29 results on '"Björn Tyrberg"'
Search Results
2. Opposing roles of the entero-pancreatic hormone urocortin-3 in glucose metabolism in rats
- Author
-
Kaare V. Grunddal, Samuel A. J. Trammell, Cecilie Bæch-Laursen, Daniel B. Andersen, Stella F. S. Xu, Helle Andersen, Matthew P. Gillum, Seyed M. Ghiasi, Ivana Novak, Björn Tyrberg, Chien Li, Mette M. Rosenkilde, Bolette Hartmann, Jens J. Holst, and Rune E. Kuhre
- Subjects
Blood Glucose ,Male ,Somatostatin/metabolism ,Blood Glucose/metabolism ,Insulin/metabolism ,Endocrinology, Diabetes and Metabolism ,Glucagon ,Rats ,Islets of Langerhans ,Glucose ,Internal Medicine ,Urocortins/metabolism ,Glucose/metabolism ,Animals ,Insulin ,Glucagon/metabolism ,Islets of Langerhans/metabolism ,Somatostatin ,Urocortins - Abstract
Aim/hypothesis Urocortin-3 (UCN3) is a glucoregulatory peptide produced in the gut and pancreatic islets. The aim of this study was to clarify the acute effects of UCN3 on glucose regulation following an oral glucose challenge and to investigate the mechanisms involved. Methods We studied the effect of UCN3 on blood glucose, gastric emptying, glucose absorption and secretion of gut and pancreatic hormones in male rats. To supplement these physiological studies, we mapped the expression of UCN3 and the UCN3-sensitive receptor, type 2 corticotropin-releasing factor receptor (CRHR2), by means of fluorescence in situ hybridisation and by gene expression analysis. Results In rats, s.c. administration of UCN3 strongly inhibited gastric emptying and glucose absorption after oral administration of glucose. Direct inhibition of gastrointestinal motility may be responsible because UCN3’s cognate receptor, CRHR2, was detected in gastric submucosal plexus and in interstitial cells of Cajal. Despite inhibited glucose absorption, post-challenge blood glucose levels matched those of rats given vehicle in the low-dose UCN3 group, because UCN3 concomitantly inhibited insulin secretion. Higher UCN3 doses did not further inhibit gastric emptying, but the insulin inhibition progressed resulting in elevated post-challenge glucose and lipolysis. Incretin hormones and somatostatin (SST) secretion from isolated perfused rat small intestine was unaffected by UCN3 infusion; however, UCN3 infusion stimulated secretion of somatostatin from delta cells in the isolated perfused rat pancreas which, unlike alpha cells and beta cells, expressed Crhr2. Conversely, acute antagonism of CRHR2 signalling increased insulin secretion by reducing SST signalling. Consistent with these observations, acute drug-induced inhibition of CRHR2 signalling improved glucose tolerance in rats to a similar degree as administration of glucagon-like peptide-1. UCN3 also powerfully inhibited glucagon secretion from isolated perfused rat pancreas (perfused with 3.5 mmol/l glucose) in a SST-dependent manner, suggesting that UCN3 may be involved in glucose-induced inhibition of glucagon secretion. Conclusions/interpretation Our combined data indicate that UCN3 is an important glucoregulatory hormone that acts through regulation of gastrointestinal and pancreatic functions. Graphical abstract
- Published
- 2021
- Full Text
- View/download PDF
3. MCPIP1 is a novel link between diabetogenic conditions and impaired insulin secretory capacity
- Author
-
Tenna Holgersen Bryde, Anne Jörns, Alessia Dunst, Yadi Tang, Ewa Gurgul-Convey, Lorella Marselli, Karolina Tyka, Miriam Cnop, Michal Marzec, Anna Walentinsson, Björn Tyrberg, and Ilir Mehmeti
- Subjects
0301 basic medicine ,Beta-cells ,medicine.medical_specialty ,RNase P ,medicine.medical_treatment ,Glucose uptake ,Regulator ,030209 endocrinology & metabolism ,Inflammation ,FOXO1 ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Ribonucleases ,Internal medicine ,Insulin-Secreting Cells ,Insulin Secretion ,medicine ,Diabetes Mellitus ,Animals ,Humans ,Insulin ,RNA, Messenger ,Molecular Biology ,3' Untranslated Regions ,Diabetes ,Human islets ,Insulin production and secretion ,MCPIP1 ,Chemistry ,Forkhead Box Protein O1 ,Wild type ,Sciences bio-médicales et agricoles ,Rats ,030104 developmental biology ,Endocrinology ,Glucose ,Molecular Medicine ,PDX1 ,Cytokines ,Calcium ,medicine.symptom ,Transcription Factors - Abstract
SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2021
4. Endoplasmic Reticulum Chaperone Glucose-Regulated Protein 94 Is Essential for Proinsulin Handling
- Author
-
Seyed Mojtaba Ghiasi, Sólrun Petersen, Kristian Klindt, Oana Cheta, Thomas Mandrup-Poulsen, Marcelo J. Perone, Michal Marzec, Peter Arvan, Anders Tengholm, Sophie Emilie Bresson, Caroline Hede Andersen, Clara Prats, Sebastian Barg, Mingyu Yang, Muhammad Saad Khilji, Leena Haataja, Muhmmad Omar-Hmeadi, Björn Tyrberg, Celina Pihl, and Tina Dahlby
- Subjects
0301 basic medicine ,endocrine system ,Protein Folding ,endocrine system diseases ,Glucose-regulated protein ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,030209 endocrinology & metabolism ,Apoptosis ,GRP94 ,Endoplasmic Reticulum ,Exocytosis ,03 medical and health sciences ,eIF-2 Kinase ,0302 clinical medicine ,Cell Line, Tumor ,Insulin-Secreting Cells ,Insulin Secretion ,Internal Medicine ,medicine ,Animals ,Humans ,Insulin ,HSP70 Heat-Shock Proteins ,Protein kinase A ,Proinsulin ,biology ,Chemistry ,Kinase ,Endoplasmic reticulum ,Membrane Proteins ,purl.org/becyt/ford/3.1 [https] ,Endoplasmic Reticulum Stress ,Cell biology ,Rats ,030104 developmental biology ,Islet Studies ,Diabetes Mellitus, Type 2 ,Chaperone (protein) ,biology.protein ,Chaperone binding ,purl.org/becyt/ford/3 [https] ,GP96 ,hormones, hormone substitutes, and hormone antagonists - Abstract
Although endoplasmic reticulum (ER) chaperone binding to mutant proinsulin has been reported, the role of protein chaperones in the handling of wild-type proinsulin is underinvestigated. Here, we have explored the importance of glucose-regulated protein 94 (GRP94), a prominent ER chaperone known to fold insulin-like growth factors, in proinsulin handling within b-cells. We found that GRP94 coimmunoprecipitated with proinsulin and that inhibition of GRP94 function and/or expression reduced glucose-dependent insulin secretion, shortened proinsulin half-life, and lowered intracellular proinsulin and insulin levels. This phenotype was accompanied by post-ER proinsulin misprocessing and higher numbers of enlarged insulin granules that contained amorphic material with reduced immunogold staining for mature insulin. Insulin granule exocytosis was accelerated twofold, but the secreted insulin had diminished bioactivity. Moreover, GRP94 knockdown or knockout in b-cells selectively activated protein kinase R–like endoplasmic reticulum kinase (PERK), without increasing apoptosis levels. Finally, GRP94 mRNA was overexpressed in islets from patients with type 2 diabetes. We conclude that GRP94 is a chaperone crucial for proinsulin handling and insulin secretion. Fil: Ghiasi, Seyed Mojtaba. Universidad de Copenhagen; Dinamarca Fil: Dahlby, Tina. Universidad de Copenhagen; Dinamarca Fil: Andersen, Caroline Hede. Universidad de Copenhagen; Dinamarca Fil: Haataja, Leena. University of Michigan; Estados Unidos Fil: Petersen, Sólrun. Universidad de Copenhagen; Dinamarca Fil: Omar-Hmeadi, Muhmmad. Uppsala Universitet; Suecia Fil: Yang, Mingyu. Uppsala Universitet; Suecia Fil: Pihl, Celina. Universidad de Copenhagen; Dinamarca Fil: Bresson, Sophie Emilie. Universidad de Copenhagen; Dinamarca Fil: Khilji, Muhammad Saad. Universidad de Copenhagen; Dinamarca Fil: Klindt, Kristian. Universidad de Copenhagen; Dinamarca Fil: Cheta, Oana. Universidad de Copenhagen; Dinamarca Fil: Perone, Marcelo Javier. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Parque Centenario. Instituto de Investigación en Biomedicina de Buenos Aires - Instituto Partner de la Sociedad Max Planck; Argentina. Universidad de Copenhagen; Dinamarca Fil: Tyrberg, Björn. Astrazeneca. IMED Biotech Unit; Suecia Fil: Prats, Clara. Universidad de Copenhagen; Dinamarca Fil: Barg, Sebastian. Uppsala Universitet; Suecia Fil: Tengholm, Anders. Uppsala Universitet; Suecia Fil: Arvan, Peter. University of Michigan; Estados Unidos Fil: Mandrup-Poulsen, Thomas. Universidad de Copenhagen; Dinamarca Fil: Marzec, Michal Tomasz. Universidad de Copenhagen; Dinamarca
- Published
- 2019
5. The inducible β5i proteasome subunit contributes to proinsulin degradation in GRP94-deficient β-cells and is overexpressed in type 2 diabetes pancreatic islets
- Author
-
Michal Marzec, Sarah J. Richardson, Kristian Klindt, Anna Walentinsson, Marie-Pierre Bousquet, Jette Bach Agergaard, Thomas Mandrup-Poulsen, Noel G. Morgan, Phillip Alexander Keller Andersen, Tenna Holgersen Bryde, Christian Kronborg Nielsen, Tina Dahlby, Dusan Zivkovic, Muhammad Saad Khilji, Celina Pihl, Björn Tyrberg, Sophie Emilie Bresson, Danielle Verstappen, Institut de pharmacologie et de biologie structurale (IPBS), Centre National de la Recherche Scientifique (CNRS)-Université Toulouse III - Paul Sabatier (UT3), and Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées
- Subjects
0301 basic medicine ,medicine.medical_specialty ,endocrine system ,Proteasome Endopeptidase Complex ,Protein Folding ,Physiology ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Protein subunit ,[SDV]Life Sciences [q-bio] ,GRP94 ,Protein degradation ,proinsulin degradation ,restoration of proinsulin ,03 medical and health sciences ,Gene Knockout Techniques ,Islets of Langerhans ,0302 clinical medicine ,Physiology (medical) ,Internal medicine ,Insulin-Secreting Cells ,Insulin Secretion ,medicine ,Animals ,Humans ,Proinsulin ,Membrane Glycoproteins ,biology ,Chemistry ,Pancreatic islets ,Insulin ,Endoplasmic reticulum ,Endoplasmic Reticulum-Associated Degradation ,Middle Aged ,β5i ,Endoplasmic Reticulum Stress ,Cell biology ,Rats ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,proteasome ,Proteasome ,Diabetes Mellitus, Type 2 ,030220 oncology & carcinogenesis ,Chaperone (protein) ,biology.protein ,Female - Abstract
Proinsulin is a misfolding-prone protein, and its efficient breakdown is critical when β-cells are confronted with high-insulin biosynthetic demands, to prevent endoplasmic reticulum stress, a key trigger of secretory dysfunction and, if uncompensated, apoptosis. Proinsulin degradation is thought to be performed by the constitutively expressed standard proteasome, while the roles of other proteasomes are unknown. We recently demonstrated that deficiency of the proinsulin chaperone glucose-regulated protein 94 (GRP94) causes impaired proinsulin handling and defective insulin secretion associated with a compensated endoplasmic reticulum stress response. Taking advantage of this model of restricted folding capacity, we investigated the role of different proteasomes in proinsulin degradation, reasoning that insulin secretory dynamics require an inducible protein degradation system. We show that the expression of only one enzymatically active proteasome subunit, namely, the inducible β5i-subunit, was increased in GRP94 CRISPR/Cas9 knockout (KO) cells. Additionally, the level of β5i-containing intermediate proteasomes was significantly increased in these cells, as was β5i-related chymotrypsin-like activity. Moreover, proinsulin levels were restored in GRP94 KO upon β5i small interfering RNA-mediated knockdown. Finally, the fraction of β-cells expressing the β5i-subunit is increased in human islets from type 2 diabetes patients. We conclude that β5i is an inducible proteasome subunit dedicated to the degradation of mishandled proinsulin.
- Published
- 2020
- Full Text
- View/download PDF
6. Inhibition of the prostaglandin D-2-GPR44/DP2 axis improves human islet survival and function
- Author
-
Erik Ryberg, Simen W. Schive, David M. Smith, Shadab Abadpour, Stanko Skrtic, Tina Rydén-Bergsten, Maria Sörhede Winzell, Björn Tyrberg, Olle Korsgren, Hanne Scholz, Charlotte Wennberg Huldt, and Peter Gennemark
- Subjects
0301 basic medicine ,medicine.medical_specialty ,endocrine system ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,Endocrinology and Diabetes ,Proinflammatory cytokine ,03 medical and health sciences ,chemistry.chemical_compound ,Prostaglandin D-2 ,0302 clinical medicine ,Downregulation and upregulation ,Diabetes mellitus ,Internal medicine ,Internal Medicine ,medicine ,DP2 ,GPR44 ,Human islets ,geography ,geography.geographical_feature_category ,Chemistry ,medicine.disease ,Islet ,Transplantation ,Islet apoptosis ,030104 developmental biology ,Endocrinology ,Islet survival rate ,Endokrinologi och diabetes ,Hepatocyte growth factor ,Blood sugar regulation ,Prostaglandin D2 ,medicine.drug - Abstract
Aims/hypothesis Inflammatory signals and increased prostaglandin synthesis play a role during the development of diabetes. The prostaglandin D2 (PGD2) receptor, GPR44/DP2, is highly expressed in human islets and activation of the pathway results in impaired insulin secretion. The role of GPR44 activation on islet function and survival rate during chronic hyperglycaemic conditions is not known. In this study, we investigate GPR44 inhibition by using a selective GPR44 antagonist (AZ8154) in human islets both in vitro and in vivo in diabetic mice transplanted with human islets. Methods Human islets were exposed to PGD2 or proinflammatory cytokines in vitro to investigate the effect of GPR44 inhibition on islet survival rate. In addition, the molecular mechanisms of GPR44 inhibition were investigated in human islets exposed to high concentrations of glucose (HG) and to IL-1β. For the in vivo part of the study, human islets were transplanted under the kidney capsule of immunodeficient diabetic mice and treated with 6, 60 or 100 mg/kg per day of a GPR44 antagonist starting from the transplantation day until day 4 (short-term study) or day 17 (long-term study) post transplantation. IVGTT was performed on mice at day 10 and day 15 post transplantation. After termination of the study, metabolic variables, circulating human proinflammatory cytokines, and hepatocyte growth factor (HGF) were analysed in the grafted human islets. Results PGD2 or proinflammatory cytokines induced apoptosis in human islets whereas GPR44 inhibition reversed this effect. GPR44 inhibition antagonised the reduction in glucose-stimulated insulin secretion induced by HG and IL-1β in human islets. This was accompanied by activation of the Akt–glycogen synthase kinase 3β signalling pathway together with phosphorylation and inactivation of forkhead box O-1and upregulation of pancreatic and duodenal homeobox-1 and HGF. Administration of the GPR44 antagonist for up to 17 days to diabetic mice transplanted with a marginal number of human islets resulted in reduced fasting blood glucose and lower glucose excursions during IVGTT. Improved glucose regulation was supported by increased human C-peptide levels compared with the vehicle group at day 4 and throughout the treatment period. GPR44 inhibition reduced plasma levels of TNF-α and growth-regulated oncogene-α/chemokine (C-X-C motif) ligand 1 and increased the levels of HGF in human islets. Conclusions/interpretation Inhibition of GPR44 in human islets has the potential to improve islet function and survival rate under inflammatory and hyperglycaemic stress. This may have implications for better survival rate of islets following transplantation.
- Published
- 2020
7. No direct effect of SGLT2 activity on glucagon secretion
- Author
-
Nicolai J. Wewer Albrechtsen, Fiona M. Gribble, Nils Wierup, Seyed Mojtaba Ghiasi, Lihua Chen, Jens J. Holst, Alexander Aivazidis, Alice E. Adriaenssens, Frank Reimann, Cathrine Ørskov, Thomas Mandrup-Poulsen, Daniel B. Andersen, Björn Tyrberg, and Rune E. Kuhre
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,endocrine system ,Insulin/metabolism ,Somatostatin secretion ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Blotting, Western ,Alpha (ethology) ,030209 endocrinology & metabolism ,Glucagon ,Alpha cell ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Internal medicine ,Internal Medicine ,medicine ,Animals ,Sodium-Glucose Transporter 1/genetics ,Glucagon/metabolism ,Rats, Wistar ,Pancreas/metabolism ,Delta cell ,Somatostatin/metabolism ,Chemistry ,Insulin ,Glucagon secretion ,Immunohistochemistry ,Rats ,Sodium-Glucose Transporter 2/genetics ,030104 developmental biology ,Endocrinology ,Sodium/Glucose Cotransporter 2 ,Female ,Islets of Langerhans/metabolism ,Chickens - Abstract
AIMS/HYPOTHESIS: Sodium-glucose cotransporter (SGLT) 2 inhibitors constitute a new class of glucose-lowering drugs, but they increase glucagon secretion, which may counteract their glucose-lowering effect. Previous studies using static incubation of isolated human islets or the glucagon-secreting cell line α-TC1 suggested that this results from direct inhibition of alpha cell SGLT1/2-activity. The aim of this study was to test whether the effects of SGLT2 on glucagon secretion demonstrated in vitro could be reproduced in a more physiological setting.METHODS: We explored the effect of SGLT2 activity on glucagon secretion using isolated perfused rat pancreas, a physiological model for glucagon secretion. Furthermore, we investigated Slc5a2 (the gene encoding SGLT2) expression in rat islets as well as in mouse and human islets and in mouse and human alpha, beta and delta cells to test for potential inter-species variations. SGLT2 protein content was also investigated in mouse, rat and human islets.RESULTS: Glucagon output decreased three- to fivefold within minutes of shifting from low (3.5 mmol/l) to high (10 mmol/l) glucose (4.0 ± 0.5 pmol/15 min vs 1.3 ± 0.3 pmol/15 min, p CONCLUSIONS/INTERPRETATION: Our combined data show that increased plasma glucagon during SGLT2 inhibitor treatment is unlikely to result from direct inhibition of SGLT2 in alpha cells, but instead may occur downstream of their blood glucose-lowering effects.
- Published
- 2019
- Full Text
- View/download PDF
8. Exploring the insulin secretory properties of the PGD2-GPR44/DP2 axis in vitro and in a randomized phase-1 trial of type 2 diabetes patients
- Author
-
Elaine Watkins, Charlotte Wennberg Huldt, Joanna Parkinson, Erik Ryberg, Volker Schnecke, Eva-Marie Andersson, Hans Ericsson, Björn Tyrberg, Marcus Hompesch, Stanko Skrtic, Tina Rydén-Bergsten, Linda Morrow, Lars Löfgren, M Kjaer, Alexander Aivazidis, Malin A. Broberg, and Maria Sörhede Winzell
- Subjects
0301 basic medicine ,Blood Glucose ,Male ,Indoles ,Physiology ,Prostaglandin ,medicine.medical_treatment ,Receptors, Prostaglandin ,Gene Expression ,Type 2 diabetes ,Acetates ,Biochemistry ,0302 clinical medicine ,Endocrinology ,Insulin Secretion ,Medicine and Health Sciences ,Medicine ,Insulin ,Lipid Hormones ,Receptors, Immunologic ,Multidisciplinary ,geography.geographical_feature_category ,C-Peptide ,Organic Compounds ,Pharmaceutics ,Prostaglandin D2 ,Monosaccharides ,Middle Aged ,Islet ,Type 2 Diabetes ,DNA-Binding Proteins ,Chemistry ,030220 oncology & carcinogenesis ,Physical Sciences ,Female ,Research Article ,medicine.medical_specialty ,Endocrine Disorders ,Science ,Carbohydrates ,Incretin ,Glucagon ,Cell Line ,03 medical and health sciences ,Islets of Langerhans ,Drug Therapy ,Diagnostic Medicine ,Internal medicine ,Diabetes mellitus ,Genetics ,Diabetes Mellitus ,Humans ,Diabetic Endocrinology ,geography ,Endocrine Physiology ,business.industry ,Organic Chemistry ,Antagonist ,Chemical Compounds ,Biology and Life Sciences ,medicine.disease ,Hormones ,030104 developmental biology ,Glucose ,Diabetes Mellitus, Type 2 ,Gene Expression Regulation ,Metabolic control analysis ,Metabolic Disorders ,Glucose Tolerance Tests ,business ,Transcription Factors - Abstract
Aims/hypothesis GPR44 (DP2, PTGDR2, CRTh2) is the receptor for the pro-inflammatory mediator prostaglandin D2 (PGD2) and it is enriched in human islets. In rodent islets, PGD2 is produced in response to glucose, suggesting that the PGD2-GPR44/DP2 axis may play a role in human islet function during hyperglycemia. Consequently, the aim of this work was to elucidate the insulinotropic role of GPR44 antagonism in vitro in human beta-cells and in type 2 diabetes (T2DM) patients. Methods We determined the drive on PGD2 secretion by glucose and IL-1beta, as well as, the impact on insulin secretion by pharmacological GPR44/DP2 antagonism (AZD1981) in human islets and beta-cells in vitro. To test if metabolic control would be improved by antagonizing a hyperglycemia-driven increased PGD2 tone, we performed a proof-of-mechanism study in 20 T2DM patients (average 54 years, HbA1c 9.4%, BMI 31.6 kg/m2). The randomized, double-blind, placebo-controlled cross-over study consisted of two three-day treatment periods (AZD1981 or placebo) separated by a three-day wash-out period. Mixed meal tolerance test (MMTT) and intravenous graded glucose infusion (GGI) was performed at start and end of each treatment period. Assessment of AZD1981 pharmacokinetics, glucose, insulin, C-peptide, glucagon, GLP-1, and PGD2 pathway biomarkers were performed. Results We found (1) that PGD2 is produced in human islet in response to high glucose or IL-1beta, but likely by stellate cells rather than endocrine cells; (2) that PGD2 suppresses both glucose and GLP-1 induced insulin secretion in vitro; and (3) that the GPR44/DP2 antagonist (AZD1981) in human beta-cells normalizes insulin secretion. However, AZD1981 had no impact on neither glucose nor incretin dependent insulin secretion in humans (GGI AUC C-peptide 1-2h and MMTT AUC Glucose 0-4h LS mean ratios vs placebo of 0.94 (80% CI of 0.90-0.98, p = 0.12) and 0.99 (90% CI of 0.94-1.05, p = 0.45), despite reaching the expected antagonist exposure. Conclusion/interpretation Pharmacological inhibition of the PGD2-GPR44/DP2 axis has no major impact on the modulation of acute insulin secretion in T2DM patients. Trial registration ClinicalTrials.gov NCT02367066.
- Published
- 2018
9. The No-Go and Nonsense-Mediated RNA Decay Pathways Are Regulated by Inflammatory Cytokines in Insulin-Producing Cells and Human Islets and Determine β-Cell Insulin Biosynthesis and Survival
- Author
-
Nicolai Krogh, Thomas Mandrup-Poulsen, Seyed Mojtaba Ghiasi, and Björn Tyrberg
- Subjects
0301 basic medicine ,Cell Survival ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,RNA Stability ,Blotting, Western ,030209 endocrinology & metabolism ,Apoptosis ,Proinflammatory cytokine ,Cell Line ,03 medical and health sciences ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Downregulation and upregulation ,Insulin-Secreting Cells ,Internal Medicine ,medicine ,Animals ,Humans ,Insulin ,Phosphorylation ,Gene knockdown ,Messenger RNA ,biology ,Chemistry ,Reverse Transcriptase Polymerase Chain Reaction ,Endoplasmic reticulum ,Nuclear Proteins ,Blotting, Northern ,Cell biology ,Rats ,Nitric oxide synthase ,030104 developmental biology ,Exoribonucleases ,biology.protein ,Unfolded protein response ,Cytokines ,RNA ,Signal Transduction - Abstract
Stress-related changes in β-cell mRNA levels result from a balance between gene transcription and mRNA decay. The regulation of RNA decay pathways has not been investigated in pancreatic β-cells. We found that no-go and nonsense-mediated RNA decay pathway components (RDPCs) and exoribonuclease complexes were expressed in INS-1 cells and human islets. Pelo, Dcp2, Dis3L2, Upf2, and Smg1/5/6/7 were upregulated by inflammatory cytokines in INS-1 cells under conditions where central β-cell mRNAs were downregulated. These changes in RDPC mRNA or corresponding protein levels were largely confirmed in INS-1 cells and rat/human islets. Cytokine-induced upregulation of Pelo, Xrn1, Dis3L2, Upf2, and Smg1/6 was reduced by inducible nitric oxide synthase inhibition, as were endoplasmic reticulum (ER) stress, inhibition of Ins1/2 mRNA, and accumulated insulin secretion. Reactive oxygen species inhibition or iron chelation did not affect RDPC expression. Pelo or Xrn1 knockdown (KD) aggravated, whereas Smg6 KD ameliorated, cytokine-induced INS-1 cell death without affecting ER stress; both increased insulin biosynthesis and medium accumulation but not glucose-stimulated insulin secretion in cytokine-exposed INS-1 cells. In conclusion, RDPCs are regulated by inflammatory stress in β-cells. RDPC KD improved insulin biosynthesis, likely by preventing Ins1/2 mRNA clearance. Pelo/Xrn1 KD aggravated, but Smg6 KD ameliorated, cytokine-mediated β-cell death, possibly through prevention of proapoptotic and antiapoptotic mRNA degradation, respectively.
- Published
- 2018
10. Genetic Control of β-Cell Mass Homeostasis
- Author
-
Ulrika Bergstrom, Mark A. Atkinson, Mangala M. Soundarapandian, Roxane Pasquier, Björn Tyrberg, and Maria L. Nieves
- Subjects
medicine.medical_specialty ,Cell growth ,Cell ,Cell generation ,Biology ,biology.organism_classification ,Neogenesis ,Cell biology ,medicine.anatomical_structure ,Endocrinology ,Internal medicine ,medicine ,Genetic predisposition ,Glucose homeostasis ,Homeostasis ,Cell mass - Abstract
Control of -cell function and mass is tightly linked to glucose homeostasis. Failing -cells inevitably lead to diabetes. Recently, several contradictory studies have been published arguing against or in favor of various mechanisms controlling -cell mass regulation. Here we review the literature on control of adult -cell mass and aim to reconcile thereby the contradictions. We discuss the role of � -cell proliferation and neogenesis, both in mice and man. We also discuss the influence of genetic predisposition on -cell mass control. We conclude that -cell generation in the adult human and mouse likely depends on many paths to assure sufficient numbers of -cells at any given time, thereby balancing mechanisms for negative regulation of -cell numbers. A simple model with only one pathway does not fit the current literature.
- Published
- 2010
- Full Text
- View/download PDF
11. Promoting islet cell function after transplantation
- Author
-
Carina Carlsson, Björn Tyrberg, Richard F. Olsson, Arne Andersson, Magnus Johansson, Linda Tillmar, Nils Welsh, Fredrik Palm, Göran Mattsson, Per-Ola Carlsson, Astrid Nordin, Örjan Källskog, and Leif Jansson
- Subjects
endocrine system ,medicine.medical_specialty ,Stromal cell ,endocrine system diseases ,medicine.medical_treatment ,Cell Culture Techniques ,Islets of Langerhans Transplantation ,Biophysics ,Mice, Obese ,Enteroendocrine cell ,Biology ,Revascularization ,Biochemistry ,Islets of Langerhans ,Mice ,Internal medicine ,medicine ,Animals ,Humans ,Hypoxia ,Cells, Cultured ,geography ,geography.geographical_feature_category ,Dose-Response Relationship, Drug ,Pancreatic islets ,Cell Biology ,General Medicine ,Hypoxia (medical) ,Islet ,Capillaries ,Mice, Inbred C57BL ,Oxygen ,Transplantation ,surgical procedures, operative ,Endocrinology ,medicine.anatomical_structure ,Hypertension ,Stromal Cells ,medicine.symptom ,Reinnervation - Abstract
Engraftment (i.e., the adaptation of transplanted pancreatic islets to their new surroundings with regard to revascularization, reinnervation, and reorganization of other stromal compartments) is of crucial importance for the survival and function of the endocrine cells. Previous studies suggest that transplantation induces both vascular and stromal dysfunctions in the implanted islets when compared with endogenous islets. Thus the vascular density and the blood perfusion of islet grafts is decreased and accompanied with a capillary hypertension. This leads to hypoxic conditions, with an associated shift toward anaerobic metabolism in grafted islets. An improved engraftment will prevent or compensate for the vascular/stromal dysfunction seen in transplanted islets and thereby augment survival of the islet implant. By such means the number of islets needed to cure the recipient will be lessened. This will increase the number of patients that can be transplanted with the limited material available.
- Published
- 2004
- Full Text
- View/download PDF
12. c-Myc Controls ProliferationVersusDifferentiation in Human Pancreatic Endocrine Cells
- Author
-
Björn Tyrberg, Fred Levine, Rich Jarvis, Carla Demeterco, Lance P. Ford, and Pamela Itkin-Ansari
- Subjects
Adult ,medicine.medical_specialty ,Cell division ,Cell Survival ,Recombinant Fusion Proteins ,Endocrinology, Diabetes and Metabolism ,Cellular differentiation ,Clinical Biochemistry ,Gene Expression ,Enteroendocrine cell ,Cell Communication ,Biology ,Biochemistry ,Translocation, Genetic ,Cell Line ,Proto-Oncogene Proteins c-myc ,Islets of Langerhans ,Fetus ,Endocrinology ,Internal medicine ,medicine ,Humans ,Insulin ,Receptor ,Transcription factor ,Homeodomain Proteins ,Cell growth ,Biochemistry (medical) ,Cell Differentiation ,Pancreatic Hormones ,medicine.anatomical_structure ,Receptors, Estrogen ,Cell culture ,Trans-Activators ,Pancreas ,Cell Division ,HeLa Cells ,Transcription Factors - Abstract
Using immortalized human pancreatic endocrine cell lines, we have shown previously that differentiation into hormoneexpressing cells requires cell-cell contact acting in synergy with the homeodomain transcription factor pancreatic duodenal homeobox-1 (PDX-1). Although differentiation is associated with a decrease in cell proliferation, the mechanisms behind this relationship are not known. Using TRM-6, a cell line, andlox5, a-cell line, we show here that cell-cell contact and subsequent endocrine differentiation lead to a down-regulation of the c-myc protooncogene. Overexpression of c-Myc obtained with an inducible c-Myc-estrogen receptor fusion protein results in an increase in cell proliferation and the ablation of hormone expression. Moreover, we show that although c-Myc is expressed in a subset of cells from the human fetal and adult pancreas, it is absent in differentiated endocrine cells. The mechanism by which c-Myc interferes with hormone expression may be through effects on the homeodomain transcription factor PDX-1, as immunostaining for PDX-1 in cells with activated c-Myc revealed a redistribution of PDX-1 from the nucleus to the cytoplasm. These results suggest that c-Myc plays a central role in a cell-cell contactmediated switch mechanism by which cell division vs. differentiation in endocrine cells is determined. (J Clin Endocrinol Metab 87: 3475–3485, 2002)
- Published
- 2002
- Full Text
- View/download PDF
13. Sweet Taste Receptors Regulate Basal Insulin Secretion and Contribute to Compensatory Insulin Hypersecretion During the Development of Diabetes in Male Mice
- Author
-
Kathleen R. Smith, Richard E. Pratley, George Kyriazis, Björn Tyrberg, and Tania Hussain
- Subjects
Male ,medicine.medical_specialty ,medicine.medical_treatment ,Type 2 diabetes ,Biology ,Real-Time Polymerase Chain Reaction ,Receptors, G-Protein-Coupled ,Mice ,Endocrinology ,Internal medicine ,Diabetes mellitus ,Insulin-Secreting Cells ,Insulin Secretion ,medicine ,Hyperinsulinemia ,Animals ,Humans ,Insulin ,Secretion ,Cells, Cultured ,Sensory mechanism ,Diabetes-Insulin-Glucagon-Gastrointestinal ,medicine.disease ,humanities ,Insulin oscillation ,Basal (medicine) ,Diabetes Mellitus, Type 2 - Abstract
β-Cells rapidly secrete insulin in response to acute increases in plasma glucose but, upon further continuous exposure to glucose, insulin secretion progressively decreases. Although the mechanisms are unclear, this mode of regulation suggests the presence of a time-dependent glucosensory system that temporarily attenuates insulin secretion. Interestingly, early-stage β-cell dysfunction is often characterized by basal (ie, fasting) insulin hypersecretion, suggesting a disruption of these related mechanisms. Because sweet taste receptors (STRs) on β-cells are implicated in the regulation of insulin secretion and glucose is a bona fide STR ligand, we tested whether STRs mediate this sensory mechanism and participate in the regulation of basal insulin secretion. We used mice lacking STR signaling (T1R2−/− knockout) and pharmacologic inhibition of STRs in human islets. Mouse and human islets deprived of STR signaling hypersecrete insulin at short-term fasting glucose concentrations. Accordingly, 5-hour fasted T1R2−/− mice have increased plasma insulin and lower glucose. Exposure of isolated wild-type islets to elevated glucose levels reduced STR expression, whereas islets from diabetic (db/db) or diet-induced obese mouse models show similar down-regulation. This transcriptional reprogramming in response to hyperglycemia correlates with reduced STR function in these mouse models, leading to insulin hypersecretion. These findings reveal a novel mechanism by which insulin secretion is physiologically regulated by STRs and also suggest that, during the development of diabetes, STR function is compromised by hyperglycemia leading to hyperinsulinemia. These observations further suggest that STRs might be a promising therapeutic target to prevent and treat type 2 diabetes.
- Published
- 2014
14. Species Differences in Susceptibility of Transplanted and Cultured Pancreatic Islets to the β-Cell Toxin Alloxan
- Author
-
Borg La, Björn Tyrberg, and Agneta Andersson
- Subjects
Male ,Necrosis ,endocrine system diseases ,Swine ,Islets of Langerhans Transplantation ,Rats, Sprague-Dawley ,Mice ,chemistry.chemical_compound ,Endocrinology ,Alloxan ,Insulin Secretion ,Insulin ,geography.geographical_feature_category ,Middle Aged ,Islet ,Immunohistochemistry ,medicine.anatomical_structure ,Rabbits ,medicine.symptom ,Cell Division ,Adult ,endocrine system ,medicine.medical_specialty ,Adolescent ,Guinea Pigs ,Mice, Nude ,Biology ,Guinea pig ,Islets of Langerhans ,Dogs ,Organ Culture Techniques ,Species Specificity ,In vivo ,Internal medicine ,medicine ,Animals ,Humans ,geography ,Pancreatic islets ,nutritional and metabolic diseases ,In vitro ,Rats ,Mice, Inbred C57BL ,Microscopy, Electron ,chemistry ,Apoptosis ,Animal Science and Zoology ,Lysosomes - Abstract
The beta-cell toxin alloxan, which produces oxygen radicals, is a model substance in studies of type 1 diabetes. Recently, human beta-cells have been found to be relatively resistant to this toxin. To clarify species differences in alloxan diabetogenicity, and oxygen radical toxicity, mouse, rat, rabbit, dog, pig, human and guinea pig islets have been studied after alloxan exposure. Using a standardized in vivo model, where islets were transplanted to nude mice, the different islets were compared. The results demonstrated that mouse and rat islet grafts were morphologically disturbed by alloxan and ROS. Rabbit and dog islet graft morphology was reasonably intact; and human, porcine, and guinea pig islet grafts were all well preserved. Furthermore, ultrastructural signs of apoptosis and necrosis, disturbances in the insulin secretory pattern during and after an alloxan perifusion, and islet lysosomal enzyme activities were studied in vitro in islets from some species. Guinea pig beta-cells were affected by alloxan, but a regeneration process compensated for the observed apoptotic and necrotic cell death. Human islets did not show any signs of alloxan-induced damage in the different models studied. Finally, no correlation between high alloxan sensitivity and high lysosomal enzyme activity was found. Thus, the beta-cell lysosomes are hardly specific targets for alloxan.
- Published
- 2001
- Full Text
- View/download PDF
15. Stimulated Endocrine Cell Proliferation and Differentiation in Transplanted Human Pancreatic Islets
- Author
-
Timo Otonkoski, Björn Tyrberg, Arne Andersson, and Jarkko Ustinov
- Subjects
geography ,medicine.medical_specialty ,Kidney ,geography.geographical_feature_category ,Cell growth ,Endocrinology, Diabetes and Metabolism ,Cellular differentiation ,Pancreatic islets ,Biology ,Islet ,biology.organism_classification ,Neogenesis ,Transplantation ,Endocrinology ,medicine.anatomical_structure ,Internal medicine ,Internal Medicine ,medicine ,Hepatocyte growth factor ,medicine.drug - Abstract
Neogenesis is crucial for the maintenance of β-cell mass in the human pancreas and possibly for the outcome of clinical islet transplantation. To date, no studies have reported a stimulation of human β-cell neogenesis in vivo. Therefore, we investigated whether human α-, β-, and duct cell growth can be stimulated when human islets are xenotransplanted to obese hyperglycemic-hyperinsulinemic ob/ob mice immuno-suppressed with anti-lymphocyte serum. Moreover, we wanted to study whether β-cell growth and duct-to-β-cell differentiation were induced in the hepatocyte growth factor (HGF)-dependent compensatory kidney growth model. For that purpose, we evaluated human islets grafted to nude (nu/nu) mice before uninephrectomy of the contralateral kidney for DNA-synthesis and duct cell expression of the β-cell-specific transcription factor Nkx 6.1 as an estimate of differentiation. Human islet grafts were well preserved after 2 weeks when transplanted to ob/ob mice during anti-lymphocyte immunosuppression. Both human β-cells (P < 0.01) and duct cells (P < 0.001) were growth stimulated when islets were transplanted to ob/ob mice. We also observed a correlation between increased duct cell proliferation and increased organ donor age (P =0.02). Moreover, duct (P < 0.05) and β-cell (P
- Published
- 2001
- Full Text
- View/download PDF
16. 35th Annual Meeting of the European Association for the Study of Diabetes
- Author
-
Phil Chowienczyk, Henri Martens, Federico Mallo, Edward Boyko, Matem Tunçdemir, Adam Tabak, Stephan Lortz, Marina TRENTO, Mauro DI LEO, Mathias Uhlen, Peter Butler, Jacek Golański, Nicolaas Schaper, Marja-Riitta Taskinen, Björn Tyrberg, Adam Kretowski, Guy Rutter, Mithieux Gilles, Marco Songini, Clive Petry, Morvarid Kabir, John Pernow, Frédéric Lemaigre, Nicolae Hancu, Alexandra Kautzky-Willer, Campistol Josep M, Mikael Knip, Chantal Mathieu, Jan Bolinder, Violeta Iotova, FRANCOISE MACARI, Solomon Tesfaye, Dominique DEVILLE de PERIERE, Christophe Hirtz, Angelina Passaro, Zdenek Sumnik, SONIA GAZTAMBIDE, Christophe Magnan, Karsten Buschard, Jaume Marrugat, Wladyslaw Grzeszczak, and Philippe Amouyel
- Subjects
medicine.medical_specialty ,biology ,business.industry ,Endocrinology, Diabetes and Metabolism ,Methylenetetrahydrofolate reductase ,Internal medicine ,Internal Medicine ,biology.protein ,Medicine ,business ,Gastroenterology ,Non diabetic ,Subclinical infection - Published
- 1999
- Full Text
- View/download PDF
17. Reduced sensitivity of inducible nitric oxide synthase-deficient mice to multiple low-dose streptozotocin-induced diabetes
- Author
-
Steven J. Sandler, Björn Tyrberg, Malin Flodström, and Decio L. Eizirik
- Subjects
medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Nitric Oxide Synthase Type II ,Mice, Inbred Strains ,Drug Administration Schedule ,Streptozocin ,Diabetes Mellitus, Experimental ,Nitric oxide ,Islets of Langerhans ,Mice ,chemistry.chemical_compound ,In vivo ,Internal medicine ,Internal Medicine ,medicine ,Animals ,RNA, Messenger ,Pancreas ,Nitrites ,Proinsulin ,geography ,geography.geographical_feature_category ,Dose-Response Relationship, Drug ,biology ,Insulin ,Streptozotocin ,Islet ,Nitric oxide synthase ,Endocrinology ,chemistry ,Mutation ,Knockout mouse ,biology.protein ,Disease Susceptibility ,Nitric Oxide Synthase ,Interleukin-1 ,medicine.drug - Abstract
Nitric oxide (NO), synthesized by the inducible isoform of nitric oxide synthase (iNOS), has been proposed as a mediator of immune-induced beta-cell destruction in type 1 diabetes. To evaluate the role of iNOS for beta-cell dysfunction and death, we investigated the sensitivity of beta-cells from mice genetically deficient in this enzyme (iNOS-/-, background C57BL/6x129SvEv, H-2b) both to interleukin (IL)-1beta-induced beta-cell dysfunction in vitro and to multiple low-dose streptozotocin (MLDS)-induced diabetes in vivo. Exposure of islets isolated from C57BL/6 mice to IL-1beta for 24 h in vitro resulted in an induction of iNOS mRNA expression, an increase in nitrite formation, and a decrease in insulin release and proinsulin biosynthesis as compared with untreated C57BL/6 islets. IL-1beta failed to induce iNOS mRNA expression and increase nitrite formation by islets isolated from iNOS knockout mice (iNOS-/-), and no impairment in islet function was observed. The iNOS-/- mice showed a reduced incidence of hyperglycemia after treatment with MLDS as compared with wild-type C57BL/6 (H-2b) and 129 SvEv (H-2b) mice. On day 21 after the first streptozotocin (STZ) injection, 75% of the C57BL/6 mice and 100% of the 129SvEv mice had blood glucose levels >11 mmol/l, whereas the corresponding number for iNOS-/- mice was only 23%. This protection was not due to a delay in the onset of hyperglycemia, since no increase in number of hyperglycemic iNOS-/- mice was observed when the animals were followed up to 42 days. Moreover, islets isolated from iNOS-/- mice were susceptible to the in vitro deleterious effects of STZ. In conclusion, the present study provides evidence that iNOS may contribute to beta-cell damage after exposure to IL-1beta in vitro and treatment with MLDS in vivo.
- Published
- 1999
- Full Text
- View/download PDF
18. Sweet Taste Receptor Deficient Mice Have Decreased Adiposity and Increased Bone Mass
- Author
-
William P. Cawthorn, Erica L. Scheller, Sebastian D. Parlee, Hiroyuki Mori, Björn Tyrberg, Ormond A. MacDougald, Yanfei L. Ma, Brian S. Learman, Xiaomin Ning, Becky R. Simon, and Venkatesh Krishnan
- Subjects
Male ,Taste ,Anatomy and Physiology ,Mouse ,lcsh:Medicine ,Adipose tissue ,Biochemistry ,Bone remodeling ,Receptors, G-Protein-Coupled ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Bone Density ,Adipocyte ,Adipocytes ,lcsh:Science ,Receptor ,Musculoskeletal System ,Adiposity ,2. Zero hunger ,Mice, Knockout ,0303 health sciences ,Multidisciplinary ,GLUCAGON-LIKE PEPTIDE-1 ,MARROW FAT ,Animal Models ,Taste Buds ,Adipose Tissue ,Adipogenesis ,Knockout mouse ,Medicine ,PPAR-GAMMA ,Bone Remodeling ,T1R3 ,Research Article ,EXPRESSION ,medicine.medical_specialty ,Carbohydrate metabolism ,Biology ,INSULIN-SECRETION ,Bone and Bones ,OSTEOBLAST DIFFERENTIATION ,03 medical and health sciences ,Model Organisms ,Internal medicine ,medicine ,Animals ,Obesity ,Bone ,030304 developmental biology ,MAMMALIAN SWEET ,Nutrition ,Cell Size ,REGULATE SECRETION ,lcsh:R ,Diet ,Endocrinology ,Metabolism ,Glucose ,chemistry ,CELLS ,lcsh:Q ,Physiological Processes ,Energy Metabolism ,030217 neurology & neurosurgery - Abstract
Functional expression of sweet taste receptors (T1R2 and T1R3) has been reported in numerous metabolic tissues, including the gut, pancreas, and, more recently, in adipose tissue. It has been suggested that sweet taste receptors in these nongustatory tissues may play a role in systemic energy balance and metabolism. Smaller adipose depots have been reported in T1R3 knockout mice on a high carbohydrate diet, and sweet taste receptors have been reported to regulate adipogenesis in vitro. To assess the potential contribution of sweet taste receptors to adipose tissue biology, we investigated the adipose tissue phenotypes of T1R2 and T1R3 knockout mice. Here we provide data to demonstrate that when fed an obesogenic diet, both T1R2 and T1R3 knockout mice have reduced adiposity and smaller adipocytes. Although a mild glucose intolerance was observed with T1R3 deficiency, other metabolic variables analyzed were similar between genotypes. In addition, food intake, respiratory quotient, oxygen consumption, and physical activity were unchanged in T1R2 knockout mice. Although T1R2 deficiency did not affect adipocyte number in peripheral adipose depots, the number of bone marrow adipocytes is significantly reduced in these knockout animals. Finally, we present data demonstrating that T1R2 and T1R3 knockout mice have increased cortical bone mass and trabecular remodeling. This report identifies novel functions for sweet taste receptors in the regulation of adipose and bone biology, and suggests that in these contexts, T1R2 and T1R3 are either dependent on each other for activity or have common independent effects in vivo.
- Published
- 2014
- Full Text
- View/download PDF
19. Human pancreatic beta-cell deoxyribonucleic acid-synthesis in islet grafts decreases with increasing organ donor age but increases in response to glucose stimulation in vitro
- Author
-
Decio L. Eizirik, Claes Hellerström, Agneta Andersson, Daniel Pipeleers, and Björn Tyrberg
- Subjects
Adult ,Male ,Aging ,medicine.medical_specialty ,Adolescent ,medicine.medical_treatment ,Transplantation, Heterologous ,Islets of Langerhans Transplantation ,Mice, Nude ,Biology ,Islets of Langerhans ,Mice ,chemistry.chemical_compound ,Endocrinology ,Culture Techniques ,Internal medicine ,Alloxan ,medicine ,Animals ,Humans ,Organ donation ,Child ,geography ,Kidney ,geography.geographical_feature_category ,Insulin ,Osmolar Concentration ,DNA ,Middle Aged ,Islet ,Tissue Donors ,Glucose ,medicine.anatomical_structure ,chemistry ,Growth Hormone ,Hyperglycemia ,Beta cell ,Pancreas ,Hyperglycemic agent ,Thymidine - Abstract
Human pancreatic beta-cell proliferation may be crucial for the success of islet transplantation. The aim of this study was to test the hypothesis that adult human beta-cells proliferate in vitro and in vivo and respond with increased rates of replication to factors known to promote rodent islet-cell proliferation, i.e. glucose, human recombinant GH, and FCS. For this purpose, human islets were prepared from a total of 19 adult heart-beating organ donors and cultured for 48 h with or without the additives described above. 3H-thymidine was added to the medium during the last 60 min of culture. After immunohistochemical staining for insulin and autoradiography, the labeling index (LI; i.e. % of labeled beta-cells over total number of beta-cells) was estimated by light microscopy. Islets also were transplanted under the kidney capsule of normal or alloxan-diabetic nude mice. After 2 weeks, 3H-thymidine was injected and the islet grafts prepared for determination of LI, as described above. Islets cultured at 5.6 mM glucose showed an increased beta-cell proliferation compared with islets cultured at 2.8 mM glucose (P0.05). However, culture at 11 mM glucose failed to further increase beta-cell proliferation. Addition of GH (1 microg/ml) to the medium, in the presence of 1% FCS and 5.6 mM glucose, did not influence the rate of beta-cell proliferation. In islets transplanted to hyperglycemic nude mice, beta-cell proliferation was similar to that observed in islets grafted into normoglycemic nude mice. Proliferation, however, decreased with increasing organ donor age. This study shows that pancreatic beta-cells from adult man are able to proliferate both in vitro and in vivo. Moreover, beta-cells from adult human donors respond with increased proliferation to glucose in vitro and show a decreased proliferation in vivo with increasing donor age.
- Published
- 1996
- Full Text
- View/download PDF
20. Sweet taste receptor signaling in beta cells mediates fructose-induced potentiation of glucose-stimulated insulin secretion
- Author
-
George Kyriazis, Björn Tyrberg, and Mangala M. Soundarapandian
- Subjects
Male ,medicine.medical_specialty ,medicine.medical_treatment ,Intracellular Space ,TRPM Cation Channels ,Fructose ,Receptors, G-Protein-Coupled ,Mice ,Enzyme activator ,Insulin-Secreting Cells ,Insulin receptor substrate ,Internal medicine ,Insulin Secretion ,medicine ,Animals ,Humans ,Insulin ,TRPM3 ,TRPM5 ,Multidisciplinary ,biology ,Postprandial Period ,Enzyme Activation ,Insulin receptor ,Glucose ,Endocrinology ,PNAS Plus ,Taste ,Type C Phospholipases ,biology.protein ,Calcium ,Beta cell ,Signal transduction ,Gene Deletion ,Signal Transduction - Abstract
Postprandial insulin release is regulated by glucose, but other circulating nutrients may target beta cells and potentiate glucose-stimulated insulin secretion via distinct signaling pathways. We demonstrate that fructose activates sweet taste receptors (TRs) on beta cells and synergizes with glucose to amplify insulin release in human and mouse islets. Genetic ablation of the sweet TR protein T1R2 obliterates fructose-induced insulin release and its potentiating effects on glucose-stimulated insulin secretion in vitro and in vivo. TR signaling in beta cells is triggered, at least in part, in parallel with the glucose metabolic pathway and leads to increases in intracellular calcium that are dependent on the activation of phospholipase C (PLC) and transient receptor potential cation channel, subfamily M, member 5 (TRPM5). Our results unveil a pathway for the regulation of insulin release by postprandial nutrients that involves beta cell sweet TR signaling.
- Published
- 2012
- Full Text
- View/download PDF
21. T-cadherin (Cdh13) in association with pancreatic beta-cell granules contributes to second phase insulin secretion
- Author
-
Martin S. Denzel, Barbara Ranscht, Edward Monosov, Fred Levine, Philip D.G. Miles, Björn Tyrberg, Krist T. Azizian, and Maria L. Nieves
- Subjects
Male ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Blotting, Western ,Biology ,Islets of Langerhans ,Mice ,Endocrinology ,Insulin-Secreting Cells ,Internal medicine ,Insulin Secretion ,medicine ,Animals ,Humans ,Insulin ,Glucose homeostasis ,Microscopy, Immunoelectron ,Mice, Knockout ,geography ,Glucose tolerance test ,Microscopy, Confocal ,geography.geographical_feature_category ,Adiponectin ,medicine.diagnostic_test ,Glucose Tolerance Test ,Glucose clamp technique ,Cadherins ,Islet ,Immunohistochemistry ,Insulin oscillation ,Mice, Inbred C57BL ,Insulin receptor ,Glucose Clamp Technique ,biology.protein ,Research Paper - Abstract
Glucose homeostasis depends on adequate control of insulin secretion. We report the association of the cell-adhesion and adiponectin (APN)-binding glycoprotein T-cadherin (Cdh13) with insulin granules in mouse and human beta-cells. Immunohistochemistry and electron microscopy of islets in situ and targeting of RFP-tagged T-cadherin to GFP-labeled insulin granules in isolated beta-cells demonstrate this unusual location. Analyses of T-cadherin-deficient (Tcad-KO) mice show normal islet architecture and insulin content. However, T-cadherin is required for sufficient insulin release in vitro and in vivo. Primary islets from Tcad-KO mice were defective in glucose-induced but not KCl-mediated insulin secretion. In vivo, second phase insulin release in T-cad-KO mice during a hyperglycemic clamp was impaired while acute first phase release was unaffected. Tcad-KO mice showed progressive glucose intolerance by 5 mo of age without concomitant changes in peripheral insulin sensitivity. Our analyses detected no association of APN with T-cadherin on beta-cell granules although colocalization was observed on the pancreatic vasculature. These data identify T-cadherin as a novel component of insulin granules and suggest that T-cadherin contributes to the regulation of insulin secretion independently of direct interactions with APN.
- Published
- 2011
22. NeuroD1 in the endocrine pancreas: localization and dual function as an activator and repressor
- Author
-
Carla Demeterco, Ifat Geron, Pamela Itkin-Ansari, Björn Tyrberg, Ergeng Hao, Jacqueline E. Lee, C. Padilla, Edoardo Marcora, Andrew B. Leiter, Fred Levine, and Christelle Ratineau
- Subjects
medicine.medical_specialty ,Aging ,Amyloid ,Transcription, Genetic ,Repressor ,Enteroendocrine cell ,Nerve Tissue Proteins ,Biology ,Cell Line ,Islets of Langerhans ,Mice ,Internal medicine ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Somatostatin receptor 2 ,Animals ,Humans ,Insulin ,Somatostatin receptor 1 ,Promoter Regions, Genetic ,Pancreas ,Homeodomain Proteins ,Delta cell ,Gene Expression Profiling ,Nuclear Proteins ,DNA ,Zebrafish Proteins ,Cell biology ,Protein Structure, Tertiary ,DNA-Binding Proteins ,Repressor Proteins ,medicine.anatomical_structure ,Endocrinology ,Somatostatin ,Homeobox Protein Nkx-2.2 ,Animals, Newborn ,Trans-Activators ,Ectopic expression ,Developmental Biology ,Transcription Factors - Abstract
The basic helix–loop–helix transcription factor NeuroD1 regulates cell fate in the nervous system but previously has not been considered to function similarly in the endocrine pancreas due to its reported expression in all islet cell types in the newborn mouse. Because we found that NeuroD1 potently represses somatostatin expression in vitro, its pattern of expression was examined in both strains of mice in which lacZ has been introduced into the NeuroD1 locus by homologous recombination. Analysis of adult transgenic mice revealed that NeuroD1 is predominantly expressed in β-cells and either absent or expressed below the limit of lacZ detection in mature α-, δ-, or PP cells. Consistent with a previous report, NeuroD1 colocalizes with glucagon as well as insulin in immature islets of the newborn mouse. However, no colocalization of NeuroD1with somatostatin was detected in the newborn. In vitro, ectopic expression of NeuroD1 in TRM-6/PDX-1, a human pancreatic δ-cell line, resulted in potent repression of somatostatin concomitant with induction of the β-cell hormones insulin and islet amyloid polypeptide. Additionally, NeuroD1 induced expression of Nkx2.2, a transcription factor expressed in β- but not δ-cells. Transfection studies using insulin and somatostatin promoters confirm the ability of NeuroD1 to act as both a transcriptional repressor and activator in the same cell, suggesting a more complex role for NeuroD1 in the establishment and/or maintenance of mature endocrine cells than has been recognized previously. Developmental Dynamics 233:946–953, 2005. © 2005 Wiley-Liss, Inc.
- Published
- 2005
23. Islet expression of the DNA repair enzyme 8-oxoguanosine DNA glycosylase (Ogg1) in human type 2 diabetes
- Author
-
Fred Levine, Sergio Atala Dib, Jessica Wang-Rodriguez, Björn Tyrberg, Kun-Ho Yoon, and Kamen A Anachkov
- Subjects
medicine.medical_specialty ,Type 1 diabetes ,geography ,geography.geographical_feature_category ,lcsh:RC648-665 ,business.industry ,DNA damage ,DNA repair ,Endocrinology, Diabetes and Metabolism ,General Medicine ,Type 2 diabetes ,medicine.disease ,Islet ,medicine.disease_cause ,lcsh:Diseases of the endocrine glands. Clinical endocrinology ,Endocrinology ,Diabetes mellitus ,Internal medicine ,medicine ,business ,Oxidative stress ,Epigenetics of diabetes Type 2 ,Research Article - Abstract
Background It has become increasingly clear that β-cell failure plays a critical role in the pathogenesis of type 2 diabetes. Free-radical mediated β-cell damage has been intensively studied in type 1 diabetes, but not in human type 2 diabetes. Therefore, we studied the protein expression of the DNA repair enzyme Ogg1 in pancreases from type 2 diabetics. Ogg1 was studied because it is the major enzyme involved in repairing 7,8-dihydro-8-oxoguanosine DNA adducts, a lesion previously observed in a rat model of type 2 diabetes. Moreover, in a gene expression screen, Ogg1 was over-expressed in islets from a human type 2 diabetic. Methods Immunofluorescent staining of Ogg1 was performed on pancreatic specimens from healthy controls and patients with diabetes for 2–23 years. The intensity and islet area stained for Ogg1 was evaluated by semi-quantitative scoring. Results Both the intensity and the area of islet Ogg1 staining were significantly increased in islets from the type 2 diabetic subjects compared to the healthy controls. A correlation between increased Ogg1 fluorescent staining intensity and duration of diabetes was also found. Most of the staining observed was cytoplasmic, suggesting that mitochondrial Ogg1 accounts primarily for the increased Ogg1 expression. Conclusion We conclude that oxidative stress related DNA damage may be a novel important factor in the pathogenesis of human type 2 diabetes. An increase of Ogg1 in islet cell mitochondria is consistent with a model in which hyperglycemia and consequent increased β-cell oxidative metabolism lead to DNA damage and the induction of Ogg1 expression.
- Published
- 2002
24. Beta-cell differentiation from a human pancreatic cell line in vitro and in vivo
- Author
-
Ergeng Hao, Dominique Dufayet de la Tour, Stuart Bossie, Pamela Itkin-Ansari, Mary Loy, Tanya L. Halvorsen, Soon-Jib Yoo, Björn Tyrberg, Carla Demeterco, and Fred Levine
- Subjects
Blood Glucose ,medicine.medical_specialty ,PAX6 Transcription Factor ,Transcription, Genetic ,Cell Transplantation ,Cellular differentiation ,medicine.medical_treatment ,Transplantation, Heterologous ,Mice, Nude ,Biology ,Glucagon-Like Peptide-1 Receptor ,Cell Line ,Islets of Langerhans ,Mice ,Endocrinology ,Downregulation and upregulation ,Internal medicine ,Glucokinase ,Insulin Secretion ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Receptors, Glucagon ,Animals ,Humans ,Insulin ,Paired Box Transcription Factors ,Eye Proteins ,Molecular Biology ,Transcription factor ,Activating Transcription Factor 1 ,Homeodomain Proteins ,C-Peptide ,Cell Differentiation ,General Medicine ,Cell biology ,Up-Regulation ,Transplantation ,DNA-Binding Proteins ,Repressor Proteins ,medicine.anatomical_structure ,Cell culture ,Trans-Activators ,Signal transduction ,Pancreas ,Somatostatin ,Transcription Factors - Abstract
Cell transplantation therapy for diabetes is limited by an inadequate supply of cells exhibiting glucose-responsive insulin secretion. To generate an unlimited supply of human β-cells, inducibly transformed pancreatic β-cell lines have been created by expression of dominant oncogenes. The cell lines grow indefinitely but lose differentiated function. Induction of β-cell differentiation was achieved by stimulating the signaling pathways downstream of the transcription factor PDX-1, cell-cell contact, and the glucagon-like peptide (GLP-1) receptor. Synergistic activation of those pathways resulted in differentiation into functional β-cells exhibiting glucose-responsive insulin secretion in vitro. Both oncogene-expressing and oncogene-deleted cells were transplanted into nude mice and found to exhibit glucose-responsive insulin secretion in vivo. The ability to grow unlimited quantities of human β-cells is a major step toward developing a cell transplantation therapy for diabetes.
- Published
- 2001
25. Human islets in mixed islet grafts protect mouse pancreatic beta-cells from alloxan toxicity
- Author
-
Ole D. Madsen, Björn Tyrberg, Stefan L. Marklund, Arne Andersson, Beata T. Olejnicka, and Decio L. Eizirik
- Subjects
Male ,endocrine system ,medicine.medical_specialty ,endocrine system diseases ,Ratón ,Health, Toxicology and Mutagenesis ,Mice, Nude ,Toxicology ,Superoxide dismutase ,chemistry.chemical_compound ,Islets of Langerhans ,Mice ,Alloxan ,Internal medicine ,medicine ,Animals ,Humans ,B cell ,Pharmacology ,chemistry.chemical_classification ,geography ,Glutathione Peroxidase ,geography.geographical_feature_category ,biology ,Superoxide Dismutase ,Glutathione peroxidase ,nutritional and metabolic diseases ,Hydrogen Peroxide ,Islet ,Immunohistochemistry ,Coculture Techniques ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Toxicity ,biology.protein ,Pancreas - Abstract
We have previously shown that human beta-cells are resistant to the toxic effects of alloxan. In order to further clarify this characteristic of human islets, we investigated whether these cells might transfer their alloxan resistance to alloxan-sensitive rat or mouse islets. Islets from two species (human-mouse or rat-mouse) were mixed into one graft, which was implanted into the subcapsular kidney space of nude mice. Alloxan or saline was injected intravenously two weeks after implantation and one week thereafter the mice were killed. The number of grafted and endogenous beta-cells were evaluated by a semi-quantitative method after immunohistochemistry. Human islet production of the scavenging enzymes extracellular superoxide dismutase and plasma glutathione peroxidase were analyzed with ELISA-techniques, and mouse and human islet hydrogen peroxide breakdown activity were monitored with a horseradish peroxidase-dependent assay. Mouse beta-cells transplanted together with human islets were protected against alloxan cytotoxicity. Rat islets did not protect mouse beta-cells against alloxan, suggesting that the mixing procedure as such did not impose the protection. Production of extracellular superoxide dismutase and plasma glutathione peroxidase by human islets was very low. Moreover, H2O2 breakdown in vitro, did not differ between human and mouse islets. Alloxan-insensitive human islets protect mouse beta-cells against alloxan-induced lesions, suggesting that yet to be identified extracellular factors are involved in human islet resistance to alloxan toxicity.
- Published
- 2000
26. Differences between humans and rodents in the susceptibility to pancreatic cell injury. Implications for the pathogenesis of diabetes
- Author
-
Carol A. Delaney, L. A. Håkan Borg, Stellan Sandler, Nils Welsh, Björn Tyrberg, Claes Hellerström, Arne Andersson, Daniel Pipeleers, Malin Flodström, and Decio L. Eizirik
- Subjects
Pathogenesis ,Endocrinology ,business.industry ,Endocrinology, Diabetes and Metabolism ,Diabetes mellitus ,Immunology ,Internal Medicine ,Cell injury ,medicine ,General Medicine ,medicine.disease ,business - Published
- 2009
- Full Text
- View/download PDF
27. Engraftment and growth of transplanted pancreatic islets
- Author
-
Aileen King, Richard F. Olsson, Leif Jansson, Per Liss, Björn Tyrberg, Göran Mattsson, Claes Hellerström, Örjan Källskog, Stellan Sandler, Arne Andersson, Fredrik Palm, Per-Ola Carlsson, Erika Höglund, and Carina Carlsson
- Subjects
endocrine system ,medicine.medical_specialty ,Microdialysis ,geography ,geography.geographical_feature_category ,endocrine system diseases ,business.industry ,Pancreatic islets ,Islets of Langerhans Transplantation ,Enteroendocrine cell ,General Medicine ,Islet ,Transplantation ,Islets of Langerhans ,Endocrinology ,medicine.anatomical_structure ,In vivo ,Internal medicine ,medicine ,Cancer research ,Animals ,Humans ,business ,Reinnervation ,Hormone - Abstract
Transplantation of pancreatic islets may provide a cure for type 1 diabetes. However, this treatment can currently be offered only to very few patients. To improve transplantation success we need to understand better the mechanisms of how the implanted islets survive, grow and/or maintain adequate function. We herein report on our studies to evaluate the factors responsible for the engraftment, i.e. revascularization, reinnervation etc., of transplanted islets and relate these factors to the metabolism and growth of the islets. Graft metabolism can be monitored by microdialysis probes that allow for the measurement of minute amounts of islet metabolites and hormonal products. Growth of the endocrine cells can be stimulated both in vitro before implantation and in vivo post-transplantation. Another problem is rejection of transplanted islets, which may be overcome by the microencapsulation of islets. The knowledge gained by the present studies will enable us to elucidate the optimal treatment of islets to ensure a maximal survival of the transplanted islets, and may be applied also to clinical islet transplantation.
28. Sensitivity of human pancreatic islets to peroxynitrite-induced cell dysfunction and death
- Author
-
Carol A. Delaney, Decio L. Eizirik, Björn Hellman, Björn Tyrberg, Hamid Vaghef, Luc Bouwens, Pathological Anatomy, and Vrije Universiteit Brussel
- Subjects
Programmed cell death ,medicine.medical_specialty ,Necrosis ,Biophysics ,Apoptosis ,Biology ,Biochemistry ,Peroxynitrite ,Nitric oxide ,chemistry.chemical_compound ,Islets of Langerhans ,Species Specificity ,Structural Biology ,Internal medicine ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Comet assay ,Cells, Cultured ,geography ,geography.geographical_feature_category ,Nitrates ,Cell Death ,Superoxide ,Pancreatic islets ,Cell Biology ,Islet ,Rats ,Endocrinology ,medicine.anatomical_structure ,Glucose ,chemistry ,medicine.symptom ,Oxidation-Reduction ,DNA Damage - Abstract
Nitric oxide and peroxynitrite (generated by the reaction of nitric oxide with the superoxide anion) may both be mediators of β-cell damage in early insulin-dependent diabetes mellitus. We observed that acute exposure of primary cultured human pancreatic islets to peroxynitrite results in a significant decrease in glucose oxidation and islet retrieval. DNA strand breaks in single human and rat islet cells are detectable after acute peroxynitrite exposure, followed by a decrease in islet cell survival after 1 h and 24 h. Cell death appeared to occur via a toxic cell death mechanism (necrosis) rather than apoptosis, as suggested by vital staining and ultrastructural evidence of early membrane and organelle degradation, mitochondrial swelling and loss of matrix. This study demonstrates for the first time that cultured human pancreatic islets are susceptible to the noxious effects of peroxynitrite.
29. �Tasting� fructose with pancreatic beta-cells: modulation of insulin release by sweet taste receptor signaling and its role in metabolic diseases
- Author
-
Kathleen R Mosure, Richard E. Pratley, Mangala M. Soundarapandian, George Kyriazis, and Björn Tyrberg
- Subjects
medicine.medical_specialty ,medicine.medical_treatment ,Protein subunit ,lcsh:Medicine ,Stimulation ,Bioinformatics ,General Biochemistry, Genetics and Molecular Biology ,Transient receptor potential channel ,chemistry.chemical_compound ,Internal medicine ,medicine ,Monosaccharide ,lcsh:Science ,Receptor ,chemistry.chemical_classification ,business.industry ,Insulin ,lcsh:R ,Fructose ,General Medicine ,In vitro ,Endocrinology ,chemistry ,Poster Presentation ,lcsh:Q ,business - Abstract
Background Although glucose is indispensable for the stimulation of insulin release, numerous other insulin secretagogues have been identified. For instance, the dietary monosaccharide fructose potentiates insulin secretion in vitro, but the mechanism and physiological significance remains unclear. The T1R2-T1R3 heterodimer of G protein-coupled receptors mediates sweet sensing in the tongue and ablation of either subunit obliterates sweet taste. We hypothesized that the effects of fructose on insulin release may be mediated by sweet taste receptors (TRs) on beta-cells.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.