SwePub
Sök i SwePub databas

  Extended search

Träfflista för sökning "WFRF:(Regazzi Romano) "

Search: WFRF:(Regazzi Romano)

  • Result 1-6 of 6
Sort/group result
   
EnumerationReferenceCoverFind
1.
  • Cheviet, Severine, et al. (author)
  • Tomosyn-1 is involved in a post-docking event required for pancreatic beta-cell exocytosis
  • 2006
  • In: Journal of Cell Science. - : The Company of Biologists. - 0021-9533 .- 1477-9137. ; 119:14, s. 2912-2920
  • Journal article (peer-reviewed)abstract
    • Although the assembly of a ternary complex between the SNARE proteins syntaxin-1, SNAP25 and VAMP2 is known to be crucial for insulin exocytosis, the mechanisms controlling this key event are poorly understood. We found that pancreatic beta-cells express different isoforms of tomosyn-1, a syntaxin-1-binding protein possessing a SNARE-like motif. Using atomic force microscopy we show that the SNARE-like domain of tomosyn-1 can form a complex with syntaxin-1 and SNAP25 but displays binding forces that are weaker than those observed for VAMP2 (237 +/- 13 versus 279 +/- 3 pN). In pancreatic beta-cells tomosyn-1 was found to be concentrated in cellular compartments enriched in insulin-containing secretory granules. Silencing of tomosyn-1 in the rat beta-cell line INS-1E by RNA interference did not affect the number of secretory granules docked at the plasma membrane but led to a reduction in stimulus-induced exocytosis. Replacement of endogenous tomosyn-1 with mouse tomosyn-1, which differs in the nucleotide sequence from its rat homologue and escapes silencing, restored a normal secretory rate. Taken together, our data suggest that tomosyn-1 is involved in a post-docking event that prepares secretory granules for fusion and is necessary to sustain exocytosis of pancreatic beta-cells in response to insulin secretagogues.
  •  
2.
  • Eliasson, Lena, et al. (author)
  • Micro(RNA) Management and Mismanagement of the Islet
  • 2020
  • In: Journal of Molecular Biology. - : Elsevier BV. - 0022-2836. ; 432:5, s. 1419-1428
  • Research review (peer-reviewed)abstract
    • Pancreatic β-cells located within the islets of Langerhans play a central role in metabolic control. The main function of these cells is to produce and secrete insulin in response to a rise in circulating levels of glucose and other nutrients. The release of insufficient insulin to cover the organism needs results in chronic hyperglycemia and diabetes development. β-cells insure a highly specialized task and to efficiently accomplish their function they need to express a specific set of genes. MicroRNAs (miRNAs) are small noncoding RNAs and key regulators of gene expression. Indeed, by partially pairing to specific sequences in the 3′ untranslated regions of target mRNAs, each of them can control the translation of hundreds of transcripts. In this review, we focus on few key miRNAs controlling islet function and discuss: their differential expression in Type 2 diabetes (T2D), their regulation by genetic and environmental factors, and their therapeutic potential. Genetic and epigenetic changes or prolonged exposure to hyperglycemia and/or hyperlipidemia can affect the β-cell miRNA expression profile, resulting in impaired β-cell function and survival leading to the development of T2D. Experimental approaches permitting to correct the level of misexpressed miRNAs have been shown to prevent or treat T2D in animal models, suggesting that these small RNAs may become interesting therapeutic targets. However, translation of these experimental findings to the clinics will necessitate the development of innovative strategies allowing safe and specific delivery of compounds modulating the level of the relevant miRNAs to the β-cells.
  •  
3.
  •  
4.
  • Ivarsson, Rosita, et al. (author)
  • Myosin 5a controls insulin granule recruitment during late-phase secretion.
  • 2005
  • In: Traffic: the International Journal of Intracellular Transport. - : Wiley. - 1398-9219. ; 6:11, s. 1027-1035
  • Journal article (peer-reviewed)abstract
    • We have examined the importance of the actin-based molecular motor myosin 5a for insulin granule transport and insulin secretion. Expression of myosin 5a was downregulated in clonal INS-1E cells using RNAinterference. Stimulated hormone secretion was reduced by 46% and single-cell exocytosis, measured by capacitance recordings, was inhibited by 42% after silencing. Silencing of Slac-2c/MYRIP, which links insulin granules to myosin 5a, resulted in similar inhibition of single-cell exocytosis. Antibody inhibition of the myosin 5a-Slac-2c/MYRIP interaction significantly reduced the recruitment of insulin granules for release. The pool of releasable granules independent of myosin 5a activity was estimated to approximately 550 granules. Total internal reflection microscopy was then applied to directly investigate granule recruitment to the plasma membrane. Silencing of myosin 5a inhibited granule recruitment during late phase of insulin secretion. In conclusion, we propose a model where insulin granules are transported through the actin network via both myosin 5a-mediated transport and via passive diffusion, with the former playing the major role during stimulatory conditions.
  •  
5.
  • Motterle, Anna, et al. (author)
  • Identification of islet-enriched long non-coding RNAs contributing to β-cell failure in type 2 diabetes
  • 2017
  • In: Molecular Metabolism. - : Elsevier BV. - 2212-8778. ; 6:11, s. 1407-1418
  • Journal article (peer-reviewed)abstract
    • Objective: Non-coding RNAs constitute a major fraction of the β-cell transcriptome. While the involvement of microRNAs is well established, the contribution of long non-coding RNAs (lncRNAs) in the regulation of β-cell functions and in diabetes development remains poorly understood. The aim of this study was to identify novel islet lncRNAs differently expressed in type 2 diabetes models and to investigate their role in β-cell failure and in the development of the disease. Methods: Novel transcripts dysregulated in the islets of diet-induced obese mice were identified by high throughput RNA-sequencing coupled with de novo annotation. Changes in the level of the lncRNAs were assessed by real-time PCR. The functional role of the selected lncRNAs was determined by modifying their expression in MIN6 cells and primary islet cells. Results: We identified about 1500 novel lncRNAs, a number of which were differentially expressed in obese mice. The expression of two lncRNAs highly enriched in β-cells, βlinc2, and βlinc3, correlated to body weight gain and glycemia levels in obese mice and was also modified in diabetic db/. db mice. The expression of both lncRNAs was also modulated in vitro in isolated islet cells by glucolipotoxic conditions. Moreover, the expression of the human orthologue of βlinc3 was altered in the islets of type 2 diabetic patients and was associated to the BMI of the donors. Modulation of the level of βlinc2 and βlinc3 by overexpression or downregulation in MIN6 and mouse islet cells did not affect insulin secretion but increased β-cell apoptosis. Conclusions: Taken together, the data show that lncRNAs are modulated in a model of obesity-associated type 2 diabetes and that variations in the expression of some of them may contribute to β-cell failure during the development of the disease.
  •  
6.
  • Stoll, Lisa, et al. (author)
  • A circular RNA generated from an intron of the insulin gene controls insulin secretion
  • 2020
  • In: Nature Communications. - : Springer Science and Business Media LLC. - 2041-1723. ; 11:1
  • Journal article (peer-reviewed)abstract
    • Fine-tuning of insulin release from pancreatic β-cells is essential to maintain blood glucose homeostasis. Here, we report that insulin secretion is regulated by a circular RNA containing the lariat sequence of the second intron of the insulin gene. Silencing of this intronic circular RNA in pancreatic islets leads to a decrease in the expression of key components of the secretory machinery of β-cells, resulting in impaired glucose- or KCl-induced insulin release and calcium signaling. The effect of the circular RNA is exerted at the transcriptional level and involves an interaction with the RNA-binding protein TAR DNA-binding protein 43 kDa (TDP-43). The level of this circularized intron is reduced in the islets of rodent diabetes models and of type 2 diabetic patients, possibly explaining their impaired secretory capacity. The study of this and other circular RNAs helps understanding β-cell dysfunction under diabetes conditions, and the etiology of this common metabolic disorder.
  •  
Skapa referenser, mejla, bekava och länka
  • Result 1-6 of 6

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 Close

Copy and save the link in order to return to this view