Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cancer Sci ; 114(6): 2485-2498, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36854451

RESUMEN

Cancer tissues generally have molecular oxygen and serum component deficiencies because of poor vascularization. Recently, we revealed that ICAM1 is strongly activated through lipophagy in ovarian clear cell carcinoma (CCC) cells in response to starvation of long-chain fatty acids and oxygen and confers resistance to apoptosis caused by these harsh conditions. CD69 is a glycoprotein that is synthesized in immune cells and is associated with their activation through cellular signaling pathways. However, the expression and function of CD69 in nonhematological cells is unclear. Here, we report that CD69 is induced in CCC cells as in ICAM1. Mass spectrometry analysis of phosphorylated peptides followed by pathway analysis revealed that CD69 augments CCC cell binding to fibronectin (FN) in association with the phosphorylation of multiple cellular signaling molecules including the focal adhesion pathway. Furthermore, CD69 synthesized in CCC cells could facilitate cell survival because the CD69-FN axis can induce epithelial-mesenchymal transition. Experiments with surgically removed tumor samples revealed that CD69 is predominantly expressed in CCC tumor cells compared with other histological subtypes of epithelial ovarian cancer. Overall, our data suggest that cancer cell-derived CD69 can contribute to CCC progression through FN.


Asunto(s)
Fibronectinas , Neoplasias Ováricas , Humanos , Femenino , Oxígeno , Neoplasias Ováricas/patología , Transducción de Señal , Lípidos , Línea Celular Tumoral
2.
Cancer Sci ; 111(12): 4381-4392, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32979884

RESUMEN

Interaction of cancer cells with cancer-associated fibroblasts (CAFs) plays critical roles in tumor progression. Recently we proposed a new tumor invasion mechanism in which invasive cancer cells individually migrate on elongate protrusions of CAFs (CAF fibers) in 3-D collagen matrix. In this mechanism, cancer cells interact with fibronectin fibrils assembled on CAFs mainly through integrin-α5ß1. Here we tested whether this mechanism is applicable to the collective invasion of cancer cells, using two E-cadherin-expressing adenocarcinoma cell lines, DLD-1 (colon) and MCF-7 (breast). When hybrid spheroids of DLD-1 cells with CAFs were embedded into collagen gel, DLD-1 cells collectively but very slowly migrated through the collagen matrix in contact with CAFs. Epidermal growth factor and tumor necrosis factor-α promoted the collective invasion, possibly by reducing the E-cadherin junction, as did the transforming growth factor-ß inhibitor SB431542 by stimulating the outgrowth of CAFs. Transforming growth factor-ß itself inhibited the cancer cell invasion. Efficient collective invasion of DLD-1 cells required large CAF fibers or their assembly as stable adhesion substrates. Experiments with function-blocking Abs and siRNAs confirmed that DLD-1 cells adhered to fibronectin fibrils on CAFs mainly through integrin-α5ß1. Anti-E-cadherin Ab promoted the single cell invasion of DLD-1 cells by dissociating the E-cadherin junction. Although the binding affinity of MCF-7 cells to CAFs was lower than DLD-1, they also collectively invaded the collagen matrix in a similar fashion to DLD-1 cells. Our results suggest that the direct interaction with CAFs, as well as environmental cytokines, contributes to the collective invasion of cancers.


Asunto(s)
Fibroblastos Asociados al Cáncer/patología , Colágeno/metabolismo , Fibroblastos/patología , Fibronectinas/metabolismo , Integrina alfa5beta1/metabolismo , Invasividad Neoplásica/patología , Células A549 , Adenocarcinoma/patología , Amidas/farmacología , Benzamidas/farmacología , Fibroblastos Asociados al Cáncer/metabolismo , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Cromonas/farmacología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Tejido Conectivo/patología , Dioxoles/farmacología , Factor de Crecimiento Epidérmico/metabolismo , Fibroblastos/metabolismo , Humanos , Inmunohistoquímica/métodos , Células MCF-7 , Morfolinas/farmacología , Invasividad Neoplásica/fisiopatología , Piridinas/farmacología , Esferoides Celulares/patología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
3.
Biol Reprod ; 100(5): 1215-1227, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30649202

RESUMEN

A decellularized uterine scaffold (DUS) prepared from rats permits recellularization and regeneration of uterine tissues when placed onto a partially excised uterus and supports pregnancy in a fashion comparable to the intact uterus. The underlying extracellular matrix (ECM) together with an acellular, perfusable vascular architecture preserved in DUS is thought to be responsible for appropriate regeneration of the uterus. To investigate this concept, we examined the effect of the orientation of the DUS-preserving ECM and the vascular architecture on uterine regeneration through placement of a DUS onto a partially defective uterine area in the reversed orientation such that the luminal face of the DUS was outside and the serosal face was inside. We characterized the tissue structure and function of the regenerated uterus, comparing the outcome to that when the DUS was placed in the correct orientation. Histological analysis revealed that aberrant structures including ectopic location of glands and an abnormal lining of smooth muscle layers were observed significantly more frequently in the reversed group than in the correct group (70% vs. 30%, P < 0.05). Despite the changes in tissue topology, the uteri regenerated with an incorrectly oriented DUS could achieve pregnancy in a way similar to uteri regenerated with a correctly oriented DUS. These results suggest that DUS-driven ECM orientation determines the regenerated uterus structure. Using DUS in the correct orientation is preferable when clinically applied. The disoriented DUS may deteriorate the tissue topology leading to structural disease of the uterus even though the fertility potential is not immediately affected.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Polaridad Celular/fisiología , Matriz Extracelular/fisiología , Regeneración/fisiología , Andamios del Tejido , Útero/citología , Útero/fisiología , Animales , Técnicas de Cultivo de Célula/veterinaria , Células Cultivadas , Matriz Extracelular/química , Femenino , Intestino Delgado/citología , Intestino Delgado/ultraestructura , Embarazo , Ratas , Ratas Sprague-Dawley , Ingeniería de Tejidos/métodos , Ingeniería de Tejidos/veterinaria , Andamios del Tejido/química , Útero/ultraestructura
4.
Cancer Sci ; 107(12): 1909-1918, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27685891

RESUMEN

The laminin γ2 chain, a subunit of laminin-332 (α3ß3γ2), is a molecular marker for invasive cancer cells, but its pathological roles in tumor progression remain to be clarified. It was recently found that the most N-terminal, domain V (dV) of γ2 chain has activities to bind CD44 and stimulate tumor cell migration and vascular permeability. In the present study, we prepared a mAb recognizing γ2 dV. Immunoblotting with this antibody, for the first time, showed that proteolytic fragments containing dV in a range of 15-80 kDa were highly produced in various human cancer cell lines and lung cancer tissues. In immunohistochemistry of adenocarcinomas and squamous cell carcinomas of the lung, this antibody immunostained the cytoplasm of invasive tumor cells and adjacent stroma much more strongly than a widely used antibody recognizing the C-terminal core part of the processed γ2 chain. This suggests that the dV fragments are highly accumulated in tumor cells and stroma compared to the processed γ2 protein. The strong tumor cell staining with the dV antibody correlated with the tumor malignancy grade. We also found that the laminin ß3 and α3 chains were frequently overexpressed in tumor cells and tumor stroma, respectively. The cytoplasmic dV detection was especially prominent in tumor cells infiltrating stroma, but low in the cells surrounded by basement membranes, suggesting that the active tumor-stroma interaction is critical for the aberrant γ2 expression. The present study suggests important roles of laminin γ2 N-terminal fragments in tumor progression.


Asunto(s)
Laminina/metabolismo , Neoplasias Pulmonares/metabolismo , Animales , Anticuerpos Monoclonales , Biomarcadores , Línea Celular Tumoral , Humanos , Inmunohistoquímica , Laminina/química , Neoplasias Pulmonares/patología , Ratones , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas
5.
Biol Reprod ; 93(4): 84, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26316062

RESUMEN

Uterine endometrium is one of the most important organs for species preservation. However, the physiology of human endometrium remains poorly understood, because the human endometrium undergoes rapid and large changes during each menstrual cycle and it is very difficult to investigate human endometrium as one organ. This remarkable regenerative capacity of human endometrium strongly suggests the existence of adult stem cells, and physiology of endometrium cannot be explained without adult stem cells. Therefore, investigating endometrial stem/progenitor cells should lead to a breakthrough in understanding the normal endometrial physiology and the pathophysiology of endometrial neoplastic disorders, such as endometriosis and endometrial cancer. Several cell populations have been discovered as putative endometrial stem/progenitor cells. Emerging evidence reveals that the endometrial side population (SP) is one of the potential endometrial stem/progenitor populations. Of all the endometrial stem/progenitor cell candidates, the endometrial SP (ESP) is best investigated in vitro and in vivo, and has the largest number of references. In this review, we provide an overview of the accumulating evidence for the ESP cells, both directly from human endometria and from cultured endometrial cells. Furthermore, SP cells are compared to other potential stem/progenitor cells, and we discuss their stem cell properties. We also discuss the difficulties and unsolved issues in endometrial stem cell biology.


Asunto(s)
Endometrio/citología , Células de Población Lateral/fisiología , Células Madre/fisiología , Animales , Femenino , Humanos
6.
Biol Reprod ; 93(2): 37, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26108791

RESUMEN

Repeated and dramatic pregnancy-induced uterine enlargement and remodeling throughout reproductive life suggests the existence of uterine smooth muscle stem/progenitor cells. The aim of this study was to isolate and characterize stem/progenitor-like cells from human myometrium through identification of specific surface markers. We here identify CD49f and CD34 as markers to permit selection of the stem/progenitor cell-like population from human myometrium and show that human CD45(-) CD31(-) glycophorin A(-) and CD49f(+) CD34(+) myometrial cells exhibit stem cell-like properties. These include side population phenotypes, an undifferentiated status, high colony-forming ability, multilineage differentiation into smooth muscle cells, osteoblasts, adipocytes, and chondrocytes, and in vivo myometrial tissue reconstitution following xenotransplantation. Furthermore, CD45(-) CD31(-) glycophorin A(-) and CD49f(+) CD34(+) myometrial cells proliferate under hypoxic conditions in vitro and, compared with the untreated nonpregnant myometrium, show greater expansion in the estrogen-treated nonpregnant myometrium and further in the pregnant myometrium in mice upon xenotransplantation. These results suggest that the newly identified myometrial stem/progenitor-like cells influenced by hypoxia and sex steroids may participate in pregnancy-induced uterine enlargement and remodeling, providing novel insights into human myometrial physiology.


Asunto(s)
Antígenos CD34/genética , Antígenos CD34/fisiología , Integrina alfa6/genética , Integrina alfa6/fisiología , Miometrio/metabolismo , Células Madre/fisiología , Útero/fisiología , Animales , Diferenciación Celular , Hipoxia de la Célula , Linaje de la Célula/genética , Femenino , Glicoforinas/biosíntesis , Glicoforinas/genética , Células Madre Hematopoyéticas , Humanos , Ratones , Miometrio/citología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/biosíntesis , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Embarazo
7.
Exp Cell Res ; 326(2): 267-79, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24780821

RESUMEN

Interaction between tumor cells and stromal fibroblasts plays essential roles in tumor progression. However, its detailed molecular mechanism remains unclear. To understand the mechanism, we investigated molecules mediating this interaction using the three-dimensional (3D) co-culture system of Panc-1 pancreatic carcinoma cells with normal fibroblasts. When the two kinds of cells were placed on the top of collagen gel, the tumor cells scattered into the fibroblast layer, apparently undergoing epithelial-mesenchymal transition. When fibroblasts were placed within collagen gel, Panc-1 cells actively invaded into the collagen gel, extending a microtubule-based long protrusion. Although transforming growth factor-ß (TGF-ß) and hepatocyte growth factor (HGF) individually stimulated the tumor cell invasion into collagen gel without fibroblasts, TGF-ß signaling inhibitors (SB431542 and LY2157299) significantly enhanced the Panc-1 cell invasion in the 3D co-culture with fibroblasts. Experiments with HGF/Met signaling inhibitors or with the fibroblast conditioned medium revealed that HGF was a major invasion-promoting factor secreted from fibroblasts and SB431542 increased the HGF secretion by blocking the HGF-suppressing activity of cancer cell-derived TGF-ß. These results indicate that HGF and TGF-ß are critical regulators for both tumor-stroma interaction and tumor invasion. The results also suggest that TGF-ß signaling inhibitors may promote tumor progression under some pathological conditions.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Invasividad Neoplásica/fisiopatología , Neoplasias Pancreáticas/metabolismo , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Benzamidas/farmacología , Línea Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Colágeno/metabolismo , Dioxoles/farmacología , Transición Epitelial-Mesenquimal , Fibroblastos/metabolismo , Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Humanos , Modelos Estadísticos , Invasividad Neoplásica/patología , Neoplasias Pancreáticas/patología , Transducción de Señal , Células del Estroma/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
8.
Endocr J ; 62(1): 93-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25312800

RESUMEN

The objective of this study was to assess the potential predictive factors for follicle growth, ovulation, and pregnancy rate in patients with primary ovarian insufficiency/premature ovarian failure (POI/POF). We enrolled 25 POI patients with desired fertility who were treated and monitored for a minimum of 7 months between the years of 2000-2009 into this retrospective study. The clinical, endocrinologic, chromosomal, and autoimmunologic parameters of these patients were collected. Furthermore, hormonal backgrounds on each of 620 treatment cycles were investigated. The main outcome measures were follicle growth, ovulation, and pregnancy rate. Four of 25 patients (16%) conceived while being monitored and undergoing treatment. Follicle growth, ovulation, and pregnancy rate were not significantly different as a function of parity, iatrogenic history (e.g., chemotherapy), age of disease onset, serum estradiol (E(2))/follicle stimulating hormone (FSH) level at the time of diagnosis, chromosomal abnormality, and positive autoantibody titer. The serum E2 levels on days 1-5 of withdrawal bleeding (Day 1-5 E(2)) were significantly higher in the cycles with successful follicle growth and ovulation than unsuccessful cycles (P<0.05). Receiver-operator characteristic curve analysis revealed the cut-off value of the Day 1-5 E(2) to be 15.5 pg/mL, and an area under the curve (AUC) value of 0.674 for follicle growth and 0.752 for ovulation. The results suggest that cycles with a Day 1-5 E(2)≥15.5 pg/mL have a higher rate of follicle growth and ovulation in patients with POI.


Asunto(s)
Estradiol/sangre , Infertilidad Femenina/etiología , Metrorragia/etiología , Ovario/fisiopatología , Insuficiencia Ovárica Primaria/sangre , Adulto , Biotransformación , Estrógenos/farmacocinética , Estrógenos/uso terapéutico , Femenino , Fármacos para la Fertilidad Femenina/farmacocinética , Fármacos para la Fertilidad Femenina/uso terapéutico , Estudios de Seguimiento , Humanos , Infertilidad Femenina/prevención & control , Japón/epidemiología , Metrorragia/prevención & control , Oogénesis/efectos de los fármacos , Ovario/efectos de los fármacos , Ovario/metabolismo , Ovulación/efectos de los fármacos , Embarazo , Índice de Embarazo , Insuficiencia Ovárica Primaria/tratamiento farmacológico , Insuficiencia Ovárica Primaria/fisiopatología , Curva ROC , Estudios Retrospectivos , Adulto Joven
9.
J Biol Chem ; 288(13): 9066-76, 2013 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-23395821

RESUMEN

Synthetic inhibitors of matrix metalloproteinases (MMPs), designed previously, as well as tissue inhibitors of metalloproteinases (TIMPs) lack enzyme selectivity, which has been a major obstacle for developing inhibitors into safe and effective MMP-targeted drugs. Here we designed a fusion protein named APP-IP-TIMP-2, in which the ten amino acid residue sequence of APP-derived MMP-2 selective inhibitory peptide (APP-IP) is added to the N terminus of TIMP-2. The APP-IP and TIMP-2 regions of the fusion protein are designed to interact with the active site and the hemopexin-like domain of MMP-2, respectively. The reactive site of the TIMP-2 region, which has broad specificity against MMPs, is blocked by the APP-IP adduct. The recombinant APP-IP-TIMP-2 showed strong inhibitory activity toward MMP-2 (Ki(app) = 0.68 pm), whereas its inhibitory activity toward MMP-1, MMP-3, MMP-7, MMP-8, MMP-9, or MT1-MMP was six orders of magnitude or more weaker (IC50 > 1 µm). The fusion protein inhibited the activation of pro-MMP-2 in the concanavalin A-stimulated HT1080 cells, degradation of type IV collagen by the cells, and the migration of stimulated cells. Compared with the decapeptide APP-IP (t½ = 30 min), APP-IP-TIMP-2 (t½ ≫ 96 h) showed a much longer half-life in cultured tumor cells. Therefore, the fusion protein may be a useful tool to evaluate contributions of proteolytic activity of MMP-2 in various pathophysiological processes. It may also be developed as an effective anti-tumor drug with restricted side effects.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Metaloproteinasa 2 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Amiloide/química , Sitios de Unión , Línea Celular Tumoral , Colágeno Tipo IV/metabolismo , Concanavalina A/farmacología , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Fluoresceína-5-Isotiocianato/farmacología , Regulación Neoplásica de la Expresión Génica , Vectores Genéticos , Humanos , Concentración 50 Inhibidora , Cinética , Inhibidores de la Metaloproteinasa de la Matriz/química , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes/química , Factores de Tiempo
10.
Cancer Sci ; 105(2): 168-75, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24238220

RESUMEN

Laminin γ2 (Lmγ2) chain, a subunit of laminin-332, is a typical molecular marker of invading cancer cells, and its expression correlates with poor prognosis of cancer patients. It was previously found that forced expression of Lmγ2 in cancer cells promotes their invasive growth in nude mice. However, the mechanism of the tumor-promoting activity of Lmγ2 remains unknown. Here we investigated the interaction between Lmγ2 and vascular endothelial cells. When treated with an N-terminal proteolytic fragment of γ2 (γ2pf), HUVECs became markedly retracted or shrunken. The overexpression of Lmγ2 or treatment with γ2pf stimulated T-24 bladder carcinoma cells to invade into the HUVEC monolayer and enhanced their transendothelial migration in vitro. Moreover, γ2pf increased endothelial permeability in vitro and in vivo. As the possible mechanisms, γ2pf activated ERK and p38 MAPK but inactivated Akt in HUVECs. Such effects of γ2pf led to prominent actin stress fiber formation in HUVECs, which was blocked by a ROCK inhibitor. In addition, γ2pf induced delocalization of VE-cadherin and ß-catenin from the intercellular junction. As possible receptors, γ2pf interacted with heparan sulfate proteoglycans on the surface of HUVECs. Moreover, we localized the active site of γ2pf to the N-terminal epidermal growth factor-like repeat. These data suggest that the interaction between γ2pf and heparan sulfate proteoglycans induces cytoskeletal changes of endothelial cells, leading to the loss of endothelial barrier function and the enhanced transendothelial migration of cancer cells. These activities of Lmγ2 seem to support the aberrant growth of cancer cells.


Asunto(s)
Permeabilidad Capilar/fisiología , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Laminina/metabolismo , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular/fisiología , Citoesqueleto/metabolismo , Citoesqueleto/patología , Citoesqueleto/fisiología , Células Endoteliales/fisiología , Endotelio Vascular/patología , Endotelio Vascular/fisiología , Factor de Crecimiento Epidérmico/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Uniones Intercelulares/metabolismo , Uniones Intercelulares/patología , Uniones Intercelulares/fisiología , Laminina/farmacología , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos BALB C , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Recombinantes/farmacología , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología , beta Catenina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
J Biol Chem ; 287(7): 4441-50, 2012 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-22174415

RESUMEN

Human embryo implantation is a critical multistep process consisting of embryo apposition/adhesion, followed by penetration and invasion. Through embryo penetration, the endometrial epithelial cell barrier is disrupted and remodeled by an unknown mechanism. We have previously developed an in vitro model for human embryo implantation employing the human choriocarcinoma cell line JAR and the human endometrial adenocarcinoma cell line Ishikawa. Using this model we have shown that stimulation with ovarian steroid hormones (17ß-estradiol and progesterone, E2P4) and suberoylanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor, enhances the attachment and adhesion of JAR spheroids to Ishikawa. In the present study we showed that the attachment and adhesion of JAR spheroids and treatment with E2P4 or SAHA individually induce the epithelial-mesenchymal transition (EMT) in Ishikawa cells. This was evident by up-regulation of N-cadherin and vimentin, a mesenchymal cell marker, and concomitant down-regulation of E-cadherin in Ishikawa cells. Stimulation with E2P4 or SAHA accelerated Ishikawa cell motility, increased JAR spheroid outgrowth, and enhanced the unique redistribution of N-cadherin, which was most prominent in proximity to the adhered spheroids. Moreover, an N-cadherin functional blocking antibody attenuated all events but not JAR spheroid adhesion. These results collectively provide evidence suggesting that E2P4- and implanting embryo-induced EMT of endometrial epithelial cells may play a pivotal role in the subsequent processes of human embryo implantation with functional control of N-cadherin.


Asunto(s)
Implantación del Embrión/fisiología , Embrión de Mamíferos/metabolismo , Endometrio/metabolismo , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Modelos Biológicos , Antígenos CD/biosíntesis , Cadherinas/biosíntesis , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Implantación del Embrión/efectos de los fármacos , Embrión de Mamíferos/citología , Endometrio/citología , Células Epiteliales/citología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/fisiología , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Progesterona/farmacología , Progestinas/farmacología , Esferoides Celulares/citología , Esferoides Celulares/metabolismo , Vimentina/biosíntesis , Vorinostat
12.
Endocr J ; 60(6): 791-7, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23445562

RESUMEN

Women with primary ovarian insufficiency (POI)/premature ovarian failure exhibit hypergonadotropic hypogonadism due to follicle dysfunction and depletion before the age of 40 years. Because ovulation is extremely rare and thought to be unpredictable in women with POI and because no ovulation induction regimens have been shown to be efficacious, oocyte donation is the only evidence-based treatment for women with POI with desired fertility. Oocyte donation is, however, extremely limited in several countries including Japan. Here, we report four women with POI who achieved pregnancies resulting from timed intercourse or intrauterine insemination in combination with cyclic estrogen/progesterone therapy and close monitoring of follicle development. These four patients were diagnosed with POI at the mean age of 27.5 ± 8.5 (mean ± SD; range, 19-35), subjected to follicle monitoring at the mean age of 29.8 ± 5.7 (23-35), and conceived at the mean age of 34.5 ± 3.9 (29-38). The interval between the initiation of follicle monitoring and pregnancy was 4.8 ± 2.8 (2-8) years. In one of the patients, her most recent ovulation occurred after a three-year interval. All four patients had uncomplicated pregnancies with term deliveries. In the event that oocyte donation and adoption are not available and/or various treatments with intensive ovulation induction have been unsuccessful, close and continuous monitoring of follicle growth to identify very rare ovulatory events might be considered for patients with POI and desired fertility.


Asunto(s)
Fertilización , Infertilidad Femenina/etiología , Infertilidad Femenina/terapia , Folículo Ovárico/fisiología , Ovulación , Insuficiencia Ovárica Primaria/complicaciones , Adulto , Femenino , Humanos , Inseminación Artificial , Monitoreo Fisiológico , Inducción de la Ovulación/métodos , Embarazo , Adulto Joven
13.
Endocr J ; 60(10): 1155-64, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23883529

RESUMEN

Nerve growth factor (NGF) has been recently proposed as one of the key factors responsible not only for promotion of nerve fiber growth but also for the onset and maintenance of pain in a variety of diseases. The aim of this study was to investigate the role of NGF in the pelvic pain associated with endometriosis. Tissue and peritoneal fluid samples were collected from 95 women with laparoscopically and histopathologically confirmed endometriosis and 59 control women without endometriosis. Expression levels of NGF mRNA and protein were examined using real-time RT-PCR and immunohistochemistry, respectively. Concentration of NGF in the peritoneal fluid (PF-NGF) was measured using ELISA. The degree of dyspareunia and dysmenorrhea was evaluated using a verbal rating scale. Real-time RT-PCR analysis revealed that NGF mRNA was significantly more abundant in the ovarian endometriomas and peritoneal endometriosis than in the normal control endometrium. Immunohistochemical analyses demonstrated that NGF was prominently expressed and preferentially localized to the glands of the ovarian endometriomas and peritoneal endometriosis, whereas it was only weakly detectable in the normal endometrium. Although PF-NGF was undetectable in some normal subjects and endometriosis patients, elevated PF-NGF in the peritoneal fluid was more frequently observed in endometriosis patients with severe pain than in those with less severe pain. Our results suggest that NGF produced locally in the peritoneal cavity may be involved in the generation of endometriosis-associated pelvic pain.


Asunto(s)
Dismenorrea/etiología , Dispareunia/etiología , Endometriosis/complicaciones , Factor de Crecimiento Nervioso/fisiología , Dolor Pélvico/etiología , Adulto , Líquido Ascítico/química , Endometriosis/fisiopatología , Femenino , Humanos , Factor de Crecimiento Nervioso/biosíntesis , Regulación hacia Arriba
14.
Reprod Med Biol ; 12(3): 79-84, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29699134

RESUMEN

To achieve a successful pregnancy in humans, sperm is required for capacitation, followed by binding to and entry into an oocyte. Maternal endometrial epithelial cells (EECs) prepare the appropriate implantation environment through regulation of immune cells and endometrial cells. After acquiring endometrial receptivity, a successful pregnancy consists of complex and finely regulated steps involving apposition, adhesion, invasion, and penetration. Glycodelin is a secretory glycoprotein that affects cell proliferation, differentiation, adhesion, and motility. Glycodelin has four glycoforms (glycodelin-A, -S, -F. and -C); differences in glycosylation affect each characteristic function. Glycodelin has a unique temporospatial pattern of expression, primarily in the reproductive tract where glycodelin is mid-secretory phase-dominant. Recent studies have demonstrated that glycodelin protein has the potential to regulate various processes, including immunosuppression, fertilization, and implantation. This review details the orchestrated regulation of successful pregnancy by glycodelin as well as a discussion of the basic characteristics of glycodelin.

15.
J Biol Chem ; 286(38): 33236-43, 2011 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-21813640

RESUMEN

Unlike other synthetic or physiological inhibitors for matrix metalloproteinases (MMPs), the ß-amyloid precursor protein-derived inhibitory peptide (APP-IP) having an ISYGNDALMP sequence has a high selectivity toward MMP-2. Our previous study identified amino acid residues of MMP-2 essential for its selective inhibition by APP-IP and demonstrated that the N to C direction of the decapeptide inhibitor relative to the substrate-binding cleft of MMP-2 is opposite that of substrate. However, detailed interactions between the two molecules remained to be clarified. Here, we determined the crystal structure of the catalytic domain of MMP-2 in complex with APP-IP. We found that APP-IP in the complex is indeed embedded into the substrate-binding cleft of the catalytic domain in the N to C direction opposite that of substrate. With the crystal structure, it was first clarified that the aromatic side chain of Tyr(3) of the inhibitor is accommodated into the S1' pocket of the protease, and the carboxylate group of Asp(6) of APP-IP coordinates bidentately to the catalytic zinc of the enzyme. The Ala(7) to Pro(10) and Tyr(3) to Ile(1) strands of the inhibitor extend into the nonprime and the prime sides of the cleft, respectively. Therefore, the decapeptide inhibitor has long range contact with the substrate-binding cleft of the protease. This mode of interaction is probably essential for the high MMP-2 selectivity of the inhibitor because MMPs share a common architecture in the vicinity of the catalytic center, but whole structures of their substrate-binding clefts have sufficient variety for the inhibitor to distinguish MMP-2 from other MMPs.


Asunto(s)
Metaloproteinasa 2 de la Matriz/química , Inhibidores de la Metaloproteinasa de la Matriz , Proteínas/metabolismo , Secuencia de Aminoácidos , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Proteínas/química
16.
Cancer Sci ; 103(4): 691-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22321149

RESUMEN

Angiomodulin (AGM/IGFBP-rP1), a glycoprotein of about 30 kDa, is overexpressed in tumor vasculature as well as some human cancer cell lines, but it has been suggested to be a tumor suppressor. To elucidate roles of angiomodulin (AGM) in tumor progression, we here examined distribution of AGM in three types of human cancer tissues by immunohistochemistry. The results showed that AGM was overexpressed in the stroma as well as the vasculature surrounding tumor cells in the human cancer tissues. AGM and α-smooth muscle actin (α-SMA) as an activated fibroblast marker were often colocalized in cancer-associated fibroblasts (CAFs). In vitro analysis indicated that transforming growth factor (TGF)-ß1 might be an important inducer of AGM in normal human fibroblasts. AGM strongly stimulated the expression of fibronectin and weakly that of α-SMA in normal fibroblasts. AGM significantly stimulated the proliferation and migration of fibroblasts. The AGM-induced expression of fibronectin and α-SMA was blocked by a TGF-ß signal inhibitor but neither the stimulation of cell growth nor migration. These results imply that AGM activates normal fibroblasts by TGF-ß-dependent and independent mechanisms. These findings also suggest that AGM and TGF-ß1 cooperatively or complementarily contribute to the stromal activation and connective tissue formation in human cancer tissues, contributing to tumor progression.


Asunto(s)
Fibroblastos/fisiología , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Neoplasias/metabolismo , Células del Estroma/metabolismo , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Humanos , Neoplasias/irrigación sanguínea , Factor de Crecimiento Transformador beta/farmacología
17.
Stem Cell Res Ther ; 13(1): 225, 2022 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-35659728

RESUMEN

BACKGROUND: Distinct subsets of cancer stem cells (CSCs) drive the initiation and progression of malignant tumors via enhanced self-renewal and development of treatment/apoptosis resistance. Endometrial CSC-selective drugs have not been successfully developed because most endometrial cell lines do not contain a sufficient proportion of stable CSCs. Here, we aimed to identify endometrial CSC-containing cell lines and to search for endometrial CSC-selective drugs. METHODS: We first assessed the presence of CSCs by identifying side populations (SPs) in several endometrial cancer cell lines. We then characterized cell viability, colony-formation, transwell invasion and xenotransplantion capability using the isolated SP cells. We also conducted real-time RT-PCR, immunoblot and immunofluorescence analyses of the cells' expression of CSC-associated markers. Focusing on 14 putative CSC-selective drugs, we characterized their effects on the proliferation and apoptosis of endometrial cancer cell lines, examining cell viability and annexin V staining. We further examined the inhibitory effects of the selected drugs, focusing on proliferation, invasion, expression of CSC-associated markers and tumor formation. RESULTS: We focused on HHUA cells, an endometrial cancer cell line derived from a well-differentiated endometrial adenocarcinoma. HHUA cells contained a sufficient proportion of stable CSCs with an SP phenotype (HHUA-SP). HHUA-SP showed greater proliferation, colony-formation, and invasive capabilities compared with the main population of HHUA cells (HHUA-MP). HHUA-SP generated larger tumors with higher expression of proliferation-related markers, Ki67, c-MYC and phosphorylated ERK compared with HHUA-MP when transplanted into immunodeficient mice. Among the 14 candidate drugs, sorafenib, an inhibitor of RAF pathways and multiple kinase receptors, inhibited cell proliferation and invasion in both HHUA-SP and -MP, but more profoundly in HHUA-SP. In vivo treatment with sorafenib for 4 weeks reduced the weights of HHUA-SP-derived tumors and decreased the expression of Ki67, ZEB1, and RAF1. CONCLUSIONS: Our results suggest that HHUA is a useful cell line for discovery and identification of endometrial CSC-selective drugs, and that sorafenib may be an effective anti-endometrial cancer drug targeting endometrial CSCs.


Asunto(s)
Neoplasias Endometriales , Sistema de Señalización de MAP Quinasas , Animales , Carcinogénesis/patología , Línea Celular Tumoral , Proliferación Celular , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Femenino , Humanos , Antígeno Ki-67/metabolismo , Ratones , Células Madre Neoplásicas/metabolismo , Sorafenib/metabolismo , Sorafenib/farmacología
18.
J Biol Chem ; 285(37): 28862-73, 2010 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-20605794

RESUMEN

Localization of secreted matrix metalloproteinases (MMPs) on the cell surface is required not only for processing of cell surface proteins, but also for controlled degradation of the extracellular matrix (ECM). Our previous study demonstrated that binding of MMP-7 (matrilysin) to cell surface cholesterol sulfate (CS) is essential for the cell membrane-associated proteolytic action of this MMP. In this study, we investigated the role of CS in the MMP-7-catalyzed degradation of protein components of ECM. We found that the degradation of laminin-332 (laminin-5) catalyzed by MMP-7 was accelerated dramatically in the presence of CS, whereas the sulfated lipid inhibited the degradation of casein catalyzed by the protease. The MMP-7-catalyzed degradation of fibronectin was partially inhibited in the presence of low concentrations of CS, whereas it was accelerated significantly at high concentrations of the lipid. Therefore, it is likely that CS alters the substrate preference of MMP-7. We also found that the proteins of which MMP-7-catalyzed degradation were accelerated by CS also had affinities for CS, suggesting that CS facilitates the proteolyses by cross-linking MMP-7 to its substrates. Moreover, MMP-7 tethered to cancer cell surface via CS degraded fibronectin and laminin-332 coated on a culture plate. The degradations of the adhesive proteins led to significant detachment of the cells from the plate. Taken together, our findings provide a novel mechanism in which cell surface CS promotes the proteolytic activities of MMP-7 toward selective substrates in the pericellular ECM, thereby contributing to cancer cell migration and metastasis.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Ésteres del Colesterol/farmacología , Fibronectinas/metabolismo , Metaloproteinasa 7 de la Matriz/metabolismo , Inhibidores de Serina Proteinasa/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Metástasis de la Neoplasia , Neoplasias/metabolismo , Neoplasias/patología , Especificidad por Sustrato/efectos de los fármacos , Kalinina
19.
J Biol Chem ; 285(45): 35068-78, 2010 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-20805229

RESUMEN

The basement membrane (BM) proteins laminins, which consist of α, ß, and γ chains, support tissue structures and cellular functions. To date only α4 and α5 types of laminins have been identified in the BMs of blood vessels. Our recent study suggested the presence of novel α3B-containing laminins in vascular BMs. Here we identified and characterized the third member of vascular laminins, laminin-3B11 (Lm3B11). RT-PCR analysis showed that microvascular endothelial (MVE) cells and umbilical vein endothelial cells expressed the messages for the α3B, ß1, ß2, and γ1 chains. In the culture of MVE cells, α3B was associated with ß1 and γ1, producing Lm3B11. Recombinant Lm3B11 was overexpressed by introducing the cDNAs of the three chains into HEK-293 cells and purified to homogeneity. Purified Lm3B11 exhibited relatively weak cell adhesion activity through both α3ß1 and α6ß1 integrins. Most characteristically, Lm3B11 strongly stimulated MVE cells to extend many lamellipodial protrusions. This pseudopodial branching was blocked by an inhibitor for Src or phosphatidylinositol 3-kinase. Consistently, Lm3B11 stimulated the phosphorylation of Src and Akt more strongly than other laminins, suggesting that the integrin-derived signaling is mediated by these factors. The unique activity of Lm3B11 appears to be favorable to the branching of capillaries and venules.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Células Endoteliales/metabolismo , Microvasos/metabolismo , Seudópodos/metabolismo , Moléculas de Adhesión Celular/genética , Línea Celular , Células Endoteliales/citología , Humanos , Microvasos/citología , Proteína Oncogénica pp60(v-src)/genética , Proteína Oncogénica pp60(v-src)/metabolismo , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Seudópodos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Kalinina
20.
Cancer Sci ; 102(5): 1095-100, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21276136

RESUMEN

Laminins present in the basement membranes (BM) of blood vessels are involved in angiogenesis and other vascular functions that are critical for tumor growth and metastasis. Two major vascular laminins, the α4 (laminin-411/421) and α5 (laminin-511/521) types, have been well characterized. We recently found a third type of vascular laminin, laminin-3B11, consisting of the α3B, ß1 and γ1 chains, and revealed its biological activity. Laminin-3B11 potently stimulates vascular endothelial cells to extend lamellipodial protrusions. To understand the roles of laminin-3B11 in blood vessel functions and tumor growth, we examined localization of the laminin α3B chain in normal mammary glands and breast cancers, in comparison with the α4 and α5 laminins. In the immunohistochemical analysis, the α3B laminin was co-localized with the α4 and α5 laminins in the BM of venules and capillaries of normal breast tissues, but α3B was scarcely detected in vessels near invasive breast carcinoma cells. In contrast, the α4 laminin was overexpressed in capillaries of invasive carcinomas, where a large number of macrophages were found. The α5 laminin appeared to be weakly downregulated in cancer tissues, especially in capillary vessels. Furthermore, our in vitro analysis indicated that TNF-α significantly suppressed the laminin α3B expression in vascular endothelial cells, while it, as well as IL-1ß and TGF-α, upregulated the α4 expression. These results suggest that Lm3B11/3B21 may be required for normal mature vessels and interfere with tumor angiogenesis.


Asunto(s)
Membrana Basal/metabolismo , Neoplasias de la Mama/metabolismo , Regulación hacia Abajo , Células Endoteliales/metabolismo , Laminina/metabolismo , Neoplasias de la Mama/patología , Femenino , Humanos , Inmunohistoquímica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA