Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Prev Res (Phila) ; 15(2): 75-86, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34844992

RESUMO

Recent studies suggest that the fallopian tube epithelium (FTE) harbors the precursor for high-grade ovarian cancer, creating opportunities for targeting the FTE for ovarian cancer prevention. Preclinical evidence supports progestins as ovarian cancer preventives, but the effect of progestins on the FTE is not well characterized. The murine oviduct-specific glycoprotein promotor-driven simian virus 40 large T-Antigen (mogp-TAg) transgenic mouse model develops neoplastic lesions in the fallopian tube in a manner similar to that described in human fallopian tube and ovarian cancers. In this study, we investigated the inhibitory effects of the progestin depo-medroxyprogesterone acetate (DMPA) on fallopian tube carcinogenesis following treatment for 3 and 7 weeks in 5-week-old mogp-TAg mice. Overall, compared with vehicle-treated mice, the fallopian tube of DMPA-treated mice was significantly smaller (P < 0.0005), accumulated fewer p53-positive cells, had normal distribution of ciliated cells, less nuclear pleomorphism and epithelial tufting, and had a significantly lower proliferative index (P = 0.001). Accumulation of p53 signatures and serous tubal intraepithelial carcinomas (STIC) in the fallopian tube was significantly reduced in the DMPA (P < 0.0005) treatment group. Moreover, the fallopian tube of the DMPA-treated mice developed significantly less adenocarcinoma compared with vehicle (P < 0.005) at both treatment time points. DMPA treatment significantly induced cleaved caspase-3 (P < 0.0005) in the FTE compared with vehicle suggesting that apoptosis is involved in DMPA-related clearance of abnormal cells from the fallopian tube. These data demonstrate that DMPA targets early events in fallopian tube carcinogenesis by clearing genetically damaged cells, leading to marked reduction in adenocarcinoma, supporting progestins as chemopreventive agents for fallopian tube and ovarian cancers. PREVENTION RELEVANCE: The fallopian tube is thought to harbor the cell of origin for most ovarian cancers. We show in a mouse model of fallopian tube cancer that progestin eradicates the earliest known precancerous lesions and markedly inhibits fallopian tube carcinogenesis, adding to growing preclinical evidence supporting progestins as potent ovarian cancer chemopreventive agents.


Assuntos
Cistadenocarcinoma Seroso , Neoplasias das Tubas Uterinas , Neoplasias Ovarianas , Animais , Carcinogênese/patologia , Cistadenocarcinoma Seroso/tratamento farmacológico , Cistadenocarcinoma Seroso/patologia , Cistadenocarcinoma Seroso/prevenção & controle , Neoplasias das Tubas Uterinas/tratamento farmacológico , Neoplasias das Tubas Uterinas/patologia , Neoplasias das Tubas Uterinas/prevenção & controle , Tubas Uterinas/patologia , Feminino , Humanos , Camundongos , Camundongos Transgênicos , Neoplasias Ovarianas/patologia , Progestinas/farmacologia , Progestinas/uso terapêutico , Proteína Supressora de Tumor p53
2.
Gynecol Oncol ; 154(2): 432-440, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31178149

RESUMO

OBJECTIVE: Recent data show that simvastatin (SIM) and metformin (MET) have anti-proliferative effects in endometrial cancer cells. The combination (MET+SIM) inhibits tumor growth and metastasis in prostate cancer cells which possess similar molecular alterations to many early endometrial cancers. We tested the hypothesis that the anti-proliferative effects of MET+SIM in endometrial cancer cells would be greater than the effects of each agent alone. METHODS: RL95-2, HEC1B, and Ishikawa endometrial cancer cell lines were treated with MET and/or SIM. Growth inhibition was measured by MTS cell proliferation assays. Apoptosis was evaluated by caspase-3, Annexin V, and TUNEL assays and by apoptosis markers (BAX, Bcl-2, Bim) using western blot. Bim was silenced using Bim siRNA to confirm this apoptotic pathway. Treatment effects on the mTOR pathway were investigated by western blot using antibodies to phosphorylated (phospho)-AMPK and phospho-S6. RESULTS: MET+SIM synergistically inhibited growth in all three cell lines. The combination induced apoptosis as measured by TUNEL, Annexin V, and caspase-3 assays. Bim siRNA transfection abrogated this effect-silencing Bim in MET+SIM-treated RL95-2 cells rescued cell viability in MTS assays and reduced caspase-3 activity compared with control siRNA-transfected cells. Combination treatment upregulated phosphorylated AMPK and downregulated downstream phosphorylated S6, suggesting mTOR inhibition as a mechanism for these anti-proliferative effects. CONCLUSIONS: MET+SIM treatment synergistically inhibits endometrial cancer cell viability. This may be mediated by apoptosis and mTOR pathway inhibition. Our results provide preclinical evidence that the combination of these well-tolerated drugs may warrant further clinical investigation for endometrial cancer treatment.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias do Endométrio/tratamento farmacológico , Metformina/farmacologia , Sinvastatina/farmacologia , Western Blotting , Caspase 3/metabolismo , Linhagem Celular Tumoral/efeitos dos fármacos , Quimioterapia Combinada , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Serina-Treonina Quinases TOR/metabolismo
3.
Oncotarget ; 7(47): 77576-77590, 2016 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-27769055

RESUMO

Here, we evaluated the expression of CYP24A1, a protein that inactivates vitamin D in tissues. CYP24A1 expression was increased in advanced-stage endometrial tumors compared to normal tissues. Similarly, endometrial cancer cells expressed higher levels of CYP24A1 than immortalized endometrial epithelial cells. RT-PCR and Western blotting were used to examine CYP24A1 mRNA and protein levels in endometrial cancer cells after 8, 24, 72, and 120 h of exposure to progesterone, progestin derivatives and calcitriol, either alone or in combination. Progestins inhibited calcitriol-induced expression of CYP24A1 and splice variant CYP24SV mRNA and protein in cancer cells. Furthermore, actinomycin D, but not cycloheximide, blocked calcitriol-induced CYP24A1 splicing. siRNA-induced knockdown of CYP24A1 expression sensitized endometrial cancer cells to calcitriol-induced growth inhibition. These data suggest that CYP24A1 overexpression reduces the antitumor effects of calcitriol in cancer cells and that progestins may be beneficial for maintaining calcitriol's anti-endometrial cancer activity.


Assuntos
Calcitriol/farmacologia , Neoplasias do Endométrio/patologia , Progesterona/farmacologia , Vitamina D3 24-Hidroxilase/genética , Vitamina D3 24-Hidroxilase/metabolismo , Adulto , Idoso , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Pessoa de Meia-Idade , Gradação de Tumores
4.
Gynecol Oncol ; 143(1): 159-167, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27106018

RESUMO

OBJECTIVES: Previously we have shown in endometrial cells that progesterone (P4) and calcitriol (CAL, 1,25(OH)2D3) synergistically promote apoptosis and that progestins induce expression of the vitamin D receptor. In the current study we examined the progestin/vitamin D combination in ovarian cells and searched for other progestin-related effects on vitamin D metabolism that may underlie the novel interaction between progestins and vitamin D, including whether progestins inhibit CYP24A1, the enzyme that renders CAL inactive. METHODS: We investigated the impact of P4 on CAL-induced CYP24A1 expression in cancer cell lines expressing progesterone receptors (PRs), [OVCAR-5, OVCAR-3-PGR (PR-transfected OVCAR-3 ovarian line), and T47D-WT, T47D-A and T47D-B (breast lines expressing PRs or individual PR isoforms)] or lines that do not express PRs (OVCAR-3 and T47D-Y). We examined CYP24A1 expression using RT-PCR and western blotting, and apoptosis by TUNEL. We also investigated P4 inhibition of Cyp24a1 in ovaries from CAL-treated mice. RESULTS: CAL treatment induced CYP24A1 expression. When co-treated with P4, cell lines expressing PRs showed marked inhibition of CYP24A1 expression (p<0.001), along with increased apoptosis (p<0.01); cells not expressing PRs did not. Mouse ovaries showed a significant reduction in CAL-induced Cyp24a1 mRNA (p<0.001) and protein (p<0.01) in response to P4. CONCLUSIONS: We show for the first time that progestins and vitamin D synergistically reduce cell viability and induce apoptosis in ovarian cells and that progestins PR-dependently inhibit CAL-induced CYP24A1, thus extending CAL activity. The combination of progestins and vitamin D deserves further consideration as a strategy for inhibiting ovarian carcinogenesis.


Assuntos
Calcitriol/farmacologia , Quimioprevenção , Neoplasias Ovarianas/tratamento farmacológico , Progesterona/farmacologia , Vitamina D3 24-Hidroxilase/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Ovarianas/patologia , Ovário/enzimologia , Receptores de Progesterona/análise , Receptores de Progesterona/fisiologia
5.
Cancer Prev Res (Phila) ; 6(12): 1283-92, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24136864

RESUMO

Epidemiologic, laboratory, and animal evidence suggests that progestins and vitamin D may be potent ovarian cancer preventives. Our objectives were to evaluate progestins as reproductive tract cancer chemopreventives in the chicken, determine whether restricted ovulation affected the incidence of reproductive tract tumors, and assess whether vitamin D would confer cancer protection either alone or in addition to progestin. A total of 2,400 two-year-old Single Comb White Leghorns were randomized into six groups (400 each) with hormonal and dietary manipulation for 2 years as follows: (i) no intervention, regular feed/caloric intake, (ii) control, (iii) vitamin D, (iv) the progestin levonorgestrel, (v) vitamin D plus levonorgestrel, and (vi) the progestin Provera (medroxyprogesterone acetate). Groups 2 to 6 were caloric restricted to inhibit ovulation. Our results indicated that caloric restriction decreased egg production by more than 60%, and was associated with a greater than 70% decrease in reproductive tract cancers. Ovulatory events did not differ among the caloric-restricted groups (groups 2-6), except for the group receiving levonorgestrel, which had fewer ovulatory events than controls (P = 0.046). After correcting for egg production, birds receiving progestins had significantly fewer reproductive tract cancers [OR, 0.61; confidence interval (CI), 0.39-0.95; P = 0.03], with similar proportionate reductions in tumors arising in either the ovary or oviduct. Vitamin D did not significantly affect cancer incidence overall, or add to the cancer preventive effect of progestins. This study suggests a protective effect of progestins against ovarian and oviductal cancers. These data support the concept that progestins provide a chemopreventive effect unrelated to ovulation.


Assuntos
Adenocarcinoma/prevenção & controle , Neoplasias Ovarianas/prevenção & controle , Oviposição/efeitos dos fármacos , Ovulação/efeitos dos fármacos , Progestinas/uso terapêutico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica , Galinhas , Suplementos Nutricionais , Ovos , Feminino , Vitamina D/administração & dosagem
6.
Cancer Prev Res (Phila) ; 6(7): 731-43, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23682076

RESUMO

Human studies suggest that progesterone and calcitriol may prove beneficial in preventing or inhibiting oncogenesis, but the underlying mechanism is not fully understood. The current study investigates the effects of progesterone, calcitriol, and their combination on immortalized human endometrial epithelial cells and endometrial cancer cells and identifies their targets of action. Combination treatment with both agents enhanced vitamin D receptor expression and inhibited cell proliferation through caspase-3 activation and induction of G0-G1 cell-cycle arrest with associated downregulation of cyclins D1 and D3 and p27 induction. We used mass spectrometry-based proteomics to measure protein abundance differences between calcitriol-, progesterone-, or combination-exposed endometrial cells. A total of 117 proteins showed differential expression among these three treatments. Four proteins were then selected for validation studies: histone H1.4 (HIST1H1E), histidine triad nucleotide-binding protein 2 (HINT2), IFN-induced, double-stranded RNA-activated protein kinase (EIF2AK2), and Bcl-2-associated X protein (BAX). Abundance levels of selected candidates were low in endometrial cancer cell lines versus the immortalized endometrial epithelial cell line. All four proteins displayed elevated expression in cancer cells upon exposure to calcitriol, progesterone, or the combination. Further BAX analysis through gain- or loss-of-function experiments revealed that upregulation of BAX decreased cell proliferation by changing the BAX:BCL-2 ratio. Knockdown of BAX attenuated progesterone- and calcitriol-induced cell growth inhibition. Our results showed that progesterone and calcitriol upregulate the expression of BAX along with other apoptosis-related proteins, which induce inhibition of endometrial cancer cell growth by apoptosis and cell-cycle arrest.


Assuntos
Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/metabolismo , Calcitriol/farmacologia , Neoplasias do Endométrio/patologia , Progesterona/farmacologia , Receptores de Calcitriol/metabolismo , Western Blotting , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cromatografia Líquida , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/metabolismo , Endométrio/citologia , Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Feminino , Histonas/metabolismo , Humanos , Proteínas Mitocondriais/metabolismo , Proteômica , Espectrometria de Massas em Tandem , Proteína X Associada a bcl-2/metabolismo , eIF-2 Quinase/metabolismo
7.
Am J Obstet Gynecol ; 206(3): 253.e1-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22206747

RESUMO

OBJECTIVE: There is growing evidence that progestins and nonsteroidal antiinflammatory drugs (NSAIDs) may prevent ovarian cancer. Because both induce apoptosis, we investigated the potential for synergistic impact of combined drug treatment on cell death. STUDY DESIGN: Using normal and malignant human ovarian epithelial cells and an NSAID-sensitive human colon cancer cell line, we evaluated the effects of progestins and NSAIDs alone and in combination on apoptosis. RESULTS: Both progestins and NSAIDs dose dependently inhibited cell growth (P < .0001). Doses of NSAIDs or progestins that independently reduced cell viability by less than 30% synergistically reduced cell viability by 70-95% when combined. Similarly, the NSAID/progestin combination conferred 4- to 18-fold (P < .05) increased apoptosis over either treatment alone. CONCLUSION: Our results suggest it may be possible to combine progestins and NSAIDs to achieve ovarian cancer prevention at lower doses of each than are required for single administration, thereby lessening the risk of side effects posed by these agents.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Ovarianas/prevenção & controle , Ovário/efeitos dos fármacos , Ovário/patologia , Progestinas/farmacologia , Anti-Inflamatórios não Esteroides/administração & dosagem , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Humanos , Progestinas/administração & dosagem
8.
Cancer Prev Res (Phila) ; 2(2): 114-21, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19174584

RESUMO

We examined alterations in the p53 tumor suppressor gene and the ras and HER-2/neu oncogenes in chicken ovarian cancers to determine if these tumors have genetic alterations similar to those in human ovarian adenocarcinomas. Mutations in the p53 tumor suppressor gene and the H-ras and K-ras oncogenes were assessed by direct sequencing in 172 ovarian cancers obtained from 4-year-old birds enrolled at age 2 in two separate 2-year chemoprevention trials. Birds in trial B had approximately twice as many lifetime ovulations as those in trial A. Immunohistochemical staining for the HER-2/neu oncogene was done on a subset of avian ovarian and oviductal adenocarcinomas. Alterations in p53 were detected in 48% of chicken ovarian cancers. Incidence of p53 alterations varied according to the number of lifetime ovulations, ranging from 14% in trial A to 96% in trial B (P < 0.01). No mutations were seen in H-ras, and only 2 of 172 (1.2%) tumors had K-ras mutations. Significant HER-2/neu staining was noted in 10 of 19 ovarian adenocarcinomas but in only 1 of 17 oviductal adenocarcinomas. Similar to human ovarian cancers, p53 alterations are common in chicken ovarian adenocarcinomas and correlate with the number of lifetime ovulations. Ras mutations are rare, similar to high-grade human ovarian cancers. HER-2/neu overexpression is common and may represent a marker to exclude an oviductal origin in cancers involving both the ovary and oviduct.


Assuntos
Adenocarcinoma/genética , Genes ras/genética , Mutação/genética , Neoplasias Ovarianas/genética , Receptor ErbB-2/metabolismo , Proteína Supressora de Tumor p53/genética , Adenocarcinoma/patologia , Animais , Biomarcadores Tumorais/genética , Galinhas/genética , Galinhas/metabolismo , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Técnicas Imunoenzimáticas , Neoplasias Ovarianas/patologia , Oviductos/metabolismo , Oviductos/patologia , Reação em Cadeia da Polimerase
9.
Cancer Epidemiol Biomarkers Prev ; 17(3): 578-84, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18349275

RESUMO

BACKGROUND: Epidemiologic, animal, and human data suggest that progestins are potent endometrial cancer preventive agents. In the ovarian surface epithelium, progestins have been hypothesized to confer a cancer preventive effect via apoptosis and modulation of transforming growth factor-beta (TGF-beta). Given that the ovarian epithelium and endometrium share a common embryologic origin and similar reproductive and hormonal risk factors for malignancy, we tested the hypothesis that progestins confer biological effects in the endometrium similar to those in the ovary. METHODS: Postmenopausal female macaques (n = 78) were randomized into four groups to receive a diet for 36 months containing no hormone versus conjugated equine estrogen (CEE), medroxyprogesterone acetate (MPA), or CEE + MPA. The endometrium was then examined immunohistochemically for treatment-specific changes using antibodies to activated caspase-3 (for apoptosis), Ki-67 (proliferation), and the TGF-beta1, TGF-beta2, and TGF-beta3 isoforms. RESULTS: Percentages of caspase-positive endometrial glandular cells were 3- to 5-fold higher in CEE + MPA-treated animals compared with all others (P < 0.05). Caspase-expressing cells were six times more numerous in the endometrial stroma of animals treated with MPA alone relative to other groups (P < 0.0001). Induction of endometrial glandular cell apoptosis in the CEE + MPA-treated group was associated with a dramatic increase in expression of TGF-beta2 and TGF-beta3 in the stromal compartment of the endometrium (P < 0.0001). CONCLUSION: Progestin treatment activates chemopreventive biological effects in the endometrium that are similar to those in the ovarian surface epithelium. These data may facilitate identification of a chemopreventive approach that dramatically lessens the risk of both uterine and ovarian cancer.


Assuntos
Apoptose/efeitos dos fármacos , Progestinas/farmacologia , Fator de Crescimento Transformador beta/efeitos dos fármacos , Análise de Variância , Animais , Caspase 3/metabolismo , Feminino , Antígeno Ki-67/metabolismo , Modelos Logísticos , Macaca fascicularis , Pós-Menopausa , Estudos Prospectivos , Distribuição Aleatória
10.
J Surg Oncol ; 79(1): 17-29, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11754373

RESUMO

BACKGROUND AND OBJECTIVES: We propose that a growth factor receptor tyrosine kinase (RTK) inhibitor, such as tyrphostin A47, could serve as an adjunct to estrogen replacement therapy (ERT) for postmenopausal breast cancer survivors. Tyrphostins have been shown to block estrogen (E2)-induced proliferation in the human breast cancer MCF-7 cell line. Therefore, the effects of A47 on signal transduction, cell cycle progression, and apoptosis in E2-mediated breast cancer cell growth in vitro were investigated. METHODS: Cell growth was determined by MTT proliferation assay, cell cycle analysis assessed by flow cytometry, and RTK activation by Western blot. Apoptosis assays included nuclear staining, TdT-mediated dUTP-X nick end labeling, and caspase 3 activation. RESULTS: We find A47 selectively inhibits epidermal growth factor (EGF) and basic fibroblast growth factor but not insulin growth factor-1 proliferation. Although A47 inhibits EGF-induced phosphorylation of the EGF receptor in A431 cells, it does not consistently block MAP kinase phosphorylation. CONCLUSIONS: Taken together, A47 blocks E2/EGF-induced activation of EGFR and therefore interferes with the proximal EGFR signaling pathway. A47 also arrests the cells at the G1-S transition of the cell cycle and induces cell death by apoptosis. Thus, a growth factor RTK may be useful in blocking hormone-dependent tumor growth in an elevated E2 environment.


Assuntos
Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Fator de Crescimento Epidérmico/efeitos dos fármacos , Proteínas Tirosina Quinases/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Tirfostinas/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Neoplasias da Mama/prevenção & controle , Terapia de Reposição de Estrogênios , Estrogênios , Feminino , Fatores de Crescimento de Fibroblastos/efeitos dos fármacos , Citometria de Fluxo , Humanos , Marcação In Situ das Extremidades Cortadas , Recidiva Local de Neoplasia/prevenção & controle , Neoplasias Hormônio-Dependentes/prevenção & controle , Pós-Menopausa , Sobreviventes , Células Tumorais Cultivadas/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...