Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Gynecol Pathol ; 33(6): 598-606, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25272299

RESUMO

Anti-N-methyl-D-aspartate receptor (anti-NMDAR) encephalitis is the most common antibody-mediated limbic encephalitis and is associated with underlying ovarian teratoma. Previous studies suggest that expression of NMDAR on teratoma neural tissue initiates an autoimmune response to NMDAR in the brain. As some teratomas of patients with anti-NMDAR encephalitis lack neuronal tissue, we questioned if there could be an alternate mechanism of the disease. We performed immunohistochemical analyses for NMDAR and correlated its expression with histology on 10 control teratomas and 5 teratomas associated with anti-NMDAR encephalitis. Both control and case teratomas expressed NMDAR-bearing neural tissue. All 15 teratomas contained large amounts of NMDAR bearing squamous epithelium; in 2 cases this was the only tissue expressing NMDAR. NMDAR-bearing neural tissue is not the sole source of encephalitis in all patients. Furthermore, we speculate that NMDAR expression by squamous epithelium may contribute to the disease development in some patients.


Assuntos
Encefalite Antirreceptor de N-Metil-D-Aspartato/metabolismo , Células Epiteliais/metabolismo , Neoplasias Ovarianas/metabolismo , Receptores de N-Metil-D-Aspartato/biossíntese , Teratoma/metabolismo , Adolescente , Adulto , Encefalite Antirreceptor de N-Metil-D-Aspartato/etiologia , Feminino , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Neoplasias Ovarianas/complicações , Neoplasias Ovarianas/patologia , Receptores de N-Metil-D-Aspartato/análise , Teratoma/complicações , Teratoma/patologia , Adulto Jovem
2.
Am J Reprod Immunol ; 72(4): 392-402, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24898804

RESUMO

PROBLEM: The anti-inflammatory impact of three polyunsaturated fatty acids (3-PUFA) in endometriosis is incompletely understood. The effect of 3-PUFA on endometriosis-like lesions is evaluated as a potential anti-inflammatory treatment target. METHOD OF STUDY: Wild Type (WT) and transgenic Fat-1 mice (high levels of endogenous 3-PUFA) were utilized in a uterine tissue transplant endometriosis model. Experimental donor×host pairs included: WT×WT (WW), WT×Fat-1 (WF), and Fat-1×Fat-1 (FF). Cytokine content (IL-1ß, IL-2, IL-4, IL-6, IL-10, IL-12, IL-17A, IFN-γ, TNF-γ, MCP-1 and RANTES) and immunocellular composition in lesions was determined. RESULTS: Intralesion IL-6 in WF hosts was 99-fold lower than WW hosts (P=0.03). Compared to WW host lesions, Cox-2 levels were decreased in WF [1.5-fold (P=0.02)] and FF [1.2-fold (P=0.01)] host lesions, respectively, and intralesion VEGF expression was increased [1.8-fold; P=0.02 (WF) and 1.5-fold; P=0.01 (FF)]. Lesions in FF hosts demonstrated reduced phosphohistone 3 expression (70%; P=0.03) compared to WW control hosts. CONCLUSIONS: Systemic host 3-PUFA levels influence immune, angiogenic, and proliferative factors implicated in the early establishment of endometriosis.


Assuntos
Anti-Inflamatórios/farmacologia , Citocinas/biossíntese , Endometriose/dietoterapia , Endometriose/tratamento farmacológico , Ácidos Graxos Ômega-3/farmacologia , Animais , Linfócitos B/imunologia , Ciclo-Oxigenase 2/biossíntese , Citocinas/genética , Dieta , Feminino , Inflamação/dietoterapia , Inflamação/tratamento farmacológico , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosfoproteínas/análise , Linfócitos T/imunologia , Útero/transplante , Fator A de Crescimento do Endotélio Vascular/biossíntese
3.
Int J Cancer ; 134(9): 2041-50, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24519534

RESUMO

The KRAS oncogene influences angiogenesis, metastasis and chemoresistance in colorectal cancers (CRCs), and these processes are all enhanced in hypoxic conditions. To define functional activities of mutant KRAS in a hypoxic microenvironment, we first performed cDNA microarray experiments in isogenic DKs5 and DKO3 colon cancer cell lines that differ only by their expression of mutant KRAS (K-ras(D13)). Adrenomedullin (ADM) was identified as one of the most significantly upregulated genes in DKs5 cells that express the KRAS oncogene in hypoxia (3.2-fold, p = 1.47 × 10(-5)). Ectopic expression of mutant KRAS (K-ras(V12)) in Caco-2 cells (K-ras(WT)) induced ADM, whereas selective knockdown of mutant KRAS alleles (K-ras(D13) or K-ras(V12)) in HCT116, DLD1 and SW480 colon cancer cells suppressed the expression of ADM in hypoxia. Knockdown of ADM in colon tumor xenografts blocked angiogenesis and stimulated apoptosis, resulting in tumor suppression. Furthermore, ADM also regulated colon cancer cell invasion in vitro. Among 56 patients with CRC, significantly higher expression levels of ADM were observed in samples harboring a KRAS mutation. Collectively, ADM is a new target of oncogenic KRAS in the setting of hypoxia. This observation suggests that therapeutic targets may differ depending upon the specific tumor microenvironment.


Assuntos
Adrenomedulina/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas ras/genética , Animais , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Feminino , Humanos , Imuno-Histoquímica , Camundongos Nus , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas p21(ras) , Microambiente Tumoral/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
4.
J Ovarian Res ; 4(1): 8, 2011 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-21605417

RESUMO

BACKGROUND: Metformin, an oral biguanide traditionally used for the treatment of type 2 diabetes, is widely used for the management of polycystic ovary syndrome (PCOS)-related anovulation. Because of the significant prevalence of insulin resistance and glucose intolerance in PCOS patients, and their putative role in ovulatory dysfunction, the use of metformin was touted as a means to improve ovulatory function and reproductive outcomes in PCOS patients. To date, there has been inconsistent evidence to demonstrate a favorable effect of metformin on oocyte quality and competence in women with PCOS. Given the heterogeneous nature of this disorder, we hypothesized that metformin may be beneficial in mice with aberrant metabolic characteristics similar to a significant number of PCOS patients. The aim of this study was to gain insight into the in vitro and in vivo effects of metformin on oocyte development and ovulatory function. METHODS: We utilized metformin treatment in the transgenic ob/ob and db/db mutant murine models which demonstrate metabolic and reproductive characteristics similar to women with PCOS. RESULTS: Metformin did not improve in vitro oocyte maturation nor did it have an appreciable effect on in vitro granulosa cell luteinization (progesterone production) in any genotype studied. Although both mutant strains have evidence of hyperandrogenemia, anovulation, and hyperinsulinemia, only db/db mice treated with metformin had a greater number of mature oocytes and total overall oocytes compared to control. There was no observed impact on body mass, or serum glucose and androgens in any genotype. CONCLUSIONS: Our data provide evidence to suggest that metformin may optimize ovulatory performance in mice with a specific reproductive and metabolic phenotype shared by women with PCOS. The only obvious difference between the mutant murine models is that the db/db mice have elevated leptin levels raising the questions of whether their response to metformin is related to elevated leptin levels and/or if a subset of PCOS women with hyperleptinemia may be responsive to metformin therapy. Further study is needed to better define a subset of women with PCOS that may be responsive to metformin.

5.
Int J Gynecol Pathol ; 28(2): 127-33, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19188823

RESUMO

MUC4 is a transmembrane glycoprotein more highly expressed in cervical dysplasia than benign cervical epithelium. We sought to determine whether MUC4 expression differs between benign and malignant cervical tissue. Fifty-eight patients with benign, dysplastic, or malignant cervical pathology were identified retrospectively, and representative sections were stained with a mouse monoclonal anti-MUC4 antibody. Semiquantitative analysis was performed on benign, dysplastic, and malignant regions by scoring staining intensity (0: negative, 1: weak, 2: moderate, and 3: strong) and distribution (focal <10%, multifocal=10%-60%, diffuse > or =60%). In samples with benign glycogenated squamous epithelium, only the parabasal cells had MUC4 staining, and 48.5% had an intensity of 2 or 3. All samples with immature squamous metaplasia were positive through the entire epithelial thickness. Cervical intraepithelial neoplasia (CIN) 1 samples had variable staining with an intensity similar to glycogenated squamous epithelium but distribution similar to squamous metaplasia. All CIN 3 (n=21) and invasive squamous cell carcinomas (n=17) had increased MUC4 staining intensity (P<0.001 and P<0.001) and increased diffuse staining (P<0.001 and P<0.001) compared with the limited staining in glycogenated squamous epithelium. In contrast, no differences in staining were observed between benign endocervical glands, adenocarcinoma in situ, and invasive adenocarcinoma. These expression patterns suggest that MUC4 is a lineage marker in benign cervical tissue that may have aberrant expression in squamous dysplasia and carcinoma. Further studies may elucidate the role of MUC4 in the development of squamous cell cervical cancer.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma de Células Escamosas/patologia , Mucina-4/biossíntese , Neoplasias do Colo do Útero/patologia , Carcinoma de Células Escamosas/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Regulação para Cima , Displasia do Colo do Útero/metabolismo , Displasia do Colo do Útero/patologia , Neoplasias do Colo do Útero/metabolismo
6.
Int J Cancer ; 124(4): 763-71, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19030186

RESUMO

Hypoxia-inducible factor (HIF)-1 and HIF-2 are heterodimeric transcription factors that mediate the cellular response to hypoxia. Their key regulatory subunits, HIF-1alpha and HIF-2alpha, are induced similarly by hypoxia, but their functional roles in cancer may be distinct and isoform-specific. SW480 colon cancer cells with stable expression of siRNA to HIF-1alpha or HIF-2alpha or both were established. HIF-1alpha-deficient cells displayed lower rates of proliferation and migration, but HIF-2alpha-deficient cells exhibited enhanced anchorage independent growth in a soft agar assay. Xenograft studies revealed that HIF-1alpha deficiency inhibited overall tumor growth, whereas deficiency of HIF-2alpha stimulated tumor growth. In human colon cancer tissues, expression of HIF-1alpha and to a lesser extent, HIF-2alpha, was linked to upregulation of VEGF and tumor angiogenesis. However, loss of expression of HIF-2alpha but not HIF-1alpha was strongly correlated with advanced tumor stage. DNA microarray analysis identified distinct sets of HIF-1alpha and HIF-2alpha target genes that may explain these phenotypic differences. Collectively, these findings suggest that HIF isoforms may have differing cellular functions in colon cancer. In particular, HIF-1alpha promoted the growth of SW480 colon cancer cells but HIF-2alpha appeared to restrain growth. Consequently, therapeutic approaches that target HIF may need to consider these isoform-specific properties.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Neoplasias do Colo/metabolismo , Regulação Neoplásica da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Neovascularização Patológica , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias
7.
Int J Cancer ; 123(12): 2782-90, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18798554

RESUMO

Several proangiogenic/proinflammatory factors involved in endometrial cancer are regulated by leptin, but the signaling mechanisms responsible for these leptin-induced actions are largely unknown. Here, we report that in benign (primary and HES) and cancerous-endometrial epithelial cells (EEC) (An3Ca, SK-UT2 and Ishikawa), leptin in a dose-dependent manner regulates vascular endothelial growth factor, (VEGF); interleukin-1 beta, (IL-1beta); leukemia inhibitory factor, (LIF) and their respective receptors, VEGFR2, IL-1R tI and LIFR. Remarkably, leptin induces a greater increase in VEGF/VEGFR2 and LIF levels in cancer than in benign cells. However, IL-1beta was only increased by leptin in benign primary-EEC. Cancer-EEC expressed higher levels of leptin receptor (full-length OB-Rb and short isoforms) in contrast to benign primary-EEC. Leptin-mediated activation of JAK2 (janus kinase 2) was upstream to the activation of PI-3K (phosphatidylinositol-3 kinase) and/or MAPK (mitogen-activated protein kinase) signaling pathways. Leptin induction of cytokines/receptors generally involved JAK2 and MAPK activation, but PI-3K phosphorylation was required for leptin increase of LIF, IL-1/IL-1R tI. Leptin-mediated activation of mTOR (mammalian target of Rapamycin), mainly linked to MAPK, played a central role in leptin regulation of all cytokines and receptors. These results suggest that leptin's effects are cell-specific and could confer a proliferative or cell survival advantage or possibly promote endometrial thickness. Leptin's effects on proangiogenic molecules were more evident in malignant versus benign cells and may imply that there is an underlying shift in leptin-induced cell signaling pathways in endometrial cancer cells.


Assuntos
Adenocarcinoma/metabolismo , Neoplasias do Endométrio/metabolismo , Endométrio/metabolismo , Leptina/metabolismo , Neovascularização Patológica/metabolismo , Proteínas Quinases/metabolismo , Adenocarcinoma/irrigação sanguínea , Western Blotting , Neoplasias do Endométrio/irrigação sanguínea , Endométrio/irrigação sanguínea , Feminino , Humanos , Interleucina-1beta/metabolismo , Fator Inibidor de Leucemia/metabolismo , Receptores de Interleucina-1/metabolismo , Receptores de OSM-LIF/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
8.
Proc Natl Acad Sci U S A ; 105(22): 7670-5, 2008 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-18505843

RESUMO

TNF is well known for its role in inflammation, including direct effects on the vasculature. TNF also is implicated in the regulation of reproduction by its actions to affect ovarian steroidogenic cells and to induce apoptosis of corpus luteum (CL)-derived endothelial cells in vitro. We hypothesized that the disruption of TNF signaling would postpone the regression of the highly vascularized CL in vivo, and this effect could be replicated in mutant mouse models lacking TNF receptor (TNFRI(-/-)) and/or a critical enzyme of TNF signaling, acid sphingomyelinase (ASMase(-/-)). In the current study, the treatment of pseudopregnant mice with the luteolytic mediator prostaglandin F2-alpha (PGF) significantly increased TNF in the ovaries when compared with saline-treated controls. Treatment with PGF also reduced serum progesterone (P4) concentrations and caused involution of the CL. However, pretreatment of pseudopregnant mice with Etanercept (ETA), a TNF-neutralizing antibody, inhibited the PGF-induced decrease in P4 and delayed luteal regression. A similar outcome was evident in pseudopregnant TNFRI(-/-) animals. Treatment of luteal microvascular endothelial cells (MVECs) with TNF provoked a significant increase in ASMase activity when compared with the corresponding controls. Furthermore, TNF-induced MVEC death was inhibited in the ASMase(-/-) mice. The ASMase(-/-) mice displayed no obvious evidence of luteal regression 24 h after treatment with PGF and were resistant to the PGF-induced decrease in P4. Together these data provide evidence that TNF plays an active role in luteolysis. Further studies are required to determine the deleterious effects of anti-inflammatory agents on basic ovarian processes.


Assuntos
Corpo Lúteo/fisiologia , Luteólise/metabolismo , Progesterona/metabolismo , Esfingomielina Fosfodiesterase/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Apoptose , Capilares/citologia , Capilares/efeitos dos fármacos , Capilares/metabolismo , Ceramidas/metabolismo , Corpo Lúteo/citologia , Corpo Lúteo/metabolismo , Dinoprosta/farmacologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Etanercepte , Feminino , Imunoglobulina G/farmacologia , Luteólise/efeitos dos fármacos , Luteólise/genética , Camundongos , Camundongos Knockout , Progesterona/sangue , Receptores do Fator de Necrose Tumoral , Receptores Tipo I de Fatores de Necrose Tumoral/antagonistas & inibidores , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Esfingomielina Fosfodiesterase/genética , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/farmacologia
9.
Cancer Biol Ther ; 7(2): 180-88, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18059193

RESUMO

Cables 1, a cyclin-dependent kinase binding protein, is primarily involved in cell cycle regulation. Loss of nuclear Cables 1 expression is observed in human colon, lung and endometrial cancers. We previously reported that loss of nuclear Cables 1 expression was also observed with high frequency in a limited sample set of human ovarian carcinomas, although the mechanisms underlying loss of nuclear Cables 1 expression remained unknown. Our present objective was to examine Cables 1 expression in ovarian cancer in greater detail, and determine the predominant mechanisms of Cables 1 loss. We assessed potential genetic and epigenetic modifications of the Cables 1 locus through analyses of mutation, polymorphisms, loss of heterozygosity and DNA methylation. We observed a marked loss of nuclear Cables 1 expression in serous and endometrioid ovarian carcinomas that correlated with decreased Cables 1 mRNA levels. Although we detected no Cables 1 mutations, there was evidence of LOH at the Cables 1 locus and epigenetic modification of the Cables 1 promoter region in a subset of ovarian carcinomas and established cancer cell lines. From a functional perspective, over-expression of Cables 1 induced apoptosis, whereas, knockdown of Cables 1 negated this effect. Together these findings suggest that multiple mechanisms underlie the loss of Cables 1 expression in ovarian cancer cells, supporting the hypothesis that Cables 1 is a tumor suppressor in human ovarian cancer.


Assuntos
Proteínas de Transporte/genética , Ciclinas/genética , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Perda de Heterozigosidade , Neoplasias Ovarianas/genética , Fosfoproteínas/genética , Estudos de Casos e Controles , Linhagem Celular Tumoral , Proliferação de Células , Cromossomos Humanos Par 18 , Metilação de DNA , Epigênese Genética , Feminino , Humanos , Imuno-Histoquímica , Neoplasias Ovarianas/classificação , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Regiões Promotoras Genéticas , Análise de Sequência de DNA , Proteínas Supressoras de Tumor
10.
J Biol Chem ; 281(36): 26320-8, 2006 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-16825198

RESUMO

To gain insight into the mechanism(s) by which leptin contributes to mammary tumor (MT) development we investigated the effects of leptin, kinase inhibitors, and/or leptin receptor antagonists (LPrA2) on 4T1 mouse mammary cancer cells in vitro and LPrA2 on 4T1-MT development in vivo. Leptin increases the expression of vascular endothelial growth factor (VEGF), its receptor (VEGF-R2), and cyclin D1 through phosphoinositide 3-kinase, Janus kinase 2/signal transducer and activator of transcription 3, and/or extracellular signal-activated kinase 1/2 signaling pathways. In contrast to leptin-induced levels of cyclin D1 the changes in VEGF or VEGF-R2 were more dependent on specific signaling pathways. Incubation of 4T1 cells with anti-VEGF-R2 antibody increased leptin-mediated VEGF expression suggesting an autocrine/paracrine loop. Pretreatment of syngeneic mice with LPrA2 prior to inoculation with 4T1 cells delayed the development and slowed the growth of MT (up to 90%) compared with controls. Serum VEGF levels and VEGF/VEGF-R2 expression in MT were significantly lower in mice treated with LPrA2. Interestingly, LPrA2-induced effects were more pronounced in vivo than in vitro suggesting paracrine actions in stromal, endothelial, and/or inflammatory cells that may impact the growth of MT. Although all the mechanism(s) by which leptin contributes to tumor development are unknown, it appears leptin stimulates an increase in cell numbers, and the expression of VEGF/VEGF-R2. Together, these results provide further evidence suggesting leptin is a MT growth-promoting factor. The inhibition of leptin signaling could serve as a potential adjuvant therapy for treatment of breast cancer and/or provide a new target for the designing strategies to prevent MT development.


Assuntos
Leptina/metabolismo , Neoplasias Mamárias Animais/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Linhagem Celular Tumoral , Ciclina D1/metabolismo , Feminino , Humanos , Leptina/genética , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos/genética , Peptídeos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/genética , Receptores para Leptina , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
11.
Nat Med ; 11(9): 992-7, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16127434

RESUMO

Hypoxia inducible factor-1 (HIF-1) is considered a crucial mediator of the cellular response to hypoxia through its regulation of genes that control angiogenesis. It represents an attractive therapeutic target in colon cancer, one of the few tumor types that shows a clinical response to antiangiogenic therapy. But it is unclear whether inhibition of HIF-1 alone is sufficient to block tumor angiogenesis. In HIF-1alpha knockdown DLD-1 colon cancer cells (DLD-1(HIF-kd)), the hypoxic induction of vascular endothelial growth factor (VEGF) was only partially blocked. Xenografts remained highly vascularized with microvessel densities identical to DLD-1 tumors that had wild-type HIF-1alpha (DLD-1(HIF-wt)). In addition to the preserved expression of VEGF, the proangiogenic cytokine interleukin (IL)-8 was induced by hypoxia in DLD-1(HIF-kd) but not DLD-1(HIF-wt) cells. This induction was mediated by the production of hydrogen peroxide and subsequent activation of NF-kappaB. Furthermore, the KRAS oncogene, which is commonly mutated in colon cancer, enhanced the hypoxic induction of IL-8. A neutralizing antibody to IL-8 substantially inhibited angiogenesis and tumor growth in DLD-1(HIF-kd) but not DLD-1(HIF-wt) xenografts, verifying the functional significance of this IL-8 response. Thus, compensatory pathways can be activated to preserve the tumor angiogenic response, and strategies that inhibit HIF-1alpha may be most effective when IL-8 is simultaneously targeted.


Assuntos
Neoplasias do Colo/irrigação sanguínea , Interleucina-8/fisiologia , Neovascularização Patológica/fisiopatologia , Fatores de Transcrição/fisiologia , Animais , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Peróxido de Hidrogênio/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia , Interleucina-8/biossíntese , Camundongos , Camundongos Nus , Oxigênio/fisiologia , Fatores de Transcrição/genética , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/biossíntese
12.
Cancer Biol Ther ; 4(1): 103-7, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15662117

RESUMO

Loss of Cables expression is associated with a high incidence of endometrial hyperplasia and endometrial adenocarcinoma in humans. The Cables mutant mouse develops endometrial hyperplasia and following exposure to chronic estrogen develops early endometrial adenocarcinoma. The objectives of the current study were to determine if: (1) loss of Cables expression occurred in high grade endometrioid adenocarcinoma, uterine serous and clear cell carcinoma as observed in endometrial hyperplasia and low grade endometrial adenocarcinoma; (2) overexpression of Cables inhibited cell proliferation in endometrial cancer (EC) cells in vitro and in vivo; and (3) progesterone could regulate the expression of Cables mRNA. Hyperplastic endometrium and low and high grade endometrioid adenocarcinoma showed loss of Cables expression when compared to benign control secretory endometrium. Loss of Cables expression in serous and clear cell tumors was similar to that observed in endometrioid adenocarcinomas with greater than 80% showing loss of protein expression. Treatment of EC lines with progesterone increased cables expression in low-grade EC whereas it had no effect on cables expression in cells derived from high-grade EC. The progesterone-induced increase in cables was abrogated in the presence of a progesterone receptor (PR) antagonist, suggesting the PR mediates the increase. Cables overexpression inhibited cell proliferation of well differentiated EC cells and had no effect on the poorly differentiated EC cells. The capacity to form tumors was dramatically reduced in the Cables overexpressing cell lines compared to those cells containing the control vector. Collectively these results suggest that Cables is an important regulator of cell proliferation and loss of Cables expression contributes to the development of all types of EC.


Assuntos
Adenocarcinoma de Células Claras/genética , Adenocarcinoma de Células Claras/patologia , Adenocarcinoma/genética , Adenocarcinoma/patologia , Proliferação de Células , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Perfilação da Expressão Gênica , Animais , Proteínas de Transporte , Ciclinas , Feminino , Humanos , Camundongos , Fosfoproteínas , Receptores de Progesterona/fisiologia , Células Tumorais Cultivadas
13.
Cancer Res ; 64(1): 202-8, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14729625

RESUMO

Endometrial cancer is the most common gynecological cancer in Western industrialized countries. Cables, a cyclin-dependent kinase binding protein, plays a role in proliferation and/or differentiation. Cables mutant mice are viable, but develop endometrial hyperplasia and carcinoma in situ at a young age. Exposure to chronic low levels of estrogen results in development of endometrial cancer, similar to that observed in the postmenopausal female. In vitro and in vivo studies demonstrate that levels of Cables mRNA in benign human endometrial epithelium are up-regulated by progesterone and down-regulated by estrogen. Furthermore, nuclear immunostaining for Cables is lost in a high percentage of cases of human endometrial hyperplasia and adenocarcinoma, which are likely the product of unopposed estrogen. The loss of Cables immunostaining in the human endometrial cancer samples correlates with a marked decrease in Cables mRNA. Ectopic expression of Cables in human endometrial cells dramatically slows cell proliferation. Collectively, these data provide evidence that Cables is hormonally regulated and is involved in regulating endometrial cell proliferation. In addition, loss or suppression of Cables may be an early step in the development of endometrial cancer.


Assuntos
Proteínas de Transporte/genética , Ciclinas , Neoplasias do Endométrio/genética , Endométrio/patologia , Regulação Neoplásica da Expressão Gênica , Perda de Heterozigosidade , Fosfoproteínas/genética , Animais , Clonagem Molecular , Neoplasias do Endométrio/patologia , Feminino , Mutação em Linhagem Germinativa , Humanos , Hiperplasia , Camundongos , RNA Mensageiro/genética , Mapeamento por Restrição , Células-Tronco/fisiologia , Transcrição Gênica
14.
Cancer Biol Ther ; 2(4): 392-5, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14508111

RESUMO

We have shown that CD95-mediated cell death requires a clustering of the receptor in distinct sphingolipid-rich domains of the cell membrane (Grassme et al., 2000, Cremesti et al., 2000). These domains form in response to acid sphingomyelinase (ASM)-induced ceramide generation. However, recent studies challenged the finding of early CD95 clustering (Algeciras-Schimnich et al., 2002). Here, six independent groups tested clustering of CD95 in diverse cell type including primary cells ex vivo and established cell lines. The studies show clustering of CD95 within seconds to minutes in all cell types tested by the different groups. In addition, clustering of CD95 was detected after stimulation of cells using three agonistic anti-CD95 antibodies (CH11, APO-1-3 and JO2), CD95 ligand and stimuli that induce an upregulation and activation of the endogenous CD95/CD95 ligand system. The data confirm our previous studies and suggest rapid, i.e., within seconds to minutes, CD95 clustering as a general phenomenon occurring in many cell types.


Assuntos
Apoptose , Linfócitos/metabolismo , Glicoproteínas de Membrana/metabolismo , Baço/metabolismo , Receptor fas/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Células Cultivadas , Proteína Ligante Fas , Humanos , Ligantes , Camundongos , Regulação para Cima , Receptor fas/imunologia
15.
Reprod Biol Endocrinol ; 1: 15, 2003 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-12657159

RESUMO

We recently demonstrated that caspase-3 is important for apoptosis during spontaneous involution of the corpus luteum (CL). These studies tested if prostaglandin F2alpha (PGF2alpha) or FAS regulated luteal regression, utilize a caspase-3 dependent pathway to execute luteal cell apoptosis, and if the two receptors work via independent or potentially shared intracellular signaling components/pathways to activate caspase-3. Wild-type (WT) or caspase-3 deficient female mice, 25-26 days old, were given 10 IU equine chorionic gonadotropin (eCG) intraperitoneally (IP) followed by 10 IU human chorionic gonadotropin (hCG) IP 46 h later to synchronize ovulation. The animals were then injected with IgG (2 micrograms, i.v.), the FAS-activating antibody Jo2 (2 micrograms, i.v.), or PGF2alpha (10 micrograms, i.p.) at 24 or 48 h post-ovulation. Ovaries from each group were collected 8 h later for assessment of active caspase-3 enzyme and apoptosis (measured by the TUNEL assay) in the CL. Regardless of genotype or treatment, CL in ovaries collected from mice injected 24 h after ovulation showed no evidence of active caspase-3 or apoptosis. However, PGF2alpha or Jo2 at 48 h post-ovulation and collected 8 h later induced caspase-3 activation in 13.2 +/- 1.8% and 13.7 +/- 2.2 % of the cells, respectively and resulted in 16.35 +/- 0.7% (PGF2alpha) and 14.3 PlusMinus; 2.5% TUNEL-positive cells when compared to 1.48 +/- 0.8% of cells CL in IgG treated controls. In contrast, CL in ovaries collected from caspase-3 deficient mice whether treated with PGF2alpha, Jo2, or control IgG at 48 h post-ovulation showed little evidence of active caspase-3 or apoptosis. CL of WT mice treated with Jo2 at 48 h post-ovulation had an 8-fold increase in the activity of caspase-8, an activator of caspase-3 that is coupled to the FAS death receptor. Somewhat unexpectedly, however, treatment of WT mice with PGF2alpha at 48 h post-ovulation resulted in a 22-fold increase in caspase-8 activity in the CL, despite the fact that the receptor for PGF2alpha has not been shown to be directly coupled to caspase-8 recruitment and activation. We hypothesize that PGF2alpha initiates luteolysis in vivo, at least in part, by increasing the bioactivity or bioavailability of cytokines, such as FasL and that multiple endocrine factors work in concert to activate caspase-3-driven apoptosis during luteolysis.


Assuntos
Apoptose/efeitos dos fármacos , Caspases/fisiologia , Corpo Lúteo/efeitos dos fármacos , Dinoprosta/farmacologia , Luteólise/efeitos dos fármacos , Receptor fas/farmacologia , Animais , Animais Congênicos , Apoptose/fisiologia , Caspase 3 , Caspase 8 , Caspases/deficiência , Caspases/genética , Corpo Lúteo/citologia , Corpo Lúteo/enzimologia , Dinoprosta/antagonistas & inibidores , Dinoprosta/imunologia , Ativação Enzimática/efeitos dos fármacos , Feminino , Luteólise/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Progesterona/sangue , Transdução de Sinais/efeitos dos fármacos , Receptor fas/imunologia
16.
Reprod Biol Endocrinol ; 1: 17, 2003 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-12646059

RESUMO

The microvasculature of the corpus luteum (CL), which comprises greater than 50% of the total number of cells in the CL, is thought to be the first structure to undergo degeneration via apoptosis during luteolysis. These studies compared the apoptotic potential of various cytokines (tumor necrosis factor alpha, TNFalpha; interferon gamma, IFNgamma; soluble Fas ligand, sFasL), a FAS activating antibody (FasAb), and the luteolytic hormone prostaglandin F2alpha (PGF2alpha) on CL-derived endothelial (CLENDO) cells. Neither sFasL, FasAb nor PGF2alpha had any effect on CLENDO cell viability. Utilizing morphological and biochemical parameters it was evident that TNFalpha and IFNgamma initiated apoptosis in long-term cultures. However, TNFalpha was the most potent stimulus for CLENDO cell apoptosis at early time points. Unlike many other studies described in non-reproductive cell types, TNFalpha induced apoptosis of CLENDO cells occurs in the absence of inhibitors of protein synthesis. TNFalpha-induced death is typically associated with acute activation of distinct intracellular signaling pathways (e.g. MAPK and sphingomyelin pathways). Treatment with TNFalpha for 5-30 min activated MAPKs (ERK, p38, and JNK), and increased ceramide accumulation. Ceramide, a product of sphingomyelin hydrolysis, can serve as an upstream activator of members of the MAPK family independently in numerous cell types, and is a well-established pro-apoptotic second messenger. Like TNFalpha, treatment of CLENDO cells with exogenous ceramide significantly induced endothelial apoptosis. Ceramide also activated the JNK pathway, but had no effect on ERK and p38 MAPKs. Pretreatment of CLENDO cells with glutathione (GSH), an intracellular reducing agent and known inhibitor of reactive oxygen species (ROS) or TNFalpha-induced apoptosis, significantly attenuated TNFalpha-induced apoptosis. It is hypothesized that TNFalpha kills CLENDO cells through elevation of reactive oxygen species, and intracellular signals that promote apoptosis.


Assuntos
Corpo Lúteo/irrigação sanguínea , Endotélio Vascular/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Animais , Apoptose/efeitos dos fármacos , Capilares/citologia , Bovinos , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Ceramidas/biossíntese , Ceramidas/farmacologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/citologia , Feminino , Glutationa/farmacologia , Humanos , Interferon gama/farmacologia , Luteólise , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Oxirredução , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Receptor fas/imunologia , Receptor fas/fisiologia
17.
Endocrinology ; 143(4): 1495-501, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11897708

RESUMO

Because caspase-3 is considered a primary executioner of apoptosis and has been implicated as a mediator of luteal regression, we hypothesized that corpora lutea (CL) derived from caspase-3 null mice would exhibit a delayed onset of apoptosis during luteal regression, when compared with CL derived from wild-type (WT) mice. To test this hypothesis, ovulation was synchronized in immature (postpartum d 24-27) WT and caspase-3-deficient female littermates by exogenous gonadotropins. Individual CL were isolated by manual dissection, 30 h after ovulation, and placed in organ culture dishes in the absence of serum and growth factors. At the time of isolation (0 h) and after 24, 48, and 72 h in culture, the CL were removed and assessed for the presence of processed (active) caspase-3 enzyme and for apoptosis by multiple criteria. There was no evidence of active caspase-3 enzyme or apoptosis in either WT or caspase-3-deficient CL before culture. However, CL derived from the WT mice exhibited a time-dependent increase in the level of active caspase-3 and apoptosis during culture. By comparison, CL derived from caspase-3-deficient mice, cultured in parallel, failed to exhibit any detectable active caspase-3 and showed attenuated rates of apoptosis. To extend these findings derived from ex vivo culture experiments, ovaries were collected from WT and caspase-3 null female littermates at 2, 4, or 6 d post ovulation, and the occurrence of apoptosis within the CL was analyzed. Whereas ovaries of WT mice had only residual luteal tissue at d 6 post ovulation, ovaries collected from caspase-3-deficient mice retained many CL, at d 6 post ovulation, that were similar in size to those observed in the early luteal phase of WT mice. Importantly, there was no dramatic increase in apoptosis in CL of caspase-3-deficient mice at any time point examined post ovulation, indicating that the involution process had indeed been delayed. In contrast, the levels of progesterone declined regardless of genotype. These data provide the first direct evidence that caspase-3 is functionally required for apoptosis to proceed normally during luteal regression. However, caspase-3 is not a direct mediator of the decrease in steroidogenesis associated with luteolysis.


Assuntos
Apoptose/fisiologia , Caspases/fisiologia , Corpo Lúteo/fisiologia , Ovário/fisiologia , Animais , Caspase 3 , Caspases/genética , Corpo Lúteo/citologia , Corpo Lúteo/enzimologia , DNA/biossíntese , DNA/genética , DNA/isolamento & purificação , Fragmentação do DNA/fisiologia , Eletroforese em Gel de Poliacrilamida , Feminino , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Knockout , Técnicas de Cultura de Órgãos , Ovário/citologia , Ovário/enzimologia , Ovulação/fisiologia , Progesterona/metabolismo , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...