SwePub
Tyck till om SwePub Sök här!
Sök i SwePub databas

  Utökad sökning

Träfflista för sökning "WFRF:(Rorsman Patrik) ;lar1:(gu)"

Sökning: WFRF:(Rorsman Patrik) > Göteborgs universitet

  • Resultat 1-10 av 45
Sortera/gruppera träfflistan
   
NumreringReferensOmslagsbildHitta
1.
  • Adam, J., et al. (författare)
  • Fumarate Hydratase Deletion in Pancreatic beta Cells Leads to Progressive Diabetes
  • 2017
  • Ingår i: Cell Reports. - : Elsevier BV. - 2211-1247. ; 20:13, s. 3135-3148
  • Tidskriftsartikel (refereegranskat)abstract
    • We explored the role of the Krebs cycle enzyme fumarate hydratase (FH) in glucose-stimulated insulin secretion (GSIS). Mice lacking Fh1 in pancreatic beta cells (Fh1 beta KO mice) appear normal for 6-8 weeks but then develop progressive glucose intolerance and diabetes. Glucose tolerance is rescued by expression of mitochondrial or cytosolic FH but not by deletion of Hif1 alpha or Nrf2. Progressive hyperglycemia in Fh1bKO mice led to dysregulated metabolism in b cells, a decrease in glucose-induced ATP production, electrical activity, cytoplasmic [Ca2+](i) elevation, and GSIS. Fh1 loss resulted in elevated intracellular fumarate, promoting succination of critical cysteines in GAPDH, GMPR, and PARK 7/DJ-1 and cytoplasmic acidification. Intracellular fumarate levels were increased in islets exposed to high glucose and in islets from human donors with type 2 diabetes (T2D). The impaired GSIS in islets from diabetic Fh1bKO mice was ameliorated after culture under normoglycemic conditions. These studies highlight the role of FH and dysregulated mitochondrial metabolism in T2D.
  •  
2.
  • Briant, L. J. B., et al. (författare)
  • CPT1a-Dependent Long-Chain Fatty Acid Oxidation Contributes to Maintaining Glucagon Secretion from Pancreatic Islets
  • 2018
  • Ingår i: Cell Reports. - : Elsevier BV. - 2211-1247. ; 23:11, s. 3300-3311
  • Tidskriftsartikel (refereegranskat)abstract
    • Glucagon, the principal hyperglycemic hormone, is secreted from pancreatic islet a cells as part of the counter-regulatory response to hypoglycemia. Hence, secretory output from a cells is under high demand in conditions of low glucose supply. Many tissues oxidize fat as an alternate energy substrate. Here, we show that glucagon secretion in low glucose conditions is maintained by fatty acid metabolism in both mouse and human islets, and that inhibiting this metabolic pathway profoundly decreases glucagon output by depolarizing alpha cell membrane potential and decreasing action potential amplitude. We demonstrate, by using experimental and computational approaches, that this is not mediated by the K-ATP channel, but instead due to reduced operation of the Na+-K+ pump. These data suggest that counter-regulatory secretion of glucagon is driven by fatty acid metabolism, and that the Na+-K+ pump is an important ATP-dependent regulator of alpha cell function.
  •  
3.
  • Kellard, J. A., et al. (författare)
  • Reduced somatostatin signalling leads to hypersecretion of glucagon in mice fed a high-fat diet
  • 2020
  • Ingår i: Molecular Metabolism. - : Elsevier BV. - 2212-8778. ; 40
  • Tidskriftsartikel (refereegranskat)abstract
    • Objectives: Elevated plasma glucagon is an early symptom of diabetes, occurring in subjects with impaired glucose regulation. Here, we explored alpha-cell function in female mice fed a high-fat diet (HFD). Methods: Female mice expressing the Ca2+ indicator GCaMP3 specifically in alpha-cells were fed a high-fat or control (CTL) diet. We then conducted in vivo phenotyping of these mice, as well as experiments on isolated (ex vivo) islets and in the in situ perfused pancreas. Results: In HFD-fed mice, fed plasma glucagon levels were increased and glucagon secretion from isolated islets and in the perfused mouse pancreas was also elevated. In mice fed a CTL diet, increasing glucose reduced intracellular Ca2+ ([Ca2+](i)) oscillation frequency and amplitude. This effect was also observed in HFD mice; however, both the frequency and amplitude of the [Ca2+](i) oscillations were higher than those in CTL alpha-cells. Given that alpha-cells are under strong paracrine control from neighbouring somatostatin-secreting delta-cells, we hypothesised that this elevation of alpha-cell output was due to a lack of somatostatin (SST) secretion. Indeed, SST secretion in isolated islets from HFD-fed mice was reduced but exogenous SST also failed to suppress glucagon secretion and [Ca2+](i) activity from HFD alpha-cells, in contrast to observations in CTL mice. Conclusions: These findings suggest that reduced delta-cell function, combined with intrinsic changes in alpha-cells including sensitivity to somatostatin, accounts for the hyperglucagonaemia in mice fed a HFD. (C) 2020 The Author(s). Published by Elsevier GmbH.
  •  
4.
  • Shigeto, Makoto, et al. (författare)
  • GLP-1 stimulates insulin secretion by PKC-dependent TRPM4 and TRPM5 activation
  • 2015
  • Ingår i: Journal of Clinical Investigation. - : American Society for Clinical Investigation. - 0021-9738 .- 1558-8238. ; 125:12, s. 4714-4728
  • Tidskriftsartikel (refereegranskat)abstract
    • Strategies aimed at mimicking or enhancing the action of the incretin hormone glucagon-like peptide 1 (GLP-1) therapeutically improve glucose-stimulated insulin secretion (GSIS); however, it is not clear whether GLP-1 directly drives insulin secretion in pancreatic islets. Here, we examined the mechanisms by which GLP-1 stimulates insulin secretion in mouse and human islets. We found that GLP-1 enhances GSIS at a half-maximal effective concentration of 0.4 pM. Moreover, we determined that GLP-1 activates PLC, which increases submembrane diacylglycerol and thereby activates PKC, resulting in membrane depolarization and increased action potential firing and subsequent stimulation of insulin secretion. The depolarizing effect of GLP-1 on electrical activity was mimicked by the PKC activator PMA, occurred without activation of PKA, and persisted in the presence of PKA inhibitors, the K-ATP channel blacker tolbutamide, and the L-type Ca2+ channel blacker isradipine; however, depolarization was abolished by lowering extracellular Na+. The PKC-dependent effect of GLP-1 on membrane potential and electrical activity was mediated by activation of NW-permeable TRPM4 and TRPM5 channels by mobilization of intracellular Ca2+ from thapsigargin-sensitive Ca2+ stores. Concordantly, GLP-1 effects were negligible in Trpm4 or Trpm5 KO islets. These data provide important insight into the therapeutic action of GLP-1 and suggest that circulating levels of this hormone directly stimulate insulin secretion by beta cells.
  •  
5.
  • Tarasov, A. I., et al. (författare)
  • Monitoring real-time hormone release kinetics: Via high-content 3-D imaging of compensatory endocytosis
  • 2018
  • Ingår i: Lab on a Chip. - : Royal Society of Chemistry (RSC). - 1473-0197 .- 1473-0189. ; 18:18, s. 2838-2848
  • Tidskriftsartikel (refereegranskat)abstract
    • High-content real-time imaging of hormone secretion in tissues or cell populations is a challenging task, which is unlikely to be resolved directly, despite immense translational value. We approach this problem indirectly, using compensatory endocytosis, a process that closely follows exocytosis in the cell, as a surrogate read-out for secretion. The tissue is immobilized in an open-air perifusion chamber and imaged using a two-photon microscope. A fluorescent polar tracer, perifused through the experimental circuit, gets trapped into the cells via endocytosis, and is quantified using a feature-detection algorithm. The signal of the tracer that accumulates into the endocytotic system reliably reflects stimulated exocytosis, which is demonstrated via co-imaging of the latter using existing reporters. A high signal-to-noise ratio and compatibility with multisensor imaging affords the real-time quantification of the secretion at the tissue/population level, whereas the cumulative nature of the signal allows imprinting of the “secretory history” within each cell. The technology works for several cell types, reflects disease progression and can be used for human tissue.
  •  
6.
  • Vergari, Elisa, et al. (författare)
  • Insulin inhibits glucagon release by SGLT2-induced stimulation of somatostatin secretion
  • 2019
  • Ingår i: Nature Communications. - : Springer Science and Business Media LLC. - 2041-1723. ; 10:1
  • Tidskriftsartikel (refereegranskat)abstract
    • Hypoglycaemia (low plasma glucose) is a serious and potentially fatal complication of insulin-treated diabetes. In healthy individuals, hypoglycaemia triggers glucagon secretion, which restores normal plasma glucose levels by stimulation of hepatic glucose production. This counterregulatory mechanism is impaired in diabetes. Here we show in mice that therapeutic concentrations of insulin inhibit glucagon secretion by an indirect (paracrine) mechanism mediated by stimulation of intra-islet somatostatin release. Insulin's capacity to inhibit glucagon secretion is lost following genetic ablation of insulin receptors in the somatostatin-secreting δ-cells, when insulin-induced somatostatin secretion is suppressed by dapagliflozin (an inhibitor of sodium-glucose co-tranporter-2; SGLT2) or when the action of secreted somatostatin is prevented by somatostatin receptor (SSTR) antagonists. Administration of these compounds in vivo antagonises insulin's hypoglycaemic effect. We extend these data to isolated human islets. We propose that SSTR or SGLT2 antagonists should be considered as adjuncts to insulin in diabetes therapy.
  •  
7.
  • Vergari, Elisa, et al. (författare)
  • Somatostatin secretion by Na+-dependent Ca2+-induced Ca2+ release in pancreatic delta-cells.
  • 2020
  • Ingår i: Nature metabolism. - : Springer Science and Business Media LLC. - 2522-5812. ; 2:1, s. 32-40
  • Tidskriftsartikel (refereegranskat)abstract
    • Pancreatic islets are complex micro-organs consisting of at least three different cell types: glucagon-secreting α-, insulin-producing β- and somatostatin-releasing δ-cells1. Somatostatin is a powerful paracrine inhibitor of insulin and glucagon secretion2. In diabetes, increased somatostatinergic signalling leads to defective counter-regulatory glucagon secretion3. This increases the risk of severe hypoglycaemia, a dangerous complication of insulin therapy4. The regulation of somatostatin secretion involves both intrinsic and paracrine mechanisms5 but their relative contributions and whether they interact remains unclear. Here we show that dapagliflozin-sensitive glucose- and insulin-dependent sodium uptake stimulates somatostatin secretion by elevating the cytoplasmic Na+ concentration ([Na+]i) and promoting intracellular Ca2+-induced Ca2+ release (CICR). This mechanism also becomes activated when [Na+]i is elevated following the inhibition of the plasmalemmal Na+-K+ pump by reductions of the extracellular K+ concentration emulating those produced by exogenous insulin in vivo6. Islets from some donors with type-2 diabetes hypersecrete somatostatin, leading to suppression of glucagon secretion that can be alleviated by a somatostatin receptor antagonist. Our data highlight the role of Na+ as an intracellular second messenger, illustrate the significance of the intraislet paracrine network and provide a mechanistic framework for pharmacological correction of the hormone secretion defects associated with diabetes that selectively target the δ-cells.
  •  
8.
  • Zhang, Q., et al. (författare)
  • Na+ current properties in islet alpha- and beta-cells reflect cell-specific Scn3a and Scn9a expression
  • 2014
  • Ingår i: Journal of Physiology-London. - : Wiley. - 0022-3751 .- 1469-7793. ; 592:21, s. 4677-4696
  • Tidskriftsartikel (refereegranskat)abstract
    • - and -cells express both Na(v)1.3 and Na(v)1.7 Na+ channels but in different relative amounts. The differential expression explains the different properties of Na+ currents in - and -cells. Na(v)1.3 is the functionally important Na+ channel subunit in both - and -cells. Islet Na(v)1.7 channels are locked in an inactive state due to an islet cell-specific factor. Mouse pancreatic - and -cells are equipped with voltage-gated Na+ currents that inactivate over widely different membrane potentials (half-maximal inactivation (V-0.5) at -100mV and -50mV in - and -cells, respectively). Single-cell PCR analyses show that both - and -cells have Na(v)1.3 (Scn3) and Na(v)1.7 (Scn9a) subunits, but their relative proportions differ: -cells principally express Na(v)1.7 and -cells Na(v)1.3. In -cells, genetically ablating Scn3a reduces the Na+ current by 80%. In -cells, knockout of Scn9a lowers the Na+ current by >85%, unveiling a small Scn3a-dependent component. Glucagon and insulin secretion are inhibited in Scn3a(-/-) islets but unaffected in Scn9a-deficient islets. Thus, Na(v)1.3 is the functionally important Na+ channel subunit in both - and -cells because Na(v)1.7 is largely inactive at physiological membrane potentials due to its unusually negative voltage dependence of inactivation. Interestingly, the Na(v)1.7 sequence in brain and islets is identical and yet the V-0.5 for inactivation is >30mV more negative in -cells. This may indicate the presence of an intracellular factor that modulates the voltage dependence of inactivation.
  •  
9.
  • Amisten, Stefan, et al. (författare)
  • Anti-diabetic action of all- trans retinoic acid and the orphan G protein coupled receptor GPRC5C in pancreatic beta-cells
  • 2017
  • Ingår i: Endocrine Journal. - 0918-8959. ; 64:3, s. 325-338
  • Tidskriftsartikel (refereegranskat)abstract
    • Pancreatic islets express high levels of the orphan G-protein coupled receptor C5C (GPRC5C), the function of which remains to be established. Here we have examined the role of GPRC5C in the regulation of insulin secretion and beta-cell survival and proliferation using human and mouse pancreatic islets. The expression of GPRC5C was analysed by RNA-sequencing, qPCR, western blotting and confocal microscopy. Insulin secretion and cell viability were determined by RIA and MTS assays, respectively. GPRC5C mRNA expression and protein level were reduced in the islets from type-2 diabetic donors. RNA sequencing in human islets revealed GPRC5C expression correlated with the expression of genes controlling apoptosis, cell survival and proliferation. A reduction in Gprc5c mRNA and protein expression was observed in islets isolated from old mice (>46 weeks of age) compared to that in islets from newborn (<3 weeks) mice. Down-regulation of Gprc5c led to both moderately reduced glucose-stimulated insulin release and also reduced cAMP content in mouse islets. Potentiation of glucose-stimulated insulin secretion concomitant with enhanced islet cAMP level by all-trans retinoic acid (ATRA) was attenuated upon Gprc5c-KD. ATRA also increased [Ca+2](i) in Huh7-cells. Gprc5c over expression in Huh7 cells was associated with increased ERK1/2 activity. Gprc5c-KD in clonal MIN6c4 cells reduced cell proliferation and in murine islets increased apoptosis and the sensitivity of primary islet cells to a cocktail of pro-apoptotic cytokines. Our results demonstrate that agents activating GPRC5C represent a novel modality for the treatment and/or prevention of diabetes by restoring and/or maintaining functional beta-cell mass.
  •  
10.
  • Armour, Sarah L., et al. (författare)
  • Glucose Controls Glucagon Secretion by Regulating Fatty Acid Oxidation in Pancreatic α-Cells
  • 2023
  • Ingår i: DIABETES. - 0012-1797 .- 1939-327X. ; 72:10, s. 1446-1459
  • Tidskriftsartikel (refereegranskat)abstract
    • Whole-body glucose homeostasis is coordinated through secretion of glucagon and insulin from pancreatic islets. When glucose is low, glucagon is released from alpha-cells to stimulate hepatic glucose production. However, the mechanisms that regulate glucagon secretion from pancreatic alpha-cells remain unclear. Here we show that in alpha-cells, the interaction between fatty acid oxidation and glucose metabolism controls glucagon secretion. The glucose-dependent inhibition of glucagon secretion relies on pyruvate dehydrogenase and carnitine palmitoyl transferase 1a activity and lowering of mitochondrial fatty acid oxidation by increases in glucose. This results in reduced intracellular ATP and leads to membrane repolarization and inhibition of glucagon secretion. These findings provide a new framework for the metabolic regulation of the alpha-cell, where regulation of fatty acid oxidation by glucose accounts for the stimulation and inhibition of glucagon secretion.Article Highlights It has become clear that dysregulation of glucagon secretion and alpha-cell function plays an important role in the development of diabetes, but we do not know how glucagon secretion is regulated. Here we asked whether glucose inhibits fatty acid oxidation in alpha-cells to regulate glucagon secretion. We found that fatty acid oxidation is required for the inhibitory effects of glucose on glucagon secretion through reductions in ATP. These findings provide a new framework for the regulation of glucagon secretion by glucose.
  •  
Skapa referenser, mejla, bekava och länka
  • Resultat 1-10 av 45
Typ av publikation
tidskriftsartikel (44)
forskningsöversikt (1)
Typ av innehåll
refereegranskat (45)
Författare/redaktör
Rorsman, Patrik, 195 ... (43)
Zhang, Q. (12)
Salehi, Albert (11)
Tarasov, A. I. (9)
Ramracheya, R. (7)
Chibalina, M. V. (7)
visa fler...
Dou, Haiqiang, 1984 (7)
Ashcroft, Frances M. (6)
Eliasson, Lena (6)
Wernstedt Asterholm, ... (6)
Clark, A. (5)
Rorsman, N. J. G. (4)
Chibalina, Margarita ... (4)
Hamilton, Alexander (4)
Knudsen, Jakob G. (4)
Wu, Yanling, 1985 (4)
Olofsson, Charlotta ... (4)
Benrick, Anna, 1979- (4)
Vergari, E. (4)
Briant, L. J. B. (4)
Johnson, P. R. V. (4)
Knudsen, J. G. (4)
Miranda, Caroline (4)
Hamilton, A. (3)
Lundquist, Ingmar (3)
Adam, J. (3)
Renström, Erik (3)
Zhang, Quan (3)
Ramracheya, Reshma (3)
Chanclón, Belén (3)
Reimann, F. (3)
van de Bunt, M (3)
Reinbothe, Thomas, 1 ... (3)
Takahashi, H. (2)
Banke, Elin (2)
Li, Dai-Qing (2)
Seino, S. (2)
Do, H. W. (2)
Adam, Julie (2)
Tarasov, Andrei I. (2)
Rorsman, Nils J.G. (2)
Bengtsson, Martin (2)
Santos, Cristiano, 1 ... (2)
Galvanovskis, Juris (2)
Gandasi, Nikhil (2)
Zhang, Enming (2)
Bauzá-Thorbrügge, Ma ... (2)
Saliha, Musovic, 199 ... (2)
Gloyn, A. L. (2)
Kellard, J. A. (2)
visa färre...
Lärosäte
Lunds universitet (13)
Linköpings universitet (3)
Karolinska Institutet (2)
Uppsala universitet (1)
Chalmers tekniska högskola (1)
Språk
Engelska (45)
Forskningsämne (UKÄ/SCB)
Medicin och hälsovetenskap (43)
Naturvetenskap (3)

År

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Stäng

Kopiera och spara länken för att återkomma till aktuell vy