Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Clin Pharmacol Ther ; 89(5): 726-34, 2011 May.
Article in English | MEDLINE | ID: mdl-21451509

ABSTRACT

The safety and pharmacokinetic (PK)/pharmacodynamic (PD) profile of the novel CCR1 antagonist CCX354 was evaluated in double-blind, placebo-controlled, single- and multiple-dose phase I studies (1-300 mg/day oral doses). CCX354 was well tolerated and displayed a linear dose-exposure profile, with half-life approaching 7 h at the 300-mg dose. The extent of CCR1 receptor blockade on blood monocytes, which correlated well with plasma concentrations of the drug, was assessed using fluorescently labeled CCL3 binding in whole blood from phase I subjects. High levels of receptor coverage at the 12-h time point were achieved after a single dose of 100 mg CCX354. Preclinical studies indicate that effective blockade of inflammatory cell infiltration into tissues requires ≥90% CCR1 inhibition on blood leukocytes at all times. The comparison of the properties of CCX354 with those published for other CCR1 antagonists has informed the dose selection for ongoing clinical development of CCX354 in rheumatoid arthritis (RA).


Subject(s)
Inflammation Mediators/pharmacology , Inflammation Mediators/pharmacokinetics , Quinoxalines/pharmacology , Quinoxalines/pharmacokinetics , Receptors, CCR1/antagonists & inhibitors , Adult , Animals , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Inflammation Mediators/administration & dosage , Male , Middle Aged , Monocytes/drug effects , Monocytes/metabolism , Monocytes/pathology , Protein Binding/physiology , Quinoxalines/administration & dosage , Rabbits , Rats , Rats, Wistar , Receptors, CCR1/metabolism , Young Adult
2.
J Virol ; 77(19): 10404-13, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12970425

ABSTRACT

Human cytomegalovirus (CMV) US28 (and the related open reading frame [ORF] US27) are G-protein-coupled receptor homologs believed to play a role in viral pathogenesis. In vitro, US28 has been shown to bind and internalize ligands, as well as activate intracellular signaling in response to certain chemokines, and to initiate the migration of smooth muscle cells to chemokine gradients. To assess the role of US28 in vivo, we examined the rhesus model and sequenced and characterized the rhesus CMV US28 locus. We found that rhesus CMV carries five tandem homologs of US28, all widely divergent from US28 and from each other. By reverse transcription-PCR and Northern analysis, we demonstrated expression of these ORFs in infected cells. With stable cell lines expressing these ORFs, we analyzed the homolog's binding and signaling characteristics across a wide range of chemokines and found one (RhUS28.5) to have a ligand binding profile similar to that of US28. In addition, we localized US28 and the rhesus CMV homolog RhUS28.5 to the envelope of infectious virions, suggesting a role in viral entry or cell tropism.


Subject(s)
Macaca mulatta/virology , Receptors, Chemokine/genetics , Viral Proteins/genetics , Amino Acid Sequence , Animals , Calcium/metabolism , Chromosome Mapping , Glycoproteins , Humans , Immediate-Early Proteins/genetics , Membrane Proteins , Molecular Sequence Data , Open Reading Frames , Receptors, Chemokine/chemistry , Receptors, Chemokine/physiology , Viral Proteins/chemistry , Viral Proteins/physiology
3.
J Immunol ; 164(6): 2851-6, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10706668

ABSTRACT

Searching for new receptors of dendritic cell- and T cell-active chemokines, we used a combination of techniques to interrogate orphan chemokine receptors. We report here on human CCX CKR, previously represented only by noncontiguous expressed sequence tags homologous to bovine PPR1, a putative gustatory receptor. We employed a two-tiered process of ligand assignment, where immobilized chemokines constructed on stalks (stalkokines) were used as bait for adhesion of cells expressing CCX CKR. These cells adhered to stalkokines representing ELC, a chemokine previously thought to bind only CCR7. Adhesion was abolished in the presence of soluble ELC, SLC (CCR7 ligands), and TECK (a CCR9 ligand). Complete ligand profiles were further determined by radiolabeled ligand binding and competition with >80 chemokines. ELC, SLC, and TECK comprised high affinity ligands (IC50 <15 nM); lower affinity ligands include BLC and vMIP-II (IC50 <150 nM). With its high affinity for CC chemokines and homology to CC receptors, we provisionally designate this new receptor CCR10.


Subject(s)
Chemokines, CC/metabolism , Dendritic Cells/metabolism , Receptors, Chemokine/isolation & purification , Receptors, Chemokine/metabolism , T-Lymphocytes/metabolism , Amino Acid Sequence , Animals , Cattle , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Line , Chemokine CCL19 , Chemokine CCL21 , Cloning, Molecular , Humans , Leukocytes/immunology , Leukocytes/metabolism , Mice , Molecular Sequence Data , Organ Specificity/genetics , Organ Specificity/immunology , Protein Binding , RNA, Messenger/biosynthesis , Radioligand Assay , Receptors, CCR10 , Receptors, Chemokine/biosynthesis , Receptors, Chemokine/genetics , Transfection
4.
Proc Natl Acad Sci U S A ; 96(19): 10881-6, 1999 Sep 14.
Article in English | MEDLINE | ID: mdl-10485920

ABSTRACT

Chemokine homologs are encoded by many large DNA viruses, suggesting that they contribute to control of host leukocyte transmigration and trafficking during viral infection. Murine cytomegalovirus carries a CC (beta) chemokine homolog gene giving rise to two related proteins, murine cytomegalovirus chemokine 1 and 2 (MCK-1 and MCK-2). MCK-1 peptide was found to induce calcium signaling and adherence in murine peritoneal macrophages. Cells bearing human chemokine receptor CCR3 and the human macrophage THP1 cell line were responsive to MCK-1. This pattern suggested that MCK-1 might act as an agonist, promoting leukocyte trafficking during viral infection. Consistent with this prediction, MCK-1/MCK-2 mutant viruses exhibit dramatically reduced peak levels of monocyte-associated viremia in experimentally infected mice. Thus, MCK-1/MCK-2 appears to promote host leukocyte migration to initial sites of infection and may be responsible for attracting monocytes or macrophages that efficiently disseminate virus in the host.


Subject(s)
Chemokines, CC/physiology , Chemokines/genetics , Chemokines/metabolism , Cytomegalovirus/genetics , Monocytes/metabolism , Viral Proteins , Viremia/genetics , Amino Acid Sequence , Animals , Calcium/metabolism , Chemokines, CC/genetics , Cytomegalovirus/metabolism , Humans , Mice , Mice, Inbred BALB C , Models, Biological , Models, Genetic , Molecular Sequence Data , Monocytes/virology , Peritoneum/metabolism , Peritoneum/virology , Recombination, Genetic , Sequence Homology, Amino Acid , Time Factors , Viremia/metabolism
5.
Proc Natl Acad Sci U S A ; 96(17): 9839-44, 1999 Aug 17.
Article in English | MEDLINE | ID: mdl-10449781

ABSTRACT

Cytomegalovirus is a widespread opportunistic pathogen affecting immunocompromised individuals in whom neutrophils may mediate virus dissemination and contribute to progression of disease. Recent sequence analysis suggests that genes absent or altered in attenuated strains may influence pathogenesis. We have found two genes, UL146 and UL147, whose products have sequence similarity to alpha (CXC) chemokines. UL146 encodes a protein, designated vCXC-1, that is a 117-aa glycoprotein secreted into the culture medium as a late gene product, where its presence correlates with the ability to attract human neutrophils. Recombinant vCXC-1 is a fully functional chemokine, inducing calcium mobilization, chemotaxis, and degranulation of neutrophils. High-affinity vCXC-1 binding is shown to be mediated via CXCR2, but not CXCR1. vCXC-1 exhibits a potency approaching that of human IL-8. As the first example of a virus-encoded alpha chemokine, vCXC-1 may ensure the active recruitment of neutrophils during cytomegalovirus infection, thereby providing for efficient dissemination during acute infection and accounting for the prominence of this leukocyte subset in cytomegalovirus disease.


Subject(s)
Chemokines, CXC/metabolism , Cytomegalovirus Infections/metabolism , Cytomegalovirus/metabolism , Receptors, Chemokine/metabolism , Receptors, Interleukin/metabolism , Viral Proteins , Amino Acid Sequence , Chemokines, CXC/chemistry , Chemokines, CXC/genetics , Cloning, Molecular , Cytomegalovirus/genetics , DNA, Viral/chemistry , Fibroblasts/metabolism , Fibroblasts/virology , Humans , Lung/cytology , Molecular Sequence Data , Receptors, Interleukin-8B
6.
J Biol Chem ; 274(31): 21569-74, 1999 Jul 30.
Article in English | MEDLINE | ID: mdl-10419462

ABSTRACT

Uncertainty regarding viral chemokine function is mirrored by an incomplete knowledge of host chemokine receptor usage by the virally encoded proteins. One such molecule is vMIP-I, a C-C type chemokine of undefined function and binding specificity, encoded by the Kaposi's sarcoma herpesvirus HHV-8. We report here that vMIP-I binds to and induces cytosolic [Ca(2+)] signals in human T cells selectively through CCR8, a CC chemokine receptor associated with Th2 lymphocytes. Furthermore, using a panel of 65 different human, viral, and rodent chemokines, we have established a comprehensive ligand binding "fingerprint" for CCR8. The receptor exhibits marked "high" affinity (K(d) < 15 nM) only for four chemokines, three of them of viral origin: vMIP-I, vMIP-II, vMCC-I, and human I-309. A previously unreported second class of lower affinity ligands includes MCP-3 and possibly two other viral chemokines. vMIP-I and I-309 appear to act as CCR8 agonists: binding to and inducing cytosolic [Ca(2+)] elevation through the receptor. By contrast, vMIP-II and vMCC-I act as potent antagonists: binding without inducing signaling, and blocking the effects of I-309 and vMIP-I. These results suggest a ligand hierarchy for CCR8, identifying vMIP-I as a selective viral chemokine agonist. CCR8 may thus engage a specific subset of chemokines with the potential to regulate each other during viral infection and immune regulation.


Subject(s)
Chemokines, CC/physiology , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/immunology , Macrophage Inflammatory Proteins/physiology , Receptors, Chemokine/physiology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Viral Proteins , Animals , Calcium/metabolism , Cells, Cultured , Chemokines, CC/genetics , Chemokines, CC/pharmacology , Flow Cytometry , Humans , Interleukin-2/pharmacology , Macrophage Inflammatory Proteins/genetics , Mice , Receptors, CCR8 , Receptors, Chemokine/genetics , Recombinant Proteins/metabolism , Signal Transduction , Structure-Activity Relationship , T-Lymphocytes/drug effects
7.
Blood ; 93(10): 3233-40, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10233874

ABSTRACT

Eotaxin has been characterized as a chemokine involved in eosinophil activation; however, mRNA for this C-C chemokine has been shown to be constitutively expressed in thymus. Immunohistochemical analysis showed a punctate distribution pattern, with eotaxin expression localized mainly in the medulla and in Hassle's corpuscles. Moreover, the receptor for eotaxin, CCR-3, was detected on thymocytes, with the highest level of expression being on the CD8 single-positive population. Equilibrium binding analyses on unfractionated thymocytes demonstrated specific 125I-eotaxin binding profiles comparable with CCR-3 transfectants. Eotaxin induced cell migration and mobilization of intracellular calcium in all thymocytes except the immature CD4(-)/CD8(-) population. Eotaxin also induced the secretion of the chemokines interleukin-8, RANTES, and macrophage inflammatory protein-1beta from thymocyte cultures in vitro. These results suggest that eotaxin-induced thymocyte activation may have important physiological implications for lymphocyte mobilization within and from this lymphoid organ.


Subject(s)
Chemokines, CC , Cytokines/immunology , Receptors, Chemokine/genetics , T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Calcium/metabolism , Cells, Cultured , Chemokine CCL11 , Chemokine CCL4 , Chemokine CCL5/biosynthesis , Chemokine CCL5/genetics , Chemotactic Factors, Eosinophil/immunology , Chemotactic Factors, Eosinophil/pharmacology , Chemotaxis, Leukocyte , Cytokines/genetics , Cytokines/pharmacology , Humans , Infant , Infant, Newborn , Interleukin-8/biosynthesis , Interleukin-8/genetics , Macrophage Inflammatory Proteins/biosynthesis , Macrophage Inflammatory Proteins/genetics , Receptors, CCR3 , Receptors, Chemokine/biosynthesis , Receptors, HIV/genetics , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/immunology , T-Lymphocytes/drug effects , Thymus Gland/immunology
8.
FEBS Lett ; 439(3): 203-7, 1998 Nov 20.
Article in English | MEDLINE | ID: mdl-9845323

ABSTRACT

Fractalkine is the only as yet known member of a novel class of chemokines. Besides its novel Cys-X-X-X-Cys motif, fractalkine exhibits features which have not been described for any other member of the chemokine family, including its unusual size (397 amino acids human, 395 mouse) and the possession of a transmembrane anchor, from which a soluble form may be released by extracellular cleavage. This report demonstrates the abundant mRNA and fractalkine protein expression in neuronal cells. The neuronal expression of fractalkine mRNA is unaffected by experimentally induced inflammation of central nervous tissue.


Subject(s)
Chemokines, CX3C , Chemokines, CXC/biosynthesis , Encephalomyelitis, Autoimmune, Experimental/metabolism , Membrane Proteins/biosynthesis , Neurons/metabolism , Animals , Cells, Cultured , Chemokine CX3CL1 , Chemokines, CXC/analysis , Chemokines, CXC/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Immunohistochemistry , Membrane Proteins/analysis , Membrane Proteins/genetics , Mice , Neurons/pathology , RNA, Messenger/metabolism , Rats
9.
Blood ; 91(8): 2905-13, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9531601

ABSTRACT

The CC chemokine macrophage inflammatory protein 1beta (MIP-1beta), has been shown to be a chemoattractant preferentially activating CD4(+) CD45RA+ T lymphocytes. Further analysis of chemokine action on lymphocytic cells has shown the potent migration-promoting capacity of MIP-1beta on human thymocytes. The responding cells were the CD4(+) and CD8(+) single-positive (SP), as well as the CD4(+) CD8(+) double-positive (DP) populations, with little if any migratory activity on the double-negative (DN) population. The activation of thymocytes by MIP-1beta appeared to be a direct, receptor-mediated event as evidenced by the rapid mobilization of intracellular calcium, increase in proteins phosphorylated on tyrosine, and activation of the mitogen-activated protein kinase (MAPK) pathway. Radioligand binding analyses showed specific and displaceable binding of MIP-1beta to thymocytes with a Kd of approximately 1 nmol/L, a profile that was comparable with MIP-1beta binding to CCR-5-transfected NIH 3T3 cells. In addition, CCR-5 mRNA was detected in total thymocyte populations indicating that activation of thymocytes by MIP-1beta may occur through binding to CCR-5. Further dissection of the subpopulations showed that only the DP and CD8(+) SP populations expressed CCR-5 and expression data on these two populations was confirmed using anti-CCR-5 monoclonal antibody. These data may be suggestive of a role for MIP-1beta in human thymocyte activation, and show a potential route for HIV infectivity in the developing immune system.


Subject(s)
Chemotaxis, Leukocyte/drug effects , Lymphocyte Activation/drug effects , Macrophage Inflammatory Proteins/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/physiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Chemokine CCL4 , Child, Preschool , Humans , Infant , Infant, Newborn , Radioligand Assay , Receptors, CCR5/metabolism , Signal Transduction/drug effects
10.
J Immunol ; 160(1): 426-33, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9552000

ABSTRACT

The chemokine RANTES induces a unique biphasic cytoplasmic Ca2+ signal in T cells. The first phase of this signal, similar to that of other chemokines, is G-protein mediated and chemotaxis associated. The second phase of this signal, unique to RANTES and evident at concentrations greater than 100 nM, is tyrosine kinase linked and results in a spectrum of responses similar to those seen with antigenic stimulation of T cells. We show here that certain jurkat T cells responded to RANTES solely through this latter pathway. A direct correlation between the RANTES-induced second phase response and CD3 expression was demonstrated in these cells. Sorting the Jurkat cells into CD3(high) and CD3(low) populations revealed that only the CD3(high) cells were responsive to RANTES. Furthermore, stimulation of these Jurkat cells with anti-CD3 mAb significantly depresses their subsequent response to RANTES. While a RANTES-specific chemokine receptor is expressed at a low level on these Jurkat cells, the RANTES-induced activation is dependent on the presence of the TCR. Thus, stimulation through TCR may partially account for RANTES' unique pattern of signaling in T cells.


Subject(s)
CD3 Complex/physiology , Chemokine CCL5/physiology , Jurkat Cells/physiology , T-Lymphocytes/physiology , Benzoquinones , Calcium/physiology , Down-Regulation , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/physiology , Humans , Lactams, Macrocyclic , Lymphocyte Activation , Protein-Tyrosine Kinases/physiology , Quinones/pharmacology , Rifabutin/analogs & derivatives , Signal Transduction/drug effects , Virulence Factors, Bordetella/pharmacology
11.
J Immunol ; 160(4): 1894-900, 1998 Feb 15.
Article in English | MEDLINE | ID: mdl-9469451

ABSTRACT

The chemokine RANTES is a potent agonist of T cell activation. In an investigation of signal-transduction events activated by this chemokine, we have shown that RANTES stimulates dose-dependent phospholipase D (PLD) activity in Jurkat cells. Equilibrium-binding analyses using 125I-labeled RANTES indicated the presence of a receptor for RANTES on these cells, which has a Kd of 0.1 nM, is expressed at approximately 600 sites per cell, and a binding specificity that was not comparable with that of any of the known chemokine receptors, since 125I-labeled RANTES was displaced by macrophage-inflammatory protein-1 beta (but not macrophage-inflammatory protein-1 alpha), monocyte-chemotactic protein-1 (MCP-1), MCP-3, MCP-4, and eotaxin. RANTES-induced PLD activation was augmented by GTP gamma S, but not GDP beta S, and inhibited by the protein kinase C inhibitor bisindolylmaleimide, as well as the fungal metabolite brefeldin A, and C3 exoenzyme (Clostridium botulinum), implicating the activation of RhoA. RANTES also induced GTP-GDP exchange of immunoprecipitated RhoA. RANTES-stimulated PLD activity was dependent on an ADP-ribosylation factor(s), as assessed by inhibition studies using a synthetic inhibitory peptide of the N-terminal 16 amino acids of ADP-ribosylation factor 1. These studies indicate the potential existence of a novel receptor-mediated mechanism for activation of T cells by the chemokine RANTES.


Subject(s)
Chemokine CCL5/pharmacology , GTP-Binding Proteins/physiology , Phospholipase D/metabolism , T-Lymphocytes/enzymology , ADP-Ribosylation Factors , Amino Acid Sequence , Brefeldin A , Chemokine CCL5/antagonists & inhibitors , Cyclopentanes/pharmacology , Drug Synergism , Enzyme Activation/drug effects , Epoxy Compounds/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/physiology , Humans , Jurkat Cells , Molecular Sequence Data , Phospholipase D/antagonists & inhibitors , Protein Binding , Protein Kinase C/physiology , Spiro Compounds/pharmacology , T-Lymphocytes/drug effects , rhoA GTP-Binding Protein
12.
J Biol Chem ; 272(45): 28206-9, 1997 Nov 07.
Article in English | MEDLINE | ID: mdl-9353270

ABSTRACT

The CC chemokine receptor 3 (CCR3) plays an important role in the regulation of the migration of eosinophils, a leukocyte population involved in many inflammatory pathologies including asthma. CCR3 binds to the CC chemokine eotaxin, a promigratory cytokine originally isolated as the key component in a model of eosinophil-induced airway inflammation. We show here that eotaxin/CCR3 binding interactions exhibit a marked sensitivity to relatively small changes in the extracellular environment. In particular, modest variations in the pH and the level of sodium chloride over a range of physiologic and near physiologic conditions had dramatic effects on eotaxin binding and CCR3-mediated cytoplasmic Ca2+ mobilization. These biochemical indices were reflected at the functional level as well; small changes in pH and salt also resulted in striking changes in the migration of primary human eosinophils in vitro. These results reveal that relatively small perturbations in extracellular buffer conditions can yield widely disparate interpretations of CCR3 ligand binding and affinities and suggest that modulation of the tissue microenvironment might be utilized to control the affinity and efficacy of chemokine-mediated cell migration.


Subject(s)
Chemokines, CC , Receptors, Chemokine/metabolism , Calcium/metabolism , Cell Movement , Chemokine CCL11 , Chemotactic Factors, Eosinophil/metabolism , Cytokines/metabolism , Eosinophils/cytology , Eosinophils/metabolism , Humans , Hydrogen-Ion Concentration , Ligands , Osmolar Concentration , Protein Binding , Receptors, CCR3 , Sodium Chloride/metabolism
13.
J Exp Med ; 186(6): 837-44, 1997 Sep 15.
Article in English | MEDLINE | ID: mdl-9294138

ABSTRACT

Dendritic cells initiate immune responses by ferrying antigen from the tissues to the lymphoid organs for presentation to lymphocytes. Little is known about the molecular mechanisms underlying this migratory behavior. We have identified a chemokine receptor which appears to be selectively expressed in human dendritic cells derived from CD34+ cord blood precursors, but not in dendritic cells derived from peripheral blood monocytes. When stably expressed as a recombinant protein in a variety of host cell backgrounds, the receptor shows a strong interaction with only one chemokine among 25 tested: the recently reported CC chemokine macrophage inflammatory protein 3alpha. Thus, we have designated this receptor as the CC chemokine receptor 6. The cloning and characterization of a dendritic cell CC chemokine receptor suggests a role for chemokines in the control of the migration of dendritic cells and the regulation of dendritic cell function in immunity and infection.


Subject(s)
Dendritic Cells/immunology , Macrophage Inflammatory Proteins/metabolism , Receptors, Chemokine , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , Chromosome Mapping , Cloning, Molecular , Cricetinae , DNA, Complementary/genetics , Dendritic Cells/metabolism , Gene Expression , Humans , Hybrid Cells , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, CCR6 , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Transfection
14.
J Mol Biol ; 249(1): 11-28, 1995 May 26.
Article in English | MEDLINE | ID: mdl-7776365

ABSTRACT

Human mitochondrial transcription factor A (h-mtTFA) is essential for initiation of transcription from the two promoters located in the displacement-loop region of human mitochondrial DNA. This 25 kDa protein contains two tandem, HMG box DNA-binding domains separated by a 27 amino acid residue linker region and followed by a 25 residue carboxyl-terminal tail; both the linker and tail are rich in basic amino acid residues. Mutational analysis of h-mtTFA revealed that the tail region is important for specific DNA recognition and essential for transcriptional activation. The critical role of the human tail in transcription was confirmed by constructing chimeric proteins that exchanged similar regions between h-mtTFA and its Saccharomyces cerevisiae homolog, sc-mtTFA. Wild-type sc-mtTFA is unable to activate transcription from the human mitochondrial light-strand promoter (LSP). Addition of the human tail region to sc-mtTFA conferred LSP-specific promoter activation. In all of the different h-mtTFA mutations tested, transcriptional activation was correlated with specific DNA-binding activity, suggesting that these two functions may be inseparable, a situation entirely consistent with previous mutational analyses of human mitochondrial promoters.


Subject(s)
DNA-Binding Proteins/genetics , High Mobility Group Proteins/genetics , Mitochondrial Proteins , Nuclear Proteins , Trans-Activators/genetics , Transcription Factors/genetics , Base Sequence , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Promoter Regions, Genetic/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism
15.
Biochim Biophys Acta ; 1271(1): 127-34, 1995 May 24.
Article in English | MEDLINE | ID: mdl-7599198

ABSTRACT

The two major promoters for transcription of the human mitochondrial genome are located near each other in the displacement-loop region of the molecule. Previous work has localized these promoters to regions of < 100 nucleotides each; the DNA sequence at the transcription start site is stringently required, as is the region from -10 to -40 base pairs upstream of each respective start site. Each upstream site is recognized and bound by human mitochondrial transcription factor A (h-mtTFA), an event previously shown to be important for transcriptional activation. We report here results using recombinant h-mtTFA that demonstrate the dependence of transcription initiation of h-mtTFA. In addition, altering the distance between the h-mtTFA binding site and the transcription start site greatly impairs transcription initiation efficiency. The decrease in transcription initiation efficiency was shown to be a consequence of altering the position of h-mtTFA binding as opposed to the strength of h-mtTFA binding, as judged by DNA footprinting ability. Analysis of a chimeric yeast-human promoter revealed that the yeast mtTFA homologue cannot substitute for the human protein, even when bound at an appropriate position upstream of the human transcription start site.


Subject(s)
DNA Replication , DNA, Mitochondrial/biosynthesis , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/metabolism , Mitochondrial Proteins , Nuclear Proteins , Promoter Regions, Genetic , Transcription Factors/metabolism , Transcription, Genetic , Base Composition , Base Sequence , Chimera , Cloning, Molecular , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/isolation & purification , DNA-Directed RNA Polymerases/metabolism , Deoxyribonuclease I , Escherichia coli , Gene Expression , Humans , KB Cells , Molecular Sequence Data , Mutagenesis, Insertional , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Templates, Genetic , Transcription Factors/biosynthesis , Transcription Factors/isolation & purification
16.
J Mol Evol ; 37(4): 338-46, 1993 Oct.
Article in English | MEDLINE | ID: mdl-7508515

ABSTRACT

RNase MRP is a site-specific ribonucleoprotein endoribonuclease that cleaves mitochondrial RNA from the origin of leading-strand DNA synthesis contained within the displacement-loop region. Bovine mitochondrial DNA maintains the typical gene content and order of mammalian mitochondrial DNAs but differs in the nature of sequence conservation within this displacement-loop regulatory region. This markedly different sequence arrangement raises the issue of the degree to which a bovine RNase MRP would reflect the physical and functional properties ascribed to the enzymes previously characterized from mouse and human. We find that bovine RNase MRP exists as a ribonucleoprotein, with an RNA component of 279 nucleotides that is homologous to that of mouse or human RNase MRP RNA. Characterization of the nuclear gene for bovine RNase MRP RNA showed conservation of sequence extending 5' of the RNase MRP RNA coding sequence, including the presence of a cis-acting element known to be important for the expression of some mitochondrial protein-coding nuclear genes. Bovine or mouse RNase MRP cleaves a standard mouse mitochondrial RNA substrate in the same manner; each also cleaves a bovine mitochondrial RNA substrate identically. Since bovine and mouse RNase MRPs process both bovine and mouse substrates, we conclude that the structural features of the mitochondrial RNA substrate required for enzymatic cleavage have been well conserved despite significant overall primary sequence divergence. Inspection of the bovine RNA substrate reveals conservation of only the most critical portion of the primary sequence as indicated by earlier studies with mouse and human RNase MRPs.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Endoribonucleases/metabolism , RNA/metabolism , Animals , Base Sequence , Blotting, Northern , Cattle , Cell Line , DNA , Humans , Mice , Molecular Sequence Data , Polymerase Chain Reaction , RNA/chemistry , RNA, Mitochondrial , Sequence Homology, Nucleic Acid
17.
FASEB J ; 7(1): 208-13, 1993 Jan.
Article in English | MEDLINE | ID: mdl-7678563

ABSTRACT

RNase MRP is a ribonucleoprotein endoribonuclease that has been shown to cleave mitochondrial primer RNA sequences from a variety of sources. The bulk of RNase MRP activity is found in the nucleus where its function remains unknown. Two different approaches have resulted in predictions of distinct secondary structures for RNase MRP RNA. In order to analyze more definitively the higher-order structure of RNase MRP RNA, we have conducted a phylogenetic comparison of the available RNase MRP RNA sequences from human, mouse, rat, cow, toad, and yeast. The resulting secondary structure shares features in common with previously described structures for prokaryotic and eukaryotic RNase P RNAs (1) and RNase MRP RNAs (2, 3). In addition, the phylogenetic structure is consistent with available chemical modification data on RNase MRP RNA and with the detailed analysis of the To antigen binding domain located near the 5' end of the RNase MRP RNA. The structure is not limited to RNase MRP RNAs, but can be expanded to cover both eukaryotic RNase P RNAs and RNase P/MRP RNAs from plants.


Subject(s)
Endoribonucleases/chemistry , Mitochondria/chemistry , Nucleic Acid Conformation , Animals , Base Sequence , Binding Sites , Endoribonucleases/genetics , Humans , Molecular Sequence Data , Phylogeny , RNA, Bacterial/chemistry , RNA, Catalytic/chemistry , Ribonuclease P , Sequence Alignment , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...