Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Clin Endocrinol Metab ; 109(2): 413-423, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-37671625

ABSTRACT

BACKGROUND: Macroencapsulated pancreatic endoderm cells (PECs) can reverse diabetes in rodents and preclinical studies revealed that thyroid hormones in vitro and in vivo bias PECs to differentiate into insulin-producing cells. In an ongoing clinical trial, PECs implanted in macroencapsulation devices into patients with type 1 diabetes were safe but yielded heterogeneous outcomes. Though most patients developed meal responsive C-peptide, levels were heterogeneous and explanted grafts had variable numbers of surviving cells with variable distribution of endocrine cells. METHODS: We measured circulating triiodothyronine and thyroxine levels in all patients treated at 1 of the 7 sites of the ongoing clinical trial and determined if thyroid hormone levels were associated with the C-peptide or glucagon levels and cell fate of implanted PECs. RESULTS: Both triiodothyronine and thyroxine levels were significantly associated with the proportion of cells that adopted an insulin-producing fate with a mature phenotype. Thyroid hormone levels were inversely correlated to circulating glucagon levels after implantation, suggesting that thyroid hormones lead PECs to favor an insulin-producing fate over a glucagon-producing fate. In mice, hyperthyroidism led to more rapid maturation of PECs into insulin-producing cells similar in phenotype to PECs in euthyroid mice. CONCLUSION: These data highlight the relevance of thyroid hormones in the context of PEC therapy in patients with type 1 diabetes and suggest that a thyroid hormone adjuvant therapy may optimize cell outcomes in some PEC recipients.


Subject(s)
Diabetes Mellitus, Type 1 , Humans , Mice , Animals , Diabetes Mellitus, Type 1/metabolism , C-Peptide/metabolism , Thyroxine/metabolism , Triiodothyronine/metabolism , Endoderm/metabolism , Endoderm/transplantation , Glucagon/metabolism
2.
Endocr Rev ; 44(2): 222-253, 2023 03 04.
Article in English | MEDLINE | ID: mdl-36111962

ABSTRACT

For the past century, insulin injections have saved millions of lives, but glycemic instability is still a persistent challenge for people with diabetes, leading to tremendous morbidity and premature mortality. Research in the field of islet transplantation has demonstrated that replacing insulin-producing ß cells can restore euglycemia comparable to individuals without diabetes. However, a short supply of cadaveric islet donors, the technically challenging process of isolating islets, and the requirement for chronic immune suppression have impeded widespread clinical adoption. Rather than relying on cadaveric cells, pluripotent stem cells could serve as a virtually unlimited supply of insulin-producing ß cells. Protocols have been developed that mimic the normal in vivo development of the human pancreas to generate pancreatic progenitor cells in vitro. Ongoing investigations have yielded progressively more mature ß-like cells in vitro that produce insulin but do not yet fully mimic healthy mature ß cells. Alongside development of differentiation protocols, other work has provided insight into potential implantation sites for stem cell-derived islet cells including the subcutaneous space, portal vein, and omentum. To optimize implanted cell survival and function, development of immune modulation therapies is ongoing, including selection of immunomodulatory medications and genetic modification of implanted cells to evade immune responses. Further, macroencapsulation or microencapsulation devices could be used to contain and/or immunoprotect implanted cells from the immune response including by using 3-dimensional bioprinting to facilitate the process. Remarkably, ongoing clinical trials have now yielded the first patient relying on differentiated stem cells rather than syringes as their insulin replacement therapy.


Subject(s)
Insulin-Secreting Cells , Islets of Langerhans , Humans , Insulin , Stem Cells , Cell Differentiation , Cadaver
3.
Endocrinology ; 163(6)2022 06 01.
Article in English | MEDLINE | ID: mdl-35435956

ABSTRACT

Up to 6% of diabetes has a monogenic cause including mutations in the insulin gene, and patients are candidates for a gene therapy. Using a mouse model of permanent neonatal diabetes, we assessed the efficacy of an adeno-associated virus (AAV)-mediated gene therapy. We used AAVs with a rat insulin 1 promoter (Ins1) regulating a human insulin gene (INS; AAV Ins1-INS) or native mouse insulin 1 (Ins1; AAV Ins-Ins1) to deliver an insulin gene to ß-cells of constitutive insulin null mice (Ins1-/-Ins2-/-) and adult inducible insulin-deficient mice [Ins1-/-Ins2f/f PdxCreER and Ins1-/-Ins2f/f mice administered AAV Ins1-Cre)]. Although AAV Ins1-INS could successfully infect and confer insulin expression to ß-cells, insulin null ß-cells had a prohormone processing defect. Secretion of abundant proinsulin transiently reversed diabetes. We reattempted therapy with AAV Ins1-Ins1, but Ins1-/-Ins2-/- ß-cells still had a processing defect of both replaced Ins1 and pro-islet amyloid polypeptide (proIAPP). In adult inducible models, ß-cells that lost insulin expression developed a processing defect that resulted in impaired proIAPP processing and elevated circulating proIAPP, and cells infected with AAV Ins1-Ins1 to rescue insulin expression secreted proinsulin. We assessed the subcellular localization of prohormone convertase 1/3 (PC1/3) and detected defective sorting of PC1/3 to glycogen-containing vacuoles and retention in the endoplasmic reticulum as a potential mechanism underlying defective processing. We provide evidence that persistent production of endogenous proinsulin within ß-cells is necessary for ß-cells to be able to properly store and process proinsulin.


Subject(s)
Insulin-Secreting Cells , Proinsulin , Animals , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Mice , Mice, Knockout , Proinsulin/genetics , Proinsulin/metabolism , Rats
4.
Physiol Rev ; 102(1): 155-208, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34280055

ABSTRACT

Peptide hormones are first produced as larger precursor prohormones that require endoproteolytic cleavage to liberate the mature hormones. A structurally conserved but functionally distinct family of nine prohormone convertase enzymes (PCs) are responsible for cleavage of protein precursors, of which PC1/3 and PC2 are known to be exclusive to neuroendocrine cells and responsible for prohormone cleavage. Differential expression of PCs within tissues defines prohormone processing; whereas glucagon is the major product liberated from proglucagon via PC2 in pancreatic α-cells, proglucagon is preferentially processed by PC1/3 in intestinal L cells to produce glucagon-like peptides 1 and 2 (GLP-1, GLP-2). Beyond our understanding of processing of islet prohormones in healthy islets, there is convincing evidence that proinsulin, pro-islet amyloid polypeptide (proIAPP), and proglucagon processing is altered during prediabetes and diabetes. There is predictive value of elevated circulating proinsulin or proinsulin-to-C-peptide ratio for progression to type 2 diabetes, and elevated proinsulin or proinsulin-to-C-peptide ratio is predictive for development of type 1 diabetes in at-risk groups. After onset of diabetes, patients have elevated circulating proinsulin and proIAPP, and proinsulin may be an autoantigen in type 1 diabetes. Furthermore, preclinical studies reveal that α-cells have altered proglucagon processing during diabetes, leading to increased GLP-1 production. We conclude that despite strong associative data, current evidence is inconclusive on the potential causal role of impaired prohormone processing in diabetes and suggest that future work should focus on resolving the question of whether altered prohormone processing is a causal driver or merely a consequence of diabetes pathology.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucagon/metabolism , Proglucagon/metabolism , Proinsulin/metabolism , Animals , Glucagon-Like Peptide 1/metabolism , Humans , Protein Precursors/metabolism
5.
Cell Stem Cell ; 28(12): 2047-2061.e5, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34861146

ABSTRACT

An open-label, first-in-human phase 1/2 study is being conducted to evaluate the safety and efficacy of pancreatic endoderm cells (PECs) implanted in non-immunoprotective macroencapsulation devices for the treatment of type 1 diabetes. We report an analysis on 1 year of data from the first cohort of 15 patients from a single trial site that received subcutaneous implantation of cell products combined with an immunosuppressive regimen. Implants were well tolerated with no teratoma formation or severe graft-related adverse events. After implantation, patients had increased fasting C-peptide levels and increased glucose-responsive C-peptide levels and developed mixed meal-stimulated C-peptide secretion. There were immunosuppression-related transient increases in circulating regulatory T cells, PD1high T cells, and IL17A+CD4+ T cells. Explanted grafts contained cells with a mature ß cell phenotype that were immunoreactive for insulin, islet amyloid polypeptide, and MAFA. These data, and associated findings (Shapiro et al., 2021), are the first reported evidence of meal-regulated insulin secretion by differentiated stem cells in patients.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , C-Peptide , Cell Differentiation , Diabetes Mellitus, Type 1/therapy , Endoderm , Glucose , Humans , Insulin
6.
Cell Rep Med ; 2(11): 100434, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34841287

ABSTRACT

miRNAs have crucial functions in many biological processes and are candidate biomarkers of disease. Here, we show that miR-216a is a conserved, pancreas-specific miRNA with important roles in pancreatic islet and acinar cells. Deletion of miR-216a in mice leads to a reduction in islet size, ß-cell mass, and insulin levels. Single-cell RNA sequencing reveals a subpopulation of ß-cells with upregulated acinar cell markers under a high-fat diet. miR-216a is induced by TGF-ß signaling, and inhibition of miR-216a increases apoptosis and decreases cell proliferation in pancreatic cells. Deletion of miR-216a in the pancreatic cancer-prone mouse line KrasG12D;Ptf1aCreER reduces the propensity of pancreatic cancer precursor lesions. Notably, circulating miR-216a levels are elevated in both mice and humans with pancreatic cancer. Collectively, our study gives insights into how ß-cell mass and acinar cell growth are modulated by a pancreas-specific miRNA and also suggests miR-216a as a potential biomarker for diagnosis of pancreatic diseases.


Subject(s)
Disease Progression , Gene Deletion , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Animals , Apoptosis , Base Sequence , Cell Line, Tumor , Cell Movement , Diet, High-Fat , Humans , Insulin Secretion , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/metabolism , Organ Specificity , Rats
7.
Sci Rep ; 10(1): 10518, 2020 06 29.
Article in English | MEDLINE | ID: mdl-32601405

ABSTRACT

In vivo genetic manipulation is used to study the impact of gene deletion or re-expression on ß-cell function and organism physiology. Cre-LoxP is a system wherein LoxP sites flanking a gene are recognized by Cre recombinase. Cre transgenic mice are the most prevalent technology used to deliver Cre but many models have caveats of off-target recombination, impaired ß-cell function, and high cost of animal production. Inducible estrogen receptor conjugated Cre models face leaky recombination and confounding effects of tamoxifen. As an alternative, we characterize an adeno associated virus (AAV) with a rat insulin 1 promoter driving Cre recombinase (AAV8 Ins1-Cre) that is economical and rapid to implement, and has limited caveats. Intraperitoneal AAV8 Ins1-Cre produced efficient ß-cell recombination, alongside some hepatic, exocrine pancreas, α-cell, δ-cell, and hypothalamic recombination. Delivery of lower doses via the pancreatic duct retained good rates of ß-cell recombination and limited rates of off-target recombination. Unlike inducible Cre in transgenic mice, AAV8 Ins1-Cre required no tamoxifen and premature recombination was avoided. We demonstrate the utility of this technology by inducing hyperglycemia in inducible insulin knockout mice (Ins1-/-;Ins2f/f). AAV-mediated expression of Cre in ß-cells provides an effective alternative to transgenic approaches for inducible knockout studies.


Subject(s)
Dependovirus , Insulin-Secreting Cells/metabolism , Insulin/genetics , Promoter Regions, Genetic , Recombination, Genetic , Animals , Insulin/metabolism , Integrases , Mice , Mice, Transgenic
8.
Diabetes ; 69(7): 1451-1462, 2020 07.
Article in English | MEDLINE | ID: mdl-32291281

ABSTRACT

Insulin is first produced in pancreatic ß-cells as the precursor prohormone proinsulin. Defective proinsulin processing has been implicated in the pathogenesis of both type 1 and type 2 diabetes. Though there is substantial evidence that mouse ß-cells process proinsulin using prohormone convertase 1/3 (PC1/3) and then prohormone convertase 2 (PC2), this finding has not been verified in human ß-cells. Immunofluorescence with validated antibodies revealed that there was no detectable PC2 immunoreactivity in human ß-cells and little PCSK2 mRNA by in situ hybridization. Similarly, rat ß-cells were not immunoreactive for PC2. In all histological experiments, PC2 immunoreactivity in neighboring α-cells acted as a positive control. In donors with type 2 diabetes, ß-cells had elevated PC2 immunoreactivity, suggesting that aberrant PC2 expression may contribute to impaired proinsulin processing in ß-cells of patients with diabetes. To support histological findings using a biochemical approach, human islets were used for pulse-chase experiments. Despite inhibition of PC2 function by temperature blockade, brefeldin A, chloroquine, and multiple inhibitors that blocked production of mature glucagon from proglucagon, ß-cells retained the ability to produce mature insulin. Conversely, suppression of PC1/3 blocked processing of proinsulin but not proglucagon. By demonstrating that healthy human ß-cells process proinsulin by PC1/3 but not PC2, we suggest that there is a need to revise the long-standing theory of proinsulin processing.


Subject(s)
Insulin-Secreting Cells/metabolism , Proinsulin/metabolism , Proprotein Convertase 1/physiology , Proprotein Convertase 2/physiology , Animals , Diabetes Mellitus, Type 2/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Proprotein Convertase 1/analysis , Proprotein Convertase 2/analysis , Proprotein Convertase 2/metabolism
9.
Endocrinology ; 159(1): 83-102, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29029025

ABSTRACT

Insulin receptor (IR) insufficiency in ß-cells leads to impaired insulin secretion and reduced ß-cell hyperplasia in response to hyperglycemia. Selective IR deficiency in ß-cells in later embryological development may lead to compensatory ß-cell hyperplasia. Although these findings suggest insulin signaling on the ß-cell is important for ß-cell function, they are confounded by loss of signaling by the insulinlike growth factors through the IR. To determine whether insulin itself is necessary for ß-cell development and maturation, we performed a characterization of pancreatic islets in mice with deletions of both nonallelic insulin genes (Ins1-/-Ins2-/-). We immunostained neonatal Ins1-/-Ins2-/- and Ins1+/+Ins2+/+ pancreata and performed quantitative polymerase chain reaction on isolated neonatal islets. Insulin-deficient islets had reduced expression of factors normally expressed in maturing ß-cells, including muscoloaponeurotic fibrosarcoma oncogene homolog A, homeodomain transcription factor 6.1, and glucose transporter 2. Ins1-/-Ins2-/-ß-cells expressed progenitor factors associated with stem cells or dedifferentiated ß-cells, including v-myc avian myolocytomatosis viral oncogene lung carcinoma derived and homeobox protein NANOG. We replaced insulin by injection or islet transplantation to keep mice alive into adulthood to determine whether insulin replacement was sufficient for the completed maturation of insulin-deficient ß-cells. Short-term insulin glargine (Lantus®) injections partially rescued the ß-cell phenotype, whereas long-term replacement of insulin by isogenic islet transplantation supported the formation of more mature ß-cells. Our findings suggest that tightly regulated glycemia, insulin species, or other islet factors are necessary for ß-cell maturation.


Subject(s)
Hyperglycemia/surgery , Insulin-Secreting Cells/metabolism , Insulin/deficiency , Islets of Langerhans Transplantation , Animals , Animals, Newborn , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Size/drug effects , Female , Fibrosis , Gene Expression Regulation, Developmental/drug effects , Hormone Replacement Therapy/adverse effects , Hyperglycemia/drug therapy , Hyperglycemia/etiology , Hyperglycemia/pathology , Injections, Subcutaneous , Insulin/genetics , Insulin/metabolism , Insulin Glargine/administration & dosage , Insulin Glargine/adverse effects , Insulin Glargine/therapeutic use , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Tissue Culture Techniques
10.
Nat Rev Gastroenterol Hepatol ; 14(10): 612-628, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28811674

ABSTRACT

The pancreas is a complex organ with exocrine and endocrine components. Many pathologies impair exocrine function, including chronic pancreatitis, cystic fibrosis and pancreatic ductal adenocarcinoma. Conversely, when the endocrine pancreas fails to secrete sufficient insulin, patients develop diabetes mellitus. Pathology in either the endocrine or exocrine pancreas results in devastating economic and personal consequences. The current standard therapy for treating patients with type 1 diabetes mellitus is daily exogenous insulin injections, but cell sources of insulin provide superior glycaemic regulation and research is now focused on the goal of regenerating or replacing ß cells. Stem-cell-based models might be useful to study exocrine pancreatic disorders, and mesenchymal stem cells or secreted factors might delay disease progression. Although the standards that bioengineered cells must meet before being considered as a viable therapy are not yet established, any potential therapy must be acceptably safe and functionally superior to current therapies. Here, we describe progress and challenges in cell-based methods to restore pancreatic function, with a focus on optimizing the site for cell delivery and decreasing requirements for immunosuppression through encapsulation. We also discuss the tools and strategies being used to generate exocrine pancreas and insulin-producing ß-cell surrogates in situ and highlight obstacles to clinical application.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Cellular Reprogramming Techniques , Pancreatic Diseases/physiopathology , Pancreatic Diseases/therapy , Regenerative Medicine , Humans
11.
ACS Appl Mater Interfaces ; 2(2): 385-90, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20356183

ABSTRACT

This paper reports a novel approach to designing advanced solid Li ion electrolytes for application in various solid state ionic devices, including Li ion secondary batteries, gas sensors, and electrochromic displays. The employed methodology involves a solid-solution reaction between the two best-known fast Li ion conductors in the garnet-family of compounds Li(6)BaLa(2)M(2)O(12) (M = Nb, Ta) and Li(7)La(3)Zr(2)O(12). Powder X-ray diffraction (PXRD), scanning electron microscopy (SEM), AC impedance, and (7)Li nuclear magnetic resonance (Li NMR) spectroscopy were employed to characterize phase formation, morphology, ionic conductivity, and Li ion coordination in Li(6.5)La(2.5)BaZrMO(12). PXRD shows for formation of a cubic garnet-like structure and AC impedance data is consistent with other known solid Li ion electrolytes. Li(6.5)La(2.5)BaZrTaO(12) exhibits a fast Li ion conductivity of about 6 x 10(-3) S cm(-1) at 100 degrees C, which is comparable to that of currently employed organic polymer electrolytes value at room temperature. The Nb analogue shows an order of magnitude lower ionic conductivity than that of the corresponding Ta member, which is consistent with the trend in garnet-type electrolytes reported in the literature. Samples sintered at 1100 degrees C shows the highest electrical conductivity compared to that of 900 degrees C. (7)Li MAS NMR shows a sharp single peak at 0 ppm with respect to LiCl, which may be attributed to fast migration of ions between various sites in the garnets, and also suggesting average distributions of Li ions at average octahedral coordination in Li(6.5)La(2.5)BaZrMO(12). The present work together with literature used to establish very important fundamental relationship of functional property-Li concentration-crystal structure-Li diffusion coefficient in the garnet family of Li ion electrolytes.

SELECTION OF CITATIONS
SEARCH DETAIL
...