Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
New Phytol ; 221(3): 1345-1358, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30267580

RESUMEN

Although cell number generally correlates with organ size, the role of cell cycle control in growth regulation is still largely unsolved. We studied kip related protein (krp) 4, 6 and 7 single, double and triple mutants of Arabidopsis thaliana to understand the role of cell cycle inhibitory proteins in leaf development. We performed leaf growth and seed size analysis, kinematic analysis, flow cytometery, transcriptome analysis and mathematical modeling of G1/S and G2/M checkpoint progression of the mitotic and endoreplication cycle. Double and triple mutants progressively increased mature leaf size, because of elevated expression of cell cycle and DNA replication genes stimulating progression through the division and endoreplication cycle. However, cell number was also already increased before leaf emergence, as a result of an increased cell number in the embryo. We show that increased embryo and seed size in krp4/6/7 results from seed abortion, presumably reducing resource competition, and that seed size differences contribute to the phenotype of several large-leaf mutants. Our results provide a new mechanistic understanding of the role of cell cycle regulation in leaf development and highlight the contribution of the embryo to the development of leaves after germination in general.


Asunto(s)
Proteínas de Arabidopsis/genética , Arabidopsis/anatomía & histología , Arabidopsis/genética , Regulación de la Expresión Génica de las Plantas , Hojas de la Planta/anatomía & histología , Arabidopsis/citología , Arabidopsis/embriología , Proteínas de Arabidopsis/metabolismo , Fenómenos Biomecánicos , Recuento de Células , Ciclo Celular/genética , División Celular , ADN de Plantas/biosíntesis , Regulación hacia Abajo/genética , Endorreduplicación , Perfilación de la Expresión Génica , Cinética , Mutación/genética , Tamaño de los Órganos , Fenotipo , Hojas de la Planta/citología , Hojas de la Planta/crecimiento & desarrollo , Plantas Modificadas Genéticamente , Ploidias , Semillas/anatomía & histología , Semillas/fisiología , Regulación hacia Arriba/genética
2.
Cell Physiol Biochem ; 43(3): 1064-1076, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28977799

RESUMEN

BACKGROUND/AIMS: Physiological role of luteinizing hormone (LH) and its receptor (LHCGR) in adrenal remains unknown. In inhibin-α/Simian Virus 40 T antigen (SV40Tag) (inhα/Tag) mice, gonadectomy-induced (OVX) elevated LH triggers the growth of transcription factor GATA4 (GATA4)-positive adrenocortical tumors in a hyperplasia-adenoma-adenocarcinoma sequence. METHODS: We investigated the role of LHCGR in tumor induction, by crossbreeding inhα/Tag with Lhcgr knockout (LuRKO) mice. By knocking out Lhcgr and Gata4 in Cα1 adrenocortical cells (Lhcgr-ko, Gata4-ko) we tested their role in tumor progression. RESULTS: Adrenal tumors of OVX inhα/Tag mice develop from the hyperplastic cells localized in the topmost layer of zona fasciculata. OVX inhα/Tag/LuRKO only developed SV40Tag positive hyperplastic cells that were GATA4 negative, cleaved caspase-3 positive and did not progress into adenoma. In contrast to Lhcgr-ko, Gata4-ko Cα1 cells presented decreased proliferation, increased apoptosis, decreased expression of Inha, SV40Tag and Lhcgr tumor markers, as well as up-regulated adrenal- and down-regulated sex steroid gene expression. Both Gata4-ko and Lhcgr-ko Cα1 cells had decreased expression of steroidogenic genes resulting in decreased basal progesterone production. CONCLUSION: Our data indicate that LH/LHCGR signaling is critical for the adrenal cell reprogramming by GATA4 induction prompting adenoma formation and gonadal-like phenotype of the adrenocortical tumors in inhα/Tag mice.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/patología , Factor de Transcripción GATA4/metabolismo , Hormona Luteinizante/metabolismo , Neoplasias de la Corteza Suprarrenal/etiología , Neoplasias de la Corteza Suprarrenal/metabolismo , Glándulas Suprarrenales/metabolismo , Glándulas Suprarrenales/patología , Animales , Antígenos Transformadores de Poliomavirus/genética , Antígenos Transformadores de Poliomavirus/metabolismo , Apoptosis , Sistemas CRISPR-Cas/genética , Caspasa 3/metabolismo , Proliferación Celular , Transformación Celular Neoplásica , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Regulación hacia Abajo , Femenino , Fluoroinmunoensayo , Factor de Transcripción GATA4/deficiencia , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA6/metabolismo , Gónadas/cirugía , Inhibinas/genética , Inhibinas/metabolismo , Hormona Luteinizante/sangre , Ratones , Ratones Noqueados , Ratones Transgénicos , Fenotipo , Fosfoproteínas/metabolismo , Receptores de HL/deficiencia , Receptores de HL/genética , Factor Esteroidogénico 1/metabolismo
3.
Cells ; 11(17)2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-36078114

RESUMEN

The aim of this study was to evaluate the effect of acute aldosterone (ALDO) administration on the vascular permeability of skin. ALDO was injected intradermally into rats, and vascular permeability was measured. Eplerenone (EPL), a selective mineralocorticoid receptor (MR) antagonist, was used. Skin biopsies were carried out for immunohistochemical (IHC) staining, and polymerase chain reactions were performed to analyze the expression of MR, 11ß-hydroxysteroid dehydrogenase type 2, von Willebrand factor (vWF), vascular endothelial growth factor (VEGF), and zonula occludens 1. Our study showed the presence of MR in the rat skin vasculature for the first time. It was found that ALDO injection resulted in a more than 30% increase in vascular permeability and enhanced the endothelial exocytosis of vWF. The effect of ALDO diminished after EPL administration. An accumulation of vWF and a reduction in VEGF IHC staining were observed following chronic EPL administration. No effect of ALDO or EPL on the mRNA expression of the studied genes or skin structure was observed. The results suggest that ALDO increases vascular permeability in the skin via an MR-dependent mechanism. This effect of ALDO on skin microcirculation may have important therapeutic implications for diseases characterized by increased levels of ALDO and coexisting skin microangiopathy.


Asunto(s)
Aldosterona , Permeabilidad Capilar , Aldosterona/metabolismo , Aldosterona/farmacocinética , Aldosterona/farmacología , Animales , Permeabilidad Capilar/efectos de los fármacos , Eplerenona , Antagonistas de Receptores de Mineralocorticoides , Ratas , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor de von Willebrand/metabolismo
4.
EBioMedicine ; 47: 170-183, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31466918

RESUMEN

BACKGROUND: Recent clinical trials on ovarian cancer with mifepristone (MF) have failed, despite in vitro findings on its strong progesterone (P4) antagonist function. METHODS: Ovarian cancer human and murine cell lines, cultured high-grade human primary epithelial ovarian cancer (HG-hOEC) cells and their explants; as well as in vivo transgenic mice possessing ovarian cancer were used to assess the molecular mechanism underlying mifepristone (MF) agonistic actions in ovarian cancer progression. FINDINGS: Herein, we show that ovarian cancer cells express traceable/no nuclear P4 receptor (PGR), but abundantly P4 receptor membrane component 1 (PGRMC1). MF significantly stimulated ovarian cancer cell migration, proliferation and growth in vivo, and the translocation of PGRMC1 into the nucleus of cancer cells; the effects inhibited by PGRMC1 inhibitor. The beneficial antitumor effect of high-doses MF could not be achieved in human cancer tissue, and the low tissue concentrations achieved with the therapeutic doses only promoted the growth of ovarian cancers. INTERPRETATION: Our results indicate that treatment of ovarian cancer with MF and P4 may induce similar adverse agonistic effects in the absence of classical nuclear PGRs in ovarian cancer. The blockage of PGRMC1 activity may provide a novel treatment strategy for ovarian cancer. FUND: This work was supported by grants from the National Science Centre, Poland (2013/09/N/NZ5/01831 to DP-T; 2012/05/B/NZ5/01867 to MC), Academy of Finland (254366 to NAR), Moikoinen Cancer Research Foundation (to NAR) and EU PARP Cluster grant (UDA-POIG.05.01.00-005/12-00/NCREMFP to SW).


Asunto(s)
Antineoplásicos Hormonales/farmacología , Mifepristona/farmacología , Animales , Antineoplásicos Hormonales/administración & dosificación , Antineoplásicos Hormonales/farmacocinética , Biomarcadores , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/metabolismo , Carcinoma Epitelial de Ovario/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Células Cultivadas , Femenino , Humanos , Inmunohistoquímica , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Mifepristona/administración & dosificación , Mifepristona/farmacocinética , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
5.
Endocr Relat Cancer ; 26(1): 103-117, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30400009

RESUMEN

Aberrantly expressed G protein-coupled receptors in tumors are considered as potential therapeutic targets. We analyzed the expressions of receptors of gonadotropin-releasing hormone (GNRHR), luteinizing hormone/chorionic gonadotropin (LHCGR) and follicle-stimulating hormone (FSHR) in human adrenocortical carcinomas and assessed their response to GnRH antagonist therapy. We further studied the effects of the GnRH antagonist cetrorelix acetate (CTX) on cultured adrenocortical tumor (ACT) cells (mouse Cα1 and Y-1, and human H295R), and in vivo in transgenic mice (SV40 T-antigen expression under inhibin α promoter) bearing Lhcgr and Gnrhr in ACT. Both models were treated with control (CT), CTX, human chorionic gonadotropin (hCG) or CTX+hCG, and their growth and transcriptional changes were analyzed. In situ hybridization and qPCR analysis of human adrenocortical carcinomas (n = 11-13) showed expression of GNRHR in 54/73%, LHCGR in 77/100% and FSHR in 0%, respectively. CTX treatment in vitro decreased cell viability and proliferation, and increased caspase 3/7 activity in all treated cells. In vivo, CTX and CTX+hCG (but not hCG alone) decreased ACT weights and serum LH and progesterone concentrations. CTX treatment downregulated the tumor markers Lhcgr and Gata4. Upregulated genes included Grb10, Rerg, Nfatc and Gnas, all recently found to be abundantly expressed in healthy adrenal vs ACT. Our data suggest that CTX treatment may improve the therapy of human adrenocortical carcinomas by direct action on GNRHR-positive cancer cells inducing apoptosis and/or reducing gonadotropin release, directing tumor cells towards a healthy adrenal gene expression profile.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/tratamiento farmacológico , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Antagonistas de Hormonas/uso terapéutico , Neoplasias de la Corteza Suprarrenal/genética , Neoplasias de la Corteza Suprarrenal/metabolismo , Neoplasias de la Corteza Suprarrenal/patología , Adulto , Anciano , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica , Hormona Liberadora de Gonadotropina/farmacología , Hormona Liberadora de Gonadotropina/uso terapéutico , Antagonistas de Hormonas/farmacología , Humanos , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Receptores de HFE/genética , Receptores de HFE/metabolismo , Receptores de HL/genética , Receptores de HL/metabolismo , Receptores LHRH/genética , Receptores LHRH/metabolismo
6.
Endocrinology ; 159(1): 297-309, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29029022

RESUMEN

The goal of this study was to characterize the function of microRNA-27a-3p (miR-27a-3p) in polycystic ovary syndrome (PCOS). miR-27a-3p expression was analyzed in excised granulosa cells (GCs) from 21 patients with PCOS and 12 normal patients undergoing in vitro fertilization cycle treatments and in 17 nontreated cuneiform ovarian resection PCOS samples and 13 control ovarian samples from patients without PCOS. We found that the expression of miR-27a-3p was significantly increased in both excised GCs and the ovaries of patients with PCOS compared with the controls. Insulin treatment of the human granulosa-like tumor cell line (KGN) resulted in decreased downregulated expression of miR-27a-3p, and this effect appeared to be mediated by signal transducer and activator of transcription STAT1 and STAT3. The overexpression of miR-27a-3p in KGN cells inhibited SMAD5, which in turn decreased cell proliferation and promoted cell apoptosis. After KGN cells were stimulated with insulin for 48 hours, there was increased expression of SMAD5 protein and decreased apoptosis. Additionally, knockdown/overexpression of SMAD5 in KGN cells reduced/increased cell number and promoted/inhibited cell apoptosis. Insulin-stimulated primary GCs isolated from patients with PCOS, in contrast to normal GCs or KGN cells, did not exhibit decreased miR-27a-3p expression. The differences in the expression levels in KGN cells and human PCOS GCs are likely explained by increased miR-27a-3p expression in the GCs caused by insulin resistance in PCOS. Taken together, our data provided evidence for a functional role of miR-27a-3p in the GCs' dysfunction that occurs in patients with PCOS.


Asunto(s)
Apoptosis , Regulación de la Expresión Génica , Células de la Granulosa/metabolismo , MicroARNs/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Proteína Smad5/antagonistas & inhibidores , Adulto , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Tumor de Células de la Granulosa/tratamiento farmacológico , Tumor de Células de la Granulosa/metabolismo , Tumor de Células de la Granulosa/patología , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/patología , Humanos , Hipoglucemiantes/farmacología , Insulina/farmacología , Resistencia a la Insulina , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Síndrome del Ovario Poliquístico/patología , ARN/metabolismo , Interferencia de ARN , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Proteína Smad5/agonistas , Proteína Smad5/genética , Proteína Smad5/metabolismo
7.
Sci Rep ; 6: 37095, 2016 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-27848975

RESUMEN

Expression of follicle-stimulation hormone receptor (FSHR) is confined to gonads and at low levels to some extragonadal tissues like human umbilical vein endothelial cells (HUVEC). FSH-FSHR signaling was shown to promote HUVEC angiogenesis and thereafter suggested to have an influential role in pregnancy. We revisited hereby the expression and functionality of FSHR in HUVECs angiogenesis, and were unable to reproduce the FSHR expression in human umbilical cord, HUVECs or immortalized HUVECs (HUV-ST). Positive controls as granulosa cells and HEK293 cells stably transfected with human FSHR cDNA expressed FSHR signal. In contrast to positive control VEGF, FSH treatment showed no effects on tube formation, nitric oxide production, wound healing or cell proliferation in HUVEC/HUV-ST. Thus, it remains open whether the FSH-FSHR activation has a direct regulatory role in the angiogenesis of HUVECs.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Hormona Folículo Estimulante/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Receptores de HFE/biosíntesis , Femenino , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Embarazo , Receptores de HFE/genética
8.
J Clin Endocrinol Metab ; 101(7): 2905-14, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27224263

RESUMEN

CONTEXT: FSH receptor (FSHR), besides being expressed in gonads, is also expressed in some extragonadal tissues at low levels. OBJECTIVE: We examined the functional expression of FSHR in different types of endometriotic lesions. DESIGN: Extensive studies were carried out to detect functional FSHR expression and FSH-stimulated estrogen production in ovarian endometriomas and recto-vaginal endometriotic nodules (RVEN). Normal endometrium, ovary, and myometrium tissues from nonpregnant cycling women served as controls. SETTINGS: This laboratory-based study was carried out on tissue specimens from patients with endometriosis and healthy donors. RESULTS: Endometriotic lesions and normal secretory-phase endometrium showed FSHR expression at both mRNA and protein level. RVEN and ovarian endometrioma demonstrated up-regulated CYP19A1, dependent on the activation of CYP19A1 proximal promoter II. Estrogen receptor-ß (ESR2) expression was significantly increased in RVEN vs normal endometrium. Recombinant human FSH stimulation of RVEN explants significantly increased estradiol production and CYP19A1 and ESR2 expression. FSHR was up-regulated in recombinant human FSH-stimulated endometrial and decidualized stromal cells with increased CYP19A1 expression. CONCLUSIONS: We described a novel functional FSHR expression, where FSH-stimulated CYP19A1 expression and estrogen production in RVEN are demonstrated. This locally FSH-induced estrogen production may contribute to the pathology, development, progression, and severity of RVEN.


Asunto(s)
Aromatasa/genética , Endometriosis/genética , Endometrio/metabolismo , Receptores de HFE/genética , Enfermedades del Recto/genética , Enfermedades Vaginales/genética , Adulto , Aromatasa/metabolismo , Estudios de Casos y Controles , Endometriosis/patología , Endometrio/efectos de los fármacos , Endometrio/patología , Estradiol/metabolismo , Receptor beta de Estrógeno/fisiología , Femenino , Hormona Folículo Estimulante/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Enfermedades del Ovario/genética , Enfermedades del Ovario/patología , Regiones Promotoras Genéticas/efectos de los fármacos , Receptores de HFE/metabolismo , Enfermedades del Recto/patología , Enfermedades Vaginales/patología , Adulto Joven
9.
J Plant Physiol ; 171(14): 1306-14, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25014266

RESUMEN

The exposition of 7-day-old pea seedlings to dehydration induced sudden changes in the concentration of monosaccharides and sucrose in epicotyl and roots tissues. During 24h of dehydration, the concentration of glucose and, to a lesser extent, fructose in seedling tissues decreased. The accumulation of sucrose was observed in roots after 4h and in epicotyls after 8h of stress. Epicotyls and roots also began to accumulate galactinol and raffinose after 8h of stress, when small changes in the water content of tissues occurred. The accumulation of galactinol and raffinose progressed parallel to water withdrawal from tissues, but after seedling rehydration both galactosides disappeared. The synthesis of galactinol and raffinose by an early induction (during the first hour of treatment) of galactinol synthase (PsGolS) and raffinose synthase (PsRS) gene expression as well as a later increase in the activity of both enzymes was noted. Signals possibly triggering the induction of PsGolS and PsRS gene expression and accumulation of galactinol and raffinose in seedlings are discussed.


Asunto(s)
Galactosiltransferasas/genética , Regulación de la Expresión Génica de las Plantas , Pisum sativum/enzimología , Pisum sativum/genética , Proteínas de Plantas/genética , Secuencia de Bases , Desecación , Galactosiltransferasas/metabolismo , Datos de Secuencia Molecular , Monosacáridos/metabolismo , Pisum sativum/metabolismo , Hojas de la Planta/enzimología , Proteínas de Plantas/metabolismo , Raíces de Plantas/enzimología , Plantones/enzimología , Sacarosa/metabolismo
10.
Reprod Biol ; 14(1): 25-31, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24607252

RESUMEN

Granulosa cell tumors are rare, 3-7.6% of primary ovarian tumors, although with poor prognosis as the tumor-related mortality rate is 37.3%, with 80% of deaths occurring on recurrence. We have created a transgenic (TG) murine model for gonadal somatic cell tumors by expressing the powerful viral oncogene, Simian Virus 40 T-antigen (Tag), under the regulation of murine inhibin α-subunit 6 kb promoter (inhα/Tag). Gonadotropin dependent ovarian granulosa cell tumors were formed in females by the age of 5-6 months, with a 100% penetrance. We have successfully used the inhα/Tag model to test different treatment strategies for ovarian tumors. With a gene therapy trial in inhα/Tag mice crossbred with inhα/HSV-TK (herpes simplex virus thymidine kinase) mice (double TG), we proved the principle that targeted expression of HSV-TK gene in gonadal somatic cell tumors enabled tumor ablation by anti-herpes treatment. When we aimed at targeted destruction of luteinizing hormone/chorionic gonadotropin receptor (LHCGR) expressing inhα/Tag tumor cells in vivo by a lytic peptide Hecate-CGß conjugate, we could successfully kill the tumor cells, sparing the normal cells. We recently found high zona pellucida glycoprotein 3 (ZP3) expression in inhα/Tag granulosa cell tumors, as well as in human granulosa cell tumors. We tested the concept of treating the ovarian tumors of inhα/Tag mice by vaccination against the ectopically expressed ZP3. Immunotherapy with recombinant human (rh) ZP3 was highly successful with no objective side effects in inhα/Tag females, suggesting rhZP3 immunization as a novel strategy for the immunotherapy of ovarian granulosa cell tumors.


Asunto(s)
Tumor de Células de la Granulosa/terapia , Inhibinas/genética , Neoplasias Ováricas/terapia , Regiones Promotoras Genéticas , Animales , Antígenos Virales de Tumores/genética , Antígenos Virales de Tumores/metabolismo , Modelos Animales de Enfermedad , Femenino , Tumor de Células de la Granulosa/genética , Tumor de Células de la Granulosa/metabolismo , Ratones , Ratones Transgénicos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Virus 40 de los Simios/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA