Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Mol Biol Rep ; 47(7): 5377-5383, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32623618

RESUMO

Antimicrobial irrigation solutions are widely used under clinical settings. Their effect on dental tissue is a subject of recent research, which aims for a safer irrigant for clinical use. In this regard, here our goal was to evaluate the cytotoxicity and the genotoxicity of calcium hypochlorite (Ca(OCl)2) solution, along with NaOCl, on Mouse embryonic fibroblast cells (NIH3T3). First, Cells were treated either with NaOCl or Ca(OCl)2 in a time- and dose-dependent manner for cytotoxicity by 3-(4,5-dimethylthiazolyl-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, then cell viability was calculated according to cell proliferation plots. Secondly, genotoxicity was assessed by Comet assay. Data were statistically analyzed by Tukey's test (P < .05). NaOCl and Ca(OCl)2 had similar effects on cellular viability at 3 and 6 h treatments. Cell viability of Ca(OCl)2 at concentrations of 0.0125%, 0.025%, 0.05%, or 0.125% was significantly lower than that of NaOCl at 24 h treatment (P < .05).Comparing Ca(OCl)2 and NaOCl treatments at all time points and concentrations, the damaged cell number of Ca(OCl)2 was almost fourfold higher than that of NaOCl. In conclusion, both, NaOCl and Ca(OCl)2 solutions were cytotoxic and genotoxic to NIH3T3, however, Ca(OCl)2 had a significantly higher damaged cell percentage than NaOCl at all time points and concentrations investigated.


Assuntos
Compostos de Cálcio/farmacologia , Compostos de Cálcio/toxicidade , Animais , Anti-Infecciosos/farmacologia , Compostos de Cálcio/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Ensaio Cometa , Camundongos , Células NIH 3T3 , Hipoclorito de Sódio/metabolismo , Hipoclorito de Sódio/farmacologia , Hipoclorito de Sódio/toxicidade
2.
Environ Sci Pollut Res Int ; 29(10): 14767-14779, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34617235

RESUMO

Microplastics and associated adverse effects have been on the global agenda in recent years. Because of its importance as a model organism for studies on developmental biology, Xenopus laevis has been chosen as the study animal in in vitro teratogenesis studies. FETAX test uses early-stage embryos of X. laevis to measure the potential of substances to cause mortality, malformation, and growth inhibition in developing embryos. The aim of this study was to examine the effects of high molecular weight polyvinyl chloride (HMW-PVC) on parental X. laevis frogs and their embryos using the FETAX test. To this purpose, a HMW-PVC dose of 1% of body weight/twice each week was provided to frogs by oral gavage throughout 6 weeks. After the procedure, oocytes and sperms of HMW-PVC-exposed frogs were fertilized and FETAX was applied to selected embryos. After the completion of a 96-h incubation period, tadpoles were examined, their live/dead status were determined, their lengths were measured, and their anomalies were photographed. Besides, excised organs of the parental frogs were referred to histopathology examination. On the other hand, the mRNA expression levels of Hsp70, Myf5, Bmp4, Pax6, and Esr1 genes were determined by applying real-time quantitative PCR method to cDNA which was synthesized from the total RNA of embryos. The results showed that treatment with HMW-PVC dose of 1% of body weight/twice each week caused malformations and decreased viability. Hsp70 and Pax6 gene expression levels significantly decreased in all assay groups, as compared with controls. Lung and intestine tissues showed normal appearance in histopatological examination. Further research is required to explain the whole effects of HMW-PVC exposure on X. laevis embryos.


Assuntos
Embrião não Mamífero , Cloreto de Polivinila , Animais , Desenvolvimento Embrionário , Masculino , Peso Molecular , Plásticos , RNA Mensageiro/genética , Teratogênicos , Xenopus laevis/genética
3.
J Clin Endocrinol Metab ; 93(5): 1915-23, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18270252

RESUMO

CONTEXT: The enzyme 17beta-hydroxysteroid dehydrogenase type 2 (HSD17B2) exerts a local antiestrogenic effect by metabolizing biologically active estradiol to inactive estrone in endometrial epithelial cells. Retinoic acid (RA) induces HSD17B2 expression, but the underlying mechanism is not known. OBJECTIVE: Our objective was to elucidate the molecular mechanisms responsible for HSD17B2 expression in human endometrial cells. METHOD: Human endometrial Ishikawa and RL95-2 cell lines were cultured in the presence or absence of RA to analyze endogenous HSD17B2 expression, transcription factor complex formation, and promoter activity. RESULTS: RA induced HSD17B2 mRNA levels in a dose- and time-dependent manner in endometrial cells. The RA antagonist ANG11273 abolished RA-induced HSD17B2 expression. Small interfering RNA ablation of RA receptor (RAR)alpha or retinoid X receptor (RXR)alpha completely blocked RA-induced HSD17B2 gene expression. Analysis of serial deletion and site-directed mutants of the HSD17B2 promoter fused to a reporter gene indicated that RA induction requires a cis-regulatory sequence that binds the specificity protein (SP) class of transcription factors. Chromatin-immunoprecipitation-PCR and gel-shift assays showed that RARalpha/RXRalpha and SP1/SP3 interact with this HSD17B2 promoter sequence. Small interfering RNA ablation of SP1 and SP3 expression markedly decreased HSD17B2 basal expression and blocked RA-induced expression. Finally, immunoprecipitationimmunoblotting demonstrated RA-induced interactions between RARalpha/RXRalpha and SP1/SP3 in intact endometrial cells. CONCLUSIONS: In endometrial epithelial cells, RA stimulates formation of a multimeric complex comprised of RARalpha/RXRalpha tethered to transcription factors SP1 and SP3 on the HSD17B2 promoter. Assembly of this transcriptional complex is necessary for RA induction of HSD17B2 expression and may be an important mechanism for local estradiol inactivation in the endometrium.


Assuntos
17-Hidroxiesteroide Desidrogenases/genética , Endométrio/enzimologia , Estradiol/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Receptores do Ácido Retinoico/fisiologia , Fator de Transcrição Sp1/fisiologia , Fator de Transcrição Sp3/fisiologia , Tretinoína/farmacologia , Linhagem Celular Tumoral , Células Epiteliais/enzimologia , Estradiol Desidrogenases , Feminino , Humanos , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas , RNA Interferente Pequeno/farmacologia
4.
J Clin Endocrinol Metab ; 92(5): 1979-82, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17341559

RESUMO

CONTEXT: Uterine leiomyomata are common tumors that cause irregular uterine bleeding and pregnancy loss and depend on estrogen for growth. Aromatase catalyzes the conversion of androgens to estrogens. Aromatase expression is regulated via alternatively used promoters in the placenta (I.1 and I.2a), fat (I.4, I.3, and II), bone (I.6), and gonads (II). A prostaglandin E(2)/cAMP-dependent pathway regulates coordinately the proximal promoters I.3/II, whereas glucocorticoids and cytokines regulate the distal promoter I.4. Use of each promoter gives rise to a population of aromatase mRNA species with unique 5'-untranslated regions (5'-UTRs). Uterine leiomyoma tissue, but not normal myometrium, overexpresses aromatase leading to estrogen-stimulated cell proliferation. Aromatase inhibitor treatment shrank uterine leiomyomata in a few women. OBJECTIVE AND DESIGN: Promoter I.4 was reported to regulate aromatase expression in uterine leiomyomata from a group of Japanese women. Here, we used two independent techniques to identify the promoters that regulate aromatase expression in uterine leiomyomata (n = 30) from 23 African-American, Hispanic, and white women. RESULTS: Rapid amplification of 5'-cDNA ends of aromatase mRNA species revealed the following distribution of promoter usage in leiomyomata: promoters I.3/II, 61.5%; I.2a, 15.4%; I.6, 15.4%; and I.4, 7.7%. Real-time PCR, which quantifies mRNA species with promoter-specific 5'-UTRs, revealed the following distribution for each 5'-UTR as a fraction of total aromatase mRNA: I.3/II, 69.6%; I.4, 7.3%; and other promoters, 23.1%. CONCLUSIONS: The primary in vivo aromatase promoter in leiomyoma tissues in non-Asian U.S. women is the prostaglandin E(2)/cAMP-responsive I.3/II region. Alternative signals may stimulate aromatase expression that is a common biological phenotype in uterine leiomyomata.


Assuntos
Aromatase/biossíntese , Aromatase/genética , Leiomioma/enzimologia , Leiomioma/genética , Regiões Promotoras Genéticas/genética , Neoplasias Uterinas/enzimologia , Neoplasias Uterinas/genética , Regiões 5' não Traduzidas/genética , Adulto , Feminino , Amplificação de Genes , Regulação Enzimológica da Expressão Gênica/fisiologia , Humanos , Pessoa de Meia-Idade , RNA Complementar/biossíntese , RNA Complementar/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
J Steroid Biochem Mol Biol ; 106(1-5): 81-96, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17590327

RESUMO

Pathogenesis and growth of three common women's cancers (breast, endometrium and ovary) are linked to estrogen. A single gene encodes the key enzyme for estrogen biosynthesis named aromatase, inhibition of which effectively eliminates estrogen production in the entire body. Aromatase inhibitors successfully treat breast cancer, whereas their roles in endometrial and ovarian cancers are less clear. Ovary, testis, adipose tissue, skin, hypothalamus and placenta express aromatase normally, whereas breast, endometrial and ovarian cancers overexpress aromatase and produce local estrogen exerting paracrine and intracrine effects. Tissue-specific promoters distributed over a 93-kb regulatory region upstream of a common coding region alternatively control aromatase expression. A distinct set of transcription factors regulates each promoter in a signaling pathway- and tissue-specific manner. In cancers of breast, endometrium and ovary, aromatase expression is primarly regulated by increased activity of the proximally located promoter I.3/II region. Promoters I.3 and II lie 215 bp from each other and are coordinately stimulated by PGE(2) via a cAMP-PKA-dependent pathway. In breast adipose fibroblasts exposed to PGE(2) secreted by malignant epithelial cells, PKC is also activated, and this potentiates cAMP-PKA-dependent induction of aromatase. Thus, inflammatory substances such as PGE(2) may play important roles in inducing local production of estrogen that promotes tumor growth.


Assuntos
Aromatase/metabolismo , Neoplasias da Mama/enzimologia , Neoplasias do Endométrio/enzimologia , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Neoplasias Ovarianas/enzimologia , Animais , Aromatase/genética , Neoplasias da Mama/genética , Neoplasias do Endométrio/genética , Feminino , Humanos , Neoplasias Ovarianas/genética
6.
Semin Reprod Med ; 22(1): 5-9, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15083376

RESUMO

The human CYP19 (p450arom) gene is located in the 21.2 region on the long arm of chromosome 15 (15q21.2). This gene spans a region that consists of a 30 kb coding region and a 93 kb regulatory region ( approximately 123 kb total length). Its regulatory region contains at least 10 distinct promoters regulated in a tissue- or signaling pathway-specific manner. The Human Genome Project data published in 2000 enabled us to accurately align these promoters within the 93 kb regulatory region of the p450arom gene. Each promoter is regulated by a distinct set of regulatory sequences in DNA and transcription factors that bind to these specific sequences. In most vertebrates, p450arom expression is under the control of gonad- and brain-specific promoters. In humans, however, there are at least eight additional promoters that were apparently recruited throughout evolution, possibly via alterations in DNA. A critical mechanism that permits the use of such a large number of promoters seems to be the extremely promiscuous nature of the common splice acceptor site because, with activation of each promoter, an untranslated first exon is spliced onto this common junction immediately upstream of the translation start site in the coding region. These partially tissue-specific promoters are used in the gonads, bone, brain, vascular tissue, adipose tissue, skin, fetal liver, and placenta for estrogen biosynthesis necessary for human physiology. Ovary and testis use promoter II, which is located immediately upstream of the coding region. The adipose tissue in general, including adipose tissue of the disease-free breast, on the other hand, maintains low levels of aromatase expression primarily via promoter I.4, which lies 73 kb upstream of the common coding region. Promoters I.3 and II are used only minimally in normal breast adipose tissue. Promoter II and I.3 activities in breast cancer tissue, however, are strikingly increased. Additionally, the endothelial-type promoter I.7 is also upregulated in breast cancer. Therefore, breast tumor tissue takes advantage of four promoters (II, I.3, I.7, and I.4) for aromatase expression and estrogen production. The sum of p450arom mRNA species arising from these four promoters contributes significantly to elevated levels of total p450arom mRNA in breast cancer in contrast to the normal breast that uses promoter I.4. Because each mRNA species contains the identical coding region regardless of the variable untranslated first exon, the encoded protein functions as the aromatase enzyme in each case.


Assuntos
Aromatase/genética , Aromatase/metabolismo , Neoplasias da Mama/enzimologia , Neoplasias da Mama/metabolismo , DNA/genética , Bases de Dados Factuais , Estrogênios/metabolismo , Evolução Molecular , Feminino , Genoma Humano , Humanos , Regiões Promotoras Genéticas
7.
Semin Reprod Med ; 22(1): 45-50, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15083380

RESUMO

Aromatase p450 (p450arom) is the key enzyme for biosynthesis of estrogen, which is an essential hormone for the establishment and growth of endometriosis. There is no detectable aromatase enzyme activity in normal endometrium; therefore, estrogen is not locally produced in endometrium. Endometriosis tissue, however, contains very high levels of aromatase enzyme, which leads to production of significant quantities of estrogen. Moreover, one of the best-known mediators of inflammation and pain, prostaglandin E (2), strikingly induces aromatase enzyme activity and formation of local estrogen in this tissue. Additionally, estrogen itself stimulates cyclo-oxygenase-2 and therefore increases the formation of prostaglandin E (2) in endometriosis. We were able to target this positive feedback cycle in endometriosis using aromatase inhibitors. In fact, pilot trials showed that aromatase inhibitors could decrease pelvic pain associated with endometriosis.


Assuntos
Aromatase/metabolismo , Endometriose/enzimologia , Endometriose/tratamento farmacológico , Inibidores Enzimáticos/uso terapêutico , Estrogênios/biossíntese , Feminino , Regulação da Expressão Gênica , Humanos , Células Estromais/enzimologia
8.
Genet Test Mol Biomarkers ; 15(10): 701-7, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21699408

RESUMO

AIM: The purpose of this article was to evaluate the reliability of maternal serum triple marker screening of alpha-fetoprotein, human chorionic gonadotropin, and unconjugated estriol for the prenatal diagnosis of fetal chromosomal abnormalities in Turkish pregnant women. METHOD: Medical records were used to analyze indications of amniocentesis and quantitative fluorescent-polymerase chain reaction. Anomaly screening was performed for all patients between 13 and 22 weeks of pregnancy. A total of 1725 pregnancies with chromosomal abnormality risk according to triple test screening were accepted for fetal chromosome analysis and quantitative fluorescent-polymerase chain reaction. RESULTS: Chromosomal aberrations were observed in 56 (3.2%) cases. About 44.6% of the abnormalities detected were numerical aberrations; however, 55.3% of the abnormalities were structural aberrations. Abnormalities detected were inversion of chromosome 9 in 20 cases, trisomy 21 in 14 cases, 46,XX/47,XX, +21 in 1 case, trisomy 18 in 2 cases, trisomy 13 in 1 case, 47,XXY, in 1 case, 45,X, in 1 case, structural abnormalities in 12 cases, and mosaic or tetraploidy in 6 cases. CONCLUSION: Second trimester triple test is an effective screening tool for detecting fetal Down syndrome in Turkish women.


Assuntos
Aberrações Cromossômicas , Análise Citogenética/métodos , Doenças Fetais/diagnóstico , Diagnóstico Pré-Natal/métodos , Adolescente , Adulto , Feminino , Doenças Fetais/genética , Humanos , Recém-Nascido , Masculino , Gravidez , Reprodutibilidade dos Testes , Soro/metabolismo , Turquia , Adulto Jovem
9.
Biol Reprod ; 75(4): 605-14, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16807381

RESUMO

The opposing actions of estrogen and progesterone during the menstrual cycle regulate the cyclical and predictable endometrial proliferation and differentiation that is required for implantation. Progesterone indirectly stimulates the expression of 17beta hydroxysteroid dehydrogenase type 2 (HSD17B2), which catalyzes the conversion of biologically potent estradiol to weakly estrogenic estrone in the endometrial epithelium. We previously demonstrated upregulation of the HSD17B2 gene in human endometrial epithelial cells by factors secreted from endometrial stromal cells in response to progesterone. We investigated the underlying mechanism by which these stroma-derived, progesterone-induced paracrine factors stimulate HSD17B2 expression. Here, we show that transcription factors SP1 and SP3 interact with specific motifs in HSD17B2 promoter to upregulate enzyme expression in human endometrial epithelial cell lines. Conditioned medium (CM) from progestin-treated stromal cells increased levels of SP1 and SP3 in endometrial epithelial cells and induced HSD17B2 mRNA expression. Mithramycin A, an inhibitor of SP1-DNA interaction, reduced epithelial HSD17B2 promoter activity in a dose-dependent manner. Serial deletion and site-directed mutants of the HSD17B2 promoter demonstrated that two overlapping SP1 motifs (nt -82/-65) are essential for induction of promoter activity by CM or overexpression of SP1/SP3. CM markedly enhanced, whereas anti-SP1/SP3 antibodies inhibited, binding of nuclear proteins to this region of the HSD17B2 promoter. In vivo, we demonstrated a significant spatiotemporal association between epithelial SP1/SP3 and HSD17B2 levels in human endometrial biopsies. Taken together, these data suggest that HSD17B2 expression in endometrial epithelial cells, and, therefore, estrogen inactivation, is regulated by SP1 and SP3, which are downstream targets of progesterone-dependent paracrine signals originating from endometrial stromal cells.


Assuntos
17-Hidroxiesteroide Desidrogenases/genética , Endométrio/metabolismo , Progesterona/metabolismo , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição Sp3/metabolismo , 17-Hidroxiesteroide Desidrogenases/efeitos dos fármacos , 17-Hidroxiesteroide Desidrogenases/metabolismo , Adulto , Sequência de Bases , Sítios de Ligação , Células Cultivadas , Células Epiteliais/metabolismo , Estradiol Desidrogenases , Feminino , Sequência Rica em GC , Humanos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Plicamicina/análogos & derivados , Plicamicina/farmacologia , Gravidez , Regiões Promotoras Genéticas , Deleção de Sequência , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp3/genética
10.
Pharmacol Rev ; 57(3): 359-83, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16109840

RESUMO

A single gene encodes the key enzyme for estrogen biosynthesis termed aromatase, inhibition of which effectively eliminates estrogen production. Aromatase inhibitors successfully treat breast cancer and endometriosis, whereas their roles in endometrial cancer, uterine fibroids, and aromatase excess syndrome are less clear. Ovary, testis, adipose tissue, skin, hypothalamus, and placenta express aromatase normally, whereas breast and endometrial cancers, endometriosis, and uterine fibroids overexpress aromatase and produce local estrogen that exerts paracrine and intracrine effects. Tissue-specific promoters distributed over a 93-kilobase regulatory region upstream of a common coding region alternatively control aromatase expression. A distinct set of transcription factors regulates each promoter in a signaling pathway- and tissue-specific manner. Three mechanisms are responsible for aromatase overexpression in a pathologic tissue versus its normal counterpart. First, cellular composition is altered to increase aromatase-expressing cell types that use distinct promoters (breast cancer). Second, molecular alterations in stromal cells favor binding of transcriptional enhancers versus inhibitors to a normally quiescent aromatase promoter and initiate transcription (breast/endometrial cancer, endometriosis, and uterine fibroids). Third, heterozygous mutations, which cause the aromatase coding region to lie adjacent to constitutively active cryptic promoters that normally transcribe other genes, result in excessive estrogen formation owing to the overexpression of aromatase in many tissues.


Assuntos
Aromatase/biossíntese , Neoplasias da Mama/enzimologia , Estrogênios/metabolismo , Regulação Enzimológica da Expressão Gênica , Doenças Ovarianas/enzimologia , Animais , Aromatase/genética , Inibidores da Aromatase/farmacologia , Neoplasias da Mama/metabolismo , Feminino , Humanos , Doenças Ovarianas/metabolismo , Regiões Promotoras Genéticas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA