Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cell Rep Methods ; 3(9): 100590, 2023 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-37714158

RESUMEN

Non-human primates (NHPs) are the closest animal model to humans; thus, gene engineering technology in these species holds great promise for the elucidation of higher brain functions and human disease models. Knockin (KI) gene targeting is a versatile approach to modify gene(s) of interest; however, it generally suffers from the low efficiency of homology-directed repair (HDR) in mammalian cells, especially in non-expressed gene loci. In the current study, we generated a tyrosine hydroxylase (TH)-2A-Cre KI model of the common marmoset monkey (marmoset; Callithrix jacchus) using an HDR-biased CRISPR-Cas9 genome editing approach using Cas9-DN1S and RAD51. This model should enable labeling and modification of a specific neuronal lineage using the Cre-loxP system. Collectively, the current study paves the way for versatile gene engineering in NHPs, which may be a significant step toward further biomedical and preclinical applications.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Animales , Sistemas CRISPR-Cas/genética , Tirosina 3-Monooxigenasa/genética , Primates/genética , Mamíferos/genética
2.
Neurosci Res ; 185: 49-61, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36075457

RESUMEN

Alzheimer's disease (AD) is the leading cause of dementia which afflicts tens of millions of people worldwide. Despite many scientific progresses to dissect the AD's molecular basis from studies on various mouse models, it has been suffered from evolutionary species differences. Here, we report generation of a non-human primate (NHP), common marmoset model ubiquitously expressing Amyloid-beta precursor protein (APP) transgenes with the Swedish (KM670/671NL) and Indiana (V717F) mutations. The transgene integration of generated two transgenic marmosets (TG1&TG2) was thoroughly investigated by genomic PCR, whole-genome sequencing, and fluorescence in situ hybridization. By reprogramming, we confirmed the validity of transgene expression in induced neurons in vitro. Moreover, we discovered structural changes in specific brain regions of transgenic marmosets by magnetic resonance imaging analysis, including in the entorhinal cortex and hippocampus. In immunohistochemistry, we detected increased Aß plaque-like structures in TG1 brain at 7 years old, although evident neuronal loss or glial inflammation was not observed. Thus, this study summarizes our attempt to establish an NHP AD model. Although the transgenesis approach alone seemed not sufficient to fully recapitulate AD in NHPs, it may be beneficial for drug development and further disease modeling by combination with other genetically engineered models and disease-inducing approaches.


Asunto(s)
Enfermedad de Alzheimer , Precursor de Proteína beta-Amiloide , Animales , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Callithrix/genética , Modelos Animales de Enfermedad , Hibridación Fluorescente in Situ , Ratones Transgénicos , Transgenes
3.
Dev Growth Differ ; 64(6): 325-341, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35841539

RESUMEN

Potentials of immortal proliferation and unlimited differentiation into all the three germ layers and germ cells in induced pluripotent stem cells (iPSCs) render them important bioresources for in vitro reconstitution and modeling of intravital tissues and organs in various animal models, thus contributing to the elucidation of pathomechanisms, drug discovery and stem cell-based regenerative medicine. We previously reported promising approaches for deriving transgene-free iPSCs from somatic fibroblasts of multiple mammalian species by episomal vector or RNA transfection, although the respective step-by-step protocols and the combinatorial usage of these methods, which achieved high induction efficiency, have not been described in the literature so far. Here, we provide a detailed step-by-step description of these methods with critical tips and slight modifications (improvements) to previously reported methods. We also report a novel method for the establishment of iPSCs from the Syrian hamster (also known as golden hamster; Mesocricetus auratus), a unique animal model of hibernation. We anticipate this methodology will contribute to stem cell biology and regenerative medicine research.


Asunto(s)
Células Madre Pluripotentes Inducidas , Animales , Diferenciación Celular , Reprogramación Celular , Fibroblastos , Mamíferos , Transgenes
4.
Neural Dev ; 17(1): 6, 2022 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-35524278

RESUMEN

BACKGROUND: Fine-tuned cochlear development is essential for hearing. Owing to the difficulty in using early human fetal samples, most of our knowledge regarding cochlear development has been obtained from rodents. However, several inter-species differences in cochlear development between rodents and humans have been reported. To bridge these differences, we investigated early otic development of a non-human primate model animal, the common marmoset (Callithrix jacchus). METHODS: We examined 20 genes involved in early cochlear development and described the critical developmental steps for morphogenesis, which have been reported to vary between rodents and marmosets. RESULTS: The results revealed that several critical genes involved in prosensory epithelium specifications showed higher inter-species differences, suggesting that the molecular process for hair cell lineage acquisition in primates differs considerably from that of rodents. We also observed that the tempo of cochlear development was three times slower in the primate than in rodents. CONCLUSIONS: Our data provide new insights into early cochlear development in primates and humans and imply that the procedures used for manipulating rodent cochlear sensory cells cannot be directly used for the research of primate cells due to the intrinsic inter-species differences in the cell fate determination program.


Asunto(s)
Callithrix , Cóclea , Animales , Callithrix/genética , Diferenciación Celular , Humanos
5.
Stem Cell Reports ; 16(4): 754-770, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33798453

RESUMEN

Induced pluripotent stem cells (iPSCs) are capable of providing an unlimited source of cells from all three germ layers and germ cells. The derivation and usage of iPSCs from various animal models may facilitate stem cell-based therapy, gene-modified animal production, and evolutionary studies assessing interspecies differences. However, there is a lack of species-wide methods for deriving iPSCs, in particular by means of non-viral and non-transgene-integrating (NTI) approaches. Here, we demonstrate the iPSC derivation from somatic fibroblasts of multiple mammalian species from three different taxonomic orders, including the common marmoset (Callithrix jacchus) in Primates, the dog (Canis lupus familiaris) in Carnivora, and the pig (Sus scrofa) in Cetartiodactyla, by combinatorial usage of chemical compounds and NTI episomal vectors. Interestingly, the fibroblasts temporarily acquired a neural stem cell-like state during the reprogramming. Collectively, our method, robustly applicable to various species, holds a great potential for facilitating stem cell-based research using various animals in Mammalia.


Asunto(s)
Fibroblastos/citología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Mamíferos/metabolismo , Transgenes , Animales , Callithrix , Perros , Perfilación de la Expresión Génica , Vectores Genéticos/metabolismo , Estratos Germinativos/metabolismo , Células-Madre Neurales/metabolismo , Plásmidos/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , RNA-Seq , Especificidad de la Especie , Porcinos , Virus
6.
Cell Transplant ; 30: 963689721992066, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33588604

RESUMEN

Tumorigenicity of induced pluripotent stem cells (iPSCs) is anticipated when cells derived from iPSCs are transplanted. It has been reported that iPSCs formed a teratoma in vivo in autologous transplantation in a nonhuman primate model without immunosuppression. However, there has been no study on tumorigenicity in major histocompatibility complex (MHC)-matched allogeneic iPSC transplantation with immune-competent hosts. To examine the tumorigenicity of allogeneic iPSCs, we generated four iPSC clones carrying a homozygous haplotype of the MHC. Two clones were derived from female fibroblasts by using a retrovirus and the other two clones were derived from male peripheral blood mononuclear cells by using Sendai virus (episomal approach). The iPSC clones were transplanted into allogenic MHC-matched immune-competent cynomolgus macaques. After transplantation of the iPSCs into subcutaneous tissue of an MHC-matched female macaque and into four testes of two MHC-matched male macaques, histological analysis showed no tumor, inflammation, or regenerative change in the excised tissues 3 months after transplantation, despite the results that iPSCs formed teratomas in immune-deficient mice and in autologous transplantation as previously reported. The results in the present study suggest that there is no tumorigenicity of iPSCs in MHC-matched allogeneic transplantation in clinical application.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Pluripotentes Inducidas , Complejo Mayor de Histocompatibilidad , Trasplante Homólogo , Animales , Femenino , Masculino , Carcinogénesis , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Macaca fascicularis , Complejo Mayor de Histocompatibilidad/inmunología , Trasplante Homólogo/métodos , Ratones
7.
Sci Rep ; 10(1): 21516, 2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33299078

RESUMEN

GPR56, a member of the adhesion G protein-coupled receptor family, is abundantly expressed in cells of the developing cerebral cortex, including neural progenitor cells and developing neurons. The human GPR56 gene has multiple presumptive promoters that drive the expression of the GPR56 protein in distinct patterns. Similar to coding mutations of the human GPR56 gene that may cause GPR56 dysfunction, a 15-bp homozygous deletion in the cis-regulatory element upstream of the noncoding exon 1 of GPR56 (e1m) leads to the cerebral cortex malformation and epilepsy. To clarify the expression profile of the e1m promoter-driven GPR56 in primate brain, we generated a transgenic marmoset line in which EGFP is expressed under the control of the human minimal e1m promoter. In contrast to the endogenous GPR56 protein, which is highly enriched in the ventricular zone of the cerebral cortex, EGFP is mostly expressed in developing neurons in the transgenic fetal brain. Furthermore, EGFP is predominantly expressed in GABAergic neurons, whereas the total GPR56 protein is evenly expressed in both GABAergic and glutamatergic neurons, suggesting the GABAergic neuron-preferential activity of the minimal e1m promoter. These results indicate a possible pathogenic role for GABAergic neuron in the cerebral cortex of patients with GPR56 mutations.


Asunto(s)
Neuronas GABAérgicas/metabolismo , Regiones Promotoras Genéticas/genética , Receptores Acoplados a Proteínas G/genética , Animales , Animales Modificados Genéticamente/genética , Secuencia de Bases/genética , Encéfalo/metabolismo , Callithrix/genética , Callithrix/metabolismo , Movimiento Celular/genética , Corteza Cerebral/metabolismo , Expresión Génica/genética , Homocigoto , Humanos , Mutación/genética , Células-Madre Neurales/metabolismo , Polimicrogiria/genética , Polimicrogiria/metabolismo , Polimicrogiria/patología , Receptores Acoplados a Proteínas G/metabolismo , Eliminación de Secuencia/genética
8.
Stem Cells Dev ; 29(12): 761-773, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32188344

RESUMEN

Mammalian pluripotent stem cells are thought to exist in two states: naive and primed. Generally, unlike those in rodents, pluripotent stem cells in primates, including humans, are regarded as being in the primed pluripotent state. Recently, several groups reported the existence of naive pluripotent stem cells in humans. In this study, we report the conversion of primed state embryonic stem cells from common marmoset, a New World monkey, to the naive state using transgenes. The cells showed typical naive state features, including dome-like colony morphology, growth factor requirement, gene expression profile, X chromosome activation state, and energy metabolic status. Moreover, interspecies chimeric embryo formation ability with mouse embryos was increased in the naive state. This technique can be applied in basic medical research using nonhuman primates, such as preclinical use of naive pluripotent stem cells and generating genetically modified primates.


Asunto(s)
Células Madre Embrionarias/metabolismo , Ingeniería Genética/métodos , Transgenes , Animales , Callithrix , Línea Celular , Forma de la Célula , Quimera/genética , Quimera/metabolismo , Células Madre Embrionarias/citología , Metabolismo Energético , Transcriptoma , Inactivación del Cromosoma X
9.
Biochem Biophys Res Commun ; 515(4): 593-599, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31178141

RESUMEN

The common marmoset (marmoset; Callithrix jacchus) shows anatomical and physiological features that are in common with humans. Establishing induced pluripotent stem cells (iPSCs) from marmosets holds promise for enhancing the utility of the animal model for biomedical and preclinical studies. However, in spite of the presence of some previous reports on marmoset iPSCs, the reprogramming technology in marmosets is still under development. In particular, the efficacy of RNA-based reprogramming has not been thoroughly investigated. In this study, we attempted RNA-based reprogramming for deriving iPSCs from marmoset fibroblasts. Although we failed to derive iPSC colonies from marmoset fibroblasts by using a conventional RNA-based reprogramming method previously validated in human fibroblasts, we succeeded in deriving colony-forming cells with a modified induction medium supplemented with a novel set of small molecules. Importantly, following one-week culture of the colony-forming cells in conventional embryonic stem cell (ESC) medium, we obtained iPSCs which express endogenous pluripotent markers and show a differentiation potential into all three germ layers. Taken together, our results indicate that RNA-based reprogramming, which is valuable for deriving transgene-free iPSCs, is applicable to marmosets.


Asunto(s)
Técnicas de Cultivo de Célula , Reprogramación Celular , Fibroblastos/citología , Células Madre Pluripotentes Inducidas/citología , ARN/química , Animales , Callithrix , Diferenciación Celular , Células Madre Embrionarias/citología , Femenino , Regulación de la Expresión Génica , Humanos , Transcriptoma , Transgenes
10.
Sci Rep ; 9(1): 1528, 2019 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-30728412

RESUMEN

Genome editing technology greatly facilitates the genetic modification of various cells and animals. The common marmoset (Callithrix jacchus), a small non-human primate which exhibits high reproductive efficiency, is a widely used animal model in biomedical research. Developing genome editing techniques in the common marmoset will further enhance its utility. Here, we report the successful establishment of a knock-in (KI) method for marmoset embryonic stem cells (ESCs), which is based on the CRISPR-Cas9 system. The use of CRISPR-Cas9, mediated by homologous recombination (HR), enhanced the KI efficiency in marmoset ESCs. Furthermore, we succeeded in performing KI in early-stage marmoset embryos. In the course of the experiments, we found that HR in the marmoset ESCs is innately highly efficient. This suggested that the marmoset possesses a repair mechanism for DNA double-strand breaks. The current study will facilitate the generation of genetically modified marmosets and gene function analysis in the marmoset.


Asunto(s)
Sistemas CRISPR-Cas , Roturas del ADN de Doble Cadena , Embrión de Mamíferos/citología , Células Madre Embrionarias/citología , Edición Génica , Técnicas de Sustitución del Gen/métodos , Células-Madre Neurales/citología , Animales , Callithrix , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Femenino , Factores de Transcripción Forkhead/antagonistas & inhibidores , Factores de Transcripción Forkhead/genética , Marcación de Gen , Recombinación Homóloga , Humanos , Masculino , Modelos Animales , Proteína Proteolipídica de la Mielina/antagonistas & inhibidores , Proteína Proteolipídica de la Mielina/genética , Células-Madre Neurales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA