of mice and rats, and the enzyme was found to be fully active. Using recombinant adenoassociated virus vectors (serotype 5) carrying our NDI1 gene, we were able to express the Ndi1 protein in the substantia nigra (SN) of C57BL/6 mice with an expression period of two months. The results show that the AAV serotype 5 was highly efficient in expressing Ndi1 in the SN, when compared to a previous model using serotype 2, which led to nearly 100% protection when using an acute MPTP model. It is conceivable that the AAV-serotype5 carrying the NDI1 gene is a powerful tool for proof-of-concept study to demonstrate complex I defects as the causable factor in diseases of the brain.

15.
Biochemistry ; 49(47): 10072-80, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-20979355

ABSTRACT

The prokaryotic proton-translocating NADH-quinone oxidoreductase (NDH-1) is an L-shaped membrane-bound enzyme that contains 14 subunits (NuoA-NuoN or Nqo1-Nqo14). All subunits have their counterparts in the eukaryotic enzyme (complex I). NDH-1 consists of two domains: the peripheral arm (NuoB, -C, -D, -E, -F, -G, and -I) and the membrane arm (NuoA, -H, -J, -K, -L, -M, and -N). In Escherichia coli NDH-1, the hydrophilic subunits NuoC/Nqo5/30k and NuoD/Nqo4/49k are fused together in a single polypeptide as the NuoCD subunit. The NuoCD subunit is the only subunit that does not bear a cofactor in the peripheral arm. While some roles for inhibitor and quinone association have been reported for the NuoD segment, structural and functional roles of the NuoC segment remain mostly elusive. In this work, 14 highly conserved residues of the NuoC segment were mutated and 21 mutants were constructed using the chromosomal gene manipulation technique. From the enzymatic assays and immunochemical and blue-native gel analyses, it was found that residues Glu-138, Glu-140, and Asp-143 that are thought to be in the third α-helix are absolutely required for the energy-transducing NDH-1 activities and the assembly of the whole enzyme. Together with available information for the hydrophobic subunits, we propose that Glu-138, Glu-140, and Asp-143 of the NuoC segment may have a pivotal role in the structural stability of NDH-1.


Subject(s)
Quinone Reductases/chemistry , Amino Acid Sequence , Aspartic Acid/chemistry , Escherichia coli/enzymology , Escherichia coli Proteins/chemistry , Glutamic Acid/chemistry , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Protein Subunits/chemistry , Protons , Sequence Alignment
16.
PLoS One ; 5(7): e11472, 2010 Jul 08.
Article in English | MEDLINE | ID: mdl-20628600

ABSTRACT

BACKGROUND: Leber's hereditary optic neuropathy (LHON) is a maternally inherited disorder with point mutations in mitochondrial DNA which result in loss of vision in young adults. The majority of mutations reported to date are within the genes encoding the subunits of the mitochondrial NADH-quinone oxidoreductase, complex I. Establishment of animal models of LHON should help elucidate mechanism of the disease and could be utilized for possible development of therapeutic strategies. METHODOLOGY/PRINCIPAL FINDINGS: We established a rat model which involves injection of rotenone-loaded microspheres into the optic layer of the rat superior colliculus. The animals exhibited the most common features of LHON. Visual loss was observed within 2 weeks of rotenone administration with no apparent effect on retinal ganglion cells. Death of retinal ganglion cells occurred at a later stage. Using our rat model, we investigated the effect of the yeast alternative NADH dehydrogenase, Ndi1. We were able to achieve efficient expression of the Ndi1 protein in the mitochondria of all regions of retinal ganglion cells and axons by delivering the NDI1 gene into the optical layer of the superior colliculus. Remarkably, even after the vision of the rats was severely impaired, treatment of the animals with the NDI1 gene led to a complete restoration of the vision to the normal level. Control groups that received either empty vector or the GFP gene had no effects. CONCLUSIONS/SIGNIFICANCE: The present study reports successful manifestation of LHON-like symptoms in rats and demonstrates the potential of the NDI1 gene therapy on mitochondrial optic neuropathies. Our results indicate a window of opportunity for the gene therapy to be applied successfully after the onset of the disease symptoms.


Subject(s)
Genetic Therapy/methods , NADH Dehydrogenase/metabolism , Optic Atrophy, Hereditary, Leber/therapy , Yeasts/enzymology , Animals , Cell Line , Male , Microscopy, Electron , Mitochondria , NADH Dehydrogenase/genetics , Optic Atrophy, Hereditary, Leber/chemically induced , Optic Atrophy, Hereditary, Leber/pathology , Optic Nerve/metabolism , Optic Nerve/pathology , Optic Nerve/ultrastructure , Rats , Rats, Long-Evans , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/ultrastructure , Rotenone/toxicity
17.
Proc Natl Acad Sci U S A ; 107(20): 9105-10, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20435911

ABSTRACT

Mutations in mitochondrial oxidative phosphorylation complex I are associated with multiple pathologies, and complex I has been proposed as a crucial regulator of animal longevity. In yeast, the single-subunit NADH dehydrogenase Ndi1 serves as a non-proton-translocating alternative enzyme that replaces complex I, bringing about the reoxidation of intramitochondrial NADH. We have created transgenic strains of Drosophila that express yeast NDI1 ubiquitously. Mitochondrial extracts from NDI1-expressing flies displayed a rotenone-insensitive NADH dehydrogenase activity, and functionality of the enzyme in vivo was confirmed by the rescue of lethality resulting from RNAi knockdown of complex I. NDI1 expression increased median, mean, and maximum lifespan independently of dietary restriction, and with no change in sirtuin activity. NDI1 expression mitigated the aging associated decline in respiratory capacity and the accompanying increase in mitochondrial reactive oxygen species production, and resulted in decreased accumulation of markers of oxidative damage in aged flies. Our results support a central role of mitochondrial oxidative phosphorylation complex I in influencing longevity via oxidative stress, independently of pathways connected to nutrition and growth signaling.


Subject(s)
Aging/metabolism , Drosophila melanogaster/physiology , Electron Transport Complex I/metabolism , Longevity/physiology , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Animals , Blotting, Western , Caloric Restriction , Drosophila melanogaster/enzymology , Electron Transport Complex I/genetics , Histocytochemistry , Longevity/genetics , Mitochondria/metabolism , Oxidative Stress/genetics , Oxidative Stress/physiology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
18.
Biochemistry ; 49(13): 2973-80, 2010 Apr 06.
Article in English | MEDLINE | ID: mdl-20192260

ABSTRACT

The Ndi1 enzyme found in the mitochondrial membrane of Saccharomyces cerevisiae is an NDH-2-type alternative NADH-quinone oxidoreductase. As Ndi1 is expected to be a possible remedy for complex I defects of mammalian mitochondria, a detailed biochemical characterization of the enzyme is needed. To identify the ubiquinone (UQ) binding site in Ndi1, we conducted photoaffinity labeling using a photoreactive biotinylated UQ mimic (compound 2) synthesized following a concept of the least possible modification of the substituents on the quinone ring. Cleavage with CNBr of Ndi1 cross-linked by 2 revealed the UQ ring of 2 to be specifically cross-linked to the Phe281-Met410 region (130 amino acids). Digestion of the CNBr fragment with V8 protease and lysylendopeptidase (Lys-C) gave approximately 8 and approximately 4 kDa peptides, respectively. The approximately 8 kDa V8 digest was identified as the Thr329-Glu399 region (71 amino acids) by an N-terminal sequence analysis. Although the approximately 4 kDa Lys-C digest could not be identified by N-terminal sequence analysis, the band was thought to cover the Gly374-Lys405 region (32 amino acids). Taken together, the binding site of the Q ring of 2 must be located in a common region of the V8 protease, and Lys-C digests Gly374-Glu399 (26 amino acids). Superimposition of the Ndi1 sequence onto a three-dimensional structural model of NDH-2 from Escherichia coli suggested that the C-terminal portion of this region is close to the isoalloxazine ring of FAD.


Subject(s)
Electron Transport Complex I/chemistry , Photoaffinity Labels/chemistry , Quinone Reductases/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Ubiquinone/metabolism , Binding Sites , Electron Transport Complex I/metabolism , Mitochondrial Proteins/chemistry , Peptide Mapping/methods , Photoaffinity Labels/chemical synthesis , Protein Conformation , Quinone Reductases/metabolism , Saccharomyces cerevisiae Proteins/metabolism
19.
FEBS Lett ; 584(5): 883-8, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20074573

ABSTRACT

NADH:ubiquinone oxidoreductase (complex I) is the entry enzyme of mitochondrial oxidative phosphorylation. To obtain the structural information on inhibitor/quinone binding sites, we synthesized [3H]benzophenone-asimicin ([3H]BPA), a photoaffinity analogue of asimicin, which belongs to the acetogenin family known as the most potent complex I inhibitor. We found that [3H]BPA was photo-crosslinked to ND2, ND1 and ND5 subunits, by the three dimensional separation (blue-native/doubled SDS-PAGE) of [3H]BPA-treated bovine heart submitochondrial particles. The cross-linking was blocked by rotenone. This is the first finding that ND2 was photo-crosslinked with a potent complex I inhibitor, suggesting its involvement in the inhibitor/quinone-binding.


Subject(s)
Benzophenones/chemistry , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/chemistry , Enzyme Inhibitors/chemistry , Furans/chemistry , Animals , Cattle , Electron Transport Complex I/metabolism , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Immunoblotting , Multienzyme Complexes/metabolism , NADH, NADPH Oxidoreductases/metabolism , Protein Subunits , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
20.
J Bioenerg Biomembr ; 41(6): 493-7, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19904590

ABSTRACT

Mitochondrial impairment has been collecting more and more attention as a contributing factor to the etiology of Parkinson's disease. Above all, the NADH-quinone oxidoreductase, complex I, of the respiratory chain seems to be most culpable. Complex I dysfunction is translated to an increased production of reactive oxygen species and a decreased energy supply. In the brain, the dopaminergic neurons are one of the most susceptible cells. Their death is directly linked to the disease apparition. Developing an effective gene therapy is challenged by harmful actions of reactive oxygen species. To overcome this problem a therapeutic candidate must be able to restore the NADH-quinone oxidoreductase activity regardless of how complex I is impaired. Here we discuss the potency of the yeast alternative NADH dehydrogenase, the Ndi1 protein, to reinstate the mitochondrial respiratory chain compensating for disabled complex I and the benefit Ndi1 brings toward retardation of Parkinson's disease.


Subject(s)
Electron Transport Complex I/physiology , Parkinson Disease/therapy , Saccharomyces cerevisiae Proteins/therapeutic use , Animals , Electron Transport Complex I/therapeutic use , Genetic Therapy , Humans , Mitochondrial Proteins , Parkinson Disease/etiology , Quinone Reductases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...