Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 280
Filter
1.
Proc Natl Acad Sci U S A ; 119(27): e2123090119, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35759670

ABSTRACT

Energy-converting NADH:ubiquinone oxidoreductase, respiratory complex I, is essential for cellular energy metabolism coupling NADH oxidation to proton translocation. The mechanism of proton translocation by complex I is still under debate. Its membrane arm contains an unusual central axis of polar and charged amino acid residues connecting the quinone binding site with the antiporter-type subunits NuoL, NuoM, and NuoN, proposed to catalyze proton translocation. Quinone chemistry probably causes conformational changes and electrostatic interactions that are propagated through these subunits by a conserved pattern of predominantly lysine, histidine, and glutamate residues. These conserved residues are thought to transfer protons along and across the membrane arm. The distinct charge distribution in the membrane arm is a prerequisite for proton translocation. Remarkably, the central subunit NuoM contains a conserved glutamate residue in a position that is taken by a lysine residue in the two other antiporter-type subunits. It was proposed that this charge asymmetry is essential for proton translocation, as it should enable NuoM to operate asynchronously with NuoL and NuoN. Accordingly, we exchanged the conserved glutamate in NuoM for a lysine residue, introducing charge symmetry in the membrane arm. The stably assembled variant pumps protons across the membrane, but with a diminished H+/e- stoichiometry of 1.5. Thus, charge asymmetry is not essential for proton translocation by complex I, casting doubts on the suggestion of an asynchronous operation of NuoL, NuoM, and NuoN. Furthermore, our data emphasize the importance of a balanced charge distribution in the protein for directional proton transfer.


Subject(s)
Cell Membrane , Electron Transport Complex I , Escherichia coli Proteins , NADH Dehydrogenase , Amino Acid Substitution , Cell Membrane/chemistry , Conserved Sequence , Electron Transport Complex I/chemistry , Electron Transport Complex I/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Glutamates/chemistry , Glutamates/genetics , Lysine/chemistry , Lysine/genetics , NADH Dehydrogenase/chemistry , NADH Dehydrogenase/genetics , Protons , Quinones/chemistry
2.
Sci Rep ; 11(1): 12641, 2021 06 16.
Article in English | MEDLINE | ID: mdl-34135385

ABSTRACT

NADH:ubiquinone oxidoreductase (respiratory complex I) plays a major role in energy metabolism by coupling electron transfer from NADH to quinone with proton translocation across the membrane. Complex I deficiencies were found to be the most common source of human mitochondrial dysfunction that manifest in a wide variety of neurodegenerative diseases. Seven subunits of human complex I are encoded by mitochondrial DNA (mtDNA) that carry an unexpectedly large number of mutations discovered in mitochondria from patients' tissues. However, whether or how these genetic aberrations affect complex I at a molecular level is unknown. Here, we used Escherichia coli as a model system to biochemically characterize two mutations that were found in mtDNA of patients. The V253AMT-ND5 mutation completely disturbed the assembly of complex I, while the mutation D199GMT-ND1 led to the assembly of a stable complex capable to catalyze redox-driven proton translocation. However, the latter mutation perturbs quinone reduction leading to a diminished activity. D199MT-ND1 is part of a cluster of charged amino acid residues that are suggested to be important for efficient coupling of quinone reduction and proton translocation. A mechanism considering the role of D199MT-ND1 for energy conservation in complex I is discussed.


Subject(s)
Electron Transport Complex I/genetics , Escherichia coli/growth & development , Mitochondrial Proteins/genetics , Mutation , NADH Dehydrogenase/genetics , Adult , Benzoquinones/metabolism , Electron Transport Complex I/chemistry , Electron Transport Complex I/metabolism , Escherichia coli/genetics , Humans , Infant, Newborn , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Models, Molecular , NADH Dehydrogenase/chemistry , NADH Dehydrogenase/metabolism , Operon , Plasmids/genetics
3.
Mitochondrion ; 54: 57-64, 2020 09.
Article in English | MEDLINE | ID: mdl-32659360

ABSTRACT

Pathogenic point mutations of mitochondrial DNA (mtDNA) are associated with a large number of heterogeneous diseases involving multiple systems with which patients may present with a wide range of clinical phenotypes. In this study, we describe a novel heteroplasmic missense mutation, m.11406 T > A, of the ND4 gene encoding the subunit 4 of mitochondrial complex I in a 32-year-old woman with recurrent epileptic seizure, headache and bilateral hearing loss. Skeletal muscle histochemistry demonstrated that approximately 20% of fibers were cytochrome C oxidase (COX) deficient with increased activity of succinate dehydrogenase (SDH). Further investigations in muscle specimens showed significantly reduced level of ND4 protein. It is interesting that the subunits of complex I (ND1 and NDFUB8) and complex IV(CO1) were also remarkably decreased. These findings indicate that ND1, NDFUB8 and CO1 are more susceptible than other subunits to mutations in the mitochondrial ND4 gene.


Subject(s)
Hearing Loss, Bilateral/etiology , MELAS Syndrome/diagnostic imaging , Mutation, Missense , NADH Dehydrogenase/genetics , Seizures/etiology , Adult , Female , Genetic Predisposition to Disease , Hearing Loss, Bilateral/genetics , Humans , MELAS Syndrome/genetics , Magnetic Resonance Imaging , Male , Models, Molecular , NADH Dehydrogenase/chemistry , Pedigree , Polymorphism, Single Nucleotide , Seizures/genetics
4.
Mitochondrion ; 54: 21-25, 2020 09.
Article in English | MEDLINE | ID: mdl-32652230

ABSTRACT

Tuberculosis caused by Mycobacterium tuberculosis is one of the main global health concerns. In this study, the entire mitochondrial genome from blood samples of tuberculosis patients was analyzed to understand the possible mtDNA variants. The potential impact of non-synonymous substitutions on protein functions were determined using prediction tools. 28 non- synonymous variants were found of which 2 variants (MT-ND2 g. A > G4824 p.T119A and MT-ND6 g. T > C14180 p.Y165C) were found to be deleterious among the cases only. Majority of the variants lie in the D-loop of the non-protein coding region of the mitochondrial DNA. We propose that mutations in the mitochondrial genome need to be validated further to understand their association with tuberculosis.


Subject(s)
Mitochondria/genetics , NADH Dehydrogenase/genetics , Polymorphism, Single Nucleotide , Tuberculosis/genetics , Genetic Predisposition to Disease , Genome, Mitochondrial , High-Throughput Nucleotide Sequencing , Humans , India , NADH Dehydrogenase/chemistry , Protein Domains , Sequence Analysis, DNA , White People/genetics
5.
J Biol Chem ; 295(9): 2544-2554, 2020 02 28.
Article in English | MEDLINE | ID: mdl-31974161

ABSTRACT

Mammalian mitochondria assemble four complexes of the respiratory chain (RCI, RCIII, RCIV, and RCV) by combining 13 polypeptides synthesized within mitochondria on mitochondrial ribosomes (mitoribosomes) with over 70 polypeptides encoded in nuclear DNA, translated on cytoplasmic ribosomes, and imported into mitochondria. We have previously observed that mitoribosome assembly is inefficient because some mitoribosomal proteins are produced in excess, but whether this is the case for other mitochondrial assemblies such as the RCs is unclear. We report here that pulse-chase stable isotope labeling with amino acids in cell culture (SILAC) is a valuable technique to study RC assembly because it can reveal considerable differences in the assembly rates and efficiencies of the different complexes. The SILAC analyses of HeLa cells indicated that assembly of RCV, comprising F1/Fo-ATPase, is rapid with little excess subunit synthesis, but that assembly of RCI (i.e. NADH dehydrogenase) is far less efficient, with dramatic oversynthesis of numerous proteins, particularly in the matrix-exposed N and Q domains. Unassembled subunits were generally degraded within 3 h. We also observed differential assembly kinetics for individual complexes that were immunoprecipitated with complex-specific antibodies. Immunoprecipitation with an antibody that recognizes the ND1 subunit of RCI co-precipitated a number of proteins implicated in FeS cluster assembly and newly synthesized ubiquinol-cytochrome c reductase Rieske iron-sulfur polypeptide 1 (UQCRFS1), the Rieske FeS protein in RCIII, reflecting some coordination between RCI and RCIII assemblies. We propose that pulse-chase SILAC labeling is a useful tool for studying rates of protein complex assembly and degradation.


Subject(s)
Electron Transport Complex I/genetics , Iron-Sulfur Proteins/genetics , Mitochondria/genetics , NADH Dehydrogenase/genetics , Proton-Translocating ATPases/genetics , Cell Culture Techniques/methods , Cell Nucleus/genetics , DNA/genetics , Electron Transport/genetics , Electron Transport Complex I/chemistry , HeLa Cells , Humans , Isotope Labeling/methods , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Ribosomes/metabolism , NADH Dehydrogenase/chemistry , Peptides/genetics , Protein Transport/genetics , Proton-Translocating ATPases/chemistry
6.
Anal Chem ; 92(1): 1363-1371, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31794197

ABSTRACT

High-throughput drug discovery is highly dependent on the targets available to accelerate the process of candidates screening. Traditional chemical proteomics approaches for the screening of drug targets usually require the immobilization/modification of the drug molecules to pull down the interacting proteins. Recently, energetics-based proteomics methods provide an alternative way to study drug-protein interaction by using complex cell lysate directly without any modification of the drugs. In this study, we developed a novel energetics-based proteomics strategy, the solvent-induced protein precipitation (SIP) approach, to profile the interaction of drugs with their target proteins by using quantitative proteomics. The method is easy to use for any laboratory with the common chemical reagents of acetone, ethanol, and acetic acid. The SIP approach was able to identify the well-known protein targets of methotrexate, SNS-032, and a pan-kinase inhibitor of staurosporine in cell lysate. We further applied this approach to discover the off-targets of geldanamycin. Three known protein targets of the HSP90 family were successfully identified, and several potential off-targets including NADH dehydrogenase subunits NDUFV1 and NDUFAB1 were identified for the first time, and the NDUFV1 was validated by using Western blotting. In addition, this approach was capable of evaluating the affinity of the drug-target interaction. The data collectively proved that our approach provides a powerful platform for drug target discovery.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Methotrexate/pharmacology , NADH Dehydrogenase/antagonists & inhibitors , Oxazoles/pharmacology , Proteomics , Staurosporine/pharmacology , Thiazoles/pharmacology , Acetic Acid/chemistry , Acetone/chemistry , Cells, Cultured , Drug Discovery , Drug Evaluation, Preclinical , Ethanol/chemistry , HEK293 Cells , HSP90 Heat-Shock Proteins/chemistry , HeLa Cells , High-Throughput Screening Assays , Humans , Methotrexate/chemistry , NADH Dehydrogenase/chemistry , NADH Dehydrogenase/metabolism , Oxazoles/chemistry , Solvents/chemistry , Staurosporine/chemistry , Thiazoles/chemistry
7.
Anal Chim Acta ; 1095: 219-225, 2020 Jan 25.
Article in English | MEDLINE | ID: mdl-31864626

ABSTRACT

Herein we present a general and turn-on strategy for enzymatic bioassays on the basis of redox state dependent emission of gold nanoclusters (AuNCs). The photoluminescence of AuNCs was quenched obviously by the oxidative ferricyanide while unaffected by its corresponding reduced state, i.e., ferrocyanide. The distinctive quenching abilities for AuNCs by the redox couple (ferricyanide/ferrocyanide) enabled their utility as new fluorescent sensing platforms to detect redox-related phenomena. The proposed protocols were conducted by using the model oxidoreductases of glucose oxidase (GOx) and the enzyme cascade of lactate dehydrogenase (LDH)/diaphorase to catalytically convert ferricyanide to ferrocyanide, which switched on fluorescence of the detection systems. The detection limit for glucose and lactate was found to be as low as 0.12 and 0.09 µM, respectively. This work features the first use of the redox couple of ferricyanide/ferrocyanide in fluorescent bioanalysis, which enables versatile, signal on and highly sensitive/selective detections as compared to the state of the art fluorescently enzymatic sensing platforms. Importantly, considering the significance of ferricyanide/ferrocyanide involves in numerous other oxidoreductases mediated biocatalysis, this protocol has wide versatility that enables combination with oxidoreductases related reactions for biosensing.


Subject(s)
Fluorescent Dyes/chemistry , Glucose/analysis , Lactic Acid/analysis , Metal Nanoparticles/chemistry , Spectrometry, Fluorescence/methods , Animals , Cattle , Ferricyanides/chemistry , Fluorescence , Glucose/chemistry , Glucose Oxidase/chemistry , Gold/chemistry , Humans , L-Lactate Dehydrogenase/chemistry , Lactic Acid/chemistry , Limit of Detection , NADH Dehydrogenase/chemistry , Oxidation-Reduction , Serum Albumin, Bovine/chemistry
8.
Plant Sci ; 288: 110205, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31521217

ABSTRACT

Maize kernel size and weight are essential contributors to its yield. So the identification of the genes controlling kernel size and weight can give us a chance to gain the yield. Here, we identified a small kernel mutant, Zea mays small kernel 9 (Zmsmk9), in maize. Cytological observation showed that the development of the endosperm and embryo was delayed in Zmsmk9 mutants at the early stages, resulting in a small kernel phenotype. Interestingly, despite substantial variation in kernel size, the germination of Zmsmk9 seeds was comparable to that of WT, and could develop into normal plants with upright leaf architecture. We cloned Zmsmk9 via map-based cloning. ZmSMK9 encodes a P-type pentatricopeptide repeat protein that targets to mitochondria, and is involved in RNA splicing in mitochondrial NADH dehydrogenase5 (nad5) intron-1 and intron-4. Consistent with the delayed development phenotype, transcriptome analysis of 12-DAP endosperm showed that starch and zeins biosynthesis related genes were dramatically down regulated in Zmsmk9, while cell cycle and cell growth related genes were dramatically increased. As a result, ZmSMK9 is a novel gene required for the splicing of nad5 intron-1 and intron-4, kernel development, and plant architecture in maize.


Subject(s)
Gene Expression Regulation, Plant , NADH Dehydrogenase/genetics , Plant Proteins/genetics , Zea mays/genetics , Amino Acid Sequence , Gene Expression Profiling , Germination/genetics , Introns , Mitochondrial Proteins/metabolism , NADH Dehydrogenase/chemistry , NADH Dehydrogenase/metabolism , Plant Proteins/chemistry , Plant Proteins/metabolism , Seeds/physiology , Zea mays/growth & development
9.
Cells ; 8(10)2019 09 25.
Article in English | MEDLINE | ID: mdl-31557978

ABSTRACT

Complex I (CI) is the first enzyme of the mitochondrial respiratory chain and couples the electron transfer with proton pumping. Mutations in genes encoding CI subunits can frequently cause inborn metabolic errors. We applied proteome and metabolome profiling of patient-derived cells harboring pathogenic mutations in two distinct CI genes to elucidate underlying pathomechanisms on the molecular level. Our results indicated that the electron transfer within CI was interrupted in both patients by different mechanisms. We showed that the biallelic mutations in NDUFS1 led to a decreased stability of the entire N-module of CI and disrupted the electron transfer between two iron-sulfur clusters. Strikingly interesting and in contrast to the proteome, metabolome profiling illustrated that the pattern of dysregulated metabolites was almost identical in both patients, such as the inhibitory feedback on the TCA cycle and altered glutathione levels, indicative for reactive oxygen species (ROS) stress. Our findings deciphered pathological mechanisms of CI deficiency to better understand inborn metabolic errors.


Subject(s)
Electron Transport/genetics , Energy Metabolism/genetics , Metabolism, Inborn Errors/genetics , Metabolome/genetics , NADH Dehydrogenase/genetics , Case-Control Studies , Cells, Cultured , Cellular Reprogramming/genetics , DNA Mutational Analysis , Female , Humans , Infant , Infant, Newborn , Male , Metabolism, Inborn Errors/metabolism , Metabolism, Inborn Errors/pathology , Models, Molecular , NADH Dehydrogenase/chemistry , NADH Dehydrogenase/metabolism , Pregnancy , Primary Cell Culture , Protein Conformation , Proteome/genetics
10.
Phys Chem Chem Phys ; 21(33): 18105-18118, 2019 Aug 21.
Article in English | MEDLINE | ID: mdl-31396604

ABSTRACT

With the emergence of drug-resistant Plasmodium falciparum, the treatment of malaria has become a significant challenge; therefore, the development of antimalarial drugs acting on new targets is extremely urgent. In Plasmodium falciparum, type II nicotinamide adenine dinucleotide (NADH) dehydrogenase (NDH-2) is responsible for catalyzing the transfer of two electrons from NADH to flavin adenine dinucleotide (FAD), which in turn transfers the electrons to coenzyme Q (CoQ). As an entry enzyme for oxidative phosphorylation, NDH-2 has become one of the popular targets for the development of new antimalarial drugs. In this study, reliable motion trajectories of the NDH-2 complex with its co-factors (NADH and FAD) and inhibitor, RYL-552, were obtained by comparative molecular dynamics simulations. The influence of cofactor binding on the global motion of NDH-2 was explored through conformational clustering, principal component analysis and free energy landscape. The molecular interactions of NDH-2 before and after its binding with the inhibitor RYL-552 were analyzed, and the key residues and important hydrogen bonds were also determined. The results show that the association of RYL-552 results in the weakening of intramolecular hydrogen bonds and large allosterism of NDH-2. There was a significant positive correlation between the angular change of the key pocket residues in the NADH-FAD-pockets that represents the global functional motion and the change in distance between NADH-C4 and FAD-N5 that represents the electron transfer efficiency. Finally, the possible non-competitive inhibitory mechanism of RYL-552 was proposed. Specifically, the association of inhibitors with NDH-2 significantly affects the global motion mode of NDH-2, leading to widening of the distance between NADH and FAD through cooperative motion induction; this reduces the electron transfer efficiency of the mitochondrial respiratory chain. The simulation results provide useful theoretical guidance for subsequent antimalarial drug design based on the NDH-2 structure and the respiratory chain electron transfer mechanism.


Subject(s)
Antimalarials/chemistry , Ketones/chemistry , NADH Dehydrogenase/antagonists & inhibitors , Plasmodium falciparum/enzymology , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/chemistry , Quinolines/chemistry , Electron Transport , Flavin-Adenine Dinucleotide/chemistry , Hydrogen Bonding , Models, Molecular , Molecular Structure , NAD/chemistry , NADH Dehydrogenase/chemistry , Oxidation-Reduction , Protein Binding , Structure-Activity Relationship , Thermodynamics
11.
Sci Rep ; 9(1): 9987, 2019 07 10.
Article in English | MEDLINE | ID: mdl-31292494

ABSTRACT

Mitochondrial complex I (CI) is the largest multi-subunit oxidative phosphorylation (OXPHOS) protein complex. Recent availability of a high-resolution human CI structure, and from two non-human mammals, enabled predicting the impact of mutations on interactions involving each of the 44 CI subunits. However, experimentally assessing the impact of the predicted interactions requires an easy and high-throughput method. Here, we created such a platform by cloning all 37 nuclear DNA (nDNA) and 7 mitochondrial DNA (mtDNA)-encoded human CI subunits into yeast expression vectors to serve as both 'prey' and 'bait' in the split murine dihydrofolate reductase (mDHFR) protein complementation assay (PCA). We first demonstrated the capacity of this approach and then used it to examine reported pathological OXPHOS CI mutations that occur at subunit interaction interfaces. Our results indicate that a pathological frame-shift mutation in the MT-ND2 gene, causing the replacement of 126 C-terminal residues by a stretch of only 30 amino acids, resulted in loss of specificity in ND2-based interactions involving these residues. Hence, the split mDHFR PCA is a powerful assay for assessing the impact of disease-causing mutations on pairwise protein-protein interactions in the context of a large protein complex, thus offering a possible mechanistic explanation for the underlying pathogenicity.


Subject(s)
Frameshift Mutation , Genetic Predisposition to Disease/genetics , NADH Dehydrogenase/chemistry , NADH Dehydrogenase/genetics , Binding Sites , Cell Nucleus/genetics , Cloning, Molecular , Electron Transport Complex I/chemistry , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Humans , Mitochondria/genetics , Models, Molecular , NADH Dehydrogenase/metabolism , Protein Binding
12.
Sci Rep ; 9(1): 2775, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30808983

ABSTRACT

Sequence comparison is an essential part of modern molecular biology research. In this study, we estimated the parameters of Markov chain by considering the frequencies of occurrence of the all possible amino acid pairs from each alignment-free protein sequence. These estimated Markov chain parameters were used to calculate similarity between two protein sequences based on a fuzzy integral algorithm. For validation, our result was compared with both alignment-based (ClustalW) and alignment-free methods on six benchmark datasets. The results indicate that our developed algorithm has a better clustering performance for protein sequence comparison.


Subject(s)
Proteins/chemistry , Algorithms , Amino Acid Sequence , Electron Transport Complex I/chemistry , Electron Transport Complex I/classification , Humans , Markov Chains , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/classification , NADH Dehydrogenase/chemistry , NADH Dehydrogenase/classification , Phylogeny , Proteins/classification , Sequence Alignment
13.
Hum Mol Genet ; 28(9): 1515-1529, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30597069

ABSTRACT

Mitochondrial DNA (mtDNA) mutations have been associated with Leber's hereditary optic neuropathy (LHON) and their pathophysiology remains poorly understood. In this study, we investigated the pathophysiology of a LHON susceptibility allele (m.3394T>C, p.30Y>H) in the Mitochondrial (MT)-ND1 gene. The incidence of m.3394T>C mutation was 2.7% in the cohort of 1741 probands with LHON. Extremely low penetrances of LHON were observed in 26 pedigrees carrying only m.3394T>C mutation, while 21 families bearing m.3394T>C, together with m.11778G>A or m.14484T>C mutation, exhibited higher penetrance of LHON than those in families carrying single mtDNA mutation(s). The m.3394T>C mutation disrupted the specific electrostatic interactions between Y30 of p.MT-ND1 with the sidechain of E4 and backbone carbonyl group of M1 of NDUFA1 (NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 1) of complex I, thereby altering the structure and function of complex I. We demonstrated that these cybrids bearing only m.3394T>C mutation caused mild mitochondrial dysfunctions and those harboring both m.3394T>C and m.11778G>A mutations exhibited greater mitochondrial dysfunctions than cybrids carrying only m.11778G>A mutation. In particular, the m.3394T>C mutation altered the stability of p.MT-ND1 and complex I assembly. Furthermore, the m.3394T>C mutation decreased the activities of mitochondrial complexes I, diminished mitochondrial ATP levels and membrane potential and increased the production of reactive oxygen species in the cybrids. These m.3394T>C mutation-induced alterations aggravated mitochondrial dysfunctions associated with the m.11778G>A mutation. These resultant biochemical defects contributed to higher penetrance of LHON in these families carrying both mtDNA mutations. Our findings provide new insights into the pathophysiology of LHON arising from the synergy between mitochondrial ND1 and ND4 mutations.


Subject(s)
Alleles , Mitochondria/genetics , Mitochondria/metabolism , Mutation , NADH Dehydrogenase/genetics , Optic Atrophy, Hereditary, Leber/diagnosis , Optic Atrophy, Hereditary, Leber/genetics , Phenotype , Amino Acid Sequence , Animals , Axons/metabolism , Cell Line , Genes, Mitochondrial , Genetic Association Studies , Genetic Predisposition to Disease , Mice , NADH Dehydrogenase/chemistry , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Phosphorylation , Synaptic Transmission , Synaptic Vesicles/metabolism
14.
BMC Bioinformatics ; 20(1): 739, 2019 Dec 30.
Article in English | MEDLINE | ID: mdl-31888452

ABSTRACT

BACKGROUND: Recent advances in genome sequencing technologies and the cost drop in high-throughput sequencing continue to give rise to a deluge of data available for downstream analyses. Among others, evolutionary biologists often make use of genomic data to uncover phenotypic diversity and adaptive evolution in protein-coding genes. Therefore, multiple sequence alignments (MSA) and phylogenetic trees (PT) need to be estimated with optimal results. However, the preparation of an initial dataset of multiple sequence file(s) (MSF) and the steps involved can be challenging when considering extensive amount of data. Thus, it becomes necessary the development of a tool that removes the potential source of error and automates the time-consuming steps of a typical workflow with high-throughput and optimal MSA and PT estimations. RESULTS: We introduce LMAP_S (Lightweight Multigene Alignment and Phylogeny eStimation), a user-friendly command-line and interactive package, designed to handle an improved alignment and phylogeny estimation workflow: MSF preparation, MSA estimation, outlier detection, refinement, consensus, phylogeny estimation, comparison and editing, among which file and directory organization, execution, manipulation of information are automated, with minimal manual user intervention. LMAP_S was developed for the workstation multi-core environment and provides a unique advantage for processing multiple datasets. Our software, proved to be efficient throughout the workflow, including, the (unlimited) handling of more than 20 datasets. CONCLUSIONS: We have developed a simple and versatile LMAP_S package enabling researchers to effectively estimate multiple datasets MSAs and PTs in a high-throughput fashion. LMAP_S integrates more than 25 software providing overall more than 65 algorithm choices distributed in five stages. At minimum, one FASTA file is required within a single input directory. To our knowledge, no other software combines MSA and phylogeny estimation with as many alternatives and provides means to find optimal MSAs and phylogenies. Moreover, we used a case study comparing methodologies that highlighted the usefulness of our software. LMAP_S has been developed as an open-source package, allowing its integration into more complex open-source bioinformatics pipelines. LMAP_S package is released under GPLv3 license and is freely available at https://lmap-s.sourceforge.io/.


Subject(s)
User-Computer Interface , Amino Acid Sequence , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/classification , Humans , Mitochondrial Proton-Translocating ATPases/chemistry , Mitochondrial Proton-Translocating ATPases/classification , NADH Dehydrogenase/chemistry , NADH Dehydrogenase/classification , Phylogeny , Sequence Alignment
15.
Biochim Biophys Acta Bioenerg ; 1860(3): 201-208, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30448269

ABSTRACT

NDH-1 is a gigantic redox-driven proton pump linked with respiration and cyclic electron flow in cyanobacterial cells. Based on experimentally resolved X-ray and cryo-EM structures of the respiratory complex I, we derive here molecular models of two isoforms of the cyanobacterial NDH-1 complex involved in redox-driven proton pumping (NDH-1L) and CO2-fixation (NDH-1MS). Our models show distinct structural and dynamic similarities to the core architecture of the bacterial and mammalian respiratory complex I. We identify putative plastoquinone-binding sites that are coupled by an electrostatic wire to the proton pumping elements in the membrane domain of the enzyme. Molecular simulations suggest that the NDH-1L isoform undergoes large-scale hydration changes that support proton-pumping within antiporter-like subunits, whereas the terminal subunit of the NDH-1MS isoform lacks such structural motifs. Our work provides a putative molecular blueprint for the complex I-analogue in the photosynthetic energy transduction machinery and demonstrates that general mechanistic features of the long-range proton-pumping machinery are evolutionary conserved in the complex I-superfamily.


Subject(s)
Cyanobacteria/enzymology , Molecular Dynamics Simulation , NADH Dehydrogenase/chemistry , Antiporters , Bacterial Proteins/chemistry , Binding Sites , Electron Transport Complex I/chemistry , Energy Transfer , Molecular Structure , Photosynthesis , Plastoquinone/metabolism , Protein Isoforms , Protein Subunits , Proton Pumps
16.
J Mol Graph Model ; 85: 242-249, 2018 10.
Article in English | MEDLINE | ID: mdl-30227369

ABSTRACT

Breast cancer is the second cause of death among women all around the world. One out of every eight women is diagnosed with breast cancer in Iran. There are many reasons for cancer, one of which is the mutations in the mitochondrial genome observed in most breast cancer studies. However, the aim of this study is to evaluate the genetic region of NADH dehydrogenase subunit 4 in patients with breast cancer. First, the genomic DNA was extracted from a tissue. The NADH dehydrogenase subunit 4 coding region was amplified by PCR, and then the SSCP was sequenced. After that, the molecular dynamics were employed. The association between the mutations and the prognostic factors such as ER, PR, HER-2, and age were statistically examined. The sequence of the ND4 area was determined in 24 suspected patients, and 15 nucleotide changes were reported. The role of this variations was investigated by in-silico. The harmful mutations were predicted based on some servers. The molecular dynamics results showed that there is a significant relationship between the mutant protein and the changes in the structural conformation. Our results showed that the mutation in the ND4 area plays an important role in developing breast cancer. So, it can be concluded that the mitochondrial NADH dehydrogenase analysis may help to detect breast cancer in the early stages.


Subject(s)
Breast Neoplasms/genetics , Molecular Dynamics Simulation , NADH Dehydrogenase/chemistry , NADH Dehydrogenase/genetics , Polymorphism, Single Nucleotide , Biomarkers, Tumor , Breast Neoplasms/metabolism , Evolution, Molecular , Female , High-Throughput Nucleotide Sequencing , Humans , Iran , Mutation , NADH Dehydrogenase/metabolism , Protein Conformation
17.
Eur J Hum Genet ; 26(11): 1582-1587, 2018 11.
Article in English | MEDLINE | ID: mdl-29976978

ABSTRACT

Medical genomics research performed in diverse population facilitates a better understanding of the genetic basis of developmental disorders, with regional implications for community genetics. Autosomal recessive mitochondrial complex I deficiency (MCID) accounts for a constellation of clinical features, including encephalopathies, myopathies, and Leigh Syndrome. Using whole-exome sequencing, we identified biallelic missense variants in NDUFV1 that encodes the 51-kD subunit of complex I (NADH dehydrogenase) NDUFV1. Mapping the variants on published crystal structures of mitochondrial complex I demonstrate that the novel c.1118T > C (p.(Phe373Ser)) variant is predicted to diminish the affinity of the active pocket of NDUFV1 for FMN that correlates to an early onset of debilitating MCID symptoms. The c.1156C > T (p.(Arg386Cys)) variant is predicted to alter electron shuttling required for energy production and correlate to a disease onset in childhood. NDUFV1 c.1156C > T (p.(Arg386Cys)) represents a founder variant in South Asian populations that have value in prioritizing this variant in a population-specific manner for genetic diagnostic evaluation. In conclusion, our results demonstrate the advantage of analyzing population-specific sequences to understand the disease pathophysiology and prevalence of inherited risk variants in the underrepresented populations.


Subject(s)
Electron Transport Complex I/deficiency , Mitochondrial Diseases/genetics , Mutation, Missense , NADH Dehydrogenase/genetics , Binding Sites , Child , Electron Transport Complex I/genetics , Female , Humans , Infant , Male , Mitochondrial Diseases/pathology , NADH Dehydrogenase/chemistry
18.
Nat Struct Mol Biol ; 25(7): 548-556, 2018 07.
Article in English | MEDLINE | ID: mdl-29915388

ABSTRACT

Complex I (NADH:ubiquinone oxidoreductase) uses the reducing potential of NADH to drive protons across the energy-transducing inner membrane and power oxidative phosphorylation in mammalian mitochondria. Recent cryo-EM analyses have produced near-complete models of all 45 subunits in the bovine, ovine and porcine complexes and have identified two states relevant to complex I in ischemia-reperfusion injury. Here, we describe the 3.3-Å structure of complex I from mouse heart mitochondria, a biomedically relevant model system, in the 'active' state. We reveal a nucleotide bound in subunit NDUFA10, a nucleoside kinase homolog, and define mechanistically critical elements in the mammalian enzyme. By comparisons with a 3.9-Å structure of the 'deactive' state and with known bacterial structures, we identify differences in helical geometry in the membrane domain that occur upon activation or that alter the positions of catalytically important charged residues. Our results demonstrate the capability of cryo-EM analyses to challenge and develop mechanistic models for mammalian complex I.


Subject(s)
Electron Transport Complex I/chemistry , Mitochondria, Heart/enzymology , Animals , Binding Sites , Cryoelectron Microscopy , Electron Transport Complex I/metabolism , Electron Transport Complex I/ultrastructure , Enzyme Activation , Mice , Models, Molecular , NADH Dehydrogenase/chemistry , NADH Dehydrogenase/metabolism , NADH Dehydrogenase/ultrastructure , Nucleotides/chemistry , Nucleotides/metabolism , Phospholipids/chemistry , Phospholipids/metabolism , Protein Structural Elements , Protein Structure, Quaternary , Protein Subunits , Ubiquinone/chemistry , Ubiquinone/metabolism
19.
Bioorg Med Chem Lett ; 28(13): 2239-2243, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29859905

ABSTRACT

Energy generation is a promising area of drug discovery for both bacterial pathogens and parasites. Type II NADH dehydrogenase (NDH-2), a vital respiratory membrane protein, has attracted attention as a target for the development of new antitubercular and antimalarial agents. To date, however, no potent, specific inhibitors have been identified. Here, we performed a site-directed screening technique, tethering-fragment based drug discovery, against wild-type and mutant forms of NDH-2 containing engineered active-site cysteines. Inhibitory fragments displayed IC50 values between 3 and 110 µM against NDH-2 mutants. Possible binding poses were investigated by in silico modelling, providing a basis for optimisation of fragment binding and improved potency against NDH-2.


Subject(s)
Bacterial Proteins/metabolism , Drug Design , Enzyme Inhibitors/metabolism , Membrane Proteins/metabolism , NADH Dehydrogenase/metabolism , Bacillaceae/enzymology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Cysteine/chemistry , Cysteine/genetics , Enzyme Inhibitors/chemistry , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Docking Simulation , Mutagenesis, Site-Directed , Mutation , NADH Dehydrogenase/antagonists & inhibitors , NADH Dehydrogenase/chemistry , NADH Dehydrogenase/genetics , Protein Binding
20.
ACS Infect Dis ; 4(6): 954-969, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29522317

ABSTRACT

Mycobacterium tuberculosis ( MTb) possesses two nonproton pumping type II NADH dehydrogenase (NDH-2) enzymes which are predicted to be jointly essential for respiratory metabolism. Furthermore, the structure of a closely related bacterial NDH-2 has been reported recently, allowing for the structure-based design of small-molecule inhibitors. Herein, we disclose MTb whole-cell structure-activity relationships (SARs) for a series of 2-mercapto-quinazolinones which target the ndh encoded NDH-2 with nanomolar potencies. The compounds were inactivated by glutathione-dependent adduct formation as well as quinazolinone oxidation in microsomes. Pharmacokinetic studies demonstrated modest bioavailability and compound exposures. Resistance to the compounds in MTb was conferred by promoter mutations in the alternative nonessential NDH-2 encoded by ndhA in MTb. Bioenergetic analyses revealed a decrease in oxygen consumption rates in response to inhibitor in cells in which membrane potential was uncoupled from ATP production, while inverted membrane vesicles showed mercapto-quinazolinone-dependent inhibition of ATP production when NADH was the electron donor to the respiratory chain. Enzyme kinetic studies further demonstrated noncompetitive inhibition, suggesting binding of this scaffold to an allosteric site. In summary, while the initial MTb SAR showed limited improvement in potency, these results, combined with structural information on the bacterial protein, will aid in the future discovery of new and improved NDH-2 inhibitors.


Subject(s)
Mycobacterium tuberculosis/enzymology , NADH Dehydrogenase/chemistry , Quinazolinones/chemistry , Molecular Structure , NADH Dehydrogenase/antagonists & inhibitors , Quinazolinones/chemical synthesis , Quinazolinones/pharmacology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...