Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(7)2024 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-38612451

RESUMEN

Reproduction is the important process of transmitting one's genetic information to the next generation [...].


Asunto(s)
Células Germinativas , Neoplasias , Genitales , Reproducción , Biología
2.
Int J Mol Sci ; 23(13)2022 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-35806199

RESUMEN

This Special Issue is intended to provide up-to-date information on reproduction, including the reproduction of germ cells and reproductive organs (ovary, testis, and uterus) [...].


Asunto(s)
Reproducción , Testículo , Femenino , Células Germinativas , Humanos , Masculino , Ovario , Útero
3.
Int J Mol Sci ; 23(7)2022 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-35409214

RESUMEN

The uterus is essential for embryo implantation and fetal development. During the estrous cycle, the uterine endometrium undergoes dramatic remodeling to prepare for pregnancy. Angiogenesis is an essential biological process in endometrial remodeling. Steroid hormones regulate the series of events that occur during such remodeling. Researchers have investigated the potential factors, including angiofactors, involved in endometrial remodeling. The Hippo signaling pathway discovered in the 21st century, plays important roles in various cellular functions, including cell proliferation and cell death. However, its role in the endometrium remains unclear. In this review, we describe the female reproductive system and its association with the Hippo signaling pathway, as well as novel Hippo pathway genes and potential target genes.


Asunto(s)
Endometrio , Vía de Señalización Hippo , Implantación del Embrión/fisiología , Endometrio/metabolismo , Ciclo Estral/fisiología , Femenino , Humanos , Embarazo , Útero/metabolismo
4.
Int J Mol Sci ; 23(22)2022 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-36430821

RESUMEN

In mice, zygotic genome activation (ZGA) occurs in two steps: minor ZGA at the one-cell stage and major ZGA at the two-cell stage. Regarding the regulation of gene transcription, minor ZGA is known to have unique features, including a transcriptionally permissive state of chromatin and insufficient splicing processes. The molecular characteristics may originate from extremely open chromatin states in the one-cell stage zygotes, yet the precise underlying mechanism has not been well studied. Recently, the R-loop, a triple-stranded nucleic acid structure of the DNA/RNA hybrid, has been implicated in gene transcription and DNA replication. Therefore, in the present study, we examined the changes in R-loop dynamics during mouse zygotic development, and its roles in zygotic transcription or DNA replication. Our analysis revealed that R-loops persist in the genome of metaphase II oocytes and preimplantation embryos from the zygote to the blastocyst stage. In particular, zygotic R-loop levels dynamically change as development proceeds, showing that R-loop levels decrease as pronucleus maturation occurs. Mechanistically, R-loop dynamics are likely linked to ZGA, as inhibition of either DNA replication or transcription at the time of minor ZGA decreases R-loop levels in the pronuclei of zygotes. However, the induction of DNA damage by treatment with anticancer agents, including cisplatin or doxorubicin, does not elicit genome-wide changes in zygotic R-loop levels. Therefore, our study suggests that R-loop formation is mechanistically associated with the regulation of mouse ZGA, especially minor ZGA, by modulating gene transcription and DNA replication.


Asunto(s)
Estructuras R-Loop , Cigoto , Ratones , Animales , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Cromatina/genética
5.
Int J Mol Sci ; 23(23)2022 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-36499727

RESUMEN

Precise regulation of the cell cycle of embryonic stem cells (ESCs) is critical for their self-maintenance and differentiation. The cell cycle of ESCs differs from that of somatic cells and is different depending on the cell culture conditions. However, the cell cycle regulation in ESCs via epigenetic mechanisms remains unclear. Here, we showed that the ATP-dependent chromatin remodeler Ino80 regulates the cell cycle genes in ESCs under primed conditions. Ino80 loss led to a significantly extended length of the G1-phase in ESCs grown under primed culture conditions. Ino80 directly bound to the transcription start site and regulated the expression of cell cycle-related genes. Furthermore, Ino80 loss induced cell apoptosis. However, the regulatory mechanism of Ino80 in differentiating ESC cycle slightly differed; an extended S-phase was detected in differentiating inducible Ino80 knockout ESCs. RNA-seq analysis of differentiating ESCs revealed that the expression of genes associated with organ development cell cycle is persistently altered in Ino80 knockout cells, suggesting that cell cycle regulation by Ino80 is not limited to undifferentiated ESCs. Therefore, our study establishes the function of Ino80 in ESC cycle via transcriptional regulation, at least partly. Moreover, this Ino80 function may be universal to other cell types.


Asunto(s)
Células Madre Embrionarias de Ratones , Animales , Ratones , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Puntos de Control del Ciclo Celular , Diferenciación Celular/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica
6.
Int J Mol Sci ; 23(17)2022 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-36077170

RESUMEN

The dynamics of uterine endometrium is important for successful establishment and maintenance of embryonic implantation and development, along with extensive cell differentiation and proliferation. The tissue event is precisely and complicatedly regulated as several signaling pathways are involved including two main hormones, estrogen and progesterone signaling. We previously showed a novel signaling molecule, Serine/threonine protein kinase 3/4 (STK3/4), which is responded to hormone in the mouse uterine epithelium. However, the role and regulation of its target, YES-associated protein (YAP) remains unknown. In this study, we investigated the expression and regulation of YAP in mouse endometrium. We found that YAP was periodically expressed in the endometrium during the estrous cycle. Furthermore, periodic expression of YAP was shown to be related to the pathway under hormone treatment. Interestingly, estrogen was shown to positively modulate YAP via endometrial epithelial receptors. In addition, the knockdown of YAP showed that YAP regulated various target genes in endometrial cells. The knockdown of YAP down-regulated numerous targets including ADAMTS1, AMOT, AMOTL1, ANKRD1, CTNNA1, MCL1. On the other hand, the expressions of AREG and AXL were increased by its knockdown. These findings imply that YAP responds via Hippo signaling under various intrauterine signals and is considered to play a role in the expression of factors important for uterine endometrium dynamic regulation.


Asunto(s)
Estrógenos , Proteínas Serina-Treonina Quinasas , Útero/metabolismo , Proteínas Señalizadoras YAP/metabolismo , Animales , Estrógenos/metabolismo , Femenino , Ratones , Progesterona/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal
7.
Int J Mol Sci ; 22(17)2021 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-34502315

RESUMEN

Cluster of differentiation 73 (CD73, also known as ecto-5'-nucleotidase) is an enzyme that converts AMP into adenosine. CD73 is a surface enzyme bound to the outside of the plasma membrane expressed in several cells and regulates immunity and inflammation. In particular, it is known to inhibit T cell-mediated immune responses. However, the regulation of CD73 expression by hormones in the uterus is not yet clearly known. In this study, we investigated the expression of CD73 in ovariectomized mice treated with estrogen or progesterone and its regulation in the mouse uterus during the estrous cycle. The level of CD73 expression was dynamically regulated in the uterus during the estrous cycle. CD73 protein expression was high in proestrus, estrus, and diestrus, whereas it was relatively low in the metestrus stage. Immunofluorescence revealed that CD73 was predominantly expressed in the cytoplasm of the luminal and glandular epithelium and the stroma of the endometrium. The expression of CD73 in ovariectomized mice was gradually increased by progesterone treatment. However, estrogen injection did not affect its expression. Moreover, CD73 expression was increased when estrogen and progesterone were co-administered and was inhibited by the pretreatment of the progesterone receptor antagonist RU486. These findings suggest that the expression of CD73 is dynamically regulated by estrogen and progesterone in the uterine environment, and that there may be a synergistic effect of estrogen and progesterone.


Asunto(s)
5'-Nucleotidasa/metabolismo , Estrógenos/farmacología , Ciclo Estral/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Progesterona/farmacología , Útero/metabolismo , 5'-Nucleotidasa/genética , Animales , Ciclo Estral/efectos de los fármacos , Femenino , Ratones , Ratones Endogámicos ICR , Progestinas/farmacología , Útero/efectos de los fármacos
8.
Int J Mol Sci ; 21(2)2020 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-31936679

RESUMEN

The thermoplasmonic properties of platinum nanoparticles (PtNPs) render them desirable for use in diagnosis, detection, therapy, and surgery. However, their toxicological effects and impact at the molecular level remain obscure. Nanotoxicology is mainly focused on the interactions of nanostructures with biological systems, particularly with an emphasis on elucidating the relationship between the physical and chemical properties such as size and shape. Therefore, we hypothesized whether these unique anisotropic nanoparticles could induce cytotoxicity similar to that of spherical nanoparticles and the mechanism involved. Thus, we synthesized unique and distinct anisotropic PtNPs using lycopene as a biological template and investigated their biological activities in model human acute monocytic leukemia (THP-1) macrophages. Exposure to PtNPs for 24 h dose-dependently decreased cell viability and proliferation. Levels of the cytotoxic markers lactate dehydrogenase and intracellular protease significantly and dose-dependently increased with PtNP concentration. Furthermore, cells incubated with PtNPs dose-dependently produced oxidative stress markers including reactive oxygen species (ROS), malondialdehyde, nitric oxide, and carbonylated protein. An imbalance in pro-oxidants and antioxidants was confirmed by significant decreases in reduced glutathione, thioredoxin, superoxide dismutase, and catalase levels against oxidative stress. The cell death mechanism was confirmed by mitochondrial dysfunction and decreased ATP levels, mitochondrial copy numbers, and PGC-1α expression. To further substantiate the mechanism of cell death mediated by endoplasmic reticulum stress (ERS), we determined the expression of the inositol-requiring enzyme (IRE1), (PKR-like ER kinase) PERK, activating transcription factor 6 (ATF6), and activating transcription factor 4 ATF4, the apoptotic markers p53, Bax, and caspase 3, and the anti-apoptotic marker Bcl-2. PtNPs could activate ERS and apoptosis mediated by mitochondria. A proinflammatory response to PtNPs was confirmed by significant upregulation of interleukin-1-beta (IL-1ß), interferon γ (IFNγ), tumor necrosis factor alpha (TNFα), and interleukin (IL-6). Transcriptomic and molecular pathway analyses of THP-1 cells incubated with the half maximal inhibitory concentration (IC50) of PtNPs revealed the altered expression of genes involved in protein misfolding, mitochondrial function, protein synthesis, inflammatory responses, and transcription regulation. We applied transcriptomic analyses to investigate anisotropic PtNP-induced toxicity for further mechanistic studies. Isotropic nanoparticles are specifically used to inhibit non-specific cellular uptake, leading to enhanced in vivo bio-distribution and increased targeting capabilities due to the higher radius of curvature. These characteristics of anisotropic nanoparticles could enable the technology as an attractive platform for nanomedicine in biomedical applications.


Asunto(s)
Apoptosis/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Inflamación/genética , Leucemia Monocítica Aguda/patología , Nanopartículas del Metal/toxicidad , Platino (Metal)/toxicidad , Transducción de Señal/efectos de los fármacos , Transcriptoma/genética , Adenosina Trifosfato/metabolismo , Anisotropía , Antioxidantes/farmacología , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Leucemia Monocítica Aguda/genética , Peroxidación de Lípido/efectos de los fármacos , Licopeno/química , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Nanopartículas del Metal/ultraestructura , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Óxido Nítrico/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Carbonilación Proteica/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
9.
Mol Reprod Dev ; 86(12): 1993-2004, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31680343

RESUMEN

It has long been known that pig conceptuses produce interferon-γ (IFNG) at the time of implantation, but the role of IFNG and its mechanism of action at the maternal-conceptus interface are not fully understood. Accordingly, we analyzed the expression and regulation of IFNG receptors IFNGR1 and IFNGR2 in the endometrium during the estrous cycle and pregnancy in pigs. Levels of IFNGR1 and IFNGR2 messenger RNA (mRNA) expression changed in the endometrium, with the highest levels during mid pregnancy for IFNGR1 and on Day 12 of pregnancy for IFNGR2. The expression of IFNGR1 and IFNGR2 mRNAs was also detected in conceptuses during early pregnancy and chorioallantoic tissues during mid to late pregnancy. IFNGR1 and IFNGR2 mRNAs were localized to endometrial epithelial and stromal cells and to the chorionic membrane during pregnancy. IFNGR2 protein was also localized to endometrial epithelial and stromal cells, and increased epithelial expression of IFNGR2 mRNA and protein was detectable during early pregnancy than the estrous cycle. Explant culture studies showed that estrogen increased levels of IFNGR2, but not IFNGR1, mRNAs, while interleukin-1ß did not affect levels of IFNGR1 and IFNGR2 mRNAs. Furthermore, IFNG increased levels of IRF1, IRF2, STAT1, and STAT2 mRNAs in the endometrial explants. These results in pigs indicate that IFNGR1 and IFNGR2 are expressed in a stage of pregnancy- and cell-type specific manner in the endometrium and that sequential cooperative action of conceptus signals estrogen and IFNG may be critical for endometrial responsiveness to IFNs for the establishment of pregnancy in pigs.


Asunto(s)
Endometrio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Interferón gamma/biosíntesis , Placenta/metabolismo , Receptores de Interferón/biosíntesis , Transducción de Señal/fisiología , Porcinos/metabolismo , Animales , Femenino , Embarazo , Receptor de Interferón gamma
10.
Cell Physiol Biochem ; 45(5): 2009-2020, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29518769

RESUMEN

BACKGROUND/AIMS: Cyclic adenosine monophosphate (cAMP)-dependent type 2 regulatory subunit beta (Prkar2b) is a regulatory isoform of cAMP-dependent protein kinase (PKA), which is the primary target for cAMP actions. In oocytes, PKA and the pentose phosphate pathway (PPP) have important roles during the germinal vesicle (GV) stage arrest of development. Although the roles of the PKA signal pathway have been studied in the development of oocyte, there has been no report on the function of PRKAR2B, a key regulator of PKA. METHODS: Using reverse transcription polymerase chain reaction (RT-PCR), quantitative real-time PCR (qRT-PCR), immunohistochemistry, and immunofluorescence, we determined the relative expression of Prkar2b in various tissues, including ovarian follicles, during oocyte maturation. Prkar2b-interfering RNA (RNAi) microinjection was conducted to confirm the effect of Prkar2b knockdown, and immunofluorescence, qRT-PCR, and time-lapse video microscopy were used to analyze Prkar2b-deficient oocytes. RESULTS: Prkar2b is strongly expressed in the ovarian tissues, particularly in the growing follicle. During oocyte maturation, the highest expression of Prkar2b was during metaphase I (MI), with a significant decrease at metaphase II (MII). RNAi-mediated Prkar2b suppression resulted in MI-stage arrest during oocyte development, and these oocytes exhibited abnormal spindle formation and chromosome aggregation. Expression of other members of the PKA family (except for Prkaca) were decreased, and the majority of the PPP factors were also reduced in Prkar2b-deficient oocytes. CONCLUSION: These results suggest that Prkar2b is closely involved in the maturation of oocytes by controlling spindle formation and PPP-mediated metabolism.


Asunto(s)
Subunidad RIIbeta de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Interferencia de ARN , Animales , Subunidad RIIbeta de la Proteína Quinasa Dependiente de AMP Cíclico/antagonistas & inhibidores , Subunidad RIIbeta de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Metafase , Ratones , Ratones Endogámicos ICR , Microscopía Fluorescente , Microscopía por Video , Oocitos/crecimiento & desarrollo , Oocitos/metabolismo , Oogénesis , Folículo Ovárico/metabolismo , Folículo Ovárico/patología , ARN Bicatenario/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Imagen de Lapso de Tiempo
11.
Hum Mol Genet ; 24(4): 1127-41, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25305081

RESUMEN

Deciphering the molecular basis of neuronal cell death is a central issue in the etiology of neurodegenerative diseases, such as Parkinson's and Alzheimer's. Dysregulation of p53 levels has been implicated in neuronal apoptosis. The role of histone deacetylase 3 (HDAC3) in suppressing p53-dependent apoptosis has been recently emphasized; however, the molecular basis of modulation of p53 function by HDAC3 remains unclear. Here we show that PTEN-induced putative kinase 1 (PINK1), which is linked to autosomal recessive early-onset familial Parkinson's disease, phosphorylates HDAC3 at Ser-424 to enhance its HDAC activity in a neural cell-specific manner. PINK1 prevents H2O2-induced C-terminal cleavage of HDAC3 via phosphorylation of HDAC3 at Ser-424, which is reversed by protein phosphatase 4c. PINK1-mediated phosphorylation of HDAC3 enhances its direct association with p53 and causes subsequent hypoacetylation of p53. Genetic deletion of PINK1 partly impaired the suppressive role of HDAC3 in regulating p53 acetylation and transcriptional activity. However, depletion of HDAC3 fully abolished the PINK1-mediated p53 inhibitory loop. Finally, ectopic expression of phosphomometic-HDAC3(S424E) substantially overcomes the defective action of PINK1 against oxidative stress in dopaminergic neuronal cells. Together, our results uncovered a mechanism by which PINK1-HDAC3 network mediates p53 inhibitory loop in response to oxidative stress-induced damage.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Histona Desacetilasas/metabolismo , Proteínas Quinasas/metabolismo , Acetilación/efectos de los fármacos , Animales , Caspasa 7/metabolismo , Muerte Celular/genética , Línea Celular , Citoplasma/metabolismo , Neuronas Dopaminérgicas/patología , Activación Enzimática , Histona Desacetilasas/genética , Humanos , Peróxido de Hidrógeno/farmacología , Ratones , Especificidad de Órganos , Fosforilación , Proteínas Quinasas/genética , Proteolisis , Proteína p53 Supresora de Tumor/metabolismo
12.
J Pineal Res ; 63(3)2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28658519

RESUMEN

Premature ovarian failure during chemotherapy is a serious problem for young women with cancer. To preserve the fertility of these patients, approaches to prevent chemotherapy-induced ovarian failure are needed. In a previous study, we reported that melatonin treatment prevents the depletion of the dormant follicle pool via repression of the simultaneous activation of dormant primordial follicles by cisplatin. However, melatonin's protective effect was only partial and thus insufficient. In this study, we found that the hormone ghrelin enhances the protective effect of melatonin against cisplatin-induced ovarian failure in mouse model. Co-administration of melatonin and ghrelin more effectively prevented cisplatin-induced follicle disruption. Simultaneous treatment with melatonin and ghrelin almost restored the number of primordial follicles and the corpus luteum in cisplatin-treated ovaries, compared with single administration. We found melatonin and ghrelin receptors on the cell membrane of premature oocytes of primordial follicles. In addition, melatonin and ghrelin co-administration inhibited the cisplatin-induced phosphorylation of PTEN and FOXO3a that induces cytoplasmic translocation of FOXO3a. Inhibition of FOXO3a phosphorylation by melatonin and ghrelin increased the binding affinity of FOXO3a for the p27Kip1 promoter in primordial follicles. Co-administration of melatonin and ghrelin in cisplatin-treated ovaries restored the expression of p27Kip1 , which is critical for retention of the dormant status of primordial follicles. In conclusion, these findings suggest that melatonin and ghrelin co-administration is suitable for use as a fertoprotective adjuvant therapy during cisplatin chemotherapy in young female cancer patients.


Asunto(s)
Antioxidantes/uso terapéutico , Ghrelina/uso terapéutico , Melatonina/uso terapéutico , Ovario/efectos de los fármacos , Insuficiencia Ovárica Primaria/prevención & control , Animales , Antineoplásicos/efectos adversos , Antioxidantes/farmacología , Cisplatino/efectos adversos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Evaluación Preclínica de Medicamentos , Quimioterapia Combinada , Femenino , Proteína Forkhead Box O3/metabolismo , Ghrelina/farmacología , Humanos , Melatonina/farmacología , Ratones Endogámicos ICR , Ovario/metabolismo , Insuficiencia Ovárica Primaria/inducido químicamente , Receptores de Ghrelina/metabolismo , Receptores de Melatonina/metabolismo
13.
Int J Mol Sci ; 18(6)2017 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-28590419

RESUMEN

Premature ovarian failure is one of the side effects of chemotherapy in pre-menopausal cancer patients. Preservation of fertility has become increasingly important in improving the quality of life of completely recovered cancer patients. Among the possible strategies for preserving fertility such as ovarian tissue cryopreservation, co-treatment with a pharmacological adjuvant is highly effective and poses less of a burden on the human body. Melatonin is generally produced in various tissues and acts as a universally acting antioxidant in cells. Melatonin is now more widely used in various biological processes including treating insomnia and an adjuvant during chemotherapy. In this review, we summarize the information indicating that melatonin may be useful for reducing and preventing premature ovarian failure in chemotherapy-treated female patients. We also mention that many adjuvants other than melatonin are developed and used to inhibit chemotherapy-induced infertility. This information will give us novel insights on the clinical use of melatonin and other agents as fertoprotective adjuvants for female cancer patients.


Asunto(s)
Antineoplásicos/efectos adversos , Preservación de la Fertilidad , Melatonina/farmacología , Neoplasias/complicaciones , Insuficiencia Ovárica Primaria/etiología , Insuficiencia Ovárica Primaria/prevención & control , Sustancias Protectoras/farmacología , Animales , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Antioxidantes/farmacología , Femenino , Humanos , Neoplasias/tratamiento farmacológico , Insuficiencia Ovárica Primaria/metabolismo
14.
Biochim Biophys Acta ; 1853(2): 409-21, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25450970

RESUMEN

The tripartite motif containing (TRIM) proteins are a large family of proteins that have been implicated in many biological processes including cell differentiation, apoptosis, transcriptional regulation, and signaling pathways. Here, we show that TRIM15 co-localized to focal adhesions through homo-dimerization and significantly suppressed cell migration. Domain mapping analysis indicated that B-box2 and PRY domains were essential for TRIM15 localization to focal adhesions and inhibition of cell migration. Our protein-protein interaction screen of TRIM15 with the integrin adhesome identified several TRIM15 interacting proteins including coronin 1B, cortactin, filamin binding LIM protein1, and vasodilator-stimulated phosphoprotein, which are involved in actin cytoskeleton dynamics. TRIM15 expression was tissue-restricted and downregulated in colon cancer. Level of TRIM15 expression was associated with colon cancer cell migration, as well as both in vitro and in vivo tumor growth. These data provide novel insights into the role of TRIM15 as an additional component of the integrin adhesome, regulating cell migration, and suggest that TRIM15 may function as a tumor suppressor of colon cancer.


Asunto(s)
Carcinogénesis/genética , Carcinogénesis/patología , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Proteínas de Unión al ADN/metabolismo , Adhesiones Focales/metabolismo , Actinas/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Cortactina/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Células Epiteliales/metabolismo , Matriz Extracelular/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones Desnudos , Fosforilación , Unión Proteica , Multimerización de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas , Relación Estructura-Actividad
15.
Biochim Biophys Acta ; 1853(10 Pt A): 2722-30, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26239118

RESUMEN

Mitogen-inducible gene 6 (MIG6) is a tumor suppressor implicated in the development of human cancers; however, the regulatory mechanisms of MIG6 remain unknown. Here, using a yeast two-hybrid screen, we identified DnaJ homolog subfamily B member I (DNAJB1) as a novel MIG6-interacting protein. We found that DNAJB1 binds to and decreases MIG6 protein, but not mRNA, levels. DNAJB1 overexpression dosage-dependently decreased MIG6 protein levels. Conversely, DNAJB1 knockdown increased MIG6 protein levels. DNAJB1 destabilizes MIG6 by enhancing K48-linked ubiquitination of MIG6. However, knocking-down of DNAJB1 reduced the ubiquitination of MIG6. DNAJB1 positively regulates the epidermal growth factor receptors (EGFR) signaling pathway via destabilization of MIG6; however, DNAJB1 knockdown diminishes activation of EGFR signaling as well as elevation of MIG6. Importantly, the increased levels of MIG6 by DNAJB1 knockdown greatly enhanced the gefitinib sensitivity in A549 cells. Thus, our study provides a new molecular mechanism to regulate EGFR signaling through modulation of MIG6 by DNAJB1 as a negative regulator.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Receptores ErbB/metabolismo , Proteínas del Choque Térmico HSP40/metabolismo , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/metabolismo , Ubiquitinación/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular Tumoral , Receptores ErbB/genética , Técnicas de Silenciamiento del Gen , Proteínas del Choque Térmico HSP40/genética , Humanos , Unión Proteica , Proteínas Supresoras de Tumor/genética
16.
Biochim Biophys Acta ; 1853(5): 1060-72, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25603536

RESUMEN

Programmed cell death 5 (PDCD5) plays a crucial role in TP53-mediated apoptosis, but the regulatory mechanism of PDCD5 itself during apoptosis remains obscure. We identified YY1-associated factor 2 (YAF2) as a novel PDCD5-interacting protein in a yeast two-hybrid screen for PDCD5-interacting proteins. We found that YY1-associated factor 2 (YAF2) binds to and increases PDCD5 stability by inhibiting the ubiquitin-dependent proteosomal degradation pathway. However, knocking-down of YAF2 diminishes the levels of PDCD5 protein but not the levels of PDCD5 mRNA. Upon genotoxic stress response, YAF2 promotes TP53 activation via association with PDCD5. Strikingly, YAF2 failed to promote TP53 activation in the deletion of PDCD5, whereas restoration of wild-type PDCD5WT efficiently reversed the ineffectiveness of YAF2 on TP53 activation. Conversely, PDCD5 efficiently overcame the knockdown effect of YAF2 on ET-induced TP53 activation. Finally, impaired apoptosis upon PDCD5 ablation was substantially rescued by restoration of PDCD5WT but not YAF2-interacting defective PDCD5E4D nor TP53-interacting defective PDCD5E16D mutant. Our findings uncovered an apoptotic signaling cascade linking YAF2, PDCD5, and TP53 during genotoxic stress responses.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Daño del ADN , Proteínas Musculares/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Represoras/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular Tumoral , Humanos , Leupeptinas/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Modelos Biológicos , Unión Proteica , Multimerización de Proteína , Estabilidad Proteica/efectos de los fármacos , Ubiquitinación
17.
Cell Physiol Biochem ; 40(6): 1289-1302, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27997888

RESUMEN

BACKGROUND: Ras dexamethasone-induced protein (RASD1) is a member of Ras superfamily of small GTPases. RASD1 regulates various signaling pathways involved in iron homeostasis, growth hormone secretion, and circadian rhythm. However, RASD1 function in oocyte remains unknown. METHODS: Using immunohistochemistry, immunofluorescence, and quantitative real-time RT-PCR, RASD1 expression in mouse ovary and RASD1 role in oocyte maturation-related gene expression, spindle formation, and chromosome alignment were analyzed. RNAi microinjection and time-lapse video microscopy were used to examine the effect of Rasd1 knockdown on oocyte maturation. RESULTS: RASD1 was highly detected in oocytes transitioning from primordial to secondary follicles. Rasd1 was highly expressed in germinal vesicle (GV), during GV breakdown, and in metaphase I (MI) stage as oocytes mature, and its expression was significantly downregulated in MII stage. With knockdown of Rasd1, maturation in GV oocytes was arrested at MI stage, showing disrupted meiotic spindling and chromosomal misalignment. In addition, Obox4 and Arp2/3, engaged in MI-MII transition and cytokinesis, respectively, were misregulated in GV oocytes by Rasd1 knockdown. CONCLUSION: These findings suggest that RASD1 is a novel factor in MI-MII oocyte transition and may be involved in regulating the progression of cytokinesis and spindle formation, controlling related signaling pathways during oocyte maturation.


Asunto(s)
Diferenciación Celular , Técnicas de Silenciamiento del Gen , Oocitos/citología , Oocitos/metabolismo , Proteínas ras/genética , Animales , Diferenciación Celular/genética , Cromosomas de los Mamíferos/metabolismo , Citocinesis , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Metafase/genética , Ratones Endogámicos ICR , Especificidad de Órganos/genética , Interferencia de ARN , Huso Acromático , Proteínas ras/metabolismo
18.
Biochem Biophys Res Commun ; 472(3): 489-95, 2016 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-26940739

RESUMEN

The sine oculis homeobox 1 (SIX1) is a member of the Six gene family. SIX1 is involved in tissue development by regulating proliferation, apoptosis, and differentiation. However, function of SIX1 in the uterus remains unknown. Here, we found that Six1 expression is regulated along the estrous cycle in mouse uterus. Six1 expression was significantly increased at estrus stage and decreased at the rest of stages. SIX1 is detected in the luminal and glandular epithelium of uterine endometrium at the estrus stage. Estrogen injection increased Six1 expression in the ovariectomized mouse uterus, whereas progesterone had no effect on its expression. Estrogen receptor antagonist inhibited estrogen-induced Six1 expression. Our findings imply that SIX1 may play a role as an important regulator to orchestrate the dynamic of uterine endometrium in response to estrogen level during the estrous cycle. These results will give us a better understanding of uterine biology.


Asunto(s)
Estrógenos/metabolismo , Ciclo Estral/fisiología , Proteínas de Homeodominio/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Femenino , Regulación de la Expresión Génica/fisiología , Ratones , Ratones Endogámicos ICR
19.
J Pineal Res ; 60(3): 336-47, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26882203

RESUMEN

Premature ovarian failure (POF) is a major side effect of chemotherapy in young cancer patients. To develop pharmaceutical agents for preserving fertility, it is necessary to understand the mechanisms responsible for chemotherapy-induced follicle loss. Here, we show that treatment with cisplatin, a widely used anticancer drug, depleted the dormant follicle pool in mouse ovaries by excessive activation of the primordial follicles, without inducing follicular apoptosis. Moreover, we show that co-treatment with the antioxidant melatonin prevented cisplatin-induced disruption of the follicle reserve. We quantified the various stages of growing follicles, including primordial, primary, secondary, and antral, to demonstrate that cisplatin treatment alone significantly decreased, whereas melatonin co-treatment preserved, the number of primordial follicles in the ovary. Importantly, analysis of the PTEN/AKT/FOXO3a pathway demonstrated that melatonin significantly decreased the cisplatin-mediated inhibitory phosphorylation of PTEN, a key negative regulator of dormant follicle activation. Moreover, melatonin prevented the cisplatin-induced activating phosphorylation of AKT, GSK3ß, and FOXO3a, all of which trigger follicle activation. Additionally, we show that melatonin inhibited the cisplatin-induced inhibitory phosphorylation and nuclear export of FOXO3a, which is required in the nucleus to maintain dormancy of the primordial follicles. These findings demonstrate that melatonin attenuates cisplatin-induced follicle loss by preventing the phosphorylation of PTEN/AKT/FOXO3a pathway members; thus, melatonin is a potential therapeutic agent for ovarian protection and fertility preservation during chemotherapy in female cancer patients.


Asunto(s)
Cisplatino/efectos adversos , Proteína Forkhead Box O3/metabolismo , Melatonina/farmacología , Folículo Ovárico/metabolismo , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Cisplatino/farmacología , Femenino , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Ratones , Folículo Ovárico/patología
20.
Asian-Australas J Anim Sci ; 29(7): 944-51, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26954166

RESUMEN

Tdrd12 is one of tudor domain containing (Tdrd) family members. However, the expression pattern of Tdrd12 has not been well studied. To compare the expression levels of Tdrd12 in various tissues, real time-polymerase chain reaction was performed using total RNAs from liver, small intestine, heart, brain, kidney, lung, spleen, stomach, uterus, ovary, and testis. Tdrd12 mRNA was highly expressed in testis. Antibody against mouse TDRD12 were generated using amino acid residues SQRPNEKPLRLTEKKDC of TDRD12 to investigate TDRD12 localization in testis. Immunostaining assay shows that TDRD12 is mainly localized at the spermatid in the seminiferous tubules of adult testes. During postnatal development, TDRD12 is differentially expressed. TDRD12 was detected in early spermatocytes at 2 weeks and TDRD12 was localized at acrosome of the round spermatids. TDRD12 expression was not co-localized with TDRD1 which is an important component of piRNA pathway in germ cells. Our results indicate that TDRD12 may play an important role in spermatids and function as a regulator of spermatogenesis in dependent of TDRD1.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA