Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Intervalo de año de publicación
1.
JHEP Rep ; 4(4): 100446, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35284810

RESUMEN

Background & Aims: The truncating mutations in tight junction protein 2 (TJP2) cause progressive cholestasis, liver failure, and hepatocyte carcinogenesis. Due to the lack of effective model systems, there are no targeted medications for the liver pathology with TJP2 deficiency. We leveraged the technologies of patient-specific induced pluripotent stem cells (iPSC) and CRISPR genome-editing, and we aim to establish a disease model which recapitulates phenotypes of patients with TJP2 deficiency. Methods: We differentiated iPSC to hepatocyte-like cells (iHep) on the Transwell membrane in a polarized monolayer. Immunofluorescent staining of polarity markers was detected by a confocal microscope. The epithelial barrier function and bile acid transport of bile canaliculi were quantified between the two chambers of Transwell. The morphology of bile canaliculi was measured in iHep cultured in the Matrigel sandwich system using a fluorescent probe and live-confocal imaging. Results: The iHep differentiated from iPSC with TJP2 mutations exhibited intracellular inclusions of disrupted apical membrane structures, distorted canalicular networks, altered distribution of apical and basolateral markers/transporters. The directional bile acid transport of bile canaliculi was compromised in the mutant hepatocytes, resembling the disease phenotypes observed in the liver of patients. Conclusions: Our iPSC-derived in vitro hepatocyte system revealed canalicular membrane disruption in TJP2 deficient hepatocytes and demonstrated the ability to model cholestatic disease with TJP2 deficiency to serve as a platform for further pathophysiologic study and drug discovery. Lay summary: We investigated a genetic liver disease, progressive familial intrahepatic cholestasis (PFIC), which causes severe liver disease in newborns and infants due to a lack of gene called TJP2. By using cutting-edge stem cell technology and genome editing methods, we established a novel disease modeling system in cell culture experiments. Our experiments demonstrated that the lack of TJP2 induced abnormal cell polarity and disrupted bile acid transport. These findings will lead to the subsequent investigation to further understand disease mechanisms and develop an effective treatment.

2.
Nat Commun ; 10(1): 3350, 2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31350390

RESUMEN

The liver parenchyma is composed of hepatocytes and bile duct epithelial cells (BECs). Controversy exists regarding the cellular origin of human liver parenchymal tissue generation during embryonic development, homeostasis or repair. Here we report the existence of a hepatobiliary hybrid progenitor (HHyP) population in human foetal liver using single-cell RNA sequencing. HHyPs are anatomically restricted to the ductal plate of foetal liver and maintain a transcriptional profile distinct from foetal hepatocytes, mature hepatocytes and mature BECs. In addition, molecular heterogeneity within the EpCAM+ population of freshly isolated foetal and adult human liver identifies diverse gene expression signatures of hepatic and biliary lineage potential. Finally, we FACS isolate foetal HHyPs and confirm their hybrid progenitor phenotype in vivo. Our study suggests that hepatobiliary progenitor cells previously identified in mice also exist in humans, and can be distinguished from other parenchymal populations, including mature BECs, by distinct gene expression profiles.


Asunto(s)
Hígado/citología , Transcripción Genética , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Molécula de Adhesión Celular Epitelial/genética , Molécula de Adhesión Celular Epitelial/metabolismo , Feto/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Análisis de la Célula Individual , Células Madre/citología , Células Madre/metabolismo
3.
Stem Cells Transl Med ; 8(2): 124-137, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30456803

RESUMEN

Recent advancements in the production of hepatocytes from human pluripotent stem cells (hPSC-Heps) afford tremendous possibilities for treatment of patients with liver disease. Validated current good manufacturing practice (cGMP) lines are an essential prerequisite for such applications but have only recently been established. Whether such cGMP lines are capable of hepatic differentiation is not known. To address this knowledge gap, we examined the proficiency of three recently derived cGMP lines (two hiPSC and one hESC) to differentiate into hepatocytes and their suitability for therapy. hPSC-Heps generated using a chemically defined four-step hepatic differentiation protocol uniformly demonstrated highly reproducible phenotypes and functionality. Seeding into a 3D poly(ethylene glycol)-diacrylate fabricated inverted colloid crystal scaffold converted these immature progenitors into more advanced hepatic tissue structures. Hepatic constructs could also be successfully encapsulated into the immune-privileged material alginate and remained viable as well as functional upon transplantation into immune competent mice. This is the first report we are aware of demonstrating cGMP-compliant hPSCs can generate cells with advanced hepatic function potentially suitable for future therapeutic applications. Stem Cells Translational Medicine 2019;8:124&14.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/normas , Hepatocitos/citología , Células Madre Pluripotentes/citología , Animales , Técnicas de Cultivo de Célula/normas , Diferenciación Celular/fisiología , Línea Celular , Humanos , Hígado/citología , Ratones
4.
Biomaterials ; 182: 299-311, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30149262

RESUMEN

Generation of human organoids from induced pluripotent stem cells (iPSCs) offers exciting possibilities for developmental biology, disease modelling and cell therapy. Significant advances towards those goals have been hampered by dependence on animal derived matrices (e.g. Matrigel), immortalized cell lines and resultant structures that are difficult to control or scale. To address these challenges, we aimed to develop a fully defined liver organoid platform using inverted colloid crystal (ICC) whose 3-dimensional mechanical properties could be engineered to recapitulate the extracellular niche sensed by hepatic progenitors during human development. iPSC derived hepatic progenitors (IH) formed organoids most optimally in ICC scaffolds constructed with 140 µm diameter pores coated with type I collagen in a two-step process mimicking liver bud formation. The resultant organoids were closer to adult tissue, compared to 2D and 3D controls, with respect to morphology, gene expression, protein secretion, drug metabolism and viral infection and could integrate, vascularise and function following implantation into livers of immune-deficient mice. Preliminary interrogation of the underpinning mechanisms highlighted the importance of TGFß and hedgehog signalling pathways. The combination of functional relevance with tuneable mechanical properties leads us to propose this bioengineered platform to be ideally suited for a range of future mechanistic and clinical organoid related applications.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Hígado/citología , Organoides/citología , Polietilenglicoles/química , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Materiales Biocompatibles/química , Células Cultivadas , Cristalización , Humanos , Células Madre Pluripotentes Inducidas/metabolismo
6.
J Hepatol ; 58(3): 625-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23131523

RESUMEN

Advances in basic hepatology have been constrained for many years by the inability to culture primary hepatocytes in vitro, until just over five years ago when the scientific playing field was changed beyond recognition with the demonstration that human skin fibroblasts could be reprogrammed to resemble embryonic cells. The reprogrammed cells, known as induced pluripotent stem cells (iPSCs), were then shown to have the capacity to re-differentiate into almost any human cell type, including hepatocytes. The unlimited number and isogenic nature of the cells that can be generated from tiny fragments of tissue have massive implications for the study of human liver diseases in vitro. Of more immediate clinical importance were recent data demonstrating precision gene therapy on patient specific iPSCs, which opens up the real and exciting possibility of autologous hepatocyte transplantation as a substitute for allogeneic whole liver transplantation, which has been an effective approach to end-stage liver disease, but one that has now been outstripped by demand. In this review, we describe the historical development, current technology and potential clinical applications of induced pluripotency, concluding with a perspective on possible future directions in this dynamic field.


Asunto(s)
Hepatocitos/trasplante , Células Madre Pluripotentes Inducidas/citología , Premio Nobel , Animales , Diferenciación Celular , Enfermedad Hepática en Estado Terminal/cirugía , Humanos , Medicina Regenerativa
7.
J Proteome Res ; 11(8): 4052-64, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22694338

RESUMEN

Activation of hepatic stellate cells (HSCs) and subsequent uncontrolled accumulation of altered extracellular matrix (ECM) underpin liver fibrosis, a wound healing response to chronic injury, which can lead to organ failure and death. We sought to catalogue the components of fibrotic liver ECM to obtain insights into disease etiology and aid identification of new biomarkers. Cell-derived ECM was isolated from the HSC line LX-2, an in vitro model of liver fibrosis, and compared to ECM from human foreskin fibroblasts (HFFs) as a control. Mass spectrometry analyses of cell-derived ECMs identified, with ≥99% confidence, 61 structural ECM or secreted proteins (48 and 31 proteins for LX-2 and HFF, respectively). Gene ontology enrichment analysis confirmed the enrichment of ECM proteins, and hierarchical clustering coupled with protein-protein interaction network analysis revealed a subset of proteins enriched to fibrotic ECM, highlighting the existence of cell type-specific ECM niches. Thirty-six proteins were enriched to LX-2 ECM as compared to HFF ECM, of which Wnt-5a and CYR61 were validated by immunohistochemistry in human and murine fibrotic liver tissue. Future studies will determine if these and other components may play a role in the etiology of hepatic fibrosis, serve as novel disease biomarkers, or open up new avenues for drug discovery.


Asunto(s)
Proteína 61 Rica en Cisteína/metabolismo , Matriz Extracelular/metabolismo , Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática/metabolismo , Proteoma/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Wnt/metabolismo , Animales , Línea Celular , Análisis por Conglomerados , Proteína 61 Rica en Cisteína/aislamiento & purificación , Fibroblastos/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Proteoma/aislamiento & purificación , Proteómica , Proteínas Proto-Oncogénicas/aislamiento & purificación , Proteínas Wnt/aislamiento & purificación , Proteína Wnt-5a
8.
Stem Cell Res Ther ; 3(1): 4, 2012 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-22340671

RESUMEN

Human induced pluripotent stem cells offer the possibility of generating unlimited quantities of cells for autologous transplantation. By correcting the genetic defect underlying Z-allele α1-antitrypsin deficiency, we recently provided the first proof of principle for application of human induced pluripotent stem cells in the treatment of inherited genetic disorders. Several important safety concerns will need to be addressed before this can be translated into clinical practice.


Asunto(s)
Trasplante de Células Madre , Células Madre/citología , Deficiencia de alfa 1-Antitripsina/terapia , Alelos , Humanos , Mutación Puntual , Trasplante Autólogo , Deficiencia de alfa 1-Antitripsina/genética
9.
Methods Enzymol ; 501: 421-66, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22078544

RESUMEN

The serpinopathies result from point mutations in members of the serine protease inhibitor or serpin superfamily. They are characterized by the formation of ordered polymers that are retained within the cell of synthesis. This causes disease by a "toxic gain of function" from the accumulated protein and a "loss of function" as a result of the deficiency of inhibitors that control important proteolytic cascades. The serpinopathies are exemplified by the Z (Glu342Lys) mutant of α1-antitrypsin that results in the retention of ordered polymers within the endoplasmic reticulum of hepatocytes. These polymers form the intracellular inclusions that are associated with neonatal hepatitis, cirrhosis, and hepatocellular carcinoma. A second example results from mutations in the neurone-specific serpin-neuroserpin to form ordered polymers that are retained as inclusions within subcortical neurones as Collins' bodies. These inclusions underlie the autosomal dominant dementia familial encephalopathy with neuroserpin inclusion bodies or FENIB. There are different pathways to polymer formation in vitro but not all form polymers that are relevant in vivo. It is therefore essential that protein-based structural studies are interpreted in the context of human samples and cell and animal models of disease. We describe here the biochemical techniques, monoclonal antibodies, cell biology, animal models, and stem cell technology that are useful to characterize the serpin polymers that form in vivo.


Asunto(s)
Biofisica/métodos , Epilepsias Mioclónicas/metabolismo , Trastornos Heredodegenerativos del Sistema Nervioso/metabolismo , Procesamiento de Imagen Asistido por Computador/métodos , Pulmón/metabolismo , Neuropéptidos/metabolismo , Mutación Puntual , Serpinas/metabolismo , alfa 1-Antitripsina/metabolismo , Animales , Técnicas de Cultivo de Célula , Línea Celular , Epilepsias Mioclónicas/genética , Epilepsias Mioclónicas/patología , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Trastornos Heredodegenerativos del Sistema Nervioso/patología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Pulmón/patología , Ratones , Ratones Transgénicos , Microscopía Electrónica , Neuropéptidos/química , Neuropéptidos/genética , Neutrófilos/citología , Neutrófilos/metabolismo , Fragmentos de Péptidos , Polimerizacion , Unión Proteica , Conformación Proteica , Proteolisis , Serpinas/química , Serpinas/genética , Transfección , alfa 1-Antitripsina/química , alfa 1-Antitripsina/genética , Neuroserpina
10.
Nature ; 478(7369): 391-4, 2011 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-21993621

RESUMEN

Human induced pluripotent stem cells (iPSCs) represent a unique opportunity for regenerative medicine because they offer the prospect of generating unlimited quantities of cells for autologous transplantation, with potential application in treatments for a broad range of disorders. However, the use of human iPSCs in the context of genetically inherited human disease will require the correction of disease-causing mutations in a manner that is fully compatible with clinical applications. The methods currently available, such as homologous recombination, lack the necessary efficiency and also leave residual sequences in the targeted genome. Therefore, the development of new approaches to edit the mammalian genome is a prerequisite to delivering the clinical promise of human iPSCs. Here we show that a combination of zinc finger nucleases (ZFNs) and piggyBac technology in human iPSCs can achieve biallelic correction of a point mutation (Glu342Lys) in the α(1)-antitrypsin (A1AT, also known as SERPINA1) gene that is responsible for α(1)-antitrypsin deficiency. Genetic correction of human iPSCs restored the structure and function of A1AT in subsequently derived liver cells in vitro and in vivo. This approach is significantly more efficient than any other gene-targeting technology that is currently available and crucially prevents contamination of the host genome with residual non-human sequences. Our results provide the first proof of principle, to our knowledge, for the potential of combining human iPSCs with genetic correction to generate clinically relevant cells for autologous cell-based therapies.


Asunto(s)
Células Madre Pluripotentes Inducidas/fisiología , Reparación del Gen Blanco , Deficiencia de alfa 1-Antitripsina/genética , alfa 1-Antitripsina/genética , Animales , Línea Celular , Elementos Transponibles de ADN/genética , Hepatocitos/metabolismo , Hepatocitos/trasplante , Humanos , Hígado/citología , Ratones , Albúmina Sérica/genética , Albúmina Sérica/metabolismo , Albúmina Sérica Humana , Factores de Tiempo , alfa 1-Antitripsina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA