Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
Más filtros












Intervalo de año de publicación
1.
Nat Commun ; 15(1): 5550, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38956014

RESUMEN

Oocyte in vitro maturation is a technique in assisted reproductive technology. Thousands of genes show abnormally high expression in in vitro maturated metaphase II (MII) oocytes compared to those matured in vivo in bovines, mice, and humans. The mechanisms underlying this phenomenon are poorly understood. Here, we use poly(A) inclusive RNA isoform sequencing (PAIso-seq) for profiling the transcriptome-wide poly(A) tails in both in vivo and in vitro matured mouse and human oocytes. Our results demonstrate that the observed increase in maternal mRNA abundance is caused by impaired deadenylation in in vitro MII oocytes. Moreover, the cytoplasmic polyadenylation of dormant Btg4 and Cnot7 mRNAs, which encode key components of deadenylation machinery, is impaired in in vitro MII oocytes, contributing to reduced translation of these deadenylase machinery components and subsequently impaired global maternal mRNA deadenylation. Our findings highlight impaired maternal mRNA deadenylation as a distinct molecular defect in in vitro MII oocytes.


Asunto(s)
Oocitos , Poliadenilación , Oocitos/metabolismo , Animales , Humanos , Femenino , Ratones , Poli A/metabolismo , Técnicas de Maduración In Vitro de los Oocitos , ARN Mensajero/metabolismo , ARN Mensajero/genética , Transcriptoma , ARN Mensajero Almacenado/metabolismo , ARN Mensajero Almacenado/genética , Metafase , Exorribonucleasas , Proteínas Represoras , Proteínas de Ciclo Celular
2.
Curr Biol ; 34(11): R519-R523, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38834020

RESUMEN

Rapid cleavage divisions and the transition from maternal to zygotic control of gene expression are the hallmarks of early embryonic development in most species. Early development in insects, fish and amphibians is characterized by several short cell cycles with no gap phases, necessary for the rapid production of cells prior to patterning and morphogenesis. Maternal mRNAs and proteins loaded into the egg during oogenesis are essential to drive these rapid early divisions. Once the function of these maternal inputs is complete, the maternal-to-zygotic transition (MZT) marks the handover of developmental control to the gene products synthesized from the zygotic genome. The MZT requires three major events: the removal of a subset of maternal mRNAs, the initiation of zygotic transcription, and the remodeling of the cell cycle. In each species, the MZT occurs at a highly reproducible time during development due to a series of feedback mechanisms that tightly couple these three processes. Dissecting these feedback mechanisms and their spatiotemporal control will be essential to understanding the control of the MZT. In this primer, we outline the mechanisms that govern the major events of the MZT across species and highlight the role of feedback mechanisms that ensure the MZT is precisely timed and orchestrated.


Asunto(s)
Cigoto , Cigoto/metabolismo , Cigoto/crecimiento & desarrollo , Animales , Regulación del Desarrollo de la Expresión Génica , Desarrollo Embrionario , Femenino , ARN Mensajero Almacenado/metabolismo , ARN Mensajero Almacenado/genética
3.
Open Biol ; 14(6): 240065, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38896085

RESUMEN

The transition from oocyte to embryo requires translation of maternally provided transcripts that in Drosophila is activated by Pan Gu kinase to release a rapid succession of 13 mitotic cycles. Mitotic entry is promoted by several protein kinases that include Greatwall/Mastl, whose Endosulfine substrates antagonize Protein Phosphatase 2A (PP2A), facilitating mitotic Cyclin-dependent kinase 1/Cyclin B kinase activity. Here we show that hyperactive greatwallScant can not only be suppressed by mutants in its Endos substrate but also by mutants in Pan Gu kinase subunits. Conversely, mutants in me31B or trailer hitch, which encode a complex that represses hundreds of maternal mRNAs, enhance greatwallScant . Me31B and Trailer Hitch proteins, known substrates of Pan Gu kinase, copurify with Endos. This echoes findings that budding yeast Dhh1, orthologue of Me31B, associates with Igo1/2, orthologues of Endos and substrates of the Rim15, orthologue of Greatwall. endos-derived mutant embryos show reduced Me31B and elevated transcripts for the mitotic activators Cyclin B, Polo and Twine/Cdc25. Together, our findings demonstrate a previously unappreciated conservation of the Greatwall-Endosulfine pathway in regulating translational repressors and its interactions with the Pan Gu kinase pathway to regulate translation and/or stability of maternal mRNAs upon egg activation.


Asunto(s)
Proteínas de Drosophila , Regulación del Desarrollo de la Expresión Génica , Oocitos , Proteína Fosfatasa 2 , Animales , Femenino , ARN Helicasas DEAD-box , Drosophila melanogaster/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Embrión no Mamífero/metabolismo , Mutación , Oocitos/metabolismo , Oocitos/citología , Biosíntesis de Proteínas , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Estabilidad del ARN , ARN Mensajero Almacenado/metabolismo , ARN Mensajero Almacenado/genética
4.
Biochem Soc Trans ; 52(2): 861-871, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38477334

RESUMEN

A large number of mRNAs of maternal origin are produced during oogenesis and deposited in the oocyte. Since transcription stops at the onset of meiosis during oogenesis and does not resume until later in embryogenesis, maternal mRNAs are the only templates for protein synthesis during this period. To ensure that a protein is made in the right place at the right time, the translation of maternal mRNAs must be activated at a specific stage of development. Here we summarize our current understanding of the sophisticated mechanisms that contribute to the temporal repression of maternal mRNAs, termed maternal mRNA dormancy. We discuss mechanisms at the level of the RNA itself, such as the regulation of polyadenine tail length and RNA modifications, as well as at the level of RNA-binding proteins, which often block the assembly of translation initiation complexes at the 5' end of an mRNA or recruit mRNAs to specific subcellular compartments. We also review microRNAs and other mechanisms that contribute to repressing translation, such as ribosome dormancy. Importantly, the mechanisms responsible for mRNA dormancy during the oocyte-to-embryo transition are also relevant to cellular quiescence in other biological contexts.


Asunto(s)
Oocitos , Oogénesis , Animales , Humanos , Oocitos/metabolismo , Oogénesis/genética , ARN Mensajero/metabolismo , ARN Mensajero/genética , MicroARNs/genética , MicroARNs/metabolismo , ARN Mensajero Almacenado/metabolismo , ARN Mensajero Almacenado/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Biosíntesis de Proteínas , Regulación del Desarrollo de la Expresión Génica , Femenino , Desarrollo Embrionario/genética
5.
Free Radic Biol Med ; 217: 157-172, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38552928

RESUMEN

Obesity has significant repercussions for female reproductive health, including adverse effects on oocyte quality, fertility, embryo development and offspring health. Here, we showed that intermittent fasting (IF) has several notable effects on follicular development, oocyte development and maturation and offspring health in obese mice. IF treatment prevents obesity-associated germline-soma communication defects, mitochondrial dysfunction, oxidative damage, apoptosis, and spindle/chromosomal disruption. RNA-sequencing analysis of oocytes from normal diet (ND), high-fat diet (HFD), and HFD + IF mice indicated that IF treatment improved mitochondrial oxidative phosphorylation function and mRNA storage and translation, which was potentially mediated by the Smith-like family member 14 B (LSM14B). Knockdown of LSM14B by siRNA injection in oocytes from ND mice recapitulates all the translation, mitochondrial dysfunction and meiotic defect phenotypes of oocytes from HFD mice. Remarkably, the injection of Lsm14b mRNA into oocytes from HFD mice rescued the translation, mitochondrial dysfunction and meiotic defect phenotypes. These results demonstrated that dysfunction in the oocyte translation program is associated with obesity-induced meiotic defects, while IF treatment increased LSM14B expression and maternal mRNA translation and restored oocyte quality. This research has important implications for understanding the effects of obesity on female reproductive health and offers a potential nonpharmacological intervention to improve oocyte quality and fertility in obese individuals.


Asunto(s)
Ayuno Intermitente , ARN Mensajero Almacenado , Animales , Femenino , Ratones , Meiosis , Ratones Obesos , Enfermedades Mitocondriales/metabolismo , Obesidad/metabolismo , Oocitos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Mensajero Almacenado/metabolismo
6.
EMBO Rep ; 25(1): 404-427, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38177902

RESUMEN

Maternal mRNAs are essential for protein synthesis during oogenesis and early embryogenesis. To adapt translation to specific needs during development, maternal mRNAs are translationally repressed by shortening the polyA tails. While mRNA deadenylation is associated with decapping and degradation in somatic cells, maternal mRNAs with short polyA tails are stable. Here we report that the germline-specific eIF4E paralog, eIF4E1b, is essential for zebrafish oogenesis. eIF4E1b localizes to P-bodies in zebrafish embryos and binds to mRNAs with reported short or no polyA tails, including histone mRNAs. Loss of eIF4E1b results in reduced histone mRNA levels in early gonads, consistent with a role in mRNA storage. Using mouse and human eIF4E1Bs (in vitro) and zebrafish eIF4E1b (in vivo), we show that unlike canonical eIF4Es, eIF4E1b does not interact with eIF4G to initiate translation. Instead, eIF4E1b interacts with the translational repressor eIF4ENIF1, which is required for eIF4E1b localization to P-bodies. Our study is consistent with an important role of eIF4E1b in regulating mRNA dormancy and provides new insights into fundamental post-transcriptional regulatory principles governing early vertebrate development.


Asunto(s)
ARN Mensajero Almacenado , Pez Cebra , Animales , Humanos , Ratones , ARN Mensajero Almacenado/genética , ARN Mensajero Almacenado/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Histonas/metabolismo , Factor 4E Eucariótico de Iniciación/genética , Factor 4E Eucariótico de Iniciación/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Biosíntesis de Proteínas
7.
Trends Genet ; 40(3): 238-249, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38262796

RESUMEN

Maternal mRNAs accumulate during egg growth and must be judiciously degraded or translated to ensure successful development of mammalian embryos. In this review we integrate recent investigations into pathways controlling rapid degradation of maternal mRNAs during the maternal-to-zygotic transition. Degradation is not indiscriminate, and some mRNAs are selectively protected and rapidly translated after fertilization for reprogramming the zygotic genome during early embryogenesis. Oocyte specific cofactors and pathways have been illustrated to control different futures of maternal mRNAs. We discuss mechanisms that control the fate of maternal mRNAs during late oogenesis and after fertilization. Issues to be resolved in current maternal mRNA research are described, and future research directions are proposed.


Asunto(s)
Desarrollo Embrionario , ARN Mensajero Almacenado , Animales , ARN Mensajero Almacenado/genética , ARN Mensajero Almacenado/metabolismo , Desarrollo Embrionario/genética , Oocitos , Oogénesis/genética , Cigoto , Regulación del Desarrollo de la Expresión Génica/genética , Mamíferos/genética
8.
Physiol Genomics ; 56(1): 9-31, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-37842744

RESUMEN

Oocyte maturation is accompanied by changes in abundances of thousands of mRNAs, many degraded and many preferentially stabilized. mRNA stability can be regulated by diverse features including GC content, codon bias, and motifs within the 3'-untranslated region (UTR) interacting with RNA binding proteins (RBPs) and miRNAs. Many studies have identified factors participating in mRNA splicing, bulk mRNA storage, and translational recruitment in mammalian oocytes, but the roles of potentially hundreds of expressed factors, how they regulate cohorts of thousands of mRNAs, and to what extent their functions are conserved across species has not been determined. We performed an extensive in silico cross-species analysis of features associated with mRNAs of different stability classes during oocyte maturation (stable, moderately degraded, and highly degraded) for five mammalian species. Using publicly available RNA sequencing data for germinal vesicle (GV) and MII oocyte transcriptomes, we determined that 3'-UTR length and synonymous codon usage are positively associated with stability, while greater GC content is negatively associated with stability. By applying machine learning and feature selection strategies, we identified RBPs and miRNAs that are predictive of mRNA stability, including some across multiple species and others more species-restricted. The results provide new insight into the mechanisms regulating maternal mRNA stabilization or degradation.NEW & NOTEWORTHY Conservation across species of mRNA features regulating maternal mRNA stability during mammalian oocyte maturation was analyzed. 3'-Untranslated region length and synonymous codon usage are positively associated with stability, while GC content is negatively associated. Just three RNA binding protein motifs were predicted to regulate mRNA stability across all five species examined, but associated pathways and functions are shared, indicating oocytes of different species arrive at comparable physiological destinations via different routes.


Asunto(s)
MicroARNs , ARN Mensajero Almacenado , Animales , Mamíferos/genética , Mamíferos/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Oocitos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Mensajero Almacenado/genética , ARN Mensajero Almacenado/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Regiones no Traducidas , Femenino
9.
J Reprod Dev ; 70(1): 10-17, 2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38057116

RESUMEN

Poly(A)-binding proteins (PABPs) play roles in mRNA maturation, translational activity, and decay. The functions of PABPs, especially PABPN1 and PABPC1, in somatic cells have been well-studied. However, little is known about the roles of PABPs in oocytes because of the unique mechanisms of mRNA metabolism in oocytes. This study focused on PABPN1L and generated Pabpn1l knockout (KO) mice using the CRISPR/Cas9 system. After mating tests, we found that Pabpn1l KO females were infertile due to the failure of the embryos to develop to the 4-cell stage. RNA-seq analysis revealed aberrant mRNA persistence in Pabpn1l KO-MII oocytes, which indicates impaired mRNA degradation during the germinal vesicle (GV) to MII transition. We also revealed that the exogenous expression of Pabpn1l mRNA in KO-GV oocytes recovered defects of embryonic development. PABPN1L is partly indispensable for female fertility in mice, owing to its necessity for embryonic development, which is supported by mRNA degradation during GV to MII maturation.


Asunto(s)
Oocitos , ARN Mensajero Almacenado , Embarazo , Femenino , Animales , Ratones , ARN Mensajero Almacenado/metabolismo , Oocitos/metabolismo , Meiosis , ARN Mensajero/metabolismo , Estabilidad del ARN
10.
Semin Cell Dev Biol ; 154(Pt B): 88-98, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36894378

RESUMEN

Transcriptional and post-transcriptional regulations control gene expression in most cells. However, critical transitions during the development of the female gamete relies exclusively on regulation of mRNA translation in the absence of de novo mRNA synthesis. Specific temporal patterns of maternal mRNA translation are essential for the oocyte progression through meiosis, for generation of a haploid gamete ready for fertilization and for embryo development. In this review, we will discuss how mRNAs are translated during oocyte growth and maturation using mostly a genome-wide perspective. This broad view on how translation is regulated reveals multiple divergent translational control mechanisms required to coordinate protein synthesis with progression through the meiotic cell cycle and with development of a totipotent zygote.


Asunto(s)
Oocitos , Biosíntesis de Proteínas , Oocitos/metabolismo , Oocitos/crecimiento & desarrollo , Humanos , Animales , Biosíntesis de Proteínas/genética , ARN Mensajero Almacenado/genética , ARN Mensajero Almacenado/metabolismo , Femenino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Oogénesis/genética , Genoma/genética , Regulación del Desarrollo de la Expresión Génica/genética , Meiosis/genética
11.
Cell Mol Life Sci ; 80(12): 372, 2023 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-38001238

RESUMEN

Postovulatory aging leads to the decline in oocyte quality and subsequent impairment of embryonic development, thereby reducing the success rate of assisted reproductive technology (ART). Potential preventative strategies preventing oocytes from aging and the associated underlying mechanisms warrant investigation. In this study, we identified that cordycepin, a natural nucleoside analogue, promoted the quality of oocytes aging in vitro, as indicated by reduced oocyte fragmentation, improved spindle/chromosomes morphology and mitochondrial function, as well as increased embryonic developmental competence. Proteomic and RNA sequencing analyses revealed that cordycepin inhibited the degradation of several crucial maternal proteins and mRNAs caused by aging. Strikingly, cordycepin was found to suppress the elevation of DCP1A protein by inhibiting polyadenylation during postovulatory aging, consequently impeding the decapping of maternal mRNAs. In humans, the increased degradation of DCP1A and total mRNA during postovulatory aging was also inhibited by cordycepin. Collectively, our findings demonstrate that cordycepin prevents postovulatory aging of mammalian oocytes by inhibition of maternal mRNAs degradation via suppressing polyadenylation of DCP1A mRNA, thereby promoting oocyte developmental competence.


Asunto(s)
Poliadenilación , ARN Mensajero Almacenado , Humanos , Animales , ARN Mensajero Almacenado/metabolismo , Proteómica , Oocitos/metabolismo , Envejecimiento , ARN Mensajero/genética , ARN Mensajero/metabolismo , Mamíferos/metabolismo , Endorribonucleasas/metabolismo , Transactivadores/metabolismo
12.
Development ; 150(21)2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37767629

RESUMEN

Control of mRNA poly(A) tails is essential for regulation of mRNA metabolism, specifically translation efficiency and mRNA stability. Gene expression in maturing oocytes relies largely on post-transcriptional regulation, as genes are transcriptionally silent during oocyte maturation. The CCR4-NOT complex is a major mammalian deadenylase, which regulates poly(A) tails of maternal mRNAs; however, the function of the CCR4-NOT complex in translational regulation has not been well understood. Here, we show that this complex suppresses translational activity of maternal mRNAs during oocyte maturation. Oocytes lacking all CCR4-NOT deadenylase activity owing to genetic deletion of its catalytic subunits, Cnot7 and Cnot8, showed a large-scale gene expression change caused by increased translational activity during oocyte maturation. Developmental arrest during meiosis I in these oocytes resulted in sterility of oocyte-specific Cnot7 and Cnot8 knockout female mice. We further showed that recruitment of CCR4-NOT to maternal mRNAs is mediated by the 3'UTR element CPE, which suppresses translational activation of maternal mRNAs. We propose that suppression of untimely translational activation of maternal mRNAs via deadenylation by CCR4-NOT is essential for proper oocyte maturation.


Asunto(s)
Oocitos , ARN Mensajero Almacenado , Animales , Ratones , Femenino , ARN Mensajero Almacenado/metabolismo , Oocitos/metabolismo , Oogénesis/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Meiosis , Ratones Noqueados , Mamíferos/genética , Exorribonucleasas/genética , Exorribonucleasas/metabolismo , Proteínas Represoras/metabolismo
13.
J Ovarian Res ; 16(1): 90, 2023 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-37165445

RESUMEN

BACKGROUND: Most of the resources that support the early development of the embryo are stored in the oocyte. Clearing of maternal resources and activation of the embryonic genome to produce its own mRNA transcripts marks the maternal-to-embryo transition. Dependence on stored mRNA can last from a few hours to several days, depending on animal species. The mechanisms regulating stabilization and recruitment of stored maternal transcripts have not yet been described in full detail but are known to involve reversible polyadenylation and modulation of 3'UTR-mediated elements. RNA epigenetic modifications, new players in this field, have an important role in RNA regulation and stabilization. RESULTS: The objectives of this study were first to determine if some of post-transcriptional methylation of stored mRNA is greater in oocytes than in somatic cells. We found that m6A, known to be the most prevalent and involved in various aspects of RNA metabolism and physiological functions, is particularly abundant in porcine oocyte mRNA compared to liver used as a somatic tissue reference. The second objective was to compare the epitranscriptome machinery, such as methyltransferases ("writers"), binding proteins ("readers") and demethylases ("erasers") catalyzing the different process, in follicles and oocytes of different mammalian species by immunofluorescence and confocal microscopy. The expression and localization patterns of these proteins differ between mice, pigs and cows ovaries and oocytes. m5C-associated proteins were generally less abundant. In contrast, m6A-associated proteins were expressed strongly during the early and late stages of folliculogenesis. Transzonal projections were found to contain more granules bearing the m5C mark in mice but both m5C and m6A methylation marks in association with mature oocytes of pigs and cows. Eraser proteins showed the greatest interspecies diversity in terms of distribution in the germinal tissues. CONCLUSIONS: So far, few studies have looked at the oocyte and ovarian epitranscriptomic profile. Our findings indicate that a hitherto unrecognized species-specific layer of transcript regulation occurs at the RNA level and might be consequential during the oocyte transcriptional silencing period.


Asunto(s)
ARN Mensajero Almacenado , ARN , Femenino , Animales , Bovinos , Porcinos , Ratones , ARN/metabolismo , ARN Mensajero Almacenado/metabolismo , Oocitos/metabolismo , Folículo Ovárico/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Mamíferos/genética , Mamíferos/metabolismo
14.
Development ; 150(12)2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37218508

RESUMEN

The accumulation and storage of maternal mRNA is crucial for oocyte maturation and embryonic development. PATL2 is an oocyte-specific RNA-binding protein, and previous studies have confirmed that PATL2 mutation in humans and knockout mice cause oocyte maturation arrest or embryonic development arrest, respectively. However, the physiological function of PATL2 in the process of oocyte maturation and embryonic development is largely unknown. Here, we report that PATL2 is highly expressed in growing oocytes and couples with EIF4E and CPEB1 to regulate maternal mRNA expression in immature oocytes. The germinal vesicle oocytes from Patl2-/- mice exhibit decreasing maternal mRNA expression and reduced levels of protein synthesis. We further confirmed that PATL2 phosphorylation occurs in the oocyte maturation process and identified the S279 phosphorylation site using phosphoproteomics. We found that the S279D mutation decreased the protein level of PATL2 and led to subfertility in Palt2S279D knock-in mice. Our work reveals the previously unrecognized role of PATL2 in regulating the maternal transcriptome and shows that phosphorylation of PATL2 leads to the regulation of PATL2 protein levels via ubiquitin-mediated proteasomal degradation in oocytes.


Asunto(s)
Factor 4E Eucariótico de Iniciación , Proteínas Nucleares , ARN Mensajero Almacenado , Proteínas de Unión al ARN , Animales , Femenino , Humanos , Ratones , Embarazo , Factor 4E Eucariótico de Iniciación/metabolismo , Homeostasis , Ratones Noqueados , Factores de Escisión y Poliadenilación de ARNm/metabolismo , Proteínas Nucleares/metabolismo , Oocitos/metabolismo , ARN Mensajero/metabolismo , ARN Mensajero Almacenado/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo
15.
Genes Dev ; 37(9-10): 418-431, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37257918

RESUMEN

Translation of maternal mRNAs is detected before transcription of zygotic genes and is essential for mammalian embryo development. How certain maternal mRNAs are selected for translation instead of degradation and how this burst of translation affects zygotic genome activation remain unknown. Using gene-edited mice, we document that the oocyte-specific eukaryotic translation initiation factor 4E family member 1b (eIF4E1b) is the regulator of maternal mRNA expression that ensures subsequent reprogramming of the zygotic genome. In oocytes, eIF4E1b binds to transcripts encoding translation machinery proteins, chromatin remodelers, and reprogramming factors to promote their translation in zygotes and protect them from degradation. The protein products are thought to establish an open chromatin landscape in one-cell zygotes to enable transcription of genes required for cleavage stage development. Our results define a program for rapid resetting of the zygotic epigenome that is regulated by maternal mRNA expression and provide new insights into the mammalian maternal-to-zygotic transition.


Asunto(s)
ARN Mensajero Almacenado , Cigoto , Animales , Ratones , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Oocitos , Biosíntesis de Proteínas , ARN Mensajero Almacenado/genética , ARN Mensajero Almacenado/metabolismo , Cigoto/metabolismo
16.
Adv Sci (Weinh) ; 10(18): e2300043, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37083226

RESUMEN

Mammalian oogenesis features reliance on the mRNAs produced and stored during early growth phase. These are essential for producing an oocyte competent to undergo meiotic maturation and embryogenesis later when oocytes are transcriptionally silent. The fate of maternal mRNAs hence ensures the success of oogenesis and the quality of the resulting eggs. Nevertheless, how the fate of maternal mRNAs is determined remains largely elusive. RNA-binding proteins (RBPs) are crucial regulators of oogenesis, yet the identity of the full complement of RBPs expressed in oocytes is unknown. Here, a global view of oocyte-expressed RBPs is presented: mRNA-interactome capture identifies 1396 RBPs in mouse oocytes. An analysis of one of these RBPs, LSM family member 14 (LSM14B), demonstrates that this RBP is specific to oocytes and associated with many networks essential for oogenesis. Deletion of Lsm14b results in female-specific infertility and a phenotype characterized by oocytes incompetent to complete meiosis and early embryogenesis. LSM14B serves as an interaction hub for proteins and mRNAs throughout oocyte development and regulates translation of a subset of its bound mRNAs. Therefore, RNP complexes tethered by LSM14B are found exclusively in oocytes and are essential for the control of maternal mRNA fate and oocyte development.


Asunto(s)
Oocitos , ARN Mensajero Almacenado , Femenino , Animales , Ratones , ARN Mensajero Almacenado/genética , ARN Mensajero Almacenado/metabolismo , Oocitos/metabolismo , Oogénesis/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Mamíferos/metabolismo
17.
Reprod Biol Endocrinol ; 21(1): 40, 2023 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-37101140

RESUMEN

BACKGROUND: Studies have shown that sperm-borne microRNAs (miRNAs) are involved in mammalian preimplantation embryonic development. In humans, spermatozoan miR-34c levels are correlated with in vitro fertilization outcomes, such as embryo quality and the clinical pregnancy and live birth rates. In rabbits and cows, miR-34c improves the developmental competence of embryos generated by somatic cell nuclear transfer. However, the mechanisms underlying the regulation of embryonic development by miR-34c remain unknown. METHODS: Female C57BL/6 mice (6-8 weeks old) were superovulated, and pronucleated zygotes were collected and microinjected with an miR-34c inhibitor or a negative-control RNA. The embryonic development of the microinjected zygotes was evaluated, and the messenger RNA (mRNA) expression profiles of the embryos at the two-cell, four-cell and blastocyst stages (five embryos per group) were determined by RNA sequencing analysis. Gene expression levels were verified by reverse transcription-quantitative polymerase chain reaction. Cluster analysis and heat map visualization were performed to detect differentially expressed mRNAs. Pathway and process enrichment analyses were performed using ontology resources. Differentially expressed mRNAs were systematically analyzed using the Search Tool for the Retrieval of Interacting Genes/Proteins database to determine their biological functions. RESULTS: Embryonic developmental potential was significantly reduced in zygotes microinjected with the miR-34c inhibitor compared with those microinjected with a negative-control RNA. Two-cell stage embryos microinjected with an miR-34c inhibitor presented altered transcriptomic profiles, with upregulated expression of maternal miR-34c target mRNAs and classical maternal mRNAs. Differentially expressed transcripts were mainly of genes associated with lipid metabolism and cellular membrane function at the two-cell stage, with cell-cycle phase transition and energy metabolism at the four-cell stage; and with vesicle organization, lipid biosynthetic process and endomembrane system organization at the blastocyst stage. We also showed that genes related to preimplantation embryonic development, including Alkbh4, Sp1, Mapk14, Sin3a, Sdc1 and Laptm4b, were significantly downregulated after microinjection of an miR-34c inhibitor. CONCLUSIONS: Sperm-borne miR-34c may regulate preimplantation embryonic development by affecting multiple biological processes, such as maternal mRNA degradation, cellular metabolism, cell proliferation and blastocyst implantation. Our data demonstrate the importance of sperm-derived miRNAs in the development of preimplantation embryos.


Asunto(s)
MicroARNs , ARN Mensajero Almacenado , Humanos , Embarazo , Masculino , Animales , Femenino , Ratones , Bovinos , Conejos , ARN Mensajero Almacenado/genética , ARN Mensajero Almacenado/metabolismo , Ratones Endogámicos C57BL , Semen/metabolismo , Desarrollo Embrionario/genética , Espermatozoides/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Blastocisto , ARN Mensajero/genética , ARN Mensajero/metabolismo , Estabilidad del ARN , Mamíferos , Proteínas de la Membrana/metabolismo , Proteínas Oncogénicas/metabolismo
18.
Mol Hum Reprod ; 29(6)2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37068378

RESUMEN

Strategies to maximize individual fertility chances are constant requirements of ART. In vitro folliculogenesis may represent a valid option to create a large source of immature ovarian follicles in ART. Efforts are being made to set up mammalian follicle culture protocols with suitable FSH stimuli. In this study, a new type of recombinant FSH (KN015) with a prolonged half-life is proposed as an alternative to canonical FSH. KN015 supports the in vitro development of mouse follicles from primary to preovulatory stage with higher efficiency than canonical FSH and enhanced post-fertilization development rates of the ovulated oocytes. The use of KN015 also allows us to compare the dynamic transcriptome changes in oocytes and granulosa cells at different stages, in vivo and in vitro. In particular, KN015 facilitates mRNA accumulation in growing mouse oocytes and prevents spontaneous luteinization of granulosa cells in vitro. Novel analyses of transcriptome changes in this study reveal that the in vivo oocytes were more efficient than in vitro oocytes in terms of maternal mRNA clearing during meiotic maturation. KN015 promotes the degradation of maternal mRNA during in vitro oocyte maturation, improves cytoplasmic maturation and, therefore, enhances embryonic developmental potential. These findings establish new transcriptome data for oocyte and granulosa cells at the key stages of follicle development, and should help to widen the use of KN015 as a valid and commercially available hormonal support enabling optimized in vitro development of follicles and oocytes.


Asunto(s)
ARN Mensajero Almacenado , Transcriptoma , Femenino , Ratones , Animales , ARN Mensajero Almacenado/metabolismo , Oogénesis/genética , Oocitos/metabolismo , Células de la Granulosa , Hormona Folículo Estimulante/genética , Hormona Folículo Estimulante/farmacología , Hormona Folículo Estimulante/metabolismo , Meiosis , Mamíferos
19.
Adv Sci (Weinh) ; 10(11): e2205500, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36755190

RESUMEN

Maternal messenger ribonucleic acids (mRNAs) are driven by a highly orchestrated scheme of recruitment to polysomes and translational activation. However, selecting and regulating individual mRNAs for the translation from a competitive pool of mRNAs are little-known processes. This research shows that the maternal eukaryotic translation initiation factor 4e1b (Eif4e1b) expresses during the oocyte-to-embryo transition (OET), and maternal deletion of Eif4e1b leads to multiple defects concerning oogenesis and embryonic developmental competence during OET. The linear amplification of complementary deoxyribonucleic acid (cDNA) ends, and sequencing (LACE-seq) is used to identify the distinct subset of mRNA and its CG-rich binding sites within the 5' untranslated region (UTR) targeted by eIF4E1B. The proteomics analyses indicate that eIF4E1B-specific bound genes show stronger downregulation at the protein level, which further verify a group of proteins that plays a crucial role in oocyte maturation and embryonic developmental competence is insufficiently synthesized in Eif4e1b-cKO oocytes during OET. Moreover, the biochemical results in vitro are combined to further confirm the maternal-specific translation activation model assembled by eIF4E1B and 3'UTR-associated mRNA binding proteins. The findings demonstrate the indispensability of eIF4E1B for selective translation activation in mammalian oocytes and provide a potential network regulated by eIF4E1B in OET.


Asunto(s)
Factor 4E Eucariótico de Iniciación , Oocitos , ARN Mensajero Almacenado , Animales , Ratones , Sitios de Unión , Factor 4E Eucariótico de Iniciación/metabolismo , Mamíferos/metabolismo , Oocitos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Mensajero Almacenado/metabolismo , Proteínas de Unión al ARN/metabolismo
20.
Nat Struct Mol Biol ; 30(2): 200-215, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36646905

RESUMEN

Poly(A)-tail-mediated post-transcriptional regulation of maternal mRNAs is vital in the oocyte-to-embryo transition (OET). Nothing is known about poly(A) tail dynamics during the human OET. Here, we show that poly(A) tail length and internal non-A residues are highly dynamic during the human OET, using poly(A)-inclusive RNA isoform sequencing (PAIso-seq). Unexpectedly, maternal mRNAs undergo global remodeling: after deadenylation or partial degradation into 3'-UTRs, they are re-polyadenylated to produce polyadenylated degradation intermediates, coinciding with massive incorporation of non-A residues, particularly internal long consecutive U residues, into the newly synthesized poly(A) tails. Moreover, TUT4 and TUT7 contribute to the incorporation of these U residues, BTG4-mediated deadenylation produces substrates for maternal mRNA re-polyadenylation, and TENT4A and TENT4B incorporate internal G residues. The maternal mRNA remodeling is further confirmed using PAIso-seq2. Importantly, maternal mRNA remodeling is essential for the first cleavage of human embryos. Together, these findings broaden our understanding of the post-transcriptional regulation of maternal mRNAs during the human OET.


Asunto(s)
Oocitos , ARN Mensajero Almacenado , Humanos , ARN Mensajero Almacenado/metabolismo , Oocitos/metabolismo , ARN Mensajero/metabolismo , Regulación de la Expresión Génica , Poliadenilación , Poli A/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...