Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Transplant ; 16(6): 1751-65, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26714197

RESUMO

Bacterial infections after lung transplantation cause airway epithelial injury and are associated with an increased risk of developing bronchiolitis obliterans syndrome. The damaged epithelium is a source of alarmins that activate the innate immune system, yet their ability to activate fibroblasts in the development of bronchiolitis obliterans syndrome has not been evaluated. Two epithelial alarmins were measured longitudinally in bronchoalveolar lavages from lung transplant recipients who developed bronchiolitis obliterans syndrome and were compared to stable controls. In addition, conditioned media from human airway epithelial cells infected with Pseudomonas aeruginosa was applied to lung fibroblasts and inflammatory responses were determined. Interleukin-1 alpha (IL-1α) was increased in bronchoalveolar lavage of lung transplant recipients growing P. aeruginosa (11.5 [5.4-21.8] vs. 2.8 [0.9-9.4] pg/mL, p < 0.01) and was significantly elevated within 3 months of developing bronchiolitis obliterans syndrome (8.3 [1.4-25.1] vs. 3.6 [0.6-17.1] pg/mL, p < 0.01), whereas high mobility group protein B1 remained unchanged. IL-1α positively correlated with elevated bronchoalveolar lavage IL-8 levels (r(2)  = 0.6095, p < 0.0001) and neutrophil percentage (r(2)  = 0.25, p = 0.01). Conditioned media from P. aeruginosa infected epithelial cells induced a potent pro-inflammatory phenotype in fibroblasts via an IL-1α/IL-1R-dependent signaling pathway. In conclusion, we propose that IL-1α may be a novel therapeutic target to limit Pseudomonas associated allograft injury after lung transplantation.


Assuntos
Lesão Pulmonar Aguda/etiologia , Bronquiolite Obliterante/etiologia , Células Epiteliais/microbiologia , Fibroblastos/patologia , Rejeição de Enxerto/etiologia , Transplante de Pulmão/efeitos adversos , Pseudomonas aeruginosa/patogenicidade , Mucosa Respiratória/microbiologia , Lesão Pulmonar Aguda/patologia , Adulto , Aloenxertos , Bronquiolite Obliterante/patologia , Líquido da Lavagem Broncoalveolar , Células Epiteliais/imunologia , Células Epiteliais/patologia , Feminino , Rejeição de Enxerto/patologia , Humanos , Inflamação/etiologia , Inflamação/patologia , Interleucina-1alfa/imunologia , Masculino , Pessoa de Meia-Idade , Neutrófilos/imunologia , Infecções por Pseudomonas/complicações , Infecções por Pseudomonas/microbiologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/patologia , Estudos Retrospectivos , Adulto Jovem
2.
Development ; 128(14): 2649-60, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11526072

RESUMO

Sex-lethal is the Drosophila melanogaster sex determination master switch. It is also required in female germ cells to control mitosis and meiotic recombination. As early germ cells mature, distinct changes in both Sex-lethal protein levels and localization occur. By manipulating the levels of Hedgehog and making germline clones of components in the hedgehog signaling pathway, we demonstrate that Hedgehog affects the nuclear translocation of Sex-lethal and the levels of the protein in early germ cells. This effect is mediated primarily through degradation. Consistent with the Hedgehog pathway regulating Sex-lethal, we find Sex-lethal in a complex with Fused and Costal-2, both downstream components of the pathway. This is the first demonstration that downstream components of the Hedgehog signaling pathway regulate a target other than Cubitus interruptus.


Assuntos
Proteínas de Drosophila , Proteínas de Insetos/metabolismo , Proteínas de Ligação a RNA/metabolismo , Receptores Acoplados a Proteínas G , Processos de Determinação Sexual , Transdução de Sinais , Animais , Sobrevivência Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Citoplasma/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Feminino , Expressão Gênica , Células Germinativas , Proteínas Hedgehog , Proteínas de Insetos/genética , Cinesinas/genética , Proteínas de Membrana/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Ligação a RNA/genética , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Proteínas Repressoras/metabolismo , Receptor Smoothened , Tubulina (Proteína)/genética
3.
Genetics ; 155(4): 1741-56, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10924471

RESUMO

It has been suggested that sexual identity in the germline depends upon the combination of a nonautonomous somatic signaling pathway and an autonomous X chromosome counting system. In the studies reported here, we have examined the role of the sexual differentiation genes transformer (tra) and doublesex (dsx) in regulating the activity of the somatic signaling pathway. We asked whether ectopic somatic expression of the female products of the tra and dsx genes could feminize the germline of XY animals. We find that Tra(F) is sufficient to feminize XY germ cells, shutting off the expression of male-specific markers and activating the expression of female-specific markers. Feminization of the germline depends upon the constitutively expressed transformer-2 (tra-2) gene, but does not seem to require a functional dsx gene. However, feminization of XY germ cells by Tra(F) can be blocked by the male form of the Dsx protein (Dsx(M)). Expression of the female form of dsx, Dsx(F), in XY animals also induced germline expression of female markers. Taken together with a previous analysis of the effects of mutations in tra, tra-2, and dsx on the feminization of XX germ cells in XX animals, our findings indicate that the somatic signaling pathway is redundant at the level tra and dsx. Finally, our studies call into question the idea that a cell-autonomous X chromosome counting system plays a central role in germline sex determination.


Assuntos
Proteínas de Drosophila , Drosophila/genética , Drosophila/fisiologia , Células Germinativas/fisiologia , Processos de Determinação Sexual , Cromossomo X , Alelos , Animais , Animais Geneticamente Modificados , Western Blotting , DNA Complementar/metabolismo , Proteínas de Ligação a DNA/genética , Feminino , Imunofluorescência , Proteínas de Insetos/genética , Masculino , Modelos Genéticos , Mutagênese , Proteínas Nucleares/genética , Splicing de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleoproteínas/genética , beta-Galactosidase/metabolismo
4.
Med Sci Sports Exerc ; 31(3): 393-9, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10188743

RESUMO

PURPOSE: The purpose of this study was to examine the effect of high and low glycemic index (GI) carbohydrate (CHO) pre-exercise meals on endurance running capacity. METHODS: Eight active subjects (five male and three female) ran on a treadmill at approximately 70% VO2max to exhaustion on two occasions separated by 7 d. Three hours before the run after an overnight fast, each subject was given in a single-blind, random order, isoenergetic meal of 850+/-21 kcal (mean+/-SEM; 67% carbohydrate, 30% protein, and 3% fat) containing either high (HGI) or low (LGI) GI carbohydrate foods providing 2.0 g CHO.kg(-1) body weight. RESULTS: Ingestion of the HGI meal resulted in a 580% and 330% greater incremental area under the 3-h blood glucose and serum insulin response curves, respectively. Performance times were not different between the HGI and LGI trials (113+/-4 min and 111+/-5 min, respectively). During the first 80 min of exercise in the LGI trial, CHO oxidation was 12% lower and fat oxidation was 118% higher than in the HGI trial. Although serum insulin concentrations did not differ between trials, blood glucose at 20 min into exercise in the HGI trial was lower than that during the LGI trial at the same time (3.6+/-0.3 mmol.L(-1) vs 4.3+/-0.3 mmol.L(-1); P < 0.05). During exercise, plasma glycerol and serum free fatty acid concentrations were lower in the HGI trial than in the LGI trial. CONCLUSIONS: This results demonstrate that although there is a relative shift in substrate utilization from CHO to fat when a low GI meal is ingested before exercise compared with that for a high GI meal, there is no difference in endurance running capacity.


Assuntos
Carboidratos da Dieta/administração & dosagem , Resistência Física/fisiologia , Corrida/fisiologia , Adulto , Feminino , Humanos , Masculino
5.
Development ; 122(3): 971-82, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8631274

RESUMO

In D. melanogaster the binary switch gene Sex-lethal (Sxl) plays a pivotal role in somatic sex determination -- when the Sxl gene is on the female pathway is followed, while the male pathway is followed when the gene is off. In the present study we have asked whether the Sxl gene is present in other species of the genus Drosophila and whether it is subject to a similar sex-specific on-off regulation. Sxl proteins were found in all of the drosophilids examined, and they display a sex-specific pattern of expression. Furthermore, characterization of the Sxl gene in the distant drosophilan relative, D. virilis, reveals that the structure and sequence organization of the gene has been well conserved and that, like melanogaster, alternative RNA processing is responsible for its sex-specific expression. Hence, this posttranscriptional on-off regulatory mechanism probably existed before the separation of the drosophilan and sophophoran subgenera and it seems likely that Sxl functions as a sex determination switch gene in most species in the Drosophila genus. Although alternative splicing appears to be responsible for the on-off regulation of the Sxl gene in D. virilis, this species is unusual in that Sxl proteins are present not only in females but also in males. The D. virilis female and male proteins appear to be identical over most of the length except for the amino-terminal approx. 25 aa which are encoded by the differentially spliced exons. In transcriptionally active polytene chromosomes, the male and female proteins bind to the same cytogenetic loci, including the sites corresponding to the D. virilis Sxl and tra genes. Hence, though the male proteins are able to interact with appropriate target pre-mRNAs, they are apparently incapable of altering the splicing pattern of these pre-mRNAs.


Assuntos
Proteínas de Drosophila , Drosophila/embriologia , Proteínas de Ligação a RNA/fisiologia , Análise para Determinação do Sexo , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sequência de Bases , Mapeamento Cromossômico , Sequência Consenso , Primers do DNA/química , Drosophila/genética , Éxons , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes de Insetos , Masculino , Dados de Sequência Molecular , Ligação Proteica , Splicing de RNA , RNA Mensageiro/genética
6.
RNA ; 2(1): 1-10, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8846292

RESUMO

mRNAs from the early Sex-lethal promoter, Sxl-Pe, encode embryonic Sxl proteins that function to activate the Sxl autoregulatory loop. They do so by directing the female-specific splicing of the first transcripts expressed from the late or maintenance promoter, Sxl-Pm. The early promoter is located, however, upstream not downstream of the translation terminating male-specific exon, L3, and upstream of the second Sxl-Pm exon, L2. If the Sxl proteins expressed from Sxl-Pe are to provide the mechanism for bypassing the normal requirement for Sxl protein in the female-specific splicing of transcripts from Sxl-Pm, then what is the mechanism for skipping L2 and L3 in the processing of transcripts from Sxl-Pe? In the studies reported here, we have generated a report construct to examine the splicing of Sxl-Pe transcripts. Our results indicate that neither specific maternal products, Sxl protein, nor an X chromosome to autosome ratio of 1 are required for the processing of the embryonic mRNAs. We also found that none of the three genes, snf, virilizer, and fl(2)d, which when mutated alter the female-specific processing of Sxl-Pm transcripts, alter the generation of the early splice. Skipping to intervening exons to generate an open reading frame that will encode the Sxl early proteins appears to be an intrinsic property of initiating the early Sxl RNAs within the first intron of the Sxl-Pm maintenance transcription unit.


Assuntos
Proteínas de Drosophila , Drosophila/genética , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Animais , Sequência de Bases , Drosophila/embriologia , Drosophila/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Dados de Sequência Molecular , Splicing de RNA , Análise para Determinação do Sexo , Transcrição Gênica
7.
Genetics ; 141(4): 1521-35, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8601491

RESUMO

Unlike sex determination in the soma, which is an autonomous process, sex determination in the germline of Drosophila has both inductive and autonomous components. In this paper, we examined how sexual identity is selected and maintained in the Drosophila germline. We show that female-specific expression of genes in the germline is dependent on a somatic signaling pathway. This signaling pathway requires the sex-non-specific transformer 2 gene but, surprisingly, does not appear to require the sex-specific genes, transformer and doublesex. Moreover, in contrast to the soma where pathway initiation and maintenance are independent processes, the somatic signaling pathway appears to function continuously from embryogenesis to the larval stages to select and sustain female germline identity. We also show that the primary target for the somatic signaling pathway in germ cells can not be the Sex-lethal gene.


Assuntos
Drosophila/embriologia , Células Germinativas , Análise para Determinação do Sexo , Animais , Sequência de Bases , Primers do DNA , Feminino , Masculino , Dados de Sequência Molecular
8.
Genes Dev ; 8(5): 598-613, 1994 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-7523244

RESUMO

The orb gene of Drosophila encodes sex-specific germ-line proteins that contain two RRM-type RNA-binding domains. Here we report the distribution of Orb protein in wild-type, tumorous, and orb mutant ovaries. The wild-type distribution of Orb protein during oogenesis resembles that of its RNA, preferentially accumulating in the cytoplasm of the developing oocyte shortly after the formation of the 16-cell cyst. As anticipated from its germ-line expression, mutations in orb lead to female sterility. Analysis of the effect of orb mutants on the distribution of RNAs known to be required for oocyte differentiation and polarity suggests that orb functions in RNA localization at multiple points during oogenesis. In addition, phenotypic characterization of the orb mutants indicates that the gene is required early in oogenesis for formation of the 16-cell cyst. It then functions in the differentiation of the oocyte and is required for the three-dimensional reorganization of the germ cells in the cyst as well as for the establishment of normal germ-line-soma interactions in the egg chamber.


Assuntos
Proteínas de Drosophila , Drosophila/genética , Genes de Insetos , Oogênese , Proteínas de Ligação a RNA/fisiologia , Animais , Western Blotting , Feminino , Masculino , Mutação , Ovário/metabolismo , Óvulo/metabolismo , RNA , Testículo/metabolismo
9.
Mol Cell Biol ; 13(12): 7734-46, 1993 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8246990

RESUMO

The on/off state of the binary switch gene Sex-lethal (Sxl), which controls somatic sexual development in Drosophila melanogaster, is regulated at the level of alternative splicing. In males, in which the gene is off, the default splicing machinery produces nonfunctional mRNAs; in females, in which the gene is on, the autoregulatory activity of the Sxl proteins directs the splicing machinery to produce functional mRNAs. We have used germ line transformation to analyze the mechanism of default and regulated splicing. Our results demonstrate that a blockage mechanism is employed in Sxl autoregulation. However, in contrast to transformer, in which Sxl appears to function by preventing the interaction of splicing factors with the default 3' splice site, a different strategy is used in autoregulation. (i) Multiple cis-acting elements, both upstream and downstream of the male exon, are required. (ii) These cis-acting elements are distant from the splice sites they regulate, suggesting that the Sxl protein cannot function in autoregulation by directly competing with splicing factors for interaction with the regulated splice sites. (iii) The 5' splice site of the male exon appears to be dominant in regulation while the 3' splice site plays a subordinate role.


Assuntos
Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/genética , Éxons , Splicing de RNA/genética , Diferenciação Sexual/genética , Animais , Sequência de Bases , Primers do DNA/genética , Feminino , Genes Letais , Genes de Troca , Homeostase/genética , Íntrons , Masculino , Modelos Genéticos , Dados de Sequência Molecular , Poli U/genética , RNA Mensageiro/genética
10.
Development ; 118(3): 797-812, 1993 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8076518

RESUMO

In addition to controlling somatic sexual development in Drosophila melanogaster, the Sex-lethal (Sxl) gene is required for proper differentiation of female germ cells. To investigate its role in germ-line development, we have examined the expression of Sxl in wild-type ovaries and ovaries that are defective in early steps of germ cell differentiation. As in the soma, the basic mechanism for on/off regulation of Sxl relies on sex-specific processing of its transcripts in germ cells. One class of female-sterile mutations, which includes fs(1)1621 and the tumorous-ovary-producing allele of the ovarian tumor gene, otu1, is defective in the splicing process. These mutants have germ lines with high amounts of Sxl RNA spliced in the male mode and a severe reduction of protein levels in the germ cells. Another class of female-sterile mutations produces a phenotype similar to that seen in fs(1)1621 and otu1 but appears to express normal levels of Sxl protein in the germ cells. However, this second class does not show the changes in protein distribution normally observed in wild-type germ cells. In the wild-type germarium, the non-differentiated germ cells show a strong cytoplasmic accumulation of Sxl protein followed, as the germ cells differentiate, by a dramatic reduction and redistribution of the protein into nuclear foci. Interestingly, two female-sterile alleles of Sxl, Sxlf4 and Sxlf5 belong to the second class, which shows persistent cytoplasmic accumulation of Sxl protein. These Sxl female-sterile mutants encode an altered protein indicating that Sxl regulates processes that eventually lead to the changes in Sxl protein distribution. Lastly, we demonstrate that during the final stages of oogenesis several mechanisms must operate to prevent the progeny from inheriting Sxl protein. Conceivably, this regulation safeguards the inadvertent activation of the Sxl autoregulatory feedback loop in the male zygote.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster/genética , Regulação da Expressão Gênica , Genes de Insetos , Genes Letais , Hormônios de Inseto/genética , Oogênese/genética , Proteínas de Ligação a RNA , Animais , Sequência de Bases , Diferenciação Celular , Drosophila melanogaster/fisiologia , Feminino , Infertilidade Feminina/genética , Hormônios de Inseto/biossíntese , Hormônios de Inseto/fisiologia , Masculino , Dados de Sequência Molecular , Mutação , Neoplasias Ovarianas/genética , Ovário/citologia , Reação em Cadeia da Polimerase , Splicing de RNA , Análise para Determinação do Sexo
11.
Mol Cell Biol ; 13(3): 1408-14, 1993 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8441386

RESUMO

In Drosophila melanogaster, sex determination in somatic cells is controlled by a cascade of genes whose expression is regulated by alternative splicing [B. S. Baker, Nature (London) 340:521-524, 1989; J. Hodgkin, Cell 56:905-906, 1989]. The master switch gene in this hierarchy is Sex-lethal. Sex-lethal is turned on only in females, and an autoregulatory feedback loop which controls alternative splicing maintains this state (L. R. Bell, J. I. Horabin, P. Schedl, and T. W. Cline, Cell 65:229-239, 1991; L. N. Keyes, T. W. Cline, and P. Schedl, Cell 68:933-943, 1992). Sex-lethal also promotes female differentiation by controlling the splicing of RNA from the next gene in the hierarchy, transformer. Sosnowski et al. (B. A. Sosnowski, J. M. Belote, and M. McKeown, Cell 58:449-459, 1989) have shown that the mechanism for generating female transformer transcripts is not through the activation of the alternative splice site but by the blockage of the default splice site. We have tested whether an activation or a blockage mechanism is involved in Sex-lethal autoregulation. The male exon of Sex-lethal with flanking splice sites was placed into the introns of heterologous genes. Our results support the blockage mechanism. The poly(U) run at the male exon 3' splice site is required for sex-specific splicing. However, unlike transformer, default splicing to the male exon is sensitive to the sequence context within which the exon resides. This and the observation that the splice signals at the exon are suboptimal are discussed with regard to alternate splicing.


Assuntos
Processamento Alternativo , Drosophila melanogaster/genética , Genes de Insetos/genética , Genes Letais/genética , Diferenciação Sexual/genética , Animais , Sequência de Bases , Éxons/genética , Feminino , Masculino , Dados de Sequência Molecular , Poli U/genética , Precursores de RNA/metabolismo , Análise de Sequência
12.
Cell ; 65(2): 229-39, 1991 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-2015624

RESUMO

Sex-lethal is a binary switch gene that controls all aspects of Drosophila sexual dimorphism. It must be active in females and inactive in males. The on/off regulation reflects alternative RNA splicing in which full-length proteins are produced only in females. Here we investigate the role of Sxl in maintaining sexual pathway commitments. By ectopic expression of a female Sxl cDNA in transgenic male flies, we show that Sxl protein induces a rapid switch from male- to female-specific splicing. The ectopically expressed Sxl protein wil trans-activate an endogenous wild-type Sxl gene. This establishes a feedback loop in which Sxl proteins induce their own synthesis by directing the female-specific splicing of Sxl transcripts. We conclude that the female determined state is maintained by Sxl through positive autoregulation, while the male determined state is maintained by default.


Assuntos
Proteínas de Drosophila , Drosophila/genética , Regulação da Expressão Gênica , Genes de Troca , Splicing de RNA , Proteínas de Ligação a RNA , Análise para Determinação do Sexo , Transformação Genética , Alelos , Animais , Feminino , Homeostase , Hormônios de Inseto/análise , Hormônios de Inseto/genética , Hormônios de Inseto/fisiologia , Masculino , Mutação , Fenótipo , Reação em Cadeia da Polimerase/métodos , Mapeamento por Restrição , Transcrição Gênica , Ativação Transcricional
13.
J Biol Chem ; 263(23): 11575-83, 1988 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-2457024

RESUMO

A 55-amino acid segment, normally present between residues 241 and 295 of the 348-residue gene I protein of the filamentous bacteriophage f1, acts as an internal signal sequence for gene I protein or, when present in fusion proteins, for EcoRI endonuclease or alkaline phosphatase. The resulting proteins are inserted so that they span the membrane with sequences on the amino side of the 55-residue segment in the cytoplasm and those near the carboxy side outside the cytoplasmic membrane. The presence of these proteins in the membrane results in the rapid inhibition of cell growth, probably from a loss of the membrane potential. We describe some of the elements in this 55-residue segment that appear to be crucial for its interaction with the membrane.


Assuntos
Potenciais da Membrana , Proteínas Virais/análise , Sequência de Aminoácidos , Sequência de Bases , Enzimas de Restrição do DNA/metabolismo , DNA Viral/análise , Desoxirribonuclease EcoRI , Desoxirribonuclease HpaII , Dados de Sequência Molecular , Peso Molecular , Plasmídeos , Proteínas Virais de Fusão/análise , Proteínas Virais de Fusão/genética , Proteínas Virais/genética
14.
J Mol Biol ; 188(3): 403-13, 1986 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-3525845

RESUMO

We have cloned the gene I sequence of the filamentous bacteriophage f1 downstream from the lambda leftward promoter on a plasmid that also contains the temperature-sensitive lambda repressor, cI857. Temperature induction of gene I protein (pI) resulted in rapid cessation of growth. This inhibition appears to involve a rapid decrease in synthesis of host protein and RNA. The ability of pI to cause this inhibition is not dependent on thioredoxin, a host factor that is necessary for phage morphogenesis and has been shown by genetic data to interact with pI. The inhibition does not appear to be mediated by the amino half of the protein, as induction of an identical plasmid construction of an amber mutant positioned two-thirds along gene I, does not affect cell growth. Analysis of the transcription products from the cloned gene I confirmed previous suggestions that a transcription terminator exists in the amino-terminal portion of the gene. In addition, there is no detectable promoter activity in the 152 bases immediately upstream from the gene. These data and the inability to overproduce pI argue for down-regulation of pI production. Radioactive labeling of proteins in maxi-cells and normal Escherichia coli cells identifies pI as a protein of about 39,000 Mr that partitions with the cell envelope. Pulse-chase experiments suggest that pI is not processed to any appreciable extent.


Assuntos
Bacteriófagos/genética , Genes Virais , Proteínas Virais/genética , Bacteriófagos/crescimento & desenvolvimento , Clonagem Molecular , Eletroforese em Gel de Poliacrilamida , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Morfogênese , Biossíntese de Proteínas , Regiões Terminadoras Genéticas , Transcrição Gênica , Proteínas Virais/biossíntese
15.
J Bacteriol ; 155(3): 1417-25, 1983 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-6224774

RESUMO

An Escherichia coli DNA fragment containing the structural gene serU132 for the nonsense suppressor tRNASer2am was identified and purified by being cloned into a plasmid vector. Information obtained from DNA sequence analysis was used to select a serU132 fragment for insertion downstream from the bacteriophage lambda pL promoter in two pBR322-lambda derivatives. In nonsense mutant strains bearing the resulting serU132 hybrid plasmids, the presence of the lambda cI857 repressor gene carried on the same plasmid or in a prophage genome permits thermal regulation of suppressor synthesis.


Assuntos
Bacteriófago lambda/genética , Escherichia coli/genética , Óperon , Supressão Genética , Sequência de Bases , Clonagem Molecular , Mutação , Plasmídeos , Aminoacil-RNA de Transferência/genética , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...