21
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Intracellular free fatty acid upholds β-cell glucose competence: The role of peroxisome proliferator-activated receptor δ and mitochondrial metabolism

      article-commentary

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          What is Known Regarding β-Cell Intracellular Fatty Acid in Relation to Glucose-Stimulated insulin Secretion? Elevation of extracellular glucose causes fusion of the β granules and the plasma membrane as a result of increased submembrane Ca2+ concentration. A raised adenosine triphosphate (ATP)-to-adenosine diphosphate (ADP) ratio is pivotal to this process, as it causes closure of the ATP-sensitive K+ (KATP) channel, membrane depolarization, opening of voltage-dependent calcium channels and finally Ca2+ influx from the cell exterior. Soon thereafter, glucose-stimulated insulin secretion (GSIS) is augmented as the releasable pool of β granules is replenished. Although the molecular basis of this augmentation has not been fully defined, free fatty acid (FFA) has been strongly implicated as having an indispensable role in this process. Quantities of FFA in the micromolar range, and therefore too minute to generate classic metabolic coupling factor(s), such as ATP for insulin exocytosis, enhance GSIS when added to the incubation in conjunction with a stimulatory concentration of glucose1. Furthermore, pre-exposure of β-cells to a similarly low concentration of FFA in the absence of a stimulatory concentration of glucose primes the cells so that the insulin release (IR) subsequently provoked by any stimulation is enhanced compared with that in β-cells not treated with FFA1. Continued exposure of β-cells to a high concentration of glucose causes an anaplerotic output of citrate from the tricarboxylic acid (TCA) cycle, elevating cytosolic malonyl-CoA, which causes suppression of carnitine palmitoyltransferase 1 leading to decreased FFA entry to mitochondria2. The subsequent accumulation of cytosolic FFA might increase insulin secretion through fatty acylation of key protein(s) involved in exocytosis3. In contrast, increased cellular fatty acyl-CoA (FA-CoA) could also enhance IR through the glycerolipid/FFA cycle2. Knockdown of desnutrin/adipose triglyceride lipase (ATGL) in β-cells by short hairpin ribonucleic acid suppressed GSIS4. All of these data show that acute elevation and lowering of cellular FFA in β-cells, respectively, enhanced and suppressed GSIS. However, the downstream effector or effectors of FFA regulation of insulin secretion have remained elusive, especially under the conditions of sustained β-cell specific lowering of intracellular FFA. Newer Insights Obtained by β-Cell Specific Desnutrin/ATGL Knockout In an elegant recent study, Tang et al.5 identified a novel downstream signaling pathway for intracellular FFA in pan-creatic β-cells: activation of peroxis-ome proliferator-activated receptor δ (PPARδ) followed by enhanced mitochondrial metabolism. This was accomplished by using β-cell specific desnutrin/ATGL knockout (desnutrin βKO) mice (Figure1). The desnutrin βKO mouse showed postprandial and postglucose hyperglycemia with blunted nutrient- or glucose-induced insulin secretion. Interestingly, the islets were enlarged and the insulin content increased by approximately 50%, while there was no evidence of increased insulin synthesis, so that islet insulin appeared to have increased as a result of decreased IR. Because desnutrin hydrolyzes triacylglycerol (TAG), TAG accumulated and the FFA content and glycerol output were reduced in the islets of the KO mouse. Extracellular application of oleate (concentration unspecified) in the presence of a stimulatory concentration of glucose failed to acutely elevate insulin exocytosis in the β-cells of the KO mouse. This indicated that a constitutive, normal level of intracellular FFA by desnutrin is required to uphold the β-cell machinery for IR competence in response not only to glucose, but also to fatty acid. IR directly triggered by high K+-induced membrane depolarization in the presence or absence of the KATP channel opener, diazoxide, remained unaffected in the KO mouse. In other words, the FFA produced by desnutrin tonically maintains the nutrient-induced IR in the β-cells. Using mitochondrial membrane potential markers, Tang et al.5 further showed that the loss of desnutrin/reduced TAG hydrolysis was associated with impaired mitochondrial metabolism, which was in this case as a result of defective activation of PPARδ. Adenoviral transfection of desnutrin and pharmacological activation of PPARδ mostly restored the altered phenotype of the desnutrin KO mouse, implying that the absence of desnutrin was in fact causal for impaired PPARδ activation and the series of phenotypic, cellular and subcellular abnormalities in the β-cell specific desnutrin KO mouse/islets. Feeding a normal mouse on a high-fat diet for 8 weeks caused 50% suppression of desnutrin protein and messenger ribonucleic acid in the islet cells. Figure 1 A proposed newer view on the free fatty acid (FFA) signaling in (a) pancreatic β-cells, on the basis of the findings in (b) the β-cell specific desnutrin knockout mouse. The novel pathway shown by Tang et al.5 is shown in blue. Downward or upward arrows indicate increase or decrease, respectively, in the substrate or signaling. (b) Faintness in blue implies attenuated signaling. The scheme is focused on the topic of the work of Tang et al.5, and is not intended to cover all aspects of β-cell metabolism-secretion coupling. ADP, adenosine diphosphate; ATP, adenosine triphosphate; KATP channel, adenosine triphosphate-sensitive K+ channel; LC-CoA, long-chain acyl-CoA; PPARδ, peroxisome proliferator-activated receptor-δ; TAG, triacylglycerol; VDCC, voltage-dependent calcium channel. Critical Appraisal The β-cell specific desnutrin KO mouse shows a very unique phenotype from the viewpoint of islet physiology and pathophysiology. The desnutrin βKO mouse has large islets with increased insulin content in the absence of attenuated insulin sensitivity in vivo. Having large islets with increased insulin content is a typical phenotype for animals fed a high-fat diet, and decreased whole-body insulin sensitivity with increased insulin synthesis is the rule under this condition. Nevertheless, in the desnutrin βKO mouse, there was no enhancement of insulin synthesis as indexed by expression of the messenger ribonucleic acid. Despite increased insulin islet content, basal insulin secretion was not at all elevated either in vivo or in vitro. Although GSIS by the islets was depressed, the IR triggered by a depolarizing concentration of K+ was normal, with no increase. Taken together, these findings imply that, in the KO mouse: (i) the islet insulin content increases mostly, if not entirely, as a consequence of reduced insulin secretion; that is, the accumulation of insulin in the β-cells caused by impaired secretion; and (ii) the distribution of β granules clearly shifts to the reserve, or poorly releasable, pool. In other words, there is a diminution in the size of the readily releasable pool of β granules. It should be noted that TAG hydrolysis by desnutrin was not the only source of FFA in the cells. In addition to TAG hydrolysis by desnutrin, an influx of FFA from outside the cells, a de novo accumulation of FFA in part as a result of the inhibition of carnitine palmitoyltransferase-1 (CPT-1) by malonyl-CoA and hydrolysis of diacyl glycerol by hormone sensitive lipase all contributed to maintaining cellular FFA in the β-cells. Accordingly, the level of islet free FFA in the desnutrin KO mouse was modestly (approximately 40%), but not radically, suppressed. Therefore, it can be inferred that the deranged in vivo and in vitro phenotype of the KO mouse was a result of attenuation, not total obliteration, of FFA signaling. Thus, it can be assumed that a certain level of intracellular FFA is a required positive effector for the maintenance of the glucose competence of the IR machinery. There is a similarity between the data for the β-cell specific desnutrin KO mouse and the β-cells of fasted rats6. In the latter case, nutrient-induced, but not depolarization-induced, IR is preferentially suppressed with selective impairment of the mitochondrial metabolism. However, there is a crucial difference between the two conditions in that islet insulin content increased in the former, but decreased in the latter. The data from the IR experiments was presented ‘per islet’; that is, it was not adjusted for increased islet insulin content. The values for basal insulin output in the presence of substimulatory concentrations of glucose, so called constitutive release, might have been significantly lower if they had been expressed ‘per islet insulin content’. Also, the degree of suppression in GSIS would have been much more pronounced if the data had been expressed as ‘per islet insulin content’. Similarly, IR directly triggered by high K+ could have been more appropriately interpreted as ‘decreased’ if the increased insulin content of the islets had been taken into account. The authors concluded that the treatment with PPARδ agonist of the KO mouse for 2 weeks caused ‘normalization’ of GSIS in the islets of the KO mouse. However, knowing that insulin content is grossly increased in the islets of the KO mouse, we are afraid that this interpretation might have been too simplistic. In other words, it appears that treatment with the PPARδ agonist only partially restored the relative decrease of the releasable pool of β granules. This finding strongly indicates the existence of PPARδ-independent, insulinotropic signaling of FFA in the β-cells. If the imbalance in the distribution of β granules had been completely corrected by the PPARδ agonist, GSIS in the agonist-treated islets should have been significantly greater than in the control islets. Nevertheless, the novel findings in the present study, especially the PPARδ mediation of the effects of FFA, constitute a valuable addition to previously accumulated knowledge relating to the role of FFA in β-cells. Because the effect of PPARδ would have been manifested through changes in gene transcription and protein synthesis within a timeframe of several hours, this mechanism provides an elegant explanation for the chronic, sustained effects of FFA in β-cells. If the data from the heterozygous desnutrin KO mouse and from those fed a high-fat diet with a halving of desnutrin had been somewhere in between homozygotes and the control mice, desnutrin might have been appropriately described as a ‘regulator’ of IR. Although activation of PPARδ and the mitochondrial metabolism, especially raised ATP/ADP, were implicated as downstream effectors of FFA, pharmacological activation of PPARδ in flox mouse did not cause upregulation of IR. Conclusions Free fatty acid signaling in islet β-cells is complicated. Tang et al.5 have identified PPARδ-mediated activation of mitochondrial metabolism as a novel downstream effector of FFA in a comprehensive study of the β-cell specific desnutrin KO mouse. Specific pharmacological modulation of this signaling branch, if possible, would provide an innovative treatment for type 2 diabetes.

          Related collections

          Most cited references6

          • Record: found
          • Abstract: found
          • Article: not found

          Metabolic signaling in fuel-induced insulin secretion.

          The pancreatic islet β cell senses circulating levels of calorigenic nutrients to secrete insulin according to the needs of the organism. Altered insulin secretion is linked to various disorders such as diabetes, hypoglycemic states, and cardiometabolic diseases. Fuel stimuli, including glucose, free fatty acids, and amino acids, promote insulin granule exocytosis primarily via their metabolism in β cells and the production of key signaling metabolites. This paper reviews our current knowledge of the pathways involved in both positive and negative metabolic signaling for insulin secretion and assesses the role of established and candidate metabolic coupling factors, keeping recent developments in focus. Copyright © 2013 Elsevier Inc. All rights reserved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Desnutrin/ATGL activates PPARδ to promote mitochondrial function for insulin secretion in islet β cells.

            Excessive caloric intake leading to obesity is associated with insulin resistance and dysfunction of islet β cells. High-fat feeding decreases desnutrin (also called ATGL/PNPLA2) levels in islets. Here we show that desnutrin ablation via RIP-Cre (βKO) or RIP-CreER results in hyperglycemia with impaired glucose-stimulated insulin secretion (GSIS). Due to decreased lipolysis, islets have higher TAG content but lower free FA levels. βKO islets exhibit impaired mitochondrial respiration and lower production of ATP required for GSIS, along with decreased expression of PPARδ target genes involved in mitochondrial oxidation. Furthermore, synthetic PPARδ, but not PPARα, agonist restores GSIS and expression of mitochondrial oxidative genes in βKO mice, revealing that desnutrin-catalyzed lipolysis generates PPARδ ligands. Finally, adenoviral expression of desnutrin in βKO islets restores all defects of βKO islet phenotype and function, including GSIS and mitochondrial defects, demonstrating the critical role of the desnutrin-PPARδ-mitochondrial oxidation axis in regulating islet β cell GSIS.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Adipose triglyceride lipase is implicated in fuel- and non-fuel-stimulated insulin secretion.

              Reduced lipolysis in hormone-sensitive lipase-deficient mice is associated with impaired glucose-stimulated insulin secretion (GSIS), suggesting that endogenous beta-cell lipid stores provide signaling molecules for insulin release. Measurements of lipolysis and triglyceride (TG) lipase activity in islets from HSL(-/-) mice indicated the presence of other TG lipase(s) in the beta-cell. Using real time-quantitative PCR, adipose triglyceride lipase (ATGL) was found to be the most abundant TG lipase in rat islets and INS832/13 cells. To assess its role in insulin secretion, ATGL expression was decreased in INS832/13 cells (ATGL-knockdown (KD)) by small hairpin RNA. ATGL-KD increased the esterification of free fatty acid (FFA) into TG. ATGL-KD cells showed decreased glucose- or Gln + Leu-induced insulin release, as well as reduced response to KCl or palmitate at high, but not low, glucose. The K(ATP)-independent/amplification pathway of GSIS was considerably reduced in ATGL-KD cells. ATGL(-/-) mice were hypoinsulinemic and hypoglycemic and showed decreased plasma TG and FFAs. A hyperglycemic clamp revealed increased insulin sensitivity and decreased GSIS and arginine-induced insulin secretion in ATGL(-/-) mice. Accordingly, isolated islets from ATGL(-/-) mice showed reduced insulin secretion in response to glucose, glucose + palmitate, and KCl. Islet TG content and FFA esterification into TG were increased by 2-fold in ATGL(-/-) islets, but glucose usage and oxidation were unaltered. The results demonstrate the importance of ATGL and intracellular lipid signaling for fuel- and non-fuel-induced insulin secretion.
                Bookmark

                Author and article information

                Journal
                J Diabetes Investig
                J Diabetes Investig
                jdi
                Journal of Diabetes Investigation
                BlackWell Publishing Ltd (Oxford, UK )
                2040-1116
                2040-1124
                March 2015
                15 July 2014
                : 6
                : 2
                : 133-136
                Affiliations
                [1 ]Division of Diabetes Endocrinology and Metabolism, Department of Internal Medicine, Shinshu University School of Medicine Matsumoto, Japan
                [2 ]Diabetes Center, Aizawa Hospital Matsumoto, Japan
                Author notes
                * Corresponding author. Toru Aizawa, Tel.: +81-263-33-8600, Fax: +81-263-33-8609, E-mail address: taizawax@ 123456ai-hosp.or.jp
                Article
                10.1111/jdi.12257
                4364846
                77386f42-372f-47d1-ae73-350d2dac6257
                © 2014 The Authors. Journal of Diabetes Investigation published by Asian Association of the Study of Diabetes (AASD) and Wiley Publishing Asia Pty Ltd

                This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.

                History
                : 11 May 2014
                : 30 May 2014
                : 02 June 2014
                Categories
                Commentaries

                Comments

                Comment on this article