Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(23)2022 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-36499089

RESUMEN

Non-obstructive azoospermia is a major clinical issue associated with male infertility that remains to be addressed. Although neogenin is reportedly abundantly expressed in the testis, its role in mammalian spermatogenesis is unknown. We systematically investigated the role of neogenin during spermatogenesis by performing loss-of-function studies. Testis-specific neogenin conditional knock-out (cKO) mice were generated using CRISPR/Cas9 and neogenin-targeting guide RNAs. We analyzed the expression profiles of germ cell factors by RT-PCR and Western blotting. Neogenin localized mainly to spermatogonia in seminiferous tubules of mouse testes. RT-PCR and Western blot analyses further demonstrated that neogenin expression varied during spermatogenesis and was dramatically increased at postnatal day 12-25 during the pubertal stage. In neogenin-cKO mouse testes, the ratio of primary and secondary spermatocytes was significantly decreased compared with the control, while the number of apoptotic testicular cells was significantly increased. Taken together, these results suggest that neogenin plays a pivotal role in the maintenance and proliferation of spermatogonia during the early stage of spermatogenesis in mice.


Asunto(s)
Espermatogénesis , Espermatogonias , Humanos , Masculino , Ratones , Animales , Regulación hacia Abajo , Espermatogonias/metabolismo , Espermatogénesis/genética , Testículo/metabolismo , Diferenciación Celular/genética , Ratones Noqueados , Proliferación Celular , Mamíferos
2.
Int J Mol Sci ; 22(10)2021 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-34064719

RESUMEN

Inflammation is a major cause of several chronic diseases and is reported to be recovered by the immuno-modulation of mesenchymal stem cells (MSCs). While most studies have focussed on the anti-inflammatory roles of MSCs in stem cell therapy, the impaired features of MSCs, such as the loss of homeostasis by systemic aging or pathologic conditions, remain incompletely understood. In this study, we investigated whether the altered phenotypes of human placenta-derived MSCs (hPD-MSCs) exposed to inflammatory cytokines, including TNF-α and IFN-γ, could be protected by MIT-001, a small anti-inflammatory and anti-necrotic molecule. MIT-001 promoted the spindle-like shape and cytoskeletal organization extending across the long cell axis, whereas hPD-MSCs exposed to TNF-α/IFN-γ exhibited increased morphological heterogeneity with an abnormal cell shape and cytoskeletal disorganization. Importantly, MIT-001 improved mitochondrial distribution across the cytoplasm. MIT-001 significantly reduced basal respiration, ATP production, and cellular ROS levels and augmented the spare respiratory capacity compared to TNF-α/IFN-γ-exposed hPD-MSCs, indicating enhanced mitochondrial quiescence and homeostasis. In conclusion, while TNF-α/IFN-γ-exposed MSCs lost homeostasis and mitochondrial quiescence by becoming over-activated in response to inflammatory cytokines, MIT-001 was able to rescue mitochondrial features and cellular phenotypes. Therefore, MIT-001 has therapeutic potential for clinical applications to treat mitochondrion-related inflammatory diseases.


Asunto(s)
Citoesqueleto/fisiología , Células Madre Mesenquimatosas/fisiología , Mitocondrias/fisiología , Compuestos Orgánicos/farmacología , Placenta/citología , Citoesqueleto/efectos de los fármacos , Femenino , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Consumo de Oxígeno , Placenta/efectos de los fármacos , Placenta/metabolismo , Embarazo , Especies Reactivas de Oxígeno/metabolismo
3.
Int J Mol Sci ; 22(11)2021 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-34200157

RESUMEN

The purpose of this study was to investigate whether polymorphisms in five microRNAs (miRNAs), miR-604A>G, miR-608C>G, 631I/D, miR-938G>A, and miR-1302-3C>T, are associated with the risk of idiopathic recurrent pregnancy loss (RPL). Blood samples were collected from 388 patients with idiopathic RPL (at least two consecutive spontaneous abortions) and 227 control participants. We found the miR-604 AG and AG + GG genotypes of miR-604, the miR-938 GA and GA + AA genotypes of miR-938, and the miR-1302-3CT and CT + TT genotypes of miR-1302-3 are less frequent than the wild-type (WT) genotypes, miR-604AA, miR-938GG, and miR-1302-3CC, respectively, in RPL patients. Using allele-combination multifactor dimensionality reduction (MDR) analysis, we found that eight haplotypes conferred by the miR-604/miR-608/miR-631/miR-938/miR-1302-3 allele combination, A-C-I-G-T, A-C-I-A-C, G-C-I-G-C, G-C-I-G-T, G-G-I-G-C, G-G-I-G-T, G-G-I-A-C, G-G-D-G-C, three from the miR-604/miR-631/miR-938/miR-1302-3 allele combination, A-I-G-T, G-I-G-C, G-I-A-T, one from the miR-604/miR-631/miR-1302-3 allele combination, G-I-C, and two from the miR-604/miR-1302-3 allele combination, G-C and G-T, were less frequent in RPL patients, suggesting protective effects (all p < 0.05). We also identified the miR-604A>G and miR-938G>A polymorphisms within the seed sequence of the mature miRNAs and aligned the seed sequences with the 3'UTR of putative target genes, methylenetetrahydrofolate reductase (MTHFR) and gonadotropin-releasing hormone receptor (GnRHR), respectively. We further found that the binding affinities between miR-604/miR-938 and the 3'UTR of their respective target genes (MTHFR, GnRHR) were significantly different for the common (miR-604A, miR-938G) and variant alleles (miR-604G, miR-938A). These results reveal a significant association between the miR-604A>G and miR-938G>A polymorphisms and idiopathic RPL and suggest that miRNAs can affect RPL in Korean women.


Asunto(s)
Aborto Habitual/patología , Predisposición Genética a la Enfermedad , MicroARNs/genética , Polimorfismo de Nucleótido Simple , Regiones no Traducidas 3' , Aborto Habitual/etiología , Adulto , Estudios de Casos y Controles , Implantación del Embrión , Femenino , Estudios de Asociación Genética , Genotipo , Humanos , Embarazo
4.
Cell Physiol Biochem ; 45(1): 37-53, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29316553

RESUMEN

BACKGROUND/AIMS: Previously, we found that silencing of growth arrest-specific gene 6 (Gas6) in oocytes impaired cytoplasmic maturation, resulting in failure of sperm chromatin decondensation (SCD) and pronuclear (PN) formation after fertilization. Thus, we conducted this study to determine the effect of Gas6 RNAi on downstream genes and to elucidate the working mechanism of Gas6 on oocyte cytoplasmic maturation and SCD. METHODS: Using RT-PCR, Western blot and immunofluorescence, the expression levels of various target genes and the localization of heparan sulfate (HS) were analyzed after Gas6 RNAi. The roles of Gas6 in HS biosynthesis, production of ATP and GSH, ROS generation and ΔΨm were also investigated. SCD and micrococcal nuclease (MNase) analyses were used to examine the effects of HS on the open chromatin state in sperm and somatic cell nuclei, respectively. RESULTS: Disruption of Gas6 expression led to the inhibition of HS biosynthesis through the reduction of several HS biosynthetic enzymes. The rescue experiment, HS treatment in vitro, significantly recovered SCD and PN formation, confirming that HS had the ability to induce sperm head remodeling during fertilization. Interestingly, excessive mitochondrial activation in Gas6-depleted MII oocytes caused ROS generation and glutathione (GSH) degradation via mitochondrial activation, such as elevated ΔΨm and ATP production. Indeed, HS-treated NIH3T3 cell nuclei showed an open chromatin state, as determined by diffuse DAPI staining and increased sensitivity to MNase. CONCLUSION: We propose that the addition of HS to sperm and/or oocyte maturation would improve the efficiency of in vitro fertilization and somatic cell nuclear transfer (SCNT) reprogramming.


Asunto(s)
Ensamble y Desensamble de Cromatina/efectos de los fármacos , Cromatina/metabolismo , Citoplasma/metabolismo , Heparitina Sulfato/farmacología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Animales , Núcleo Celular/metabolismo , Cromatina/química , Cromatina/efectos de los fármacos , Femenino , Fertilización In Vitro , Glutatión/metabolismo , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos ICR , Microscopía Confocal , Células 3T3 NIH , Oocitos/metabolismo , Interferencia de ARN , ARN Bicatenario/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Espermatozoides/efectos de los fármacos , Espermatozoides/fisiología
5.
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
6.
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
7.
Int J Mol Sci ; 18(6)2017 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-28604625

RESUMEN

MicroRNAs (miRNAs) post-transcriptionally regulate gene expression in animals and plants. The aim of this study was to investigate whether polymorphisms in miR-938 are associated with the risk of primary ovarian insufficiency (POI) and POI-related target gene regulation. We identified the miR-938G>A polymorphisms within the seed sequence of mature miRNA and aligned the seed sequence with the 3' untranslated region (UTR) of the gonadotropin-releasing hormone receptor (GnRHR) mRNA, a miR-938 target gene. We found that the binding of miR-938 to the 3'-UTR of GnRHR mRNA was significantly different between normal and variant alleles. Our data suggests that the dysregulation of miR-938G>A influences the binding to GnRHR and that miR-938G>A polymorphisms might contribute to regulation of POI-related target genes.


Asunto(s)
Predisposición Genética a la Enfermedad , MicroARNs/genética , Polimorfismo de Nucleótido Simple , Insuficiencia Ovárica Primaria/metabolismo , Receptores LHRH/genética , Regiones no Traducidas 3' , Adulto , Femenino , Regulación de la Expresión Génica , Humanos , MicroARNs/metabolismo , Insuficiencia Ovárica Primaria/genética , Receptores LHRH/metabolismo
8.
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
9.
Acta Neuropathol ; 132(4): 577-92, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27221146

RESUMEN

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder of the central nervous system (CNS) that is defined by a CAG expansion in exon 1 of the huntingtin gene leading to the production of mutant huntingtin (mHtt). To date, the disease pathophysiology has been thought to be primarily driven by cell-autonomous mechanisms, but, here, we demonstrate that fibroblasts derived from HD patients carrying either 72, 143 and 180 CAG repeats as well as induced pluripotent stem cells (iPSCs) also characterized by 143 CAG repeats can transmit protein aggregates to genetically unrelated and healthy host tissue following implantation into the cerebral ventricles of neonatal mice in a non-cell-autonomous fashion. Transmitted mHtt aggregates gave rise to both motor and cognitive impairments, loss of striatal medium spiny neurons, increased inflammation and gliosis in associated brain regions, thereby recapitulating the behavioural and pathological phenotypes which characterizes HD. In addition, both in vitro work using co-cultures of mouse neural stem cells with 143 CAG fibroblasts and the SH-SY5Y human neuroblastoma cell line as well as in vivo experiments conducted in newborn wild-type mice suggest that exosomes can cargo mHtt between cells triggering the manifestation of HD-related behaviour and pathology. This is the first evidence of human-to-mouse prion-like propagation of mHtt in the mammalian brain; a finding which will help unravel the molecular bases of HD pathology as well as to lead to the development of a whole new range of therapies for neurodegenerative diseases of the CNS.


Asunto(s)
Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Células Madre Pluripotentes Inducidas/citología , Proteínas Mutantes/metabolismo , Neuronas/citología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Niño , Modelos Animales de Enfermedad , Humanos , Enfermedad de Huntington/terapia , Células Madre Pluripotentes Inducidas/patología , Masculino , Ratones , Neuronas/patología
10.
Reproduction ; 151(4): 369-78, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26762402

RESUMEN

Mouse oocytes begin to mature in vitro once liberated from ovarian follicles. Previously, we showed that oocyte-specific homeobox 4 (Obox4) is critical for maintaining the intact nuclear membrane of the germinal vesicle (GV) in oocytes and for completing meiosis at the metaphase I-II (MI-MII) transition. This study further examines the molecular mechanisms of OBOX4 in regulating GV nuclear membrane breakdown. Maturation-promoting factor (MPF) and MAPK are normally inactive in GV stage oocytes but were activated prematurely in arrested GV stage oocytes by 3-isobutyl-1-metyl-xanthine (IBMX) in vitro after Obox4 RNA interference (RNAi). Furthermore, signal transducer and activator of transcription 3 (STAT3) was significantly activated by Obox4 RNAi. We confirmed that this Obox4 RNAi-induced premature STAT3 and MPF/MAPK activation at the GV stage provoked subsequent GV breakdown (GVBD) despite the opposing force of high cAMP in the IBMX-supplemented medium to maintain intact GV. When cumulus-oocyte complexes were exposed to interferon α (IFNA), a STAT3 activator, oocytes matured and cumulus cells expanded to resume nuclear maturation in IBMX-supplemented medium, suggesting that STAT3 activation is sufficient for stimulating the continuation of meiosis. Using Stattic, a specific STAT3 inhibitor, we confirmed that GVBD involves STAT3 activation in Obox4-silenced oocytes. Based on these findings, we concluded that i) Obox4 is an important upstream regulator of MPF/MAPK and STAT3 signaling, and ii) Obox4 is a key regulator of the GV arrest mechanism in oocytes.


Asunto(s)
Proteínas Ligadas a GPI/metabolismo , Silenciador del Gen , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Membrana Nuclear/metabolismo , Oocitos/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Western Blotting , Núcleo Celular/metabolismo , Células Cultivadas , Femenino , Técnica del Anticuerpo Fluorescente , Proteínas Ligadas a GPI/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Técnicas para Inmunoenzimas , Meiosis/fisiología , Mesotelina , Ratones , Proteínas Quinasas Activadas por Mitógenos/genética , Oocitos/citología , Folículo Ovárico/citología , Folículo Ovárico/metabolismo , Fosforilación , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética
11.
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
12.
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.

13.
Cytotherapy ; 17(8): 1090-103, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26031742

RESUMEN

BACKGROUND AIMS: Adipose-derived mesenchymal stromal cells (AD-MSCs) have high proliferative capacity and ability to secrete trophic factors. Although intra-arterial (IA) transplantation of stem cells induces efficient engraftment to the host brain, it is unclear whether engrafted cells exert their long-term therapeutic effects through a bystander mechanism or a cell replacement mechanism. METHODS: After induction of ischemia in rats by middle cerebral artery occlusion, we transplanted human AD-MSCs into their carotid arteries with the use of a micro-needle, and we then investigated the therapeutic effects during the early and late phases of ischemia by means of in vivo magnetic resonance imaging, functional and histological analyses. RESULTS: During the early phase of cerebral ischemia, IA transplantation of AD-MSCs attenuated inflammation and enhanced endogenous neurogenesis. Transplanted animals showed a marked improvement in functional tests during the early phase of cerebral ischemia that was less prominent but still significant during the late phase of cerebral ischemia. Although the transplanted cells effectively migrated to the infarct area, only a small number of engrafted cells survived at 8 weeks after transplantation and differentiated into neuronal, glial and endothelial cells. CONCLUSIONS: IA transplantation of human AD-MSCs provides an effective therapeutic modality in a rodent model of stroke, of which the main effects are mediated by a bystander mechanism at the early phase of ischemia.


Asunto(s)
Isquemia Encefálica/cirugía , Infarto de la Arteria Cerebral Media/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Neuroprotección/fisiología , Accidente Cerebrovascular/cirugía , Tejido Adiposo/citología , Adulto , Animales , Efecto Espectador , Diferenciación Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Células Madre Mesenquimatosas/citología , Neurogénesis/fisiología , Ratas , Ratas Sprague-Dawley , Adulto Joven
14.
Arch Gynecol Obstet ; 291(2): 347-54, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25115280

RESUMEN

PURPOSE: The present study aims to evaluate whether multiplex ligation-dependent probe amplification (MLPA) technique with subtelomeric probes is to be an alternative method of routine G-banding chromosome analysis from pregnancy loss. METHODS: A review of 5 years (from 2005 to 2009) of karyotype for products of conception (POCs) was carried out. From June 2010 to June 2012, MLPA was performed in parallel with karyotype analysis on 347 miscarriages. Karyotyped miscarriages served as controls in this blinded study. Abnormal results were confirmed by fluorescence in situ hybridization. RESULTS: A review of 5 years of karyotype results for POCs indicated that 11.46 % of cases failed to karyotyping. In the study periods, MLPA results were successfully obtained from all cases including 51 (14.7 %) culture failed cases, chromosomal abnormalities were detected in 27 (52.9 %) of cases which failed to grow or could not be cultivated. It took 3 weeks by conventional karyotyping, but it required at least 24 h and at most a week by MLPA from tissue sampling to final reporting. 47 cases showed discordant results between karyotyping and MLPA because of maternal cell contamination, polyploidy, mosaicism, or balanced translocation. CONCLUSIONS: MLPA technique is relatively low cost, less labor intensive and reduces waiting time with high accuracy compared with conventional cytogenetic analysis. Therefore, MLPA can be the first approach for chromosome analysis from pregnancy loss.


Asunto(s)
Aborto Espontáneo/genética , Aberraciones Cromosómicas , Hibridación Fluorescente in Situ/métodos , Reacción en Cadena de la Polimerasa Multiplex/métodos , Estudios Transversales , Método Doble Ciego , Femenino , Humanos , Cariotipo , Cariotipificación , Masculino , Mosaicismo , Técnicas de Amplificación de Ácido Nucleico/métodos , Poliploidía , Embarazo , Estudios Prospectivos , Estudios Retrospectivos
15.
Stem Cells ; 30(8): 1703-13, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22696466

RESUMEN

Recent studies suggest that epigenetic modifications, such as DNA methylation and histone modification, can alter the differentiation potential of stem cells or progenitor cells. Specifically, coactivator-associated arginine methyltransferase 1 (CARM1) is known to act as a coactivator for various transcription factors and to regulate gene expression by chromatin remodeling through histone methylation. Here, for the first time, we have used direct protein delivery of CARM1 using cell-penetrating peptide (CPP) to regulate the differentiation potential of human mesenchymal stem cells (hMSCs). Immunofluorescence showed that the CPP-CARM1 protein is successfully delivered into the nuclei of hMSCs. Further experiments using immunofluorescence and Western blotting showed that the delivered CARM1 protein can effectively methylate the arginine 17 residue of histone H3 in both bone marrow (BM)- and adipose-derived (AD)-hMSCs, thus suggesting that the CARM1 protein delivered by the CPP system is biologically active in hMSCs. Chromatin immunoprecipitation (ChIP) assay and genome-wide gene expression profiling supported the result that delivered CARM1 protein can cause chromatin remodeling through histone methylation. Finally, the CPP-CARM1 protein efficiently elevated the differentiation efficiency of BM-hMSCs and AD-hMSCs into adipogenic, osteogenic, and myogenic cell lineages in vitro. Altered expression of critical genes after hMSC differentiation was reconfirmed by real-time reverse transcription polymerase chain reaction (qRT-PCR). Collectively, our results suggest that CPP-CARM1 can elevate the differentiation potential of hMSCs into various cell types, and that this system using CPP is a useful tool for exogenous protein delivery in clinical applications of cell-based therapy.


Asunto(s)
Péptidos de Penetración Celular/metabolismo , Células Madre Mesenquimatosas/citología , Proteína-Arginina N-Metiltransferasas/metabolismo , Diferenciación Celular/fisiología , Péptidos de Penetración Celular/genética , Expresión Génica , Humanos , Células Madre Mesenquimatosas/enzimología , Células Madre Mesenquimatosas/metabolismo , Análisis por Micromatrices , Regiones Promotoras Genéticas , Proteína-Arginina N-Metiltransferasas/genética , Transcripción Genética
16.
Cells ; 11(7)2022 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-35406698

RESUMEN

Millions of people around the world suffer from infertility, with the number of infertile couples and individuals increasing every year. Assisted reproductive technologies (ART) have been widely developed in recent years; however, some patients are unable to benefit from these technologies due to their lack of functional germ cells. Therefore, the development of alternative methods seems necessary. One of these methods is to create artificial oocytes. Oocytes can be generated in vitro from the ovary, fetal gonad, germline stem cells (GSCs), ovarian stem cells, or pluripotent stem cells (PSCs). This approach has raised new hopes in both basic research and medical applications. In this article, we looked at the principle of oocyte development, the landmark studies that enhanced our understanding of the cellular and molecular mechanisms that govern oogenesis in vivo, as well as the mechanisms underlying in vitro generation of functional oocytes from different sources of mouse and human stem cells. In addition, we introduced next-generation ART using somatic cells with artificial oocytes. Finally, we provided an overview of the reproductive application of in vitro oogenesis and its use in human fertility.


Asunto(s)
Infertilidad , Células Madre Pluripotentes , Femenino , Células Germinativas/fisiología , Humanos , Oocitos/fisiología , Oogénesis/fisiología , Ovario/fisiología , Células Madre Pluripotentes/fisiología
17.
Cell Prolif ; 55(7): e13274, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35698260

RESUMEN

OBJECTIVES: Patient-derived induced pluripotent stem cells (iPSCs) are materials that can be used for autologous stem cell therapy. We screened mtDNA mutations in iPSCs and iPSC-derived neuronal cells from patients with Alzheimer's disease (AD). Also, we investigated whether the mutations could affect mitochondrial function and deposition of ß-amyloid (Aß) in differentiated neuronal cells. MATERIALS AND METHODS: mtDNA mutations were measured and compared among iPSCs and iPSC-derived neuronal cells. The selected iPSCs carrying mtDNA mutations were subcloned, and then their growth rate and neuronal differentiation pattern were analyzed. The differentiated cells were measured for mitochondrial respiration and membrane potential, as well as deposition of Aß. RESULTS: Most iPSCs from subjects with AD harbored ≥1 mtDNA mutations, and the number of mutations was significantly higher than that from umbilical cord blood. About 35% and 40% of mutations in iPSCs were shared with isogenic iPSCs and their differentiated neuronal precursor cells, respectively, with similar or different heteroplasmy. Furthermore, the mutations in clonal iPSCs were stable during extended culture and neuronal differentiation. Finally, mtDNA mutations could induce a growth advantage with higher viability and proliferation, lower mitochondrial respiration and membrane potential, as well as increased Aß deposition. CONCLUSION: This study demonstrates that mtDNA mutations in patients with AD could lead to mitochondrial dysfunction and accelerated Aß deposition. Therefore, early screening for mtDNA mutations in iPSC lines would be essential for developing autologous cell therapy or drug screening for patients with AD.


Asunto(s)
Enfermedad de Alzheimer , Genoma Mitocondrial , Células Madre Pluripotentes Inducidas , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Diferenciación Celular/genética , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Genoma Humano , Humanos , Mutación/genética
18.
J Proteome Res ; 10(1): 269-76, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21047053

RESUMEN

Small cell lung cancer (SCLC) is the leading cause of cancer death, with a high propensity for aggressiveness and metastasis even in an early stage. Thus, identification of biomarkers as early diagnostics and treatment is needed. In this study, we investigated differentially regulated proteins between human SCLC tissues and normal bronchial epithelium by proteomic analysis using two-dimensional electrophoresis (2-DE) and MALDI-TOF mass spectrometry. Seven proteins and protein isoforms, including, γ-actin, tubulin α-1B, laminin B1, coactosin-like protein-1 (COTL-1), ubiquitin carboxyl-terminal esterase L1, ubiquitin-conjugating enzyme E2-25K, and carbonic anhydrase 1, were up-regulated more than 2 fold in SCLC tissues. In particular, up-regulated COTL-1 expression was validated by Western blot analysis, immunohistochemistry, and reverse transcription quantitative polymerase chain reaction (RT-qPCR). Moreover, most SCLC tissues (93%; 28/30) were COTL-1-positive in immunohistochemistry, whereas only 16% (10/64) of nonsmall cell lung cancer (NSLC) tissues were. Taken together, this SCLC proteomic data may help in establishing a human SCLC proteome database. COTL-1 may be a biomarker or a therapeutic target in SCLC patients.


Asunto(s)
Biomarcadores de Tumor/química , Neoplasias Pulmonares/metabolismo , Proteínas de Microfilamentos/química , Proteómica/métodos , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Bronquios/citología , Electroforesis en Gel Bidimensional , Femenino , Humanos , Inmunohistoquímica , Masculino , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Persona de Mediana Edad , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Reproducibilidad de los Resultados , Mucosa Respiratoria/química , Mucosa Respiratoria/citología , Mucosa Respiratoria/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Regulación hacia Arriba
19.
Biochem Biophys Res Commun ; 415(4): 567-72, 2011 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-22062548

RESUMEN

MicroRNAs (miRNAs) are small non-coding RNAs that regulate diverse biological processes. We cloned novel small RNA from human mesenchymal stem cells (hMSCs) and termed microRNA-5787 (hsa-miR-5787) that met the criteria for a miRNA. The level of miR-5787 was elevated in senescent fibroblasts. Based on the target prediction algorithm and results that were obtained, we find that eukaryotic translation initiation factor 5 (eIF5) is a target of miR-5787. Similar to the over-expression of miR-5787, we showed that repression of eIF5 in fibroblasts negatively affected cell growth. Therefore, we propose that the miR-5787 represses cell growth, in part, by targeting eIF5.


Asunto(s)
Proliferación Celular , MicroARNs/metabolismo , Factores de Iniciación de Péptidos/genética , Proteínas de Unión al ARN/genética , Células Cultivadas , Senescencia Celular/genética , Fibroblastos/metabolismo , Fibroblastos/fisiología , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , Regulación hacia Arriba , Factor 5A Eucariótico de Iniciación de Traducción
20.
Mitochondrion ; 58: 135-146, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33639272

RESUMEN

Mesenchymal stem cells (MSCs) are multipotent cells with critical roles in homeostasis and regeneration. MSCs undergo aging in response to various stresses, and this causes many diseases including degenerative disorders. Thus, regulation of aging factors is crucial for healthy aging. Mitochondrial open reading frame of the 12S rRNA-c (MOTS-c) was recently reported to regulate metabolic homeostasis. Here, we investigated the restorative effects of MOTS-c on aged human placenta-derived MSCs (hPD-MSCs). MOTS-c promoted the morphology of old hPD-MSCs. MOTS-c significantly activated AMP-activated protein kinase, which is the main target pathway of MOTS-c, and inhibited its antagonistic effector mTORC1. MOTS-c considerably enhanced mitochondrial homeostasis by decreasing oxygen consumption and reactive oxygen species production. The mitochondrial state of MOTS-c-treated old hPD-MSCs was more similar to that of young hPD-MSCs than the mitochondrial state of non-treated old hPD-MSCs. MOTS-c also decreased lipid synthesis. In conclusion, we demonstrated that MOTS-c promotes homeostasis in aged hPD-MSCs.


Asunto(s)
Homeostasis/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Proteínas Mitocondriales/farmacología , Placenta/efectos de los fármacos , Femenino , Humanos , Técnicas In Vitro , Células Madre Mesenquimatosas/metabolismo , Mitocondrias/metabolismo , Placenta/citología , Placenta/metabolismo , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA