Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Commun Med (Lond) ; 4(1): 111, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38862768

RESUMEN

BACKGROUND: Biliary atresia (BA) is an intractable disease of unknown cause that develops in the neonatal period. It causes jaundice and liver damage due to the destruction of extrahepatic biliary tracts,. We have found that heterozygous knockout mice of the SRY related HMG-box 17 (Sox17) gene, a master regulator of stem/progenitor cells in the gallbladder wall, exhibit a condition like BA. However, the precise contribution of hypoplastic gallbladder wall to the pathogenesis of hepatobiliary disease in Sox17 heterozygous embryos and human BA remains unclear. METHODS: We employed cholangiography and histological analyses in the mouse BA model. Furthermore, we conducted a retrospective analysis of human BA. RESULTS: We show that gallbladder wall hypoplasia causes abnormal multiple connections between the hilar hepatic bile ducts and the gallbladder-cystic duct in Sox17 heterozygous embryos. These multiple hilar extrahepatic ducts fuse with the developing intrahepatic duct walls and pull them out of the liver parenchyma, resulting in abnormal intrahepatic duct network and severe cholestasis. In human BA with gallbladder wall hypoplasia (i.e., abnormally reduced expression of SOX17), we also identify a strong association between reduced gallbladder width (a morphometric parameter indicating gallbladder wall hypoplasia) and severe liver injury at the time of the Kasai surgery, like the Sox17-mutant mouse model. CONCLUSIONS: Together with the close correlation between gallbladder wall hypoplasia and liver damage in both mouse and human cases, these findings provide an insight into the critical role of SOX17-positive gallbladder walls in establishing functional bile duct networks in the hepatic hilus of neonates.


Biliary atresia (BA) is a disease in newborns that causes a serious liver condition due to damage to the bile ducts (the pathways that carry bile juice). Although reduced function of a key gene called Sox17, which is essential for forming the gallbladder wall, has been observed in some BA cases, the link between gallbladder issues and liver damage is unknown. This study has shown how damage spreads through the bile ducts in the liver around the time of birth when there are problems in the gallbladder wall due to reduced SOX17 function. The findings indicate that proper growth of the gallbladder wall during this critical period is essential for forming a normal network of bile ducts in the developing liver. This discovery is promising for early diagnosis and better treatment of BA in newborns.

2.
Nat Commun ; 15(1): 3366, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38684678

RESUMEN

Autologous skin grafting is a standard treatment for skin defects such as burns. No artificial skin substitutes are functionally equivalent to autologous skin grafts. The cultured epidermis lacks the dermis and does not engraft deep wounds. Although reconstituted skin, which consists of cultured epidermal cells on a synthetic dermal substitute, can engraft deep wounds, it requires the wound bed to be well-vascularized and lacks skin appendages. In this study, we successfully generate complete skin grafts with pluripotent stem cell-derived epidermis with appendages on p63 knockout embryos' dermis. Donor pluripotent stem cell-derived keratinocytes encroach the embryos' dermis by eliminating p63 knockout keratinocytes based on cell-extracellular matrix adhesion mediated cell competition. Although the chimeric skin contains allogenic dermis, it is engraftable as long as autologous grafts. Furthermore, we could generate semi-humanized skin segments by human keratinocytes injection into the amnionic cavity of p63 knockout mice embryos. Niche encroachment opens the possibility of human skin graft production in livestock animals.


Asunto(s)
Dermis , Queratinocitos , Ratones Noqueados , Trasplante de Piel , Animales , Trasplante de Piel/métodos , Queratinocitos/citología , Queratinocitos/trasplante , Humanos , Dermis/citología , Dermis/trasplante , Ratones , Epidermis/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/trasplante , Piel Artificial , Células Epidérmicas/trasplante , Células Epidérmicas/citología , Matriz Extracelular/metabolismo , Piel/citología
3.
Nat Commun ; 14(1): 4022, 2023 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-37419903

RESUMEN

Biomechanical cues are instrumental in guiding embryonic development and cell differentiation. Understanding how these physical stimuli translate into transcriptional programs will provide insight into mechanisms underlying mammalian pre-implantation development. Here, we explore this type of regulation by exerting microenvironmental control over mouse embryonic stem cells. Microfluidic encapsulation of mouse embryonic stem cells in agarose microgels stabilizes the naive pluripotency network and specifically induces expression of Plakoglobin (Jup), a vertebrate homolog of ß-catenin. Overexpression of Plakoglobin is sufficient to fully re-establish the naive pluripotency gene regulatory network under metastable pluripotency conditions, as confirmed by single-cell transcriptome profiling. Finally, we find that, in the epiblast, Plakoglobin was exclusively expressed at the blastocyst stage in human and mouse embryos - further strengthening the link between Plakoglobin and naive pluripotency in vivo. Our work reveals Plakoglobin as a mechanosensitive regulator of naive pluripotency and provides a paradigm to interrogate the effects of volumetric confinement on cell-fate transitions.


Asunto(s)
Desarrollo Embrionario , Estratos Germinativos , Animales , Ratones , Humanos , gamma Catenina/genética , gamma Catenina/metabolismo , Diferenciación Celular/genética , Estratos Germinativos/metabolismo , Desarrollo Embrionario/genética , Perfilación de la Expresión Génica , Blastocisto/metabolismo , Mamíferos/genética
4.
Nat Biotechnol ; 40(12): 1780-1793, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35760914

RESUMEN

Most methods for single-cell transcriptome sequencing amplify the termini of polyadenylated transcripts, capturing only a small fraction of the total cellular transcriptome. This precludes the detection of many long non-coding, short non-coding and non-polyadenylated protein-coding transcripts and hinders alternative splicing analysis. We, therefore, developed VASA-seq to detect the total transcriptome in single cells, which is enabled by fragmenting and tailing all RNA molecules subsequent to cell lysis. The method is compatible with both plate-based formats and droplet microfluidics. We applied VASA-seq to more than 30,000 single cells in the developing mouse embryo during gastrulation and early organogenesis. Analyzing the dynamics of the total single-cell transcriptome, we discovered cell type markers, many based on non-coding RNA, and performed in vivo cell cycle analysis via detection of non-polyadenylated histone genes. RNA velocity characterization was improved, accurately retracing blood maturation trajectories. Moreover, our VASA-seq data provide a comprehensive analysis of alternative splicing during mammalian development, which highlighted substantial rearrangements during blood development and heart morphogenesis.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Transcriptoma , Ratones , Animales , Análisis de Secuencia de ARN/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Empalme Alternativo/genética , ARN/metabolismo , Perfilación de la Expresión Génica/métodos , Mamíferos/genética
6.
Cell ; 185(5): 777-793.e20, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-35196500

RESUMEN

In development, lineage segregation is coordinated in time and space. An important example is the mammalian inner cell mass, in which the primitive endoderm (PrE, founder of the yolk sac) physically segregates from the epiblast (EPI, founder of the fetus). While the molecular requirements have been well studied, the physical mechanisms determining spatial segregation between EPI and PrE remain elusive. Here, we investigate the mechanical basis of EPI and PrE sorting. We find that rather than the differences in static cell surface mechanical parameters as in classical sorting models, it is the differences in surface fluctuations that robustly ensure physical lineage sorting. These differential surface fluctuations systematically correlate with differential cellular fluidity, which we propose together constitute a non-equilibrium sorting mechanism for EPI and PrE lineages. By combining experiments and modeling, we identify cell surface dynamics as a key factor orchestrating the correct spatial segregation of the founder embryonic lineages.


Asunto(s)
Blastocisto , Embrión de Mamíferos , Endodermo , Animales , Blastocisto/metabolismo , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Membrana Celular/metabolismo , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Endodermo/metabolismo , Mamíferos , Ratones , Transporte de Proteínas
7.
Cell Stem Cell ; 28(6): 1016-1022.e4, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-33957081

RESUMEN

Human naive pluripotent cells can differentiate into extraembryonic trophectoderm and hypoblast. Here we describe a human embryo model (blastoid) generated by self-organization. Brief induction of trophectoderm leads to formation of blastocyst-like structures within 3 days. Blastoids are composed of three tissue layers displaying exclusive lineage markers, mimicking the natural blastocyst. Single-cell transcriptome analyses confirm segregation of trophectoderm, hypoblast, and epiblast with high fidelity to the human embryo. This versatile and scalable system provides a robust experimental model for human embryo research.


Asunto(s)
Blastocisto , Embrión de Mamíferos , Diferenciación Celular , Linaje de la Célula , Estratos Germinativos , Humanos , Células Madre
8.
Cell Stem Cell ; 28(6): 1040-1056.e6, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-33831366

RESUMEN

Classic embryological experiments have established that the early mouse embryo develops via sequential lineage bifurcations. The first segregated lineage is the trophectoderm, essential for blastocyst formation. Mouse naive epiblast and derivative embryonic stem cells are restricted accordingly from producing trophectoderm. Here we show, in contrast, that human naive embryonic stem cells readily make blastocyst trophectoderm and descendant trophoblast cell types. Trophectoderm was induced rapidly and efficiently by inhibition of ERK/mitogen-activated protein kinase (MAPK) and Nodal signaling. Transcriptome comparison with the human embryo substantiated direct formation of trophectoderm with subsequent differentiation into syncytiotrophoblast, cytotrophoblast, and downstream trophoblast stem cells. During pluripotency progression lineage potential switches from trophectoderm to amnion. Live-cell tracking revealed that epiblast cells in the human blastocyst are also able to produce trophectoderm. Thus, the paradigm of developmental specification coupled to lineage restriction does not apply to humans. Instead, epiblast plasticity and the potential for blastocyst regeneration are retained until implantation.


Asunto(s)
Blastocisto , Estratos Germinativos , Animales , Diferenciación Celular , Linaje de la Célula , Desarrollo Embrionario , Células Madre Embrionarias , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones
9.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33452132

RESUMEN

OCT4 is a fundamental component of the molecular circuitry governing pluripotency in vivo and in vitro. To determine how OCT4 establishes and protects the pluripotent lineage in the embryo, we used comparative single-cell transcriptomics and quantitative immunofluorescence on control and OCT4 null blastocyst inner cell masses at two developmental stages. Surprisingly, activation of most pluripotency-associated transcription factors in the early mouse embryo occurs independently of OCT4, with the exception of the JAK/STAT signaling machinery. Concurrently, OCT4 null inner cell masses ectopically activate a subset of trophectoderm-associated genes. Inspection of metabolic pathways implicates the regulation of rate-limiting glycolytic enzymes by OCT4, consistent with a role in sustaining glycolysis. Furthermore, up-regulation of the lysosomal pathway was specifically detected in OCT4 null embryos. This finding implicates a requirement for OCT4 in the production of normal trophectoderm. Collectively, our findings uncover regulation of cellular metabolism and biophysical properties as mechanisms by which OCT4 instructs pluripotency.


Asunto(s)
Linaje de la Célula/genética , Desarrollo Embrionario/inmunología , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor de Transcripción STAT3/genética , Animales , Masa Celular Interna del Blastocisto/metabolismo , Embrión de Mamíferos , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica/genética , Glucólisis/genética , Ratones , Células Madre Pluripotentes/metabolismo , Transducción de Señal/genética , Análisis de la Célula Individual
10.
Cell Stem Cell ; 28(2): 273-284.e6, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33217323

RESUMEN

Cell fate transitions are frequently accompanied by changes in cell shape and mechanics. However, how cellular mechanics affects the instructive signaling pathways controlling cell fate is poorly understood. To probe the interplay between shape, mechanics, and fate, we use mouse embryonic stem cells (ESCs), which change shape as they undergo early differentiation. We find that shape change is regulated by a ß-catenin-mediated decrease in RhoA activity and subsequent decrease in the plasma membrane tension. Strikingly, preventing a decrease in membrane tension results in early differentiation defects in ESCs and gastruloids. Decreased membrane tension facilitates the endocytosis of FGF signaling components, which activate ERK signaling and direct the exit from the ESC state. Increasing Rab5a-facilitated endocytosis rescues defective early differentiation. Thus, we show that a mechanically triggered increase in endocytosis regulates early differentiation. Our findings are of fundamental importance for understanding how cell mechanics regulates biochemical signaling and therefore cell fate.


Asunto(s)
Células Madre Embrionarias , Células Madre Embrionarias de Ratones , Animales , Diferenciación Celular , Endocitosis , Ratones , Transducción de Señal
11.
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
12.
Stem Cell Reports ; 11(4): 988-997, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30245211

RESUMEN

In the case of organ transplantation accompanied by vascular anastomosis, major histocompatibility complex mismatched vascular endothelial cells become a target for graft rejection. Production of a rejection-free, transplantable organ, therefore, requires simultaneous generation of vascular endothelial cells within the organ. To generate pluripotent stem cell (PSC)-derived vascular endothelial cells, we performed blastocyst complementation with a vascular endothelial growth factor receptor-2 homozygous mutant blastocyst. This mutation is embryonic lethal at embryonic (E) day 8.5-9.5 due to an early defect in endothelial and hematopoietic cells. The Flk-1 homozygous knockout chimeric mice survived to adulthood for over 1 year without any abnormality, and all vascular endothelial cells and hematopoietic cells were derived from the injected PSCs. This approach could be used in conjunction with other gene knockouts which induce organ deficiency to produce a rejection-free, transplantable organ in which all the organ's cells and vasculature are PSC derived.


Asunto(s)
Blastocisto/citología , Células Endoteliales/citología , Células Madre Hematopoyéticas/citología , Envejecimiento/metabolismo , Animales , Blastocisto/metabolismo , Quimera , Células Endoteliales/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Fisiológica , Pericitos/citología , Pericitos/metabolismo , Fenotipo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
13.
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
14.
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
15.
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
16.
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.

17.
Methods Mol Biol ; 1357: 295-310, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-25697415

RESUMEN

Stem cells have the unique properties of self-renewal and multipotency (producing progeny belonging to two or more lineages). Induced pluripotent stem (iPS) cells can be generated from somatic cells by simultaneous expression of pluripotent factors (Oct3/4, Klf4, Sox2, and c-Myc). They share the same properties as embryonic stem (ES) cells and can differentiate into several tissue cells, i.e., neurons, hematopoietic cells, and liver cells. Therefore, iPS cells are suitable candidate cells for regenerative medicine and analyses of disease mechanisms.The liver is the major organ that regulates a multitude of metabolic functions. Hepatocytes are the major cell type populating the liver parenchyma and express several metabolic enzymes that are necessary for liver functions. Although hepatocytes are essential for maintaining homeostasis, it is difficult to alter artificial and transplanted cells because of their multifunctionality, donor shortage, and immunorejection risk. During liver development, hepatic progenitor cells in the fetal liver differentiate into both mature hepatocytes and cholangiocytes. As hepatic progenitor cells have bipotency and high proliferation ability, they could present a potential source for generating transplantable cells or as a liver study model. Here we describe the induction and purification of hepatic progenitor cells derived from human iPS cells. These cells can proliferate for a long term under suitable culture conditions.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Hepatocitos/citología , Células Madre Pluripotentes Inducidas/citología , Animales , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Reprogramación Celular , Fibroblastos/citología , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Factor 4 Similar a Kruppel , Ratones , Ratones Endogámicos ICR , Mitomicina/farmacología , Piel/citología
18.
Sci Rep ; 5: 14989, 2015 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-26456005

RESUMEN

Hepatic stem/progenitor cells, hepatoblasts, have a high proliferative ability and can differentiate into mature hepatocytes and cholangiocytes. Therefore, these cells are considered to be useful for regenerative medicine and drug screening for liver diseases. However, it is problem that in vitro maturation of hepatoblasts is insufficient in the present culture system. In this study, a novel regulator to induce hepatic differentiation was identified and the molecular function of this factor was examined in embryonic day 13 hepatoblast culture with maturation factor, oncostatin M and extracellular matrices. Overexpression of the basic helix-loop-helix type transcription factor, Mist1, induced expression of mature hepatocytic markers such as carbamoyl-phosphate synthetase1 and several cytochrome P450 (CYP) genes in this culture system. In contrast, Mist1 suppressed expression of cholangiocytic markers such as Sox9, Sox17, Ck19, and Grhl2. CYP3A metabolic activity was significantly induced by Mist1 in this hepatoblast culture. In addition, Mist1 induced liver-enriched transcription factors, CCAAT/enhancer-binding protein α and Hepatocyte nuclear factor 1α, which are known to be involved in liver functions. These results suggest that Mist1 partially induces mature hepatocytic expression and function accompanied by the down-regulation of cholangiocytic markers.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Hepatocitos/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Transcripción Genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Carbamoil-Fosfato Sintasa (Amoniaco)/genética , Carbamoil-Fosfato Sintasa (Amoniaco)/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citocromo P-450 CYP3A , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Embrión de Mamíferos , Feto , Regulación de la Expresión Génica , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Queratina-19/genética , Queratina-19/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/efectos de los fármacos , Oncostatina M/farmacología , Cultivo Primario de Células , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Stem Cells Dev ; 24(21): 2525-35, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26181762

RESUMEN

Hepatoblasts are somatic progenitor cells in the fetal liver, which retain a high proliferative capacity and differentiate into both hepatocytes and cholangiocytes in vivo. Although efficient expansion of hepatoblasts in vitro has been difficult without genetic modification, we have previously demonstrated that the interaction with mesenchymal cells is important for expansion of hepatoblasts in vitro. In this study, we show cell signaling pathways regulating the long-term proliferative ability of hepatoblasts. Individual primary hepatoblasts derived from mouse fetal livers formed large colonies when cocultured with mesenchymal feeder cells; however, secondary colony formation was unsuccessful, indicating that in vitro culture could induce short-term, but not long-term, proliferation. When the MEK inhibitor, PD0325901, was added to these cultures, hepatoblasts formed large colonies containing many Ki-67-positive cells. Expression of p16/19(cdkn2a), a cyclin-dependent kinase inhibitor, was induced after 3-6 days culture of hepatoblasts, whereas PD0325901 significantly suppressed this expression. Consistent with these observations, fetal hepatoblasts derived from p16/19(cdkn2a) knockout mice showed long-term proliferation without PD0325901, suggesting that MEK activity induced cell cycle arrest through accumulation of p16/19(cdkn2a). In transplantation assays, we could demonstrate that in vitro expanded hepatoblasts could proliferate and differentiate into hepatocytic and cholangiocytic cells in injured livers. It should also be noted that ERK in primary hepatoblasts was not highly activated during fetal liver development. Collectively, all these findings suggest that the MEK/ERK-independent pathway in the fetal liver is involved in hepatoblast proliferation to avoid accumulation of cyclin-dependent kinase inhibitor.


Asunto(s)
Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Feto/citología , Regulación del Desarrollo de la Expresión Génica/fisiología , Hepatocitos/citología , Sistema de Señalización de MAP Quinasas/fisiología , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Células Nutrientes/metabolismo , Hígado/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Transgénicos
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...