Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 10 de 10
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Appl Microbiol ; 123(6): 1584-1596, 2017 Dec.
Article de Anglais | MEDLINE | ID: mdl-28940494

RÉSUMÉ

AIMS: Test the choice of 16S rRNA gene amplicon and data analysis method on the accuracy of identification of clinically important bacteria utilizing a benchtop sequencer. METHODS AND RESULTS: Nine 16S rRNA amplicons were tested on an Ion Torrent PGM to identify 41 strains of clinical importance. The V1-V2 region identified 40 of 41 isolates to the species level. Three data analysis methods were tested, finding that the Ribosomal Database Project's SequenceMatch outperformed BLAST and the Ion Reporter Metagenomics analysis pipeline. Lastly, 16S rRNA gene sequencing mixtures of four species through a six log range of dilution showed species were identifiable even when present as 0·1% of the mixture. CONCLUSIONS: Sequencing the V1-V2 16S rRNA gene region, made possible by the increased read length Ion Torrent PGM sequencer's 400 base pair chemistry, may be a better choice over other commonly used regions for identifying clinically important bacteria. In addition, the SequenceMatch algorithm, freely available from the Ribosomal Database Project, is a good choice for matching filtered reads to organisms. Lastly, 16S rRNA gene sequencing's sensitivity to the presence of a bacterial species at 0·1% of a mixture suggests it has sufficient sensitivity for samples in which important bacteria may be rare. SIGNIFICANCE AND IMPACT OF THE STUDY: We have validated 16S rRNA gene sequencing on a benchtop sequencer including simple mixtures of organisms; however, our results highlight deficits for clinical application in place of current identification methods.


Sujet(s)
Bactéries/classification , ARN ribosomique 16S/composition chimique , Analyse de séquence d'ADN/méthodes , Bactéries/isolement et purification , Séquence nucléotidique , ARN ribosomique 16S/génétique , Analyse de séquence d'ADN/instrumentation
2.
Oncogene ; 25(39): 5405-15, 2006 Aug 31.
Article de Anglais | MEDLINE | ID: mdl-16636674

RÉSUMÉ

Activating enhancer-binding protein 2alpha (AP-2alpha) and activating enhancer-binding protein 2gamma (AP-2gamma) are transcription factors that bind GC-rich consensus sequences and regulate the expression of many downstream genes. AP-2alpha and AP-2gamma interact with p53 both physically and functionally. Expression microarray results in human breast carcinoma cells with forced p53 expression revealed AP-2gamma as a putative transcriptional target of p53. To confirm and extend these findings we measured the effects of forced p53 expression in human breast carcinoma cells by real-time reverse transcription-PCR, Western blotting, electrophoretic gel mobility shift assays, promoter reporter, chromatin immunoprecipitation and chromatin accessibility assays. Wild-type p53 expression rapidly induced not only AP-2gamma but also AP-2alpha mRNA. The subsequent increase in these proteins led to increased AP-2 DNA-binding and transactivating activity. Candidate p53-binding sites were identified in the AP-2alpha and AP-2gamma promoters. p53 binding to these cis-elements in vivo was also observed, together with a relaxation of chromatin structure in these regions. Finally, expression of either AP-2alpha or gamma inhibited growth of human breast carcinoma cells in vitro. Taken together, our findings indicate that these AP-2 genes are targets for transcriptional activation by p53 and suggest that AP-2 proteins may mediate some of the downstream effects of p53 expression such as inhibition of proliferation.


Sujet(s)
Gènes p53 , Facteur de transcription AP-2/génétique , Adénocarcinome , Tumeurs du sein/génétique , Division cellulaire , Lignée cellulaire tumorale , Amorces ADN , Femelle , Gènes rapporteurs , Humains , Séquençage par oligonucléotides en batterie , Plasmides , RT-PCR , Facteur de transcription AP-2/métabolisme , Transcription génétique , Transfection
3.
J Pharmacol Exp Ther ; 311(3): 968-81, 2004 Dec.
Article de Anglais | MEDLINE | ID: mdl-15302897

RÉSUMÉ

Dysregulation of epigenetic control is an important participant in carcinogenesis. The PML/RAR alpha translocation in acute promyelocytic leukemia (APL) is an example where the resultant fusion protein recruits histone deacetylase complexes to target genes resulting in their inappropriate transcriptional repression. All-trans-retinoic acid (ATRA) acts as a ligand that relieves this repression and produces an epigenetic transcriptional reprogramming of the cancer cell. CpG island microarrays were used to analyze the DNA methylation and histone acetylation state of the human APL cell line NB4 before and after differentiation with ATRA as well as normal peripheral blood mononuclear cells (PBMC). Over 70 CpG islands within 1 kb of transcription start of a known gene are aberrantly methylated in NB4 cells compared with PBMC; however, no changes in cytosine methylation were detected following ATRA-induced differentiation. With respect to histone H4 acetylation, over 100 single-copy CpG islands within 1 kb of transcription start of a known human gene became hyperacetylated following ATRA-induced differentiation. One CpG island was aberrantly methylated in NB4 cells, but became hyperacetylated and was induced following ATRA treatment and was associated with the HoxA1 gene, suggesting it may be a target gene of ATRA in APL. In addition to single-copy sequences, a selective increase in acetylation was detected in satellite DNA when compared with other high-copy sequences, such as Alu or rDNA. In summary, ATRA stimulates complex epigenomic changes during leukemic cell differentiation, and monitoring these changes may help to identify new targets of epigenetic dysfunction.


Sujet(s)
Ilots CpG/génétique , Cytosine/métabolisme , Histone/métabolisme , Leucémies/génétique , Leucémies/anatomopathologie , Séquençage par oligonucléotides en batterie , Acétylation , Différenciation cellulaire/génétique , Chromatine/métabolisme , ADN/génétique , ADN/isolement et purification , Bases de données génétiques , Banque de gènes , Humains , Immunoprécipitation , Hybridation in situ , Kératolytiques/pharmacologie , Méthylation , RT-PCR , Sulfites/composition chimique , Trétinoïne/pharmacologie
4.
Leukemia ; 17(2): 451-7, 2003 Feb.
Article de Anglais | MEDLINE | ID: mdl-12592346

RÉSUMÉ

Mutations of the ras gene are among the most commonly identified transforming events in human cancers, including multiple myeloma. Farnesyltransferase inhibitors (FTI) were developed to prevent Ras processing and induce cancer cell death. Several FTIs are in phase II and one is in phase III clinical trials. Preclinically, most of the focus has been on solid tumors, and the effects of FTIs in multiple myeloma have not been investigated. In this study we examined the cytotoxic activity and inhibition of Ras processing in three myeloma cell lines with differing Ras mutation status. H929 cells with activated N-Ras were more sensitive to FTI-277 treatment than 8226 and U266 cells with activated K-Ras or wild-type Ras, respectively. A combination of FTI-277 and a geranylgeranyltransferase I inhibitor (GGTI)-2166 inhibited K-Ras processing and enhanced cell death in 8226 cells. U266 cells and Bcl-x(L) transfectants were equally sensitive to FTI-277 treatment. Similarly, 8226 cells selected for resistance to various chemotherapeutic agents, which resulted in either P-glycoprotein overexpression, altered topoisomerase II activity, or elevated glutathione levels, were equally sensitive to FTI-277. These preclinical studies suggest that prenylation inhibitors may represent new therapeutic agents for the treatment of refractory or drug-resistant multiple myeloma.


Sujet(s)
Alkyl et aryl transferases/antagonistes et inhibiteurs , Apoptose/effets des médicaments et des substances chimiques , Division cellulaire/effets des médicaments et des substances chimiques , Antienzymes/pharmacologie , Méthionine/analogues et dérivés , Méthionine/pharmacologie , Survie cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques , Farnesyltranstransferase , Gènes ras/effets des médicaments et des substances chimiques , Humains , Myélome multiple/génétique , Cellules cancéreuses en culture , Test clonogénique de cellules souches tumorales
5.
Clin Cancer Res ; 7(12): 4262-71, 2001 Dec.
Article de Anglais | MEDLINE | ID: mdl-11751528

RÉSUMÉ

Our previous work demonstrated that the Janus kinase (JAK)-Stat3 pathway regulates expression of Bcl-x(L) in the U266 human multiple myeloma cell line and prevents Fas-mediated apoptosis. Inhibition of this pathway by the JAK selective kinase inhibitor AG490 or dominant-negative Stat3 protein results in down-regulation of Bcl-x(L) expression and enhanced sensitivity to Fas-mediated apoptosis. Because Bcl-x(L) has also been implicated in resistance to chemotherapeutic drugs, we investigated whether inhibition of the JAK-Stat3 pathway and subsequent reduction in Bcl-x(L) expression would also enhance cytotoxic drug activity. Contrary to this prediction, pretreatment of U266 myeloma cells with AG490, followed by exposure to topoisomerase II- inhibiting agents, antagonized drug-induced apoptosis. This effect correlated with reduced cyclin D1 expression and cell cycle arrest. The cell cycle arrest following AG490 pretreatment further correlated with reduced mitoxantrone-induced DNA double-strand breaks and reduced cell death, findings consistent with the critical requirement of DNA damage for drug cytotoxicity. These studies demonstrate that inhibition of the JAK-Stat3 pathway can result in paradoxical effects relative to cytotoxic drug response. These paradoxical responses may be explained by the findings that JAK-Stat3 signaling regulates the expression of multiple genes involved in controlling cell proliferation and apoptosis. Thus, understanding the cellular context of inhibiting signal transduction pathways is essential for the design of novel combination therapies for cancer.


Sujet(s)
Apoptose/physiologie , Survie cellulaire/effets des médicaments et des substances chimiques , Antienzymes/pharmacologie , Protéines proto-oncogènes , Inhibiteurs de la topoisomérase-II , Antigènes CD95/physiologie , Antinéoplasiques/pharmacologie , Régulation de l'expression des gènes tumoraux , Humains , Janus kinase 1 , Kinase Janus-2 , Janus kinase 3 , Myélome multiple/anatomopathologie , Protein-tyrosine kinases/antagonistes et inhibiteurs , Protéines proto-oncogènes c-bcl-2/génétique , Protéines recombinantes/antagonistes et inhibiteurs , Transfection , Cellules cancéreuses en culture , Tyrphostines/pharmacologie , Protéine bcl-X
6.
Blood ; 98(3): 805-13, 2001 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-11468182

RÉSUMÉ

Multiple myeloma (MM) is a clonal B-cell malignancy characterized by slow-growing plasma cells in the bone marrow (BM). Patients with MM typically respond to initial chemotherapies; however, essentially all progress to a chemoresistant state. Factors that contribute to the chemorefractory phenotype include modulation of free radical scavenging, increased expression of drug efflux pumps, and changes in gene expression that allow escape from apoptotic signaling. Recent data indicate that arsenic trioxide (As(2)O(3)) induces remission of refractory acute promyelocytic leukemia and apoptosis of cell lines overexpressing Bcl-2 family members; therefore, it was hypothesized that chemorefractory MM cells would be sensitive to As(2)O(3). As(2)O(3) induced apoptosis in 4 human MM cell lines: 8226/S, 8226/Dox40, U266, and U266/Bcl-x(L). The addition of interleukin-6 had no effect on cell death. Glutathione (GSH) has been implicated as an inhibitor of As(2)O(3)-induced cell death either through conjugating As(2)O(3) or by sequestering reactive oxygen induced by As(2)O(3). Consistent with this possibility, increasing GSH levels with N-acetylcysteine attenuated As(2)O(3) cytotoxicity. Decreases in GSH have been associated with ascorbic acid (AA) metabolism. Clinically relevant doses of AA decreased GSH levels and potentiated As(2)O(3)-mediated cell death of all 4 MM cell lines. Similar results were obtained in freshly isolated human MM cells. In contrast, normal BM cells displayed little sensitivity to As(2)O(3) alone or in combination with AA. Together, these data suggest that As(2)O(3) and AA may be effective antineoplastic agents in refractory MM and that AA might be a useful adjuvant in GSH-sensitive therapies. (Blood. 2001;98:805-813)


Sujet(s)
Composés de l'arsenic/pharmacologie , Acide ascorbique/pharmacologie , Myélome multiple/anatomopathologie , Oxydes/pharmacologie , Acétylcystéine/pharmacologie , Antinéoplasiques/pharmacologie , Antioxydants/pharmacologie , Trioxyde d'arsenic , Mort cellulaire/effets des médicaments et des substances chimiques , Synergie des médicaments , Glutathion/effets des médicaments et des substances chimiques , Humains , Peroxyde d'hydrogène/métabolisme , Plasmocytes/effets des médicaments et des substances chimiques , Superoxydes/métabolisme , Cellules cancéreuses en culture/effets des médicaments et des substances chimiques
8.
Blood ; 94(1): 265-74, 1999 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-10381522

RÉSUMÉ

We have previously shown that selection for resistance to the anthracenes, doxorubicin or mitoxantrone, results in coselection for resistance to CD95-mediated apoptosis (Landowski et al: Blood 89:1854, 1997). In the present study, we were interested in determining if the converse is also true; that is, does selection for CD95 resistance coselect for resistance to chemotherapeutic drugs. To address this question, we used two isogenic models of CD95-resistant versus CD95-sensitive cell lines: 8226/S myeloma cells selected for resistance to CD95-mediated apoptosis; and K562 cells expressing ectopic CD95. Repeated exposure of the CD95-sensitive human myeloma cell line, 8226/S, to agonistic anti-CD95 antibody resulted in a cell line devoid of CD95 receptor surface expression and completely resistant to CD95-mediated apoptosis. Multiple clonal populations derived from the CD95-resistant cell line showed no difference in sensitivity to doxorubicin, mitoxantrone, Ara-C, or etoposide, demonstrating that cross-resistance between Fas-mediated apoptosis and drug-induced apoptosis occurs only when cytotoxic drugs are used as the selecting agent. Using the inverse approach, we transfected the CD95-negative cell line, K562, with a CD95 expression vector. Clones expressing variable levels of cell-surface CD95 were isolated by limiting dilution, and analyzed for sensitivity to CD95-mediated apoptosis and response to chemotherapeutic drugs. We show that CD95 surface expression confers sensitivity to CD95-mediated apoptosis; however, it does not alter response to chemotherapeutic drugs. Similarly, doxorubicin-induced activation of caspases 3 and 8 was identical in the CD95-sensitive and CD95-resistant cell lines in both isogenic cell systems. In addition, prior treatment with the CD95 receptor-blocking antibody, ZB4, inhibited CD95-activated apoptosis in 8226/S cells, but had no effect on doxorubicin cytotoxicity. These results show that CD95 and chemotherapeutic drugs use common apoptotic effectors, but the point of convergence in these two pathways is downstream of CD95 receptor/ligand interaction.


Sujet(s)
Apoptose , Caspases/métabolisme , Résistance aux médicaments antinéoplasiques , Myélome multiple/anatomopathologie , Antigènes CD95/métabolisme , Anthracènes/pharmacologie , Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Doxorubicine/pharmacologie , Activation enzymatique , Humains , Cellules K562 , Mitoxantrone/pharmacologie , Myélome multiple/traitement médicamenteux , Myélome multiple/métabolisme , Cellules cancéreuses en culture
9.
Immunity ; 10(1): 105-15, 1999 Jan.
Article de Anglais | MEDLINE | ID: mdl-10023775

RÉSUMÉ

Interleukin 6 (IL-6) is the major survival factor for myeloma tumor cells and induces signaling through the STAT proteins. We report that one STAT family member, Stat3, is constitutively activated in bone marrow mononuclear cells from patients with multiple myeloma and in the IL-6-dependent human myeloma cell line U266. Moreover, U266 cells are inherently resistant to Fas-mediated apoptosis and express high levels of the antiapoptotic protein Bcl-xL. Blocking IL-6 receptor signaling from Janus kinases to the Stat3 protein inhibits Bcl-xL expression and induces apoptosis, demonstrating that Stat3 signaling is essential for the survival of myeloma tumor cells. These findings provide evidence that constitutively activated Stat3 signaling contributes to the pathogenesis of multiple myeloma by preventing apoptosis.


Sujet(s)
Apoptose/immunologie , Protéines de liaison à l'ADN/métabolisme , Myélome multiple/métabolisme , Transduction du signal/immunologie , Transactivateurs/métabolisme , Cellules 3T3 , Animaux , Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Protéines de liaison à l'ADN/physiologie , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Humains , Immunité innée/effets des médicaments et des substances chimiques , Souris , Myélome multiple/génétique , Myélome multiple/immunologie , Myélome multiple/anatomopathologie , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Protein-tyrosine kinases/physiologie , Protéines proto-oncogènes c-bcl-2/antagonistes et inhibiteurs , Protéines proto-oncogènes c-bcl-2/biosynthèse , Récepteurs à l'interleukine-6/physiologie , Facteur de transcription STAT-3 , Transduction du signal/génétique , Transactivateurs/physiologie , Transcription génétique/effets des médicaments et des substances chimiques , Transfection , Cellules cancéreuses en culture , Tyrphostines/pharmacologie , Protéine bcl-X
10.
Ann Nutr Metab ; 36(3): 167-74, 1992.
Article de Anglais | MEDLINE | ID: mdl-1530286

RÉSUMÉ

The effects of zinc deficiency were studied in mice submandibular salivary glands (SMG). Zn-restricted mice (Zn-) were maintained from weaning until adult age (60 days) with a powdered diet containing 3 mg Zn2+/kg. Pair-fed animals (30 mg Zn2+/kg powdered diet) and control animals fed a regular pelleted diet were also used. Total protein content and proteolytic activity of SMG did not differ among the groups, but morphometric evaluations revealed significant alterations in the nucleus/cytoplasm size ratios, most likely due to an absolute reduction in nuclear volume (control = 122.5 +/- 6.4; Zn- = 91.6 +/- 10.5; pair-fed = 125.1 +/- 6.8 microns 3) paralleled by an increase of the height of the duct epithelium (control = 70.5 +/- 3.0; Zn- = 90.5 +/- 4.2; pair-fed = 81.7 +/- 3.0 microns). The altered food consistency could be responsible for these morphological changes. In order to assess the subcellular distribution of SMG androgen receptors in conditions of chronic Zn deficiency, Zn- animals were mated and the F1 generation was fed as their dams until the age of 45 days. Cytosolic (in 105,000 g supernatants) and nuclear (KCl-extracted) SMG receptors were determined with [3H]R1881. The Zn- animals had reduced nuclear/cytosolic ratios of androgen receptors (control = 0.62; Zn- = 0.14), as an indication that chronically deficient Zn intake determines a sort of destabilization of the interactions of androgen-receptor complexes with target cell nucleus.


Sujet(s)
Androgènes/métabolisme , Glande submandibulaire/métabolisme , Zinc/déficit , Animaux , Noyau de la cellule/métabolisme , Noyau de la cellule/ultrastructure , Cytoplasme/métabolisme , Cytoplasme/ultrastructure , Fèces/composition chimique , Femelle , Mâle , Souris , Taille d'organe , Récepteurs aux androgènes/métabolisme , Glande submandibulaire/anatomopathologie , Testostérone/sang
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...