Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1872 word(s) 1872 2021-07-31 17:24:01

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Von Loeffelholz, C. AMPK. Encyclopedia. Available online: https://encyclopedia.pub/entry/13299 (accessed on 25 April 2024).
Von Loeffelholz C. AMPK. Encyclopedia. Available at: https://encyclopedia.pub/entry/13299. Accessed April 25, 2024.
Von Loeffelholz, Christian. "AMPK" Encyclopedia, https://encyclopedia.pub/entry/13299 (accessed April 25, 2024).
Von Loeffelholz, C. (2021, August 18). AMPK. In Encyclopedia. https://encyclopedia.pub/entry/13299
Von Loeffelholz, Christian. "AMPK." Encyclopedia. Web. 18 August, 2021.
AMPK
Edit

5′AMP-activated protein kinase (AMPK) is known as metabolic sensor in mammalian cells that becomes activated by an increasing adenosine monophosphate (AMP)/adenosine triphosphate (ATP) ratio. The heterotrimeric AMPK protein comprises three subunits, each of which has multiple phosphorylation sites, playing an important role in the regulation of essential molecular pathways. By phosphorylation of downstream proteins and modulation of gene transcription AMPK functions as a master switch of energy homeostasis in tissues with high metabolic turnover, such as the liver, skeletal muscle, and adipose tissue.

lipotoxicity insulin resistance free fatty acids steatohepatitis type 2 diabetes skeletal muscle adipose tissue diacylglycerol

1. Introduction

The healthy liver displays remarkable metabolic plasticity with hepatocytes in concert with a variety of other cell types, readily switching between various metabolic tasks. These transitions are regulated by numerous factors including hormones, nutrients, and further influences. However, with the onset of non-alcoholic fatty liver disease (NAFLD), this highly balanced physiological homeostasis becomes compromised [1].

It was shown in vivo that de novo lipogenesis (DNL) contributes substantially to liver fat storage in humans [2]. One hallmark finding in that matter has been the discovery of the regulation of lipid metabolism by 5′AMP-activated protein kinase (AMPK) via phosphorylation and inactivation of acetyl CoA carboxylase (ACC), the enzyme catalyzing the first and rate limiting step in DNL [3][4][5][6]. Moreover, a close association of ectopic lipid deposition with insulin resistance is well recognized. This supports the hypothesis of a crucial role of AMPK as molecular node between the epidemic pathologies obesity, type 2 diabetes (T2D), and NAFLD [7][8][9][10][11][12][13][14].

2. Structure and Physiological Regulation of AMPK

Eukaryotic cells adapt to environmental energetic fluctuations by constantly managing energy consumption and the capacity to produce adenosine triphosphate (ATP). ATP is broken down into adenosine diphosphate (ADP) and can be further converted to adenosine monophosphate (AMP). When intracellular ATP levels decrease while AMP levels rise, cellular metabolism must shift towards ATP-producing catabolic pathways [15]. AMPK was recognized as mammalian homolog of a stress sensing kinase in yeast that is critical for cellular survival under conditions of marginal energy supply [5][16][17]). AMPK becomes allosterically activated by binding adenine nucleotides and thereby functions as a sensor of intracellular energy homeostasis [18][19].
From a biochemical perspective AMPK is a phylogenetically highly conserved heterotrimeric serine/threonine protein kinase complex consisting of one catalytic (α) and two regulatory subunits (β, γ). Various isoforms of each subunit are encoded by individual genes [18][20][21][22]. They are expressed tissue specific and the resulting AMPK subunits can either exert overlapping or distinct functions [23][24][25][26][27][28][29][30]. Depending on isoform expression and combination, twelve heterotrimeric holoenzyme-variants can theoretically be deduced [22]. Excellent reviews on structure, tissue related subunit expression, the multitude of phosphorylation sites, and subcellular localization are available, to which the specifically interested reader is referred [18][22][30][31][32][33].
Multiple endocrine, (patho-) physiological, and pharmacological stimuli are known to modulate AMPK . The phosphorylation of a conserved threonine-172 (Thr172) residue in the activation loop of the N-terminal region of the α-subunit is known to be required for full AMPK activation by upstream kinases in mammalian cells (reviewed in [30]). Using purified recombinant enzyme preparations, Suter et al. have shown that upstream kinase activation in the presence of saturating AMP effects resulted in a 1000-fold increased activation of α1-β1-γ1- and α2-β2-γ1 AMPK heterotrimers [34]. Interestingly, AMP did not enforce kinase dependent phosphorylation, at least under the research conditions chosen by the authors, but protein phosphatase mediated deactivation by site specific dephosphorylation was prevented [34]. Meanwhile, these findings have been challenged and complemented by a broad body of further research, and AMP binding is suggested to modify the AMPK protein by allowing more effective upstream stimulation. Moreover, augmented allosteric activation of still Thr172 phosphorylated AMPK by AMP and further mechanisms have been discussed [20][35][36][37]. Together, available evidence generally supports the view that for full enzyme activation adenine nucleotide (AMP, ADP) binding is essential for Thr172 phosphorylation in the activation loop of the α-catalytic domain [18][36]. Adenine nucleotide binding is realized by three adenine nucleotide binding sites (ANBS), which are localized in three out of four cystathionine-β-synthase repeats (CBS1-4) in the AMPK γ-subunit [30]. CBS4 was shown to exclusively and permanently bind AMP [38][39], while CBS1 and CBS3 competitively bind AMP, ADP, or ATP, depending on their respective cellular abundance [18]. This elegantly enables the AMPK protein complex to rapidly sense changes in AMP:ATP ratio and immediately balance cellular energy supply [35]. Another structural key element in the AMPK γ-subunit is CBS2. CBS2 is discussed to be closely related to ACC phosphorylation [40]. The β-subunit is of extraordinary importance for mediating metabolic effects and contrasting with humans, the rodent liver predominately expresses β1, while the β2-subunit appears to be of minor importance in this species [20][24][41][42][43][44]. The β-subunit enables AMPK to bind glycogen via a carbohydrate binding module (β-CBM), augmenting its energy sensitizing properties [35]. Moreover, it was actually shown by Steinberg’s group that activated long chain fatty acid esters can allosterically activate β1-subunit containing isoforms and thereby increase lipid oxidation through phosphorylation of ACC [45].
Due to the core functional position in metabolism, AMPK was recognized as a promising pharmacologic target structure. Metfomin, a drug widely used in the therapy of T2D, exerts some of its main effects by AMPK activation in the liver and skeletal muscle [46][47]. Increasing AMPK activity is widely viewed as viable therapeutic strategy [48]. Stimulation of AMPK not alone inhibits ACC1, but further leads to concomitant phosphorylation of ACC2 and activation of malonyl CoA decarboxylase (MCD), resulting in an acute reduction of cytosolic malonyl CoA [49]. Malonyl CoA is well known as negative regulator of fatty acid oxidation by virtue of its ability to inhibit carnitine palmitoyl-transferase 1 (CPT1). Thus, when malonyl CoA levels decrease, CPT1 remains uninhibited and fatty acid oxidation becomes accelerated [50][51][52][53].

3. The Role of AMPK in Human Fat Depots

Based on the close association of NAFLD and obesity, AT expansion is common in fatty liver patients. The shift of fat storage away from subcutaneous (SCAT) towards visceral white adipose tissue (VAT) is considered a predictor of increased risk profile [54][55].
AMPK is known to be expressed in AT and to exert relevant metabolic functions (reviewed in [48]). Under physiological conditions AMPK is primarily discussed to contribute to AT insulin sensitivity [48]. Expression appears to be higher in human SCAT compared to VAT, at least in morbid obesity (BMI ≥ 40 kg/m2; n = 17), and after short term starvation [56]. Reduced AMPK activity, as estimated by Thr172 phosphorylation of αAMPK in relation to total protein, positively correlated with insulin resistance in obese humans [57][58]. However, the number of studied subjects was limited, all were morbidly obese, and the homeostasis model of insulin resistance (HOMA-IR) as a relatively rough estimate was used to evaluate the efficacy of insulin signaling [57][58]. Our group has studied AMPK expression and activation in VAT and SCAT of NAFLD patients . However, we were unable to observe any significant differences between groups, yet subjects were matched for main confounding factors. Contrasting with the studies of Ruderman’s group [57][58], our patients were not morbidly obese, and NAFLD was evaluated by gold standard methods. However, we did also use the HOMA-IR for estimating insulin resistance and did not perform functional analyses. Moreover, our control subjects were overweight and we cannot exclude that this could have biased the results. Grisouard et al. differentiated human preadipocytes in vitro and studied the effects of metformin stimulation on glucose uptake [59]. Metformin increased glucose uptake more than twofold, accompanied by a rise in GLUT4 and glucose oxidation. Silencing of AMPKα1 counteracted the effects [59]. Available evidence therefore supports the hypothesis that AMPK contributes to peripheral insulin sensitivity. Pharmacological AMPK activation in a manner supporting cellular glucose uptake with consecutively enforced FFA esterification could help to control for hyperlipolysis. Insulin mediated lipolysis suppression showed a continuous worsening with rising liver fat accumulation [60] and hyperlipolysis is considered a major contributor of NAFLD (reviewed in [61][62]). Hyperlipolysis is further aggravated when adipocytes under conditions of a repetitively positive energy balance become hypertrophic. This can result in systemic low-grade inflammation, as promoted by macrophages residing in AT with worsening of insulin resistance [14][61][63][64][65][66]. However, according to numerous experimental and animal models, the role of AMPK on AT lipolysis remains inconclusive, with existing evidence supporting both enhanced and suppressed lipolysis by AMPK [30][67][68][69][70][71][72][73][74]. We are aware of no research data specifically addressing this issue in human NAFLD. However, indirect evidence to support the hypothesis of suppressed lipolysis (and/or elevated fat oxidation) in humans via AMPK comes from a study using an AICAR stimulation protocol [75]. Given the percentage of AT in relation to total body mass, further research on this principle could provide interesting insights on the role of AMPK, particularly regarding peripheral insulin resistance in various AT depots [76]. This principle is also supported by animal data showing that abolishing AMPK in AT results in decreased whole body insulin sensitivity and enhanced liver triacylglycerol accumulation [77]. Furthermore, human adipocytes in principal have the capability of DNL [78]. However, in contrast to rodents it is known that AT is not a significant site of DNL in humans [79][80].

4. Skeletal Muscle Is a Predominant Target Tissue of Insulin and Sensitive to AMPK Activation

Skeletal muscle is the most important site of insulin stimulated glucose disposal in human body (reviewed in [62][81]). Liver triacylglycerol content can be interpreted as direct barometer of whole-body insulin resistance, with maximally impaired insulin signaling in skeletal muscle being reached when hepatic triacylglycerol levels range about 6% [60]. AMPK is expressed at high levels in skeletal muscle, and under physiological conditions the kinase contributes to insulin sensitivity and insulin independent exercise related peripheral glucose uptake.
A number of studies have addressed the role of AMPK in human skeletal muscle. The majority of trials were of short duration and included limited subject numbers. Only one study considered NAFLD [82]. The studies can be categorized in (i) descriptive studies focusing basal AMPK activity in obesity/insulin resistance [83][84][85], (ii) interventional studies using AMPK mimetics under in vivo or ex vivo conditions [75][82][84][86][87][88], and (iii) exercise studies (e.g., [87][89][90][91][92]). It was shown that after 12–18 h of starvation basal AMPK activity and expression remained comparable in muscle of lean and obese subjects [84]. This was also reported for T2D patients and controls [83][85][86]. However, patients in several of these studies were under antidiabetic medication including metformin, with no relevant drug free interval before intervention [86][88]. Thus, the results must be interpreted carefully.

5. Dysregulation of Hepatic AMPK in Humans

DNL is a main contributor to hepatic triacylglycerol accumulation under conditions of caloric oversupply, with glucose representing the main DNL substrate. The quantitatively only relevant site of DNL in human metabolism is the liver [79]. Therefore, attempts to modulate DNL in a therapeutic manner by means of AMPK activation should primarily address hepatic isoforms. Under physiological conditions AMPK is considered to balance hepatocellular energy demands, with major influence on liver insulin sensitivity and lipid homeostasis [29].
The main predictor of NAFLD is hepatic insulin resistance, defined by the failure of insulin to suppress endogenous glucose production, e.g., gluconeogenesis [61][93]. Under physiological conditions DNL, like gluconeogenesis, is under slow but potent transcriptional control by an insulin dependent mechanism [62]. When hepatocellular insulin signaling becomes attenuated DNL activation remains paradoxically uncompromised. This is referred to as selective insulin resistance, resulting in fasting hyperglycemia and uncontrolled DNL at the same time [94]. Corresponding to the human study of Lambert et al. [95], fasting hyperglycemia is a significant driver of DNL since glucose independently from insulin can enter hepatocytes in large amounts via GLUT2, and thereby enter glycolysis .

 

References

  1. Byrne, C.D.; Targher, G. NAFLD: A multisystem disease. J. Hepatol. 2015, 62, S47–S64.
  2. Donnelly, K.L.; Smith, C.I.; Schwarzenberg, S.J.; Jessurun, J.; Boldt, M.D.; Parks, E.J. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J. Clin. Investig. 2005, 115, 1343–1351.
  3. Carling, D.; Clarke, P.R.; Zammit, V.A.; Hardie, D.G. Purification and characterization of the AMP-activated protein kinase. Copurification of acetyl-CoA carboxylase kinase and 3-hydroxy-3-methylglutaryl-CoA reductase kinase activities. Eur. J. Biochem. 1989, 186, 129–136.
  4. Hardie, D.G. Regulation of fatty acid and cholesterol metabolism by the AMP-activated protein kinase. Biochim. Biophys. Acta 1992, 1123, 231–238.
  5. Woods, A.; Munday, M.R.; Scott, J.; Yang, X.; Carlson, M.; Carling, D. Yeast SNF1 is functionally related to mammalian AMP-activated protein kinase and regulates acetyl-CoA carboxylase in vivo. J. Biol. Chem. 1994, 269, 19509–19515.
  6. Fullerton, M.D.; Galic, S.; Marcinko, K.; Sikkema, S.; Pulinilkunnil, T.; Chen, Z.-P.; O’Neill, H.M.; Ford, R.J.; Palanivel, R.; O’Brien, M.; et al. Single phosphorylation sites in Acc1 and Acc2 regulate lipid homeostasis and the insulin-sensitizing effects of metformin. Nat. Med. 2013, 19, 1649–1654.
  7. Lewis, G.F.; Carpentier, A.; Adeli, K.; Giacca, A. Disordered fat storage and mobilization in the pathogenesis of insulin resistance and type 2 diabetes. Endocr. Rev. 2002, 23, 201–229.
  8. Gastaldelli, A.; Cusi, K.; Pettiti, M.; Hardies, J.; Miyazaki, Y.; Berria, R.; Buzzigoli, E.; Sironi, A.M.; Cersosimo, E.; Ferrannini, E.; et al. Relationship between hepatic/visceral fat and hepatic insulin resistance in nondiabetic and type 2 diabetic subjects. Gastroenterology 2007, 133, 496–506.
  9. Zhang, C.; Klett, E.L.; Coleman, R.A. Lipid signals and insulin resistance. Clin. Lipidol. 2013, 8, 659–667.
  10. Szendroedi, J.; Roden, M. Ectopic lipids and organ function. Curr. Opin. Lipidol. 2009, 20, 50–56.
  11. Korenblat, K.M.; Fabbrini, E.; Mohammed, B.S.; Klein, S. Liver, muscle, and adipose tissue insulin action is directly related to intrahepatic triglyceride content in obese subjects. Gastroenterology 2008, 134, 1369–1375.
  12. Birkenfeld, A.L.; Shulman, G.I. Nonalcoholic fatty liver disease, hepatic insulin resistance, and type 2 diabetes. Hepatology 2014, 59, 713–723.
  13. Häring, H.-U. Novel phenotypes of prediabetes? Diabetologia 2016, 59, 1806–1818.
  14. Samuel, V.T.; Shulman, G.I. The pathogenesis of insulin resistance: Integrating signaling pathways and substrate flux. J. Clin. Investig. 2016, 126, 12–22.
  15. Dashty, M. A quick look at biochemistry: Carbohydrate metabolism. Clin. Biochem. 2013, 46, 1339–1352.
  16. Carling, D.; Aguan, K.; Woods, A.; Verhoeven, A.J.; Beri, R.K.; Brennan, C.H.; Sidebottom, C.; Davison, M.D.; Scott, J. Mammalian AMP-activated protein kinase is homologous to yeast and plant protein kinases involved in the regulation of carbon metabolism. J. Biol. Chem. 1994, 269, 11442–11448.
  17. Hong, S.-P.; Leiper, F.C.; Woods, A.; Carling, D.; Carlson, M. Activation of yeast Snf1 and mammalian AMP-activated protein kinase by upstream kinases. Proc. Natl. Acad. Sci. USA 2003, 100, 8839–8843.
  18. Hardie, D.G.; Schaffer, B.E.; Brunet, A. AMPK: An Energy-Sensing Pathway with Multiple Inputs and Outputs. Trends Cell Biol. 2016, 26, 190–201.
  19. Garcia, D.; Shaw, R.J. AMPK: Mechanisms of Cellular Energy Sensing and Restoration of Metabolic Balance. Mol. Cell 2017, 66, 789–800.
  20. Viollet, B.; Horman, S.; Leclerc, J.; Lantier, L.; Foretz, M.; Billaud, M.; Giri, S.; Andreelli, F. AMPK inhibition in health and disease. Crit. Rev. Biochem. Mol. Biol. 2010, 45, 276–295.
  21. Schultze, S.M.; Hemmings, B.A.; Niessen, M.; Tschopp, O. PI3K/AKT, MAPK and AMPK signalling: Protein kinases in glucose homeostasis. Expert Rev. Mol. Med. 2012, 14, e1.
  22. Ross, F.A.; MacKintosh, C.; Hardie, D.G. AMP-activated protein kinase: A cellular energy sensor that comes in 12 flavours. FEBS J. 2016, 283, 2987–3001.
  23. Yan, Y.; Zhou, X.E.; Xu, H.E.; Melcher, K. Structure and Physiological Regulation of AMPK. Int. J. Mol. Sci. 2018, 19, 3534.
  24. Thornton, C.; Snowden, M.A.; Carling, D. Identification of a novel AMP-activated protein kinase beta subunit isoform that is highly expressed in skeletal muscle. J. Biol. Chem. 1998, 273, 12443–12450.
  25. Cheung, P.C.; Salt, I.P.; Davies, S.P.; Hardie, D.G.; Carling, D. Characterization of AMP-activated protein kinase gamma-subunit isoforms and their role in AMP binding. Biochem. J. 2000, 346, 659–669.
  26. Crute, B.E.; Seefeld, K.; Gamble, J.; Kemp, B.E.; Witters, L.A. Functional domains of the alpha1 catalytic subunit of the AMP-activated protein kinase. J. Biol. Chem. 1998, 273, 35347–35354.
  27. Lee, C.-W.; Wong, L.L.-Y.; Tse, E.Y.-T.; Liu, H.-F.; Leong, V.Y.-L.; Lee, J.M.-F.; Hardie, D.G.; Ng, I.O.-L.; Ching, Y.-P. AMPK promotes p53 acetylation via phosphorylation and inactivation of SIRT1 in liver cancer cells. Cancer Res. 2012, 72, 4394–4404.
  28. Fox, M.M.; Phoenix, K.N.; Kopsiaftis, S.G.; Claffey, K.P. AMP-Activated Protein Kinase α 2 Isoform Suppression in Primary Breast Cancer Alters AMPK Growth Control and Apoptotic Signaling. Genes Cancer 2013, 4, 3–14.
  29. Jeon, S.-M. Regulation and function of AMPK in physiology and diseases. Exp. Mol. Med. 2016, 48, e245.
  30. Wang, Q.; Liu, S.; Zhai, A.; Zhang, B.; Tian, G. AMPK-Mediated Regulation of Lipid Metabolism by Phosphorylation. Biol. Pharm. Bull. 2018, 41, 985–993.
  31. Steinberg, G.R.; Kemp, B.E. AMPK in Health and Disease. Physiol. Rev. 2009, 89, 1025–1078.
  32. Hardie, D.G.; Ross, F.A.; Hawley, S.A. AMPK: A nutrient and energy sensor that maintains energy homeostasis. Nat. Rev. Mol. Cell Biol. 2012, 13, 251–262.
  33. Zadra, G.; Batista, J.L.; Loda, M. Dissecting the Dual Role of AMPK in Cancer: From Experimental to Human Studies. Mol. Cancer Res. 2015, 13, 1059–1072.
  34. Suter, M.; Riek, U.; Tuerk, R.; Schlattner, U.; Wallimann, T.; Neumann, D. Dissecting the role of 5′-AMP for allosteric stimulation, activation, and deactivation of AMP-activated protein kinase. J. Biol. Chem. 2006, 281, 32207–32216.
  35. Hardie, D.G.; Carling, D.; Gamblin, S.J. AMP-activated protein kinase: Also regulated by ADP? Trends Biochem. Sci. 2011, 36, 470–477.
  36. Gowans, G.J.; Hawley, S.A.; Ross, F.A.; Hardie, D.G. AMP is a true physiological regulator of AMP-activated protein kinase by both allosteric activation and enhancing net phosphorylation. Cell Metab. 2013, 18, 556–566.
  37. Ross, F.A.; Jensen, T.E.; Hardie, D.G. Differential regulation by AMP and ADP of AMPK complexes containing different γ subunit isoforms. Biochem. J. 2016, 473, 189–199.
  38. Xiao, B.; Heath, R.; Saiu, P.; Leiper, F.C.; Leone, P.; Jing, C.; Walker, P.A.; Haire, L.; Eccleston, J.F.; Davis, C.T.; et al. Structural basis for AMP binding to mammalian AMP-activated protein kinase. Nature 2007, 449, 496–500.
  39. Xiao, B.; Sanders, M.J.; Underwood, E.; Heath, R.; Mayer, F.V.; Carmena, D.; Jing, C.; Walker, P.A.; Eccleston, J.F.; Haire, L.F.; et al. Structure of mammalian AMPK and its regulation by ADP. Nature 2011, 472, 230–233.
  40. Scott, J.W.; Ross, F.A.; Liu, J.K.D.; Hardie, D.G. Regulation of AMP-activated protein kinase by a pseudosubstrate sequence on the gamma subunit. EMBO J. 2007, 26, 806–815.
  41. Dzamko, N.; van Denderen, B.J.W.; Hevener, A.L.; Jørgensen, S.B.; Honeyman, J.; Galic, S.; Chen, Z.-P.; Watt, M.J.; Campbell, D.J.; Steinberg, G.R.; et al. AMPK beta1 deletion reduces appetite, preventing obesity and hepatic insulin resistance. J. Biol. Chem. 2010, 285, 115–122.
  42. Gruzman, A.; Babai, G.; Sasson, S. Adenosine Monophosphate-Activated Protein Kinase (AMPK) as a New Target for Antidiabetic Drugs: A Review on Metabolic, Pharmacological and Chemical Considerations. Rev. Diabet. Stud. 2009, 6, 13–36.
  43. Sanz, P.; Rubio, T.; Garcia-Gimeno, M.A. AMPKβ subunits: More than just a scaffold in the formation of AMPK complex. FEBS J. 2013, 280, 3723–3733.
  44. Steinberg, G.R.; O’Neill, H.M.; Dzamko, N.L.; Galic, S.; Naim, T.; Koopman, R.; Jørgensen, S.B.; Honeyman, J.; Hewitt, K.; Chen, Z.-P.; et al. Whole body deletion of AMP-activated protein kinase 2 reduces muscle AMPK activity and exercise capacity. J. Biol. Chem. 2010, 285, 37198–37209.
  45. Pinkosky, S.L.; Scott, J.W.; Desjardins, E.M.; Smith, B.K.; Day, E.A.; Ford, R.J.; Langendorf, C.G.; Ling, N.X.Y.; Nero, T.L.; Loh, K.; et al. Long-chain fatty acyl-CoA esters regulate metabolism via allosteric control of AMPK β1 isoforms. Nat. Metab. 2020, 2, 873–881.
  46. An, H.; He, L. Current understanding of metformin effect on the control of hyperglycemia in diabetes. J. Endocrinol. 2016, 228, R97–R106.
  47. Howell, J.J.; Hellberg, K.; Turner, M.; Talbott, G.; Kolar, M.J.; Ross, D.S.; Hoxhaj, G.; Saghatelian, A.; Shaw, R.J.; Manning, B.D. Metformin Inhibits Hepatic mTORC1 Signaling via Dose-Dependent Mechanisms Involving AMPK and the TSC Complex. Cell Metab. 2017, 25, 463–471.
  48. Smith, B.K.; Marcinko, K.; Desjardins, E.M.; Lally, J.S.; Ford, R.J.; Steinberg, G.R. Treatment of nonalcoholic fatty liver disease: Role of AMPK. Am. J. Physiol. Endocrinol. Metab. 2016, 311, E730–E740.
  49. Hegarty, B.D.; Turner, N.; Cooney, G.J.; Kraegen, E.W. Insulin resistance and fuel homeostasis: The role of AMP-activated protein kinase. Acta Physiol. 2009, 196, 129–145.
  50. McGarry, J.D.; Stark, M.J.; Foster, D.W. Hepatic malonyl-CoA levels of fed, fasted and diabetic rats as measured using a simple radioisotopic assay. J. Biol. Chem. 1978, 253, 8291–8293.
  51. McGarry, J.D. The mitochondrial carnitine palmitoyltransferase system: Its broadening role in fuel homoeostasis and new insights into its molecular features. Biochem. Soc. Trans. 1995, 23, 321–324.
  52. Saha, A.K.; Laybutt, D.R.; Dean, D.; Vavvas, D.; Sebokova, E.; Ellis, B.; Klimes, I.; Kraegen, E.W.; Shafrir, E.; Ruderman, N.B. Cytosolic citrate and malonyl-CoA regulation in rat muscle in vivo. Am. J. Physiol. 1999, 276, E1030–E1037.
  53. Båvenholm, P.N.; Pigon, J.; Saha, A.K.; Ruderman, N.B.; Efendic, S. Fatty acid oxidation and the regulation of malonyl-CoA in human muscle. Diabetes 2000, 49, 1078–1083.
  54. Fabbrini, E.; Tiemann Luecking, C.; Love-Gregory, L.; Okunade, A.L.; Yoshino, M.; Fraterrigo, G.; Patterson, B.W.; Klein, S. Physiological Mechanisms of Weight Gain-Induced Steatosis in People with Obesity. Gastroenterology 2016, 150, 79–81.e2.
  55. Bedossa, P.; Tordjman, J.; Aron-Wisnewsky, J.; Poitou, C.; Oppert, J.-M.; Torcivia, A.; Bouillot, J.-L.; Paradis, V.; Ratziu, V.; Clément, K. Systematic review of bariatric surgery liver biopsies clarifies the natural history of liver disease in patients with severe obesity. Gut 2017, 66, 1688–1696.
  56. Martínez-Agustin, O.; Hernández-Morante, J.J.; Martínez-Plata, E.; Sánchez de Medina, F.; Garaulet, M. Differences in AMPK expression between subcutaneous and visceral adipose tissue in morbid obesity. Regul. Pept. 2010, 163, 31–36.
  57. Gauthier, M.-S.; O’Brien, E.L.; Bigornia, S.; Mott, M.; Cacicedo, J.M.; Xu, X.J.; Gokce, N.; Apovian, C.; Ruderman, N. Decreased AMP-activated protein kinase activity is associated with increased inflammation in visceral adipose tissue and with whole-body insulin resistance in morbidly obese humans. Biochem. Biophys. Res. Commun. 2011, 404, 382–387.
  58. Xu, X.J.; Gauthier, M.-S.; Hess, D.T.; Apovian, C.M.; Cacicedo, J.M.; Gokce, N.; Farb, M.; Valentine, R.J.; Ruderman, N.B. Insulin sensitive and resistant obesity in humans: AMPK activity, oxidative stress, and depot-specific changes in gene expression in adipose tissue. J. Lipid Res. 2012, 53, 792–801.
  59. Grisouard, J.; Timper, K.; Radimerski, T.M.; Frey, D.M.; Peterli, R.; Kola, B.; Korbonits, M.; Herrmann, P.; Krähenbühl, S.; Zulewski, H.; et al. Mechanisms of metformin action on glucose transport and metabolism in human adipocytes. Biochem. Pharmacol. 2010, 80, 1736–1745.
  60. Bril, F.; Barb, D.; Portillo-Sanchez, P.; Biernacki, D.; Lomonaco, R.; Suman, A.; Weber, M.H.; Budd, J.T.; Lupi, M.E.; Cusi, K. Metabolic and histological implications of intrahepatic triglyceride content in nonalcoholic fatty liver disease. Hepatology 2017, 65, 1132–1144.
  61. Tilg, H.; Moschen, A.R.; Roden, M. NAFLD and diabetes mellitus. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 32–42.
  62. Petersen, M.C.; Shulman, G.I. Mechanisms of Insulin Action and Insulin Resistance. Physiol. Rev. 2018, 98, 2133–2223.
  63. Weisberg, S.P.; McCann, D.; Desai, M.; Rosenbaum, M.; Leibel, R.L.; Ferrante, A.W. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Investig. 2003, 112, 1796–1808.
  64. Ogawa, W.; Kasuga, M. Cell signaling. Fat stress and liver resistance. Science 2008, 322, 1483–1484.
  65. Elbein, S.C.; Kern, P.A.; Rasouli, N.; Yao-Borengasser, A.; Sharma, N.K.; Das, S.K. Global gene expression profiles of subcutaneous adipose and muscle from glucose-tolerant, insulin-sensitive, and insulin-resistant individuals matched for BMI. Diabetes 2011, 60, 1019–1029.
  66. Sun, K.; Kusminski, C.M.; Scherer, P.E. Adipose tissue remodeling and obesity. J. Clin. Investig. 2011, 121, 2094–2101.
  67. Nielsen, T.S.; Jessen, N.; Jørgensen, J.O.L.; Møller, N.; Lund, S. Dissecting adipose tissue lipolysis: Molecular regulation and implications for metabolic disease. J. Mol. Endocrinol. 2014, 52, R199–R222.
  68. Yin, W.; Mu, J.; Birnbaum, M.J. Role of AMP-activated protein kinase in cyclic AMP-dependent lipolysis in 3T3-L1 adipocytes. J. Biol. Chem. 2003, 278, 43074–43080.
  69. Daval, M.; Diot-Dupuy, F.; Bazin, R.; Hainault, I.; Viollet, B.; Vaulont, S.; Hajduch, E.; Ferré, P.; Foufelle, F. Anti-lipolytic action of AMP-activated protein kinase in rodent adipocytes. J. Biol. Chem. 2005, 280, 25250–25257.
  70. Daval, M.; Foufelle, F.; Ferré, P. Functions of AMP-activated protein kinase in adipose tissue. J. Physiol. 2006, 574, 55–62.
  71. Koh, H.-J.; Hirshman, M.F.; He, H.; Li, Y.; Manabe, Y.; Balschi, J.A.; Goodyear, L.J. Adrenaline is a critical mediator of acute exercise-induced AMP-activated protein kinase activation in adipocytes. Biochem. J. 2007, 403, 473–481.
  72. Ahmadian, M.; Abbott, M.J.; Tang, T.; Hudak, C.S.S.; Kim, Y.; Bruss, M.; Hellerstein, M.K.; Lee, H.-Y.; Samuel, V.T.; Shulman, G.I.; et al. Desnutrin/ATGL is regulated by AMPK and is required for a brown adipose phenotype. Cell Metab. 2011, 13, 739–748.
  73. Kim, S.-J.; Tang, T.; Abbott, M.; Viscarra, J.A.; Wang, Y.; Sul, H.S. AMPK Phosphorylates Desnutrin/ATGL and Hormone-Sensitive Lipase To Regulate Lipolysis and Fatty Acid Oxidation within Adipose Tissue. Mol. Cell. Biol. 2016, 36, 1961–1976.
  74. Zechner, R.; Madeo, F.; Kratky, D. Cytosolic lipolysis and lipophagy: Two sides of the same coin. Nat. Rev. Mol. Cell Biol. 2017, 18, 671–684.
  75. Boon, H.; Bosselaar, M.; Praet, S.F.E.; Blaak, E.E.; Saris, W.H.M.; Wagenmakers, A.J.M.; McGee, S.L.; Tack, C.J.; Smits, P.; Hargreaves, M.; et al. Intravenous AICAR administration reduces hepatic glucose output and inhibits whole body lipolysis in type 2 diabetic patients. Diabetologia 2008, 51, 1893–1900.
  76. Ng, J.M.; Azuma, K.; Kelley, C.; Pencek, R.; Radikova, Z.; Laymon, C.; Price, J.; Goodpaster, B.H.; Kelley, D.E. PET imaging reveals distinctive roles for different regional adipose tissue depots in systemic glucose metabolism in nonobese humans. Am. J. Physiol. Endocrinol. Metab. 2012, 303, E1134–E1141.
  77. Mottillo, E.P.; Desjardins, E.M.; Crane, J.D.; Smith, B.K.; Green, A.E.; Ducommun, S.; Henriksen, T.I.; Rebalka, I.A.; Razi, A.; Sakamoto, K.; et al. Lack of Adipocyte AMPK Exacerbates Insulin Resistance and Hepatic Steatosis through Brown and Beige Adipose Tissue Function. Cell Metab. 2016, 24, 118–129.
  78. Chong, M.F.F.; Fielding, B.A.; Frayn, K.N. Metabolic interaction of dietary sugars and plasma lipids with a focus on mechanisms and de novo lipogenesis. Proc. Nutr. Soc. 2007, 66, 52–59.
  79. Ferré, P.; Foufelle, F. SREBP-1c transcription factor and lipid homeostasis: Clinical perspective. Horm. Res. 2007, 68, 72–82.
  80. Castle, J.C.; Hara, Y.; Raymond, C.K.; Garrett-Engele, P.; Ohwaki, K.; Kan, Z.; Kusunoki, J.; Johnson, J.M. ACC2 is expressed at high levels in human white adipose and has an isoform with a novel N-terminus corrected. PLoS ONE 2009, 4, e4369.
  81. Goodpaster, B.H.; Sparks, L.M. Metabolic Flexibility in Health and Disease. Cell Metab. 2017, 25, 1027–1036.
  82. Timmers, S.; Konings, E.; Bilet, L.; Houtkooper, R.H.; van de Weijer, T.; Goossens, G.H.; Hoeks, J.; van der Krieken, S.; Ryu, D.; Kersten, S.; et al. Calorie restriction-like effects of 30 days of resveratrol supplementation on energy metabolism and metabolic profile in obese humans. Cell Metab. 2011, 14, 612–622.
  83. Højlund, K.; Mustard, K.J.; Staehr, P.; Hardie, D.G.; Beck-Nielsen, H.; Richter, E.A.; Wojtaszewski, J.F.P. AMPK activity and isoform protein expression are similar in muscle of obese subjects with and without type 2 diabetes. Am. J. Physiol. Endocrinol. Metab. 2004, 286, E239–E244.
  84. Steinberg, G.R.; Smith, A.C.; van Denderen, B.J.W.; Chen, Z.; Murthy, S.; Campbell, D.J.; Heigenhauser, G.J.F.; Dyck, D.J.; Kemp, B.E. AMP-activated protein kinase is not down-regulated in human skeletal muscle of obese females. J. Clin. Endocrinol. Metab. 2004, 89, 4575–4580.
  85. Højlund, K.; Staehr, P.; Hansen, B.F.; Green, K.A.; Hardie, D.G.; Richter, E.A.; Beck-Nielsen, H.; Wojtaszewski, J.F.P. Increased phosphorylation of skeletal muscle glycogen synthase at NH2-terminal sites during physiological hyperinsulinemia in type 2 diabetes. Diabetes 2003, 52, 1393–1402.
  86. Koistinen, H.A.; Galuska, D.; Chibalin, A.V.; Yang, J.; Zierath, J.R.; Holman, G.D.; Wallberg-Henriksson, H. 5-amino-imidazole carboxamide riboside increases glucose transport and cell-surface GLUT4 content in skeletal muscle from subjects with type 2 diabetes. Diabetes 2003, 52, 1066–1072.
  87. Cuthbertson, D.J.; Babraj, J.A.; Mustard, K.J.W.; Towler, M.C.; Green, K.A.; Wackerhage, H.; Leese, G.P.; Baar, K.; Thomason-Hughes, M.; Sutherland, C.; et al. 5-aminoimidazole-4-carboxamide 1-beta-D-ribofuranoside acutely stimulates skeletal muscle 2-deoxyglucose uptake in healthy men. Diabetes 2007, 56, 2078–2084.
  88. Babraj, J.A.; Mustard, K.; Sutherland, C.; Towler, M.C.; Chen, S.; Smith, K.; Green, K.; Leese, G.; Hardie, D.G.; Rennie, M.J.; et al. Blunting of AICAR-induced human skeletal muscle glucose uptake in type 2 diabetes is dependent on age rather than diabetic status. Am. J. Physiol. Endocrinol. Metab. 2009, 296, E1042–E1048.
  89. Musi, N.; Fujii, N.; Hirshman, M.F.; Ekberg, I.; Fröberg, S.; Ljungqvist, O.; Thorell, A.; Goodyear, L.J. AMP-activated protein kinase (AMPK) is activated in muscle of subjects with type 2 diabetes during exercise. Diabetes 2001, 50, 921–927.
  90. Roepstorff, C.; Vistisen, B.; Donsmark, M.; Nielsen, J.N.; Galbo, H.; Green, K.A.; Hardie, D.G.; Wojtaszewski, J.F.P.; Richter, E.A.; Kiens, B. Regulation of hormone-sensitive lipase activity and Ser563 and Ser565 phosphorylation in human skeletal muscle during exercise. J. Physiol. 2004, 560, 551–562.
  91. Watt, M.J.; Steinberg, G.R.; Chan, S.; Garnham, A.; Kemp, B.E.; Febbraio, M.A. Beta-adrenergic stimulation of skeletal muscle HSL can be overridden by AMPK signaling. FASEB J. 2004, 18, 1445–1446.
  92. Steenberg, D.E.; Jørgensen, N.B.; Birk, J.B.; Sjøberg, K.A.; Kiens, B.; Richter, E.A.; Wojtaszewski, J.F.P. Exercise training reduces the insulin-sensitizing effect of a single bout of exercise in human skeletal muscle. J. Physiol. 2019, 597, 89–103.
  93. Bajaj, M.; Suraamornkul, S.; Piper, P.; Hardies, L.J.; Glass, L.; Cersosimo, E.; Pratipanawatr, T.; Miyazaki, Y.; Defronzo, R.A. Decreased plasma adiponectin concentrations are closely related to hepatic fat content and hepatic insulin resistance in pioglitazone-treated type 2 diabetic patients. J. Clin. Endocrinol. Metab. 2004, 89, 200–206.
  94. Brown, M.S.; Goldstein, J.L. Selective versus total insulin resistance: A pathogenic paradox. Cell Metab. 2008, 7, 95–96.
  95. Lambert, J.E.; Ramos-Roman, M.A.; Browning, J.D.; Parks, E.J. Increased de novo lipogenesis is a distinct characteristic of individuals with nonalcoholic fatty liver disease. Gastroenterology 2014, 146, 726–735.
More
Information
Subjects: Cell Biology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 486
Revision: 1 time (View History)
Update Date: 18 Aug 2021
1000/1000