Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Nature ; 567(7746): 43-48, 2019 03.
Article in English | MEDLINE | ID: mdl-30760930

ABSTRACT

Cell-identity switches, in which terminally differentiated cells are converted into different cell types when stressed, represent a widespread regenerative strategy in animals, yet they are poorly documented in mammals. In mice, some glucagon-producing pancreatic α-cells and somatostatin-producing δ-cells become insulin-expressing cells after the ablation of insulin-secreting ß-cells, thus promoting diabetes recovery. Whether human islets also display this plasticity, especially in diabetic conditions, remains unknown. Here we show that islet non-ß-cells, namely α-cells and pancreatic polypeptide (PPY)-producing γ-cells, obtained from deceased non-diabetic or diabetic human donors, can be lineage-traced and reprogrammed by the transcription factors PDX1 and MAFA to produce and secrete insulin in response to glucose. When transplanted into diabetic mice, converted human α-cells reverse diabetes and continue to produce insulin even after six months. Notably, insulin-producing α-cells maintain expression of α-cell markers, as seen by deep transcriptomic and proteomic characterization. These observations provide conceptual evidence and a molecular framework for a mechanistic understanding of in situ cell plasticity as a treatment for diabetes and other degenerative diseases.


Subject(s)
Diabetes Mellitus/pathology , Diabetes Mellitus/therapy , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/metabolism , Glucose/metabolism , Insulin/metabolism , Islets of Langerhans/pathology , Animals , Biomarkers/analysis , Cell Lineage/drug effects , Cellular Reprogramming/drug effects , Diabetes Mellitus/immunology , Diabetes Mellitus/metabolism , Disease Models, Animal , Female , Glucagon/metabolism , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/transplantation , Glucose/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Islets of Langerhans/drug effects , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Maf Transcription Factors, Large/genetics , Maf Transcription Factors, Large/metabolism , Male , Mice , Organ Specificity/drug effects , Pancreatic Polypeptide/metabolism , Pancreatic Polypeptide-Secreting Cells/cytology , Pancreatic Polypeptide-Secreting Cells/drug effects , Pancreatic Polypeptide-Secreting Cells/metabolism , Proteomics , Sequence Analysis, RNA , Trans-Activators/genetics , Trans-Activators/metabolism , Transcriptome , Transduction, Genetic
2.
Pancreas ; 46(6): 820-824, 2017 07.
Article in English | MEDLINE | ID: mdl-28609372

ABSTRACT

A partial pancreaticogastrodudenectomy was performed on a 66-year old man with type 2 diabetes mellitus because of an invasive, moderately differentiated adenocarcinoma in the head of the pancreas. In the adjacent grossly normal tissue of the uncinate process, there was a massive proliferation of pancreatic polypeptide (PP) cells confined to this region and showed invasive pattern. Strikingly, in the heaped area of his duodenum, there was a strikingly large number of PP, glucagon, a few insulin cells in a mini-islet-like patterns composed of glucagon and insulin cells. Among the etiological factors, the possible long-lasting effects of the GLP-1 analog, with which the patient was treated, are discussed. This is the first report in the literature of both the coexistence of a pancreatic adenocarcinoma and invasive PPoma and the occurrence of PP and insulin cells in human duodenal mucosa.


Subject(s)
Adenocarcinoma/complications , Cell Proliferation/drug effects , Diabetes Mellitus, Type 2/drug therapy , Duodenum/drug effects , Hypoglycemic Agents/adverse effects , Liraglutide/adverse effects , Pancreas/drug effects , Pancreatic Neoplasms/complications , Pancreatic Polypeptide-Secreting Cells/drug effects , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Aged , Chemotherapy, Adjuvant , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Duodenum/metabolism , Duodenum/pathology , Duodenum/surgery , Fatal Outcome , Humans , Hyperplasia , Immunohistochemistry , Male , Pancreas/metabolism , Pancreas/pathology , Pancreas/surgery , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Pancreatic Polypeptide-Secreting Cells/metabolism , Pancreaticoduodenectomy , Risk Factors , Time Factors , Treatment Outcome
3.
Sci Rep ; 7(1): 2505, 2017 05 31.
Article in English | MEDLINE | ID: mdl-28566744

ABSTRACT

Insulin-producing cells (IPCs) derived from a patient's own stem cells offer great potential for autologous transplantation in diabetic patients. However, the limited survival of engrafted cells remains a bottleneck in the application of this strategy. The present study aimed to investigate whether nanoparticle-based magnetic resonance (MR) tracking can be used to detect the loss of grafted stem cell-derived IPCs in a sensitive and timely manner in a diabetic monkey model. Pancreatic progenitor cells (PPCs) were isolated from diabetic monkeys and labeled with superparamagnetic iron oxide nanoparticles (SPIONs). The SPION-labeled cells presented as hypointense signals on MR imaging (MRI). The labeling procedure did not affect the viability or IPC differentiation of PPCs. Importantly, the total area of the hypointense signal caused by SPION-labeled IPCs on liver MRI decreased before the decline in C-peptide levels after autotransplantation. Histological analysis revealed no detectable immune response to the grafts and many surviving insulin- and Prussian blue-positive cell clusters on liver sections at one year post-transplantation. Collectively, this study demonstrates that SPIO nanoparticles can be used to label stem cells for noninvasive, sensitive, longitudinal monitoring of stem cell-derived IPCs in large animal models using a conventional MR imager.


Subject(s)
Cell Tracking/methods , Diabetes Mellitus, Type 1/diagnostic imaging , Diabetes Mellitus, Type 1/therapy , Magnetic Resonance Imaging , Pancreatic Polypeptide-Secreting Cells/cytology , Animals , C-Peptide/blood , Cell Differentiation , Contrast Media/administration & dosage , Contrast Media/chemistry , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Ferric Compounds/administration & dosage , Ferric Compounds/chemistry , Humans , Macaca fascicularis , Magnetite Nanoparticles/administration & dosage , Magnetite Nanoparticles/chemistry , Mesenchymal Stem Cell Transplantation/methods , Pancreatic Polypeptide-Secreting Cells/drug effects , Transplantation, Autologous
4.
Diabetes Obes Metab ; 19(9): 1267-1275, 2017 09.
Article in English | MEDLINE | ID: mdl-28345790

ABSTRACT

AIMS: Ghrelin is a gastric-derived hormone that stimulates growth hormone (GH) secretion and has a multi-faceted role in the regulation of energy homeostasis, including glucose metabolism. Circulating ghrelin concentrations are modulated in response to nutritional status, but responses to ghrelin in altered metabolic states are poorly understood. We investigated the metabolic effects of ghrelin in obesity and early after Roux-en-Y gastric bypass (RYGB). MATERIALS AND METHODS: We assessed central and peripheral metabolic responses to acyl ghrelin infusion (1 pmol kg-1 min-1 ) in healthy, lean subjects (n = 9) and non-diabetic, obese subjects (n = 9) before and 2 weeks after RYGB. Central responses were assessed by GH and pancreatic polypeptide (surrogate for vagal activity) secretion. Peripheral responses were assessed by hepatic and skeletal muscle insulin sensitivity during a hyperinsulinaemic-euglycaemic clamp. RESULTS: Ghrelin-stimulated GH secretion was attenuated in obese subjects, but was restored by RYGB to a response similar to that of lean subjects. The heightened pancreatic polypeptide response to ghrelin infusion in the obese was attenuated after RYGB. Hepatic glucose production and hepatic insulin sensitivity were not altered by ghrelin infusion in RYGB subjects. Skeletal muscle insulin sensitivity was impaired to a similar degree in lean, obese and post-RYGB individuals in response to ghrelin infusion. CONCLUSIONS: These data suggest that obesity is characterized by abnormal central, but not peripheral, responsiveness to ghrelin that can be restored early after RYGB before significant weight loss. Further work is necessary to fully elucidate the role of ghrelin in the metabolic changes that occur in obesity and following RYGB.


Subject(s)
Anti-Obesity Agents/therapeutic use , Gastric Bypass , Ghrelin/therapeutic use , Human Growth Hormone/agonists , Insulin Resistance , Obesity, Morbid/drug therapy , Obesity, Morbid/surgery , Acylation , Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/adverse effects , Anti-Obesity Agents/chemistry , Cohort Studies , Combined Modality Therapy/adverse effects , Cross-Over Studies , Energy Metabolism/drug effects , Ghrelin/administration & dosage , Ghrelin/adverse effects , Ghrelin/chemistry , Gluconeogenesis/drug effects , Glucose Clamp Technique , Human Growth Hormone/blood , Human Growth Hormone/metabolism , Humans , Infusions, Intravenous , Liver/drug effects , Liver/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Obesity, Morbid/blood , Obesity, Morbid/metabolism , Pancreatic Polypeptide/agonists , Pancreatic Polypeptide/blood , Pancreatic Polypeptide/metabolism , Pancreatic Polypeptide-Secreting Cells/drug effects , Pancreatic Polypeptide-Secreting Cells/metabolism , Pituitary Gland, Anterior/drug effects , Pituitary Gland, Anterior/metabolism , Postoperative Care , Preoperative Care , Protein Precursors/agonists , Protein Precursors/blood , Protein Precursors/metabolism , Single-Blind Method
5.
Am J Physiol Endocrinol Metab ; 311(4): E661-E670, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27530231

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) has important actions on whole body metabolic function. GIP and its receptor are also present in the central nervous system and have been linked to neurotrophic actions. Metabolic effects of central nervous system GIP signaling have not been reported. We investigated whether centrally administered GIP could increase peripheral plasma GIP concentrations and influence the metabolic response to a mixed macronutrient meal in nonhuman primates. An infusion and sampling system was developed to enable continuous intracerebroventricular (ICV) infusions with serial venous sampling in conscious nonhuman primates. Male baboons (Papio sp.) that were healthy and had normal body weights (28.9 ± 2.1 kg) were studied (n = 3). Animals were randomized to receive continuous ICV infusions of GIP (20 pmol·kg-1·h-1) or vehicle before and over the course of a 300-min mixed meal test (15 kcal/kg, 1.5g glucose/kg) on two occasions. A significant increase in plasma GIP concentration was observed under ICV GIP infusion (66.5 ± 8.0 vs. 680.6 ± 412.8 pg/ml, P = 0.04) before administration of the mixed meal. Increases in postprandial, but not fasted, insulin (P = 0.01) and pancreatic polypeptide (P = 0.04) were also observed under ICV GIP. Effects of ICV GIP on fasted or postprandial glucagon, glucose, triglyceride, and free fatty acids were not observed. Our data demonstrate that central GIP signaling can promote increased plasma GIP concentrations independent of nutrient stimulation and increase insulin and pancreatic polypeptide responses to a mixed meal.


Subject(s)
Gastric Inhibitory Polypeptide/metabolism , Insulin/metabolism , Pancreatic Polypeptide-Secreting Cells/drug effects , Pancreatic Polypeptide/metabolism , Papio/metabolism , Animals , Blood Glucose/metabolism , Eating , Gastric Inhibitory Polypeptide/genetics , Infusions, Intraventricular , Male , Postprandial Period/drug effects , Species Specificity , Stereotaxic Techniques
6.
PLoS One ; 10(12): e0144597, 2015.
Article in English | MEDLINE | ID: mdl-26658466

ABSTRACT

The transcription factor Pax6 is an important regulator of development and cell differentiation in various organs. Thus, Pax6 was shown to promote neural development in the cerebral cortex and spinal cord, and to control pancreatic endocrine cell genesis. However, the role of Pax6 in distinct endocrine cells of the adult pancreas has not been addressed. We report the conditional inactivation of Pax6 in insulin and glucagon producing cells of the adult mouse pancreas. In the absence of Pax6, beta- and alpha-cells lose their molecular maturation characteristics. Our findings provide strong evidence that Pax6 is responsible for the maturation of beta-, and alpha-cells, but not of delta-, and PP-cells. Moreover, lineage-tracing experiments demonstrate that Pax6-deficient beta- and alpha-cells are shunted towards ghrelin marked cells, sustaining the idea that ghrelin may represent a marker for endocrine cell maturation.


Subject(s)
Eye Proteins/genetics , Ghrelin/genetics , Glucagon-Secreting Cells/metabolism , Homeodomain Proteins/genetics , Insulin-Secreting Cells/metabolism , Paired Box Transcription Factors/genetics , Pancreatic Polypeptide-Secreting Cells/metabolism , Repressor Proteins/genetics , Somatostatin-Secreting Cells/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Differentiation , Cell Lineage/drug effects , Cell Lineage/genetics , Crosses, Genetic , Eye Proteins/metabolism , Female , Gene Expression Regulation, Developmental , Genes, Reporter , Ghrelin/metabolism , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/drug effects , Homeodomain Proteins/metabolism , Insulin/genetics , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Integrases/genetics , Integrases/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Knockout , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Pancreatic Polypeptide-Secreting Cells/cytology , Pancreatic Polypeptide-Secreting Cells/drug effects , Repressor Proteins/metabolism , Signal Transduction , Somatostatin-Secreting Cells/cytology , Somatostatin-Secreting Cells/drug effects , Tamoxifen/pharmacology
7.
Mol Cell Biol ; 28(20): 6373-83, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18710955

ABSTRACT

During pancreas development, transcription factors play critical roles in exocrine and endocrine differentiation. Transcriptional regulation in eukaryotes occurs within chromatin and is influenced by posttranslational histone modifications (e.g., acetylation) involving histone deacetylases (HDACs). Here, we show that HDAC expression and activity are developmentally regulated in the embryonic rat pancreas. We discovered that pancreatic treatment with different HDAC inhibitors (HDACi) modified the timing and determination of pancreatic cell fate. HDACi modified the exocrine lineage via abolition and enhancement of acinar and ductal differentiation, respectively. Importantly, HDACi treatment promoted the NGN3 proendocrine lineage, leading to an increased pool of endocrine progenitors and modified endocrine subtype lineage choices. Interestingly, treatments with trichostatin A and sodium butyrate, two inhibitors of both class I and class II HDACs, enhanced the pool of beta cells. These results highlight the roles of HDACs at key points in exocrine and endocrine differentiation. They show the powerful use of HDACi to switch pancreatic cell determination and amplify specific cellular subtypes, with potential applications in cell replacement therapies in diabetes.


Subject(s)
Cell Lineage/drug effects , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Islets of Langerhans/cytology , Pancreas/drug effects , Pancreas/enzymology , Stem Cells/cytology , Animals , Apoptosis/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Female , Hydroxamic Acids/pharmacology , Islets of Langerhans/drug effects , Islets of Langerhans/enzymology , Nerve Tissue Proteins/metabolism , Pancreas/cytology , Pancreas/embryology , Pancreas, Exocrine/cytology , Pancreas, Exocrine/drug effects , Pancreas, Exocrine/enzymology , Pancreatic Ducts/cytology , Pancreatic Ducts/drug effects , Pancreatic Ducts/enzymology , Pancreatic Polypeptide-Secreting Cells/cytology , Pancreatic Polypeptide-Secreting Cells/drug effects , Pancreatic Polypeptide-Secreting Cells/enzymology , Rats , Rats, Wistar , Stem Cells/drug effects , Valproic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...