Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 33(7): 8634-8647, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31090455

RESUMEN

Reduced expression of 2'-3'-cyclic nucleotide 3'-phosphodiesterase (Cnp) in humans and mice causes white matter inflammation and catatonic signs. These consequences are experimentally alleviated by microglia ablation via colony-stimulating factor 1 receptor (CSF1R) inhibition using PLX5622. Here we address for the first time preclinical topics crucial for translation, most importantly 1) the comparison of 2 long-term PLX5622 applications (prevention and treatment) vs. 1 treatment alone, 2) the correlation of catatonic signs and executive dysfunction, 3) the phenotype of leftover microglia evading depletion, and 4) the role of intercellular interactions for efficient CSF1R inhibition. Based on our Cnp-/- mouse model and in vitro time-lapse imaging, we report the unexpected discovery that microglia surviving under PLX5622 display a highly inflammatory phenotype including aggressive premortal phagocytosis of oligodendrocyte precursor cells. Interestingly, ablating microglia in vitro requires mixed glial cultures, whereas cultured pure microglia withstand PLX5622 application. Importantly, 2 extended rounds of CSF1R inhibition are not superior to 1 treatment regarding any readout investigated (magnetic resonance imaging and magnetic resonance spectroscopy, behavior, immunohistochemistry). Catatonia-related executive dysfunction and brain atrophy of Cnp-/- mice fail to improve under PLX5622. To conclude, even though microglia depletion is temporarily beneficial and worth pursuing, complementary treatment strategies are needed for full and lasting recovery.-Fernandez Garcia-Agudo, L., Janova, H., Sendler, L. E., Arinrad, S., Steixner, A. A., Hassouna, I., Balmuth, E., Ronnenberg, A., Schopf, N., van der Flier, F. J., Begemann, M., Martens, H., Weber, M. S., Boretius, S., Nave, K.-A., Ehrenreich, H. Genetically induced brain inflammation by Cnp deletion transiently benefits from microglia depletion.


Asunto(s)
2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa/genética , Encéfalo/patología , Encefalitis/genética , Microglía/patología , Eliminación de Secuencia/genética , Adulto , Animales , Encéfalo/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Compuestos Orgánicos/farmacología , Fenotipo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Eliminación de Secuencia/efectos de los fármacos
2.
Mutagenesis ; 34(5-6): 421-429, 2019 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-31676900

RESUMEN

The tetrahydrofuran-type abasic site analog (THF) induces large deletion mutations in human cells. To compare the large deletions induced by THF on leading and lagging strand templates, plasmid DNAs bearing the analog at a specific position outside the supF gene were introduced into human U2OS cells. The replicated DNAs recovered from the transfected cells were electroporated into an Escherichia coli indicator strain. THF on the lagging strand template produced more supF mutants than THF on the leading strand template. This unequal mutagenicity was due to the higher frequencies of not only large deletions but also untargeted base substitutions induced in the gene. These results suggested that both types of mutations occur more frequently when abasic sites are formed on the lagging strand template.


Asunto(s)
Eliminación de Secuencia/genética , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Replicación del ADN/efectos de los fármacos , Replicación del ADN/genética , ADN Bacteriano/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Humanos , Mutagénesis/efectos de los fármacos , Mutagénesis/genética , Mutágenos/farmacología , Plásmidos/genética , Eliminación de Secuencia/efectos de los fármacos , Transfección/métodos
3.
Appl Microbiol Biotechnol ; 103(3): 1465-1474, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30607491

RESUMEN

Markerless genetic engineering of bacterial genomes is commonly performed by two-step homologous recombination methods using vectors carrying flanking regions of the target gene for site-specific vector integration and counterselection markers to provide positive selection pressure on the second recombination step resulting in vector excision. Here, we provide the proof-of-principle of a novel counterselection method that exploits antagonistic activities between bactericidal and bacteriostatic antibiotics and which can provide selection pressure on the second recombination step by selective killing of bacteria retaining the antibiotic selection marker. This method was optimized for the bacterial pathogens Listeria monocytogenes and Neisseria meningitidis by screening for antagonistic activities between the bactericidal aminoglycosides kanamycin, streptomycin, and gentamicin in combination with the bacteriostatic antibiotics chloramphenicol and erythromycin. The largest difference in selective killing of both L. monocytogenes and N. meningitidis containing an antibiotic selection marker versus wild-type bacteria was observed for the combination of erythromycin, gentamicin, and ermC as antibiotic selection marker. Therefore, this combination was used to generate two markerless deletion mutants for both L. monocytogenes and N. meningitidis. After applying the dual-antimicrobial selection pressure on cultures during the second recombination step, surviving colonies were replica plated on agar with and without erythromycin. On average, 12-13% of the randomly selected bacterial colonies had lost the selection marker due to a second recombination event and approximately half of these colonies were the desired markerless in-frame deletion mutants. Therefore, this method proved to be easy and fast and should be applicable to a wide variety of bacterial species.


Asunto(s)
Antibacterianos/farmacología , Secuencia de Bases/genética , Ingeniería Genética/métodos , Listeria monocytogenes/efectos de los fármacos , Listeria monocytogenes/genética , Neisseria meningitidis/efectos de los fármacos , Neisseria meningitidis/genética , Eliminación de Secuencia/efectos de los fármacos , Cloranfenicol/farmacología , Combinación de Medicamentos , Eritromicina/farmacología , Genoma Bacteriano/genética , Gentamicinas/farmacología , Kanamicina/farmacología , Metiltransferasas/genética , Eliminación de Secuencia/genética , Estreptomicina/farmacología
4.
PLoS Genet ; 12(10): e1006368, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27755535

RESUMEN

For more than half a century, genotoxic agents have been used to induce mutations in the genome of model organisms to establish genotype-phenotype relationships. While inaccurate replication across damaged bases can explain the formation of single nucleotide variants, it remained unknown how DNA damage induces more severe genomic alterations. Here, we demonstrate for two of the most widely used mutagens, i.e. ethyl methanesulfonate (EMS) and photo-activated trimethylpsoralen (UV/TMP), that deletion mutagenesis is the result of polymerase Theta (POLQ)-mediated end joining (TMEJ) of double strand breaks (DSBs). This discovery allowed us to survey many thousands of available C. elegans deletion alleles to address the biology of this alternative end-joining repair mechanism. Analysis of ~7,000 deletion breakpoints and their cognate junctions reveals a distinct order of events. We found that nascent strands blocked at sites of DNA damage can engage in one or more cycles of primer extension using a more downstream located break end as a template. Resolution is accomplished when 3' overhangs have matching ends. Our study provides a step-wise and versatile model for the in vivo mechanism of POLQ action, which explains the molecular nature of mutagen-induced deletion alleles.


Asunto(s)
Caenorhabditis elegans/genética , Reparación del ADN por Unión de Extremidades/genética , Reparación del ADN/efectos de los fármacos , ADN Polimerasa Dirigida por ADN/genética , Animales , Caenorhabditis elegans/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de los fármacos , Reparación del ADN/genética , Replicación del ADN/efectos de los fármacos , ADN Polimerasa Dirigida por ADN/biosíntesis , Metanosulfonato de Etilo/toxicidad , Estudios de Asociación Genética , Genoma/efectos de los fármacos , Mutagénesis , Mutágenos/toxicidad , Eliminación de Secuencia/efectos de los fármacos , ADN Polimerasa theta
5.
Cell Mol Life Sci ; 74(1): 57-66, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27722768

RESUMEN

Cystic fibrosis transmembrane conductance regulator (CFTR) channel gating is predominantly regulated by protein kinase A (PKA)-dependent phosphorylation. In addition to regulating CFTR channel activity, PKA phosphorylation is also involved in enhancing CFTR trafficking and mediating conformational changes at the interdomain interfaces of the protein. The major cystic fibrosis (CF)-causing mutation is the deletion of phenylalanine at position 508 (F508del); it causes many defects that affect CFTR trafficking, stability, and gating at the cell surface. Due to the multiple roles of PKA phosphorylation, there is growing interest in targeting PKA-dependent signaling for rescuing the trafficking and functional defects of F508del-CFTR. This review will discuss the effects of PKA phosphorylation on wild-type CFTR, the consequences of CF mutations on PKA phosphorylation, and the development of therapies that target PKA-mediated signaling.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/genética , Eliminación de Secuencia , Animales , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/análisis , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Descubrimiento de Drogas , Humanos , Fosforilación/efectos de los fármacos , Mutación Puntual/efectos de los fármacos , Conformación Proteica/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Eliminación de Secuencia/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
6.
Am J Physiol Renal Physiol ; 306(5): F561-7, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24402098

RESUMEN

Epithelial Na(+) channel (ENaC) subunits (α, ß, and γ) found in functional complexes are translated from mature mRNAs that are similarly processed by the inclusion of 13 canonical exons. We examined whether individual exons 3-12, encoding the large extracellular domain, are required for functional channel expression. Human ENaCs with an in-frame deletion of a single α-subunit exon were expressed in Xenopus oocytes, and their functional properties were examined by two-electrode voltage clamp. With the exception of exon 11, deletion of an individual exon eliminated channel activity. Channels lacking α-subunit exon 11 were hyperactive. Oocytes expressing this mutant exhibited fourfold greater amiloride-sensitive whole cell currents than cells expressing wild-type channels. A parallel fivefold increase in channel open probability was observed with channels lacking α-subunit exon 11. These mutant channels also exhibited a lost of Na(+) self-inhibition, whereas we found similar levels of surface expression of mutant and wild-type channels. In contrast, in-frame deletions of exon 11 from either the ß- or γ-subunit led to a significant loss of channel activity, in association with a marked decrease in surface expression. Our results suggest that exon 11 within the three human ENaC genes encodes structurally homologous yet functionally diverse domains and that exon 11 in the α-subunit encodes a module that regulates channel gating.


Asunto(s)
Canales Epiteliales de Sodio/genética , Eliminación de Secuencia , Xenopus laevis/metabolismo , Amilorida/farmacología , Animales , Células Cultivadas , Canales Epiteliales de Sodio/metabolismo , Exones , Humanos , Oocitos/metabolismo , Técnicas de Placa-Clamp/métodos , Mutación Puntual/genética , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Eliminación de Secuencia/efectos de los fármacos , Xenopus laevis/genética
7.
Mutagenesis ; 29(1): 27-36, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24243707

RESUMEN

Ochratoxin A (OTA) is a carcinogen targeting proximal tubules at the renal outer medulla (ROM) in rodents. We previously reported that OTA increased mutant frequencies of the red/gam gene (Spi(-)), primarily deletion mutations. In the present study, Spi(-) assays and mutation spectrum analyses in the Spi(-) mutants were performed using additional samples collected in our previous study. Spi(-) assay results were similar to those in our previous study, revealing large (>1kb) deletion mutations in the red/gam gene. To clarify the molecular progression from DNA damage to gene mutations, in vivo comet assays and analysis of DNA damage/repair-related mRNA and/or protein expression was performed using the ROM of gpt delta rats treated with OTA at 70, 210 or 630 µg/kg/day by gavage for 4 weeks. Western blotting and immunohistochemical staining demonstrated that OTA increased γ-H2AX expression specifically at the carcinogenic target site. In view of the results of comet assays, we suspected that OTA was capable of inducing double-strand breaks (DSBs) at the target sites. mRNA and/or protein expression levels of homologous recombination (HR) repair-related genes (Rad51, Rad18 and Brip1), but not nonhomologous end joining-related genes, were increased in response to OTA in a dose-dependent manner. Moreover, dramatic increases in the expression of genes involved in G2/M arrest (Chek1 and Wee1) and S/G2 phase (Ccna2 and Cdk1) were observed, suggesting that DSBs induced by OTA were repaired predominantly by HR repair, possibly due to OTA-specific cell cycle regulation, consequently producing large deletion mutations at the carcinogenic target site.


Asunto(s)
Carcinógenos/toxicidad , Roturas del ADN de Doble Cadena/efectos de los fármacos , Ocratoxinas/toxicidad , Eliminación de Secuencia/efectos de los fármacos , Animales , Secuencia de Bases , Peso Corporal/efectos de los fármacos , Carcinógenos/administración & dosificación , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Ensayo Cometa , Proteínas de Escherichia coli/genética , Riñón/efectos de los fármacos , Riñón/patología , Masculino , Pruebas de Mutagenicidad/métodos , Ocratoxinas/administración & dosificación , Tamaño de los Órganos , Pentosiltransferasa/genética , Ratas , Ratas Transgénicas
8.
Zhongguo Zhong Yao Za Zhi ; 37(11): 1663-6, 2012 Jun.
Artículo en Zh | MEDLINE | ID: mdl-22994004

RESUMEN

OBJECTIVE: To study the effect of pine pollen on Kidney Mitochondria DNA Deletion Mutation (mtDNA) in senile mice. METHOD: Kunming senile mice were randomly divided into the pine pollen group, and the senile control group. And a young control group was randomly selected. Mouse in the pine pollen group were orally administered with pine pollen (750 mg x kg(-1)) daily. The young control group and the senile control group were orally administered with isometric 0.9% sodium chloride injection. After 60 days, deletion mutation of mtDNA were detected by PCR technology and photodensity scan. Relative level of MDA and activity of SOD in kidney tissues were detected. RESULT: The senile control group showed significant increase in relative level and deletion mutation of mtDNA (P < 0.05). Compared with the senile control group, the pine pollen group showed decreased depletion of kidney mtDNA (P < 0.05). Pine pollen can decrease MDA volume and increase the activity of SOD significantly (P < 0.05). CONCLUSION: Pine pollen can inhibit deletion mutation of mtDNA in senile mice, suggesting that pine pollen can reduce oxidative damage of mtDNA and protect mtDNA. Accordingly, it provides a possible mechanism of anti-aging effect of pine pollen at the molecular level.


Asunto(s)
Envejecimiento/genética , ADN Mitocondrial/genética , Medicamentos Herbarios Chinos/farmacología , Riñón/metabolismo , Pinus/química , Polen/química , Eliminación de Secuencia/efectos de los fármacos , Envejecimiento/efectos de los fármacos , Animales , Femenino , Riñón/efectos de los fármacos , Masculino , Ratones
9.
J Biol Chem ; 285(1): 123-30, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19892708

RESUMEN

The purpose of this work was to obtain information about conformational changes of the plasma membrane Ca(2+)-pump (PMCA) in the membrane region upon interaction with Ca(2+), calmodulin (CaM) and acidic phospholipids. To this end, we have quantified labeling of PMCA with the photoactivatable phosphatidylcholine analog [(125)I]TID-PC/16, measuring the shift of conformation E(2) to the auto-inhibited conformation E(1)I and to the activated E(1)A state, titrating the effect of Ca(2+) under different conditions. Using a similar approach, we also determined the CaM-PMCA dissociation constant. The results indicate that the PMCA possesses a high affinity site for Ca(2+) regardless of the presence or absence of activators. Modulation of pump activity is exerted through the C-terminal domain, which induces an apparent auto-inhibited conformation for Ca(2+) transport but does not modify the affinity for Ca(2+) at the transmembrane domain. The C-terminal domain is affected by CaM and CaM-like treatments driving the auto-inhibited conformation E(1)I to the activated E(1)A conformation and thus modulating the transport of Ca(2+). This is reflected in the different apparent constants for Ca(2+) in the absence of CaM (calculated by Ca(2+)-ATPase activity) that sharply contrast with the lack of variation of the affinity for the Ca(2+) site at equilibrium. This is the first time that equilibrium constants for the dissociation of Ca(2+) and CaM ligands from PMCA complexes are measured through the change of transmembrane conformations of the pump. The data further suggest that the transmembrane domain of the PMCA undergoes major rearrangements resulting in altered lipid accessibility upon Ca(2+) binding and activation.


Asunto(s)
Azirinas/metabolismo , Calcio/metabolismo , Calmodulina/metabolismo , Membrana Celular/enzimología , Sondas Moleculares/metabolismo , Fosfatidilcolinas/metabolismo , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Membrana Celular/efectos de los fármacos , Quimotripsina/farmacología , Activación Enzimática/efectos de los fármacos , Humanos , Cinética , Ácido Oléico/farmacología , Ácidos Fosfatidicos/farmacología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/química , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estructura Terciaria de Proteína , Eliminación de Secuencia/efectos de los fármacos , Volumetría , Clorometilcetona Tosilisina/farmacología
10.
Hum Mol Genet ; 18(6): 1028-36, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19095717

RESUMEN

Age-dependent accumulation of partially deleted mitochondrial DNA (DeltamtDNA) has been suggested to contribute to aging and the development of age-associated diseases including Parkinson's disease. However, the molecular mechanisms underlying the generation and accumulation of DeltamtDNA have not been addressed in vivo. In this study, we have developed a mouse model expressing an inducible mitochondria-targeted restriction endonuclease (PstI). Using this system, we could trigger mtDNA double-strand breaks (DSBs) in adult neurons. We found that this transient event leads to the generation of a family of DeltamtDNA with features that closely resemble naturally-occurring mtDNA deletions. The formation of these deleted species is likely to be mediated by yet uncharacterized DNA repairing machineries that participate in homologous recombination and non-homologous end-joining. Furthermore, we obtained in vivo evidence that DeltamtDNAs with larger deletions accumulate faster than those with smaller deletions, implying a replicative advantage of smaller mtDNAs. These findings identify DSB, DNA repair systems and replicative advantage as likely mechanisms underlying the generation and age-associated accumulation of DeltamtDNA in mammalian neurons.


Asunto(s)
Envejecimiento/metabolismo , ADN Mitocondrial/metabolismo , Neuronas/metabolismo , Eliminación de Secuencia , Envejecimiento/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Roturas del ADN de Doble Cadena/efectos de los fármacos , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Doxiciclina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Especificidad de Órganos/efectos de los fármacos , Eliminación de Secuencia/efectos de los fármacos , Tetraciclina/farmacología
11.
Nat Commun ; 12(1): 5111, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34433825

RESUMEN

Mutational outcomes following CRISPR-Cas9-nuclease cutting in mammalian cells have recently been shown to be predictable and, in certain cases, skewed toward single genotypes. However, the ability to control these outcomes remains limited, especially for 1-bp insertions, a common and therapeutically relevant class of repair outcomes. Here, through a small molecule screen, we identify the ATM kinase inhibitor KU-60019 as a compound capable of reproducibly increasing the fraction of 1-bp insertions relative to other Cas9 repair outcomes. Small molecule or genetic ATM inhibition increases 1-bp insertion outcome fraction across three human and mouse cell lines, two Cas9 species, and dozens of target sites, although concomitantly reducing the fraction of edited alleles. Notably, KU-60019 increases the relative frequency of 1-bp insertions to over 80% of edited alleles at several native human genomic loci and improves the efficiency of correction for pathogenic 1-bp deletion variants. The ability to increase 1-bp insertion frequency adds another dimension to precise template-free Cas9-nuclease genome editing.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Sistemas CRISPR-Cas/efectos de los fármacos , Morfolinas/farmacología , Mutagénesis Insercional/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Tioxantenos/farmacología , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Línea Celular , Edición Génica , Humanos , Eliminación de Secuencia/efectos de los fármacos
12.
BMC Genomics ; 11: 499, 2010 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-20846421

RESUMEN

BACKGROUND: Classical and quantitative linkage analyses of genetic crosses have traditionally been used to map genes of interest, such as those conferring chloroquine or quinine resistance in malaria parasites. Next-generation sequencing technologies now present the possibility of determining genome-wide genetic variation at single base-pair resolution. Here, we combine in vivo experimental evolution, a rapid genetic strategy and whole genome re-sequencing to identify the precise genetic basis of artemisinin resistance in a lineage of the rodent malaria parasite, Plasmodium chabaudi. Such genetic markers will further the investigation of resistance and its control in natural infections of the human malaria, P. falciparum. RESULTS: A lineage of isogenic in vivo drug-selected mutant P. chabaudi parasites was investigated. By measuring the artemisinin responses of these clones, the appearance of an in vivo artemisinin resistance phenotype within the lineage was defined. The underlying genetic locus was mapped to a region of chromosome 2 by Linkage Group Selection in two different genetic crosses. Whole-genome deep coverage short-read re-sequencing (Illumina Solexa) defined the point mutations, insertions, deletions and copy-number variations arising in the lineage. Eight point mutations arise within the mutant lineage, only one of which appears on chromosome 2. This missense mutation arises contemporaneously with artemisinin resistance and maps to a gene encoding a de-ubiquitinating enzyme. CONCLUSIONS: This integrated approach facilitates the rapid identification of mutations conferring selectable phenotypes, without prior knowledge of biological and molecular mechanisms. For malaria, this model can identify candidate genes before resistant parasites are commonly observed in natural human malaria populations.


Asunto(s)
Artemisininas/farmacología , Evolución Molecular Dirigida/métodos , Resistencia a Medicamentos/genética , Genoma de Protozoos/genética , Mutación/genética , Plasmodium chabaudi/genética , Análisis de Secuencia de ADN/métodos , Animales , Artemisininas/uso terapéutico , Simulación por Computador , Variaciones en el Número de Copia de ADN/genética , Genes Protozoarios , Genotipo , Humanos , Mutación INDEL/genética , Malaria/tratamiento farmacológico , Malaria/parasitología , Mutagénesis Insercional/efectos de los fármacos , Mutagénesis Insercional/genética , Parásitos/efectos de los fármacos , Parásitos/genética , Fenotipo , Filogenia , Plasmodium chabaudi/efectos de los fármacos , Mutación Puntual/genética , Pirimetamina/farmacología , Eliminación de Secuencia/efectos de los fármacos , Eliminación de Secuencia/genética
13.
Lab Invest ; 90(9): 1385-95, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20567236

RESUMEN

Tumor necrosis factor-alpha (TNF-alpha) is a key regulator of adipose tissue mass, but mechanisms underlying this effect have not been fully elucidated. We found that exposure to TNF-alpha caused a significant decrease in the number of adipocytes, but not preadipocytes. Subsequent experiments revealed that TNF-alpha selectively deleted adipocytes through induction of apoptosis. Following exposure to TNF-alpha, rapid activation of nuclear factor-kappaB (NF-kappaB) was observed only in preadipocytes, but not in adipocytes. Inhibition of NF-kappaB rendered preadipocytes susceptible to TNF-alpha-induced apoptosis, suggesting that different activity of NF-kappaB is the determinant for the distinct apoptotic responses. During adipocyte differentiation, expression and activity of peroxisome proliferator-activated receptor-gamma (PPARgamma) were upregulated. Treatment of preadipocytes with a PPARgamma agonist attenuated NF-kappaB activation and rendered the cells vulnerable to TNF-alpha-induced apoptosis. Conversely, treatment of adipocytes with a PPARgamma antagonist enhanced NF-kappaB activation and conferred resistance to TNF-alpha-induced apoptosis, suggesting involvement of PPARgamma in the suppression of NF-kappaB in adipocytes. We also found that, following differentiation, expression and activity of CCAAT/enhancer binding protein (C/EBP), especially C/EBPalpha and C/EBPbeta, were upregulated in adipocytes. Overexpression of individual C/EBPs significantly inhibited activation of NF-kappaB in preadipocytes. Furthermore, transfection with siRNA for C/EBPalpha or C/EBPbeta enhanced activity of NF-kappaB in adipocytes, suggesting that C/EBP is also involved in the repression of NF-kappaB in adipocytes. These results suggested novel mechanisms by which TNF-alpha selectively deletes adipocytes in the adipose tissue. The C/EBP- and PPARgamma-mediated suppression of NF-kappaB may contribute to TNF-alpha-related loss of adipose tissue mass under certain pathological situations, such as cachexia.


Asunto(s)
Adipocitos , Proteínas Potenciadoras de Unión a CCAAT/genética , FN-kappa B/metabolismo , PPAR gamma/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Células 3T3-L1 , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Núcleo Celular/genética , Núcleo Celular/metabolismo , Ratones , FN-kappa B/genética , FN-kappa B/farmacología , PPAR gamma/genética , PPAR gamma/farmacología , Eliminación de Secuencia/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos
14.
Mutat Res ; 696(2): 160-6, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20100592

RESUMEN

Cadmium (Cd) is a non-essential element and is a widespread environmental pollutant. Exposure to cadmium can result in cytotoxic, carcinogenic and mutagenic effects. Mutagenesis is an indicative of genetic instability and can be assayed using microsatellites instability. The aim of the present study is to investigate; based on the rat model, the effects of oral acute (single 8.8mg/kg BW, 1/10 LD(50)) and sub-chronic (2.93mg/kg BW, 1/30 LD(50), for 4 weeks) doses of cadmium chloride on microsatellite instability at D6mit3, D9mit2 and D15Mgh1 loci, which are located in three different common fragile sites (6q21, 9q32-q33 and 15p14, respectively), within rat genome. In the acute study, no microsatellite instability (MSI) was observed in all the three tested loci (D6mit3, D9mit2 and D15Mgh1). In the sub-chronic study, the MSI were observed in the three studied loci and was in the form of deletion of 2-3bp or addition of 3-6bp. These finding may indicate the sensitivity of microsatellite sequences located at the fragile sites and the sensitivity of the simple sequence repeats (SSRs) assay for the detection of small variations in DNA sequence. However, additional chronic toxicological trials are needed in order to assess genotoxic effects of chronic exposure to Cd.


Asunto(s)
Cadmio/toxicidad , Sitios Frágiles del Cromosoma/efectos de los fármacos , Inestabilidad de Microsatélites , Repeticiones de Microsatélite/efectos de los fármacos , Mutágenos/toxicidad , Animales , Ratas , Eliminación de Secuencia/efectos de los fármacos
15.
Neurology ; 94(21): e2270-e2282, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32139505

RESUMEN

OBJECTIVE: To report safety, pharmacokinetics, exon 53 skipping, and dystrophin expression in golodirsen-treated patients with Duchenne muscular dystrophy (DMD) amenable to exon 53 skipping. METHODS: Part 1 was a randomized, double-blind, placebo-controlled, 12-week dose titration of once-weekly golodirsen; part 2 is an ongoing, open-label evaluation. Safety and pharmacokinetics were primary and secondary objectives of part 1. Primary biological outcome measures of part 2 were blinded exon skipping and dystrophin protein production on muscle biopsies (baseline, week 48) evaluated, respectively, using reverse transcription PCR and Western blot and immunohistochemistry. RESULTS: Twelve patients were randomized to receive golodirsen (n = 8) or placebo (n = 4) in part 1. All from part 1 plus 13 additional patients received 30 mg/kg golodirsen in part 2. Safety findings were consistent with those previously observed in pediatric patients with DMD. Most of the study drug was excreted within 4 hours following administration. A significant increase in exon 53 skipping was associated with ∼16-fold increase over baseline in dystrophin protein expression at week 48, with a mean percent normal dystrophin protein standard of 1.019% (range, 0.09%-4.30%). Sarcolemmal localization of dystrophin was demonstrated by significantly increased dystrophin-positive fibers (week 48, p < 0.001) and a positive correlation (Spearman r = 0.663; p < 0.001) with dystrophin protein change from baseline, measured by Western blot and immunohistochemistry. CONCLUSION: Golodirsen was well-tolerated; muscle biopsies from golodirsen-treated patients showed increased exon 53 skipping, dystrophin production, and correct dystrophin sarcolemmal localization. CLINICALTRIALSGOV IDENTIFIER: NCT02310906. CLASSIFICATION OF EVIDENCE: This study provides Class I evidence that golodirsen is safe and Class IV evidence that it induces exon skipping and novel dystrophin as confirmed by 3 different assays.


Asunto(s)
Distrofina/biosíntesis , Distrofia Muscular de Duchenne/tratamiento farmacológico , Oligonucleótidos/uso terapéutico , Administración Intravenosa , Adolescente , Niño , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Distrofina/genética , Técnica del Anticuerpo Fluorescente , Humanos , Masculino , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/sangre , Distrofia Muscular de Duchenne/genética , Eliminación de Secuencia/efectos de los fármacos
16.
Biochemistry ; 48(33): 7833-41, 2009 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-19618962

RESUMEN

Tandem lesions are comprised of two contiguously damaged nucleotides. Tandem lesions make up the major family of reaction products generated from a pyrimidine nucleobase radical, which are formed in large amounts by ionizing radiation. One of these tandem lesions contains a thymidine glycol lesion flanked on its 5'-side by 2-deoxyribonolactone (LTg). The replication of this tandem lesion was investigated in Escherichia coli using single-stranded genomes. LTg is a much more potent replication block than thymidine glycol and is bypassed only under SOS-induced conditions. The adjacent thymidine glycol does not significantly affect nucleotide incorporation opposite 2-deoxyribonolactone in wild-type cells. In contrast, the misinsertion frequency opposite thymidine glycol, which is negligible in the absence of 2-deoxyribonolactone, increases to 10% in wild-type cells when LTg is flanked by a 3'-dG. Experiments in which the flanking nucleotides are varied and in cells lacking one of the SOS-induced bypass polymerases indicate that the mutations are due to a mechanism in which the primer misaligns prior to bypassing the lesion, which allows for an additional nucleotide to be incorporated across from the 3'-flanking nucleotide. Subsequent realignment and extension results in the observed mutations. DNA polymerases II and IV are responsible for misalignment induced mutations and compete with DNA polymerase V which reads through the tandem lesion. These experiments reveal that incorporation of the thymidine glycol into a tandem lesion indirectly induces increases in mutations by blocking replication, which enables the misalignment-realignment mechanism to compete with direct bypass by DNA polymerase V.


Asunto(s)
Daño del ADN , ADN Bacteriano/efectos de los fármacos , Repeticiones de Dinucleótido/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Mutagénesis , Timidina/análogos & derivados , Regiones no Traducidas 5'/efectos de los fármacos , Regiones no Traducidas 5'/genética , Bacteriófago M13/genética , Reparación del ADN/efectos de los fármacos , Reparación del ADN/genética , ADN Bacteriano/síntesis química , Repeticiones de Dinucleótido/genética , Genoma Bacteriano/efectos de los fármacos , Radical Hidroxilo/toxicidad , Respuesta SOS en Genética/efectos de los fármacos , Eliminación de Secuencia/efectos de los fármacos , Azúcares Ácidos/toxicidad , Timidina/toxicidad
17.
Biochemistry ; 48(22): 5034-41, 2009 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-19378973

RESUMEN

Because all three protein components of prothrombinase, factors (f) Xa and Va and prothrombin, bind to negatively charged membrane phospholipids, the exact role of the membrane in the prothrombinase reaction has not been fully understood. In this study, we prepared deletion derivatives of fXa and prothrombin in which both the Gla and first EGF-like domains of the protease (E2-fXa) as well as the Gla and both kringle domains of the substrate (prethrombin-2) had been deleted. The fVa-mediated catalytic activity of E2-fXa toward prethrombin-2 was analyzed in both the absence and presence of phospholipids composed of 80% phosphatidylcholine (PC) and 20% phosphatidylserine (PS). PCPS markedly accelerated the initial rate of prethrombin-2 activation by E2-fXa, with the cofactor exhibiting saturation only in the presence of phospholipids (apparent K(d) of approximately 60 nM). Competitive kinetic studies in the presence of the two exosite-1-specific ligands Tyr(63)-sulfated hirudin(54-65) and TM456 suggested that while both peptides are highly effective inhibitors of the fVa-mediated activation of prethrombin-2 by E2-fXa in the absence of PCPS, they are ineffective competitors in the presence of phospholipids. Since neither E2-fXa nor prethrombin-2 can interact with membranes, these results suggest that interaction of fVa with PCPS improves the affinity of the activation complex for proexosite-1 of the substrate. Direct binding studies employing OG(488)-EGR-labeled fXa and E2-fXa revealed that the interaction of the Gla domain of fXa with PCPS also induces conformational changes in the protease to facilitate its high-affinity interaction with fVa.


Asunto(s)
Factor V/metabolismo , Factor Va/metabolismo , Factor Xa/metabolismo , Fosfolípidos/metabolismo , Protrombina/metabolismo , Ácido 1-Carboxiglutámico/química , Ácido 1-Carboxiglutámico/genética , Ácido 1-Carboxiglutámico/metabolismo , Unión Competitiva/efectos de los fármacos , Línea Celular , Factor de Crecimiento Epidérmico/química , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Factor V/química , Factor Va/antagonistas & inhibidores , Factor Va/química , Factor Xa/química , Humanos , Fosfatidilcolinas/farmacología , Fosfatidilserinas/farmacología , Fosfolípidos/química , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/genética , Protrombina/antagonistas & inhibidores , Protrombina/química , Protrombina/fisiología , Eliminación de Secuencia/efectos de los fármacos , Electricidad Estática , Especificidad por Sustrato/efectos de los fármacos , Tromboplastina/química , Tromboplastina/metabolismo
18.
J Appl Genet ; 50(3): 301-10, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19638689

RESUMEN

During fermentation, yeast cells are exposed to a number of stresses -- such as high alcohol concentration, high osmotic pressure, and temperature fluctuation - so some overlap of mechanisms involved in the response to these stresses has been suggested. To identify the genes required for tolerance to alcohol (ethanol, methanol, and 1-propanol), heat, osmotic stress, and oxidative stress, we performed genome-wide screening by using 4828 yeast deletion mutants. Our screens identified 95, 54, 125, 178, 42, and 30 deletion mutants sensitive to ethanol, methanol, 1-propanol, heat, NaCl, and H2O2, respectively. These deleted genes were then classified based on their cellular functions, and cross-sensitivities between stresses were determined. A large number of genes involved in vacuolar H(+)-ATPase (V-ATPase) function, cytoskeleton biogenesis, and cell wall integrity, were required for tolerance to alcohol, suggesting their protective role against alcohol stress. Our results revealed a partial overlap between genes required for alcohol tolerance and those required for thermotolerance. Genes involved in cell wall integrity and the actin cytoskeleton are required for both alcohol tolerance and thermotolerance, whereas the RNA polymerase II mediator complex seems to be specific to heat tolerance. However, no significant overlap of genes required for osmotic stress and oxidative stress with those required for other stresses was observed. Interestingly, although mitochondrial function is likely involved in tolerance to several stresses, it was found to be less important for thermotolerance. The genes identified in this study should be helpful for future research into the molecular mechanisms of stress response.


Asunto(s)
ADN de Hongos/genética , Regulación Fúngica de la Expresión Génica/genética , Genoma Fúngico/genética , Saccharomyces cerevisiae/genética , Estrés Fisiológico/genética , 1-Propanol/farmacología , ADN de Hongos/efectos de los fármacos , Etanol/farmacología , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Genoma Fúngico/efectos de los fármacos , Estudio de Asociación del Genoma Completo , Peróxido de Hidrógeno/farmacología , Metanol/farmacología , Presión Osmótica/fisiología , Estrés Oxidativo/genética , Saccharomyces cerevisiae/efectos de los fármacos , Eliminación de Secuencia/efectos de los fármacos , Eliminación de Secuencia/genética
19.
G3 (Bethesda) ; 9(1): 61-71, 2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30389796

RESUMEN

In at least some unicellular organisms, mutation rates are temporarily raised upon exposure to environmental stress, potentially contributing to the evolutionary response to stress. Whether this is true for multicellular organisms, however, has received little attention. This study investigated the effects of chronic mild stress, in the form of low-level copper and nickel exposure, on mutational processes in Daphnia pulex using a combination of mutation accumulation, whole genome sequencing and life-history assays. After over 100 generations of mutation accumulation, we found no effects of metal exposure on the rates of single nucleotide mutations and of loss of heterozygosity events, the two mutation classes that occurred in sufficient numbers to allow statistical analysis. Similarly, rates of decline in fitness, as measured by intrinsic rate of population increase and of body size at first reproduction, were negligibly affected by metal exposure. We can reject the possibility that Daphnia were insufficiently stressed to invoke genetic responses as we have previously shown rates of large-scale deletions and duplications are elevated under metal exposure in this experiment. Overall, the mutation accumulation lines did not significantly depart from initial values for phenotypic traits measured, indicating the lineage used was broadly mutationally robust. Taken together, these results indicate that the mutagenic effects of chronic low-level exposure to these metals are restricted to certain mutation classes and that fitness consequences are likely minor and therefore unlikely to be relevant in determining the evolutionary responses of populations exposed to these stressors.


Asunto(s)
Daphnia/genética , Aptitud Genética/genética , Genoma/efectos de los fármacos , Reproducción/efectos de los fármacos , Animales , Cobre/toxicidad , Daphnia/efectos de los fármacos , Aptitud Genética/efectos de los fármacos , Mutación/efectos de los fármacos , Acumulación de Mutaciones , Tasa de Mutación , Níquel/toxicidad , Reproducción/genética , Eliminación de Secuencia/efectos de los fármacos
20.
J Steroid Biochem Mol Biol ; 185: 200-211, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30194976

RESUMEN

Bisphenol A (BPA), an endocrine-disrupting chemical, is capable of producing reproductive toxicity. BPA results in mitochondrial DNA (mtDNA) deletion and mitochondrial dysfunction; however, the effect of BPA on the mitochondria of ovarian granulosa cells is not clear. Further, 1,25-dihydroxyvitamin D3 (1,25D3) may play a role in reproduction, because its receptor, VDR, contributes to the inhibition of oxidative stress and predominantly exists in the nuclei of granulosa cells. Hence, the role of 1,25D3 in BPA-mediated effects on mitochondrial function was examined in this study. Primary rat granulosa cells treated with BPA, 1,25D3, or both were subjected to molecular/biochemical assays to measure cell survival, mtDNA content, mtDNA deletion, superoxide dismutase activity, levels of proteins related to mitochondrial biogenesis, and mitochondrial function. We found that cell viability was dose-dependently reduced and reactive oxygen species (ROS) levels were increased by BPA treatment. BPA administration elevated Mn-superoxide dismutase (MnSOD) expression but negatively regulated total SOD activity. 1,25D3 treatment alone increased 17ß-estradiol secretion, ATP production, and cellular oxygen consumption. In cells treated with both agents, 1,25D3 enhanced BPA-induced MnSOD protein upregulation and blocked the BPA-mediated decline in total SOD activity. Furthermore, 1,25D3 attenuated BPA-mediated mtDNA deletion but showed no effect on BPA-induced increases in mtDNA content. Although BPA had no influence on the levels of peroxisome proliferator-activated receptor-γ coactivator-1 α, nuclear respiratory factor-1, mitochondrial transcription factor A, or cytochrome c oxidase subunit IV, 1,25D3 plus BPA markedly increased mitochondrial biogenesis-related protein expression via the PI3K-Akt pathway. Moreover, BPA-mediated negative regulation of cytochrome c oxidase subunit I levels and 17ß-estradiol secretion was attenuated by 1,25D3 pre-treatment. Our results suggest that 1,25D3 attenuates BPA-induced decreases in 17ß-estradiol and that treatment with 1,25D3 plus BPA regulates granulosa cell mitochondria by elevating mitochondrial biogenesis-related protein levels.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Calcitriol/farmacología , Disruptores Endocrinos/toxicidad , Estradiol/metabolismo , Células de la Granulosa/metabolismo , Mitocondrias/patología , Fenoles/toxicidad , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , ADN Mitocondrial/genética , Complejo IV de Transporte de Electrones/metabolismo , Femenino , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Receptores de Calcitriol/metabolismo , Eliminación de Secuencia/efectos de los fármacos , Eliminación de Secuencia/genética , Superóxido Dismutasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA