Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Reproduction ; 164(4): R75-R86, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35900353

RESUMO

In brief: Trophectoderm is the first tissue to differentiate in the early mammalian embryo and is essential for hatching, implantation, and placentation. This review article discusses the roles of Ras homolog family members (RHO) and RHO-associated coiled-coil containing protein kinases (ROCK) in the molecular and cellular regulation of trophectoderm formation. Abstract: The trophectoderm (TE) is the first tissue to differentiate during the preimplantation development of placental mammals. It constitutes the outer epithelial layer of the blastocyst and is responsible for hatching, uterine attachment, and placentation. Thus, its formation is the key initial step that enables the viviparity of mammals. Here, we first describe the general features of TE formation at the morphological and molecular levels. Prospective TE cells form an epithelial layer enclosing an expanding fluid-filled cavity by establishing the apical-basal cell polarity, intercellular junctions, microlumen, and osmotic gradient. A unique set of genes is expressed in TE that encode the transcription factors essential for the development of trophoblasts of the placenta upon implantation. TE-specific gene expressions are driven by the inhibition of HIPPO signaling, which is dependent on the prior establishment of the apical-basal polarity. We then discuss the specific roles of RHO and ROCK as essential regulators of TE formation. RHO and ROCK modulate the actomyosin cytoskeleton, apical-basal polarity, intercellular junctions, and HIPPO signaling, thereby orchestrating the epithelialization and gene expressions in TE. Knowledge of the molecular mechanisms underlying TE formation is crucial for assisted reproductive technologies in human and farm animals, as it provides foundation to help improve procedures for embryo handling and selection to achieve better reproductive outcomes.


Assuntos
Polaridade Celular , Via de Sinalização Hippo , Animais , Blastocisto/metabolismo , Polaridade Celular/fisiologia , Feminino , Expressão Gênica , Humanos , Mamíferos/genética , Morfogênese , Placenta/metabolismo , Gravidez , Estudos Prospectivos , Proteínas rho de Ligação ao GTP/metabolismo , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo
2.
Mol Hum Reprod ; 27(4)2021 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-33677573

RESUMO

Early embryos are vulnerable to environmental insults, such as medications taken by the mother. Due to increasing prevalence of hypercholesterolemia, more women of childbearing potential are taking cholesterol-lowering medications called statins. Previously, we showed that inhibition of the mevalonate pathway by statins impaired mouse preimplantation development, by modulating HIPPO signaling, a key regulator for trophectoderm (TE) lineage specification. Here, we further evaluated molecular events that are altered by mevalonate pathway inhibition during the timeframe of morphogenesis and cell lineage specification. Whole transcriptome analysis revealed that statin treatment dysregulated gene expression underlying multiple processes, including cholesterol biosynthesis, HIPPO signaling, cell lineage specification and endoplasmic reticulum (ER) stress response. We explored mechanisms that link the mevalonate pathway to ER stress, because of its potential impact on embryonic health and development. Upregulation of ER stress-responsive genes was inhibited when statin-treated embryos were supplemented with the mevalonate pathway product, geranylgeranyl pyrophosphate (GGPP). Inhibition of geranylgeranylation was sufficient to upregulate ER stress-responsive genes. However, ER stress-responsive genes were not upregulated by inhibition of ras homolog family member A (RHOA), a geranylgeranylation target, although it interfered with TE specification and blastocyst cavity formation. In contrast, inhibition of Rac family small GTPase 1 (RAC1), another geranylgeranylation target, upregulated ER stress-responsive genes, while it did not impair TE specification or cavity formation. Thus, our study suggests that the mevalonate pathway regulates cellular homeostasis (ER stress repression) and differentiation (TE lineage specification) in preimplantation embryos through GGPP-dependent activation of two distinct small GTPases, RAC1 and RHOA, respectively. Translation of the findings to human embryos and clinical settings requires further investigations.


Assuntos
Estresse do Retículo Endoplasmático , Ácido Mevalônico , Animais , Blastocisto/metabolismo , Linhagem da Célula , Embrião de Mamíferos , Desenvolvimento Embrionário/fisiologia , Estresse do Retículo Endoplasmático/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Ácido Mevalônico/farmacologia , Camundongos
3.
Mol Hum Reprod ; 25(2): 43-60, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30395288

RESUMO

STUDY QUESTION: What molecular signals are required to maintain the functional trophectoderm (TE) during blastocyst expansion of the late stage of preimplantation development? SUMMARY ANSWER: The activity of ras homology family member A (RHOA) GTPases is necessary to retain the expanded blastocyst cavity and also to sustain the gene expression program specific to TE. WHAT IS KNOWN ALREADY: At the early stages of preimplantation development, the precursor of the TE lineage is generated through the molecular signals that integrate RHOA, RHO-associated coiled-coil containing protein kinase (ROCK), the apicobasal cell polarity, and the HIPPO-Yes-associated protein (YAP) signaling pathway. By contrast, molecular mechanisms regulating the maintenance of the TE characteristics at the later stage, which is crucial for blastocyst hatching and implantation, are scarcely understood. STUDY DESIGN, SIZE, DURATION: Expanding mouse blastocysts, obtained from crosses of the F1 (C57BL6 × DBA/2) strain, were exposed to chemical agents that interfere with RHOA, ROCK, or the actin cytoskeleton for up to 8 h, and effects on the blastocyst cavity, HIPPO-YAP signaling, and cell lineage-specific gene expression profiles were examined. PARTICIPANTS/MATERIALS, SETTING, METHODS: Mouse embryos at the embryonic stage E3.5 (expanding blastocysts) and E4.5 (fully expanded blastocysts) were treated with RHOA inhibitor (C3 exoenzyme), ROCK inhibitor (Y27632), or actin filament disruptors (cytochalasin B and latrunculin A). The integrity of the blastocyst cavity was evaluated based on the gross morphology. Effects on HIPPO-YAP signaling were assessed based on the presence of nuclearized YAP protein by immunofluorescence staining and the expression of YAP/TEA domain family member (TEAD) target genes by quantitative RT-PCR (qRT-PCR). The impact of these disruptors on cell lineages was evaluated based on expression of the TE-specific and inner cell mass-specific marker genes by qRT-PCR. The integrity of the apicobasal cell polarity was assessed by localization of protein kinase C zeta (PRKCZ; apical) and scribbled planar cell polarity (SCRIB; basal) proteins by immunofluorescence staining. For comparisons, cultured cell lines, NIH/3T3 (mouse fibroblast) and P19C5 (mouse embryonal carcinoma), were also treated with RHOA inhibitor, ROCK inhibitor, and actin filament disruptors for up to 8 h, and effects on HIPPO-YAP signaling were assessed based on expression of YAP/TEAD target genes by qRT-PCR. Each experiment was repeated using three independent batches of embryos (n = 40-80 per batch) or cell collections. Statistical analyses of data were performed, using one-way ANOVA and two-sample t-test. MAIN RESULTS AND THE ROLE OF CHANCE: Inhibition of RHOA deflated the cavity, diminished nuclear YAP (P < 0.01), and down-regulated the YAP/TEAD target and TE-specific marker genes in both E3.5 and E4.5 blastocysts (P < 0.05), indicating that the maintenance of the key TE characteristics is dependent on RHOA activity. However, inhibition of ROCK or disruption of actin filament only deflated the blastocyst cavity, but did not alter HIPPO-YAP signaling or lineage-specific gene expressions, suggesting that the action of RHOA to sustain the TE-specific gene expression program is not mediated by ROCK or the actomyosin cytoskeleton. By contrast, ROCK inhibitor and actin filament disruptors diminished YAP/TEAD target gene expressions in cultured cells to a greater extent than RHOA inhibitor, implicating that the regulation of HIPPO-YAP signaling in expanding blastocysts is distinctly different from that in the cell lines. Furthermore, the apicobasal cell polarity proteins in the expanding blastocyst were mislocalized by ROCK inhibition but not by RHOA inhibition, indicating that cell polarity is not linked to regulation of HIPPO-YAP signaling. Taken together, our study suggests that RHOA activity is essential to maintain the TE lineage in the expanding blastocyst and it regulates HIPPO-YAP signaling and the lineage-specific gene expression program through mechanisms that are independent of ROCK or actomyosin cytoskeleton. LARGE-SCALE DATA: Not applicable. LIMITATIONS, REASONS FOR CAUTION: This study was conducted using one species, the mouse. Direct translation of the experiments and findings to human fertility preservation and ART requires further investigations. WIDER IMPLICATIONS OF THE FINDINGS: The elucidation of the mechanisms of TE formation is highly pertinent to fertility preservation in women. Our findings may raise awareness among providers of ART that the TE is sensitive to disturbance even in the late stage of blastocyst expansion and that rational approaches should be devised to avoid conditions that may impair the TE and its function. STUDY FUNDING/COMPETING INTEREST(S): This study was funded by grants from the Ingeborg v.F. McKee Fund of the Hawaii Community Foundation (16ADVC-78882 to V.B.A.), and the National Institutes of Health (P20 GM103457 and R03 HD088839 to V.B.A.). The authors have no conflict of interest to declare.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Blastocisto/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas de Ciclo Celular/genética , Linhagem Celular , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Feminino , Via de Sinalização Hippo , Camundongos , Microscopia Confocal , Células NIH 3T3 , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Proteínas de Sinalização YAP , Proteína rhoA de Ligação ao GTP/genética
4.
Adv Anat Embryol Cell Biol ; 229: 47-68, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29177764

RESUMO

In placental mammalian development, the first cell differentiation produces two distinct lineages that emerge according to their position within the embryo: the trophectoderm (TE, placenta precursor) differentiates in the surface, while the inner cell mass (ICM, fetal body precursor) forms inside. Here, we discuss how such position-dependent lineage specifications are regulated by the RHOA subfamily of small GTPases and RHO-associated coiled-coil kinases (ROCK). Recent studies in mouse show that activities of RHO/ROCK are required to promote TE differentiation and to concomitantly suppress ICM formation. RHO/ROCK operate through the HIPPO signaling pathway, whose cell position-specific modulation is central to establishing unique gene expression profiles that confer cell fate. In particular, activities of RHO/ROCK are essential in outside cells to promote nuclear localization of transcriptional co-activators YAP/TAZ, the downstream effectors of HIPPO signaling. Nuclear localization of YAP/TAZ depends on the formation of apicobasal polarity in outside cells, which requires activities of RHO/ROCK. We propose models of how RHO/ROCK regulate lineage specification and lay out challenges for future investigations to deepen our understanding of the roles of RHO/ROCK in preimplantation development. Finally, as RHO/ROCK may be inhibited by certain pharmacological agents, we discuss their potential impact on human preimplantation development in relation to fertility preservation in women.


Assuntos
Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Quinases Associadas a rho , Animais , Blastocisto , Diferenciação Celular , Embrião de Mamíferos , Feminino , Camundongos , Gravidez , Transdução de Sinais , Quinases Associadas a rho/metabolismo
5.
Mol Hum Reprod ; 22(5): 350-63, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26908642

RESUMO

STUDY HYPOTHESIS: Statins, inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase of the mevalonate pathway and prescription drugs that treat hypercholesterolemia, compromise preimplantation mouse development via modulation of HIPPO signaling. STUDY FINDING: HMG-CoA reductase activity is required for trophectoderm specification, namely blastocyst cavity formation and Yes-associated protein (YAP) nuclear localization, through the production of isoprenoid geranylgeranyl pyrophosphate (GGPP) and the action of geranylgeranyl transferase. WHAT IS KNOWN ALREADY: Previous studies have shown that treatment of mouse embryos with mevastatin prevents blastocyst formation, but how HMG-CoA reductase is involved in preimplantation development is unknown. HIPPO signaling regulates specification of the trophectoderm lineage of the mouse blastocyst by controlling the nuclear localization of YAP. In human cell lines, the mevalonate pathway regulates YAP to mediate self-renewal and survival through geranylgeranylation of RHO proteins. These studies suggest that in preimplantation development, statins may act through HIPPO pathway to interfere with trophectoderm specification and thereby inhibit blastocyst formation. STUDY DESIGN, SAMPLES/MATERIALS, METHODS: Eight-cell stage (E2.5) mouse embryos were treated in hanging drop culture with chemical agents, namely statins (lovastatin, atorvastatin, cerivastatin and pravastatin), mevalonic acid (MVA), cholesterol, squalene, farnesyl pyrophosphate (FPP), geranylgeranyl pyrophosphate (GGPP), geranylgeranyltransferase inhibitor GGTI-298, RHO inhibitor I, and squalene synthase inhibitor YM-53601, up to the late blastocyst stage (E4.5). Efficiency of blastocyst formation was assessed based on gross morphology and the measurement of the cavity size using an image analysis software. Effects on cell lineages and HIPPO signaling were analyzed using immunohistochemistry with confocal microscopy based on the expression patterns of the lineage-specific markers and the nuclear accumulation of YAP. Effects on cell lineages were also examined by quantitative RT-PCR based on the transcript levels of the lineage-specific marker genes. Data were analyzed using one-way ANOVA and two-sample t-test. MAIN RESULTS AND THE ROLE OF CHANCE: All four statins examined inhibited blastocyst formation. The adverse impact of statins was rescued by supplementation of MVA (P < 0.01) or GGPP (P < 0.01) but not squalene nor cholesterol. Blastocyst formation was also prevented by GGTI-298 (P < 0.01). These results indicate that HMG-CoA reductase activity is required for blastocyst formation mainly through the production of GGPP but not cholesterol. Inhibition of RHO proteins, known targets of geranylgeranylation, impaired blastocyst formation, which was not reversed by GGPP supplementation. Nuclear localization of YAP was diminished by statin treatment but fully restored by supplementation of MVA (P < 0.01) or GGPP (P < 0.01). This suggests that HIPPO signaling is regulated by GGPP-dependent mechanisms, possibly geranylgeranylation of RHO, to enable trophectoderm formation. YM-53601 prevented blastocyst formation (P < 0.01), but its adverse impact was not rescued by supplementation of squalene or cholesterol, suggesting that squalene synthesis inhibition was not the cause of blastocyst defects. LIMITATIONS, REASONS FOR CAUTION: Analyses were conducted on embryos cultured ex vivo, but they enable the determination of specific concentrations that impair embryo development which can be compared with drug concentrations in the reproductive tract when testing in vivo impact of statins through animal experimentations. Also, analyses were conducted in only one species, the mouse. Epidemiological studies on the effects of various types of statins on the fertility of women are necessary. WIDER IMPLICATIONS OF THE FINDINGS: Our study reveals how the mevalonate pathway is required for blastocyst formation and intersects with HIPPO pathway to provide a mechanistic basis for the embryotoxic effect of statins. This bears relevance for women who are taking statins while trying to conceive, since statins have potential to prevent the conceptus from reaching the blastocyst stage and to cause early conceptus demise. LARGE SCALE DATA: Not applicable. STUDY FUNDING AND COMPETING INTERESTS: This study was supported by grants from the George F. Straub Trust of the Hawaii Community Foundation (13ADVC-60315 to V.B.A.) and the National Institutes of Health, USA (P20GM103457 to V.B.A.). The authors have no conflict of interest to declare.


Assuntos
Blastocisto/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Prenilação/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Benzamidas/farmacologia , Blastocisto/efeitos dos fármacos , Proteínas de Ciclo Celular , Feminino , Lovastatina/análogos & derivados , Lovastatina/farmacologia , Masculino , Ácido Mevalônico/farmacologia , Camundongos , Fosfoproteínas/metabolismo , Fosfatos de Poli-Isoprenil/farmacologia , Pravastatina/farmacologia , Quinuclidinas/farmacologia , Sesquiterpenos/farmacologia , Proteínas de Sinalização YAP
6.
Dev Biol ; 394(1): 142-55, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24997360

RESUMO

Specification of the trophectoderm (TE) and inner cell mass (ICM) lineages in the mouse blastocyst correlates with cell position, as TE derives from outer cells whereas ICM from inner cells. Differences in position are reflected by cell polarization and Hippo signaling. Only in outer cells, the apical-basal cell polarity is established, and Hippo signaling is inhibited in such a manner that LATS1 and 2 (LATS1/2) kinases are prevented from phosphorylating YAP, a key transcriptional co-activator of the TE-specifying gene Cdx2. However, the molecular mechanisms that regulate these events are not fully understood. Here, we showed that inhibition of RHO-ROCK signaling enhances ICM and suppresses TE characteristics through activation of Hippo signaling and disruption of apical-basal polarity. Embryos treated with ROCK inhibitor Y-27632 exhibited elevated expression of ICM marker NANOG and reduced expression of CDX2 at the blastocyst stage. Y-27632-treated embryos failed to accumulate YAP in the nucleus, although it was rescued by concomitant inhibition of LATS1/2. Segregation between apical and basal polarity regulators, namely PARD6B, PRKCZ, SCRIB, and LLGL1, was dampened by Y-27632 treatment, whereas some of the polarization events at the late 8-cell stage such as compaction and apical localization of p-ERM and tyrosinated tubulin occurred normally. Similar abnormalities of Hippo signaling and apical-basal polarization were also observed in embryos that were treated with RHO GTPases inhibitor. These results suggest that RHO-ROCK signaling plays an essential role in regulating Hippo signaling and cell polarization to enable proper specification of the ICM and TE lineages.


Assuntos
Massa Celular Interna do Blastocisto/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas rho de Ligação ao GTP/antagonistas & inibidores , Quinases Associadas a rho/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Amidas/farmacologia , Animais , Fator de Transcrição CDX2 , Proteínas de Ciclo Celular , Diferenciação Celular , Linhagem da Célula , Polaridade Celular/fisiologia , Proteínas do Citoesqueleto , Ectoderma/metabolismo , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento , Via de Sinalização Hippo , Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas de Homeodomínio/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Proteína Homeobox Nanog , Fosfoproteínas/biossíntese , Proteína Quinase C/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Piridinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais , Fatores de Transcrição/biossíntese , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/genética , Proteínas de Sinalização YAP , Proteínas rho de Ligação ao GTP/metabolismo , Quinases Associadas a rho/metabolismo
7.
Biol Reprod ; 89(5): 122, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23946538

RESUMO

The blastocyst consists of the outer layer of trophectoderm and pluripotent inner cell mass (ICM), the precursor of the placenta and fetus, respectively. During blastocyst expansion, the ICM adopts a compact, ovoidal shape, whose proper morphology is crucial for normal embryogenesis. Rho-associated kinase (ROCK), an effector of small GTPase RHO signaling, mediates the diverse cellular processes of morphogenesis, but its role in ICM morphogenesis is unclear. Here, we demonstrate that ROCK is required for cohesion of ICM cells and formation of segregated tissues called primitive endoderm (PrE) and epiblast (Epi) in the ICM of the mouse blastocyst. Blastocyst treatment with ROCK inhibitors Y-27632 and Fasudil caused widening or spreading of the ICM, and intermingling of PrE and Epi. Widening of ICM was independent of trophectoderm because isolated ICMs as well as colonies of mouse embryonic stem cells (mESC) also spread upon Y-27632 treatment. PrE, Epi, and trophectoderm cell numbers were similar between control and treated blastocysts, suggesting that ROCK inhibition affected ICM morphology but not lineage differentiation. Rock1 and Rock2 knockdown via RNA interference in mESC also induced spreading, supporting the conclusion that morphological defects caused by the pharmacological inhibitors were due to ROCK inactivation. When blastocysts were transferred into surrogates, implantation efficiencies were unaffected by ROCK inhibition, but treated blastocysts yielded greater fetal loss. These results show that proper ICM morphology is dependent on ROCK activity and is crucial for fetal development. Our studies have wider implication for improving efficiencies of human assisted reproductive technologies that diminish pregnancy loss and promote successful births.


Assuntos
Massa Celular Interna do Blastocisto/fisiologia , Morfogênese/genética , Quinases Associadas a rho/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Embrião de Mamíferos , Feminino , Técnicas de Silenciamento de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Gravidez , Quinases Associadas a rho/genética
8.
Differentiation ; 83(5): 282-92, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22475651

RESUMO

Activation of Wnt/ß-catenin signaling is crucial for the differentiation of pluripotent stem cells, namely the epiblast, embryonic stem, and embryonal carcinoma cells, into mesendoderm. However, downstream events of Wnt/ß-catenin signaling that control the formation of mesendoderm are still unclear. In the present study, we used mouse P19 embryonal carcinoma cells as a model, and identified a homeodomain protein Nkx1-2 as a key regulator of mesendoderm formation. In the mouse embryo, Nkx1-2 was expressed in the primitive streak, in which the nascent mesendoderm emerges. In P19 cells, the expression of Nkx1-2 was activated by Wnt/ß-catenin signaling independently of Brachyury, an evolutionary conserved early mesendoderm gene. In contrast, the expression of Nkx1-2 was both necessary and sufficient for the activation of Brachyury. Nkx1-2 acted as a transcriptional repressor to mediate the action of Wnt/ß-catenin signaling to activate the Brachyury expression. We found Tcf3 as a potential target of gene repression by Nkx1-2, and the down-regulation of Tcf3 was partly required for effective activation of Brachyury by Wnt/ß-catenin signaling. These results suggest that Nkx1-2 is a critical component of the gene regulatory network that operates downstream of Wnt/ß-catenin signaling to regulate the formation of mesendoderm.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/genética , Proteínas Fetais/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem da Célula , Células-Tronco de Carcinoma Embrionário/citologia , Células-Tronco de Carcinoma Embrionário/metabolismo , Proteínas Fetais/genética , Redes Reguladoras de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Proteínas com Domínio T/genética , Via de Sinalização Wnt
9.
Reprod Toxicol ; 121: 108475, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37748715

RESUMO

Molnupiravir is a nucleoside analog antiviral that is authorized for use in the treatment of COVID-19. For its therapeutic action, molnupiravir is converted after ingestion to the active metabolite N4-hydroxycytidine, which is incorporated into the viral genome to cause lethal mutagenesis. Molnupiravir is not recommended for use during pregnancy, because preclinical animal studies suggest that it is hazardous to developing embryos. However, the mechanisms underlying the embryotoxicity of molnupiravir are currently unknown. To gain mechanistic insights into its embryotoxic action, the effects of molnupiravir and N4-hydroxycytidine were examined on the in vitro development of mouse preimplantation embryos. Molnupiravir did not prevent blastocyst formation even at concentrations that were much higher than the therapeutic plasma levels. By contrast, N4-hyroxycytidine exhibited potent toxicity, as it interfered with blastocyst formation and caused extensive cell death at concentrations below the therapeutic plasma levels. The adverse effects of N4-hydroxycytidine were dependent on the timing of exposure, such that treatment after the 8-cell stage, but not before it, caused embryotoxicity. Transcriptomic analysis of N4-hydroxycytidine-exposed embryos, together with the examination of eIF-2a protein phosphorylation level, suggested that N4-hydroxycytidine induced the integrated stress response. The adverse effects of N4-hydroxycytidine were significantly alleviated by the co-treatment with S-(4-nitrobenzyl)-6-thioinosine, suggesting that the embryotoxic potential of N4-hydroxycytidine requires the activity of nucleoside transporters. These findings show that the active metabolite of molnupiravir impairs preimplantation development at clinically relevant concentrations, providing mechanistic foundation for further studies on the embryotoxic potential of molnupiravir and other related nucleoside antivirals.


Assuntos
COVID-19 , Nucleosídeos , Gravidez , Feminino , Camundongos , Animais , Nucleosídeos/metabolismo , Nucleosídeos/farmacologia , Blastocisto , Hidroxilaminas/metabolismo , Hidroxilaminas/farmacologia , Antivirais/toxicidade
10.
Biol Reprod ; 87(3): 62, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22674395

RESUMO

In eukaryotes, DNA synthesis is preceded by licensing of replication origins. We examined the subcellular localization of two licensing proteins, ORC2 and MCM7, in the mouse zygotes and two-cell embryos. In somatic cells ORC2 remains bound to DNA replication origins throughout the cell cycle, while MCM7 is one of the last proteins to bind to the licensing complex. We found that MCM7 but not ORC2 was bound to DNA in metaphase II oocytes and remained associated with the DNA until S-phase. Shortly after fertilization, ORC2 was detectable at the metaphase II spindle poles and then between the separating chromosomes. Neither protein was present in the sperm cell at fertilization. As the sperm head decondensed, MCM7 was bound to DNA, but no ORC2 was seen. By 4 h after fertilization, both pronuclei contained DNA bound ORC2 and MCM7. As expected, during S-phase of the first zygotic cell cycle, MCM7 was released from the DNA, but ORC2 remained bound. During zygotic mitosis, ORC2 again localized first to the spindle poles, then to the area between the separating chromosomes. ORC2 then formed a ring around the developing two-cell nuclei before entering the nucleus. Only soluble MCM7 was present in the G2 pronuclei, but by zygotic metaphase it was bound to DNA, again apparently before ORC2. In G1 of the two-cell stage, both nuclei had salt-resistant ORC2 and MCM7. These data suggest that licensing follows a unique pattern in the early zygote that differs from what has been described for other mammalian cells that have been studied.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Replicação do DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Complexo de Reconhecimento de Origem/metabolismo , Zigoto/metabolismo , Animais , Cromatina/metabolismo , Período de Replicação do DNA/fisiologia , Embrião de Mamíferos , Feminino , Fertilização/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Componente 7 do Complexo de Manutenção de Minicromossomo , Modelos Biológicos , Distribuição Tecidual , Zigoto/ultraestrutura
11.
Reprod Toxicol ; 111: 135-147, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35605700

RESUMO

Remdesivir (RDV) is the first antiviral drug to be approved by the US Food and Drug Administration for the treatment of COVID-19. While the general safety of RDV has been studied, its reproductive risk, including embryotoxicity, is largely unknown. Here, to gain insights into its embryotoxic potential, we investigated the effects of RDV on mouse preimplantation embryos cultured in vitro at the concentrations comparable to the therapeutic plasma levels. Exposure to RDV (2-8 µM) did not affect the initiation of blastocyst formation, although the maintenance of the cavity failed at 8 µM due to increased cell death. While exposure to 2-4 µM permitted the cavity maintenance, expressions of developmental regulator genes associated with the inner cell mass (ICM) lineage were significantly diminished. Adverse effects of RDV depended on the duration and timing of exposure, as treatment between the 8-cell to early blastocyst stage most sensitively affected cavity expansion, gene expressions, and cell proliferation, particularly of the ICM than the trophectoderm lineage. GS-441524, a major metabolite of RDV, did not impair blastocyst formation or cavity expansion, although it altered gene expressions in a manner differently from RDV. Additionally, RDV reduced the viability of human embryonic stem cells, which were used as a model for the human ICM lineage, more potently than GS-441524. These findings suggest that RDV is potentially embryotoxic to impair the pluripotent lineage, and will be useful for designing and interpreting further in vitro and in vivo studies on the reproductive toxicity of RDV.


Assuntos
Tratamento Farmacológico da COVID-19 , Complicações Infecciosas na Gravidez , Monofosfato de Adenosina/análogos & derivados , Alanina/análogos & derivados , Animais , Blastocisto , Desenvolvimento Embrionário/genética , Feminino , Camundongos , Gravidez , Complicações Infecciosas na Gravidez/metabolismo
12.
J Assist Reprod Genet ; 28(8): 659-68, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21617931

RESUMO

PURPOSE: To assess the impact of embryonic stem cell culture medium (ESCM) on the pre- and post-implantation development of the mouse embryo, as a mammalian model, in comparison with the conventional culture medium, a potassium simplex optimized medium (KSOM). METHODS: Development in ESCM versus KSOM was compared in terms of embryo morphology, cleavage, cavitation, hatching, cell number, expression of TE and ICM transcription factors (Cdx2 and Oct4, respectively), implantation, and development in utero. RESULTS: An enriched medium like ESCM can be beneficial for in vitro embryo development when cultured from the 8-cell stage, as evidenced by promotion of blastocyst development with respect to cavity expansion, hatching, and cell division. Such benefits were not observed when embryos were cultured from the 2-cell stage. CONCLUSIONS: ESCM may augment in vitro embryo development from the 8-cell stage. Using different culture media at different stages may be beneficial to achieve more effective human in vitro fertilization.


Assuntos
Blastocisto/citologia , Meios de Cultura , Técnicas de Cultura Embrionária/métodos , Células-Tronco Embrionárias/citologia , Animais , Blastocisto/metabolismo , Divisão Celular , Desenvolvimento Embrionário , Células-Tronco Embrionárias/metabolismo , Feminino , Fertilização in vitro , Camundongos , Técnicas de Reprodução Assistida
13.
Biol Reprod ; 83(3): 347-58, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20505164

RESUMO

In preimplantation mouse development, the first cell lineages to be established are the trophectoderm (TE) and inner cell mass. TE possesses epithelial features, including apical-basal cell polarity and intercellular junctions, which are crucial to generate a fluid-filled cavity in the blastocyst. Homologs of the partitioning defective (par) genes in Caenorhabditis elegans are critical regulators of cell polarity. However, their roles in regulating TE differentiation and blastocyst formation remain unclear. Here, the role of mouse Pard6b, a homolog of par-6 gene and a component of the PAR-atypical protein kinase C (aPKC) complex, was investigated. Pard6b expression was knocked down by microinjecting RNA interference construct into zygotes. Pard6b-knockdown embryos cleaved and compacted normally but failed to form the blastocyst cavity. The cavitation failure is likely the result of defective intercellular junctions, because Pard6b knockdown caused abnormal distribution of actin filaments and TJP1 (ZO-1) tight junction (TJ) protein and interfered with cavitation in chimeras containing cells from normal embryos. Defective TJ formation may be caused by abnormal cell polarization, because the apical localization of PRKCZ (aPKCzeta) was absent in Pard6b-knockdown embryos. Pard6b knockdown also diminished the expression of CDX2, a TE-lineage transcription factor, in the outer cells. TEAD4, a transcriptional activator that is required for Cdx2 expression and cavity formation, was not essential for the transcription of Pard6b. Taken together, Pard6b is necessary for blastocyst morphogenesis, particularly the development of TE-specific features-namely, the apical-basal cell polarity, formation of TJ, paracellular permeability sealing, and up-regulated expression of Cdx2.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Blastocisto/fisiologia , Polaridade Celular/fisiologia , Morfogênese/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Fator de Transcrição CDX2 , Linhagem Celular Tumoral , Linhagem da Célula , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Células Epiteliais/fisiologia , Imunofluorescência , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Microinjeções , Microscopia Confocal , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição de Domínio TEA , Junções Íntimas/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
14.
Reprod Toxicol ; 91: 74-91, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31711903

RESUMO

Pluripotent stem cells recapitulate many aspects of embryogenesis in vitro. Here, we established a novel culture system to differentiate human embryonic stem cell aggregates (HESCA), and evaluated its utility for teratogenicity assessment. Culture of HESCA with modulators of developmental signals induced morphogenetic and molecular changes associated with differentiation of the paraxial mesoderm and neuroectoderm. To examine impact of teratogenic exposures on HESCA differentiation, 18 compounds were tested, for which adequate information on in vivo plasma concentrations is available. HESCA treated with each compound were examined for gross morphology and transcript levels of 15 embryogenesis regulator genes. Significant alterations in the transcript levels were observed for 94% (15/16) of the teratogenic exposures within 5-fold margin, whereas no alteration was observed for 92% (11/12) of the non-teratogenic exposures. Our study demonstrates that transcriptional changes in HESCA serve as predictive indicator of teratogenicity in a manner comparable to in vivo exposure levels.


Assuntos
Técnicas de Cultura de Células , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Teratogênicos/toxicidade , Agregação Celular , Diferenciação Celular , Células Cultivadas , Desenvolvimento Embrionário/efeitos dos fármacos , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Teratogênese
15.
Genesis ; 47(2): 93-106, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19115346

RESUMO

Because of their capacity to give rise to various types of cells in vitro, embryonic stem and embryonal carcinoma (EC) cells have been used as convenient models to study the mechanisms of cell differentiation in mammalian embryos. In this study, we explored the mouse P19 EC cell line as an effective tool to investigate the factors that may play essential roles in mesoderm formation and axial elongation morphogenesis. We first demonstrated that aggregated P19 cells not only exhibited gene expression patterns characteristic of mesoderm formation but also displayed elongation morphogenesis with a distinct anterior-posterior body axis as in the embryo. We then showed by RNA interference that these processes were controlled by various regulators of Wnt signaling pathways, namely beta-catenin, Wnt3, Wnt3a, and Wnt5a, in a manner similar to normal embryo development. We further showed by inhibitor treatments that the axial elongation morphogenesis was dependent on the activity of Rho-associated kinase. Because of the convenience of these experimental manipulations, we propose that P19 cells can be used as a simple and efficient screening tool to assess the potential functions of specific molecules in mesoderm formation and axial elongation morphogenesis.


Assuntos
Carcinoma Embrionário/patologia , Mesoderma/embriologia , Modelos Biológicos , Animais , Sequência de Bases , Padronização Corporal , Carcinoma Embrionário/genética , Carcinoma Embrionário/metabolismo , Agregação Celular , Diferenciação Celular , Linhagem Celular Tumoral , Primers do DNA/genética , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Morfogênese/genética , Morfogênese/fisiologia , Gravidez , Interferência de RNA , Transdução de Sinais , Proteínas Wnt/antagonistas & inibidores , Proteínas Wnt/genética , Proteína Wnt-5a , Proteína Wnt3 , Proteína Wnt3A , beta Catenina/antagonistas & inibidores , beta Catenina/genética , Quinases Associadas a rho/metabolismo
16.
Biochim Biophys Acta ; 1780(12): 1395-402, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18675318

RESUMO

In mammals, Sry is the master regulator of male sex determination, although how it functions is still unclear. By contrast, female sex determination depends on the action of Rspo1 and Wnt4, the regulators of Wnt/beta-catenin signaling. To seek a possible interaction between male and female sex determination mechanisms, we examined whether Sry affects Wnt/beta-catenin signaling. Using the TOPFLASH reporter system to measure Lef/Tcf-dependent transcriptional activity, we showed that ectopic expression of mouse Sry strongly suppressed Wnt/beta-catenin signaling in mouse embryonal carcinoma and human embryonic kidney cell lines. This inhibition occurred downstream of beta-catenin but upstream of Lef/Tcf, and depended on both the HMG-box and the C-terminal transcriptional activation domain. By contrast, TOPFLASH was not inhibited by human SRY, which apparently lacks a transcriptional activation domain. However, a fusion construct consisting of human SRY attached to the C-terminal domain of mouse Sry was able to inhibit TOPFLASH effectively. Furthermore, Sry constructs carrying point mutations equivalent to those in human sex reversal mutations were less effective in inhibiting Wnt/beta-catenin signaling. Also, we showed that the action of Sry as a transcriptional activator was both necessary and sufficient to inhibit Wnt/beta-catenin signaling, suggesting that the transcriptional targets of Sry are responsible for the inhibition of signaling. Sox9 is a potential transcriptional target of Sry, although quantitative RT-PCR analysis indicates that the expression of Sox9 was not up-regulated by the ectopic expression of mouse Sry in mouse embryonal carcinoma cells. While the present study demonstrates an impact of mouse Sry on Wnt/beta-catenin signaling at an in vitro level, it requires further investigations to assess whether such action also takes place in vivo to regulate male sex determination.


Assuntos
Fatores de Transcrição SOXB1/biossíntese , Proteínas Wnt/fisiologia , beta Catenina/fisiologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Linhagem Celular , Linhagem Celular Tumoral , Glutamina/genética , Histidina/genética , Camundongos , Dados de Sequência Molecular , Fatores de Transcrição SOXB1/genética , Processos de Determinação Sexual , Transdução de Sinais
17.
Mol Reprod Dev ; 76(11): 1019-32, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19479991

RESUMO

The first cell lineage specification in mouse embryo development is the formation of trophectoderm (TE) and inner cell mass (ICM) of the blastocyst. This article is to review and discuss the current knowledge on the cellular and molecular mechanisms of this particular event. Several transcription factors have been identified as the critical regulators of the formation or maintenance of the two cell lineages. The establishment of TE manifests as the formation of epithelium, and is dependent on many structural and regulatory components that are commonly found and that function in many epithelial tissues. Distinct epithelial features start to emerge at the late 8-cell stage, but the fates of blastomeres are not fixed as TE or ICM until around 32-cell stage. The location of blastomeres at this stage, that is, external or internal of the embryo, in effect defines the commitment towards the TE or ICM lineage, respectively. Some studies implicate the presence of a developmental bias among blastomeres at 2- or 4-cell stage, although it is unlikely to play a decisive role in the establishment of TE and ICM. The unique mode of cell lineage specification in the mouse embryo is further discussed in comparison with the formation of initial cell lineages, namely the three germ layers, in non-mammalian embryos.


Assuntos
Massa Celular Interna do Blastocisto/citologia , Ectoderma/citologia , Embrião de Mamíferos/citologia , Animais , Massa Celular Interna do Blastocisto/fisiologia , Blastômeros/citologia , Linhagem da Célula , Embrião de Mamíferos/fisiologia , Células Epiteliais/citologia , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Microscopia Confocal , Fatores de Transcrição , Trofoblastos/citologia
18.
Mol Reprod Dev ; 75(7): 1143-53, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18196554

RESUMO

The embryonic-abembryonic (Em-Ab) axis of the mouse blastocyst has been found in several studies to align orthogonal to the first cleavage plane, raising the possibility that a developmental prepattern already exists at the two-cell stage. However, it is also possible that such alignment is not due to any developmental disparity between the two-cell stage blastomeres, but rather is caused by an extrinsic mechanical constraint that is conferred by an irregular shape of the zona pellucida (ZP). Here, we conducted a series of experiments to distinguish between these possibilities. We showed that the shape of the ZP at the two-cell stage varied among embryos, ranging from near spherical to ellipsoidal, and that the ZP shape did not change until the blastocyst stage. In those embryos with an ellipsoidal ZP, the Em-Ab axis tended to lie orthogonal to the first cleavage plane, while in those embryos with a near spherical ZP, there was no such relationship. The clonal boundary between the descendants of the two-cell stage blastomeres tended to lie orthogonal to the Em-Ab axis when the rotation of the embryo within the ZP was experimentally prevented, while the control embryos did not exhibit such tendency. These results support the possibility that an apparent correlation between the first cleavage plane and the blastocyst axis can be generated by the mechanical constraint from the ZP but not by a developmental prepattern. Moreover, recent reports indicate that the vegetal blastomere of the four-cell stage embryo that had undergone a specific type of second cleavages is destined to contribute to the abembryonic side of the blastocyst. However, our present study shows that in spite of such specific second cleavages, the vegetal blastomere did not preferentially give rise to the abembryonic side. This result implicates that the lineage of the four-cell stage blastomere is not restricted even when embryos undergo a specific type of second cleavages.


Assuntos
Blastocisto/citologia , Blastômeros/citologia , Animais , Divisão Celular , Cruzamentos Genéticos , Desenvolvimento Embrionário , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Microscopia de Vídeo , Gravidez , Estresse Mecânico
19.
Int J Dev Biol ; 50(7): 581-6; discussion 586-7, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16892171

RESUMO

Mechanism underlying mammalian preimplantation development has long been a subject of controversy and the central question has been if any "determinants" play a key role in a manner comparable to the non-mammalian "model" system. During the last decade, this issue has been revived (Pearson, 2002; Rossant and Tam, 2004) by claims that the axes of the mouse blastocyst are anticipated at the egg ("prepatterning model"; Gardner, 1997; Gardner, 2001; Piotrowska et al., 2001; Piotrowska and Zernicka-Goetz, 2001; Zernicka-Goetz, 2005), suggesting that a mechanism comparable to that operating in non-mammals may be at work. However, recent studies by other laboratories do not support these claims ("regulative model"; Alarcon and Marikawa, 2003; Chroscicka et al., 2004; Hiiragi and Solter, 2004; Alarcon and Marikawa, 2005; Louvet-Vallee et al., 2005; Motosugi et al., 2005) and the issue is currently under hot debate (Vogel, 2005). Deepening our knowledge of this issue will not only provide an essential basis for understanding mammalian development, but also directly apply to ongoing clinical practices such as intracytoplasmic sperm injection (ICSI) and preimplantation genetic diagnosis (PGD). These practices were originally supported by a classical premise that mammalian preimplantation embryos are highly regulative (Tarkowski, 1959; Tarkowski, 1961; Tarkowski and Wroblewska, 1967; Rossant, 1976), in keeping with the "regulative model". However, if the "prepatterning model" is correct, the latter will require critical reassessment.


Assuntos
Blastocisto/citologia , Padronização Corporal , Desenvolvimento Embrionário , Zigoto/crescimento & desenvolvimento , Animais , Animais não Endogâmicos , Blastômeros/citologia , Linhagem da Célula , Polaridade Celular , Fase de Clivagem do Zigoto/citologia , Cruzamentos Genéticos , Feminino , Previsões , Camundongos , Camundongos Endogâmicos CBA , Camundongos Endogâmicos , Gravidez , Interações Espermatozoide-Óvulo , Zigoto/citologia
20.
Cloning Stem Cells ; 7(1): 8-16, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15996113

RESUMO

The transcription factor Oct4 is specifically expressed in the germ line and pluripotent stem cells, and is indispensable for normal mouse development. To understand the epigenetic control of Oct4 expression, we examined the DNA methylation pattern of the Oct4 regulatory element in various types of cells. Bisulfite analysis showed that the regulatory element was unmethylated in P19 embryonal carcinoma cells, which robustly express Oct4. By contrast, the regulatory element was distinctly methylated in somatic cells, including cell lines, such as NIH3T3 embryonic fibroblast and Hepa1-6 hepatoma, as well as tissues from the adult body, such as liver, spleen, and cumulus cells. However, we found that the extent of methylation was considerably heterogeneous among the alleles in the adult somatic cells. Using a luciferase reporter construct, we demonstrated that the extent of methylation directly affects the efficiency of gene expression driven by the Oct4 regulatory element in P19 cells. These results raise the possibility that the epigenetic status of Oct4 is heterogeneous among a population of somatic cells, which may affect the efficiency of Oct4 reactivation after somatic cell nuclear transfer.


Assuntos
Clonagem Molecular/métodos , Metilação de DNA , Proteínas de Ligação a DNA/genética , Genes Reguladores , Fatores de Transcrição/genética , Alelos , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Ilhas de CpG , Feminino , Fibroblastos/metabolismo , Genes Reporter , Humanos , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Modelos Genéticos , Células NIH 3T3 , Fator 3 de Transcrição de Octâmero , Plasmídeos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sulfitos/farmacologia , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA