Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 132
Filter
2.
J Evol Biol ; 25(5): 981-94, 2012 May.
Article in English | MEDLINE | ID: mdl-22435703

ABSTRACT

There is a well-established allometric relationship between brain and body mass in mammals. Deviation of relatively increased brain size from this pattern appears to coincide with enhanced cognitive abilities. To examine whether there is a phylogenetic structure to such episodes of changes in encephalization across mammals, we used phylogenetic techniques to analyse brain mass, body mass and encephalization quotient (EQ) among 630 extant mammalian species. Among all mammals, anthropoid primates and odontocete cetaceans have significantly greater variance in EQ, suggesting that evolutionary constraints that result in a strict correlation between brain and body mass have independently become relaxed. Moreover, ancestral state reconstructions of absolute brain mass, body mass and EQ revealed patterns of increase and decrease in EQ within anthropoid primates and cetaceans. We propose both neutral drift and selective factors may have played a role in the evolution of brain-body allometry.


Subject(s)
Biological Evolution , Brain/physiology , Cetacea/physiology , Haplorhini/physiology , Phylogeny , Animals , Brain/anatomy & histology , Cetacea/classification , Cognition , Databases, Factual , Haplorhini/classification , Organ Size/physiology , Species Specificity , Time Factors
3.
Gene ; 267(1): 111-23, 2001 Apr 04.
Article in English | MEDLINE | ID: mdl-11311561

ABSTRACT

Cytochrome c oxidase (COX) contains ten nuclear encoded subunits, three of them known to show tissue isoforms in mammals. We have now found a fourth isoform, for subunit IV, in human, rat and mouse (COX IV-2). Comparison of the two human isoform genes shows a similar structural organization, including an overall size of about 8 kb, the presence of five exons, and the initiation of translation in the second exon, consistent with formation by gene duplication. Also consistent is the higher identity of precursor peptides of 78% within the new IV-2 isoform (average in the three species) compared to 44% average identity with the IV-1 isoform. Northern analysis and quantitative PCR with human and rat tissues show high IV-2 expression in adult lung and lower expression in all other tissues investigated, including fetal lung. In contrast, the IV-1 isoform is ubiquitously expressed. In situ hybridizations were performed to localize isoform transcripts in rat lung. Both isoforms are found in similar ratios in most lung cell types except for smooth muscle and respiratory epithelium, which have a IV-2 and a IV-1 preference, respectively. Structural modeling of the IV-2 isoform from human, based on the bovine crystal data, produces a conformation in which two of three conserved cysteine groups, exclusively present in the mammalian IV-2 isoform, are in close proximity. The formation of a cysteine bond and the implications for function of these sequence differences for subunit IV, which plays a pivotal role in COX regulation, are discussed.


Subject(s)
Cytochrome c Group/genetics , Protein Isoforms/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Cytochrome c Group/chemistry , DNA, Complementary/chemistry , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Gene Expression , Humans , In Situ Hybridization , Male , Mice , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Isoforms/chemistry , Protein Subunits , RNA/genetics , RNA/metabolism , Rats , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Tissue Distribution
4.
Mol Biol Evol ; 18(4): 563-9, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11264408

ABSTRACT

Mitochondrial DNA (mtDNA)-encoded proteins function in eukaryotes as subunits of respiratory complexes that also contain nuclear DNA (nDNA)-encoded subunits. The importance of functional interactions between mtDNA- and nDNA-encoded proteins was previously demonstrated by testing the survivability of cybrid cells or individuals containing nDNA and mtDNA from different populations or species. This report focuses on the multisubunit respiratory complex cytochrome c oxidase (COX), made up of both mtDNA-encoded and nDNA-encoded subunits. A combination of evolutionary and crystallographic data is employed to determine whether rates of nonsynonymous substitutions have been higher, the same, or lower for residues in close proximity that are encoded by a different genome (nDNA or mtDNA). This determination is performed by simply taking the ratio, called the interaction ratio i, of the nonsynonymous substitution rate of the close-contact residues to the nonsynonymous substitution rate of the noncontact residues. We assume that the close-contact residues (which are more likely to interact) are functionally important and that, therefore, amino acid replacements among these residues cannot escape the scrutiny of natural selection. i = 1 indicates that the close-contact residues have been under neither greater purifying selection nor greater positive selection than the noncontact residues as a specific consequence of their being encoded by separate genomes. i < 1 indicates that the close-contact residues have been under greater purifying selection but less positive selection than have the noncontact residues. Conversely, i > 1 indicates that the close-contact residues have been under less purifying but greater positive selection than have the noncontact residues. i < 1 may be referred to as a constraining interaction; i.e., the close-contact residues compared with the noncontact residues appear to be under greater structural-functional constraints. On the other hand, i > 1 may be referred to as an optimizing interaction; i.e., apparently many different amino acid replacements are required to optimize this subunit's interaction with the other subunit. A major finding is that the nDNA-encoded residues in close physical proximity to mtDNA-encoded residues evolve more slowly than the other nuclear-encoded residues (and thus display a constraining interaction), whereas the mtDNA-encoded residues in close physical proximity to nDNA-encoded residues evolve more rapidly than the other mitochondrial-encoded residues (and thus display an optimizing interaction). A possible reason for this striking difference between the nuclear- and mitochondrial-encoded COX subunits in how their functional interaction evolves is discussed.


Subject(s)
Amino Acid Substitution/genetics , Electron Transport Complex IV/genetics , Evolution, Molecular , Animals , DNA/analysis , DNA/genetics , DNA, Mitochondrial/genetics , Electron Transport Complex IV/metabolism , Humans , Macromolecular Substances , Models, Molecular , Phylogeny , Protein Subunits
5.
Mol Phylogenet Evol ; 18(1): 26-36, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11161739

ABSTRACT

As part of our goal to reconstruct human evolution at the DNA level, we have been examining changes in the biochemical machinery for aerobic energy metabolism. We find that protein subunits of two of the electron transfer complexes, complex III and complex IV, and cytochrome c, the protein carrier that connects them, have all undergone a period of rapid protein evolution in the anthropoid lineage that ultimately led to humans. Indeed, subunit IV of cytochrome c oxidase (COX; complex IV) provides one of the best examples of positively selected changes of any protein studied. The rate of subunit IV evolution accelerated in our catarrhine ancestors in the period between 40 to 18 million years ago and then decelerated in the descendant hominid lineages, a pattern of rate changes indicative of positive selection of adaptive changes followed by purifying selection acting against further changes. Besides clear evidence that adaptive evolution occurred for cytochrome c and subunits of complexes III (e.g., cytochrome c(1)) and IV (e.g., COX2 and COX4), modest rate accelerations in the lineage that led to humans are seen for other subunits of both complexes. In addition the contractile muscle-specific isoform of COX subunit VIII became a pseudogene in an anthropoid ancestor of humans but appears to be a functional gene in the nonanthropoid primates. These changes in the aerobic energy complexes coincide with the expansion of the energy-dependent neocortex during the emergence of the higher primates. Discovering the biochemical adaptations suggested by molecular evolutionary analysis will be an exciting challenge.


Subject(s)
Biological Evolution , Evolution, Molecular , Primates/genetics , Animals , Cytochrome c Group/genetics , Electron Transport , Electron Transport Complex IV/genetics , Humans , Models, Biological , Models, Genetic , Mutation , Phylogeny , Protein Isoforms
6.
Mol Phylogenet Evol ; 17(2): 294-304, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11083942

ABSTRACT

Phylogenetic analyses carried out on cytochrome c oxidase (COX) subunit I mitochondrial genes from 14 primates representing the major branches of the order and four outgroup nonprimate eutherians revealed that transversions and amino acid replacements (i.e., the more slowly occurring sequence changes) contained lower levels of homoplasy and thus provided more accurate information on cladistic relationships than transitions (i.e., the more rapidly occurring sequence changes). Several amino acids, each with a high likelihood of functionality involving the binding of cytochrome c or interaction with COX VIII, have changed in Anthropoidea, the primate suborder grouping New World monkey, Old World monkey, ape, and human lineages. They are conserved in other mammalian lineages and in nonanthropoid primates. Maximum-likelihood ancestral COX I nucleotide sequences were determined utilizing a near most parsimonious branching arrangement for the primate sequences that was consistent with previously hypothesized primate cladistic relationships based on larger and more diverse data sets. Relative rate tests of COX I mitochondrial sequences showed an elevated nonsynonymous (N) substitution rate for anthropoid-nonanthropoid comparisons. This finding for the largest mitochondrial (mt) DNA-encoded subunit is consistent with previous observations of elevated nonsynonymous substitution/synonymous substitution (S) rates in primates for mt-encoded COX II and for the nuclear-encoded COX IV and COX VIIa-H. Other COX-related proteins, including cytochrome c and cytochrome b, also show elevated amino acid replacement rates or N/S during similar time frames, suggesting that this group of interacting genes is likely to have coevolved during primate evolution.


Subject(s)
Electron Transport Complex IV/genetics , Evolution, Molecular , Primates/genetics , Amino Acid Sequence , Animals , Base Sequence , COS Cells , DNA/chemistry , DNA/genetics , DNA, Mitochondrial/chemistry , DNA, Mitochondrial/genetics , Molecular Sequence Data , Phylogeny , Primates/classification , Protein Subunits , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
7.
J Neurol Sci ; 179(S 1-2): 1-33, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11054482

ABSTRACT

Brain ischemia and reperfusion engage multiple independently-fatal terminal pathways involving loss of membrane integrity in partitioning ions, progressive proteolysis, and inability to check these processes because of loss of general translation competence and reduced survival signal-transduction. Ischemia results in rapid loss of high-energy phosphate compounds and generalized depolarization, which induces release of glutamate and, in selectively vulnerable neurons (SVNs), opening of both voltage-dependent and glutamate-regulated calcium channels. This allows a large increase in cytosolic Ca(2+) associated with activation of mu-calpain, calcineurin, and phospholipases with consequent proteolysis of calpain substrates (including spectrin and eIF4G), activation of NOS and potentially of Bad, and accumulation of free arachidonic acid, which can induce depletion of Ca(2+) from the ER lumen. A kinase that shuts off translation initiation by phosphorylating the alpha-subunit of eukaryotic initiation factor-2 (eIF2alpha) is activated either by adenosine degradation products or depletion of ER lumenal Ca(2+). Early during reperfusion, oxidative metabolism of arachidonate causes a burst of excess oxygen radicals, iron is released from storage proteins by superoxide-mediated reduction, and NO is generated. These events result in peroxynitrite generation, inappropriate protein nitrosylation, and lipid peroxidation, which ultrastructurally appears to principally damage the plasmalemma of SVNs. The initial recovery of ATP supports very rapid eIF2alpha phosphorylation that in SVNs is prolonged and associated with a major reduction in protein synthesis. High catecholamine levels induced by the ischemic episode itself and/or drug administration down-regulate insulin secretion and induce inhibition of growth-factor receptor tyrosine kinase activity, effects associated with down-regulation of survival signal-transduction through the Ras pathway. Caspase activation occurs during the early hours of reperfusion following mitochondrial release of caspase 9 and cytochrome c. The SVNs find themselves with substantial membrane damage, calpain-mediated proteolytic degradation of eIF4G and cytoskeletal proteins, altered translation initiation mechanisms that substantially reduce total protein synthesis and impose major alterations in message selection, down-regulated survival signal-transduction, and caspase activation. This picture argues powerfully that, for therapy of brain ischemia and reperfusion, the concept of single drug intervention (which has characterized the approaches of basic research, the pharmaceutical industry, and clinical trials) cannot be effective. Although rigorous study of multi-drug protocols is very demanding, effective therapy is likely to require (1) peptide growth factors for early activation of survival-signaling pathways and recovery of translation competence, (2) inhibition of lipid peroxidation, (3) inhibition of calpain, and (4) caspase inhibition. Examination of such protocols will require not only characterization of functional and histopathologic outcome, but also study of biochemical markers of the injury processes to establish the role of each drug.


Subject(s)
Brain Ischemia/metabolism , Nerve Degeneration/metabolism , Reperfusion Injury/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis/physiology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Calpain/metabolism , Cell Differentiation/physiology , Cerebrovascular Circulation/physiology , Excitatory Amino Acids/metabolism , Free Radicals/metabolism , Genes, Immediate-Early/physiology , Growth Substances/metabolism , Humans , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/biosynthesis , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Signal Transduction/physiology
8.
Biochim Biophys Acta ; 1492(1): 252-8, 2000 Jun 21.
Article in English | MEDLINE | ID: mdl-11004498

ABSTRACT

The gene for human cytochrome c oxidase subunit VIIa liver isoform (COX7AL) was isolated and its sequence determined and analyzed. The three introns of the gene are considerably larger than those of the heart isoform of subunit VIIa (COX7AH), but the position of the introns relative to the cDNA sequences is homologous between the two genes. Comparison with other isolated COX7AL genes suggests that the promoter region binding motifs for transcription factors have evolved along with the coding region. In fibroblasts cultured originally from a Leigh's disease patient, a shortened COX7AL cDNA was identified by RT-PCR, consisting of exon I joined to exon IV, omitting exons II and III. No mutation could be identified in COX7AL of the patient, suggesting that the shortened cDNA is due to an alteration of the genome during cell culture. A surprising transcription of COX7AH was observed in cultured fibroblasts, suggesting a potential utility of these cells for study of its gene expression.


Subject(s)
Electron Transport Complex IV/genetics , Genome, Human , Amino Acid Sequence , Base Sequence , Cells, Cultured , DNA/analysis , Electron Transport Complex IV/isolation & purification , Fibroblasts/physiology , Humans , Leigh Disease/genetics , Molecular Sequence Data , RNA, Messenger/metabolism , Sequence Homology, Nucleic Acid
9.
Mamm Genome ; 10(5): 506-12, 1999 May.
Article in English | MEDLINE | ID: mdl-10337626

ABSTRACT

We identified a novel human gene, NOC4 (Neighbor Of COX4), located 5' to COX4, the gene for cytochrome c oxidase subunit IV, on Chr 16q32-ter. Transcripts from this gene were identified among human expressed sequence tags. A full-length, 1.06-kb human retinal NOC4 cDNA encoded a 24-kDa, 210-amino acid hypothetical protein of unknown function. Northern hybridization analysis of human RNAs from various tissues detected NOC4 transcripts of 2.2 and 1.4 kb in all tissues examined, suggesting that NOC4 expression is ubiquitous. Transcription of both the COX4 and NOC4 genes initiates within a 250-bp intergenic promoter and occurs in opposite directions. The bidirectional promoter is G + C-rich, lacks TATA and CCAAT elements, and contains multiple potential binding sites for Sp1 and NRF-2/GABP. Two of the NRF-2/GABP sites are located within 14-bp direct repeats, a conserved feature of mammalian COX4 promoters. The NOC4 and COX4 genes are also linked in the rat, mouse, and bovine genomes. A NOC4-GFP fusion protein is located in both the nucleus and the cytoplasm, including the mitochondria.


Subject(s)
Electron Transport Complex IV/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA, Complementary , Genes, Overlapping , Humans , Microscopy, Fluorescence , Molecular Sequence Data , Promoter Regions, Genetic , Recombinant Fusion Proteins/genetics , Subcellular Fractions/metabolism
10.
Mol Biol Evol ; 16(5): 619-26, 1999 May.
Article in English | MEDLINE | ID: mdl-10335655

ABSTRACT

COX VIIa is one of 10 nuclear-encoded subunits of the COX holoenzyme, and one of three that have isoforms with tissue-specific differences in expression. Analysis of nucleotide substitution rates revealed an accelerated rate of nonsynonymous substitutions relative to that of synonymous substitutions for the heart isoform gene (COX7AH) in six primate lineages. Rate accelerations have been noted for four other COX-related genes in this time period, suggesting that the COX holoenzyme has experienced an episode of adaptive evolution. A third member of the gene family, COX7AR, has recently been described. Although its function is currently unknown, low nonsynonymous substitution/synonymous substitution (N/S) ratios in mammalian evolution suggest that COX7AR is of functional importance. When the COX7A isoforms were divided into domains, examination of nucleotide substitution rates suggested that mitochondrial targeting residues experienced an accelerated nonsynonymous substitution rate in the period following gene duplication. In contrast, paralogous comparisons of the targeting residues of each isoform show they have been relatively conserved in mammalian evolution. This pattern is consistent with the evolution of tissue-specific function.


Subject(s)
Electron Transport Complex IV/genetics , Evolution, Molecular , Multigene Family/genetics , Primates/physiology , Amino Acid Sequence , Animals , Genetic Variation , Humans , Isoenzymes/genetics , Molecular Sequence Data , Myocardium/enzymology , Organ Specificity , Sequence Homology, Amino Acid
11.
Electrophoresis ; 19(8-9): 1254-9, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9694260

ABSTRACT

The cytochrome c oxidase (COX) holoenzyme is a 13-subunit complex that carries out the terminal step in the electron transport chain. Three of the subunits, which contain the electron transfer function, are coded by mitochondrial DNA and the other ten subunits by nuclear DNA. Since the holoenzyme contains equivalent amounts of each subunit, we and others have examined transcriptional regulation of COX nuclear subunits to explore whether there is a common basis for co-regulation. Each gene is seen to have a unique pattern of recognition by regulatory factors; although some factors bind to more than one gene, not all COX genes seem to be regulated by the same set of factors. Current information about the COX promoters that have been examined is summarized, and the relation of promoter regulation to coordinate gene expression is discussed.


Subject(s)
Electron Transport Complex IV/genetics , Gene Expression Regulation, Enzymologic , Promoter Regions, Genetic , Transcription, Genetic , Animals , Base Sequence , DNA, Complementary , Humans , Mammals , Molecular Sequence Data
12.
Genomics ; 49(3): 363-70, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9615220

ABSTRACT

We have isolated and examined the gene for the heart isoform of cytochrome c oxidase subunit VIIa (COX VIIa-H) in mouse, an isoform gene previously thought to be lacking in rodents. Interspecies amino acid comparisons indicate that mouse COX VIIa-H protein displays 82.5 and 70.9% identity with the bovine and human heart isoforms of COX VIIa, but only 53.7% identity with the paralogous mouse liver isoform (COX VIIa-L). Expression in adult mouse tissues is limited to heart and skeletal muscle, as found in other species. In the early mouse embryo, Cox7al was the exclusive isoform expressed and Cox7ah mRNA was not detectable until day 17 postcoitum. That the mouse Cox7ah gene characterized in this study is orthologous to the human COX7AH gene was also suggested by its mapping to mouse chromosome 7, to a conserved region syntenic with the human chromosome location of COX7AH, 19q13.1. As a result, all three COX heart isoform genes in mouse group to chromosome 7. Interestingly, mapping of the mouse Cox7al to chromosome 9 suggests a new syntenic region between the mouse and the human genomes.


Subject(s)
Chromosome Mapping , Electron Transport Complex IV/genetics , Mice/genetics , Amino Acid Sequence , Animals , Base Sequence , Cattle , Crosses, Genetic , Electron Transport Complex IV/biosynthesis , Electron Transport Complex IV/chemistry , Genetic Markers , Humans , Isoenzymes/biosynthesis , Isoenzymes/chemistry , Isoenzymes/genetics , Macromolecular Substances , Mice, Inbred C57BL , Molecular Sequence Data , Muridae , Myocardium/enzymology , Organ Specificity , Sequence Alignment , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Transcription, Genetic
13.
Mol Biol Evol ; 14(6): 595-601, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9190060

ABSTRACT

Cytochrome c oxidase (COX) consists of 13 subunits, 3 encoded in the mitochondrial genome and 10 in the nucleus. Little is known of the role of the nuclear-encoded subunits, some of which exhibit tissue-specific isoforms. Subunit VIa is unique in having tissue-specific isoforms in all mammalian species examined. We examined relative evolutionary rates for the COX6A heart (H) and liver (L) isoform genes along the length of the molecule, specifically in relation to the tissue-specific function(s) of the two isoforms. Nonsynonymous (amino acid replacement) substitutions in the COX6AH gene occurred more frequently than in the ubiquitously expressed COX6AL gene. Maximum-parsimony analysis and sequence divergences from reconstructed ancestral sequences revealed that after the ancestral COX6A gene duplicated to yield the genes for the H and L isoforms, the sequences encoding the mitochondrial matrix region of the COX VIa protein experienced an elevated rate of nonsynonymous substitutions relative to synonymous substitutions. This is expected for relaxed selective constraints after gene duplication followed by purifying selection to preserve the replacements with tissue-specific functions.


Subject(s)
Electron Transport Complex IV/genetics , Evolution, Molecular , Genetic Variation , Amino Acid Sequence , Animals , Binding Sites , Cattle , Cloning, Molecular , Electron Transport Complex IV/metabolism , Humans , Isoenzymes , Liver/enzymology , Mice , Models, Genetic , Molecular Sequence Data , Myocardium/enzymology , Organ Specificity , Phylogeny , Rats , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
15.
J Mol Evol ; 44(5): 477-91, 1997 May.
Article in English | MEDLINE | ID: mdl-9115172

ABSTRACT

Cytochrome c oxidase (COX) is a multi-subunit enzyme complex that catalyzes the final step of electron transfer through the respiratory chain on the mitochondrial inner membrane. Up to 13 subunits encoded by both the mitochondrial (subunits I, II, and III) and nuclear genomes occur in eukaryotic organisms ranging from yeast to human. Previously, we observed a high number of amino acid replacements in the human COX IV subunit compared to mouse, rat, and cow orthologues. Here we examined COX IV evolution in the two groups of anthropoid primates, the catarrhines (hominoids, cercopithecoids) and platyrrhines (ceboids), as well as one prosimian primate (lorisiform), by sequencing PCR-amplified portions of functional COX4 genes from genomic DNAs. Phylogenetic analysis of the COX4 sequence data revealed that accelerated nonsynonymous substitution rates were evident in the early evolution of both catarrhines and, to a lesser extent, platyrrhines. These accelerated rates were followed later by decelerated rates, suggesting that positive selection for adaptive amino acid replacement became purifying selection, preserving replacements that had occurred. The evidence for positive selection was especially pronounced along the catarrhine lineage to hominoids in which the nonsynonymous rate was first faster than the synonymous rate, then later much slower. The rates of three types of "neutral DNA" nucleotide substitutions (synonymous substitutions, pseudogene nucleotide substitutions, and intron nucleotide substitutions) are similar and are consistent with previous observations of a slower rate of such substitutions in the nuclear genomes of hominoids than in the nuclear genomes of other primate and mammalian lineages.


Subject(s)
Electron Transport Complex IV/genetics , Evolution, Molecular , Primates/genetics , Amino Acid Sequence , Animals , Base Sequence , Exons/genetics , Hominidae/genetics , Humans , Introns/genetics , Molecular Sequence Data , Phylogeny , Pseudogenes/genetics , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
16.
J Biol Chem ; 272(15): 10175-81, 1997 Apr 11.
Article in English | MEDLINE | ID: mdl-9092564

ABSTRACT

Cytochrome c oxidase (COX) subunit VIIc is one of the nuclear encoded subunits of the 13-subunit holoenzyme that carries out the terminal step in the electron transport chain. We have isolated the gene for this subunit, previously shown to be ubiquitously expressed from a single copy gene in the genome, and show that 167 base pairs of DNA surrounding the transcriptional start site contain the minimal promoter of this gene. This basal promoter contains two YY1 sites and at least one site for NRF-2, which show binding to their cognate factors. Mutation of both YY1 sites eliminates most of the promoter activity. Mutation at the upstream YY1 site significantly reduces the efficiency of transcript initiation at the major start site and thus plays the dominant role in COX7C regulation. COX7C is, thus, the second nuclear gene of COX that is regulated by YY1, suggesting that it is a third common factor, along with NRF-1 and NRF-2, to be associated with COX gene regulation.


Subject(s)
DNA-Binding Proteins/chemistry , Electron Transport Complex IV/chemistry , Electron Transport Complex IV/genetics , Gene Expression Regulation, Enzymologic , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Promoter Regions, Genetic , Transcription Factors/chemistry , Zinc Fingers , Animals , Base Sequence , Cattle , DNA-Binding Proteins/metabolism , Erythroid-Specific DNA-Binding Factors , GA-Binding Protein Transcription Factor , HeLa Cells , Humans , Leucine Zippers , Molecular Sequence Data , NF-E2-Related Factor 1 , Nuclear Respiratory Factor 1 , Nuclear Respiratory Factors , Protein Conformation , Restriction Mapping , Trans-Activators/metabolism , Transcription Factors/metabolism , YY1 Transcription Factor
17.
Bioessays ; 18(12): 983-91, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8976155

ABSTRACT

Mitochondria contain a molecular genetic system to express the 13 protein components of the electron transport system encoded in the mitochondrial genome (mtDNA). Defects in the function of this system result in some diseases, many of which are multisystem disorders, prominently involving highly aerobic, postmitotic tissues. These defects can be caused by large-scale rearrangements of mtDNA, by point mutations, or by nuclear gene mutations resulting in abnormalities in mtDNA. Although any of these mutations would be expected to produce a similar clinical phenotype by compromising oxidative phosphorylation, the surprising and puzzling result is that different clinical phenotypes are generally associated with specific mtDNA mutations. Moreover, the same mutation can produce a distinct clinical phenotype in different individuals or pedigrees. MtDNA rearrangements are also found in aged individuals, but at a subclinical level, suggesting that normal and pathological processes can differ by the effect of genetic or environmental factors on the error rate of mtDNA replication.


Subject(s)
DNA, Mitochondrial/chemistry , DNA, Mitochondrial/genetics , Mitochondria/metabolism , Mitochondrial Myopathies/genetics , Aged , Base Sequence , Gene Rearrangement , Genotype , Humans , Mitochondrial Myopathies/metabolism , Molecular Sequence Data , Mutation , Oxygen Consumption , Phenotype
18.
Ann Emerg Med ; 27(5): 588-94, 1996 May.
Article in English | MEDLINE | ID: mdl-8629779

ABSTRACT

Brain damage accompanying cardiac arrest and resuscitation is frequent and devastating. Neurons in the hippocampus CA1 and CA4 zones and cortical layers III and V are selectively vulnerable to death after injury by ischemia and reperfusion. Ultrastructural evidence indicates that most of the structural damage is associated with reperfusion, during which the vulnerable neurons develop disaggregation of polyribosomes, peroxidative damage to unsaturated fatty acids in the plasma membrane, and prominent alterations in the structure of the Golgi apparatus that is responsible for membrane assembly. Reperfusion is also associated with vulnerable neurons with prominent production of messenger RNAs for stress proteins and for the proteins of the activator protein-1 complex, but these vulnerable neurons fail to efficiently translate these messages into the proteins. The inhibition of protein synthesis during reperfusion involves alteration of translation initiation factors, specifically serine phosphorylation of the alpha-subunit of eukaryotic initiation factor-2 (elF-2 alpha). Growth factors--in particular, insulin--have the potential to reverse phosphorylation of elF-2 alpha, promote effective translation of the mRNA transcripts generated in response to ischemia and reperfusion, enhance neuronal defenses against radicals, and stimulate lipid synthesis and membrane repair. There is now substantial evidence that the insulin-class growth factors have neuron-sparing effects against damage by radicals and ischemia and reperfusion. This new knowledge may provide a fundamental basis for a rational approach to "cerebral resuscitation" that will allow substantial amelioration of the often dismal neurologic outcome now associated with resuscitation from cardiac arrest.


Subject(s)
Brain Ischemia/etiology , Cardiopulmonary Resuscitation , Heart Arrest/complications , Reperfusion Injury/etiology , Brain Ischemia/metabolism , Brain Ischemia/therapy , Growth Substances/therapeutic use , Hippocampus/blood supply , Hippocampus/injuries , Humans , Oxidative Stress/physiology , Protein Biosynthesis , Reperfusion Injury/metabolism , Reperfusion Injury/therapy , Risk Factors
19.
J Biol Chem ; 271(4): 2112-20, 1996 Jan 26.
Article in English | MEDLINE | ID: mdl-8567667

ABSTRACT

Cytochrome c oxidase subunit VIIa is specified by two nuclear genes, one (COX7AH) producing a heart/muscle-specific isoform and the other (COX7AL) a form expressed in all tissues. We have isolated both genes to examine their transcriptional regulation. Here, we characterize the core promoter of COX7AL and show that a 92-base pair region flanking the 5'-end promotes most of the activity of this gene. The 92-bp basal promoter contains sites for the nuclear respiratory factors NRF-1 and NRF-2, which have been shown to contribute to the transcription of a number of nuclear genes involved in mitochondrial respiratory activity, and also at least four Sp1 motifs. We show that both the NRF-1 and NRF-2 binding sites are functional in COX7AL and present evidence suggesting that interaction between the NRF-1 site and an upstream element contributes to expression.


Subject(s)
Electron Transport Complex IV/genetics , Isoenzymes/genetics , Liver/enzymology , Promoter Regions, Genetic , Animals , Base Sequence , Binding, Competitive , Cattle , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic , Molecular Sequence Data , Mutagenesis, Site-Directed , RNA, Messenger/genetics , Restriction Mapping , Structure-Activity Relationship , Transcription Factors/genetics
20.
Biochim Biophys Acta ; 1260(3): 361-4, 1995 Feb 21.
Article in English | MEDLINE | ID: mdl-7873616

ABSTRACT

A cDNA encoding cytochrome c oxidase (COX) subunit VIa liver isoform (COX6aL) was isolated from a Mus musculus library and sequenced. The protein translated from the nucleotide sequence contains a presequence and is 91% identical to the human cognate sequence over the processed polypeptide region. Northern analysis shows the expression of COX6aL is developmentally regulated in heart, being about equally transcribed with the heart isoform (COX6aH) in 18-day embryos but consisting of less than 25% in adult heart.


Subject(s)
Electron Transport Complex IV/genetics , Liver/enzymology , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary , Electron Transport Complex IV/chemistry , Humans , Mice , Molecular Sequence Data , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...