Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 164
Filtrar
1.
Biochem Soc Trans ; 29(Pt 2): 187-91, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11356151

RESUMEN

Translesion replication is a mechanism that employs specialized DNA polymerases for promoting continued nascent strand extension at forks blocked by the presence of unrepaired DNA damage. In Saccharomyces cerevisiae at least, this process contributes only modestly to the ability of cells to tolerate DNA damage, but is a major source of DNA-damage-induced substitutions and frameshifts, and of spontaneous mutations. Translesion replication past many types of DNA damage in yeast depends on the activities of DNA polymerase zeta (pol zeta) and Rev1p. Pol zeta is found in most, but not all, eukaryotes investigated, whereas Rev1p appears to be universal. Genes encoding these enzymes are found in humans, and appear to perform functions similar to those in yeast.


Asunto(s)
Replicación del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , Proteínas Fúngicas/metabolismo , Mutagénesis , Nucleotidiltransferasas/metabolismo , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Daño del ADN/genética , Proteínas de Unión al ADN , ADN Polimerasa Dirigida por ADN/química , ADN Polimerasa Dirigida por ADN/genética , Humanos , Ligasas/metabolismo , Enzimas Ubiquitina-Conjugadoras
2.
Philos Trans R Soc Lond B Biol Sci ; 356(1405): 41-6, 2001 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-11205328

RESUMEN

DNA polymerase zeta (Pol zeta) and Rev1p carry out translesion replication in budding yeast, Saccharomyces cerevisiae, and are jointly responsible for almost all base pair substitution and frameshift mutations induced by DNA damage in this organism. In addition, Pol zeta is responsible for the majority of spontaneous mutations in yeast and has been proposed as the enzyme responsible for somatic hypermutability. Pol zeta, a non-processive enzyme that lacks a 3' to 5' exonuclease proofreading activity, is composed of Rev3p, the catalytic subunit, and a second subunit encoded by REV7. In keeping with its role, extension by Pol zeta is relatively tolerant of abnormal DNA structure at the primer terminus and is much more capable of extension from terminal mismatches than yeast DNA polymerase alpha (Pol alpha). Rev1p is a bifunctional enzyme that possesses a deoxycytidyl transferase activity that incorporates deoxycytidyl opposite abasic sites in the template and a second, at present poorly defined, activity that is required for the bypass of a variety of lesions as well as abasic sites. Human homologues of the yeast REV1 and REV3 have been identified and, based on the phenotype of cells producing antisense RNA to one or other of these genes, their products appear also to be employed in translation replication and spontaneous mutagenesis. We suggest that Pol zeta is best regarded as a replication enzyme, albeit one that is used only intermittently, that promotes extension at forks the progress of which is blocked for any reason, whether the presence of an unedited terminal mismatch or unrepaired DNA lesion.


Asunto(s)
Células Eucariotas/fisiología , Proteínas Fúngicas/fisiología , Mutagénesis , Nucleotidiltransferasas , Proteínas de Saccharomyces cerevisiae , Levaduras/genética , Animales , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Proteínas Fúngicas/genética , Humanos
3.
Cancer Res ; 60(15): 4105-11, 2000 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-10945617

RESUMEN

To determine whether N-methyl-N-nitrosourea (MNU) can induce malignant transformation of human fibroblasts and whether O6-methylguanine (O6-MeG) is involved, two populations of infinite life span cell strain MISU-1.1, differing only in level of O6-alkylguanine-DNA alkyltransferase, were treated with MNU and assayed for focus formation. MNU caused a dose-dependent increase in the frequency of foci in both groups, but the dose required was significantly lower in the cells lacking O6-alkylguanine-DNA alkyltransferase, indicating that O6-MeG was causally involved. Of 35 independent focus-derived strains assayed for p53 transactivating abilily, one was heterozygous, and 15 had lost all activity, 1 of 7 from untreated cells and 14 of 27 from MNU-treated cells. These results indicate that loss of p53 is not required for focus formation but may permit cells to form foci. Of 35 strains assayed for tumorigenicity, 10 formed malignant tumors with a short latency, all 10 lacked wild-type p53. The p53 heterozygous strain also formed tumors after a long latency, and the cells from those tumors lacked p53 transactivating ability. None of the 19 strains with wild-type p53 formed tumors. These results indicate that although loss of p53 is not sufficient for malignant transformation of MSU-1.1 cells, it may be necessary. Analysis of the p53 cDNA from several focus-derived strains lacking p53 activity revealed that each contained the same mutation, an A to G transition at codon 215, resulting in a change from serine to glycine. Because p53 can be inactivated by mutations at any one of a large number of sites, finding the same mutation in each strain assayed strongly suggests that the target population included a subpopulation of cells with this codon 215 mutation in one allele. Further analysis showed that all 15 focus-derived cells strains that lacked p53 transactivating activity contained two alleles, each with the same codon 215 mutation, and that the mutant allele in the heterozygous strain also had that mutatation. Analysis of the p arm of chromosome 17 of the focus-derived cell strains containing the codon 215 mutation revealed seven patterns of loss of heterozygosity, evidence of mitotic homologous recombination. Similar analysis of a separate series of cell strains, derived from foci induced by cobalt-60, revealed four patterns of loss of heterozygosity, only two of which had been found with those induced by MNU. These data suggest that homologous mitotic recombination, induced by O6-MeG in a subpopulation of cells heterozygous for p53 mutation, rendered the cells homozygous for loss of p53 activity, that this allowed the cells to form foci, and that although loss of p53 is not sufficient for malignant transformation, it predisposes cells to acquire the additional changes needed for such transformation.


Asunto(s)
Carcinógenos/toxicidad , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Genes p53/genética , Metilnitrosourea/toxicidad , Recombinación Genética/genética , Alelos , Línea Celular , Codón/genética , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Eliminación de Gen , Guanina/análogos & derivados , Guanina/farmacología , Homocigoto , Humanos , Pérdida de Heterocigocidad , O(6)-Metilguanina-ADN Metiltransferasa/antagonistas & inhibidores , O(6)-Metilguanina-ADN Metiltransferasa/deficiencia , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Recombinación Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética
4.
Carcinogenesis ; 21(9): 1639-46, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10964094

RESUMEN

O:(6)-methylguanine is responsible for homologous recombination induced by N:-methyl-N:'-nitro-N:-nitrosoguanidine (MNNG) [H. Zhang et al. (1996) CARCINOGENESIS:, 17, 2229]. To test the hypothesis that mismatch repair is causally involved in this process, we generated mismatch repair-deficient strains from a human fibroblast line containing a substrate for detecting intrachromosomal homologous recombination. The four strains selected for study exhibited greatly increased resistance to the cytotoxic effects of MNNG, which was not affected by depletion of O:(6)-alkylguanine-DNA alkyltransferase, and greatly increased sensitivity to the mutagenic effect of MNNG, suggesting that the mutagenic base modifications induced in these four cell strains by MNNG persist in their genomic DNA. Tests showed that their extracts are deficient in the repair of G:T mismatches. The frequency of homologous recombination induced by MNNG in three of these strains was significantly (5-7-fold) lower than that induced in the parental cell strain. This was not the result of a generalized defect in recombination, because when (+/-)-7beta,8alpha-dihydroxy-9alpha,10alpha-epox y-7,8,9, 10-tetrahydrobenzo[a]pyrene was used to induce recombination, all three lines responded with a normal or even a somewhat higher frequency than that observed in the parental strain. The lack of recombination displayed by the fourth strain was shown to result from the loss of part of the recombination substrate. The results strongly suggest that functional mismatch repair is required for MNNG-induced homologous recombination.


Asunto(s)
Disparidad de Par Base/genética , Reparación del ADN/genética , ADN/genética , Guanina/análogos & derivados , Guanina/metabolismo , Recombinación Genética/efectos de los fármacos , 7,8-Dihidro-7,8-dihidroxibenzo(a)pireno 9,10-óxido/toxicidad , Carcinógenos/toxicidad , Línea Celular , ADN/metabolismo , Aductos de ADN/biosíntesis , Daño del ADN/genética , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Humanos , Metilnitronitrosoguanidina/toxicidad , Mutágenos/toxicidad , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Reacción en Cadena de la Polimerasa , Recombinación Genética/genética
5.
Proc Natl Acad Sci U S A ; 97(8): 4186-91, 2000 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-10760286

RESUMEN

In Saccharomyces cerevisiae, most mutations induced by a wide range of mutagens arise during translesion replication employing the REV1 gene product and DNA polymerase zeta. As part of an effort to investigate mammalian mutagenic mechanisms, we have identified cDNA clones of the human homologs of the yeast REV genes and examined their function in UV mutagenesis. Previously, we described the isolation of a human homolog of yeast REV3, the catalytic subunit of pol zeta, and here report the identification and sequence of a human homolog of yeast REV1. This gene was isolated by identifying an expressed sequence tag encoding a peptide with similarity to the C terminus of yeast Rev1p, followed by sequencing of the clone and retrieval of the remaining cDNA by 5' rapid amplification of cDNA ends. The human gene encodes an expected protein of 1,251 residues, compared with 985 residues in the yeast protein. The proteins share two amino-terminal regions of approximately 100 residues with 41% and 20% identity, a region of approximately 320 residues with 31% identity, and a central motif in which 11 of 13 residues are identical. Human cells expressing high levels of an hREV1 antisense RNA grew normally, and were not more sensitive to the cytotoxic effect of 254 nm UV radiation than cells lacking antisense RNA. However, the frequencies of 6-thioguanine resistance mutants induced by UV in the cells expressing antisense hREV1 RNA were significantly lower than in the control (P = 0.01), suggesting that the human gene has a function similar to that of the yeast homolog.


Asunto(s)
Proteínas Fúngicas/genética , Mutagénesis/efectos de la radiación , Nucleotidiltransferasas , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , Secuencia de Aminoácidos , Codón , ADN Complementario , Proteínas Fúngicas/fisiología , Humanos , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , ARN sin Sentido/genética , Rayos Ultravioleta
7.
Cell Signal ; 11(4): 263-74, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10372804

RESUMEN

Human fibroblasts in culture will grow in serum-free medium containing serum replacement factors, but without protein growth factors, as long as the Ca2+ level is 1.0-2.0 mM. When the Ca2+ is reduced to 0.1 mM, the cells stop cycling, but they can be reinduced to cycle by raising the Ca2+ level to 1.0 mM Ca2+ or to higher concentrations that result in activation of mitogen-activated protein kinase (MAPK). We now report that exposure of human fibroblasts to extracellular Ca2+ increased the level of inositol (1,4,5)-trisphosphate in the cytoplasm and caused a transient rise in the concentration of intracellular free Ca2+. Ca2+-induced MAPK activation was partly abolished by treatment of the cells with pertussis toxin. It was also decreased by treatment of cells with thapsigargin, which depletes intracellular Ca2+ stores; with phorbol 12-myristyl 13-acetate (PMA), which down-regulates protein kinase C (PKC); with the calmodulin antagonists N-(6-aminohexyl)-5-chloro-1-naphthalenesulphonamide HCl (W-7), and calmidazolium (24571); as well as with lanthanum, a Ca2+ channel inhibitor. Ca2+ stimulation did not result in phosphorylation of the c-raf-1 protein. Our results suggest that extracellular Ca2+ stimulates MAPK activation through a pathway(s) involving a pertussis toxin-sensitive G protein, phospholipase C, intracellular free Ca2+, calmodulin, and PKC.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Calcio/metabolismo , Fibroblastos/metabolismo , Transducción de Señal , Animales , Calcio/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Calmodulina/antagonistas & inhibidores , División Celular , Línea Celular , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Espacio Extracelular , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Humanos , Líquido Intracelular/metabolismo , Isoenzimas/fisiología , Lantano/farmacología , Ratones , Nifedipino/farmacología , Toxina del Pertussis , Fosfolipasa C gamma , Fosforilación , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Piel/citología , Sulfonamidas/farmacología , Tapsigargina/farmacología , Fosfolipasas de Tipo C/fisiología , Verapamilo/farmacología , Factores de Virulencia de Bordetella/farmacología
8.
Oncogene ; 18(2): 335-42, 1999 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-9927190

RESUMEN

Identification and characterization of genes expressed in normal cells and decreased in their malignant counterparts is an important method for detecting candidate tumor suppressors. Using differential display of mRNAs from nontumorigenic infinite life span human fibroblast cell strain MSU-1.1 and an isogenic fibrosarcoma-derived cell line, 6A/SB1, which was derived from chemical carcinogen transformed MSU-1.1 cells, we identified a novel gene, ST7, showing sixfold lower expression in 6A/SB1 cells compared with parental MSU-1.1 cells. Molecular cloning of a near full-length cDNA revealed that the novel gene encodes a putative transmembrane protein composed of 859 amino acids: the 492 N-terminal amino acids including a fivefold cysteine-rich repeat of 40 amino acids homologous to the ligand binding repeat of the known low density lipoprotein receptor, a 24 hydrophobic amino acid stretch spanning the plasma membrane, and a C-terminal domain of 343 residues. ST7 is located on human chromosome 8, band q22.2-23.1, the same locus as the genes involved in acute myeloid leukemia and a locus of high polymorphism in cancer biopsies. The ST7 gene is widely expressed in normal human tissues and is particularly abundant in human heart and skeletal muscle. Northern analysis of 15 tumor cell lines derived from patients and 16 cell lines established from tumors formed in athymic mice by MSU-1.1 cells transformed in culture by various methods showed that 16 of the 31 cell lines have low or undetectable levels of ST7 mRNA. Furthermore, Western blotting analysis using a specific anti-peptide antibody demonstrated that the level of ST7 protein is high in normal fibroblasts and low in 12 sarcoma-derived cell lines tested. Altered expression of ST7 appears to occur at both the transcriptional and post-transcriptional levels. These studies are a first step in characterizing a novel putative receptor protein, whose expression is downregulated in some malignantly transformed cells, and which may play an important role in the transformation process of these cells.


Asunto(s)
Proteínas de la Membrana/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Western Blotting , Línea Celular Transformada , Mapeo Cromosómico , Cromosomas Humanos Par 8 , Clonación Molecular , ADN Complementario , Humanos , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Homología de Secuencia de Aminoácido , Células Tumorales Cultivadas
9.
Mol Cell Biol ; 19(1): 147-54, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9858539

RESUMEN

Xeroderma pigmentosum (XP) is a rare genetic disease characterized by a greatly increased susceptibility to sunlight-induced skin cancer. Cells from the majority of patients are defective in nucleotide excision repair. However, cells from one set of patients, XP variants, exhibit normal repair but are abnormally slow in replicating DNA containing UV photoproducts. The frequency of UV radiation-induced mutations in the XP variant cells is significantly higher than that in normal human cells. Furthermore, the kinds of UV-induced mutations differ very significantly from normal. Instead of transitions, mainly C-->T, 30% of the base substitutions consist of C-->A transversions, all arising from photoproducts located in one strand. Mutations involving cytosine in the other strand are almost all C-->T transitions. Forty-five percent of the substitutions involve thymine, and the majority are transversions. To test the hypothesis that the UV hypermutability and the abnormal spectrum of mutations result from abnormal bypass of photoproducts in DNA, we compared extracts from XP variant cells with those from HeLa cells and a fibroblast cell strain, MSU-1.2, for the ability to replicate a UV-irradiated form I M13 phage. The M13 template contains a simian virus 40 origin of replication located directly to the left or to the right of the target gene, lacZalpha, so that the template for the leading and lagging strands of DNA replication is defined. Reduction of replication to approximately 37% of the control value required only 1 photoproduct per template for XP variant cell extracts, but approximately 2.2 photoproducts for HeLa or MSU-1.2 cell extracts. The frequency of mutants induced was four times higher with XP variant cell extracts than with HeLa or MSU-1.2 cell extracts. With XP variant cell extracts, the proportion of C-->A transversions reached as high as 43% with either M13 template and arose from photoproducts located in the template for leading-strand synthesis; with HeLa or MSU-1.2 cell extracts, this value was only 5%, and these arose from photoproducts in either strand. With the XP variant extracts, 26% of the substitutions involved thymine, and virtually all were T-->A transversions. Sequence analysis of the coding region of the catalytic subunit of DNA polymerase delta in XP variant cell lines revealed two polymorphisms, but these do not account for the reduced bypass fidelity. Our data indicate that the UV hypermutability of XP variant cells results from reduced bypass fidelity and that unlike for normal cells, bypass of photoproducts involving cytosine in the template for the leading strand differs significantly from that of photoproducts in the lagging strand.


Asunto(s)
Reparación del ADN , Mutación , Xerodermia Pigmentosa/genética , Extractos Celulares , Línea Celular , ADN Polimerasa III/genética , Replicación del ADN , Células HeLa , Humanos , Análisis de Secuencia de ADN , Moldes Genéticos , Rayos Ultravioleta
10.
Radiat Res ; 150(5): 577-84, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9806600

RESUMEN

Cells from an infinite-life-span near-diploid human fibroblast cell strain, MSU-1.1, were transformed after a single exposure to 60Co gamma radiation. The frequency of transformation as measured by the number of induced foci per 10(6) cells was a linear function of dose. Cells from 13 independent foci from gamma-irradiated cell populations and one from a nonirradiated cell population were isolated, clonally expanded and assayed for characteristics of malignantly transformed cells. Eight of the 13 focus-derived cell strains from the irradiated populations formed tumors in athymic mice with latent periods (time required for the tumors to reach 1 cm in diameter) of 4-27 weeks. Of these 8 cell strains, 3 were fully growth factor-independent, formed large colonies (> 120 microm in diameter) in 0.33% agarose at a high frequency (50%), and produced malignant tumors with a mean latency of 6 weeks or less at all sites injected. Four others formed colonies in agarose at a slightly lower frequency, were only partially growth factor-independent, and produced malignant tumors with a longer mean latency (7-18 weeks). The tumor-derived cell lines from these latter 4 cell strains, when tested for growth in agarose, showed markedly enhanced anchorage independence. The eighth tumorigenic focus-derived cell strain was growth factor-independent but could not produce large colonies in agarose. It produced benign tumors (fibromas) with a mean latency of 27 weeks. All 8 tumorigenic focus-derived cell strains had lost the transactivating function of the TP53 (formerly known as p53) gene. However, loss of TP53 activity was not sufficient to cause tumorigenicity since 3 of the 6 nontumorigenic focus-derived cell strains had also lost all TP53 transactivation function. The other 3, which included a cell strain from the unirradiated control, had wild-type TP53 alleles and did not form tumors. These latter results support the hypothesis that loss of TP53 transactivating function plays a role in focus formation, but does not directly cause tumorigenicity. This is in agreement with studies that demonstrate that the loss of TP53 transactivation facilitates the other changes required for tumorigenicity.


Asunto(s)
Transformación Celular Neoplásica/efectos de la radiación , Animales , Transformación Celular Neoplásica/genética , Radioisótopos de Cobalto , Relación Dosis-Respuesta en la Radiación , Fibroblastos/citología , Fibroblastos/efectos de la radiación , Genes p53 , Humanos , Ratones , Ratones Endogámicos BALB C , Activación Transcripcional
11.
Proc Natl Acad Sci U S A ; 95(12): 6876-80, 1998 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-9618506

RESUMEN

To get a better understanding of mutagenic mechanisms in humans, we have cloned and sequenced the human homolog of the Saccharomyces cerevisiae REV3 gene. The yeast gene encodes the catalytic subunit of DNA polymerase zeta, a nonessential enzyme that is thought to carry out translesion replication and is responsible for virtually all DNA damage-induced mutagenesis and the majority of spontaneous mutagenesis. The human gene encodes an expected protein of 3,130 residues, about twice the size of the yeast protein (1,504 aa). The two proteins are 29% identical in an amino-terminal region of approximately 340 residues, 39% identical in a carboxyl-terminal region of approximately 850 residues, and 29% identical in a 55-residue region in the middle of the two genes. The sequence of the expected protein strongly predicts that it is the catalytic subunit of a DNA polymerase of the pol zeta type; the carboxyl-terminal domain possesses, in the right order, the six motifs characteristic of eukaryotic DNA polymerases, most closely resembles yeast pol zeta among all polymerases in the GenBank database, and is different from the human alpha, delta, and epsilon enzymes. Human cells expressing high levels of an hsREV3 antisense RNA fragment grow normally, but show little or no UV-induced mutagenesis and are slightly more sensitive to killing by UV. The human gene therefore appears to carry out a function similar to that of its yeast counterpart.


Asunto(s)
ADN Polimerasa Dirigida por ADN/genética , Proteínas Fúngicas/genética , Genes Fúngicos , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , Homología de Secuencia de Aminoácido , Secuencia de Aminoácidos , Secuencia de Bases , Humanos , Datos de Secuencia Molecular , Análisis de Secuencia
12.
Am J Cardiol ; 81(4A): 52B-59B, 1998 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-9526815

RESUMEN

Patients in the original Familial Atherosclerosis Treatment Study (FATS) cohort were subgrouped into those with triglyceride levels < or = 120 mg/dL (n = 26) and those with triglyceride levels > or = 190 mg/dL (n = 40). Their therapeutic responses to niacin plus colestipol, lovastatin plus colestipol, colestipol alone, or placebo were determined. Therapeutic response was also determined in the same 2 triglyceride subgroups (n = 12 and n = 27, respectively) of patients selected for low levels of high-density lipoprotein (HDL) cholesterol and coronary artery disease. These triglyceride criteria were chosen to identify patient subgroups with high likelihood of "pattern A" (normal-size low-density lipoprotein [LDL] particles and triglyceride < or = 120 mg/dL) or "pattern B" (small dense LDL and triglyceride > or = 190 mg/dL). Our findings in these small patient subgroups are consistent with the emerging understanding that coronary artery disease patients presenting with high triglyceride levels have lower HDL-C, smaller less buoyant LDL-C, and greater very low-density lipoprotein (VLDL) cholesterol and VLDL apolipoprotein B, and are more responsive to therapy as assessed by an increase in HDL-C and reduction in triglycerides, VLDL-C, and VLDL apolipoprotein B. In the FATS high-triglyceride subgroup with these characteristics, a tendency toward greater therapeutic improvement in coronary stenosis severity was observed among those treated with either of the 2 forms of intensive cholesterol-lowering therapy. This improvement is associated with therapeutic reduction of LDL-C and elevation of HDL-C, but also appears to be associated with drug-induced improvement in LDL buoyancy.


Asunto(s)
Anticolesterolemiantes/administración & dosificación , Colestipol/administración & dosificación , Hiperlipidemia Familiar Combinada/tratamiento farmacológico , Hipolipemiantes/administración & dosificación , Lovastatina/administración & dosificación , Niacina/administración & dosificación , Apolipoproteínas/sangre , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Ensayos Clínicos como Asunto , Estudios de Cohortes , Enfermedad Coronaria/etiología , Quimioterapia Combinada , Humanos , Hiperlipidemia Familiar Combinada/sangre , Factores de Riesgo , Triglicéridos/sangre
13.
Matrix Biol ; 16(8): 483-96, 1998 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9550265

RESUMEN

The matrix metalloproteinases (MMP) have been implicated in tumor invasion and metastasis both by immunohistochemical studies and from the observation that specific metalloproteinase inhibitors block tumor invasion and metastasis. Oligonucleotide primers for thirteen MMPs (MMP-1, MMP-2, MMP-3, MMP-7, MMP-8, MMP-9, MMP-10, MMP-11, MMP-12, MMP-13, MMP-14, MMP-15, MMP-16) were optimized for use in RT-PCR. A semi-quantitative RT-PCR assay was used to determine the pattern of MMP mRNA expression in 84 normal and transformed or carcinogen transformed human cell lines and strains derived from different tissues. The results demonstrate one or more cell lines which express thirteen members of the MMP family. In addition, various oncogene transfected human fibroblast cell strains were analyzed for MMP expression. We confirm that over-expression of the H-ras oncoprotein correlates with up-regulation of MMP-9 and demonstrate that over-expression of v-sis also up-regulates MMP-9. A cell line immortalized following myc expression was found to up-regulate MMP-7, MMP-11 and MMP-13. Inappropriate expression of several MMP mRNAs was detected in breast, prostate, bone, colon and oral tumor derived cell lines. Identification of at least one cell line expressing each of thirteen MMPs and the observation of oncogene induced expression of several MMPs should facilitate analysis of the transcriptional mechanisms controlling each MMP.


Asunto(s)
Matriz Extracelular/enzimología , Metaloendopeptidasas/biosíntesis , Línea Celular , Línea Celular Transformada , Colagenasas/biosíntesis , Colagenasas/genética , Cartilla de ADN/química , Proteínas de Fusión gag-onc/fisiología , Gelatinasas/biosíntesis , Gelatinasas/genética , Regulación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes myc/fisiología , Genes ras/fisiología , Humanos , Metaloproteinasa 1 de la Matriz , Metaloproteinasa 10 de la Matriz , Metaloproteinasa 11 de la Matriz , Metaloproteinasa 12 de la Matriz , Metaloproteinasa 13 de la Matriz , Metaloproteinasa 2 de la Matriz , Metaloproteinasa 3 de la Matriz/biosíntesis , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 7 de la Matriz , Metaloproteinasa 8 de la Matriz , Metaloproteinasa 9 de la Matriz , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Metaloendopeptidasas/genética , Familia de Multigenes , Oncogenes/fisiología , Reacción en Cadena de la Polimerasa , Células Tumorales Cultivadas
14.
Oncogene ; 15(18): 2159-68, 1997 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9393974

RESUMEN

Using differential display, we identified an mRNA that is markedly down-regulated in cell line 6A/SB1, derived from a fibrosarcoma formed in an athymic mouse following injection of carcinogen-transformed MSU-1.1 cells. The nontumorigenic parental cell strain, MSU-1.1, expresses high levels of this mRNA. Sequencing of the corresponding cDNA fragment revealed that it corresponded to an expressed sequence tag, which ultimately led to its identification as the fibulin-1D gene. Fibulin-1 is a cysteine-rich, calcium-binding extracellular matrix and plasma protein, which has four isoforms, A-D, derived from alternative splicing. Northern and Western blotting analysis of 16 cell lines established from tumors formed in athymic mice by MSU-1.1-derived cell strains independently transformed in culture showed that 44% exhibited low level or lack of expression of fibulin-1D mRNA and protein. In a similar analysis of 15 malignant cell lines derived from patients, 80% showed low level or no expression. To study the role of fibulin-1D in transformation, we transfected 6A/SB1 cells and a human fibrosarcoma-derived cell line (SHAC) with a fibulin-1D cDNA expression construct. Transfectants displaying high levels of fibulin-1D were isolated and characterized. Elevated expression of fibulin-1D led to reduced ability to form colonies in soft agar and reduced invasive potential as tested in a matrigel in vitro invasion assay. Furthermore, expression of fibulin-1D resulted in a markedly extended latency in tumor formation in athymic mice. These results indicate that low expression of fibulin-1D plays a role in tumor formation and invasion.


Asunto(s)
Proteínas de Unión al Calcio/biosíntesis , Colágeno , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Laminina , Proteoglicanos , Animales , Proteínas de Unión al Calcio/genética , Adhesión Celular/fisiología , División Celular/fisiología , Transformación Celular Neoplásica , Combinación de Medicamentos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , ARN Mensajero/metabolismo , Transfección , Células Tumorales Cultivadas
15.
Am J Cardiol ; 80(2): 111-5, 1997 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-9230143

RESUMEN

The efficacy, safety, and tolerability of a moderate dose, 3-drug lipid-lowering regimen were evaluated among 29 male patients with hyperlipidemia and coronary artery disease. In an initial 12-month phase, regular niacin, 500 mg qid, lovastatin, 20 mg bid, and colestipol, 10 g/bid, were given with dose adjustment for lipid targets and side effects. This was followed by 2 random sequence crossover phases (8 months each) alternating regular niacin with a polygel controlled-release formulation of niacin for use in this regimen. Lipid, lipoprotein, apoprotein, and clinical chemistry determinations were obtained at baseline, during the initial phase, at the 2 crossover phases, and at 6 weeks after therapy. A final questionnaire queried specific side effects and overall preferences. Low-/high-density lipoprotein (LDL/HDL) changed from means of 215/46 mg/dl at baseline, to 94/59 mg/dl after run-in, to 85/52 mg/dl after 8 months of controlled-release niacin, and to 98/56 mg/dl after 8 months of regular niacin (regular niacin vs controlled-release niacin, p <0.005/<0.05). The target of LDL < or = 100 mg/dl was achieved at 8 months by 83% of these patients with controlled-release niacin and by 52% with regular niacin (p <0.01). Compliance was 95% with controlled-release niacin versus 85% with regular niacin (p <0.001). The controlled-release niacin and regular niacin regimens did not differ in terms of uric acid, glucose, insulin, or asparate aminotransferase levels. Overall, 21% of patients called the 3 drugs "very easy" and 72% "fairly easy" to take. The controlled-release niacin-containing regimen was preferred by 21 patients and the regular niacin by 4. In conclusion, these regimens achieve striking lipid changes among hyperlipidemic patients. Controlled release is the preferred niacin preparation in terms of LDL reduction, compliance, patient preference, and achieving the National Cholesterol Education Program guideline of LDL < or = 100 mg/dl. The 2 niacin preparations did not differ in evidence of toxicity.


Asunto(s)
Colestipol/uso terapéutico , Hiperlipidemias/tratamiento farmacológico , Lovastatina/uso terapéutico , Niacina/administración & dosificación , LDL-Colesterol/sangre , LDL-Colesterol/efectos de los fármacos , Estudios Cruzados , Preparaciones de Acción Retardada , Quimioterapia Combinada , Humanos , Masculino , Persona de Mediana Edad , Niacina/efectos adversos , Cooperación del Paciente
16.
Mutat Res ; 376(1-2): 143-52, 1997 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-9202750

RESUMEN

The cytotoxic and mutagenic effect of 1-nitrosopyrene (1-NOP) and N-acetoxy-2-acetylaminofluorene (N-AcO-AAF) were compared with that of (+/-)-7 beta, 8 alpha-dihydroxy-9 alpha, 10-epoxy-7,8,9,10-tetrahydrobenzo[a]pyrene (BPDE) as a function of the initial frequency of adducts formed in the DNA of repair-proficient diploid human fibroblasts and the fraction remaining at the time the cells replicate their DNA. The principal adducts of all three agents involve guanine. The initial level of BPDE-, 1-NOP-, or N-AcO-AAF-induced adducts per 10(6) nucleotides required to lower the survival of these cells to 37% of the control was 8, 25, and 50, respectively. The frequency of mutants per 10(6) clonable cells induced at those levels of initial adduct formation was 160, 80, and 40, respectively. We determined the rate of excision repair of these adducts from the overall genome, from the individual strands of the hypoxanthine phosphoribosyltransferase (HPRT) gene, and in the case of 1-NOP and BPDE, at the level of individual nucleotides in the nontranscribed strand of exon 3 of that gene, a region where mutations induced by those agents are particularly frequent. 1-NOP-induced adducts were excised from the overall genome and from the individual strands of HPRT at a rate 2-3 times faster than BPDE-induced adducts. The average rate of repair of 1-NOP-induced adducts in exon 3 was also 2-3 times faster than the average rate of repair of BPDE-induced adducts. However, at particular nucleotides 1-NOP-induced adducts were repaired much faster, or slower, or in some cases at a rate equal to that of BPDE-induced adducts. Excision repair of N-AcO-AAF-induced adducts (i.e., deacetylated aminofluorene residues) was significantly slower than that of BPDE- or 1-NOP-induced adducts, and was not strand-specific. In an in vitro assay, BPDE adducts were four times more effective in blocking transcription than were 1-NOP or N-AcO-AAF-induced adducts. We conclude that the cytotoxic and mutagenic effect of these carcinogens reflect a complex interplay of adduct conformation, ability of adducts to block replication and transcription, and variation in the rate of excision repair, even at the nucleotide level.


Asunto(s)
Carcinógenos/química , Aductos de ADN/biosíntesis , Reparación del ADN , Mutágenos/química , 7,8-Dihidro-7,8-dihidroxibenzo(a)pireno 9,10-óxido/metabolismo , 7,8-Dihidro-7,8-dihidroxibenzo(a)pireno 9,10-óxido/farmacología , Acetoxiacetilaminofluoreno/metabolismo , Acetoxiacetilaminofluoreno/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Expresión Génica/efectos de los fármacos , Genes , Humanos , Hipoxantina Fosforribosiltransferasa/genética , Masculino , Pruebas de Mutagenicidad , Mutágenos/metabolismo , Mutágenos/farmacología , Pirenos/metabolismo , Pirenos/farmacología , Transcripción Genética/efectos de los fármacos
18.
Carcinogenesis ; 17(12): 2695-701, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9006108

RESUMEN

(+/-)-7beta,8alpha- Dihydroxy-9alpha,10alpha-epoxy-7,8,9,10-tetrahydrobenzo[a]py rene (BPDE) is the principal reactive metabolite of the carcinogenic environmental pollutant benzo[a]pyrene. Intensive studies of the distribution of BPDE-induced adduct formation in chromatin DNA compared to that in protein-free DNA have been conducted. However, until recently, investigation of BPDE-induced adduct formation at the nucleotide level in intact mammalian cells has not been feasible. We used ligation-mediated polymerase chain reaction (LMPCR) in conjunction with Escherichia coli UvrABC excinuclease to investigate the distribution of BPDE-induced adducts in the non-transcribed strand of exon 3 of the HPRT gene in normal human fibroblasts at the level of individual nucleotides to single nucleotide resolution using synchronized cell populations. We found that the relative distribution of BPDE adducts in the region of interest was essentially the same in cells treated in early G1 phase, S-phase, late G2/M phase, and in cells blocked at metaphase. Furthermore, for almost all nucleotide positions, the relative distribution of BPDE adducts in the intact cells was very similar to that found when purified DNA was treated with BPDE in vitro. The only exception was that in vivo, adduct formation at a region of six consecutive guanines, i.e. nucleotides 207-212, was strongly enhanced compared with that seen with DNA treated in vitro. No obvious nucleosomal structures or other protein-DNA interaction were detected within the region of interest by in vivo footprinting with micrococcal nuclease and other reagents revealed. In vitro studies mapping BPDE-induced adduct formation using Sequenase and UvrABC excinuclease suggested that this region of six consecutive guanines adopts a special DNA conformation. Therefore, we conclude that rather than reflecting protein-DNA interaction, the enhanced BPDE-induced adduct formation at nucleotides 207-212 in vivo reflects the impact of the physiological environment in the cell nucleus on the local DNA conformation, and that this effect remains constant throughout the cell cycle.


Asunto(s)
7,8-Dihidro-7,8-dihidroxibenzo(a)pireno 9,10-óxido/análisis , Aductos de ADN/análisis , Hipoxantina Fosforribosiltransferasa/genética , Ciclo Celular , Células Cultivadas , ADN/química , Exones , Humanos , Conformación de Ácido Nucleico
19.
Carcinogenesis ; 17(10): 2229-35, 1996 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8895493

RESUMEN

N-methyl-N'-nitro-N-nitrosoguanidine (MNNG), which alkylates many positions in DNA including the 06 position of guanine, efficiently induces intrachromosomal homologous recombination in mouse L-cells. To investigate the role of 06-methylguanine in the induction of homologous recombination in human cells, three cell strains containing duplicated copies of the Herpes simplex virus I thymidine kinase (Htk) gene and three cell strains containing duplicated copies of the gene coding for hygromycin phosphotransferase (hyg) were treated with MNNG. Neither the Htk genes nor the hyg genes code for a functional enzyme because each contains an insertion mutation at a unique site, i.e. 8-bp XhoI linker insertions in the Htk genes and 10-bp HindIII linker insertions in the hyg genes. These cell strains differ in their level of 06-alkylguanine-DNA alkyltransferase (AGT), which specifically removes the methyl group from the 06 position of guanine. Generation of a functional Htk or hyg gene has been shown to require intrachromosomal homologous recombination between the two mutant Htk genes or the two mutant hyg genes. In all six cell strains, MNNG induced a dose-dependent increase in the frequency of homologous recombination. In each set, there was an inverse correlation between the frequency of MNNG-induced recombination and the level of AGT activity. To further study the role of 06-methylguanine in the induction of homologous recombination, we used 06-benzylguanine to inactivate AGT in two additional human cell strains containing the hyg recombination substrate. After depletion of AGT activity by 06-benzylguanine, both cell strains showed a significantly elevated level of MNNG-induced homologous recombination. These results indicate that 06-methylguanine is the principal lesion responsible for the induction of homologous recombination in these human cells by this methylating agent.


Asunto(s)
Cinamatos , Aductos de ADN/genética , Guanina/análogos & derivados , Metilnitronitrosoguanidina/toxicidad , Recombinación Genética/efectos de los fármacos , Células Cultivadas , Conversión Génica , Guanina/química , Guanina/toxicidad , Humanos , Higromicina B/análogos & derivados , Timidina Quinasa/genética
20.
Mol Cell Biol ; 16(7): 3714-9, 1996 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8668188

RESUMEN

Studies showing that different types of DNA adducts are repaired in human cells at different rates suggest that DNA adduct conformation is the major determinant of the rate of nucleotide excision repair. However, recent studies of repair of cyclobutane pyrimidine dimers or benzo[a]pyrene diol epoxide (BPDE)-induced adducts at the nucleotide level in DNA of normal human fibroblasts indicate that the rate of repair of the same adduct at different nucleotide positions can vary up to 10-fold, suggesting an important role for local DNA conformation. To see if site-specific DNA repair is a common phenomenon for bulky DNA adducts, we determined the rate of repair of 1-nitrosopyrene (1-NOP)-induced adducts in exon 3 of the hypoxanthine phosphoribosyltransferase gene at the nucleotide level using ligation-mediated PCR. To distinguish between the contributions of adduct conformation and local DNA conformation to the rate of repair, we compared the results obtained with 1-NOP with those we obtained previously using BPDE. The principal DNA adduct formed by either agent involves guanine. We found that rates of repair of 1-NOP-induced adducts also varied significantly at the nucleotide level, but the pattern of site-specific repair differed from that of BPDE-induced adducts at the same guanine positions in the same region of DNA. The average rate of excision repair of 1-NOP adducts in exon 3 was two to three times faster than that of BPDE adducts, but at particular nucleotides the rate was slower or faster than that of BPDE adducts or, in some cases, equal to that of BPDE adducts. These results indicate that the contribution of the local DNA conformation to the rate of repair at a particular nucleotide position depends upon the specific DNA adduct involved. However, the data also indicate that the conformation of the DNA adduct is not the only factor contributing to the rate of repair at different nucleotide positions. Instead, the rate of repair at a particular nucleotide position depends on the interaction between the specific adduct conformation and the local DNA conformation at that nucleotide.


Asunto(s)
Aductos de ADN , Reparación del ADN , Proteínas de Escherichia coli , Hipoxantina Fosforribosiltransferasa/genética , Conformación de Ácido Nucleico , Pirenos , Secuencia de Bases , Células Cultivadas , Cartilla de ADN , Endodesoxirribonucleasas/metabolismo , Exones , Fibroblastos/citología , Humanos , Recién Nacido , Cinética , Masculino , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Piel/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA