Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biol Proced Online ; 26(1): 23, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38987687

RESUMEN

Mammalian oocytes accumulate more than ten thousand mRNAs, of which three to four thousand mRNAs are translationally repressed. The timings and sites of translational activation of these dormant mRNAs are crucial for promoting oocyte maturation and embryonic development. How these mRNAs are accumulated and distributed in oocytes is therefore a fundamental issue to be explored. A method that enables visualization of mRNA molecules with high resolution in a simple manner would be valuable for understanding how oocytes accumulate and regulate the dormant mRNAs. We have developed a highly sensitive whole-mount in situ hybridization method using in vitro-synthesized RNA probes and the tyramide signal amplification (TSA) system optimized for mouse oocytes and embryos. By using this method, Pou5f1/Oct4, Emi2, and cyclin B1 mRNAs were detected in immature oocytes and 2-cell stage embryos. Confocal microscopy showed that these mRNAs formed granular structures in the oocyte cytoplasm. The structures of Pou5f1/Oct4 and cyclin B1 mRNAs persisted in 2-cell stage embryos. Pou5f1/Oct4 RNA granules exhibited a solid-like property in immature oocytes and became liquid-like droplets in 2-cell stage embryos. Double-staining of cyclin B1 mRNA with Emi2 or Pou5f1/Oct4 mRNA revealed that these mRNAs were distributed as different RNA granules without overlapping each other and that the size of cyclin B1 RNA granules tended to be larger than that of Emi2 RNA granules. The structures and distribution patterns of these mRNAs were further analyzed by N-SIM super-resolution microscopy. This analysis revealed that the large-sized RNA granules consist of many small-sized granules, suggesting the accumulation and regulation of dormant mRNAs as basal-sized RNA granules. The method established in this study can easily visualize the structure and distribution of mRNAs accumulated in mammalian oocytes and embryos with high sensitivity and super-resolution. This method is useful for investigating the cellular and molecular mechanisms of translational control of mRNAs by which maturation and early developmental processes are promoted.

2.
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
3.
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
4.
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
5.
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
6.
Reprod Fertil ; 4(2)2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36943184

RESUMEN

Abstract: The transition of maternal to zygotic gene expression regulation is critical for human preimplantation embryo development. In recent years, single-cell RNA sequencing (scRNA-seq) had been applied to detect the factors that regulate human oocyte maturation and early embryo development. Here, the evaluation of transcriptomes in single blastomere from the embryo collected from patients by scRNA-seq was performed. There were 20 blastomeres biopsied from 8-cell embryos of seven patients who received more than two ART cycles due to low embryo competence. Meanwhile, ten cells were collected from 8-cell embryos of four patients who received ART treatment due to male or tubal factors. The blastomeres were then evaluated using the previously established scRNA-seq method to determine the associations between their gene expression and developmental competence. The total number of genes detected in 8-cell embryos that failed to form blastocyst including maternal and zygotic mRNAs was reduced. There were 324 differently expressed genes detected among the 8-cell embryos including 65 genes that were significantly suppressed in the 8-cell embryos that failed to form blastocyst. Further analysis found these 8-cell embryos arrested at the cleavage stage due to the dysfunction of the cell cycle, DNA transcription activity, histone methylation, and cell division-related genes such as SMCO-1, ZNF271P,ZNF679, ASF1b, BEX3, DPPA2, and ORC4. The alterations of gene expression detected in human 8-cell embryos are tightly associated with its developmental competence and could be used as targets to enhance embryo development or parameters to predict the embryo's development outcomes. Lay summary: Many females are suffering infertility due to the failure of embryonic development at early stages due to unknown causes. At the very beginning of human embryo development, the embryos start to express its own genes, which should be achieved at 8-cell stage. In current research, we isolated one cell from 8-cell embryos and detected the gene expression at single-cell level. Then the remaining cells of these embryos were cultured to form blastocyst. Meanwhile, the data was analyzed according to the outcomes of embryo development. We detected 324 differently expressed genes between the 8-cell embryos that succeeded and failed to form blastocyst. Our research showed the association between the gene expression and the developmental competence of 8-cell embryos. The findings could be used to predict the embryo quality and potential therapy target to improve the efficiency of assisted reproductive techniques.


Asunto(s)
Blastocisto , Desarrollo Embrionario , Embarazo , Femenino , Humanos , Masculino , Animales , Desarrollo Embrionario/genética , Blastocisto/metabolismo , Blastómeros/fisiología , Embrión de Mamíferos , Análisis de Secuencia de ARN/veterinaria , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo
7.
EMBO J ; 42(3): e111364, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36477743

RESUMEN

Early embryonic development depends on proper utilization and clearance of maternal transcriptomes. How these processes are spatiotemporally regulated remains unclear. Here we show that nuclear RNA-binding protein Rbm14 and maternal mRNAs co-phase separate into cytoplasmic condensates to facilitate vertebrate blastula-to-gastrula development. In zebrafish, Rbm14 condensates were highly abundant in blastomeres and markedly reduced after prominent activation of zygotic transcription. They concentrated at spindle poles by associating with centrosomal γ-tubulin puncta and displayed mainly asymmetric divisions with a global symmetry across embryonic midline in 8- and 16-cell embryos. Their formation was dose-dependently stimulated by m6 A, but repressed by m5 C modification of the maternal mRNA. Furthermore, deadenylase Parn co-phase separated with these condensates, and this was required for deadenylation of the mRNAs in early blastomeres. Depletion of Rbm14 impaired embryonic cell differentiations and full activations of the zygotic genome in both zebrafish and mouse and resulted in developmental arrest at the blastula stage. Our results suggest that cytoplasmic Rbm14 condensate formation regulates early embryogenesis by facilitating deadenylation, protection, and mitotic allocation of m6 A-modified maternal mRNAs, and by releasing the poly(A)-less transcripts upon regulated disassembly to allow their re-polyadenylation and translation or clearance.


Asunto(s)
ARN Mensajero Almacenado , Pez Cebra , Animales , Femenino , Ratones , Embarazo , Blastocisto/metabolismo , Blástula/metabolismo , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Mensajero Almacenado/genética , ARN Mensajero Almacenado/metabolismo
8.
Int J Mol Sci ; 23(15)2022 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-35955697

RESUMEN

Maternal-to-zygotic transition (MZT) of the control of early post-fertilization development is a key-event conditioning the fate of the future embryo, fetus and newborn. Because of the relative paucity of data concerning human embryos, due to ethical concerns and the poor availability of human embryos donated for research, most data have to be derived from animal models, among which those obtained using mouse embryos are most prevalent. However, data obtained by studies performed in non-mammalian specie can also provide useful information. For this reason, this review focuses on similarities and differences of MZT control mechanisms in humans and other species, with particular attention to the mouse. A number of molecular pathways controlling MZT in mice and humans are compared, pointing out those that could be at the origin of further focused experimental studies and the development of new diagnostic tools based on the translational medicine principles. Data concerning possible candidate molecules to be included in these studies are identified.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Cigoto , Animales , Embrión de Mamíferos , Desarrollo Embrionario/genética , Humanos , Ratones , Cigoto/metabolismo
9.
J Dev Biol ; 10(1)2022 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-35225963

RESUMEN

In many animal species, the body axis is determined by the relocalization of maternal determinants, organelles, or unique cell populations in a cytoskeleton-dependent manner. In the ascidian first cell cycle, the myoplasm, including mitochondria, endoplasmic reticulum (ER), and maternal mRNAs, move to the future posterior side concomitantly (called ooplasmic segregation or cytoplasmic and cortical reorganization). This translocation consists of first and second phases depending on the actin and microtubule, respectively. However, the transition from first to second phase, that is, translocation of myoplasmic components from microfilaments to microtubules, has been poorly investigated. In this study, we analyzed the relationship between these cytoskeletons and myoplasmic components during the first cell cycle and their role in morphogenesis by inhibitor experiments. Owing to our improved visualization techniques, there was unexpected F-actin accumulation at the vegetal pole during this transition period. When this F-actin was depolymerized, the microtubule structure was strongly affected, the myoplasmic components, including maternal mRNA, were mislocalized, and the anteroposterior axis formation was disordered. These results suggested the importance of F-actin during the first cell cycle and the existence of interactions between microfilaments and microtubules, implying the enigmatic mechanism of ooplasmic segregation. Solving this mystery leads us to an improved understanding of ascidian early development.

10.
Dev Genes Evol ; 232(1): 1-14, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34921621

RESUMEN

Axis formation is one of the most important events occurring at the beginning of animal development. In the ascidian egg, the antero-posterior axis is established at this time owing to a dynamic cytoplasmic movement called cytoplasmic and cortical reorganisation. During this movement, mitochondria, endoplasmic reticulum (ER), and maternal mRNAs (postplasmic/PEM RNAs) are translocated to the future posterior side. Although accumulating evidence indicates the crucial roles played by the asymmetrical localisation of these organelles and the translational regulation of postplasmic/PEM RNAs, the organisation of ER has not been described in sufficient detail to date owing to technical difficulties. In this study, we developed three different multiple staining protocols for visualising the ER in combination with mitochondria, microtubules, or mRNAs in whole-mount specimens. We defined the internally expanded "dense ER" using these protocols and described cisterna-like structures of the dense ER using focused ion beam-scanning electron microscopy. Most importantly, we described the dynamic changes in the colocalisation of postplasmic/PEM mRNAs and dense ER; for example, macho-1 mRNA was detached and excluded from the dense ER during the second phase of ooplasmic movements. These detailed descriptions of the association between maternal mRNA and ER can provide clues for understanding the translational regulation mechanisms underlying axis determination during ascidian early embryogenesis.


Asunto(s)
ARN Mensajero Almacenado , Urocordados , Animales , Desarrollo Embrionario/genética , Retículo Endoplásmico , Oocitos , ARN Mensajero/genética , ARN Mensajero Almacenado/genética , Urocordados/genética
11.
Biol Reprod ; 106(1): 83-94, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-34726234

RESUMEN

Infertility affects 10-15% of families worldwide. However, the pathogenesis of female infertility caused by abnormal early embryonic development is not clear. A recent study showed that poly(A)binding protein nuclear 1-like (PABPN1L) recruited BTG anti-proliferation factor 4 (BTG4) to mRNA 3'-poly(A) tails and was essential for maternal mRNA degradation. Here, we generated a PABPN1L-antibody and found "ring-like" PABPN1L aggregates in the cytoplasm of MII oocytes. PABPN1L-EGFP proteins spontaneously formed "ring-like" aggregates in vitro. This phenomenon is similar with CCR4-NOT catalytic subunit, CCR4-NOT transcription complex subunit 7 (CNOT7), when it starts deadenylation process in vitro. We constructed two mouse model (Pabpn1l-/- and Pabpn1l  tm1a/tm1a) simulating the intron 1-exon 2 abnormality of human PABPN1L and found that the female was sterile and the male was fertile. Using RNA-Seq, we observed a large-scale up-regulation of RNA in zygotes derived from Pabpn1l-/- MII oocytes. We found that 9222 genes were up-regulated instead of being degraded in the Pabpn1l-♀/+♂zygote. Both the Btg4 and CCR4-NOT transcription complex subunit 6 like (Cnot6l) genes are necessary for the deadenylation process and Pabpn1l-/- resembled both the Btg4 and Cnot6l knockouts, where 71.2% genes stabilized in the Btg4-♀/+♂ zygote and 84.2% genes stabilized in the Cnot6l-♀/+♂zygote were also stabilized in Pabpn1l-♀/+♂ zygote. BTG4/CNOT7/CNOT6L was partially co-located with PABPN1L in MII oocytes. The above results suggest that PABPN1L is widely associated with CCR4-NOT-mediated maternal mRNA degradation and PABPN1L variants on intron 1-exon 2 could be a genetic marker of female infertility.


Asunto(s)
Citoplasma/química , Oocitos/ultraestructura , Proteína I de Unión a Poli(A)/química , Proteína I de Unión a Poli(A)/fisiología , Agregado de Proteínas , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas Fluorescentes Verdes/química , Humanos , Infertilidad Femenina , Masculino , Ratones , Ratones Noqueados , Proteína I de Unión a Poli(A)/genética , Proteínas de Unión a Poli(A)/química , Proteínas de Unión a Poli(A)/genética , ARN Mensajero/metabolismo , Receptores CCR4/genética , Receptores CCR4/fisiología , Cigoto/metabolismo
12.
EMBO Mol Med ; 13(12): e14887, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34779126

RESUMEN

Early embryonic arrest and fragmentation (EEAF) is a common phenomenon leading to female infertility, but the genetic determinants remain largely unknown. The Moloney sarcoma oncogene (MOS) encodes a serine/threonine kinase that activates the ERK signaling cascade during oocyte maturation in vertebrates. Here, we identified four rare variants of MOS in three infertile female individuals with EEAF that followed a recessive inheritance pattern. These MOS variants encoded proteins that resulted in decreased phosphorylated ERK1/2 level in cells and oocytes, and displayed attenuated rescuing effects on cortical F-actin assembly. Using oocyte-specific Erk1/2 knockout mice, we verified that MOS-ERK signal pathway inactivation in oocytes caused EEAF as human. The RNA sequencing data revealed that maternal mRNA clearance was disrupted in human mature oocytes either with MOS homozygous variant or with U0126 treatment, especially genes relative to mitochondrial function. Mitochondrial dysfunction was observed in oocytes with ERK1/2 deficiency or inactivation. In conclusion, this study not only uncovers biallelic MOS variants causes EEAF but also demonstrates that MOS-ERK signaling pathway drives human oocyte cytoplasmic maturation to prevent EEAF.


Asunto(s)
Infertilidad Femenina , Sarcoma , Animales , Femenino , Humanos , Infertilidad Femenina/genética , Infertilidad Femenina/metabolismo , Ratones , Mutación , Oncogenes , Oocitos , Sarcoma/genética , Sarcoma/metabolismo
13.
FASEB J ; 35(11): e22006, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34694646

RESUMEN

The goal of preimplantation development is to establish the fates of the embryonic and extra-embryonic cells. However, when and how cell fates are determined during early mammalian embryonic development remains unclear. We report that the high mobility group (HMG) protein family member HMGA1 was distributed differentially in mouse two-cell blastomeres. Knockdown of Hmga1 expression in one of the two cells reduced the number of cells contributing to the inner cell mass (ICM), suggesting that differential distribution of HMGA1 in the blastomeres in two-cell mouse embryos affected the selection of embryonic cell lineages. Mechanistically, HMGA1 promotes the expression of the ICM-specific gene Sox2. The results of this study show that mouse embryos demonstrate heterogeneity as early as the two-cell stage, and that these differences are related to cell-fate differentiation in early mouse embryos.


Asunto(s)
Embrión de Mamíferos/embriología , Desarrollo Embrionario , Proteína HMGA1a/metabolismo , Oocitos , ARN Mensajero Almacenado/metabolismo , Animales , Diferenciación Celular , Línea Celular Tumoral , Femenino , Regulación del Desarrollo de la Expresión Génica , Ratones , Oocitos/citología , Oocitos/metabolismo , Embarazo
14.
Mol Hum Reprod ; 27(7)2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34152407

RESUMEN

Mammalian oocytes and embryos rely exclusively on maternal mRNAs to accomplish early developmental processes. Since oocytes and early embryos are transcriptionally silent after meiotic resumption, most of the synthesised maternal mRNA does not undergo immediate translation but is instead stored in the oocyte. Quantitative RT-PCR is commonly used to quantify mRNA levels, and correct quantification relies on reverse transcription and the choice of reference genes. Different methods for reverse transcription may affect gene expression determination in oocytes. In this study, we examined the suitability of either random or oligo(dT) primers for reverse transcription to be used for quantitative RT-PCR. We further looked for changes in poly(A) length of the maternal mRNAs during oocyte maturation. Our data indicate that depending on the method of reverse transcription, the optimal combination of reference genes for normalisation differed. Surprisingly, we observed a shortening of the poly(A) tail lengths of maternal mRNA as oocytes progressed from germinal vesicle to metaphase II. Overall, our findings suggest dynamic maternal regulation of mRNA structure and gene expression during oocyte maturation and early embryo development.


Asunto(s)
Blastómeros/metabolismo , Cartilla de ADN , Regulación del Desarrollo de la Expresión Génica , Mórula/metabolismo , Oocitos/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Transcripción Reversa , Cigoto/metabolismo , Animales , Bovinos , Cartilla de ADN/síntesis química , ADN Complementario/genética , Técnicas de Cultivo de Embriones , Genes , Poli A/análisis , ARN Mensajero/genética , ARN Mensajero/aislamiento & purificación , Estándares de Referencia , Creación de Embriones para Investigación , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico
15.
Am J Physiol Cell Physiol ; 321(1): C3-C16, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33881934

RESUMEN

Oogenesis is a complex process resulting in the production of a truly remarkable cell-the oocyte. Oocytes execute many unique processes and functions such as meiotic segregation of maternal genetic material, and essential life-generating functions after fertilization including posttranscriptional support of essential homeostatic and metabolic processes, and activation and reprogramming of the embryonic genome. An essential goal for understanding female fertility and infertility in mammals is to discover critical features driving the production of quality oocytes, particularly the complex regulation of oocyte maternal mRNAs. We report here the first in-depth meta-analysis of oocyte maturation-associated transcriptome changes, using eight datasets encompassing 94 RNAseq libraries for human, rhesus monkey, mouse, and cow. A majority of maternal mRNAs are regulated in a species-restricted manner, highlighting considerable divergence in oocyte transcriptome handling during maturation. We identified 121 mRNAs changing in relative abundance similarly across all four species (92 of high homology), and 993 (670 high homology) mRNAs regulated similarly in at least three of the four species, corresponding to just 0.84% and 6.9% of mRNAs analyzed. Ingenuity Pathway Analysis (IPA) revealed an association of these shared mRNAs with many shared pathways and functions, most prominently oxidative phosphorylation and mitochondrial function. These shared functions were reinforced further by primate-specific and species-specific differentially expressed genes (DEGs). Thus, correct downregulation of mRNAs related to oxidative phosphorylation and mitochondrial function is a major shared feature of mammalian oocyte maturation.


Asunto(s)
Fertilidad/genética , Mitocondrias/genética , Oocitos/metabolismo , Oogénesis/genética , ARN Mensajero/genética , Transcriptoma , Animales , Bovinos , Femenino , Biblioteca de Genes , Ontología de Genes , Humanos , Macaca mulatta , Meiosis , Ratones , Mitocondrias/metabolismo , Anotación de Secuencia Molecular , Oocitos/citología , Oocitos/crecimiento & desarrollo , Fosforilación Oxidativa , ARN Mensajero/clasificación , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN , Especificidad de la Especie
16.
Theriogenology ; 166: 104-111, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33721681

RESUMEN

Energy homeostasis and accomplishment of maternal-to-zygotic transition (MZT), which involves the timed processes of maternal mRNA clearance and zygotic genome activation (ZGA), are essential for mammalian embryogenesis. However, how energy substrates regulate maternal mRNA clearance and the underlying mechanisms have not yet been fully elucidated. Here, we found that mouse embryos were arrested at the 2-cell stage when the pyruvate level was reduced to one-fifth of the control level. Moreover, we observed that the mitochondrial contents and ROS levels were reduced. Interestingly, some maternal mRNA, including transcripts involved in the maternal factor-mediated mRNA decay (M-decay) pathway, was vastly degraded from 1-cell to 2-/4-cell embryos when cultured with control pyruvate levels, but the clearance of these transcripts was hindered when the pyruvate level was reduced. In contrast, some transcripts involved in the zygotic factor-mediated mRNA decay (Z-decay) pathway were vastly downregulated by the reduction in pyruvate. This effect was possibly due to a reduction in global transcription, as the embryos cultured with low-level pyruvate had lower transcription activity than embryos cultured with control pyruvate level. In summary, our findings demonstrate that low-level pyruvate inhibits maternal mRNA clearance, possibly by disrupting the M- and Z-decay pathways, extending our current understanding of the energy requirements of embryogenesis.


Asunto(s)
Ácido Pirúvico , ARN Mensajero Almacenado , Animales , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Ratones , Cigoto
17.
J Cell Sci ; 134(3)2021 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-33408246

RESUMEN

Replication-dependent histone mRNAs are the only cellular mRNAs that are not polyadenylated, ending in a stemloop instead of a polyA tail, and are normally regulated coordinately with DNA replication. Stemloop-binding protein (SLBP) binds the 3' end of histone mRNA, and is required for processing and translation. During Drosophila oogenesis, large amounts of histone mRNAs and proteins are deposited in the developing oocyte. The maternally deposited histone mRNA is synthesized in stage 10B oocytes after the nurse cells complete endoreduplication. We report that in wild-type stage 10B oocytes, the histone locus bodies (HLBs), formed on the histone genes, produce histone mRNAs in the absence of phosphorylation of Mxc, which is normally required for histone gene expression in S-phase cells. Two mutants of SLBP, one with reduced expression and another with a 10-amino-acid deletion, fail to deposit sufficient histone mRNA in the oocyte, and do not transcribe the histone genes in stage 10B. Mutations in a putative SLBP nuclear localization sequence overlapping the deletion phenocopy the deletion. We conclude that a high concentration of SLBP in the nucleus of stage 10B oocytes is essential for histone gene transcription.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Proteínas de Drosophila , Histonas , Animales , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Histonas/genética , Proteínas Nucleares/metabolismo , ARN Mensajero/genética , Proteínas de Unión al ARN , Proteínas Supresoras de Tumor , Factores de Escisión y Poliadenilación de ARNm
18.
Front Cell Dev Biol ; 8: 580367, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33117808

RESUMEN

Maternal mRNA clearance is critical for the early embryo development, which is under the tight control of RNA N6-methyladenosine (m6A). However, little information is known regarding the maternal mRNA clearance and mechanisms behind it in farm animals. In the present study, 3362 differentially expressed genes (DEGs) were found during the maternal-to-zygotic transition (MZT) and determined as maternal mRNAs in goat. Of which, 1961 was decreased at the 4-cell stage embryos, while 1401 was trigged down-regulation at the 8-cell stage embryos, which were termed as maternally encoded mRNA decay genes and zygotic genome activation (ZGA)-dependent maternal mRNAs, respectively. The expression of m6A reader YTHDF2 was increased during goat ZGA, and knockdown of YTHDF2 resulted in decreased blastocyst rate. In the 8-cell stage YTHDF2 knockdown embryos, the M-decay and Z-decay maternal mRNA clearance were impaired. Specifically, the expression of deadenylase (CNOT1 and CNOT11) and decapping enzymes (DCP1A and DCP2) was decreased. In conclusion, we ascertained maternal mRNAs and inferred that maternal mRNA clearance is also ZGA-dependent in goat. We reported that YTHDF2 is vital for goat early embryogenesis as it advances maternal mRNA clearance, which might through the recruitment of deadenylases and mRNA decapping enzymes. This work will be of great value for understanding the stochastic reprogramming events during MZT and achieving better development of goat embryos in vitro.

19.
Genetics ; 216(3): 805-821, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32928902

RESUMEN

How gene expression can evolve depends on the mechanisms driving gene expression. Gene expression is controlled in different ways in different developmental stages; here we ask whether different developmental stages show different patterns of regulatory evolution. To explore the mode of regulatory evolution, we used the early stages of embryonic development controlled by two different genomes, that of the mother and that of the zygote. During embryogenesis in all animals, initial developmental processes are driven entirely by maternally provided gene products deposited into the oocyte. The zygotic genome is activated later, when developmental control is handed off from maternal gene products to the zygote during the maternal-to-zygotic transition. Using hybrid crosses between sister species of Drosophila (Dsimulans, D. sechellia, and D. mauritiana) and transcriptomics, we find that the regulation of maternal transcript deposition and zygotic transcription evolve through different mechanisms. We find that patterns of transcript level inheritance in hybrids, relative to parental species, differ between maternal and zygotic transcripts, and maternal transcript levels are more likely to be conserved. Changes in transcript levels occur predominantly through differences in trans regulation for maternal genes, while changes in zygotic transcription occur through a combination of both cis and trans regulatory changes. Differences in the underlying regulatory landscape in the mother and the zygote are likely the primary determinants for how maternal and zygotic transcripts evolve.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Hibridación Genética , Oocitos/metabolismo , Transcriptoma , Cigoto/metabolismo , Animales , Drosophila , Evolución Molecular , Patrón de Herencia
20.
Mol Cell ; 75(6): 1188-1202.e11, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31399345

RESUMEN

The maternal-to-zygotic transition (MZT) is a conserved and fundamental process during which the maternal environment is converted to an environment of embryonic-driven development through dramatic reprogramming. However, how maternally supplied transcripts are dynamically regulated during MZT remains largely unknown. Herein, through genome-wide profiling of RNA 5-methylcytosine (m5C) modification in zebrafish early embryos, we found that m5C-modified maternal mRNAs display higher stability than non-m5C-modified mRNAs during MZT. We discovered that Y-box binding protein 1 (Ybx1) preferentially recognizes m5C-modified mRNAs through π-π interactions with a key residue, Trp45, in Ybx1's cold shock domain (CSD), which plays essential roles in maternal mRNA stability and early embryogenesis of zebrafish. Together with the mRNA stabilizer Pabpc1a, Ybx1 promotes the stability of its target mRNAs in an m5C-dependent manner. Our study demonstrates an unexpected mechanism of RNA m5C-regulated maternal mRNA stabilization during zebrafish MZT, highlighting the critical role of m5C mRNA modification in early development.


Asunto(s)
5-Metilcitosina/metabolismo , Embrión no Mamífero/embriología , Desarrollo Embrionario/fisiología , Estabilidad del ARN/fisiología , ARN Mensajero Almacenado/metabolismo , Pez Cebra/embriología , Animales , Células HeLa , Humanos , Ratones , ARN Mensajero Almacenado/genética , Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA