Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Mol Cell Biol ; 21(11): 3738-49, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11340167

ABSTRACT

The thermogenic peroxisome proliferator-activated receptor gamma (PPAR-gamma) coactivator 1 (PGC-1) has previously been shown to activate mitochondrial biogenesis in part through a direct interaction with nuclear respiratory factor 1 (NRF-1). In order to identify related coactivators that act through NRF-1, we searched the databases for sequences with similarities to PGC-1. Here, we describe the first characterization of a 177-kDa transcriptional coactivator, designated PGC-1-related coactivator (PRC). PRC is ubiquitously expressed in murine and human tissues and cell lines; but unlike PGC-1, PRC was not dramatically up-regulated during thermogenesis in brown fat. However, its expression was down-regulated in quiescent BALB/3T3 cells and was rapidly induced by reintroduction of serum, conditions where PGC-1 was not detected. PRC activated NRF-1-dependent promoters in a manner similar to that observed for PGC-1. Moreover, NRF-1 was immunoprecipitated from cell extracts by antibodies directed against PRC, and both proteins were colocalized to the nucleoplasm by confocal laser scanning microscopy. PRC interacts in vitro with the NRF-1 DNA binding domain through two distinct recognition motifs that are separated by an unstructured proline-rich region. PRC also contains a potent transcriptional activation domain in its amino terminus adjacent to an LXXLL motif. The spatial arrangement of these functional domains coincides with those found in PGC-1, supporting the conclusion that PRC and PGC-1 are structurally and functionally related. We conclude that PRC is a functional relative of PGC-1 that operates through NRF-1 and possibly other activators in response to proliferative signals.


Subject(s)
DNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/genetics , Transcriptional Activation , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , DNA, Complementary , Humans , Male , Mammals , Mice , Mice, Inbred BALB C , Molecular Sequence Data , NF-E2-Related Factor 1 , Nuclear Respiratory Factor 1 , Nuclear Respiratory Factors , Trans-Activators/genetics , Tumor Cells, Cultured
2.
Mol Cell Biol ; 21(2): 644-54, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11134350

ABSTRACT

In vitro studies have implicated nuclear respiratory factor 1 (NRF-1) in the transcriptional expression of nuclear genes required for mitochondrial respiratory function, as well as for other fundamental cellular activities. We investigated here the in vivo function of NRF-1 in mammals by disrupting the gene in mice. A portion of the NRF-1 gene that encodes the nuclear localization signal and the DNA-binding and dimerization domains was replaced through homologous recombination by a beta-galactosidase-neomycin cassette. In the mutant allele, beta-galactosidase expression is under the control of the NRF-1 promoter. Embryos homozygous for NRF-1 disruption die between embryonic days 3.5 and 6.5. beta-Galactosidase staining was observed in growing oocytes and in 2. 5- and 3.5-day-old embryos, demonstrating that the NRF-1 gene is expressed during oogenesis and during early stages of embryogenesis. Moreover, the embryonic expression of NRF-1 did not result from maternal carryover. While most isolated wild-type and NRF-1(+/-) blastocysts can develop further in vitro, the NRF-1(-/-) blastocysts lack this ability despite their normal morphology. Interestingly, a fraction of the blastocysts from heterozygous matings had reduced staining intensity with rhodamine 123 and NRF-1(-/-) blastocysts had markedly reduced levels of mitochondrial DNA (mtDNA). The depletion of mtDNA did not coincide with nuclear DNA fragmentation, indicating that mtDNA loss was not associated with increased apoptosis. These results are consistent with a specific requirement for NRF-1 in the maintenance of mtDNA and respiratory chain function during early embryogenesis.


Subject(s)
Blastocyst/cytology , Blastocyst/metabolism , DNA, Mitochondrial/genetics , DNA-Binding Proteins/genetics , Fetal Death , Mutagenesis, Insertional/genetics , Trans-Activators/genetics , Animals , Cell Division , DNA Fragmentation , DNA, Mitochondrial/analysis , DNA-Binding Proteins/metabolism , Embryonic and Fetal Development , Female , Gene Deletion , Gene Dosage , Gene Targeting , Genotype , Histocytochemistry , In Situ Nick-End Labeling , Mice , Mice, Knockout , NF-E2-Related Factor 1 , Nuclear Respiratory Factor 1 , Nuclear Respiratory Factors , Ovary/embryology , Ovary/metabolism , Ovum/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trans-Activators/metabolism
3.
J Cell Sci ; 113 Pt 23: 4263-73, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11069771

ABSTRACT

Nuclear respiratory factor-1 is a transcriptional activator that has been implicated in the nuclear control of respiratory chain expression. Yeast two-hybrid screens were performed to identify proteins that physically interact with nuclear respiratory factor-1. Saturation screening of both mouse embryo and mouse testis libraries yielded 14 independent clones, all of which represented two different isoforms of dynein light chain. In addition to using the two-hybrid method, the specificity of the nuclear respiratory factor-1/dynein light chain interaction was established by chemical crosslinking of the purified native proteins and by co-immunoprecipitation of nuclear respiratory factor-1 and dynein light chain from mammalian cells. Both two-hybrid and chemical crosslinking assays demonstrated that binding of dynein light chain required the first 26 amino acids of nuclear respiratory factor-1. Although dynein light chain is associated with dynein, a cytoplasmic motor molecule, immunolocalizations showed substantial nuclear staining using several different anti-dynein light chain antibodies. Moreover, fluorescence overlays of confocal images established that nuclear respiratory factor-1 and dynein light chain displayed a very similar nuclear staining pattern. The significance of the nuclear respiratory factor-1/dynein light chain interaction was investigated further by determining whether a similar interaction was conserved between dynein light chain and the erect wing gene product of Drosophila, a protein related to nuclear respiratory factor-1 through its DNA binding domain. Here, we establish that the erect wing gene product can bind and trans-activate transcription through authentic nuclear respiratory factor-1 binding sites. Moreover, the erect wing gene product, like nuclear respiratory factor-1, interacted specifically with dynein light chain both in vitro and in transfected cells. Thus, the interaction with dynein light chain is conserved between transcription factors that are structurally and functionally similar between humans and Drosophila.


Subject(s)
Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins , Neuropeptides/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Nucleus/metabolism , Conserved Sequence , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Drosophila , Dyneins , Humans , Mammals , Molecular Sequence Data , NF-E2-Related Factor 1 , Neuropeptides/chemistry , Neuropeptides/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Respiratory Factor 1 , Nuclear Respiratory Factors , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding/physiology , Sequence Homology, Amino Acid , Structure-Activity Relationship , Trans-Activators/chemistry , Trans-Activators/genetics , Transfection , Two-Hybrid System Techniques
4.
J Biol Chem ; 275(17): 13134-41, 2000 Apr 28.
Article in English | MEDLINE | ID: mdl-10777619

ABSTRACT

Progression through the cell cycle requires ATP for protein synthesis, cytoskeletal rearrangement, chromatin remodeling, and protein degradation. The mechanisms by which mammalian cells increase respiratory capacity and ATP production in preparation for cell division are largely unexplored. Here, we demonstrate that serum induction of cytochrome c mRNA and processed protein in quiescent BALB/3T3 fibroblasts is associated with a marked increase in mitochondrial respiration. Cytochrome c was induced in the absence of any increase in citrate synthase activity or in subunit IV of the cytochrome c oxidase complex mRNA or protein, indicating that the enhanced respiratory rate did not require a general increase in mitochondrial biogenesis or respiratory chain expression. Transfections with a series of cytochrome c promoter mutants showed that both nuclear respiratory factor 1 (NRF-1) and cAMP-response element-binding protein (CREB) binding sites contributed equally to induced expression by serum. Moreover, CREB and NRF-1 were phosphorylated sequentially in response to serum, and the NRF-1 phosphorylation was accompanied by an increase in its ability to trans-activate target gene expression. The results demonstrate that the differential transcriptional expression of cytochrome c, through sequential transcription factor phosphorylations, leads to enhanced mitochondrial respiratory capacity upon serum-induced entry to the cell cycle.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Cytochrome c Group/metabolism , DNA-Binding Proteins/metabolism , Mitochondria/metabolism , Trans-Activators/metabolism , 3T3 Cells , Animals , Blood , Cell Cycle , Cell Respiration , Cyclic AMP Response Element-Binding Protein/genetics , DNA-Binding Proteins/genetics , Electron Transport Complex IV/metabolism , Enzyme Activation , Immunoblotting , Luciferases/metabolism , Mice , Mice, Inbred BALB C , Models, Biological , NF-E2-Related Factor 1 , Nuclear Respiratory Factor 1 , Nuclear Respiratory Factors , Phosphorylation , Plasmids , Promoter Regions, Genetic , Spectrophotometry , Time Factors , Trans-Activators/genetics , Transcription, Genetic , Transfection , beta-Galactosidase
5.
J Biol Chem ; 275(23): 17578-82, 2000 Jun 09.
Article in English | MEDLINE | ID: mdl-10747996

ABSTRACT

A study is presented on the in vivo effect of elevated cAMP levels induced by cholera toxin on the phosphorylation of subunits of the mitochondrial respiratory complexes and their activities in Balb/c 3T3 mouse fibroblast cultures. Treatment of serum-starved fibroblasts with cholera toxin promoted serine phosphorylation in the 18-kDa subunit of complex I. Phosphorylation of the 18-kDa subunit, in response to cholera toxin treatment of fibroblasts, was accompanied by a 2-3-fold enhancement of the rotenone-sensitive endogenous respiration of fibroblasts, of the rotenone-sensitive NADH oxidase, and of the NADH:ubiquinone oxidoreductase activity of complex I. Direct exposure of fibroblasts to dibutyryl cAMP resulted in an equally potent stimulation of the NADH:ubiquinone oxidoreductase activity. Stimulation of complex I activity and respiration with NAD-linked substrates were also observed upon short incubation of isolated fibroblast mitoplasts with dibutyryl cAMP and ATP, which also promoted phosphorylation of the 18-kDa subunit. These observations document an extension of cAMP-mediated intracellular signal transduction to the regulation of cellular respiration.


Subject(s)
Cell Nucleus/metabolism , Cyclic AMP/physiology , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Bucladesine/pharmacology , Cattle , Culture Media, Serum-Free , Electron Transport Complex I , Enzyme Activation , Humans , Kinetics , Macromolecular Substances , Mice , Molecular Sequence Data , Molecular Weight , NADH, NADPH Oxidoreductases/chemistry , Phosphorylation , Sequence Alignment , Sequence Homology, Amino Acid
6.
Cell ; 98(1): 115-24, 1999 Jul 09.
Article in English | MEDLINE | ID: mdl-10412986

ABSTRACT

Mitochondrial number and function are altered in response to external stimuli in eukaryotes. While several transcription/replication factors directly regulate mitochondrial genes, the coordination of these factors into a program responsive to the environment is not understood. We show here that PGC-1, a cold-inducible coactivator of nuclear receptors, stimulates mitochondrial biogenesis and respiration in muscle cells through an induction of uncoupling protein 2 (UCP-2) and through regulation of the nuclear respiratory factors (NRFs). PGC-1 stimulates a powerful induction of NRF-1 and NRF-2 gene expression; in addition, PGC-1 binds to and coactivates the transcriptional function of NRF-1 on the promoter for mitochondrial transcription factor A (mtTFA), a direct regulator of mitochondrial DNA replication/transcription. These data elucidate a pathway that directly links external physiological stimuli to the regulation of mitochondrial biogenesis and function.


Subject(s)
Gene Expression Regulation , Membrane Transport Proteins , Mitochondria, Muscle/physiology , Mitochondrial Proteins , Oxygen Consumption , Transcription Factors/metabolism , 3T3 Cells , Animals , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , GA-Binding Protein Transcription Factor , Heat-Shock Proteins/metabolism , Ion Channels , Mice , Mitochondria, Muscle/ultrastructure , Models, Biological , NF-E2-Related Factor 1 , Nuclear Respiratory Factor 1 , Nuclear Respiratory Factors , Protein Biosynthesis , Proteins/genetics , Recombinant Fusion Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription, Genetic , Transcriptional Activation , Transfection , Uncoupling Protein 2
7.
Gene ; 233(1-2): 213-24, 1999 Jun 11.
Article in English | MEDLINE | ID: mdl-10375638

ABSTRACT

Nuclear respiratory factor 1 (NRF-1) is a nuclear transcription factor that has been implicated in the nuclear control of respiratory chain expression in mammalian cells. Here, we demonstrate that a complex pattern of alternative splicing contributes to sequence heterogeneity within the human NRF-1 5'-untranslated region (UTR). At least six different 5'-UTR exons (UTRs 1-6) were detected in NRF-1 transcripts. These exons were mapped to human NRF-1 genomic clones and their sequences, including donor and acceptor splice junctions, determined. Two of the human UTR exons were derived from insertions of Alu-sq family members into the NRF-1 locus. The distance between the transcription initiation sites in UTR1 and the first protein coding exon is approx. 47kb, bringing the total length of the human NRF-1 gene to approx. 104kb. In contrast to human, only two UTR exons were found in mouse. The mouse UTR1 sequence obtained is identical to human UTR1, but mouse UTR2 bears no resemblance to any of the human exons. Mutations within human UTR1 modulate NRF-1 expression by interfering with mRNA translational efficiency in transfected cells and in an in vitro translation system. The effects of the mutations are proportional to their ability to disrupt predicted mRNA secondary structures within UTR1. Thus, the unusually high sequence conservation within UTR1 in part reflects selective constraints on translational expression.


Subject(s)
5' Untranslated Regions , DNA-Binding Proteins/genetics , Exons , Protein Biosynthesis , RNA, Messenger/genetics , Trans-Activators/genetics , Animals , Base Sequence , Cell Line , DNA, Complementary , Gene Expression Regulation/genetics , Humans , Mice , Molecular Sequence Data , NF-E2-Related Factor 1 , Nuclear Respiratory Factor 1 , Nuclear Respiratory Factors , Repetitive Sequences, Nucleic Acid , Sequence Homology, Nucleic Acid
8.
Proc Natl Acad Sci U S A ; 94(21): 11399-404, 1997 Oct 14.
Article in English | MEDLINE | ID: mdl-9326621

ABSTRACT

Electrical stimulation of neonatal cardiac myocytes produces hypertrophy and cellular maturation with increased mitochondrial content and activity. To investigate the patterns of gene expression associated with these processes, cardiac myocytes were stimulated for varying times up to 72 hr in serum-free culture. The mRNA contents for genes associated with transcriptional activation [c-fos, c-jun, JunB, nuclear respiratory factor 1 (NRF-1)], mitochondrial proliferation [cytochrome c (Cyt c), cytochrome oxidase], and mitochondrial differentiation [carnitine palmitoyltransferase I (CPT-I) isoforms] were measured. The results establish a temporal pattern of mRNA induction beginning with c-fos (0.25-3 hr) and followed sequentially by c-jun (0.5-3 hr), JunB (0.5-6 hr), NRF-1 (1-12 hr), Cyt c (12-72 hr), and muscle-specific CPT-I (48-72 hr). Induction of the latter was accompanied by a marked decrease in the liver-specific CPT-I mRNA, thus supporting the developmental fidelity of this pattern of gene regulation. Consistent with a transcriptional mechanism, electrical stimulation increased c-fos, beta-myosin heavy chain, and Cyt c promoter activities. These increases coincided with a rise in their respective endogenous gene transcripts. NRF-1, cAMP response element, and Sp-1 site mutations within the Cyt c promoter reduced luciferase expression in both stimulated and nonstimulated myocytes. Mutations in the NRF-1 and CRE sites inhibited the induction by electrical stimulation (5-fold and 2-fold, respectively) whereas mutation of the Sp-1 site maintained or increased the fold induction. This finding is consistent with the appearance of NRF-1 and fos/jun mRNAs prior to that of Cyt c and suggests that induction of these transcription factors is a prerequisite for the transcriptional activation of Cyt c expression. These results support a regulatory role for NRF-1 and possibly AP-1 in the initiation of mitochondrial proliferation.


Subject(s)
Heart/physiology , Mitochondria, Heart/physiology , Myocardium/cytology , Transcription, Genetic , Animals , Animals, Newborn , Carnitine O-Palmitoyltransferase/biosynthesis , Cell Differentiation , Cell Division , Cells, Cultured , Cytochrome c Group/biosynthesis , Electric Stimulation , Electron Transport Complex IV/biosynthesis , Isoenzymes/biosynthesis , Kinetics , Liver/metabolism , Muscles/metabolism , Myocardium/metabolism , Myosin Heavy Chains/biosynthesis , Polymerase Chain Reaction , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-jun/biosynthesis , Rats , Transfection
9.
J Biol Chem ; 272(30): 18732-9, 1997 Jul 25.
Article in English | MEDLINE | ID: mdl-9228045

ABSTRACT

Nuclear respiratory factor 1 (NRF-1) is a transcriptional activator that acts on a diverse set of nuclear genes required for mitochondrial respiratory function in mammalian cells. These genes encode respiratory proteins as well as components of the mitochondrial transcription, replication, and heme biosynthetic machinery. Here, we establish that NRF-1 is a phosphoprotein in vivo. Phosphorylation occurs on serine residues within a concise NH2-terminal domain with the major sites of phosphate incorporation at serines 39, 44, 46, 47, and 52. The in vivo phosphorylation pattern can be approximated in vitro by phosphorylating recombinant NRF-1 with purified casein kinase II. Phosphate incorporation at the sites utilized in vivo results in a marked stimulation of DNA binding activity which is not observed in mutated proteins lacking these sites. Pairwise expression of the wild-type protein with each of a series of truncated derivatives in transfected cells results in the formation of a dimer between wild-type and mutant forms demonstrating that a homodimer is the active binding species. Although NRF-1 can dimerize in the absence of DNA, phosphorylation does not enhance the formation of these dimers. These findings suggest that phosphorylation results in an intrinsic change in the NRF-1 dimer enhancing its ability to bind DNA.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Leucine Zippers , Serine/metabolism , Trans-Activators/metabolism , Animals , Base Sequence , Binding Sites , COS Cells , Casein Kinase II , DNA-Binding Proteins/genetics , Dimerization , HeLa Cells , Humans , Molecular Sequence Data , NF-E2-Related Factor 1 , Nuclear Respiratory Factor 1 , Nuclear Respiratory Factors , Peptide Mapping , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Trans-Activators/genetics
10.
J Bioenerg Biomembr ; 29(2): 109-19, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9239537

ABSTRACT

The majority of gene products for mitochondrial respiratory function are encoded in the nuclear genome. These include most of the respiratory subunits and all of the proteins that regulate the mitochondrial genetic system. One approach to understanding nucleo-mitochondrial interactions in mammalian cells is to identify the nuclear transcription factors that are common to the expression of these gene products. This has led to the purification and molecular cloning of nuclear respiratory factors, NRF-1 and NRF-2. The DNA binding and transcriptional specificities of these proteins have implicated them in the expression of many respiratory subunits along with key components of the mitochondrial transcription, replication, and heme biosynthetic machinery. In addition, tissue-specific transcription factors have been linked to the coordinate synthesis of contractile proteins and muscle-specific respiratory subunits whereas other more ubiquitous factors may have a dual function in nuclear and mitochondrial gene activation. These findings provide a framework for further investigations of the nuclear genetic mechanisms that integrate the expression of the respiratory apparatus with that of other cellular systems during growth and development.


Subject(s)
Cell Nucleus/genetics , Electron Transport/genetics , Mitochondria/genetics , Mitochondrial Proteins , Nuclear Proteins , Animals , Cell Nucleus/physiology , DNA-Binding Proteins/physiology , Electron Transport/physiology , GA-Binding Protein Transcription Factor , Gene Expression Regulation , Humans , Mammals , Mitochondria/physiology , NF-E2-Related Factor 1 , Nuclear Respiratory Factor 1 , Nuclear Respiratory Factors , Trans-Activators/physiology , Transcription Factors/physiology , Transcriptional Activation
11.
Mol Cell Biol ; 16(10): 5708-16, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8816484

ABSTRACT

Nuclear respiratory factors 1 and 2 (NRF-1 and NRF-2) are ubiquitous transcription factors that have been implicated in the control of nuclear genes required for respiration, heme biosynthesis, and mitochondrial DNA transcription and replication. Recently, both factors have been found to be major transcriptional determinants for a subset of these genes that define a class of simple promoters involved in respiratory chain expression. Here, functional domains required for transactivation by NRF-1 have been defined. An atypical nuclear localization signal resides in a conserved amino-terminal region adjacent to the DNA binding domain and consists of functionally redundant clusters of basic residues. A second domain in the carboxy-terminal half of the molecule is necessary for transcriptional activation. The activation domains of both NRF-1 and NRF-2 were extensively characterized by both deletion and alanine substitution mutagenesis. The results show that these domains do not fall into known classes defined by a preponderance of amino acid residues, including glutamines, prolines, or isoleucines, as found in other eukaryotic activators. Rather, in both factors, a series of tandemly arranged clusters of hydrophobic amino acids were required for activation. Although all of the functional clusters contain glutamines, the glutamines differ from the hydrophobic residues in that they are inconsequential for activation. Unlike the NRF-2 domain, which contains its essential hydrophobic motifs within 40 residues, the NRF-1 domain spans about 40% of the molecule and appears to have a bipartite structure. The findings indicate that NRF-1 and NRF-2 utilize similar hydrophobic structural motifs for activating transcription.


Subject(s)
DNA-Binding Proteins/metabolism , Glutamine , Saccharomyces cerevisiae Proteins , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Alanine , Amino Acid Sequence , Animals , Base Sequence , COS Cells , DNA Primers , DNA, Mitochondrial/metabolism , Fungal Proteins , Heme/biosynthesis , Leucine Zippers , Molecular Sequence Data , Mutagenesis, Site-Directed , NF-E2-Related Factor 1 , NF-E2-Related Factor 2 , Nuclear Respiratory Factors , Polymerase Chain Reaction , Recombinant Fusion Proteins , Respiration , Sequence Deletion , Structure-Activity Relationship , Transcription, Genetic , Transfection
12.
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
13.
Trends Cardiovasc Med ; 6(2): 39-45, 1996 Feb.
Article in English | MEDLINE | ID: mdl-21232273

ABSTRACT

Mitochondrial oxidative pathways can be adversely affected by mutations in both nuclear and mitochondrial genomes. Recent evidence indicates that cardiac impairment is an important clinical feature of mitochondrial diseases resulting from such mutations. Understanding the regulatory interplay between nuclear and mitochondrial genetic systems may yield new insights into human genetic defects affecting cardiac function. Nuclear respiratory factors (NRF-1 and NRF-2) are transcriptional activators that act on a significant subset of nuclear genes required for mitochondrial respiration. These factors most likely participate in nuclear-mitochondrial interactions by helping to coordinate the synthesis of respiratory chain subunits with components of the mitochondrial transcription, replication, and heme biosynthetic machinery. Thus, NRFs and related factors are likely contributors to the nuclear control of mitochondrial energy production that is essential for normal myocardial function.

14.
J Biol Chem ; 270(30): 18019-25, 1995 Jul 28.
Article in English | MEDLINE | ID: mdl-7629110

ABSTRACT

Nuclear respiratory factor 1 (NRF-1) is a transcription factor that acts on nuclear genes encoding respiratory subunits and components of the mitochondrial transcription and replication machinery. Here we describe the isolation and characterization of the human gene encoding NRF-1. The human genomic sequences detected with NRF-1 cDNA probes at high stringency are all contained within seven overlapping recombinant lambda clones. The NRF-1 gene encompassed by these recombinants spans approximately 65 kilobases (kb) and has 11 exons and 10 introns that range in size from 0.8 to 15 kb. A rapid amplification of cDNA ends-polymerase chain reaction product containing the 5'-terminus of the NRF-1 cDNA has two exons from the 5'-untranslated region and terminates at a major transcription initiation site identified by S1 nuclease mapping. A genomic fragment containing a portion of the 5'-terminal exon and an additional 1 kb upstream had a functional promoter that was active in transfected COS cells, HeLa cells, and L6 myoblasts. The transcription initiation site utilized by the transfected promoter corresponded to that used by the endogenous gene in vivo. NRF-1 mRNA was expressed at very low levels in rat tissues compared with cytochrome c and, unlike cytochrome c, was most abundantly expressed in lung and testis. The NRF-1 gene was localized to human chromosome 7 by analysis of DNA from a panel of human-hamster cell hybrids with human-specific NRF-1 polymerase chain reaction primers. This assignment was further refined to 7q31 by cohybridization of NRF-1- and chromosome 7-specific probes to human metaphase chromosomes. These analyses should be useful in evaluating the potential role of NRF-1 in mitochondrial diseases resulting from defects in the nuclear control of mitochondrial function.


Subject(s)
Chromosomes, Human, Pair 7 , DNA-Binding Proteins/genetics , Trans-Activators/genetics , Animals , Base Sequence , Cell Line , Chromosome Mapping , Cricetinae , DNA, Complementary , DNA, Mitochondrial/genetics , HeLa Cells , Humans , Hybrid Cells , Mitochondrial Myopathies/genetics , Molecular Sequence Data , NF-E2-Related Factor 1 , Nuclear Respiratory Factor 1 , Nuclear Respiratory Factors , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic
15.
Mol Cell Biol ; 15(1): 102-11, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7799916

ABSTRACT

Nuclear respiratory factor 2 (NRF-2) was previously purified to near homogeneity from HeLa cells on the basis of its ability to bind tandem recognition sites in the rat cytochrome oxidase subunit IV (RCO4) promoter. It consisted of five subunits, alpha, beta 1, beta 2, gamma 1, and gamma 2. Sequencing of tryptic peptides from alpha and from mixtures of the two beta or two gamma subunits revealed sequence identities with subunits of the mouse GA-binding protein (GABP), a ubiquitously expressed ETS domain activator composed of three subunits, alpha, beta 1, and beta 2. To understand the precise relationship between NRF-2 and GABP, cDNAs for all five NRF-2 subunits have now been cloned and their products have been overexpressed. The results establish that the two additional NRF-2 subunits are molecular variants that differ from GABP beta 1 and beta 2 by having a 12-amino-acid insertion containing two serine doublets. PCR and RNase protection assays show that mRNAs for these variants are expressed in the human but not the rodent cells and tissues examined. The insertion did not alter the ability of the beta and gamma subunits to associate with alpha, the DNA-binding subunit, nor did it affect the ability of NRF-2 beta 1 or beta 2 to direct high-affinity binding of alpha to tandem sites in the RCO4 promoter. In addition, the four NRF-2 beta and gamma subunits were equally proficient in activating transcription in transfected cells when fused to a GAL4 DNA-binding domain. The domain responsible for this transcriptional activation was localized by deletion mapping to a region of approximately 70 amino acids that is conserved in all four NRF-2 beta and gamma subunits. The repeated glutamine-containing hydrophobic clusters within this region bear a strong resemblance to those recently implicated in protein-protein interactions within the transcriptional apparatus.


Subject(s)
DNA-Binding Proteins/chemistry , Trans-Activators/chemistry , Transcription Factors/chemistry , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA Primers/chemistry , GA-Binding Protein Transcription Factor , Gene Expression Regulation , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , RNA, Messenger/genetics , Rats , Sequence Alignment , Sequence Homology, Amino Acid , Structure-Activity Relationship , Transcription, Genetic
16.
Proc Natl Acad Sci U S A ; 91(4): 1309-13, 1994 Feb 15.
Article in English | MEDLINE | ID: mdl-8108407

ABSTRACT

Mitochondrial transcription factor A (mtTFA), the product of a nuclear gene, stimulates transcription from the two divergent mitochondrial promoters and is likely the principal activator of mitochondrial gene expression in vertebrates. Here we establish that the proximal promoter of the human mtTFA gene is highly dependent upon recognition sites for the nuclear respiratory factors, NRF-1 and NRF-2, for activity. These factors have been previously implicated in the activation of numerous nuclear genes that contribute to mitochondrial respiratory function. The affinity-purified factors from HeLa cells specifically bind to the mtTFA NRF-1 and NRF-2 sites through guanine nucleotide contacts that are characteristic for each site. Mutations in these contacts eliminate NRF-1 and NRF-2 binding and also dramatically reduce promoter activity in transfected cells. Although both factors contribute, NRF-1 binding appears to be the major determinant of promoter function. This dependence on NRF-1 activation is confirmed by in vitro transcription using highly purified recombinant proteins that display the same binding specificities as the HeLa cell factors. The activation of the mtTFA promoter by both NRF-1 and NRF-2 therefore provides a link between the expression of nuclear and mitochondrial genes and suggests a mechanism for their coordinate regulation during organelle biogenesis.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Mitochondria/metabolism , Mitochondrial Proteins , Nuclear Proteins , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Base Sequence , Cell Compartmentation , Chloramphenicol O-Acetyltransferase/biosynthesis , Chloramphenicol O-Acetyltransferase/genetics , DNA Mutational Analysis , DNA-Binding Proteins/biosynthesis , GA-Binding Protein Transcription Factor , HeLa Cells , Humans , Molecular Sequence Data , NF-E2-Related Factor 1 , Nuclear Respiratory Factor 1 , Nuclear Respiratory Factors , Promoter Regions, Genetic/genetics , Protein Binding , Recombinant Fusion Proteins/biosynthesis , Transcription Factors/biosynthesis
17.
J Biol Chem ; 269(1): 105-13, 1994 Jan 07.
Article in English | MEDLINE | ID: mdl-8276781

ABSTRACT

In vertebrate organisms, the molecular mechanisms by which extracellular signals regulate mitochondrial function and biogenesis are largely unknown. We have previously identified multiple cis-acting elements in both cytochrome c and cytochrome oxidase subunit IV (COXIV) genes that are likely targets for the regulated expression of respiratory chain components. We now demonstrate that cytochrome c but not COXIV mRNA is induced by cAMP through a mechanism involving transcriptional activation. Maximal induction occurs within 3 h and does not require de novo protein synthesis. The differential response of these genes is mediated by two distinct cAMP response elements (CREs) in the cytochrome c promoter region. Both elements function independently to drive cAMP-dependent expression from a heterologous promoter and within the proper cytochrome c promoter context. In addition, the binding properties of both elements to nuclear factors were characterized by competition DNase I footprinting, methylation interference footprinting, site-directed mutagenesis, and UV-induced DNA-protein cross-linking. The results are all consistent with the specific recognition of both CREs by CRE binding protein (CREB). A highly purified preparation of recombinant CREB formed a specific complex with each of the cytochrome c CREs identical to that formed with a crude nuclear fraction. In addition, the trans-activation of cytochrome c gene expression by recombinant CREB and protein kinase A in transfected cells was completely dependent on functional CREs within the promoter. These results establish that respiratory chain gene expression can be regulated directly by cAMP through a CREB-dependent signal transduction pathway.


Subject(s)
Cyclic AMP Response Element-Binding Protein/physiology , Cyclic AMP/physiology , Cytochrome c Group/genetics , Electron Transport Complex IV/genetics , Electron Transport , Signal Transduction , Animals , Base Sequence , Cells, Cultured , Cytochrome c Group/biosynthesis , Cytochrome c Group/metabolism , DNA/metabolism , Electron Transport Complex IV/biosynthesis , Electron Transport Complex IV/metabolism , Enzyme Induction , Gene Expression , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptide Fragments/genetics , Promoter Regions, Genetic , RNA, Messenger/metabolism , Transcriptional Activation
18.
Genes Dev ; 7(12A): 2431-45, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8253388

ABSTRACT

Nuclear respiratory factor 1 (NRF-1) was first discovered as an activator of the cytochrome c gene and was subsequently found to play a broader role in nuclear-mitochondrial interactions. We have now cloned a HeLa cDNA encoding NRF-1 using degenerate oligomers derived from tryptic peptide sequences for PCR amplification. The cDNA-encoded protein was indistinguishable from the authentic HeLa cell factor on denaturing gels, displayed the expected NRF-1 DNA-binding specificity, and made the same guanine nucleotide contacts as HeLa NRF-1 on binding known NRF-1 recognition sites. Antiserum raised against the highly purified recombinant protein recognized the identical DNA-protein complex formed using either a crude nuclear fraction or nearly homogeneous HeLa NRF-1. Recombinant NRF-1 also activated transcription through specific sites from several NRF-1-responsive promoters, confirming both the transcriptional activity and specificity of the cDNA product. Portions of NRF-1 are closely related to sea urchin P3A2 and the erect wing (EWG) protein of Drosophila. Both are recently identified developmental regulatory factors. The region of highest sequence identity with P3A2 and EWG was in the amino-terminal half of the molecule, which was found by deletion mapping to contain the DNA-binding domain, whereas the carboxy-terminal half of NRF-1 was highly divergent from both proteins. The DNA-binding domain in these molecules is unrelated to motifs found commonly in DNA-binding proteins; thus, NRF-1, P3A2, and EWG represent the founding members of a new class of highly conserved sequence-specific regulatory factors.


Subject(s)
DNA-Binding Proteins/physiology , Trans-Activators/physiology , Transcription, Genetic/physiology , Amino Acid Sequence , Base Sequence , Binding Sites/physiology , Cloning, Molecular , DNA-Binding Proteins/genetics , HeLa Cells , Humans , Molecular Sequence Data , NF-E2-Related Factor 1 , Nuclear Respiratory Factor 1 , Nuclear Respiratory Factors , Recombinant Proteins/metabolism , Trans-Activators/genetics
19.
Genes Dev ; 7(3): 380-92, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8383622

ABSTRACT

The ETS domain proteins are a diverse family of transcriptional activators that have been implicated recently in the expression of a number of cell-specific and viral promoters. Nuclear respiratory factor 2 (NRF-2) is a nuclear transcription factor that activates the proximal promoter of the rat cytochrome c oxidase subunit IV (RCO4) gene through tandem sequence elements. These elements conform to the consensus for high-affinity ETS domain recognition sites. We have now purified NRF-2 to homogeneity from HeLa cells and find that it consists of five polypeptides, only one of which has intrinsic DNA-binding ability. The others participate in the formation of heteromeric complexes with distinct binding properties. NRF-2 also specifically recognizes multiple binding sites in the mouse cytochrome c oxidase subunit Vb (MCO5b) gene. As in the functionally related RCO4 gene, tandemly arranged NRF-2 sites are essential for the activity of the proximal MCO5b promoter, further substantiating a role for NRF-2 in respiratory chain expression. Determination of peptide sequences from the various subunits of HeLa NRF-2 reveals a high degree of sequence identity with mouse GA-binding protein (GABP), a multisubunit ETS domain activator of herpes simplex virus immediate early genes. A cellular role in the activation of nuclear genes specifying mitochondrial respiratory function is thus assigned to an ETS domain activator of viral promoters.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/genetics , Electron Transport Complex IV/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Simplexvirus/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Line , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Cloning, Molecular , DNA/metabolism , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/metabolism , GA-Binding Protein Transcription Factor , Genetic Vectors , HeLa Cells , Humans , Macromolecular Substances , Mice , Molecular Sequence Data , Oligodeoxyribonucleotides , Proto-Oncogene Proteins c-ets , Rats , Restriction Mapping , Sequence Homology, Amino Acid , Transcription Factors/isolation & purification , Transcription Factors/metabolism , Transcription, Genetic , Transfection
20.
J Biol Chem ; 267(10): 6999-7006, 1992 Apr 05.
Article in English | MEDLINE | ID: mdl-1348057

ABSTRACT

Transcription factor nuclear respiratory factor 1 (NRF-1) was originally identified as an activator of the cytochrome c gene and subsequently found to stimulate transcription through specific sites in other nuclear genes whose products function in the mitochondria. These include subunits of the cytochrome oxidase and reductase complexes and a component of the mitochondrial DNA replication machinery. Here we establish that a functional recognition site for NRF-1 is present in the ATP synthase gamma-subunit gene extending the proposed respiratory role of NRF-1 to complex V. In addition, biologically active NRF-1 sites are found in genes encoding the eukaryotic translation initiation factor 2 alpha-subunit and tyrosine aminotransferase, both of which participate in the rate-limiting step of their respective pathways of protein biosynthesis and tyrosine catabolism. The recognition sites from each of these genes form identical complexes with NRF-1 as established by competition binding assays, methylation interference footprinting, and UV-induced DNA cross-linking. Cloned oligomers of each NRF-1 binding site also stimulate the activity of a truncated cytochrome c promoter in transfected cells. The NRF-1 binding activities for the various target sites copurified approximately 33,000-fold and resided in a single protein of 68 kDa. These observations further support a role for NRF-1 in the expression of nuclear respiratory genes and suggest it may help coordinate respiratory metabolism with other biosynthetic and degradative pathways.


Subject(s)
Eukaryotic Initiation Factor-2/genetics , Proton-Translocating ATPases/genetics , Transcription Factors/metabolism , Tyrosine Transaminase/genetics , Animals , Base Sequence , Binding Sites , Cattle , DNA, Mitochondrial/genetics , DNA, Mitochondrial/radiation effects , DNA-Binding Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Methylation , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Rats , Transfection , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...