Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Intervalo de año de publicación
1.
J Mol Cell Biol ; 15(7)2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-37533201

RESUMEN

The effective proliferation and differentiation of trophoblast stem cells (TSCs) is indispensable for the development of the placenta, which is the key to maintaining normal fetal growth during pregnancy. Kruppel-like factor 5 (Klf5) is implicated in the activation of pluripotency gene expression in embryonic stem cells (ESCs), yet its function in TSCs is poorly understood. Here, we showed that Klf5 knockdown resulted in the downregulation of core TSC-specific genes, consequently causing rapid differentiation of TSCs. Consistently, Klf5-depleted embryos lost the ability to establish TSCs in vitro. At the molecular level, Klf5 preferentially occupied the proximal promoter regions and maintained an open chromatin architecture of key TSC-specific genes. Deprivation of Klf5 impaired the enrichment of p300, a major histone acetyl transferase of H3 lysine 27 acetylation (H3K27ac), and further reduced the occupancy of H3K27ac at promoter regions, leading to decreased transcriptional activity of TSC pluripotency genes. Thus, our findings highlight a novel mechanism of Klf5 in regulating the self-renewal and differentiation of TSCs and provide a reference for understanding placental development and improving pregnancy rates.


Asunto(s)
Placenta , Factores de Transcripción , Femenino , Embarazo , Humanos , Placenta/metabolismo , Factores de Transcripción/metabolismo , Trofoblastos/metabolismo , Diferenciación Celular/genética , Células Madre Embrionarias/metabolismo
2.
J Cell Physiol ; 238(12): 2855-2866, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37942811

RESUMEN

The regulatory network between signaling pathways and transcription factors (TFs) is crucial for the maintenance of pluripotent stem cells. However, little is known about how the key TF OCT4 coordinates signaling pathways to regulate self-renewal and lineage differentiation of porcine pluripotent stem cells (pPSCs). Here, we explored the function of OCT4 in pPSCs by transcriptome and chromatin accessibility analysis. The TFs motif enrichment analysis revealed that, following OCT4 knockdown, the regions of increased chromatin accessibility were enriched with EOMES, GATA6, and FOXA1, indicating that pPSCs differentiated toward the mesoendoderm (ME) lineage. Besides, pPSCs rapidly differentiated into ME when the WNT/ß-catenin inhibitor XAV939 was removed. However, the ME differentiation of pPSCs caused by OCT4 knockdown did not rely on the activation of WNT/ß-catenin signaling because the target gene of WNT/ß-catenin signaling, AXIN2 was not upregulated after OCT4 knockdown, despite significant upregulation of WLS and some WNT ligands. Importantly, OCT4 is directly bound to the promoter and enhancers of EOMES and repressed its transcription. Overexpression of EOMES was sufficient to induce ME differentiation in the presence of XAV939. These results demonstrate that OCT4 can regulate WNT/ß-catenin signaling and prevent ME differentiation of pPSCs by repressing EOMES transcription.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes , Vía de Señalización Wnt , Animales , beta Catenina/genética , beta Catenina/metabolismo , Diferenciación Celular/genética , Cromatina/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Porcinos , Vía de Señalización Wnt/genética , Proteínas de Dominio T Box/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Línea Celular
3.
J Ethnobiol Ethnomed ; 19(1): 40, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37710305

RESUMEN

BACKGROUND: The Luchuan pig is an indigenous breed from Luchuan County, China, with cultural and genetic significance. However, traditional knowledge and conservation status have not been systematically documented. METHODS: Using ethnobiological methods, we surveyed 72 Luchuan pig farmers in 7 townships during 2021-2023. Semi-structured interviews and participant observation were conducted to document traditional knowledge and management practices. RESULTS: The locals reported 51 plant species used as pig feed, with 30 wild species. Growth-stage-specific feeding and seasonal adjustment practices were documented. We recorded 62 ethnoveterinary plant uses, mainly for treating pigs' heat stress and skin conditions. Luchuan pigs play central roles in local Hakka customs, rituals, and cuisine. Additonally, the new ecological farming models minimize the environmental impacts to the local community. However, there are still some challenges remained for conserving and promoting Luchuan pigs. CONCLUSIONS: The Luchuan Hakka people possess rich traditional knowledge and management experience in raising Luchuan pigs. Our study provides extensive documentation of traditional knowledge and recommends integrating cultural and genetic aspects for sustaining this biocultural heritage. Findings can inform initiatives supporting local breed conservation globally.


Asunto(s)
Agricultura , Cruzamiento , Humanos , Porcinos , Animales , China , Cultura , Agricultores
4.
Reproduction ; 166(3): 187-197, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37310899

RESUMEN

In brief: Normal gene expression during early embryonic development and in the placenta is crucial for a successful pregnancy. Nicotine can disrupt normal gene expression during development, leading to abnormal embryonic and placental development. Abstract: Nicotine is a common indoor air pollutant that is present in cigarette fumes. Due to its lipophilic nature, nicotine can rapidly transport through membrane barriers and spread throughout the body, which can lead to the development of diseases. However, the impact of nicotine exposure during early embryonic development on subsequent development remains elusive. In this study, we found that nicotine significantly elevated reactive oxygen species, DNA damage and cell apoptosis levels with the decrease of blastocyst formation during early embryonic development. More importantly, nicotine exposure during early embryonic development increased placental weight and disrupted placental structure. In molecular level, we also observed that nicotine exposure could specifically cause the hypermethylation of Phlda2 promoter (a maternally expressed imprinted gene associated with placental development) and reduce the mRNA expression of Phlda2. By RNA sequencing analysis, we demonstrated that nicotine exposure affected the gene expression and excessive activation of the Notch signaling pathway thereby affecting placental development. Blocking the Notch signaling pathway by DAPT treatment could recover abnormal placental weight and structure induced by nicotine exposure. Taken together, this study indicates that nicotine causes the declining quality of early embryos and leads to placental abnormalities related to over-activation of the Notch signaling pathway.


Asunto(s)
Placenta , Placentación , Embarazo , Femenino , Humanos , Placenta/metabolismo , Nicotina/toxicidad , Nicotina/metabolismo , Proteínas Nucleares/metabolismo , Transducción de Señal
5.
Environ Pollut ; 331(Pt 1): 121856, 2023 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-37211227

RESUMEN

Arsenite is commonly used as an insecticide, antiseptic and herbicide. It can enter the food chain via through soil contamination, and harm human health, including the reproductive systems. Early embryos, as the initial stage of mammalian life, are very sensitive to the environmental toxins and pollutants. However, whether and how arsenite disturbs the early embryo development remains unclear. Our study used mouse early embryos as a model and revealed that arsenite exposure did not cause reactive oxygen species production, DNA damage or apoptosis. However, arsenite exposure arrested embryonic development at the 2-cell stage by altering gene expression patterns. The transcriptional profile in the disrupted embryos showed abnormal maternal-to-zygote transition (MZT). More importantly, arsenite exposure attenuated H3K27ac modification enrichment at the promoter region of Brg1, a key gene for MZT, which inhibited its transcription, and further affected MZT and early embryonic development. In conclusion our study highlights arsenite exposure affects MZT by reducing the enrichment of H3K27ac on the embryonic genome, and ultimately induces early embryonic development arrest at the 2-cell stage.


Asunto(s)
Arsenitos , Cigoto , Embarazo , Femenino , Humanos , Animales , Ratones , Cigoto/metabolismo , Arsenitos/toxicidad , Arsenitos/metabolismo , Desarrollo Embrionario/genética , Mamíferos/genética , Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica
6.
Autophagy ; 19(1): 163-179, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35404187

RESUMEN

Macroautophagy/autophagy is a cellular and energy homeostatic mechanism that contributes to maintain the number of primordial follicles, germ cell survival, and anti-ovarian aging. However, it remains unknown whether autophagy in granulosa cells affects oocyte maturation. Here, we show a clear tendency of reduced autophagy level in human granulosa cells from women of advanced maternal age, implying a potential negative correlation between autophagy levels and oocyte quality. We therefore established a co-culture system and show that either pharmacological inhibition or genetic ablation of autophagy in granulosa cells negatively affect oocyte quality and fertilization ability. Moreover, our metabolomics analysis indicates that the adverse impact of autophagy impairment on oocyte quality is mediated by downregulated citrate levels, while exogenous supplementation of citrate can significantly restore the oocyte maturation. Mechanistically, we found that ACLY (ATP citrate lyase), which is a crucial enzyme catalyzing the cleavage of citrate, was preferentially associated with K63-linked ubiquitin chains and recognized by the autophagy receptor protein SQSTM1/p62 for selective autophagic degradation. In human follicles, the autophagy level in granulosa cells was downregulated with maternal aging, accompanied by decreased citrate in the follicular fluid, implying a potential correlation between citrate metabolism and oocyte quality. We also show that elevated citrate levels in porcine follicular fluid promote oocyte maturation. Collectively, our data reveal that autophagy in granulosa cells is a beneficial mechanism to maintain a certain degree of citrate by selectively targeting ACLY during oocyte maturation.Abbreviations: 3-MA: 3-methyladenine; ACLY: ATP citrate lyase; AMA: advanced maternal age; CG: cortical granule; CHX: cycloheximide; CQ: chloroquine; CS: citrate synthase; COCs: cumulus-oocyte-complexes; GCM: granulosa cell monolayer; GV: germinal vesicle; MII: metaphase II stage of meiosis; PB1: first polar body; ROS: reactive oxygen species; shRNA: small hairpin RNA; SQSTM1/p62: sequestosome 1; TCA: tricarboxylic acid; TOMM20/TOM20: translocase of outer mitochondrial membrane 20; UBA: ubiquitin-associated domain; Ub: ubiquitin; WT: wild-type.


Asunto(s)
ATP Citrato (pro-S)-Liasa , Macroautofagia , Femenino , Humanos , Animales , Porcinos , Proteína Sequestosoma-1/metabolismo , ATP Citrato (pro-S)-Liasa/metabolismo , Ácido Cítrico/metabolismo , Autofagia , Oocitos/metabolismo , Citratos/metabolismo , Aciltransferasas/metabolismo , Ubiquitina/metabolismo , Homeostasis
7.
Angew Chem Int Ed Engl ; 61(50): e202214142, 2022 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-36225162

RESUMEN

It is well known that charge separation is crucial for efficient photocatalytic solar conversion. Although some covalent-organic frameworks (COFs) exhibit visible-light harvest, the large exciton binding energies reduce their photocatalytic efficiencies. Herein, we developed a novel method to post-treat the olefin-linked COFs with end-capping polycyclic aromatic hydrocarbons (PAHs) for spontaneous charge separation. Interestingly, a type-II heterostructure is constructed in our perylene-modified COFs which displays drastically enhanced performance for photocatalytic CO2 reduction, with an efficiency of 8-fold higher than that of unmodified COF. A combination of electrochemical, steady-state, and time-resolved spectroscopic measurements indicates that such drastically enhanced performance should be attributed to photoinduced spontaneous charge separation in the heterostructure. These results illustrate the feasibility of engineering the charge-separation properties of crystalline porous frameworks at a molecular level for artificial photosynthesis.

8.
Adv Sci (Weinh) ; 9(23): e2200057, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35717671

RESUMEN

Early embryos undergo extensive epigenetic reprogramming to achieve gamete-to-embryo transition, which involves the loading and removal of histone variant H2A.Z on chromatin. However, how does H2A.Z regulate gene expression and histone modifications during preimplantation development remains unrevealed. Here, by using ultra-low-input native chromatin immunoprecipitation and sequencing, the genome-wide distribution of H2A.Z is delineated in mouse oocytes and early embryos. These landscapes indicate that paternal H2A.Z is removed upon fertilization, followed by unbiased accumulation on parental genomes during zygotic genome activation (ZGA). Remarkably, H2A.Z exhibits hierarchical accumulation as different peak types at promoters: promoters with double H2A.Z peaks are colocalized with H3K4me3 and indicate transcriptional activation; promoters with a single H2A.Z peak are more likely to occupy bivalent marks (H3K4me3+H3K27me3) and indicate development gene suppression; promoters with no H2A.Z accumulation exhibit persisting gene silencing in early embryos. Moreover, H2A.Z depletion changes the enrichment of histone modifications and RNA polymerase II binding at promoters, resulting in abnormal gene expression and developmental arrest during lineage commitment. Furthermore, similar transcription and accumulation patterns between mouse and porcine embryos indicate that a dual role of H2A.Z in regulating the epigenome required for proper gene expression is conserved during mammalian preimplantation development.


Asunto(s)
Código de Histonas , Histonas , Animales , Cromatina/genética , Cromatina/metabolismo , Embrión de Mamíferos/metabolismo , Código de Histonas/genética , Histonas/genética , Histonas/metabolismo , Mamíferos/genética , Mamíferos/metabolismo , Ratones , Procesamiento Proteico-Postraduccional
9.
Autophagy ; 18(12): 2946-2968, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35311460

RESUMEN

Macroautophagy/autophagy is a conserved cellular mechanism to degrade unneeded cytoplasmic proteins and organelles to recycle their components, and it is critical for embryonic stem cell (ESC) self-renewal and somatic cell reprogramming. Whereas autophagy is essential for early development of embryos, no information exists regarding its functions during the transition from naive-to-primed pluripotency. Here, by using an in vitro transition model of ESCs to epiblast-like cells (EpiLCs), we find that dynamic changes in ATG7-dependent autophagy are critical for the naive-to-primed transition, and are also necessary for germline specification. RNA-seq and ATAC-seq profiling reveal that NANOG acts as a barrier to prevent pluripotency transition, and autophagy-dependent NANOG degradation is important for dismantling the naive pluripotency expression program through decommissioning of naive-associated active enhancers. Mechanistically, we found that autophagy receptor protein SQSTM1/p62 translocated into the nucleus during the pluripotency transition period and is preferentially associated with K63 ubiquitinated NANOG for selective protein degradation. In vivo, loss of autophagy by ATG7 depletion disrupts peri-implantation development and causes increased chromatin association of NANOG, which affects neuronal differentiation by competitively binding to OTX2-specific neuroectodermal development-associated regions. Taken together, our findings reveal that autophagy-dependent degradation of NANOG plays a critical role in regulating exit from the naive state and marks distinct cell fate allocation during lineage specification.Abbreviations: 3-MA: 3-methyladenine; EpiLC: epiblast-like cell; ESC: embryonic stem cell; PGC: primordial germ cell.


Asunto(s)
Autofagia , Células Madre Embrionarias , Células Madre Embrionarias/metabolismo , Diferenciación Celular , Estratos Germinativos/metabolismo , Cromatina/metabolismo
10.
Stem Cell Reports ; 16(11): 2674-2689, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34678203

RESUMEN

Pig cloning by somatic cell nuclear transfer (SCNT) frequently undergoes incomplete epigenetic remodeling during the maternal-to-zygotic transition, which leads to a significant embryonic loss before implantation. Here, we generated the first genome-wide landscapes of histone methylation in pig SCNT embryos. Excessive H3K9me3 and H3K27me3, but not H3K4me3, were observed in the genomic regions with unfaithful embryonic genome activation and donor-cell-specific gene silencing. A combination of H3K9 demethylase KDM4A and GSK126, an inhibitor of H3K27me3 writer, were able to remove these epigenetic barriers and restore the global transcriptome in SCNT embryos. More importantly, thymine DNA glycosylase (TDG) was defined as a pig-specific epigenetic regulator for nuclear reprogramming, which was not reactivated by H3K9me3 and H3K27me3 removal. Both combined treatment and transient TDG overexpression promoted DNA demethylation and enhanced the blastocyst-forming rates of SCNT embryos, thus offering valuable methods to increase the cloning efficiency of genome-edited pigs for agricultural and biomedical purposes.


Asunto(s)
Embrión de Mamíferos/metabolismo , Epigénesis Genética , Regulación de la Expresión Génica , Histonas/metabolismo , Técnicas de Transferencia Nuclear , Timina ADN Glicosilasa/genética , Animales , Blastocisto/citología , Blastocisto/metabolismo , Metilación de ADN , Desmetilación , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/embriología , Perfilación de la Expresión Génica/métodos , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Indoles/farmacología , Lisina/metabolismo , Metilación , Piridonas/farmacología , Porcinos , Timina ADN Glicosilasa/metabolismo
11.
Theriogenology ; 158: 470-476, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33049572

RESUMEN

Signaling pathways and transcription factors are involved in porcine embryonic development. Here, we demonstrate that glycogen synthase kinase-3 (GSK3) inhibitor, CHIR99021 and recombinant porcine interleukin-6 (rpIL6) significantly promote porcine parthenogenetic blastocyst formation (49.23 ± 8.40% vs 32.34 ± 4.15%), with increased inner cell mass (ICM) cell numbers (7.72 ± 2.30 vs 4.28 ± 1.60) and higher expression of pluripotent genes, such as OCT4, SOX2 and NANOG. Furthermore, CHIR99021 and rpIL6 improve blastocyst quality with increased blastocyst hatching percentage (16.19 ± 1.96% vs 10.25 ± 1.12%) and subsequently porcine pluripotent stem cells (pPSCs) derivation efficiency. These results advance the understanding of porcine pre-implantation development and provide evidences in improving the blastocyst quality.


Asunto(s)
Glucógeno Sintasa Quinasa 3 , Interleucina-6 , Animales , Blastocisto , Desarrollo Embrionario , Interleucina-6/genética , Piridinas , Pirimidinas , Porcinos
12.
Front Cell Dev Biol ; 8: 578, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32733887

RESUMEN

Maternal regulatory factors endow the oocyte with developmental competence in vivo, which might be absent in current in vitro maturation (IVM) systems, thereby compromising oocyte quality. In the present study, by employing RNA sequencing data analysis, we expect to identify potential contributing factors to support porcine oocyte maturation through binding to their receptors on the oolemma. Here, C-X-C motif chemokine ligand 12 (CXCL12), vascular endothelial growth factor A (VEGFA), and Wingless-type MMTV integration site family member 5A (WNT5A), termed CVW, are selected and confirmed to be important maternal cytokines for porcine oocyte maturation. Combined supplementation of CVW promotes the nuclear maturation percentage from 57.2% in controls to 75.9%. More importantly, these maternal cytokines improve the developmental potential of matured oocytes by parthenogenesis, fertilization, and cloning, as their blastocyst formation efficiencies and total cell numbers are increased. CVW supplementation also enlarges perivitelline space and promotes cumulus expansion, which results in a more complete transzonal projection retraction on the zona pellucida, and a reduced incidence of polyspermy in fertilized oocytes. Meanwhile, inhibiting the CVW receptor-mediated signaling pathways severely impairs oocyte meiotic resumption and cumulus expansion during IVM. We further determine that maturation improvement by CVW is achieved through activating the MAPK pathway in advance and inhibiting the canonical WNT pathway at the end of the IVM period. These findings provide a new combination of three cytokines to promote the porcine IVM process, which also holds potential to be used in human assisted reproduction technologies as well as in other species.

13.
Development ; 147(13)2020 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-32620578

RESUMEN

In mammalian growing follicles, oocytes are arrested at the diplotene stage (which resembles the G2/M boundary in mitosis), while the granulosa cells (GCs) continue to proliferate during follicular development, reflecting a cell cycle asynchrony between oocytes and GCs. Hypoxanthine (Hx), a purine present in the follicular fluid, has been shown to induce oocytes meiotic arrest, although its role in GC proliferation remains ill-defined. Here, we demonstrate that Hx indiscriminately prevents G2-to-M phase transition in porcine GCs. However, oocyte-derived paracrine factors (ODPFs), particularly GDF9 and BMP15, maintain the proliferation of GCs, partly by activating the ERK1/2 signaling and enabling the G2/M transition that is suppressed by Hx. Interestingly, GCs with lower expression of GDF9/BMP15 receptors appear to be more sensitive to Hx-induced G2/M arrest and become easily detached from the follicular wall. Importantly, Hx-mediated inhibition of G2/M progression instigates GC apoptosis, which is ameliorated in the presence of GDF9 and/or BMP15. Therefore, our data indicate that the counterbalance of intrafollicular factors, particularly Hx and oocyte-derived GDF9/BMP15, fine-tunes the development of porcine follicles by regulating the cell cycle progression of GCs.


Asunto(s)
Células de la Granulosa/metabolismo , Hipoxantina/metabolismo , Oocitos/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Proteína Morfogenética Ósea 15/genética , Proteína Morfogenética Ósea 15/metabolismo , Proliferación Celular/genética , Proliferación Celular/fisiología , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Factor 9 de Diferenciación de Crecimiento/genética , Factor 9 de Diferenciación de Crecimiento/metabolismo , Porcinos
14.
Reprod Fertil Dev ; 32(7): 657-666, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32317091

RESUMEN

Autophagy plays an important role in embryo development; however, only limited information is available on how autophagy specifically regulates embryo development, especially under low oxygen culture conditions. In this study we used parthenogenetic activation (PA) of porcine embryos to test the hypothesis that a low oxygen concentration (5%) could promote porcine embryo development by activating autophagy. Immunofluorescence staining revealed that low oxygen tension activated autophagy and alleviated oxidative stress in porcine PA embryos. Development was significantly affected when autophagy was blocked by 3-methyladenine, even under low oxygen culture conditions, with increased reactive oxygen species levels and malondialdehyde content. Furthermore, the decreased expression of pluripotency-associated genes induced by autophagy inhibition could be recovered by treatment with the antioxidant vitamin C. Together, these results demonstrate that low oxygen-induced autophagy regulates embryo development through antioxidant mechanisms in the pig.


Asunto(s)
Autofagia/fisiología , Técnicas de Cultivo de Embriones/veterinaria , Desarrollo Embrionario/fisiología , Oxígeno/administración & dosificación , Partenogénesis/fisiología , Porcinos/embriología , Adenina/análogos & derivados , Adenina/farmacología , Animales , Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Autofagia/efectos de los fármacos , Técnicas de Cultivo de Embriones/métodos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología
15.
Endocrinology ; 161(4)2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32141513

RESUMEN

Follicle-stimulating hormone (FSH)-induced growth of ovarian follicles is independent of follicular vascularization. Recent evidence has indicated that follicular vascularization is critical to ovarian follicle development and survival. FSH, a gonadotropin that induces follicular growth and development, also acts as the major survival factor for antral follicles. FSH has been reported to stimulate angiogenesis in the theca layers mediated in part by the vascular endothelial growth factor A (VEGFA) and the transcription factor hypoxia inducible factor 1α (HIF-1α). However, it remains largely undetermined whether FSH-dependent growth and survival of antral follicles relies on FSH-induced vascularization. Here, we first demonstrated that induction of angiogenesis through the FSH-HIF-1α-VEGFA axis is not required for FSH-stimulated follicular growth in mouse ovary. FSH increased the total number of blood vessels in mouse ovarian follicles, which was correlated with elevated expression of VEGFA and HIF-1α in granulosa cells. In contrast, blocking of follicular angiogenesis using inhibitors against the HIF-1α-VEGFA pathway repressed vasculature formation in follicles despite FSH administration. Interestingly, by measuring follicular size and ovarian weight, we found that the suppression of angiogenesis via HIF-1α-VEGFA pathway did not influence FSH-mediated follicular growth. However, inhibition of FSH-induced follicular vascularization by PX-478, a small-molecule inhibitor that suppresses HIF-1α activity, blocked ovulation and triggered atresia in large follicles. On the other hand, PX-478 injection reduced oocyte quality via impairing the meiotic apparatus, showing a prominently defective spindle assembly and actin dynamics. Collectively, our findings unveiled a vascularization-independent effect of FSH on follicular growth, whereas follicular survival, ovulation, and oocyte development relies on FSH-mediated angiogenesis in the follicles.


Asunto(s)
Hormona Folículo Estimulante/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Oocitos/crecimiento & desarrollo , Folículo Ovárico/crecimiento & desarrollo , Ovulación/metabolismo , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Femenino , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/metabolismo , Ovulación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
16.
FASEB J ; 34(3): 3631-3645, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31960530

RESUMEN

In developing follicles, the granulosa cells (GCs) live in a hypoxic environment due to the devoid of blood supply. Upon hypoxic conditions, several types of mammalian cells have been reported to undergo apoptosis. Follicle-stimulating hormone (FSH) is known as the primary survival factor for antral follicles by preventing GCs apoptosis. Mitophagy is a type of organelle-specific autophagy that removes damaged or stressed mitochondria to maintain cellular health. This study provides the first evidence suggesting that FSH-mediated mitophagy protected porcine GCs from hypoxia-induced apoptosis. Our data showed that the GCs apoptosis caused by mitochondrial pathway upon hypoxia stress was markedly attenuated after FSH treatment, which was correlated with enhanced activation of mitophagy. Interestingly, FSH also stimulated mitochondrial biogenesis as suggested by increased expression of mitochondrial transcription factor A and nuclear respiratory factor 1 during hypoxia exposure. Notably, the protein level of hypoxia inducible factor-1α (HIF-1α) was significantly increased in hypoxic GCs following FSH treatment, accompanied by elevated mitophagic activity and dampened apoptotic signaling. Blocking HIF-1α inhibited mitophagy and restored hypoxia-induced apoptosis despite FSH treatment. Importantly, FSH promoted the expression of serine/threonine kinase PTEN induced putative kinase 1 (PINK1) and the E3 ligase Parkin during hypoxia stress through a HIF-1α dependent manner. This induced the mitophagic clearance of damaged mitochondria, hence inhibiting apoptosis by reducing cytochrome c releasing. The inhibition of HIF-1α and/or PINK1 using inhibitor or RNAi further confirmed the role of the FSH-HIF-1α-PINK1-Parkin-mitophagy axis in suppressing GC apoptosis under hypoxic conditions. These findings highlight a novel function of FSH in preserving GCs viability against hypoxic damage by activating HIF-1α-PINK1-Parkin-mediated mitophagy.


Asunto(s)
Apoptosis/efectos de los fármacos , Hormona Folículo Estimulante/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mitofagia/efectos de los fármacos , Proteínas Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Femenino , Microscopía Electrónica de Transmisión , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Porcinos
17.
J Cell Physiol ; 235(3): 2836-2846, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31535366

RESUMEN

Malathion (MAL) is a common organophosphorus pesticide and affects both animal and human reproduction. However, the mechanisms regarding how MAL affects the mammalian oocyte quality and how to prevent it have not been fully investigated. In this study, we used porcine oocyte as a model and proved that MAL impaired porcine oocyte quality in a dose-dependent manner during maturation. MAL decreased the first polar body extrusion, disrupted spindle assembly and chromosome alignment, impaired cortical granules (CGs) distribution, and increased reactive oxygen species (ROS) level in oocytes. RNA-seq analysis showed that MAL exposure altered the expression of 2,917 genes in the porcine maturated oocytes and most genes were related to ROS, the lipid droplet process, and the energy supplement. Nevertheless, these defects could be remarkably ameliorated by adding melatonin (MLT) into the oocyte maturation medium. MLT increased oocyte maturation rate and decreased the abnormities of spindle assembly, CGs distribution and ROS accumulation in MAL-exposed porcine oocytes. More important, MLT upregulated the expression of genes related to lipid droplet metabolism (PPARγ and PLIN2), decreased lipid droplet size and lipid peroxidation in MAL-exposed porcine oocytes. Finally, we found that MLT increased the blastocysts formation and the cell numbers of blastocysts in MAL-exposed porcine oocytes after parthenogenetic activation, which was mediated by reduction of ROS levels and maintaining lipid droplet metabolism. Taken together, our results revealed that MLT had a protective action against MAL-induced deterioration of porcine oocyte quality.


Asunto(s)
Malatión/metabolismo , Melatonina/farmacología , Oocitos/efectos de los fármacos , Oogénesis/efectos de los fármacos , Animales , Desarrollo Embrionario/efectos de los fármacos , Técnicas de Maduración In Vitro de los Oocitos/métodos , Peroxidación de Lípido/efectos de los fármacos , Meiosis/efectos de los fármacos , Oocitos/metabolismo , Partenogénesis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Porcinos
18.
Biol Reprod ; 102(1): 116-132, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-31435642

RESUMEN

As the follicle develops, the thickening of the granulosa compartment leads to progressively deficient supply of oxygen in granulosa cells (GCs) due to the growing distances from the follicular vessels. These conditions are believed to cause hypoxia in GCs during folliculogenesis. Upon hypoxic conditions, several types of mammalian cells have been reported to undergo cell cycle arrest. However, it remains unclear whether hypoxia exerts any impact on cell cycle progression of GCs. On the other hand, although the GCs may live in a hypoxic environment, their mitotic capability appears to be unaffected in growing follicles. It thus raises the question whether there are certain intraovarian factors that might overcome the inhibitory effects of hypoxia. The present study provides the first evidence suggesting that cobalt chloride (CoCl2)-mimicked hypoxia prevented G1-to-S cell cycle progression in porcine GCs. In addition, we demonstrated that the inhibitory effects of CoCl2 on GCs cell cycle are mediated through hypoxia-inducible factor-1 alpha/FOXO1/Cdkn1b pathway. Moreover, we identified insulin-like growth factor-I (IGF-I) as an intrafollicular factor required for cell cycle recovery by binding to IGF-I receptor in GCs suffering CoCl2 stimulation. Further investigations confirmed a role of IGF-I in preserving G1/S progression of CoCl2-treated GCs via activating the cyclin E/cyclin-dependent kinase2 complex through the phoshatidylinositol-3 kinase/protein kinase B (AKT)/FOXO1/Cdkn1b axis. Although the present findings were based on a hypoxia mimicking model by using CoCl2, our study might shed new light on the regulatory mechanism of GCs cell cycle upon hypoxic stimulation.


Asunto(s)
Puntos de Control del Ciclo Celular/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Puntos de Control del Ciclo Celular/fisiología , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/fisiología , Cobalto/farmacología , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Estradiol/farmacología , Femenino , Hormona Folículo Estimulante/farmacología , Proteína Forkhead Box O1/metabolismo , Células de la Granulosa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Porcinos
19.
Aging (Albany NY) ; 11(23): 11504-11519, 2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31834867

RESUMEN

Resveratrol (3,5,4'-trihydroxystilbene, RSV) is a natural potential anti-aging polyphenolic compound frequently used as a nutritional supplement against several diseases. However, the underlying mechanisms by which resveratrol regulates postovulatory aging of oocytes are still insufficiently known. In this study, we found that resveratrol could delay postovulatory aging and improve developmental competence of oocytes through activating selective mitophagy in the mouse. Resveratrol could maintain spindle morphology but it disturbed cortical granule (CG) distribution during oocyte aging. This might be due to upregulated mitophagy, since blocking mitophagy by cyclosporin A (CsA) treatment affected oocyte quality by damaging mitochondrial function and it decreased embryonic development. In addition, we also observed an involvement of FoxO3a in regulating mitophagy in aging oocytes following resveratrol treatment. Taken together, our results provide evidence that mitophagy induced by resveratrol is a potential mechanism to protect against postovulatory oocyte aging.


Asunto(s)
Mitofagia/efectos de los fármacos , Oocitos/efectos de los fármacos , Ovulación/fisiología , Resveratrol/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Animales , Ciclosporina/farmacología , Femenino , Ratones , Ratones Endogámicos ICR , Mitofagia/fisiología , Oocitos/fisiología
20.
Environ Mol Mutagen ; 60(9): 807-815, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31411769

RESUMEN

Tris(1,3-dichloro-2-propyl) phosphate (TDCPP) is a kind of additive flame retardants (FRs) and was found to affect early embryonic development in zebrafish; however, there are few studies to investigate whether TDCPP will disturb the development of early mouse embryos. In our studies, we used mouse embryos as models to study the toxicology of TDCPP on the early embryos. The results showed that TDCPP disturbed the development of early mouse embryos in a dose-dependent manner. 10 µM TDCPP decreased the blastocyst formation and 100 µM TDCPP was a lethal concentration for the mouse embryos. We proved that TDCPP was detrimental to embryonic development potential by increasing the reactive oxygen species level and inducing early apoptosis. Furthermore, TDCPP changed the DNA methylation patterns of imprinted genes in treated blastocysts. The methylation of H19 and Snrpn promoter regions was increased from 37.67% to 46.00% and 31.56% to 44.38% in treated groups, respectively. In contrast, Peg3 promoter region methylation was declined from 86.55% to 73.27% in treated embryos. Taken together, our results demonstrated that TDCPP could adversely impair the early embryonic development in mouse. Environ. Mol. Mutagen. 2019. © 2019 Wiley Periodicals, Inc.


Asunto(s)
Apoptosis/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Compuestos Organofosforados/efectos adversos , Animales , Blastocisto/efectos de los fármacos , Blastocisto/metabolismo , Retardadores de Llama/efectos adversos , Ratones , Ratones Endogámicos ICR , Regiones Promotoras Genéticas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...