Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 15: 1375177, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38650946

RESUMEN

Human allogeneic pancreatic islet transplantation is a life-changing treatment for patients with severe Type 1 Diabetes (T1D) who suffer from hypoglycemia unawareness and high risk of severe hypoglycemia. However, intensive immunosuppression is required to prevent immune rejection of the graft, that may in turn lead to undesirable side effects such as toxicity to the islet cells, kidney toxicity, occurrence of opportunistic infections, and malignancies. The shortage of cadaveric human islet donors further limits islet transplantation as a treatment option for widespread adoption. Alternatively, porcine islets have been considered as another source of insulin-secreting cells for transplantation in T1D patients, though xeno-transplants raise concerns over the risk of endogenous retrovirus transmission and immunological incompatibility. As a result, technological advancements have been made to protect transplanted islets from immune rejection and inflammation, ideally in the absence of chronic immunosuppression, to improve the outcomes and accessibility of allogeneic islet cell replacement therapies. These include the use of microencapsulation or macroencapsulation devices designed to provide an immunoprotective environment using a cell-impermeable layer, preventing immune cell attack of the transplanted cells. Other up and coming advancements are based on the use of stem cells as the starting source material for generating islet cells 'on-demand'. These starting stem cell sources include human induced pluripotent stem cells (hiPSCs) that have been genetically engineered to avoid the host immune response, curated HLA-selected donor hiPSCs that can be matched with recipients within a given population, and multipotent stem cells with natural immune privilege properties. These strategies are developed to provide an immune-evasive cell resource for allogeneic cell therapy. This review will summarize the immunological challenges facing islet transplantation and highlight recent bio-engineering and cell-based approaches aimed at avoiding immune rejection, to improve the accessibility of islet cell therapy and enhance treatment outcomes. Better understanding of the different approaches and their limitations can guide future research endeavors towards developing more comprehensive and targeted strategies for creating a more tolerogenic microenvironment, and improve the effectiveness and sustainability of islet transplantation to benefit more patients.


Asunto(s)
Diabetes Mellitus Tipo 1 , Rechazo de Injerto , Trasplante de Islotes Pancreáticos , Trasplante de Islotes Pancreáticos/métodos , Humanos , Animales , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/terapia , Rechazo de Injerto/inmunología , Rechazo de Injerto/prevención & control , Ingeniería Biomédica/métodos , Islotes Pancreáticos/inmunología
2.
Methods Mol Biol ; 2429: 215-232, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35507164

RESUMEN

Chromatin immunoprecipitation (ChIP) is a technique that has been widely used to interrogate DNA-protein interactions in cells. In recent years, human pluripotent stem cell (hPSC)-derived 3D organoids have emerged as a powerful model to understand human development and diseases. Performing ChIP in hPSC-derived 3D organoids is a useful approach to dissect the roles of transcription factors or co-factors and to understand the epigenetic landscape in human development and diseases. However, performing ChIP in 3D organoids is more challenging than monolayer cultures, and an optimized protocol is needed for interpretable data. Hence, in this chapter, we describe in detail a protocol for performing ChIP in hPSC-derived islet-like cells as an example, from organoid harvest to ChIP-qPCR data analysis. This chapter also highlights potential pitfalls and provides recommendations for troubleshooting.


Asunto(s)
Organoides , Células Madre Pluripotentes , Diferenciación Celular , Inmunoprecipitación de Cromatina , ADN , Humanos
3.
Nat Commun ; 12(1): 3133, 2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34035238

RESUMEN

Heterozygous HNF1A gene mutations can cause maturity onset diabetes of the young 3 (MODY3), characterized by insulin secretion defects. However, specific mechanisms of MODY3 in humans remain unclear due to lack of access to diseased human pancreatic cells. Here, we utilize MODY3 patient-derived human induced pluripotent stem cells (hiPSCs) to study the effect(s) of a causal HNF1A+/H126D mutation on pancreatic function. Molecular dynamics simulations predict that the H126D mutation could compromise DNA binding and gene target transcription. Genome-wide RNA-Seq and ChIP-Seq analyses on MODY3 hiPSC-derived endocrine progenitors reveal numerous HNF1A gene targets affected by the mutation. We find decreased glucose transporter GLUT2 expression, which is associated with reduced glucose uptake and ATP production in the MODY3 hiPSC-derived ß-like cells. Overall, our findings reveal the importance of HNF1A in regulating GLUT2 and several genes involved in insulin secretion that can account for the insulin secretory defect clinically observed in MODY3 patients.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Transportador de Glucosa de Tipo 2/genética , Glucosa/metabolismo , Factor Nuclear 1-alfa del Hepatocito/genética , Secreción de Insulina/genética , Células Secretoras de Insulina/metabolismo , Mutación , Células Cultivadas , Secuenciación de Inmunoprecipitación de Cromatina/métodos , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Transportador de Glucosa de Tipo 2/metabolismo , Factor Nuclear 1-alfa del Hepatocito/química , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células Secretoras de Insulina/citología , Masculino , Simulación de Dinámica Molecular , Linaje , Dominios Proteicos
4.
STAR Protoc ; 2(2): 100471, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-33997805

RESUMEN

This protocol describes the detailed procedures for utilizing human pluripotent stem cells (hPSCs) for pancreatic progenitor and hepatic differentiation, followed by the application of hPSC-derived cells in a luciferase reporter-based assay to study gene regulation. The generated hPSC-derived cells have been shown to achieve morphologies and gene expression profiles specific to their differentiated cell types, and subsequent luciferase assay has been shown to effectively elucidate the role of disease-relevant gene variants. Therefore, this protocol provides a valuable approach for pancreatic and liver disease modeling. For complete details on the use and execution of this protocol, please refer to Ng et al. (2019).


Asunto(s)
Diferenciación Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Hígado/metabolismo , Células Madre Pluripotentes/metabolismo , Células Cultivadas , Humanos , Hígado/citología , Células Madre Pluripotentes/citología
5.
Vitam Horm ; 116: 193-233, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33752818

RESUMEN

There is now a sizeable number of single cell transcriptomics studies performed on human and rodent pancreatic islets that have shed light on the unique gene signatures and level of heterogeneity within each individual islet cell type. Following closely from these studies, there is also rapidly-growing activity on characterizing islet-like cells derived from in vitro differentiation of human pluripotent stem cells (hPSCs) at the single cell level. The overall consensus across the studies so far suggests that the first few stages of differentiation are largely uniform, whereas during pancreatic endocrine commitment, cell trajectories start to diverge, resulting in multiple end-stage pancreatic cells that include progenitor-like, endocrine and non-endocrine cells. Comprehensive transcriptional profiling is important for understanding how and why islet cells, especially the insulin-secreting beta cells, exist in subpopulations that differ in maturity, proliferation rate, sensitivity to stress, and insulin secretion function. For hPSC-derived beta cells to be used confidently for cell therapy, optimal differentiation and thorough characterization is required. The key questions to address are-What is the trajectory of differentiation? Is heterogeneity a natural occurrence or is it a consequence of imperfect differentiation protocols? Can lessons be drawn from the extensive single cell transcriptomic data to help guide maturation of beta cells in vitro? This book chapter seeks to address some of these questions, and facilitate ongoing efforts in improving the beta cell differentiation pipeline or enriching for desired beta cell populations following differentiation, to make way for better mechanistic studies and future clinical translation.


Asunto(s)
Células Secretoras de Insulina , Islotes Pancreáticos , Células Neuroendocrinas , Células Madre Pluripotentes , Diferenciación Celular , Humanos , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo
6.
Cell Death Dis ; 11(5): 378, 2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32424151

RESUMEN

The differentiation of human pluripotent stem cells into pancreatic cells involves cellular proliferation and apoptosis during cell fate transitions. However, their implications for establishing cellular identity are unclear. Here, we profiled the expression of BCL-2 family of proteins during pancreatic specification and observed an upregulation of BCL-xL, downregulation of BAK and corresponding downregulation of cleaved CASP3 representative of apoptosis. Experimental inhibition of BCL-xL reciprocally increased apoptosis and resulted in a decreased gene expression of pancreatic markers despite a compensatory increase in anti-apoptotic protein BCL-2. RNA-Seq analyses then revealed a downregulation of multiple metabolic genes upon inhibition of BCL-xL. Follow-up bioenergetics assays revealed broad downregulation of both glycolysis and oxidative phosphorylation when BCL-xL was inhibited. Early perturbation of BCL-xL during pancreatic specification also had subsequent detrimental effects on the formation of INS+ pancreatic beta-like cells. In conclusion, the more differentiated pancreatic progenitors are dependent on anti-apoptotic BCL-xL for survival, whereas the less differentiated pancreatic progenitors that survived after WEHI-539 treatment would exhibit a more immature phenotype. Therefore, modulation of the expression level of BCL-xL can potentially increase the survival and robustness of pancreatic progenitors that ultimately define human pancreatic beta cell mass and function.


Asunto(s)
Apoptosis/fisiología , Células Madre Pluripotentes/citología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína bcl-X/metabolismo , Caspasa 3/metabolismo , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Humanos , Neoplasias Pancreáticas/metabolismo , Células Madre Pluripotentes/metabolismo
7.
iScience ; 16: 192-205, 2019 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-31195238

RESUMEN

Maturity-onset diabetes of the young 1 (MODY1) is a monogenic diabetes condition caused by heterozygous HNF4A mutations. We investigate how HNF4A haploinsufficiency from a MODY1/HNF4A mutation influences the development of foregut-derived liver and pancreatic cells through differentiation of human induced pluripotent stem cells from a MODY1 family down the foregut lineage. In MODY1-derived hepatopancreatic progenitors, which expressed reduced HNF4A levels and mislocalized HNF4A, foregut genes were downregulated, whereas hindgut-specifying HOX genes were upregulated. MODY1-derived hepatocyte-like cells were found to exhibit altered morphology. Hepatic and ß cell gene signatures were also perturbed in MODY1-derived hepatocyte-like and ß-like cells, respectively. As mutant HNF4A (p.Ile271fs) did not undergo complete nonsense-mediated decay or exert dominant negativity, HNF4A-mediated loss of function is likely due to impaired transcriptional activation of target genes. Our results suggest that in MODY1, liver and pancreas development is perturbed early on, contributing to altered hepatic proteins and ß cell defects in patients.

8.
J Hepatol ; 68(5): 1033-1048, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29175243

RESUMEN

The hepatocyte nuclear factors (HNFs) namely HNF1α/ß, FOXA1/2/3, HNF4α/γ and ONECUT1/2 are expressed in a variety of tissues and organs, including the liver, pancreas and kidney. The spatial and temporal manner of HNF expression regulates embryonic development and subsequently the development of multiple tissues during adulthood. Though the HNFs were initially identified individually based on their roles in the liver, numerous studies have now revealed that the HNFs cross-regulate one another and exhibit synergistic relationships in the regulation of tissue development and function. The complex HNF transcriptional regulatory networks have largely been elucidated in rodent models, but less so in human biological systems. Several heterozygous mutations in these HNFs were found to cause diseases in humans but not in rodents, suggesting clear species-specific differences in mutational mechanisms that remain to be uncovered. In this review, we compare and contrast the expression patterns of the HNFs, the HNF cross-regulatory networks and how these liver-enriched transcription factors serve multiple functions in the liver and beyond, extending our focus to the pancreas and kidney. We also summarise the insights gained from both human and rodent studies of mutations in several HNFs that are known to lead to different disease conditions.


Asunto(s)
Factores Nucleares del Hepatocito/metabolismo , Hígado/metabolismo , Animales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Factores Nucleares del Hepatocito/química , Factores Nucleares del Hepatocito/genética , Humanos , Riñón/metabolismo , Hígado/crecimiento & desarrollo , Redes y Vías Metabólicas , Mutación , Páncreas/metabolismo , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...