Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Lancet ; 402(10410): 1347-1355, 2023 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-37678290

RESUMO

BACKGROUND: The growing field of assisted reproductive techniques, including frozen-thawed embryo transfer (FET), should lead the way to the best sustainable health care without compromising pregnancy chances. Correct timing of FET is crucial to allow implantation of the thawed embryo. Nowadays, timing based on hospital-controlled monitoring of ovulation in the natural cycle of a woman is the preferred strategy because of the assumption of favourable fertility prospects. However, home-based monitoring is a simple method to prevent patient travel and any associated environmental concerns. We compared ongoing pregnancy rates after home-based monitoring versus hospital-controlled monitoring with ovulation triggering. METHODS: This open-label, multicentre, randomised, non-inferiority trial was undertaken in 23 hospitals and clinics in the Netherlands. Women aged between 18 and 44 years with a regular ovulatory menstrual cycle were randomly assigned in a 1:1 ratio via a web-based randomisation program to home-based monitoring or hospital-controlled monitoring. Those who analysed the data were masked to the groups; those collecting the data were not. All endpoints were analysed by intention to treat and per protocol. Non-inferiority was established when the lower limit of the 90% CI exceeded -4%. This study was registered at the Dutch Trial Register (Trial NL6414). FINDINGS: 1464 women were randomly assigned between April 10, 2018, and April 13, 2022, with 732 allocated to home-based monitoring and 732 to hospital-controlled monitoring. Ongoing pregnancy occurred in 152 (20·8%) of 732 in the home-based monitoring group and in 153 (20·9%) of 732 in the hospital-controlled monitoring group (risk ratio [RR] 0·99 [90% CI 0·81 to 1·22]; risk difference [RD] -0·14 [90% CI -3·63 to 3·36]). The per-protocol analysis confirmed non-inferiority (152 [21·0%] of 725 vs 153 [21·0%] of 727; RR 1·00 (90% CI 0·81 to 1·23); RD -0·08 [90% CI -3·60 to 3·44]). INTERPRETATION: Home-based monitoring of ovulation is non-inferior to hospital-controlled monitoring of ovulation to time FET. FUNDING: The Dutch Organisation for Health Research and Development.

2.
J Steroid Biochem Mol Biol ; 109(3-5): 219-23, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18434135

RESUMO

Tamoxifen is used as adjuvant treatment for postmenopausal breast cancer patients. The mechanism of action of tamoxifen in breast cancer patients is that tamoxifen inhibits growth of cancer cells by competitive antagonism for estrogens at the estrogen receptor (ER). In the endometrium, tamoxifen has an effect that varies with the ambient concentration of estrogen: in premenopausal women (high estrogen levels), tamoxifen displays an estrogen-antagonistic effect, while in postmenopausal women (low estrogen levels), tamoxifen displays an estrogen-agonistic mode of action. Here, using microarray technology we have compared estrogen signaling with tamoxifen signaling in the human endometrium. It was observed that on the one hand tamoxifen-treatment results in modulation of expression of specific genes (370 genes) and on the other hand tamoxifen-treatment results in modulation of a set of genes which are also regulated by estrogen treatment (142 genes). Upon focusing on regulation of proliferation, we found that tamoxifen-induced endometrial proliferation is largely accomplished by using the same set of genes as are regulated by estradiol. So, as far as regulation of proliferation goes, tamoxifen seems to act as estrogen agonist. Furthermore, tamoxifen-specific gene regulation may explain why tamoxifen-induced endometrial tumors behave more aggressively than sporadic endometrial tumors.


Assuntos
Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Estrogênios/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tamoxifeno/farmacologia , Análise por Conglomerados , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Pólipos/patologia
3.
J Soc Gynecol Investig ; 12(7): e55-67, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16202921

RESUMO

OBJECTIVES: In different tissues, estrogens, selective estrogen receptor modulators (SERMs), and anti-estrogens exert different biologic activities. For the endometrium, estradiol and tamoxifen induce proliferation, and because of this, tamoxifen treatment of breast cancer patients results in a two- to sevenfold increased risk for development of endometrial cancer. Use of raloxifene, or the anti-estrogen ICI182780, does not result in such an increased risk. The objective of the current study was to generate and analyze gene expression profiles that reflect the transcriptional response of the human endometrium to estradiol, SERMs like tamoxifen and raloxifene, and anti-estrogens like ICI182780. METHODS: Transient transfections were performed to analyze the transcriptional response of ECC-1 cells to estradiol, tamoxifen, raloxifene, and ICI182780. Subsequently, to reveal the molecular mechanism of action, gene expression profiles were generated and some of the observed regulated genes were confirmed by Northern blotting. Biostatistical methods were employed to analyze the expression profile results further, and amphiregulin effects on ECC-1 cell signaling were investigated using Northern and Western blotting, and 3H-thymidine incorporation. RESULTS: Analysis of the profiles revealed that estradiol, tamoxifen, raloxifene, and ICI182780 influence the same biologic processes, but they do so via regulation of different sets of genes. Upon construction of a genetic network it was observed that the largest possible network centered on epidermal growth factor (EGF) receptor signaling. Furthermore, the EGF receptor ligand amphiregulin was differentially regulated by all four ligands. Next it was shown that amphiregulin indeed could stimulate EGF receptor signaling in ECC-1 cells. Based on these results, it was hypothesized that EGF receptor signaling could differentially be affected by estrogen, tamoxifen, raloxifene, and ICI182780 because these four compounds differentially regulate the EGF receptor ligand amphiregulin. CONCLUSIONS: Regulation of amphiregulin coincides with the described in vivo effect of the four ligands on the endometrium. Therefore, it is possible that modulation of EGF receptor signaling is a significant player in estrogen-agonistic growth of the endometrium and needs to be investigated further.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias do Endométrio/patologia , Estradiol/análogos & derivados , Antagonistas de Estrogênios/farmacologia , Glicoproteínas/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Cloridrato de Raloxifeno/farmacologia , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Tamoxifeno/farmacologia , Anfirregulina , Família de Proteínas EGF , Estradiol/farmacologia , Estradiol/fisiologia , Feminino , Fulvestranto , Perfilação da Expressão Gênica , Humanos , Transcrição Gênica , Células Tumorais Cultivadas
4.
J Soc Gynecol Investig ; 12(4): 278-84, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15866121

RESUMO

OBJECTIVE: Loss of expression of progesterone receptors (PR) in endometrial cancer is related to a more invasive and metastatic phenotype. In this study we aim to investigate whether selective loss of PRA or PRB affects the invasive capacity of endometrial cancer cells. METHODS: cDNA microarrays were performed to compare gene expression profiles of a set of endometrial cancer sub-cell lines expressing PRA and/or PRB. In vitro invasion assays were performed to assess whether differences in gene expression between the lines were reflected by their invasive behavior. RESULTS: It was observed that cell lines that express only PRA express higher levels of cadherins, and show a lower level of invasion compared to cell lines that express PRB. When cadherin function was inhibited in exclusively PRA-expressing cell lines, an increase of in vitro invasion was observed. In support of these findings, it was observed that in higher grade and more invasive endometrial cancer, expression of E-cadherin decreased. CONCLUSIONS: These results indicate that relative loss of PRA during progression of endometrial cancer can have a negative impact on cadherin expression, which may lead to development of a more metastatic phenotype.


Assuntos
Neoplasias do Endométrio/patologia , Invasividade Neoplásica/genética , Invasividade Neoplásica/fisiopatologia , Receptores de Progesterona/biossíntese , Western Blotting , Caderinas/biossíntese , Feminino , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Metástase Neoplásica , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Células Tumorais Cultivadas
5.
J Soc Gynecol Investig ; 12(4): 285-92, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15866122

RESUMO

OBJECTIVE: Progesterone plays an important role in controlling proliferation and differentiation of the human endometrium. Because there are two progesterone receptor isoforms (PRA and PRB), it was important to generate tools to be able to study the role of these two progesterone receptors separately. METHODS: Using stable transfection techniques, both human progesterone receptor isoforms (hPRA and hPRB) were reintroduced into a hPR-negative subclone of the well-differentiated endometrial cancer cell line Ishikawa. Several Ishikawa subcell lines were constructed, each expressing different levels of hPRA, hPRB, or hPRA and hPRB, respectively. RESULTS: These Ishikawa subcell lines showed a marked progesterone-induced growth inhibition with induction of apoptosis after long-term culture in the presence of hormone. Upon measuring gene regulation, a clear difference in regulation of expression of the selected genes by progesterone treatment was observed between the PRA-, PRB-, or PRA/B-expressing cell lines. Integrin beta4 (ITGB4) was only regulated in PRA-expressing cells; amphiregulin was highly regulated in PRB-expressing cells; insulin-like growth factor binding protein 3 (IGFBP3) was only regulated in PRB- and PRA/B-expressing cells; and metallothionein 1L (MT1L) was highly regulated in PRA/B-expressing cells. Interestingly, based on literature data, these genes can be implicated in induction of apoptosis, but are modulated here in such a way that suggests induction of resistance against apoptosis. CONCLUSION: Reintroduction of PRs into Ishikawa cells rescued progesterone responsiveness in these cells. Furthermore, using these human endometrial cancer subcell lines, clear and distinct functional differences between the PR isoforms were observed.


Assuntos
Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Progesterona/fisiologia , Receptores de Progesterona/biossíntese , Apoptose , Diferenciação Celular , Proliferação de Células , Feminino , Perfilação da Expressão Gênica , Humanos , Isoformas de Proteínas , Transfecção , Células Tumorais Cultivadas
6.
J Soc Gynecol Investig ; 12(1): 58-64, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15629674

RESUMO

OBJECTIVES: Estrogen-stimulated proliferation of the normal and malignant human endometrium is balanced by the differentiating properties of progesterone. This study evaluated the role of insulin-like growth factor (IGF) signaling in steroid-induced modulation of endometrial cancer cell proliferation. METHODS: We used the human endometrial, estrogen-responsive ECC-1 and progesterone-responsive PRAB-36 cell lines. Proliferation studies with IGFs in combination with either estrogen or progesterone were conducted. Furthermore, the mRNA and protein expression of insulin-like growth factor-binding proteins (IGFBPs) was evaluated. RESULTS: Using the ECC-1 cell line, we observed that estrogen-induced proliferation is modulated via the IGF-receptor signaling pathway, and that IGF-1-induced stimulation of proliferation does not influence estrogen receptor signaling. Furthermore, expression of the main modulators of IGF action, the IGFBPs, was found to be regulated by estrogen and progesterone in both cell lines. IGFBP-4 was up-regulated by estrogen in the ECC-1 cell line, and IGFBP-3 and IGFBP-6 were down-regulated by progesterone in the PRAB-36 cell line. CONCLUSION: Estrogen-induced stimulation of proliferation of ECC-1 endometrial cancer cells is partly achieved via IGF signaling. Furthermore, the IGFBPs are regulated by estrogens as well as progestagens and could potentially play a role in the modulation of endometrial cancer cell proliferation.


Assuntos
Carcinoma/patologia , Proliferação de Células , Neoplasias do Endométrio/patologia , Estrogênios/farmacologia , Somatomedinas/farmacologia , Feminino , Humanos , Proteína 4 de Ligação a Fator de Crescimento Semelhante à Insulina/biossíntese , Transdução de Sinais , Células Tumorais Cultivadas , Regulação para Cima
7.
Steroids ; 68(10-13): 795-800, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14667970

RESUMO

Progestagens inhibit growth of endometrial cancer cells in vivo and in vitro, and also are reported to inhibit endometrial cancer cell invasion. The progesterone receptor (PR) isotypes PRA and PRB have different transcriptional activity. There are indications that relative over expression of PRB could lead to development of a more invasive phenotype in endometrial cancer. To study the effect of progestagens and the two PR isotypes on tumor dissemination, in vitro and in vivo models should be applied. The Ishikawa endometrial cancer cell line (clone 3H12) was transfected to stably express a high level of human PRB (hPRB), which resulted in the PRB-1 sub-cell line. Ovariectomized athymic NMRI nu/nu mice were injected intraperitoneally with these PRB-1 cells. After 3, 5 and 10 weeks, the animals were sacrificed. Spread of PRB-1 cells in and outside the peritoneal cavity was studied macroscopically and microscopically, and also by PCR detection. After 10 weeks, the PRB-1 cells had formed extensive tumor mass in the peritoneal cavity. Also, cells could be detected outside the peritoneal cavity, indicating metastatic ability of these cells. The present study describes an in vivo model that can provide a valuable tool in studying the influence of progestagens and the two PR isotypes on endometrial cancer cell invasion and metastasis.


Assuntos
Neoplasias do Endométrio/patologia , Receptores de Progesterona/fisiologia , Animais , Western Blotting , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Invasividade Neoplásica , Metástase Neoplásica , Transplante de Neoplasias , Fenótipo , Reação em Cadeia da Polimerase , Receptores de Progesterona/metabolismo , Fatores de Tempo , Transfecção
8.
Clin Cancer Res ; 9(11): 4190-9, 2003 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-14519645

RESUMO

PURPOSE: In endometrial cancer, loss of progesterone receptors (PR) is associated with more advanced disease. This study aimed to investigate the mechanism of action of progesterone and the loss of its receptors (PRA and PRB) in development of endometrial cancer. EXPERIMENTAL DESIGN: A 9600-cDNA microarray analysis was performed to study regulation of gene expression in the human endometrial cancer subcell line Ishikawa PRAB-36 by the progestagen medroxy progesterone acetate (MPA). Five MPA-regulated genes were selected for additional investigation. Expression of these genes was studied by Northern blot and by immunohistochemistry in Ishikawa subcell lines expressing different PR isoforms. Additionally, endometrial cancer tissue samples were immunohistochemically stained to study the in vivo protein expression of the selected genes. RESULTS: In the PRAB-36 cell line, MPA was found to regulate the expression of a number of invasion- and metastasis-related genes. On additional investigation of five of these genes (CD44, CSPG/Versican, Tenascin-C, Fibronectin-1, and Integrin-beta 1), it was observed that expression and progesterone regulation of expression of these genes varied in subcell lines expressing different PR isoforms. Furthermore, in advanced endometrial cancer, it was shown that loss of expression of both PR and E-cadherin was associated with increased expression CD44 and CSPG/Versican. CONCLUSION: The present study shows that progestagens exert a modulatory effect on the expression of genes involved in tumor cell invasion. As a consequence, loss of PR expression in human endometrial cancer may lead to development of a more invasive phenotype of the respective tumor.


Assuntos
Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Regulação Neoplásica da Expressão Gênica/genética , Receptores de Progesterona/genética , Adenocarcinoma/genética , Adenocarcinoma/patologia , Linhagem Celular Tumoral , Proteoglicanas de Sulfatos de Condroitina/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Receptores de Hialuronatos/genética , Lectinas Tipo C , Acetato de Medroxiprogesterona/farmacologia , Invasividade Neoplásica/genética , Proteínas de Neoplasias/genética , Tenascina/genética , Versicanas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...