Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Front Endocrinol (Lausanne) ; 12: 635405, 2021.
Article in English | MEDLINE | ID: mdl-34025576

ABSTRACT

Background: Cell therapy of diabetes aims at restoring the physiological control of blood glucose by transplantation of functional pancreatic islet cells. A potentially unlimited source of cells for such transplantations would be islet cells derived from an in vitro differentiation of human pluripotent stem cells (hESC/hiPSC). The islet-like clusters (ILC) produced by the known differentiation protocols contain various cell populations. Among these, the ß-cells that express both insulin and the transcription factor Nkx6.1 seem to be the most efficient to restore normoglycemia in diabetes animal models. Our aim was to find markers allowing selection of these efficient cells. Methods: Functional Cell-Capture Screening (FCCS) was used to identify markers that preferentially capture the cells expressing both insulin and Nkx6.1, from hESC-derived ILC cells. In order to test whether selection for such markers could improve cell therapy in diabetic mouse models, we used ILC produced from a clinical-grade line of hESC by a refined differentiation protocol adapted to up-scalable bioreactors. Re-aggregated MACS sorted cells were encapsulated in microspheres made of alginate modified to reduce foreign body reaction. Implantation was done intraperitoneally in STZ-treated C57BL/6 immuno-competent mice. Results: CD49A (integrin alpha1) was identified by FCCS as a marker for cells that express insulin (or C-peptide) as well as Nkx6.1 in ILC derived by hESC differentiation. The ILC fraction enriched in CD49A + cells rapidly reduced glycemia when implanted in diabetic mice, whereas mice receiving the CD49A depleted population remained highly diabetic. CD49A-enriched ILC cells also produced higher levels of human C-peptide in the blood of transplanted mice. However, the difference between CD49A-enriched and total ILC cells remained small. Another marker, CD26 (DPP4), was identified by FCCS as binding insulin-expressing cells which are Nkx6.1 negative. Depletion of CD26 + cells followed by enrichment for CD49A + cells increased insulin+/Nkx6.1+ cells fraction to ~70%. The CD26 - /CD49A + enriched ILC exhibited improved function over non-sorted ILC or CD49A + cells in diabetic mice and maintain prolonged blood C-peptide levels. Conclusions: Refining the composition of ILC differentiated from hPSC by negative selection to remove cells expressing CD26 and positive selection for CD49A expressing cells could enable more effective cell therapy of diabetes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Dipeptidyl Peptidase 4/biosynthesis , Integrin alpha1/biosynthesis , Islets of Langerhans Transplantation , Islets of Langerhans/metabolism , Pluripotent Stem Cells/metabolism , Animals , C-Peptide/biosynthesis , Cell Differentiation , Cell Separation , Homeodomain Proteins/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Mice , Mice, Inbred C57BL , Microspheres
2.
Methods Mol Biol ; 2211: 85-96, 2021.
Article in English | MEDLINE | ID: mdl-33336272

ABSTRACT

In this chapter, we describe a simple and unique method for the differentiation of mouse embryonic stem cells into insulin-producing cells. In addition to cytokines and growth factors, key transcription factors for pancreatic development are applied in this method through protein transduction technology. Furthermore, a combination of nanofiber plates and laminin coatings improves the yield of differentiated cells. The insulin-producing cells derived through this method express marker genes of mature ß-cells and have an ability to secrete insulin; therefore, these cells are useful for fundamental studies on pancreatic development, drug development, and regenerative medicine for diabetes.


Subject(s)
Cell Differentiation/genetics , Gene Expression , Insulin-Secreting Cells/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Transcription Factors/genetics , Transduction, Genetic , Animals , C-Peptide/biosynthesis , Cells, Cultured , Fibroblasts/metabolism , Humans , Insulin/biosynthesis , Insulin-Secreting Cells/cytology , Mice , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Transcription Factors/metabolism
3.
JCI Insight ; 6(3)2021 02 08.
Article in English | MEDLINE | ID: mdl-33351781

ABSTRACT

Clinical trials of biologic therapies in type 1 diabetes (T1D) aim to mitigate autoimmune destruction of pancreatic ß cells through immune perturbation and serve as resources to elucidate immunological mechanisms in health and disease. In the T1DAL trial of alefacept (LFA3-Ig) in recent-onset T1D, endogenous insulin production was preserved in 30% of subjects for 2 years after therapy. Given our previous findings linking exhausted-like CD8+ T cells to beneficial response in T1D trials, we applied unbiased analyses to sorted CD8+ T cells to evaluate their potential role in T1DAL. Using RNA sequencing, we found that greater insulin C-peptide preservation was associated with a module of activation- and exhaustion-associated genes. This signature was dissected into 2 CD8 memory phenotypes through correlation with cytometry data. These cells were hypoproliferative, shared expanded rearranged TCR junctions, and expressed exhaustion-associated markers including TIGIT and KLRG1. The 2 phenotypes could be distinguished by reciprocal expression of CD8+ T and NK cell markers (GZMB, CD57, and inhibitory killer cell immunoglobulin-like receptor [iKIR] genes), versus T cell activation and differentiation markers (PD-1 and CD28). These findings support previous evidence linking exhausted-like CD8+ T cells to successful immune interventions for T1D, while suggesting that multiple inhibitory mechanisms can promote this beneficial cell state.


Subject(s)
Alefacept/therapeutic use , C-Peptide/biosynthesis , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Adolescent , Adult , C-Peptide/genetics , CD57 Antigens/metabolism , CD8-Positive T-Lymphocytes/classification , CD8-Positive T-Lymphocytes/metabolism , Child , Diabetes Mellitus, Type 1/metabolism , Double-Blind Method , Female , Humans , Immunologic Factors/therapeutic use , Immunologic Memory/genetics , Immunophenotyping , Killer Cells, Natural/immunology , Lectins, C-Type/metabolism , Lymphocyte Activation , Male , Programmed Cell Death 1 Receptor/metabolism , RNA-Seq , Receptors, Immunologic/metabolism , Young Adult
4.
Cytokine ; 138: 155379, 2021 02.
Article in English | MEDLINE | ID: mdl-33271384

ABSTRACT

BACKGROUND: Blood has been the usual biological fluid for measuring analytes, but there is mounting evidence that saliva may be also useful for detecting cytokines in a noninvasive way. Thus, in this study we aimed to determine concentration of cytokines and other analytes in saliva from a population of healthy children. METHODS: We collected un-stimulated whole saliva samples from clinically healthy children, and concentration of 17 cytokines and 12 other analytes were measured in supernatants. All values were adjusted by albumin content and were log-transformed before multivariate statistical analysis. RESULTS: We included 114 children (53.5% females) between 6.0 and 11.9 years old. The highest concentrations (medians, pg/µg albumin) were seen for visfatin (183.70) and adiponectin (162.26) and the lowest for IL-13 and IL-2 (~0.003). Albumin concentration was associated with age (rS = 0.39, p < 0.001). In the multivariate analysis, five analytes (C peptide, ghrelin, GLP-1, glucagon, leptin) inversely correlated with age and positively with height-for-age. Age was also positively associated with PAI-1, while height-for-age was also positively associated with insulin and visfatin. Finally, BMI-for-age had a positive correlation with GM-CSF and insulin. CONCLUSIONS: Herein, we provided concentration values for 29 analytes in saliva from healthy children that may be useful as preliminary reference framework in the clinical research setting.


Subject(s)
Cytokines/metabolism , Saliva/metabolism , Adiponectin/biosynthesis , Age Factors , Body Height , C-Peptide/biosynthesis , Child , Cytokines/biosynthesis , Female , Ghrelin/biosynthesis , Glucagon/biosynthesis , Glucagon-Like Peptide 1/biosynthesis , Humans , Insulin/metabolism , Interleukin-13/biosynthesis , Interleukin-2/biosynthesis , Leptin/biosynthesis , Male , Multivariate Analysis , Nicotinamide Phosphoribosyltransferase/biosynthesis , Reference Values
5.
Transplantation ; 103(3): 630-637, 2019 03.
Article in English | MEDLINE | ID: mdl-30211831

ABSTRACT

BACKGROUND: When transplanted human pancreatic islets are exposed to blood during intraportal infusion, an innate immune response is triggered. This instant blood-mediated inflammatory reaction (IBMIR) activates the coagulation and complement cascades and leads to the destruction of 25% of all transplanted islets within minutes, contributing to the need, in most patients, for islets from more than 1 donor. Low molecular dextran sulfate (LMW-DS) has been shown in experimental settings to inhibit IBMIR. METHODS: The Clinical Islet Transplantation consortium 01 study was a phase II, multicenter, open label, active control, randomized study. Twenty-four subjects were randomized to peritransplant intraportal and systemic treatment with either LMW-DS or heparin, targeting an activated partial thromboplastin time of 150 ± 10 seconds and 50 ± 5 seconds, respectively. C-peptide response was measured with a mixed meal tolerance test at 75 and 365 days after transplant. RESULTS: Low molecular dextran sulfate was safe and well tolerated with similar observed adverse events (mostly attributed to immunosuppression) as in the heparin arm. There was no difference in the primary endpoint (stimulated C-peptide 75 ± 5 days after the first transplant) between the 2 arms (1.33 ± 1.10 versus 1.56 ± 1.36 ng/mL, P = 0.66). Insulin requirement, metabolic parameters, Clarke and HYPO score, quality of life, and safety were similar between the 2 treatments groups. CONCLUSIONS: Even with low dosing, LMW-DS showed similar efficacy in preventing IBMIR to promote islet engraftment when compared to "state-of-the art" treatment with heparin. Furthermore, no substantial differences in the efficacy and safety endpoints were detected, providing important information for future studies with more optimal dosing of LMW-DS for the prevention of IBMIR in islet transplantation.


Subject(s)
Dextran Sulfate/therapeutic use , Inflammation/prevention & control , Islets of Langerhans Transplantation , Adult , Aged , C-Peptide/biosynthesis , Complement Activation/drug effects , Complement System Proteins/immunology , Female , Glucose Tolerance Test , Heparin/therapeutic use , Humans , Immune Tolerance/drug effects , Immunity, Innate , Islets of Langerhans/cytology , Male , Middle Aged , Molecular Weight , Norway , Partial Thromboplastin Time , Quality of Life , Sweden
6.
Med Princ Pract ; 27(1): 86-91, 2018.
Article in English | MEDLINE | ID: mdl-29320773

ABSTRACT

OBJECTIVE: To examine the effect of infection with Enterovirus (EV) in children with type 1 diabetes (T1D) on the activities of serum antioxidant enzymes in diabetic and nondiabetic controls. SUBJECTS AND METHODS: Three hundred and eighty-two diabetic and 100 nondiabetic children were tested for EV RNA using reverse transcriptase (RT)-PCR. The activities of serum superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT) were also estimated in diabetic patients infected with EV (T1D-EV+), those not infected with EV (T1D-EV-), and in nondiabetic controls. RESULTS: The frequency of EV was higher in diabetic children (100/382; 26.2%) than in healthy controls (0/100). Levels of fasting blood glucose (FBG), glycosylated hemoglobin (HbA1c) and C-reactive protein (CRP) were significantly higher but C-peptide was significantly lower in diabetic children than in controls. CRP levels were higher in the T1D-EV+ group than in the T1D-EV- group, and higher in all diabetic children than in nondiabetic controls. The activities of the antioxidant enzymes GPx, SOD, and CAT decreased significantly in diabetic children compared to in controls. Moreover, the activities of the enzymes tested were significantly reduced in the T1D-EV+ group compared to in the T1D-EV- group. CONCLUSION: Our data indicate that EV infection correlated with a decrease in the activity of antioxidant enzymes in the T1D-EV+ group compared to in the T1D-EV- group; this may contribute to ß cell damage and increased inflammation.


Subject(s)
Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/epidemiology , Enterovirus Infections/blood , Enterovirus Infections/epidemiology , Adolescent , Blood Glucose , C-Peptide/biosynthesis , C-Reactive Protein/biosynthesis , Catalase/biosynthesis , Child , Child, Preschool , Female , Glutathione Peroxidase/biosynthesis , Glycated Hemoglobin , Humans , Male , Superoxide Dismutase/biosynthesis
7.
Cell Commun Signal ; 15(1): 43, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29041943

ABSTRACT

BACKGROUND: We evaluated the therapeutic effect and fate of high doses of human umbilical cord Wharton jelly cells (hUCWJCs) after IP administration to streptozotocin (STZ)-induced diabetic mice. METHODS: Type 1 diabetes (T1D) was induced in Kunming mice via IP injection of STZ. hUCWJCs were labeled with 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI). Diabetic animals with sustained hyperglycemia for at least 2 weeks were administered 1 × 107 Dil-hUCWJCs via intraperitoneal injection. Insulin, glucagon and PDX-1 were detected by immunofluorescence with confocal microscopy. Serum mouse and human C-peptide was assayed in blood collected via intracardiac puncture. Specific ß-cell differentiation markers and human DNA were assessed using qPCR performed with 200 ng of target DNA. RESULTS: hUCWJCs migrated to the STZ-damaged organs and contributed to lower blood glucose levels in 30% of the treated mice. Confocal microscopy revealed the presence of resident insulin-positive cells in the liver and kidneys. hUCWJC-treated mice with restored hyperglycemia also showed increased serum mouse C-peptide levels. The qPCR results, particularly in the liver, revealed that after transplantation hUCWJCs upregulated genes of endocrine precursors but failed to express endocrine stage markers. Mice with restored hyperglycemia had reduced urinary volume and lacked glomerular hypertrophy, exhibiting a morphology resembling that of normal glomeruli. Moreover, we also verified that one of the possible mechanisms by which hUCWJCs exert immunosuppressive effects is through down-regulation of the cell surface receptor HLA-1. CONCLUSIONS: We confirmed the potential of IP administration of hUCWJCs and the capability of these cells to migrate to damaged tissues and promote insulin secretion from non-pancreatic local cells and to improve renal damage. These findings confer unique therapeutic properties to hUCWJCs, suggesting a promising future in the treatment of diabetes mellitus.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/therapy , Insulin/biosynthesis , Kidney/injuries , Mesenchymal Stem Cell Transplantation , Pancreas/metabolism , Umbilical Cord/cytology , Animals , Blood Glucose/metabolism , C-Peptide/biosynthesis , Cell Differentiation , Diabetes Mellitus, Experimental/pathology , Humans , Male , Mice
8.
Sci Rep ; 7(1): 3156, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28600547

ABSTRACT

Congenital hyperinsulinism (CHI) is a rare genetic disorder characterized by excess insulin secretion, which results in hypoglycemia. Mutation of sulfonylurea receptor 1 (SUR1), encoded by the ABCC8 gene, is the main cause of CHI. Here, we captured the phenotype of excess insulin secretion through pancreatic differentiation of ABCC8-deficient stem cells generated by the CRISPR/Cas9 system. ABCC8-deficient insulin-producing cells secreted higher insulin than their wild-type counterparts, and the excess insulin secretion was rescued by nifedipine, octreotide and nicorandil. Further, we tested the role of SUR1 in response to different potassium levels and found that dysfunction of SUR1 decreased the insulin secretion rate in low and high potassium environments. Hence, pancreatic differentiation of ABCC8-deficient cells recapitulated the CHI disease phenotype in vitro, which represents an attractive model to further elucidate the function of SUR1 and to develop and screen for novel therapeutic drugs.


Subject(s)
CRISPR-Cas Systems , Human Embryonic Stem Cells/metabolism , Insulin-Secreting Cells/metabolism , Models, Biological , Sulfonylurea Receptors/genetics , C-Peptide/antagonists & inhibitors , C-Peptide/biosynthesis , Cell Differentiation , Congenital Hyperinsulinism/genetics , Congenital Hyperinsulinism/metabolism , Congenital Hyperinsulinism/pathology , Gastrointestinal Agents/pharmacology , Gene Editing/methods , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Humans , Insulin/biosynthesis , Insulin Antagonists/pharmacology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Nicorandil/pharmacology , Nifedipine/pharmacology , Octreotide/pharmacology , Phenotype , Potassium Chloride/pharmacology , Sulfonylurea Receptors/deficiency , Vasodilator Agents/pharmacology
9.
FEBS Lett ; 591(1): 129-136, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27926781

ABSTRACT

Glial fibrillary acidic protein (GFAP), expressed in peri-islet Schwann cells, is a novel target for the treatment of type 1 diabetes mellitus (T1DM). We designed a GFAP immune-tolerizing vaccine that successfully suppresses hyperglycemia and enhances C peptide secretion. The GFAP vaccine significantly prevented T cell infiltration into pancreatic islets. Moreover, after GFAP vaccination, naïve T-cell differentiation shifted from a cytotoxic Th1- to a Th2-biased humoral response. These results indicate that as a novel target, GFAP reliably predicts the development of T1DM, and that the GFAP vaccine successfully delays the progression of T1DM by regulating T-cell differentiation.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Glial Fibrillary Acidic Protein/immunology , Hemocyanins/immunology , Immune Tolerance , Vaccines/immunology , Animals , C-Peptide/biosynthesis , Female , Immunity , Immunoglobulin G/metabolism , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Mice, Inbred NOD , Pancreas/pathology , Phenotype , Th1 Cells/immunology , Th2 Cells/immunology , Vaccination
10.
J Transl Med ; 14(1): 341, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27998294

ABSTRACT

BACKGROUND: Induced pluripotent stem cells (iPSC) possess an enormous potential as both, scientific and therapeutic tools. Their application in the regenerative medicine provides new treatment opportunities for numerous diseases, including type 1 diabetes. In this work we aimed to derive insulin producing cells (IPC) from iPS cells established in defined conditions. METHODS: We optimized iPSC generation protocol and created pluripotent cell lines with stably integrated PDX1 and NKX6.1 transgenes under the transcriptional control of doxycycline-inducible promoter. These cells were differentiated using small chemical molecules and recombinant Activin A in the sequential process through the definitive endoderm, pancreatic progenitor cells and insulin producing cells. Efficiency of the procedure was assessed by quantitative gene expression measurements, immunocytochemical stainings and functional assays for insulin secretion. RESULTS: Generated cells displayed molecular markers characteristic for respective steps of the differentiation. The obtained IPC secreted insulin and produced C-peptide with significantly higher hormone release level in case of the combined expression of PDX1 and NKX6.1 induced at the last stage of the differentiation. CONCLUSIONS: Efficiency of differentiation of iPSC to IPC can be increased by concurrent expression of PDX1 and NKX6.1 during progenitor cells maturation. Protocols established in our study allow for iPSC generation and derivation of IPC in chemically defined conditions free from animal-derived components, which is of the utmost importance in the light of their prospective applications in the field of regenerative medicine.


Subject(s)
Cell Differentiation , Homeodomain Proteins/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Insulin-Secreting Cells/cytology , Trans-Activators/metabolism , Animals , C-Peptide/biosynthesis , Cells, Cultured , Cellular Reprogramming , Endoderm/cytology , Epithelial Cells/cytology , Fibroblasts/cytology , Gene Transfer Techniques , Humans , Insulin/biosynthesis , Insulin-Secreting Cells/metabolism , Transgenes
11.
PLoS One ; 9(10): e109607, 2014.
Article in English | MEDLINE | ID: mdl-25299668

ABSTRACT

OBJECTIVES: To examine whether combined vitamin D and calcium supplementation improves insulin sensitivity, insulin secretion, ß-cell function, inflammation and metabolic markers. DESIGN: 6-month randomized, placebo-controlled trial. PARTICIPANTS: Ninety-five adults with serum 25-hydroxyvitamin D [25(OH)D] ≤55 nmol/L at risk of type 2 diabetes (with prediabetes or an AUSDRISK score ≥15) were randomized. Analyses included participants who completed the baseline and final visits (treatment n = 35; placebo n = 45). INTERVENTION: Daily calcium carbonate (1,200 mg) and cholecalciferol [2,000-6,000 IU to target 25(OH)D >75 nmol/L] or matching placebos for 6 months. MEASUREMENTS: Insulin sensitivity (HOMA2%S, Matsuda index), insulin secretion (insulinogenic index, area under the curve (AUC) for C-peptide) and ß-cell function (Matsuda index x AUC for C-peptide) derived from a 75 g 2-h OGTT; anthropometry; blood pressure; lipid profile; hs-CRP; TNF-α; IL-6; adiponectin; total and undercarboxylated osteocalcin. RESULTS: Participants were middle-aged adults (mean age 54 years; 69% Europid) at risk of type 2 diabetes (48% with prediabetes). Compliance was >80% for calcium and vitamin D. Mean serum 25(OH)D concentration increased from 48 to 95 nmol/L in the treatment group (91% achieved >75 nmol/L), but remained unchanged in controls. There were no significant changes in insulin sensitivity, insulin secretion and ß-cell function, or in inflammatory and metabolic markers between or within the groups, before or after adjustment for potential confounders including waist circumference and season of recruitment. In a post hoc analysis restricted to participants with prediabetes, a significant beneficial effect of vitamin D and calcium supplementation on insulin sensitivity (HOMA%S and Matsuda) was observed. CONCLUSIONS: Daily vitamin D and calcium supplementation for 6 months may not change OGTT-derived measures of insulin sensitivity, insulin secretion and ß-cell function in multi-ethnic adults with low vitamin D status at risk of type 2 diabetes. However, in participants with prediabetes, supplementation with vitamin D and calcium may improve insulin sensitivity. TRIAL REGISTRATION: Australian New Zealand Clinical Trials Registry ACTRN12609000043235.


Subject(s)
Calcium, Dietary/administration & dosage , Cholecalciferol/administration & dosage , Diabetes Mellitus, Type 2/prevention & control , Dietary Supplements , Prediabetic State/diet therapy , Vitamin D Deficiency/diet therapy , Adiponectin/metabolism , Adult , Aged , Blood Glucose/metabolism , C-Peptide/biosynthesis , C-Reactive Protein/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Female , Humans , Insulin/biosynthesis , Insulin/pharmacology , Insulin Resistance , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Interleukin-6/metabolism , Male , Middle Aged , Osteocalcin/metabolism , Pilot Projects , Prediabetic State/metabolism , Prediabetic State/physiopathology , Tumor Necrosis Factor-alpha/metabolism , Vitamin D Deficiency/metabolism , Vitamin D Deficiency/physiopathology
12.
Cytotherapy ; 16(11): 1519-1527, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25287601

ABSTRACT

BACKGROUND AIMS: Human umbilical cord mesenchymal stromal cells (hUC-MSCs) hold great potential as a therapeutic candidate to treat diabetes, owing to their unlimited source and ready availability. METHODS: In this study, we differentiated hUC-MSCs with in vitro-synthesized pancreatic-duodenal homebox 1 (PDX1) messenger (m)RNA into islet-like cell clusters. hUC-MSCs were confirmed by both biomarker detection and functional differentiation. In vitro-synthesized PDX1 messenger RNA can be transfected into hUC-MSCs efficiently. The upregulated expression of PDX1 protein can be detected 4 h after transfection and remains detectable for 36 h. RESULTS: The induction of islet-like structures was confirmed by means of morphology and dithizone staining. Reverse transcriptase-polymerase chain reaction results revealed the expression of some key pancreatic transcription factors, such as PDX1, NeuroD, NKX6.1, Glut-2 and insulin in islet-like cell clusters. Immunofluorescence analysis showed that differentiated cells express both insulin and C-peptide. Enzyme-linked immunosorbent assay analysis validated the insulin secretion of islet-like cell clusters in response to the glucose stimulation. CONCLUSIONS: Our results demonstrate the use of in vitro-synthesized PDX1 messenger RNA to differentiate hUC-MSCs into islet-like cells and pave the way toward the development of reprogramming and directed-differentiation methods for the expression of encoded proteins.


Subject(s)
Cellular Reprogramming/genetics , Homeodomain Proteins/biosynthesis , In Vitro Techniques , Mesenchymal Stem Cells/metabolism , Trans-Activators/biosynthesis , Umbilical Cord/metabolism , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , C-Peptide/biosynthesis , Cell Differentiation/genetics , Flow Cytometry , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mesenchymal Stem Cells/cytology , Nerve Tissue Proteins/biosynthesis , RNA, Messenger/biosynthesis , Trans-Activators/genetics , Umbilical Cord/cytology
13.
Article in English | MEDLINE | ID: mdl-24932602

ABSTRACT

OBJECTIVE: The vision of potential autologous cell therapy for the cure of diabetes encourages ongoing research. According to a previously published protocol for the generation of insulin-producing cells from human monocytes, we analyzed whether the addition of growth factors could increase insulin production. This protocol was then transferred to a non-human primate model by using either blood- or spleen-derived monocytes. METHODS: Human monocytes were treated to dedifferentiate into programmable cells of monocytic origin (PCMO). In addition to the published protocol, PCMOs were then treated with either activin A, betacellulin, exendin 3 or 4. Cells were characterized by protein expression of insulin, Pdx-1, C-peptide and Glut-2. After identifying the optimal protocol, monocytes from baboon blood were isolated and the procedure was repeated. Spleen monocytes following splenectomy of a live baboon were differentiated and analyzed in the same manner and calculated in number and volume. RESULTS: Insulin content of human cells was highest when cells were treated with activin A and their insulin content was 13,000 µU/1 million cells. Insulin-producing cells form primate monocytes could successfully be generated despite using human growth factors and serum. Expression of insulin, Pdx-1, C-peptide and Glut-2 was comparable to that of human neo-islets. Total insulin content of activin A-treated baboon monocytes was 16,000 µU/1 million cells. CONCLUSION: We were able to show that insulin-producing cells can be generated from baboon monocytes with human growth factors. The amount generated from one spleen could be enough to cure a baboon from experimentally induced diabetes in an autologous cell transplant setting.


Subject(s)
Insulin/biosynthesis , Monocytes/metabolism , Activins/pharmacology , Animals , Betacellulin/pharmacology , C-Peptide/biosynthesis , Cell Dedifferentiation , Cell Differentiation , Glucose Transporter Type 2/biosynthesis , Homeodomain Proteins/biosynthesis , Humans , Monocytes/drug effects , Papio/surgery , Splenectomy , Trans-Activators/biosynthesis
14.
PLoS One ; 8(5): e64374, 2013.
Article in English | MEDLINE | ID: mdl-23741322

ABSTRACT

Availability of large amounts of in vitro generated ß-cells may support replacement therapy in diabetes. However, methods to obtain ß-cells from stem/progenitor cells are limited by inefficient endocrine differentiation. We have recently shown that the ghrelin gene product obestatin displays beneficial effects on pancreatic ß-cell survival and function. Obestatin prevents ß-cell apoptosis, preserves ß-cell mass and stimulates insulin secretion in vitro and in vivo, in both normal and diabetic conditions. In the present study, we investigated whether obestatin may promote in vitro ß-cell generation from mouse pancreatic islet-derived precursor cells. Treatment of cultured islets of Langerhans with obestatin (i) enriched cells expressing the mesenchymal/neuronal marker nestin, which is associated with pancreatic precursors; (ii) increased cell survival and reduced apoptosis during precursor selection; (iii) promoted the generation of islet-like cell clusters (ICCs) with increased insulin gene expression and C-peptide secretion. Furthermore, obestatin modulated the expression of fibroblast growth factor receptors (FGFRs), Notch receptors and neurogenin 3 (Ngn3) during islet-derived precursor cell selection and endocrine differentiation. These results indicate that obestatin improves the generation of functional ß-cells/ICCs in vitro, suggesting implications for cell-based replacement therapy in diabetes. Moreover, obestatin may play a role in regulating pathways involved in pancreas development and regeneration.


Subject(s)
Gene Expression Regulation, Developmental/drug effects , Islets of Langerhans/drug effects , Peptide Hormones/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , C-Peptide/biosynthesis , C-Peptide/metabolism , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Insulin/biosynthesis , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin/genetics , Nestin/metabolism , Peptide Hormones/metabolism , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction
15.
Diabet Med ; 30(11): 1342-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23659458

ABSTRACT

AIMS: To determine the prevalence and clinical characteristics of absolute insulin deficiency in long-standing Type 2 diabetes, using a strategy based on home urinary C-peptide creatinine ratio measurement. METHODS: We assessed the urinary C-peptide creatinine ratios, from urine samples taken at home 2 h after the largest meal of the day, in 191 insulin-treated subjects with Type 2 diabetes (diagnosis age ≥45 years, no insulin in the first year). If the initial urinary C-peptide creatinine ratio was ≤0.2 nmol/mmol (representing absolute insulin deficiency), the assessment was repeated. A standardized mixed-meal tolerance test with 90-min stimulated serum C-peptide measurement was performed in nine subjects with a urinary C-peptide creatinine ratio ≤ 0.2 nmol/mmol (and in nine controls with a urinary C-peptide creatinine ratio >0.2 nmol/mmol) to confirm absolute insulin deficiency. RESULTS: A total of 2.7% of participants had absolute insulin deficiency confirmed by a mixed-meal tolerance test. They were identified initially using urinary C-peptide creatinine ratio: 11/191 subjects (5.8%) had two consistent urinary C-peptide creatinine ratios ≤ 0.2 nmol/mmol; 9 of these 11 subjects completed a mixed-meal tolerance test and had a median stimulated serum C-peptide of 0.18 nmol/l. Five of these 9 had stimulated serum C-peptide <0.2 nmol/l and 9/9 subjects with urinary C-peptide creatinine ratio >0.2 had endogenous insulin secretion confirmed by the mixed-meal tolerance test. Compared with subjects with a urinary C-peptide creatinine ratio >0.2 nmol/mmol, those with confirmed absolute insulin deficiency had a shorter time to insulin treatment (median 2.5 vs. 6 years, P=0.005) and lower BMI (25.1 vs. 29.1 kg/m(2) , P=0.04). Two out of the five patients with absolute insulin deficiency were glutamic acid decarboxylase autoantibody-positive. CONCLUSIONS: Absolute insulin deficiency may occur in long-standing Type 2 diabetes, and cannot be reliably predicted by clinical features or autoantibodies. Absolute insulin deficiency in Type 2 diabetes may increase the risk of hypoglycaemia and ketoacidosis, as in Type 1 diabetes. Its recognition should help guide treatment, education and management. The urinary C-peptide creatinine ratio is a practical non-invasive method to aid detection of absolute insulin deficiency, with a urinary C-peptide creatinine ratio > 0.2 nmol/mmol being a reliable indicator of retained endogenous insulin secretion.


Subject(s)
C-Peptide/biosynthesis , C-Peptide/urine , Creatinine/urine , Diabetes Mellitus, Type 2/urine , Insulin/deficiency , Aged , Case-Control Studies , Female , Humans , Male , Middle Aged
17.
Nat Genet ; 44(4): 406-12, S1, 2012 Mar 11.
Article in English | MEDLINE | ID: mdl-22406641

ABSTRACT

Restoration of regulated insulin secretion is the ultimate goal of therapy for type 1 diabetes. Here, we show that, unexpectedly, somatic ablation of Foxo1 in Neurog3(+) enteroendocrine progenitor cells gives rise to gut insulin-positive (Ins(+)) cells that express markers of mature ß cells and secrete bioactive insulin as well as C-peptide in response to glucose and sulfonylureas. Lineage tracing experiments showed that gut Ins(+) cells arise cell autonomously from Foxo1-deficient cells. Inducible Foxo1 ablation in adult mice also resulted in the generation of gut Ins(+) cells. Following ablation by the ß-cell toxin streptozotocin, gut Ins(+) cells regenerate and produce insulin, reversing hyperglycemia in mice. The data indicate that Neurog3(+) enteroendocrine progenitors require active Foxo1 to prevent differentiation into Ins(+) cells. Foxo1 ablation in gut epithelium may provide an approach to restore insulin production in type 1 diabetes.


Subject(s)
Enteroendocrine Cells/metabolism , Forkhead Transcription Factors/physiology , Insulin/biosynthesis , Neuroendocrine Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , C-Peptide/biosynthesis , C-Peptide/metabolism , Cell Differentiation , Diabetes Mellitus, Experimental/metabolism , Enteroendocrine Cells/cytology , Forkhead Box Protein O1 , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Gastrointestinal Tract/cytology , Gastrointestinal Tract/metabolism , Glucose/pharmacology , Hyperglycemia/therapy , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Mice , Mice, Transgenic , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Stem Cells/cytology , Streptozocin/pharmacology , Sulfonylurea Compounds/pharmacology , Wnt Signaling Pathway
18.
Transplant Proc ; 43(9): 3231-4, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22099764

ABSTRACT

OBJECTIVE: To evaluate the effect of exenatide on gastric emptying and long-term metabolic control. METHODS: Ten islet allograft recipients treated with exenatide up to 4 years. Data from a mixed meal test with (MMT+) versus without (MMT-) administration of exenatide before boost ingestion were analyzed at 6, 12, 24, 36, or 48 months after initiation of exenatide treatment. None of the subjects were symptomatic for gastroparesis before or during the study. The c-peptide, acetaminophen absorption and glucose responses to MMT were analyzed by Student t test and analysis of variance. RESULTS: Average exenatide dose was 12.75 ± 9.46 µg/dL. The MMT subjects included two groups those with acetaminophen peak ≤120 minutes ("good gastric emptying; n = 4") versus those with an acetaminophen peak ≥180 minutes ("delayed gastric emptying"). Among the MMT+, acetaminophen absorption was the same in both groups (P = .27). Up to 48 months exenatide delayed time to peak of glucose, c-peptide, and acetaminophen as well as suppressed the glucagon response to MMT mean peak: 70.89 ± 12.45 versus 43.24 ± 4.67. The mean values of c-peptide and glucose responses to MMT were not significantly different. CONCLUSIONS: Long-term exenatide administration up to 4 years was safe in islet transplant recipients, even in the presence of delayed gastric emptying. The effects of exenatide were acute and reversible when the agent was withdrawn. The main difficulty with the use of exenatide in islet transplant subjects is their poor tolerability, although the physiological effects are clearly evident even at low doses. Approximately 63% of total subjects under exenatide treatment discontinued the drug due to nausea and vomiting. The use of new GLP1 analogs with longer half lives and fewer side effects may help to attain higher GLP1 levels, therefore improving islet function and survival.


Subject(s)
Cell Transplantation/methods , Gastric Emptying , Graft Survival/drug effects , Islets of Langerhans Transplantation/methods , Islets of Langerhans/cytology , Peptides/pharmacology , Venoms/pharmacology , Acetaminophen/pharmacokinetics , C-Peptide/biosynthesis , Cohort Studies , Dose-Response Relationship, Drug , Exenatide , Glucose/metabolism , Humans , Hypoglycemic Agents/pharmacology , Insulin/metabolism
19.
Methods Mol Biol ; 768: 3-11, 2011.
Article in English | MEDLINE | ID: mdl-21805235

ABSTRACT

Studies of the biosynthesis of insulin in a human insulinoma beginning in 1965 provided the first evidence for a precursor of insulin, the first such prohormone to be identified. Further studies with isolated rat islets then confirmed that the precursor became labeled more rapidly than insulin and later was converted to insulin by a proteolytic processing system located mainly within the secretory granules of the beta cell and was then stored or secreted. The precursor was designated "proinsulin" in 1967 and was isolated and sequenced from beef and pork sources. These structural studies confirmed that the precursor was a single polypeptide chain which began with the B chain of insulin, continued through a connecting segment of 30-35 amino acids and terminated with the A chain. Paired basic residues were identified at the sites of excision of the C-peptide. Human proinsulin and C-peptide were then similarly obtained and sequenced. The human C-peptide assay was developed and provided a useful tool for measuring insulin levels indirectly in diabetics treated with insulin. The discovery of other precursor proteins for a variety of peptide hormones, neuropeptides, or plasma proteins then followed, with all having mainly dibasic cleavage sites for processing. The subsequent discovery of a similar biosynthetic pathway in yeast led to the identification of eukaryotic families of specialized processing subtilisin-like endopeptidases coupled with carboxypeptidase B-like exopeptidases. Most neuroendocrine peptides are processed by two specialized members of this family - PC2 and/or PC1/3 - followed by carboxypeptidase E (CPE). This brief report concentrates mainly on the role of insulin biosynthesis in providing a useful early paradigm of precursor processing in the secretory pathway.


Subject(s)
Insulin/biosynthesis , Proprotein Convertase 1/metabolism , Proprotein Convertase 2/metabolism , Protein Precursors/metabolism , Protein Processing, Post-Translational/physiology , Animals , C-Peptide/biosynthesis , Carboxypeptidase H/metabolism , Cattle , History, 20th Century , Humans , Insulin-Secreting Cells/enzymology , Insulinoma/metabolism , Neuropeptides/metabolism , Proinsulin/biosynthesis , Protein Precursors/history , Rats , Saccharomyces cerevisiae , Swine
20.
Stem Cells Dev ; 20(7): 1223-31, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20942606

ABSTRACT

Islet cell replacement represents the most promising approach for the treatment of type I diabetes. However, it is limited by a shortage of pancreas donors. Here, we report that human amniotic fluid-derived stem cells (hAFSCs) can be induced to differentiate into functional insulin-producing cells by knocking down neuronal restrictive silencing factor (NRSF). In this study, lentiviral vectors were used to deliver small interference NRSF (siNRSF) RNA into hAFSCs. After infection with lentivirus containing siNRSF, hAFSCs were successfully induced to differentiate into insulin-producing cells. The differentiated siNRSF-hAFSCs expressed genes specific for islet cells, such as Pdx1, Hnf4α, Isl-1, Nkx6.1, Insulin, and Glut2. These cells also produced and released C-peptide in a glucose-responsive manner. These findings indicated that hAFSCs could be induced to differentiate into insulin-producing ß-like cells by NRSF silencing.


Subject(s)
Amniotic Fluid/cytology , C-Peptide/biosynthesis , Cell Differentiation , Insulin-Secreting Cells/metabolism , Repressor Proteins/metabolism , Stem Cells/cytology , Cells, Cultured , Down-Regulation , Female , Fluorescent Antibody Technique , Gene Expression Regulation , Genetic Vectors , Humans , Insulin-Secreting Cells/cytology , Lentivirus/metabolism , Pregnancy , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...