Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Cell ; 142(5): 787-99, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20813264

RESUMEN

The complexity of organogenesis hinders in vitro generation of organs derived from a patient's pluripotent stem cells (PSCs), an ultimate goal of regenerative medicine. Mouse wild-type PSCs injected into Pdx1(-/-) (pancreatogenesis-disabled) mouse blastocysts developmentally compensated vacancy of the pancreatic "developmental niche," generating almost entirely PSC-derived pancreas. To examine the potential for xenogenic approaches in blastocyst complementation, we injected mouse or rat PSCs into rat or mouse blastocysts, respectively, generating interspecific chimeras and thus confirming that PSCs can contribute to xenogenic development between mouse and rat. The development of these mouse/rat chimeras was primarily influenced by host blastocyst and/or foster mother, evident by body size and species-specific organogenesis. We further injected rat wild-type PSCs into Pdx1(-/-) mouse blastocysts, generating normally functioning rat pancreas in Pdx1(-/-) mice. These data constitute proof of principle for interspecific blastocyst complementation and for generation in vivo of organs derived from donor PSCs using a xenogenic environment.


Asunto(s)
Blastocisto , Quimera/embriología , Páncreas/citología , Páncreas/embriología , Células Madre Pluripotentes , Animales , Diabetes Mellitus/inducido químicamente , Diabetes Mellitus/terapia , Desarrollo Embrionario , Técnicas de Sustitución del Gen , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos , Organogénesis , Ratas , Ratas Wistar , Transactivadores/genética
2.
Nature ; 542(7640): 191-196, 2017 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-28117444

RESUMEN

Islet transplantation is an established therapy for diabetes. We have previously shown that rat pancreata can be created from rat pluripotent stem cells (PSCs) in mice through interspecies blastocyst complementation. Although they were functional and composed of rat-derived cells, the resulting pancreata were of mouse size, rendering them insufficient for isolating the numbers of islets required to treat diabetes in a rat model. Here, by performing the reverse experiment, injecting mouse PSCs into Pdx-1-deficient rat blastocysts, we generated rat-sized pancreata composed of mouse-PSC-derived cells. Islets subsequently prepared from these mouse-rat chimaeric pancreata were transplanted into mice with streptozotocin-induced diabetes. The transplanted islets successfully normalized and maintained host blood glucose levels for over 370 days in the absence of immunosuppression (excluding the first 5 days after transplant). These data provide proof-of-principle evidence for the therapeutic potential of PSC-derived islets generated by blastocyst complementation in a xenogeneic host.


Asunto(s)
Diabetes Mellitus Experimental/terapia , Xenoinjertos/fisiología , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/fisiología , Organogénesis , Animales , Blastocisto/citología , Blastocisto/metabolismo , Glucemia/metabolismo , Quimera , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Femenino , Xenoinjertos/inmunología , Proteínas de Homeodominio , Islotes Pancreáticos/citología , Islotes Pancreáticos/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Masculino , Ratones , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/trasplante , Ratas , Factores de Tiempo , Transactivadores/deficiencia
3.
J Reprod Dev ; 69(1): 48-52, 2023 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-36529517

RESUMEN

We examined various methods to enhance the accessibility of intracytoplasmic sperm injection (ICSI) technology to more users by making the technique easier, more efficient, and practical. First, the methods for artificially removing the mouse sperm tail were evaluated. Trypsin treatment was found to efficiently remove the sperm tails. The resultant sperm cells had a lower oocyte activation capacity; however, the use of activated oocytes resulted in the same fecundity as that of fresh, untreated sperm. Pre-activated oocytes were more resistant to physical damage, showed higher survival rates, and required less time per injection. Testing this method in rats yielded similar results, although the oocyte activation method was different. Remarkably, this method resulted in higher birth rates of rat progeny than with conventional methods of rat ICSI. Our method thereby streamlines mouse and rat ICSI, making it more accessible to laboratories across many disciplines.


Asunto(s)
Inyecciones de Esperma Intracitoplasmáticas , Cola del Espermatozoide , Ratones , Masculino , Ratas , Animales , Inyecciones de Esperma Intracitoplasmáticas/métodos , Tripsina , Semen , Espermatozoides/fisiología , Oocitos
4.
Development ; 142(18): 3222-30, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26023098

RESUMEN

Functional assay limitations are an emerging issue in characterizing human pluripotent stem cells (PSCs). With rodent PSCs, chimera formation using pre-implantation embryos is the gold-standard assay of pluripotency (competence of progeny to differentiate into all three germ layers). In human PSCs (hPSCs), however, this can only be monitored via teratoma formation or in vitro differentiation, as ethical concerns preclude generation of human-human or human-animal chimeras. To circumvent this issue, we developed a functional assay utilizing interspecific blastocyst injection and in vitro culture (interspecies in vitro chimera assay) that enables the development and observation of embryos up to headfold stage. The assay uses mouse pre-implantation embryos and rat, monkey and human PSCs to create interspecies chimeras cultured in vitro to the early egg-cylinder stage. Intra- and interspecific chimera assays with rodent PSC lines were performed to confirm the consistency of results in vitro and in vivo. The behavior of chimeras developed in vitro appeared to recapitulate that of chimeras developed in vivo; that is, PSC-derived cells survived and were integrated into the epiblast of egg-cylinder-stage embryos. This indicates that the interspecific in vitro chimera assay is useful in evaluating the chimera-forming ability of rodent PSCs. However, when human induced PSCs (both conventional and naïve-like types) were injected into mouse embryos and cultured, some human cells survived but were segregated; unlike epiblast-stage rodent PSCs, they never integrated into the epiblast of egg-cylinder-stage embryos. These data suggest that the mouse-human interspecies in vitro chimera assay does not accurately reflect the early developmental potential/process of hPSCs. The use of evolutionarily more closely related species as host embryos might be necessary to evaluate the developmental potency of hPSCs.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Quimera/embriología , Desarrollo Embrionario/fisiología , Técnicas In Vitro/métodos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Animales , Línea Celular , Haplorrinos , Humanos , Ratones , Análisis por Micromatrices , Microinyecciones , Ratas , Especificidad de la Especie
5.
BMC Biol ; 14(1): 103, 2016 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-27912755

RESUMEN

BACKGROUND: The specificity of synaptic connections is fundamental for proper neural circuit function. Specific neuronal connections that underlie information processing in the sensory cortex are initially established without sensory experiences to a considerable extent, and then the connections are individually refined through sensory experiences. Excitatory neurons arising from the same single progenitor cell are preferentially connected in the postnatal cortex, suggesting that cell lineage contributes to the initial wiring of neurons. However, the postnatal developmental process of lineage-dependent connection specificity is not known, nor how clonal neurons, which are derived from the same neural stem cell, are stamped with the identity of their common neural stem cell and guided to form synaptic connections. RESULTS: We show that cortical excitatory neurons that arise from the same neural stem cell and reside within the same layer preferentially establish reciprocal synaptic connections in the mouse barrel cortex. We observed a transient increase in synaptic connections between clonal but not nonclonal neuron pairs during postnatal development, followed by selective stabilization of the reciprocal connections between clonal neuron pairs. Furthermore, we demonstrate that selective stabilization of the reciprocal connections between clonal neuron pairs is impaired by the deficiency of DNA methyltransferase 3b (Dnmt3b), which determines DNA-methylation patterns of genes in stem cells during early corticogenesis. Dnmt3b regulates the postnatal expression of clustered protocadherin (cPcdh) isoforms, a family of adhesion molecules. We found that cPcdh deficiency in clonal neuron pairs impairs the whole process of the formation and stabilization of connections to establish lineage-specific connection reciprocity. CONCLUSIONS: Our results demonstrate that local, reciprocal neural connections are selectively formed and retained between clonal neurons in layer 4 of the barrel cortex during postnatal development, and that Dnmt3b and cPcdhs are required for the establishment of lineage-specific reciprocal connections. These findings indicate that lineage-specific connection reciprocity is predetermined by Dnmt3b during embryonic development, and that the cPcdhs contribute to postnatal cortical neuron identification to guide lineage-dependent synaptic connections in the neocortex.


Asunto(s)
Cadherinas/metabolismo , Neuronas/metabolismo , Sinapsis/metabolismo , Animales , Cadherinas/genética , Células Cultivadas , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Electrofisiología , Femenino , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vías Nerviosas/fisiología , Transmisión Sináptica/genética , Transmisión Sináptica/fisiología , ADN Metiltransferasa 3B
6.
Dev Biol ; 407(2): 331-43, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26165599

RESUMEN

Fetal hepatic stem/progenitor cells, hepatoblasts, are highly proliferative cells and the source of both hepatocytes and cholangiocytes. In contrast, mature hepatocytes have a low proliferative potency and high metabolic functions. Cell proliferation is regulated by cell cycle-related molecules. However, the correlation between cell cycle regulation and hepatic maturation are still unknown. To address this issue, we revealed that the cell cycle inhibitor p57(Kip2) was expressed in the hepatoblasts and mesenchymal cells of fetal liver in a spatiotemporal manner. In addition, we found that hepatoblasts in p57(Kip2)-/- mice were highly proliferative and had deficient maturation compared with those in wild-type (WT) mice. However, there were no remarkable differences in the expression levels of cell cycle- and bipotency-related genes except for Ccnd2. Furthermore, p57(Kip2)-/- hepatoblasts could differentiate into mature hepatocytes in p57(Kip2)-/- and WT chimeric mice, suggesting that the intrinsic activity of p57(Kip2) does not simply regulate hepatoblast maturation.


Asunto(s)
Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Hepatocitos/metabolismo , Hígado/embriología , Hígado/metabolismo , Animales , Ciclo Celular/genética , Diferenciación Celular/genética , Proliferación Celular , Quimera , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/deficiencia , Epitelio/embriología , Epitelio/metabolismo , Espacio Extracelular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hepatocitos/citología , Hígado/citología , Ratones Endogámicos C57BL , Factores de Transcripción/metabolismo
7.
Mol Reprod Dev ; 83(6): 488-96, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27031189

RESUMEN

Round spermatid injection (ROSI) into unfertilized oocytes enables a male with a severe spermatogenesis disorder to have children. One limitation of the application of this technique in the clinic is the identification and isolation of round spermatids from testis tissue. Here we developed an efficient and simple method to isolate rodent haploid round spermatids using flow cytometric cell sorting, based on DNA content (stained with Hoechst 33342 or Dye Cycle Violet) or by cell diameter and granularity (forward and side scatter). ROSI was performed with round spermatids selected by flow cytometry, and we obtained healthy offspring from unstained cells. This non-invasive method could therefore be an effective option for breeding domestic animals and human male infertility treatment. Mol. Reprod. Dev. 83: 488-496, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Citometría de Flujo/métodos , Espermátides/citología , Animales , Bencimidazoles/química , ADN/metabolismo , Masculino , Ratones , Ratas , Ratas Wistar , Inyecciones de Esperma Intracitoplasmáticas , Espermátides/metabolismo
8.
Hepatol Res ; 46(8): 816-28, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26584962

RESUMEN

AIM: Hepatic progenitor cells, called hepatoblasts, are highly proliferative and exhibit bipotential differentiation into hepatocytes and cholangiocytes in the fetal liver. Thus, they are the ideal source for transplantation therapy. Although several studies have been performed in vitro, the molecular mechanisms regulating hepatoblast differentiation in vivo following transplantation remain poorly understood. The aim of this study was to investigate an in vivo model to analyze hepatoblast bipotency and proliferative ability. METHODS: Hepatic transplantation model using Cre-inducible diphtheria toxin receptor-transgenic mice (iDTR), and albafpCre mice expressing Cre under the control of albumin and α-fetoprotein (AFP) regulatory elements were established. Fresh hepatoblasts were transplanted into diphtheria toxin (DT)-injected iDTRalbafpCre mice and we analyzed their differentiation and proliferation abilities by immunostaining and gene expression profiles. RESULTS: Fresh hepatoblasts transplanted into DT-injected iDTRalbafpCre mice engrafted and differentiated into both hepatocytes and cholangiocytes. Additionally, the number of engrafted hepatoblast-derived hepatocytes increased following partial hepatectomy and serial DT injections. Expression levels of hepatic functional genes in transplanted hepatoblast-derived hepatocytes were similar to that of normal hepatocytes. CONCLUSION: In our iDTRalbafpCre transplantation model, fresh hepatoblasts could differentiate into hepatocytes and cholangiocytes. In addition, these donor cells were induced to proliferate by the following liver injury stimulation. This result suggests that this model is valuable for investigating hepatoblast differentiation pathways in vivo.

9.
J Biol Chem ; 289(43): 29892-911, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25143383

RESUMEN

Identification of genes specifically expressed in stem/progenitor cells is an important issue in developmental and stem cell biology. Genome-wide gene expression analyses in liver cells performed in this study have revealed a strong expression of X-linked genes that include members of the brain-expressed X-linked (Bex) gene family in stem/progenitor cells. Bex family genes are expressed abundantly in the neural cells and have been suggested to play important roles in the development of nervous tissues. However, the physiological role of its individual members and the precise expression pattern outside the nervous system remain largely unknown. Here, we focused on Bex2 and examined its role and expression pattern by generating knock-in mice; the enhanced green fluorescence protein (EGFP) was inserted into the Bex2 locus. Bex2-deficient mice were viable and fertile under laboratory growth conditions showing no obvious phenotypic abnormalities. Through an immunohistochemical analysis and flow cytometry-based approach, we observed unique EGFP reporter expression patterns in endocrine and stem/progenitor cells of the liver, pyloric stomach, and hematopoietic system. Although Bex2 seems to play redundant roles in vivo, these results suggest the significance and potential applications of Bex2 in studies of endocrine and stem/progenitor cells.


Asunto(s)
Células Endocrinas/metabolismo , Marcación de Gen , Proteínas del Tejido Nervioso/metabolismo , Células Madre/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Linaje de la Célula/genética , Proliferación Celular , Células Endocrinas/citología , Endodermo/citología , Femenino , Feto/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Sitios Genéticos , Proteínas Fluorescentes Verdes/metabolismo , Hematopoyesis/genética , Hígado/embriología , Hígado/metabolismo , Masculino , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Especificidad de Órganos , Fenotipo , Regiones Promotoras Genéticas/genética , Células Madre/citología , Transcripción Genética
10.
Biol Reprod ; 91(4): 89, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25165118

RESUMEN

Primordial germ cells (PGCs) are germ cell progenitors in the fetal genital ridge; female PGCs give rise to definitive oocytes that contribute to the next generation. Artificial PGCs have been induced in vitro from pluripotent stem cells and gonad-like tissue has been induced in vivo by cotransplantation of PGCs with PGC-free gonadal cells. To apply these technologies to human infertility treatment or conservation of rare species, PGC transplantation must be established in xenogenic animals. Here, we established a xenogeneic transplantation model by inducing ovary-like tissue from PGCs in xenogenic animals. We transplanted enzymatically dispersed PGCs with PGC-free gonadal cells under the kidney capsule of xenogenic immunodeficient animals. The transplanted cells formed ovary-like tissues under the kidney capsule. These tissues were histologically similar to the normal gonad and expressed the oocyte markers Vasa and Stella. In addition, mouse germinal vesicle-stage oocyte-like cells collected from ovary-like tissue in rats matured to metaphase II via in vitro maturation and gave rise to offspring by intracytoplasmic sperm injection. Our studies show that rat/mouse female PGCs and PGC-free gonadal cells can develop and reconstruct ovary-like tissue containing functional oocytes in an ectopic xenogenic microenvironment.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/trasplante , Oocitos/fisiología , Animales , Benzofuranos , Femenino , Células Germinativas , Xenoinjertos , Riñón/citología , Masculino , Ratones , Ratones Endogámicos ICR , Ratones SCID , Oogénesis/fisiología , Quinolinas , Ratas , Ratas Endogámicas , Trasplante de Células Madre
11.
Transgenic Res ; 22(2): 411-6, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22875289

RESUMEN

The factors responsible for conferring germline competence in embryonic stem (ES) cell lines remain unidentified. In the present study, rat ES cell lines (n = 17) were established with 3i medium (SU5402, PD0325901, CHIR99021), 2i medium (PD0325901, CHIR99021) or 2iF medium (PD0325901, CHIR99021, forskolin), and their potential for germline transmission to the G1 generation was examined. Rat strains were divided into an albino group (F344, Wistar or CAG/Venus transgenic rats with the Wistar background) or a colored coat group (Brown-Norway, Dark-Agouti, or BLK rats selected from >F3 generations of Wistar × Dark-Agouti rats based on their black coat color). Successful germline transmission was observed in 57 % (4/7), 40 % (2/5) and 100 % (5/5) of the ES cells established with 3i, 2i and 2iF media, respectively. ES cell lines from the homozygous CAG/Venus transgenic rats were established in all three media, but only the lines established with the 2iF medium were germline-competent. Neither coat-color (albino: 64 %, 7/11; colored: 67 %, 4/6) nor gender of the ES cell lines (XX: 67 %, 2/3; XY: 64 %, 9/14) were likely to affect germline transmission.


Asunto(s)
Técnicas de Cultivo de Célula , Células Madre Embrionarias/citología , Células Germinativas/citología , Animales , Linaje de la Célula/genética , Embrión de Mamíferos/citología , Ratas , Ratas Transgénicas , Estudios Retrospectivos
12.
Mol Reprod Dev ; 79(6): 402-12, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22499253

RESUMEN

This study was undertaken to generate rat offspring via tetraploid blastocyst complementation with embryonic stem (ES) cells. Tetraploid blastocysts were prepared by electrofusion of blastomeres from two-cell stage embryos, and subsequent in vivo culture for 4 days. Microinjection into the tetraploid blastocoel of an inner cell mass isolated by immunosurgery resulted in the generation of rat offspring, suggesting the successful contribution of tetraploid blastocysts to their placenta. Tetraploid blastocyst complementation was attempted with a total of 4 ES cell lines (2 lines of female karyotype and 2 lines of male karyotype). In the rESWIv-3i-5 (XX) cell line, normal-sized fetuses with heartbeats were harvested on E11.5 (12.1%), E12.5 (9.5%), and E13.5 (9.1%), but no viable fetuses were detected on E14.5. Similarly, use of the rESWIv-3i-1 (XX) cell line resulted in no viable fetus production on E14.5. Using the rESBLK2i-1 (XY) cell line, viable fetuses were harvested not only on E11.5-E13.5 (2.6-5.5%), but also on E14.5 (3.0%). The transfer of a total of 487 tetraploid blastocysts complemented with rESBLK2i-1 cells resulted in 256 implantation sites (52.6%) on E21.5, but no viable offspring was detected. Use of the rESBLK2i-1/huKO (XY) cell line also resulted in no viable offspring production on E21.5. Analyses of the methylation pattern in differentially methylated regions and transcript level of genes that are imprinted in mice (H19, Meg3, Igf2r, Peg5, and Peg10) in the E14.5 conceptuses indicated a marked difference between the ES cell-derived and control normal fetuses, but not between the tetraploid and control diploid placenta.


Asunto(s)
Blastocisto/fisiología , Fusión Celular , Células Madre Embrionarias/fisiología , Desarrollo Fetal , Tetraploidía , Animales , Blastocisto/citología , Línea Celular , Desarrollo Embrionario , Células Madre Embrionarias/citología , Femenino , Masculino , Microinyecciones , Placenta/metabolismo , Embarazo , Ratas , Ratas Transgénicas
13.
Biol Reprod ; 85(1): 208-17, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21471297

RESUMEN

Spermatogonial stem cells (SSCs) are the only stem cells in the body with germline potential, which makes them an attractive target for germline modification. We previously showed the feasibility of homologous recombination in mouse SSCs and produced knockout (KO) mice by exploiting germline stem (GS) cells, i.e., cultured spermatogonia with SSC activity. In this study, we report the successful homologous recombination in rat GS cells, which can be readily established by their ability to form germ cell colonies on culture plates whose surfaces are hydrophilic and neutrally charged and thus limit somatic cell binding. We established a drug selection protocol for GS cells under hypoxic conditions. The frequency of the homologous recombination of the Ocln gene was 4.2% (2 out of 48 clones). However, these GS cell lines failed to produce offspring following xenogeneic transplantation into mouse testes and microinsemination, suggesting that long-term culture and drug selection have a negative effect on GS cells. Nevertheless, our results demonstrate the feasibility of gene targeting in rat GS cells and pave the way toward the generation of KO rats.


Asunto(s)
Recombinación Genética , Espermatogonias/fisiología , Células Madre/fisiología , Animales , Técnicas de Cultivo de Célula , Masculino , Ratones , Ratones Desnudos , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Selección Genética , Inyecciones de Esperma Intracitoplasmáticas
14.
Sci Rep ; 8(1): 15289, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30327488

RESUMEN

To study development of the conceptus in xenogeneic environments, we assessed interspecies chimera formation as well as tetraploid complementation between mouse and rat. Overall contribution of donor PSC-derived cells was lower in interspecies chimeras than in intraspecies chimeras, and high donor chimerism was associated with anomalies or embryonic death. Organ to organ variation in donor chimerism was greater in interspecies chimeras than in intraspecies chimeras, suggesting species-specific affinity differences among interacting molecules necessary for organogenesis. In interspecies tetraploid complementation, embryo development was near normal until the stage of placental formation, after which no embryos survived.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Desarrollo Embrionario , Organogénesis , Tetraploidía , Quimera por Trasplante , Animales , Blastocisto/citología , Femenino , Ratones , Ratones Endogámicos C57BL , Células Madre Pluripotentes/citología , Embarazo , Ratas , Ratas Wistar , Especificidad de la Especie , Quimera por Trasplante/crecimiento & desarrollo , Quimera por Trasplante/inmunología
15.
Sci Rep ; 7(1): 12765, 2017 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-28986544

RESUMEN

iNKT cells play important roles in immune regulation by bridging the innate and acquired immune systems. The functions of iNKT cells have been investigated in mice lacking the Traj18 gene segment that were generated by traditional embryonic stem cell technology, but these animals contain a biased T cell receptor (TCR) repertoire that might affect immune responses. To circumvent this confounding factor, we have generated a new strain of iNKT cell-deficient mice by deleting the Traj18 locus using CRISPR/Cas9 technology, and these animals contain an unbiased TCR repertoire. We employed these mice to investigate the contribution of iNKT cells to metabolic disease and found a pathogenic role of these cells in obesity-associated insulin-resistance. The new Traj18-deficient mouse strain will assist in studies of iNKT cell biology.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Enfermedades Metabólicas/inmunología , Células T Asesinas Naturales/inmunología , Animales , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Sitios Genéticos , Intolerancia a la Glucosa/patología , Células HEK293 , Humanos , Resistencia a la Insulina , Enfermedades Metabólicas/patología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mutación/genética , Obesidad/patología , ARN Guía de Kinetoplastida/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo
16.
Cell Stem Cell ; 19(5): 587-592, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27814480

RESUMEN

Cell types more advanced in development than embryonic stem cells, such as EpiSCs, fail to contribute to chimeras when injected into pre-implantation-stage blastocysts, apparently because the injected cells undergo apoptosis. Here we show that transient promotion of cell survival through expression of the anti-apoptotic gene BCL2 enables EpiSCs and Sox17+ endoderm progenitors to integrate into blastocysts and contribute to chimeric embryos. Upon injection into blastocyst, BCL2-expressing EpiSCs contributed to all bodily tissues in chimeric animals while Sox17+ endoderm progenitors specifically contributed in a region-specific fashion to endodermal tissues. In addition, BCL2 expression enabled rat EpiSCs to contribute to mouse embryonic chimeras, thereby forming interspecies chimeras that could survive to adulthood. Our system therefore provides a method to overcome cellular compatibility issues that typically restrict chimera formation. Application of this type of approach could broaden the use of embryonic chimeras, including region-specific chimeras, for basic developmental biology research and regenerative medicine.


Asunto(s)
Apoptosis , Quimera/metabolismo , Embrión de Mamíferos/citología , Animales , Células Madre Embrionarias/citología , Ratones , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factores de Transcripción SOX/metabolismo
17.
Stem Cells Dev ; 24(2): 182-9, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25192056

RESUMEN

Generation of functional organs from patients' own cells is one of the ultimate goals of regenerative medicine. As a novel approach to creation of organs from pluripotent stem cells (PSCs), we employed blastocyst complementation in organogenesis-disabled animals and successfully generated PSC-derived pancreas and kidneys. Blastocyst complementation, which exploits the capacity of PSCs to participate in forming chimeras, does not, however, exclude contribution of PSCs to the development of tissues-including neural cells or germ cells-other than those specifically targeted by disabling of organogenesis. This fact provokes ethical controversy if human PSCs are to be used. In this study, we demonstrated that forced expression of Mix-like protein 1 (encoded by Mixl1) can be used to guide contribution of mouse embryonic stem cells to endodermal organs after blastocyst injection. We then succeeded in applying this method to generate functional pancreas in pancreatogenesis-disabled Pdx1 knockout mice using a newly developed tetraploid-based organ-complementation method. These findings hold promise for targeted organ generation from patients' own PSCs in livestock animals.


Asunto(s)
Blastocisto/metabolismo , Expresión Génica , Proteínas de Homeodominio/biosíntesis , Células Madre Pluripotentes Inducidas/metabolismo , Riñón/embriología , Organogénesis , Páncreas/embriología , Animales , Blastocisto/citología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteínas de Homeodominio/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Riñón/citología , Ratones , Ratones Noqueados , Páncreas/citología , Transactivadores/biosíntesis
18.
PLoS One ; 9(3): e92973, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24667806

RESUMEN

The most stringent criterion for evaluating pluripotency is generation of chimeric animals with germline transmission ability. Because the quality of induced pluripotent stem cell (iPSC) lines is heterogeneous, an easy and accurate system to evaluate these abilities would be useful. In this study, we describe a simple but comprehensive system for generating and evaluating iPSCs by single transfection of multiple piggyBac (PB) plasmid vectors encoding Tet-inducible polycistronic reprogramming factors, a pluripotent-cell-specific reporter, a constitutively active reporter, and a sperm-specific reporter. Using this system, we reprogrammed 129 and NOD mouse embryonic fibroblasts into iPSCs, and then evaluated the molecular and functional properties of the resultant iPSCs by quantitative RT-PCR analysis and chimera formation assays. The iPSCs contributed extensively to chimeras, as indicated by the constitutively active TagRFP reporter, and also differentiated into sperm, as indicated by the late-spermatogenesis-specific Acr (acrosin)-EGFP reporter. Next, we established secondary MEFs from E13.5 chimeric embryos and efficiently generated secondary iPSCs by simple addition of doxycycline. Finally, we applied this system to establishment and evaluation of rat iPSCs and production of rat sperm in mouse-rat interspecific chimeras. By monitoring the fluorescence of Acr-EGFP reporter, we could easily detect seminiferous tubules containing rat iPSC-derived spermatids and sperm. And, we succeeded to obtain viable offspring by intracytoplasmic sperm injection (ICSI) using these haploid male germ cells. We propose that this system will enable robust strategies for induction and evaluation of iPSCs, not only in rodents but also in other mammals. Such strategies will be especially valuable in non-rodent species, in which verification of germline transmission by mating is inefficient and time-consuming.


Asunto(s)
Reprogramación Celular/genética , Vectores Genéticos/genética , Células Madre Pluripotentes Inducidas/citología , Transfección/métodos , Animales , Masculino , Ratones , Plásmidos/genética , Ratas , Espermátides/citología , Espermátides/metabolismo
19.
Stem Cells Dev ; 23(2): 107-14, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24010570

RESUMEN

This study was undertaken to establish rat embryonic stem (ES) cells from parthenogenetically developing blastocysts. Ten blastocysts were prepared by treatment of ovulated rat oocytes with ionomycin and cycloheximide, and three alkaline phosphatase-positive ES cell lines were established using the N2B27 medium supplemented with mitogen activated protein kinase kinase inhibitor PD0325901, glycogen synthase kinase 3 inhibitor CHIR99021, rat leukemia inhibitory factor, and forskolin. Expression of stem cell marker genes (Oct-4, rNanog, Fgf-4, and Rex-1) was confirmed in all three ES cell lines by reverse transcriptase-polymerase chain reaction (RT-PCR). Combined bisulfite restriction analysis showed that the differentially methylated region locus of five imprinted genes (H19, Meg3IG, Igf2r, Peg5, and Peg10) in these ES cells remained to be demethylated or was hypomethylated, which was similar to that in control ES cells established from normal blastocysts. Characteristics of the parthenogenetic blastocyst-derived ES cells were successfully transmitted to the next generation through a chimeric rat for one of the three ES cell lines. This is the first report on germline-competent (genuine) ES cells derived from parthenogenetically developing rat blastocysts.


Asunto(s)
Blastocisto/metabolismo , Células Madre Embrionarias/metabolismo , Oocitos/metabolismo , Partenogénesis , Adyuvantes Inmunológicos/farmacología , Animales , Benzamidas/farmacología , Ionóforos de Calcio/farmacología , Diferenciación Celular , Células Cultivadas , Colforsina/farmacología , Cicloheximida/farmacología , Metilación de ADN , Difenilamina/análogos & derivados , Difenilamina/farmacología , Femenino , Factor 4 de Crecimiento de Fibroblastos/biosíntesis , Marcadores Genéticos , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Ionomicina/farmacología , Factor Inhibidor de Leucemia/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Oocitos/efectos de los fármacos , Piridinas/farmacología , Pirimidinas/farmacología , Ratas , Factores de Transcripción/biosíntesis
20.
Stem Cells Dev ; 21(16): 2981-6, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22564063

RESUMEN

Recent discovery of a method for derivation and culture of germline-competent rat pluripotent stem cells (PSCs) enables generation of transgenic rats or knock-out rats via genetic modification of such PSCs. This opens the way to use rats, as is routine in mice, for analyses of gene functions or physiological features. In mouse or human, one widely used technique to express a gene of interest stably and ubiquitously is to insert that gene into the Rosa26 locus via gene targeting of PSCs. Rosa26 knock-in mice conditionally expressing a reporter or a toxin gene have contributed to tracing or ablation of specific cell lineages. We successfully identified a rat orthologue of the mouse Rosa26 locus. Insertion of tdTomato, a variant of red fluorescent protein, into the Rosa26 locus of PSCs of various rat strains allows ubiquitous expression of tdTomato. Through germline transmission of one Rosa26-tdTomato knock-in embryonic stem cell line, we also obtained tdTomato knock-in rats. These expressed tdTomato ubiquitously throughout their bodies, which indicates that the rat Rosa26 locus conserves functions of its orthologues in mouse and human. The new tools described here (targeting vectors, knock-in PSCs, and rats) should be useful for a variety of research using rats.


Asunto(s)
Técnicas de Sustitución del Gen/métodos , Sitios Genéticos/genética , Proteínas Luminiscentes/metabolismo , Animales , Blastocisto/citología , Blastocisto/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Marcación de Gen , Humanos , Ratones , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Ratas , Ratas Transgénicas , Proteína Fluorescente Roja
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA