Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Front Immunol ; 15: 1375177, 2024.
Article in English | MEDLINE | ID: mdl-38650946

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Type 1 , Graft Rejection , Islets of Langerhans Transplantation , Islets of Langerhans Transplantation/methods , Humans , Animals , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Graft Rejection/immunology , Graft Rejection/prevention & control , Biomedical Engineering/methods , Islets of Langerhans/immunology
2.
Elife ; 132024 Jan 02.
Article in English | MEDLINE | ID: mdl-38164941

ABSTRACT

Selection of the target site is an inherent question for any project aiming for directed transgene integration. Genomic safe harbour (GSH) loci have been proposed as safe sites in the human genome for transgene integration. Although several sites have been characterised for transgene integration in the literature, most of these do not meet criteria set out for a GSH and the limited set that do have not been characterised extensively. Here, we conducted a computational analysis using publicly available data to identify 25 unique putative GSH loci that reside in active chromosomal compartments. We validated stable transgene expression and minimal disruption of the native transcriptome in three GSH sites in vitro using human embryonic stem cells (hESCs) and their differentiated progeny. Furthermore, for easy targeted transgene expression, we have engineered constitutive landing pad expression constructs into the three validated GSH in hESCs.


Subject(s)
Genomics , Humans , Gene Expression , Transgenes , Cell Differentiation
3.
Nat Commun ; 14(1): 6119, 2023 09 30.
Article in English | MEDLINE | ID: mdl-37777536

ABSTRACT

The coding variant (p.Arg192His) in the transcription factor PAX4 is associated with an altered risk for type 2 diabetes (T2D) in East Asian populations. In mice, Pax4 is essential for beta cell formation but its role on human beta cell development and/or function is unknown. Participants carrying the PAX4 p.His192 allele exhibited decreased pancreatic beta cell function compared to homozygotes for the p.192Arg allele in a cross-sectional study in which we carried out an intravenous glucose tolerance test and an oral glucose tolerance test. In a pedigree of a patient with young onset diabetes, several members carry a newly identified p.Tyr186X allele. In the human beta cell model, EndoC-ßH1, PAX4 knockdown led to impaired insulin secretion, reduced total insulin content, and altered hormone gene expression. Deletion of PAX4 in human induced pluripotent stem cell (hiPSC)-derived islet-like cells resulted in derepression of alpha cell gene expression. In vitro differentiation of hiPSCs carrying PAX4 p.His192 and p.X186 risk alleles exhibited increased polyhormonal endocrine cell formation and reduced insulin content that can be reversed with gene correction. Together, we demonstrate the role of PAX4 in human endocrine cell development, beta cell function, and its contribution to T2D-risk.


Subject(s)
Diabetes Mellitus, Type 2 , Glucagon-Secreting Cells , Induced Pluripotent Stem Cells , Insulin-Secreting Cells , Humans , Mice , Animals , Homeodomain Proteins/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Cross-Sectional Studies , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Induced Pluripotent Stem Cells/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Glucagon-Secreting Cells/metabolism
4.
iScience ; 26(8): 107265, 2023 Aug 18.
Article in English | MEDLINE | ID: mdl-37502260

ABSTRACT

The differentiation of human pluripotent stem cells into the SOX17+ definitive endoderm (DE) germ layer is important for generating tissues for regenerative medicine. Multiple developmental and stem cell studies have demonstrated that Activin/Nodal signaling is the primary driver of definitive endoderm formation. Here, we uncover that the FGF2-FGFR-ERK1/2 signaling contributes to mesendoderm and SOX17+ DE formation. Without ERK1/2 signaling, the Activin/Nodal signaling is insufficient to drive mesendoderm and DE formation. Besides FGF2-FGFR-mediated signaling, IGF1R signaling possibly contributes to the ERK1/2 signaling for DE formation. We identified a temporal relationship between Activin/Nodal-SMAD2 and FGF2-FGFR-ERK1/2 signaling in which Activin/Nodal-SMAD2 participates in the initiation of mesendoderm and DE specification that is followed by increasing activity of FGF2-FGFR-ERK1/2 to facilitate and permit the successful generation of SOX17+ DE. Overall, besides the role of Activin/Nodal signaling for DE formation, our findings shed light on the contribution of ERK1/2 signaling for mesendoderm and DE formation.

5.
Handb Exp Pharmacol ; 281: 301-332, 2023.
Article in English | MEDLINE | ID: mdl-37306817

ABSTRACT

The long-standing goals in diabetes research are to improve ß-cell survival, functionality and increase ß-cell mass. Current strategies to manage diabetes progression are still not ideal for sustained maintenance of normoglycemia, thereby increasing demand for the development of novel drugs. Available pancreatic cell lines, cadaveric islets, and their culture methods and formats, either 2D or 3D, allow for multiple avenues of experimental design to address diverse aims in the research setting. More specifically, these pancreatic cells have been employed in toxicity testing, diabetes drug screens, and with careful curation, can be optimized for use in efficient high-throughput screenings (HTS). This has since spearheaded the understanding of disease progression and related mechanisms, as well as the discovery of potential drug candidates which could be the cornerstone for diabetes treatment. This book chapter will touch on the pros and cons of the most widely used pancreatic cells, including the more recent human pluripotent stem cell-derived pancreatic cells, and HTS strategies (cell models, design, readouts) that can be used for the purpose of toxicity testing and diabetes drug discovery.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Pluripotent Stem Cells , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/therapy , Drug Discovery , Cell Differentiation
6.
Cell Death Dis ; 14(5): 302, 2023 05 03.
Article in English | MEDLINE | ID: mdl-37137894

ABSTRACT

Renal defects in maturity onset diabetes of the young 3 (MODY3) patients and Hnf1a-/- mice suggest an involvement of HNF1A in kidney development and/or its function. Although numerous studies have leveraged on Hnf1α-/- mice to infer some transcriptional targets and function of HNF1A in mouse kidneys, species-specific differences obviate a straightforward extrapolation of findings to the human kidney. Additionally, genome-wide targets of HNF1A in human kidney cells have yet to be identified. Here, we leveraged on human in vitro kidney cell models to characterize the expression profile of HNF1A during renal differentiation and in adult kidney cells. We found HNF1A to be increasingly expressed during renal differentiation, with peak expression on day 28 in the proximal tubule cells. HNF1A ChIP-Sequencing (ChIP-Seq) performed on human pluripotent stem cell (hPSC)-derived kidney organoids identified its genome-wide putative targets. Together with a qPCR screen, we found HNF1A to activate the expression of SLC51B, CD24, and RNF186 genes. Importantly, HNF1A-depleted human renal proximal tubule epithelial cells (RPTECs) and MODY3 human induced pluripotent stem cell (hiPSC)-derived kidney organoids expressed lower levels of SLC51B. SLC51B-mediated estrone sulfate (E1S) uptake in proximal tubule cells was abrogated in these HNF1A-deficient cells. MODY3 patients also exhibit significantly higher excretion of urinary E1S. Overall, we report that SLC51B is a target of HNF1A responsible for E1S uptake in human proximal tubule cells. As E1S serves as the main storage form of nephroprotective estradiol in the human body, lowered E1S uptake and increased E1S excretion may reduce the availability of nephroprotective estradiol in the kidneys, contributing to the development of renal disease in MODY3 patients.


Subject(s)
Induced Pluripotent Stem Cells , Adult , Animals , Humans , Mice , Epithelial Cells/metabolism , Estradiol , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Induced Pluripotent Stem Cells/metabolism , Ubiquitin-Protein Ligases
7.
Acta Biomater ; 157: 49-66, 2023 02.
Article in English | MEDLINE | ID: mdl-36427686

ABSTRACT

Diabetes mellitus is a global disease requiring long-term treatment and monitoring. At present, pancreas or islet transplantation is the only reliable treatment for achieving stable euglycemia in Type I diabetes patients. However, the shortage of viable pancreata for transplantation limits the use of this therapy for the majority of patients. Organ decellularization and recellularization is emerging as a promising solution to overcome the shortage of viable organs for transplantation by providing a potential alternative source of donor organs. Several studies on decellularization and recellularization of rodent, porcine, and human pancreata have been performed, and show promise for generating usable decellularized pancreas scaffolds for subsequent recellularization and transplantation. In this state-of-the-art review, we provide an overview of the latest advances in pancreas decellularization, recellularization, and revascularization. We also discuss clinical considerations such as potential transplantation sites, donor source, and immune considerations. We conclude with an outlook on the remaining work that needs to be done in order to realize the goal of using this technology to create bioengineered pancreata for transplantation in diabetes patients. STATEMENT OF SIGNIFICANCE: Pancreas or islet transplantation is a means of providing insulin-independence in diabetes patients. However, due to the shortage of viable pancreata, whole-organ decellularization and recellularization is emerging as a promising solution to overcome organ shortage for transplantation. Several studies on decellularization and recellularization of rodent, porcine, and human pancreata have shown promise for generating usable decellularized pancreas scaffolds for subsequent recellularization and transplantation. In this state-of-the-art review, we highlight the latest advances in pancreas decellularization, recellularization, and revascularization. We also discuss clinical considerations such as potential transplantation sites, donor source, and immune considerations. We conclude with future work that needs to be done in order to realize clinical translation of bioengineered pancreata for transplantation in diabetes patients.


Subject(s)
Diabetes Mellitus, Type 1 , Tissue Engineering , Humans , Animals , Swine , Regenerative Medicine , Tissue Scaffolds , Pancreas , Extracellular Matrix
8.
Atherosclerosis ; 362: 11-22, 2022 12.
Article in English | MEDLINE | ID: mdl-36435092

ABSTRACT

BACKGROUND AND AIMS: Single nucleotide polymorphism rs6903956 has been identified as one of the genetic risk factors for coronary artery disease (CAD). However, rs6903956 lies in a non-coding locus on chromosome 6p24.1. We aim to interrogate the molecular basis of 6p24.1 containing rs6903956 risk alleles in endothelial disease biology. METHODS AND RESULTS: We generated induced pluripotent stem cells (iPSCs) from CAD patients (AA risk genotype at rs6903956) and non-CAD subjects (GG non-risk genotype at rs6903956). CRISPR-Cas9-based deletions (Δ63-89bp) on 6p24.1, including both rs6903956 and a short tandem repeat variant rs140361069 in linkage disequilibrium, were performed to generate isogenic iPSC-derived endothelial cells. Edited CAD endothelial cells, with removal of 'A' risk alleles, exhibited a global transcriptional downregulation of pathways relating to abnormal vascular physiology and activated endothelial processes. A CXC chemokine ligand on chromosome 10q11.21, CXCL12, was uncovered as a potential effector gene in CAD endothelial cells. Underlying this effect was the preferential inter-chromosomal interaction of 6p24.1 risk locus to a weak promoter of CXCL12, confirmed by chromatin conformation capture assays on our iPSC-derived endothelial cells. Functionally, risk genotypes AA/AG at rs6903956 were associated significantly with elevated levels of circulating damaged endothelial cells in CAD patients. Circulating endothelial cells isolated from patients with risk genotypes AA/AG were also found to have 10 folds higher CXCL12 transcript copies/cell than those with non-risk genotype GG. CONCLUSIONS: Our study reveals the trans-acting impact of 6p24.1 with another CAD locus on 10q11.21 and is associated with intensified endothelial injury.


Subject(s)
Coronary Artery Disease , Endothelial Cells , Humans , Coronary Artery Disease/genetics , Alleles , Genotype , Polymorphism, Single Nucleotide
9.
Methods Cell Biol ; 170: 127-146, 2022.
Article in English | MEDLINE | ID: mdl-35811096

ABSTRACT

Diabetes is a major healthcare burden globally, affecting over 463 million people today, according to the International Diabetes Federation. The most common types of diabetes are Type I diabetes (T1D) and Type II diabetes (T2D), characterized by hyperglycemia due to autoimmune destruction of ß cells (T1D) and ß cell dysfunction, usually on a background of insulin resistance (T2D). There is currently no cure for diabetes, and patients with T1D require lifelong insulin therapy. Additionally, while most cases of T2D can be managed by lifestyle and diet modifications, with or without antidiabetic drugs, severe cases of T2D may also require insulin therapy. The only means to restore stable euglycemia in these patients is now via whole pancreas or islet transplantation. However, this is limited by the scarcity of donors. In recent years, advances in human pluripotent stem cell (hPSC) technologies and pancreatic ß cell differentiation protocols have opened up new potential avenues for cell replacement therapies for diabetes. These advances have also created opportunities to use hPSC-derived ß-like cells for studies of disease mechanisms and drug discovery, which in turn have the potential to lead to better therapies for diabetes patients. Here, we describe the protocol used in our laboratory to generate ß-like cells from hPSCs to study the mechanisms underlying various types of diabetes.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Pluripotent Stem Cells , Cell Differentiation , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Humans , Insulin/metabolism , Pancreas
10.
Reprod Toxicol ; 112: 36-50, 2022 09.
Article in English | MEDLINE | ID: mdl-35697279

ABSTRACT

The advent of the technology to isolate or generate human pluripotent stem cells provided the potential to develop a wide range of human models that could enhance understanding of mechanisms underlying human development and disease. These systems are now beginning to mature and provide the basis for the development of in vitro assays suitable to understand the biological processes involved in the multi-organ systems of the human body, and will improve strategies for diagnosis, prevention, therapies and precision medicine. Induced pluripotent stem cell lines are prone to phenotypic and genotypic changes and donor/clone dependent variability, which means that it is important to identify the most appropriate characterization markers and quality control measures when sourcing new cell lines and assessing differentiated cell and tissue culture preparations for experimental work. This paper considers those core quality control measures for human pluripotent stem cell lines and evaluates the state of play in the development of key functional markers for their differentiated cell derivatives to promote assurance of reproducibility of scientific data derived from pluripotent stem cell-based systems.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Cell Culture Techniques , Cell Differentiation , Humans , Induced Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Reproducibility of Results
11.
Methods Mol Biol ; 2429: 215-232, 2022.
Article in English | MEDLINE | ID: mdl-35507164

ABSTRACT

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.


Subject(s)
Organoids , Pluripotent Stem Cells , Cell Differentiation , Chromatin Immunoprecipitation , DNA , Humans
12.
Front Digit Health ; 4: 845405, 2022.
Article in English | MEDLINE | ID: mdl-35585913

ABSTRACT

In the past one or two decades, countries across the world have successively implemented different precision medicine (PM) programs, and also cooperated to implement international PM programs. We are now in the era of PM. Singapore's National Precision Medicine (NPM) program, initiated in 2017, is now entering its second phase to generate a large genomic database for Asians. The National University of Singapore (NUS) also launched its own PM translational research program (TRP) in 2021, aimed at consolidating multidisciplinary expertise within the Yong Loo Lin School of Medicine to develop collaborative projects that can help to identify and validate novel therapeutic targets for the realization of PM. To achieve this, appropriate data collection, data processing, and results interpretation must be taken into consideration. There may be some difficulties during these processes, but with the improvement of relevant rules and the continuous development of omics-based technologies, we will be able to solve these problems, eventually achieving precise prediction, diagnosis, treatment, or even prevention of diseases.

13.
Cell Prolif ; 55(8): e13232, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35474596

ABSTRACT

The unlimited proliferative capacity of human pluripotent stem cells (hPSCs) fortifies it as one of the most attractive sources for cell therapy application in diabetes. In the past two decades, vast research efforts have been invested in developing strategies to differentiate hPSCs into clinically suitable insulin-producing endocrine cells or functional beta cells (ß cells). With the end goal being clinical translation, it is critical for hPSCs and insulin-producing ß cells to be derived, handled, stored, maintained and expanded with clinical compliance. This review focuses on the key processes and guidelines for clinical translation of human induced pluripotent stem cell (hiPSC)-derived ß cells for diabetes cell therapy. Here, we discuss the (1) key considerations of manufacturing clinical-grade hiPSCs, (2) scale-up and differentiation of clinical-grade hiPSCs into ß cells in clinically compliant conditions and (3) mandatory quality control and product release criteria necessitated by various regulatory bodies to approve the use of the cell-based products.


Subject(s)
Diabetes Mellitus , Induced Pluripotent Stem Cells , Insulins , Pluripotent Stem Cells , Cell Differentiation , Diabetes Mellitus/therapy , Humans
14.
Diabetes ; 71(3): 424-439, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34588186

ABSTRACT

The effects of imeglimin, a novel antidiabetes agent, on ß-cell function remain unclear. Here, we unveiled the impact of imeglimin on ß-cell survival. Treatment with imeglimin augmented mitochondrial function, enhanced insulin secretion, promoted ß-cell proliferation, and improved ß-cell survival in mouse islets. Imeglimin upregulated the expression of endoplasmic reticulum (ER)-related molecules, including Chop (Ddit3), Gadd34 (Ppp1r15a), Atf3, and Sdf2l1, and decreased eIF2α phosphorylation after treatment with thapsigargin and restored global protein synthesis in ß-cells under ER stress. Imeglimin failed to protect against ER stress-induced ß-cell apoptosis in CHOP-deficient islets or in the presence of GADD34 inhibitor. Treatment with imeglimin showed a significant decrease in the number of apoptotic ß-cells and increased ß-cell mass in Akita mice. Imeglimin also protected against ß-cell apoptosis in both human islets and human pluripotent stem cell-derived ß-like cells. Taken together, imeglimin modulates the ER homeostasis pathway, which results in the prevention of ß-cell apoptosis both in vitro and in vivo.


Subject(s)
Apoptosis/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Hypoglycemic Agents , Insulin-Secreting Cells/physiology , Triazines/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , Endoplasmic Reticulum Stress/drug effects , Glucose/pharmacology , Homeostasis/drug effects , Humans , Insulin Secretion/drug effects , Insulin-Secreting Cells/ultrastructure , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/physiology , Pluripotent Stem Cells , Protein Phosphatase 1/genetics , Protein Phosphatase 1/physiology , Transcription Factor CHOP/deficiency , Transcription Factor CHOP/genetics , Transcription Factor CHOP/physiology , Triazines/therapeutic use
15.
Diabetologia ; 64(11): 2534-2549, 2021 11.
Article in English | MEDLINE | ID: mdl-34448879

ABSTRACT

AIMS/HYPOTHESIS: We studied the effects of heterozygous human INS gene mutations on insulin secretion, endoplasmic reticulum (ER) stress and other mechanisms in both MIN6 and human induced pluripotent stem cells (hiPSC)-derived beta-like cells, as well as the effects of prolonged overexpression of mutant human INS in MIN6 cells. METHODS: We modelled the structure of mutant C109Y and G32V proinsulin computationally to examine the in silico effects. We then overexpressed either wild-type (WT), mutant (C109Y or G32V), or both WT and mutant human preproinsulin in MIN6 cells, both transiently and stably over several weeks. We measured the levels of human and rodent insulin secreted, and examined the transcript and protein levels of several ER stress and apoptotic markers. We also reprogrammed human donor fibroblasts heterozygous for the C109Y mutation into hiPSCs and differentiated these into pancreatic beta-like cells, which were subjected to single-cell RNA-sequencing and transcript and protein analyses for ER stress and apoptotic markers. RESULTS: The computational modelling studies, and short-term and long-term expression studies in beta cells, revealed the presence of ER stress, organelle changes and insulin processing defects, resulting in a decreased amount of insulin secreted but not the ability to secrete insulin. By 9 weeks of expression of mutant human INS, dominant-negative effects of mutant INS were evident and beta cell insulin secretory capacity declined. INS+/C109Y patient-derived beta-like cells and single-cell RNA-sequencing analyses then revealed compensatory upregulation in genes involved in insulin secretion, processing and inflammatory response. CONCLUSIONS/INTERPRETATION: The results provide deeper insights into the mechanisms of beta cell failure during INS mutation-mediated diabetes disease progression. Decreasing spliced X-box binding protein 1 (sXBP1) or inflammatory response could be avenues to restore the function of the remaining WT INS allele.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Insulin-Secreting Cells/metabolism , Insulin/genetics , Mutation , Pancreatic Diseases/metabolism , Biological Transport , Cells, Cultured , Diabetes Mellitus/metabolism , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation/physiology , Genetic Vectors , Glucose/pharmacology , Humans , Infant , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/ultrastructure , Karyotyping , Microscopy, Electron, Transmission , Pancreatic Diseases/pathology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Proinsulin/genetics , Real-Time Polymerase Chain Reaction , Transfection
16.
Sci Transl Med ; 13(600)2021 06 30.
Article in English | MEDLINE | ID: mdl-34193609

ABSTRACT

The paired box 6 (PAX6) transcription factor is crucial for normal pancreatic islet development and function. Heterozygous mutations of PAX6 are associated with impaired insulin secretion and early-onset diabetes mellitus in humans. However, the molecular mechanism of PAX6 in controlling insulin secretion in human beta cells and its pathophysiological role in type 2 diabetes (T2D) remain ambiguous. We investigated the molecular pathway of PAX6 in the regulation of insulin secretion and the potential therapeutic value of PAX6 in T2D by using human pancreatic beta cell line EndoC-ßH1, the db/db mouse model, and primary human pancreatic islets. Through loss- and gain-of-function approaches, we uncovered a mechanism by which PAX6 modulates glucose-stimulated insulin secretion (GSIS) through a cAMP response element-binding protein (CREB)/Munc18-1/2 pathway. Moreover, under diabetic conditions, beta cells and pancreatic islets displayed dampened PAX6/CREB/Munc18-1/2 pathway activity and impaired GSIS, which were reversed by PAX6 replenishment. Adeno-associated virus-mediated PAX6 overexpression in db/db mouse pancreatic beta cells led to a sustained amelioration of glycemic perturbation in vivo but did not affect insulin resistance. Our study highlights the pathophysiological role of PAX6 in T2D-associated beta cell dysfunction in humans and suggests the potential of PAX6 gene transfer in preserving and restoring beta cell function.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Islets of Langerhans , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Homeostasis , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism
17.
Trends Endocrinol Metab ; 32(8): 609-622, 2021 08.
Article in English | MEDLINE | ID: mdl-34154916

ABSTRACT

Diabetes is a severe chronic disease worldwide. In various types of diabetes, the pancreatic beta cells fail to secrete sufficient insulin, at some point, to regulate blood glucose levels. Therefore, the replacement of dysfunctional pancreas, islets of Langerhans, or even the insulin-secreting beta cells facilitates physiological regulation of blood glucose levels. However, the current lack of sufficient donor human islets for cell replacement therapy precludes a routine and absolute cure for most of the existing diabetes cases globally. It is envisioned that tissue engineering of a bioartificial pancreas will revolutionize regenerative medicine and the treatment of diabetes. In this review, we discuss the anatomy and physiology of the pancreas, and identify the clinical considerations for engineering a bioartificial pancreas. Subsequently, we dissect the bioengineering problem based on the design of the device, the biomaterial used, and the cells involved. Last but not least, we highlight current tissue engineering challenges and explore potential directions for future work.


Subject(s)
Diabetes Mellitus, Type 1 , Pancreas, Artificial , Printing, Three-Dimensional , Tissue Engineering , Blood Glucose , Diabetes Mellitus, Type 1/therapy , Humans , Insulin , Pancreas/anatomy & histology , Pancreas/physiology , Regenerative Medicine
18.
STAR Protoc ; 2(2): 100471, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33997805

ABSTRACT

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).


Subject(s)
Cell Differentiation , Gene Expression Profiling , Gene Expression Regulation , Liver/metabolism , Pluripotent Stem Cells/metabolism , Cells, Cultured , Humans , Liver/cytology , Pluripotent Stem Cells/cytology
19.
Diabetes ; 70(8): 1689-1702, 2021 08.
Article in English | MEDLINE | ID: mdl-33958328

ABSTRACT

Metformin is becoming a popular treatment before and during pregnancy, but current literature on in utero exposure to metformin lacks long-term clinical trials and mechanistic studies. Current literature on the effects of metformin on mature pancreatic ß-cells highlights its dual, opposing, protective, or inhibitory effects, depending on metabolic environment. However, the impact of metformin on developing human pancreatic ß-cells remains unknown. In this study, we investigated the potential effects of metformin exposure on human pancreatic ß-cell development and function in vitro. In the absence of metabolic challenges such as high levels of glucose and fatty acids, metformin exposure impaired the development and function of pancreatic ß-cells, with downregulation of pancreatic genes and dysfunctional mitochondrial respiration. It also affected the insulin secretion function of pancreatic ß-cells. These findings call for further in-depth evaluation of the exposure of human embryonic and fetal tissue during pregnancy to metformin and its implications for long-term offspring health.


Subject(s)
Cell Differentiation/drug effects , Human Embryonic Stem Cells/drug effects , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Metformin/pharmacology , Pancreas/drug effects , Cell Survival/drug effects , Human Embryonic Stem Cells/cytology , Humans , Insulin Secretion/drug effects , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Pancreas/cytology , Pancreas/metabolism
20.
Nat Commun ; 12(1): 3133, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34035238

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Glucose Transporter Type 2/genetics , Glucose/metabolism , Hepatocyte Nuclear Factor 1-alpha/genetics , Insulin Secretion/genetics , Insulin-Secreting Cells/metabolism , Mutation , Cells, Cultured , Chromatin Immunoprecipitation Sequencing/methods , Diabetes Mellitus, Type 2/metabolism , Female , Glucose Transporter Type 2/metabolism , Hepatocyte Nuclear Factor 1-alpha/chemistry , Hepatocyte Nuclear Factor 1-alpha/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Insulin-Secreting Cells/cytology , Male , Molecular Dynamics Simulation , Pedigree , Protein Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...