Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Vet Med Sci ; 10(4): e1521, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38952271

RESUMEN

BACKGROUND: Cynomolgus monkeys (Macaca fascicularis) are essential in biomedical research, including reproductive studies. However, the application of human estimated foetal weight (EFW) formulas using ultrasonography (USG) in these non-human primates is not well established. OBJECTIVES: This study aims to evaluate the applicability of human EFW formulas for estimating foetal weight in cynomolgus monkeys at approximately 130 days of gestation. METHODS: Our study involved nine pregnant cynomolgus monkeys. We measured foetal parameters, including biparietal diameter, head circumference, abdominal circumference and femur length using USG. The EFW was calculated using 11 human EFW formulas. The actual birthweight (ABW) was recorded following Cesarean section, the day after the EFW calculation. For comparing EFW and ABW, we employed statistical methods such as mean absolute percentage error (APE) and Bland-Altman analysis. RESULTS: The ABW ranged between 200.36 and 291.33 g. Among the 11 formulas, the Combs formula showed the lowest APE (4.3%) and highest correlation with ABW (p < 0.001). Notably, EFW and ABW differences for the Combs formula were ≤5% in 66.7% and ≤10% in 100% of cases. The Bland-Altman analysis supported these results, showing that all cases fell within the limits of agreement. CONCLUSIONS: The Combs formula is applicable for estimating the weight of cynomolgus monkey fetuses with USG at approximately 130 days of gestation. Our observations suggest that the Combs formula can be applied in the prenatal care and biomedical research of this species.


Asunto(s)
Peso al Nacer , Peso Fetal , Macaca fascicularis , Ultrasonografía Prenatal , Animales , Macaca fascicularis/embriología , Macaca fascicularis/fisiología , Femenino , Peso Fetal/fisiología , Embarazo , Ultrasonografía Prenatal/veterinaria , Humanos
2.
Nature ; 612(7941): 732-738, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36517595

RESUMEN

Our understanding of human early development is severely hampered by limited access to embryonic tissues. Due to their close evolutionary relationship with humans, nonhuman primates are often used as surrogates to understand human development but currently suffer from a lack of in vivo datasets, especially from gastrulation to early organogenesis during which the major embryonic cell types are dynamically specified. To fill this gap, we collected six Carnegie stage 8-11 cynomolgus monkey (Macaca fascicularis) embryos and performed in-depth transcriptomic analyses of 56,636 single cells. Our analyses show transcriptomic features of major perigastrulation cell types, which help shed light on morphogenetic events including primitive streak development, somitogenesis, gut tube formation, neural tube patterning and neural crest differentiation in primates. In addition, comparative analyses with mouse embryos and human embryoids uncovered conserved and divergent features of perigastrulation development across species-for example, species-specific dependency on Hippo signalling during presomitic mesoderm differentiation-and provide an initial assessment of relevant stem cell models of human early organogenesis. This comprehensive single-cell transcriptome atlas not only fills the knowledge gap in the nonhuman primate research field but also serves as an invaluable resource for understanding human embryogenesis and developmental disorders.


Asunto(s)
Gastrulación , Macaca fascicularis , Organogénesis , Análisis de la Célula Individual , Animales , Humanos , Ratones , Gastrulación/genética , Macaca fascicularis/embriología , Macaca fascicularis/genética , Organogénesis/genética , Cuerpos Embrioides , Perfilación de la Expresión Génica , Línea Primitiva/citología , Línea Primitiva/embriología , Tubo Neural/citología , Tubo Neural/embriología , Cresta Neural/citología , Cresta Neural/embriología , Vía de Señalización Hippo , Mesodermo/citología , Mesodermo/embriología , Células Madre
3.
Science ; 374(6570): eabd8887, 2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34793202

RESUMEN

X chromosome dosage compensation ensures balanced gene dosage between the X chromosome and autosomes and between the sexes, involving divergent mechanisms among mammals. We elucidated a distinct mechanism for X chromosome inactivation (XCI) in cynomolgus monkeys, a model for human development. The trophectoderm and cytotrophoblast acquire XCI around implantation through an active intermediate bearing repressive modifications and compacted structure, whereas the amnion, epiblast, and hypoblast maintain such an intermediate protractedly, attaining XCI by a week after implantation. Males achieve X chromosome up-regulation (XCU) progressively, whereas females show XCU coincidentally with XCI, both establishing the X:autosome dosage compensation by 1 week after implantation. Conversely, primordial germ cells undergo X chromosome reactivation by reversing the XCI pathway early during their development. Our findings establish a foundation for clarifying the dosage compensation mechanisms in primates, including humans.


Asunto(s)
Blastocisto/fisiología , Compensación de Dosificación (Genética) , Macaca fascicularis/embriología , Macaca fascicularis/genética , Trofoblastos/fisiología , Inactivación del Cromosoma X , Cromosoma X/genética , Animales , Desarrollo Embrionario , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Ligados a X , Células Germinativas/fisiología , Histonas/metabolismo , Metilación , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Regulación hacia Arriba , Cromosoma X/metabolismo , Cromosoma X/ultraestructura
4.
Nat Commun ; 12(1): 5126, 2021 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-34446705

RESUMEN

Embryonic development is largely conserved among mammals. However, certain genes show divergent functions. By generating a transcriptional atlas containing >30,000 cells from post-implantation non-human primate embryos, we uncover that ISL1, a gene with a well-established role in cardiogenesis, controls a gene regulatory network in primate amnion. CRISPR/Cas9-targeting of ISL1 results in non-human primate embryos which do not yield viable offspring, demonstrating that ISL1 is critically required in primate embryogenesis. On a cellular level, mutant ISL1 embryos display a failure in mesoderm formation due to reduced BMP4 signaling from the amnion. Via loss of function and rescue studies in human embryonic stem cells we confirm a similar role of ISL1 in human in vitro derived amnion. This study highlights the importance of the amnion as a signaling center during primate mesoderm formation and demonstrates the potential of in vitro primate model systems to dissect the genetics of early human embryonic development.


Asunto(s)
Amnios/metabolismo , Macaca fascicularis/embriología , Mesodermo/embriología , Amnios/embriología , Animales , Proteína Morfogenética Ósea 4/metabolismo , Desarrollo Embrionario , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Macaca fascicularis/genética , Macaca fascicularis/metabolismo , Mesodermo/metabolismo , Embarazo , Transducción de Señal
5.
Zool Res ; 42(4): 469-477, 2021 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-34213093

RESUMEN

Mutations of PTEN-induced kinase I (PINK1) cause early-onset Parkinson's disease (PD) with selective neurodegeneration in humans. However, current PINK1 knockout mouse and pig models are unable to recapitulate the typical neurodegenerative phenotypes observed in PD patients. This suggests that generating PINK1 disease models in non-human primates (NHPs) that are close to humans is essential to investigate the unique function of PINK1 in primate brains. Paired single guide RNA (sgRNA)/Cas9-D10A nickases and truncated sgRNA/Cas9, both of which can reduce off-target effects without compromising on-target editing, are two optimized strategies in the CRISPR/Cas9 system for establishing disease animal models. Here, we combined the two strategies and injected Cas9-D10A mRNA and two truncated sgRNAs into one-cell-stage cynomolgus zygotes to target the PINK1 gene. We achieved precise and efficient gene editing of the target site in three newborn cynomolgus monkeys. The frame shift mutations of PINK1 in mutant fibroblasts led to a reduction in mRNA. However, western blotting and immunofluorescence staining confirmed the PINK1 protein levels were comparable to that in wild-type fibroblasts. We further reprogramed mutant fibroblasts into induced pluripotent stem cells (iPSCs), which showed similar ability to differentiate into dopamine (DA) neurons. Taken together, our results showed that co-injection of Cas9-D10A nickase mRNA and sgRNA into one-cell-stage cynomolgus embryos enabled the generation of human disease models in NHPs and target editing by pair truncated sgRNA/Cas9-D10A in PINK1 gene exon 2 did not impact protein expression.


Asunto(s)
Modelos Animales de Enfermedad , Macaca fascicularis/genética , Enfermedad de Parkinson/veterinaria , Proteínas Quinasas/metabolismo , Animales , Animales Recién Nacidos , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/metabolismo , Técnicas de Cultivo de Embriones , Transferencia de Embrión , Fibroblastos/fisiología , Mutación del Sistema de Lectura , Regulación de la Expresión Génica , Macaca fascicularis/embriología , Enfermedades de los Monos/genética , Mutación , Enfermedad de Parkinson/genética , Proteínas Quinasas/genética , ARN Guía de Kinetoplastida
7.
Dev Dyn ; 250(7): 974-985, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33449399

RESUMEN

BACKGROUND: Genetic programs underlying preimplantation development and early lineage segregation are highly conserved across mammals. It has been suggested that nonhuman primates would be better model organisms for human embryogenesis, but a limited number of studies have investigated the monkey preimplantation development. In this study, we collect single cells from cynomolgus monkey preimplantation embryos for transcriptome profiling and compare with single-cell RNA-seq data derived from human and mouse embryos. RESULTS: By weighted gene-coexpression network analysis, we found that cynomolgus gene networks have greater conservation with human embryos including a greater number of conserved hub genes than that of mouse embryos. Consistently, we found that early ICM/TE lineage-segregating genes in monkeys exhibit greater similarity with human when compared to mouse, so are the genes in signaling pathways such as LRP1 and TCF7 involving in WNT pathway. Last, we tested the role of one conserved pre-EGA hub gene, SIN3A, using a morpholino knockdown of maternal RNA transcripts in monkey embryos followed by single-cell RNA-seq. We found that SIN3A knockdown disrupts the gene-silencing program during the embryonic genome activation transition and results in developmental delay of cynomolgus embryos. CONCLUSION: Taken together, our study provided new insight into evolutionarily conserved and divergent transcriptome dynamics during mammalian preimplantation development.


Asunto(s)
Blastómeros/metabolismo , Desarrollo Embrionario/genética , Macaca fascicularis/embriología , Adulto , Animales , Blastocisto , Blastómeros/citología , Linaje de la Célula/genética , Células Cultivadas , Embrión de Mamíferos , Desarrollo Embrionario/fisiología , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes/fisiología , Humanos , Macaca fascicularis/genética , Macaca mulatta , Masculino , Ratones , Embarazo , Complejo Correpresor Histona Desacetilasa y Sin3/genética , Complejo Correpresor Histona Desacetilasa y Sin3/fisiología , Análisis de la Célula Individual/veterinaria , Transcriptoma/genética
8.
Nat Commun ; 11(1): 2325, 2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32393762

RESUMEN

Common polygenic diseases result from compounded risk contributed by multiple genetic variants, meaning that simultaneous correction or introduction of single nucleotide variants is required for disease modeling and gene therapy. Here, we show precise, efficient, and simultaneous multiplex base editing of up to three target sites across 11 genes/loci in cynomolgus monkey embryos using CRISPR-based cytidine- and adenine-base editors. Unbiased whole genome sequencing demonstrates high specificity of base editing in monkey embryos. Our data demonstrate feasibility of multiplex base editing for polygenic disease modeling in primate zygotes.


Asunto(s)
Edición Génica/métodos , Animales , Secuencia de Bases , Embrión de Mamíferos/metabolismo , Exones/genética , Feto/metabolismo , Hígado/metabolismo , Macaca fascicularis/embriología , Mutación/genética
9.
Protein Cell ; 11(2): 97-107, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31781970

RESUMEN

Blastocyst complementation by pluripotent stem cell (PSC) injection is believed to be the most promising method to generate xenogeneic organs. However, ethical issues prevent the study of human chimeras in the late embryonic stage of development. Primate embryonic stem cells (ESCs), which have similar pluripotency to human ESCs, are a good model for studying interspecies chimerism and organ generation. However, whether primate ESCs can be used in xenogenous grafts remains unclear. In this study, we evaluated the chimeric ability of cynomolgus monkey (Macaca fascicularis) ESCs (cmESCs) in pigs, which are excellent hosts because of their many similarities to humans. We report an optimized culture medium that enhanced the anti-apoptotic ability of cmESCs and improved the development of chimeric embryos, in which domesticated cmESCs (D-ESCs) injected into pig blastocysts differentiated into cells of all three germ layers. In addition, we obtained two neonatal interspecies chimeras, in which we observed tissue-specific D-ESC differentiation. Taken together, the results demonstrate the capability of D-ESCs to integrate and differentiate into functional cells in a porcine model, with a chimeric ratio of 0.001-0.0001 in different neonate tissues. We believe this work will facilitate future developments in xenogeneic organogenesis, bringing us one step closer to producing tissue-specific functional cells and organs in a large animal model through interspecies blastocyst complementation.


Asunto(s)
Quimera , Células Madre Embrionarias/citología , Macaca fascicularis/embriología , Porcinos/embriología , Animales , Blastocisto/citología , Diferenciación Celular , Células Cultivadas , Quimera/embriología
10.
BMC Biotechnol ; 19(1): 7, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30646876

RESUMEN

BACKGROUND: Non-human primate (NHP) models can closely mimic human physiological functions and are therefore highly valuable in biomedical research. Genome editing is now developing rapidly due to the precision and efficiency offered by engineered site-specific endonuclease-based systems, such as transcription activator-like effector nucleases (TALENs) and the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein-9 nuclease (Cas9) system. It has been demonstrated that these programmable nucleases can introduce genetic changes in embryos from many species including NHPs. In 2014, we reported the first genetic editing of macaques using TALENs and CRISPR/Cas9. Subsequently, we characterized the phenotype of a methyl CpG binding protein 2 (MECP2)-mutant cynomolgus monkey model of Rett syndrome generated using the TALEN approach. These efforts not only accelerated the advance of modeling genetic diseases in NHPs, but also encouraged us to develop specific gene knock-in monkeys. In this study, we assess the possibility of homologous recombination (HR)-mediated gene replacement using TALENs in monkeys, and generate preimplantation embryos carrying an EmGFP fluorescent reporter constructed in the OCT4 gene. RESULT: We assembled a pair of TALENs specific to the first exon of the OCT4 gene and constructed a donor vector consisting of the homology arms cloned from the monkey genome DNA, flanking an EmGFP cassette. Next, we co-injected the TALENs-coding plasmid and donor plasmid into the cytoplasm of 122 zygotes 6-8 h after fertilization. Sequencing and immunofluorescence revealed that the OCT4-EmGFP knock-in allele had been successfully generated by TALENs-mediated HR at an efficiency of 11.3% (7 out of 62) or 11.1% (1 out of 9), respectively, in monkey embryos. CONCLUSION: We have successfully, for the first time, obtained OCT4-EmGFP knock-in monkey embryos via HR mediated by TALENs. Our results suggest that gene targeting through TALEN-assisted HR is a useful approach to introduce precise genetic modification in NHPs.


Asunto(s)
Edición Génica/métodos , Técnicas de Sustitución del Gen/métodos , Recombinación Homóloga , Macaca fascicularis/embriología , Macaca fascicularis/genética , Nucleasas de los Efectores Tipo Activadores de la Transcripción/genética , Animales , Animales Modificados Genéticamente , Femenino , Genotipo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo
11.
Nature ; 546(7658): 416-420, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28607482

RESUMEN

Human primordial germ cells (hPGCs), the precursors of sperm and eggs, originate during weeks 2-3 of early post-implantation development. Using in vitro models of hPGC induction, recent studies have suggested that there are marked mechanistic differences in the specification of human and mouse PGCs. This may be due in part to the divergence in their pluripotency networks and early post-implantation development. As early human embryos are not accessible for direct study, we considered alternatives including porcine embryos that, as in humans, develop as bilaminar embryonic discs. Here we show that porcine PGCs originate from the posterior pre-primitive-streak competent epiblast by sequential upregulation of SOX17 and BLIMP1 in response to WNT and BMP signalling. We use this model together with human and monkey in vitro models simulating peri-gastrulation development to show the conserved principles of epiblast development for competency for primordial germ cell fate. This process is followed by initiation of the epigenetic program and regulated by a balanced SOX17-BLIMP1 gene dosage. Our combinatorial approach using human, porcine and monkey in vivo and in vitro models provides synthetic insights into early human development.


Asunto(s)
Diferenciación Celular , Desarrollo Embrionario , Células Germinativas/citología , Macaca fascicularis/embriología , Modelos Biológicos , Células Madre Pluripotentes/citología , Porcinos/embriología , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Linaje de la Célula , Cuerpos Embrioides/citología , Epigénesis Genética , Femenino , Gastrulación , Dosificación de Gen , Células Germinativas/metabolismo , Estratos Germinativos/citología , Humanos , Técnicas In Vitro , Masculino , Modelos Animales , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Línea Primitiva/citología , Proteínas Represoras/genética , Factores de Transcripción SOXF/genética , Vía de Señalización Wnt
12.
Sci Data ; 4: 170067, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28649393

RESUMEN

In mammals, the development of pluripotency and specification of primordial germ cells (PGCs) have been studied predominantly using mice as a model organism. However, divergences among mammalian species for such processes have begun to be recognized. Between humans and mice, pre-implantation development appears relatively similar, but the manner and morphology of post-implantation development are significantly different. Nevertheless, the embryogenesis just after implantation in primates, including the specification of PGCs, has been unexplored due to the difficulties in analyzing the embryos at relevant developmental stages. Here, we present a comprehensive single-cell transcriptome dataset of pre- and early post-implantation embryo cells, PGCs and embryonic stem cells (ESCs) of cynomolgus monkeys as a model of higher primates. The identities of each transcriptome were also validated rigorously by other way such as immunofluorescent analysis. The information reported here will serve as a foundation for our understanding of a wide range of processes in the developmental biology of primates, including humans.


Asunto(s)
Células Madre Embrionarias , Macaca fascicularis , Transcriptoma , Animales , Regulación del Desarrollo de la Expresión Génica , Macaca fascicularis/embriología , Macaca fascicularis/genética , Análisis de la Célula Individual
13.
Drug Metab Pharmacokinet ; 32(1): 112-115, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28153493

RESUMEN

Cynomolgus macaques are an important primate species for drug metabolism studies; however cynomolgus CYP2C76, an important drug-metabolizing enzyme, accounts for drug metabolism differences to humans, so that CYP2C76-null animals might show drug-metabolizing properties more similar to humans. In this study, attempts were made to produce CYP2C76-null animals by assisted reproduction technology. Oocytes and sperm collected from the heterozygotes for the null allele (c.449TG > A) were subjected to intracytoplasmic sperm injection, and the embryos produced were cultured in vitro through the blastocyst stage. Preimplantation genetic diagnosis using a biopsied portion of the blastocyst revealed that none of the 32 blastocysts analyzed were homozygotes. In contrast, 2 of the 20 embryos analyzed were homozygotes at the 8-cell stage, indicating that CYP2C76-null embryos most likely stop developing between the 8-cell and blastocyst stage. By polymerase chain reaction, expression of CYP2C76 mRNA was detected in oocytes and blastocysts, but not in 2-, 4-, 8-, or 16/32-cell stage embryos. Metabolic assays showed that CYP2C76 metabolized progesterone. These results indicated that CYP2C76 null was likely embryonic lethal, suggesting its potential role during early embryogenesis in cynomolgus macaques.


Asunto(s)
Sistema Enzimático del Citocromo P-450/deficiencia , Sistema Enzimático del Citocromo P-450/metabolismo , Pérdida del Embrión/genética , Desarrollo Embrionario , Macaca fascicularis/embriología , Macaca fascicularis/genética , Animales , Sistema Enzimático del Citocromo P-450/genética , Femenino , Masculino , Oocitos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Espermatozoides/metabolismo
14.
Nature ; 537(7618): 57-62, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27556940

RESUMEN

The epiblast (EPI) is the origin of all somatic and germ cells in mammals, and of pluripotent stem cells in vitro. To explore the ontogeny of human and primate pluripotency, here we perform comprehensive single-cell RNA sequencing for pre- and post-implantation EPI development in cynomolgus monkeys (Macaca fascicularis). We show that after specification in the blastocysts, EPI from cynomolgus monkeys (cyEPI) undergoes major transcriptome changes on implantation. Thereafter, while generating gastrulating cells, cyEPI stably maintains its transcriptome over a week, retains a unique set of pluripotency genes and acquires properties for 'neuron differentiation'. Human and monkey pluripotent stem cells show the highest similarity to post-implantation late cyEPI, which, despite co-existing with gastrulating cells, bears characteristics of pre-gastrulating mouse EPI and epiblast-like cells in vitro. These findings not only reveal the divergence and coherence of EPI development, but also identify a developmental coordinate of the spectrum of pluripotency among key species, providing a basis for better regulation of human pluripotency in vitro.


Asunto(s)
Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Desarrollo Embrionario , Macaca fascicularis/embriología , Células Madre Pluripotentes/citología , Animales , Blastocisto/citología , Blastocisto/metabolismo , Diferenciación Celular/genética , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Femenino , Gastrulación/genética , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/citología , Estratos Germinativos/embriología , Estratos Germinativos/metabolismo , Humanos , Macaca fascicularis/genética , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/metabolismo , Células Madre Pluripotentes/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Especificidad de la Especie , Transcriptoma
15.
Anim Sci J ; 87(8): 1034-40, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26420680

RESUMEN

Since the available concentration of single-copy fetal genes in maternal blood DNA is sometimes lower than detection limits by PCR methods, the development of specific and quantitative PCR detection methods for fetal DNA in maternal blood is anticipated, which may broaden the methods that can be used to monitor pregnancy. We used the TaqMan qPCR amplification for DYS14 multi-copy sequence and the SRY gene in maternal blood plasma (cell-free DNA) and fractional precipitated blood cells (cellular DNA) from individual cynomolgus monkeys at 22 weeks of pregnancy. The availability of cell-free fetal DNA was higher in maternal blood plasma than that of cellular DNA from fractional precipitated blood cells. There was a significantly higher (P < 0.001) mean copy number of fetal male DYS14 from maternal plasma (4.4 × 10(4) copies/mL) than that of detected fetal cellular DNA from fractional blood cell pellets. The sensitivity of the DYS14 PCR assay was found to be higher than that of the SRY assay for the detection of fetal DNA when its presence was at a minimum. The DYS14 assay is an improved method for quantifying male fetal DNA in circulating maternal blood in the primate model.


Asunto(s)
Proteínas de Ciclo Celular/sangre , Proteínas de Ciclo Celular/genética , ADN/sangre , Dosificación de Gen/genética , Macaca fascicularis/embriología , Macaca fascicularis/genética , Reacción en Cadena de la Polimerasa/métodos , Animales , Femenino , Masculino , Embarazo
16.
J Comp Neurol ; 524(3): 535-63, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26053631

RESUMEN

Generation of the primate cortex is characterized by the diversity of cortical precursors and the complexity of their lineage relationships. Recent studies have reported miscellaneous precursor types based on observer classification of cell biology features including morphology, stemness, and proliferative behavior. Here we use an unsupervised machine learning method for Hidden Markov Trees (HMTs), which can be applied to large datasets to classify precursors on the basis of morphology, cell-cycle length, and behavior during mitosis. The unbiased lineage analysis automatically identifies cell types by applying a lineage-based clustering and model-learning algorithm to a macaque corticogenesis dataset. The algorithmic results validate previously reported observer classification of precursor types and show numerous advantages: It predicts a higher diversity of progenitors and numerous potential transitions between precursor types. The HMT model can be initialized to learn a user-defined number of distinct classes of precursors. This makes it possible to 1) reveal as yet undetected precursor types in view of exploring the significant features of precursors with respect to specific cellular processes; and 2) explore specific lineage features. For example, most precursors in the experimental dataset exhibit bidirectional transitions. Constraining the directionality in the HMT model leads to a reduction in precursor diversity following multiple divisions, thereby suggesting that one impact of bidirectionality in corticogenesis is to maintain precursor diversity. In this way we show that unsupervised lineage analysis provides a valuable methodology for investigating fundamental features of corticogenesis.


Asunto(s)
Encéfalo/citología , Encéfalo/embriología , Procesamiento de Imagen Asistido por Computador/métodos , Macaca fascicularis/embriología , Células Madre/citología , Aprendizaje Automático no Supervisado , Animales , Linaje de la Célula , Análisis por Conglomerados , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hidrozoos , Inmunohistoquímica , Cadenas de Markov , Microscopía Confocal , Reconocimiento de Normas Patrones Automatizadas/métodos , Nicho de Células Madre , Técnicas de Cultivo de Tejidos , Grabación en Video
17.
Cell Stem Cell ; 17(1): 116-24, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26119236

RESUMEN

Because of their similarity to humans, non-human primates are important models for studying human disease and developing therapeutic strategies. Establishment of chimeric animals using embryonic stem cells (ESCs) could help with these investigations, but has not so far been achieved. Here, we show that cynomolgus monkey ESCs (cESCs) grown in adjusted culture conditions are able to incorporate into host embryos and develop into chimeras with contribution in all three germ layers and in germ cell progenitors. Under the optimized culture conditions, which are based on an approach developed previously for naive human ESCs, the cESCs displayed altered growth properties, gene expression profiles, and self-renewal signaling pathways, suggestive of an altered naive-like cell state. Thus our findings show that it is feasible to generate chimeric monkeys using ESCs and open up new avenues for the use of non-human primate models to study both pluripotency and human disease.


Asunto(s)
Células Madre Embrionarias/citología , Macaca fascicularis/embriología , Quimera por Trasplante/embriología , Animales , Blastocisto/citología , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/trasplante , Femenino , Humanos , Masculino , Modelos Animales , Mórula/citología , Embarazo , Testículo/citología , Testículo/embriología , Transcriptoma
18.
J Matern Fetal Neonatal Med ; 28(15): 1822-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25260124

RESUMEN

OBJECTIVE: The monkey model is the best model to investigate some physiological response to the fetal transitory tracheal occlusion but it has never been described in Macaca monkeys. The aim of this study was to evaluate the feasibility of fetal endoscopic tracheal occlusion (FETO) in a non-human primate model. METHODS: Pregnant rhesus monkeys and cynomolgus were tested as a potential experimental model for FETO in the third trimester of pregnancy, by performing fetal tracheoscopies with and without tracheal occlusion. RESULTS: A total of 22 pregnancies were followed in 16 monkeys and underwent fetal surgery. Percutaneous endoscopic access to the uterine cavity was possible in 20 cases (91%). Of these 20 pregnant monkeys, fetal tracheoscopy could be achieved in 15 cases (75%). In rhesus monkeys, the time between the onset of endoscopy and tracheal penetration decreases as operator experience increases. Neither maternal morbidity nor mortality was related to surgery. Two fetal losses were possibly due to the procedure. CONCLUSION: FETO is feasible in the non-human primate, which closely reflects procedures in humans. The non-human primate model for FETO, specially the rhesus monkeys, may be useful for future studies concerning the mechanisms related to the lung growth after transitory fetal tracheal occlusion.


Asunto(s)
Obstrucción de las Vías Aéreas/cirugía , Modelos Animales de Enfermedad , Enfermedades Fetales/cirugía , Fetoscopía/métodos , Macaca fascicularis , Macaca mulatta , Tráquea/cirugía , Obstrucción de las Vías Aéreas/congénito , Obstrucción de las Vías Aéreas/embriología , Obstrucción de las Vías Aéreas/epidemiología , Animales , Endoscopía/métodos , Femenino , Enfermedades Fetales/patología , Macaca fascicularis/embriología , Macaca mulatta/embriología , Masculino , Embarazo , Resultado del Embarazo/epidemiología , Resultado del Embarazo/veterinaria , Tráquea/patología
19.
Cell Res ; 23(10): 1161-2, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23958583

RESUMEN

The fact that mammals are diploid sets a barrier to rapidly understand the function of non-coding and coding genes in the genome. Recently, Yang et al. reported successful derivation of monkey haploid embryonic stem cells from parthenotes, which provide an effective platform for studying mammalian gene function and enable reverse genetic screening of genes for recessive phenotypes in monkeys.


Asunto(s)
Células Madre Embrionarias/citología , Haploidia , Macaca fascicularis/embriología , Animales
20.
Cell Res ; 23(10): 1187-200, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23856644

RESUMEN

Recent success in the derivation of haploid embryonic stem cells (haESCs) from mouse via parthenogenesis and androgenesis has enabled genetic screening in mammalian cells and generation of gene-modified animals. However, whether haESCs can be derived from primates remains unknown. Here, we report the derivation of haESCs from parthenogenetic blastocysts of Macaca fascicularis monkeys. These cells, termed as PG-haESCs, are pluripotent and can differentiate to cells of three embryonic germ layers in vitro or in vivo. Interestingly, the haploidy of one monkey PG-haESC line (MPH1) is more stable compared with that of the other one (MPH2), as shown by the existence of haploid cells for more than 140 days without fluorescence-activated cell sorting (FACS) enrichment of haploid cells. Importantly, transgenic monkey PG-haESC lines can be generated by lentivirus- and piggyBac transposon-mediated gene transfer. Moreover, genetic screening is feasible in monkey PG-haESCs. Our results demonstrate that PG-haESCs can be generated from monkeys, providing an ideal tool for genetic analyses in primates.


Asunto(s)
Células Madre Embrionarias/citología , Haploidia , Macaca fascicularis/embriología , Animales , Blastocisto/citología , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/metabolismo , Macaca fascicularis/genética , Partenogénesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...