#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

PPP2R5C Couples Hepatic Glucose and Lipid Homeostasis


After a meal, dietary glucose travels through the hepatic portal vein to the liver. A substantial part of this glucose is taken up by liver, which converts it to glycogen which is stored, and lipids which are in part stored and in part secreted as VLDL particles. The rest of the organs receive whatever glucose the liver leaves in circulation, plus the secreted lipids. Hence the liver plays a crucial role in determining the balance of sugar versus lipids in the body after a meal. This balance is very important, because too much glucose in circulation leads to diabetic complications whereas too much VLDL increases risk of atherosclerosis. Little is known about how the liver strikes this balance. We identify here a phosphatase—the PP2A holoenzyme containing the PPP2R5C regulatory subunit—as a regulator of this process. We find that knockdown of PPP2R5C in mouse liver specifically causes it to uptake elevated levels of glucose, and secrete elevated levels of VLDL into circulation. This leads to a phenotype of improved glucose tolerance and insulin sensitivity. The prediction from these functional studies in mice is that elevated levels of PPP2R5C expression should lead to insulin resistance. Indeed, we find that PPP2R5C expression levels are elevated in diabetic patients, or healthy controls with visceral obesity, raising the possibility that dysregulation of PPP2R5C expression in humans may contribute towards metabolic dysfunction.


Vyšlo v časopise: PPP2R5C Couples Hepatic Glucose and Lipid Homeostasis. PLoS Genet 11(10): e32767. doi:10.1371/journal.pgen.1005561
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1005561

Souhrn

After a meal, dietary glucose travels through the hepatic portal vein to the liver. A substantial part of this glucose is taken up by liver, which converts it to glycogen which is stored, and lipids which are in part stored and in part secreted as VLDL particles. The rest of the organs receive whatever glucose the liver leaves in circulation, plus the secreted lipids. Hence the liver plays a crucial role in determining the balance of sugar versus lipids in the body after a meal. This balance is very important, because too much glucose in circulation leads to diabetic complications whereas too much VLDL increases risk of atherosclerosis. Little is known about how the liver strikes this balance. We identify here a phosphatase—the PP2A holoenzyme containing the PPP2R5C regulatory subunit—as a regulator of this process. We find that knockdown of PPP2R5C in mouse liver specifically causes it to uptake elevated levels of glucose, and secrete elevated levels of VLDL into circulation. This leads to a phenotype of improved glucose tolerance and insulin sensitivity. The prediction from these functional studies in mice is that elevated levels of PPP2R5C expression should lead to insulin resistance. Indeed, we find that PPP2R5C expression levels are elevated in diabetic patients, or healthy controls with visceral obesity, raising the possibility that dysregulation of PPP2R5C expression in humans may contribute towards metabolic dysfunction.


Zdroje

1. Donnelly KL, Smith CI, Schwarzenberg SJ, Jessurun J, Boldt MD, et al. (2005) Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J Clin Invest 115: 1343–1351. 15864352

2. Adiels M, Olofsson SO, Taskinen MR, Boren J (2008) Overproduction of very low-density lipoproteins is the hallmark of the dyslipidemia in the metabolic syndrome. Arterioscler Thromb Vasc Biol 28: 1225–1236. doi: 10.1161/ATVBAHA.107.160192 18565848

3. Chong MF, Hodson L, Bickerton AS, Roberts R, Neville M, et al. (2008) Parallel activation of de novo lipogenesis and stearoyl-CoA desaturase activity after 3 d of high-carbohydrate feeding. Am J Clin Nutr 87: 817–823. 18400702

4. de Wit NJ, Afman LA, Mensink M, Muller M (2012) Phenotyping the effect of diet on non-alcoholic fatty liver disease. J Hepatol 57: 1370–1373. doi: 10.1016/j.jhep.2012.07.003 22796155

5. Virtue S, Vidal-Puig A (2010) Adipose tissue expandability, lipotoxicity and the Metabolic Syndrome—an allostatic perspective. Biochim Biophys Acta 1801: 338–349. doi: 10.1016/j.bbalip.2009.12.006 20056169

6. Parks EJ, Parks EJ (2002) Changes in fat synthesis influenced by dietary macronutrient content. Proc Nutr Soc 61: 281–286. 12133211

7. Khanna A, Pimanda JE, Westermarck J (2013) Cancerous inhibitor of protein phosphatase 2A, an emerging human oncoprotein and a potential cancer therapy target. Cancer Res 73: 6548–6553. doi: 10.1158/0008-5472.CAN-13-1994 24204027

8. Vintonyak VV, Antonchick AP, Rauh D, Waldmann H (2009) The therapeutic potential of phosphatase inhibitors. Curr Opin Chem Biol 13: 272–283. doi: 10.1016/j.cbpa.2009.03.021 19410499

9. Pallen CJ (2003) Protein tyrosine phosphatase alpha (PTPalpha): a Src family kinase activator and mediator of multiple biological effects. Curr Top Med Chem 3: 821–835. 12678847

10. Westermarck J, Hahn WC (2008) Multiple pathways regulated by the tumor suppressor PP2A in transformation. Trends Mol Med 14: 152–160. doi: 10.1016/j.molmed.2008.02.001 18329957

11. Junttila MR, Puustinen P, Niemela M, Ahola R, Arnold H, et al. (2007) CIP2A inhibits PP2A in human malignancies. Cell 130: 51–62. 17632056

12. Walaas SI, Hemmings HC Jr., Greengard P, Nairn AC (2011) Beyond the dopamine receptor: regulation and roles of serine/threonine protein phosphatases. Front Neuroanat 5: 50. doi: 10.3389/fnana.2011.00050 21904525

13. Riedel CG, Katis VL, Katou Y, Mori S, Itoh T, et al. (2006) Protein phosphatase 2A protects centromeric sister chromatid cohesion during meiosis I. Nature 441: 53–61. 16541024

14. Shi Y (2009) Serine/threonine phosphatases: mechanism through structure. Cell 139: 468–484. doi: 10.1016/j.cell.2009.10.006 19879837

15. Margolis SS, Perry JA, Forester CM, Nutt LK, Guo Y, et al. (2006) Role for the PP2A/B56delta phosphatase in regulating 14-3-3 release from Cdc25 to control mitosis. Cell 127: 759–773. 17110335

16. Louis JV, Martens E, Borghgraef P, Lambrecht C, Sents W, et al. (2011) Mice lacking phosphatase PP2A subunit PR61/B'delta (Ppp2r5d) develop spatially restricted tauopathy by deregulation of CDK5 and GSK3beta. Proc Natl Acad Sci U S A 108: 6957–6962. doi: 10.1073/pnas.1018777108 21482799

17. Hahn K, Miranda M, Francis VA, Vendrell J, Zorzano A, et al. (2010) PP2A regulatory subunit PP2A-B' counteracts S6K phosphorylation. Cell Metab 11: 438–444. doi: 10.1016/j.cmet.2010.03.015 20444422

18. Zheng H, Chen Y, Chen S, Niu Y, Yang L, et al. (2011) Expression and distribution of PPP2R5C gene in leukemia. J Hematol Oncol 4: 21. doi: 10.1186/1756-8722-4-21 21548944

19. Chen W, Possemato R, Campbell KT, Plattner CA, Pallas DC, et al. (2004) Identification of specific PP2A complexes involved in human cell transformation. Cancer Cell 5: 127–136. 14998489

20. Francia G, Mitchell SD, Moss SE, Hanby AM, Marshall JF, et al. (1996) Identification by differential display of annexin-VI, a gene differentially expressed during melanoma progression. Cancer Res 56: 3855–3858. 8752144

21. Deichmann M, Polychronidis M, Wacker J, Thome M, Naher H (2001) The protein phosphatase 2A subunit Bgamma gene is identified to be differentially expressed in malignant melanomas by subtractive suppression hybridization. Melanoma Res 11: 577–585. 11725204

22. Nobumori Y, Shouse GP, Wu Y, Lee KJ, Shen B, et al. (2013) B56gamma tumor-associated mutations provide new mechanisms for B56gamma-PP2A tumor suppressor activity. Mol Cancer Res 11: 995–1003. doi: 10.1158/1541-7786.MCR-12-0633 23723076

23. Shouse GP, Cai X, Liu X (2008) Serine 15 phosphorylation of p53 directs its interaction with B56gamma and the tumor suppressor activity of B56gamma-specific protein phosphatase 2A. Mol Cell Biol 28: 448–456. 17967874

24. Li HH, Cai X, Shouse GP, Piluso LG, Liu X (2007) A specific PP2A regulatory subunit, B56gamma, mediates DNA damage-induced dephosphorylation of p53 at Thr55. EMBO J 26: 402–411. 17245430

25. Shouse GP, Nobumori Y, Panowicz MJ, Liu X (2011) ATM-mediated phosphorylation activates the tumor-suppressive function of B56gamma-PP2A. Oncogene 30: 3755–3765. doi: 10.1038/onc.2011.95 21460856

26. Shouse GP, Nobumori Y, Liu X (2010) A B56gamma mutation in lung cancer disrupts the p53-dependent tumor-suppressor function of protein phosphatase 2A. Oncogene 29: 3933–3941. doi: 10.1038/onc.2010.161 20473327

27. Varadkar P, Despres D, Kraman M, Lozier J, Phadke A, et al. (2014) The protein phosphatase 2A B56gamma regulatory subunit is required for heart development. Dev Dyn 243: 778–790. doi: 10.1002/dvdy.24111 24425002

28. Xu X, So JS, Park JG, Lee AH (2013) Transcriptional control of hepatic lipid metabolism by SREBP and ChREBP. Semin Liver Dis 33: 301–311. doi: 10.1055/s-0033-1358523 24222088

29. Jeon TI, Osborne TF (2012) SREBPs: metabolic integrators in physiology and metabolism. Trends Endocrinol Metab 23: 65–72. doi: 10.1016/j.tem.2011.10.004 22154484

30. Kulozik P, Jones A, Mattijssen F, Rose AJ, Reimann A, et al. (2011) Hepatic deficiency in transcriptional cofactor TBL1 promotes liver steatosis and hypertriglyceridemia. Cell Metab 13: 389–400. doi: 10.1016/j.cmet.2011.02.011 21459324

31. Benhamed F, Denechaud PD, Lemoine M, Robichon C, Moldes M, et al. (2012) The lipogenic transcription factor ChREBP dissociates hepatic steatosis from insulin resistance in mice and humans. J Clin Invest 122: 2176–2194. doi: 10.1172/JCI41636 22546860

32. Dashty M (2013) A quick look at biochemistry: carbohydrate metabolism. Clin Biochem 46: 1339–1352. doi: 10.1016/j.clinbiochem.2013.04.027 23680095

33. Roux KJ, Kim DI, Raida M, Burke B (2012) A promiscuous biotin ligase fusion protein identifies proximal and interacting proteins in mammalian cells. J Cell Biol 196: 801–810. doi: 10.1083/jcb.201112098 22412018

34. Ogris E, Mudrak I, Mak E, Gibson D, Pallas DC (1999) Catalytically inactive protein phosphatase 2A can bind to polyomavirus middle tumor antigen and support complex formation with pp60(c-src). J Virol 73: 7390–7398. 10438829

35. Soderberg O, Gullberg M, Jarvius M, Ridderstrale K, Leuchowius KJ, et al. (2006) Direct observation of individual endogenous protein complexes in situ by proximity ligation. Nat Methods 3: 995–1000. 17072308

36. Kinoshita E, Kinoshita-Kikuta E, Koike T (2009) Separation and detection of large phosphoproteins using Phos-tag SDS-PAGE. Nat Protoc 4: 1513–1521. doi: 10.1038/nprot.2009.154 19798084

37. Essaghir A, Toffalini F, Knoops L, Kallin A, van Helden J, et al. (2010) Transcription factor regulation can be accurately predicted from the presence of target gene signatures in microarray gene expression data. Nucleic Acids Res 38: e120. doi: 10.1093/nar/gkq149 20215436

38. Iizuka K, Horikawa Y (2008) ChREBP: a glucose-activated transcription factor involved in the development of metabolic syndrome. Endocr J 55: 617–624. 18490833

39. Bahary N, Leibel RL, Joseph L, Friedman JM (1990) Molecular mapping of the mouse db mutation. Proc Natl Acad Sci U S A 87: 8642–8646. 1978328

40. Bluher M (2014) Are metabolically healthy obese individuals really healthy? Eur J Endocrinol 171: R209–219. doi: 10.1530/EJE-14-0540 25012199

41. Hardie DG, Alessi DR (2013) LKB1 and AMPK and the cancer-metabolism link—ten years after. BMC Biol 11: 36. doi: 10.1186/1741-7007-11-36 23587167

42. Chen JQ, Russo J (2012) Dysregulation of glucose transport, glycolysis, TCA cycle and glutaminolysis by oncogenes and tumor suppressors in cancer cells. Biochim Biophys Acta 1826: 370–384. doi: 10.1016/j.bbcan.2012.06.004 22750268

43. Foretz M, Ancellin N, Andreelli F, Saintillan Y, Grondin P, et al. (2005) Short-term overexpression of a constitutively active form of AMP-activated protein kinase in the liver leads to mild hypoglycemia and fatty liver. Diabetes 54: 1331–1339. 15855317

44. Andreelli F, Foretz M, Knauf C, Cani PD, Perrin C, et al. (2006) Liver adenosine monophosphate-activated kinase-alpha2 catalytic subunit is a key target for the control of hepatic glucose production by adiponectin and leptin but not insulin. Endocrinology 147: 2432–2441. 16455782

45. Semenza GL (2013) HIF-1 mediates metabolic responses to intratumoral hypoxia and oncogenic mutations. J Clin Invest 123: 3664–3671. doi: 10.1172/JCI67230 23999440

46. Wang XL, Suzuki R, Lee K, Tran T, Gunton JE, et al. (2009) Ablation of ARNT/HIF1beta in liver alters gluconeogenesis, lipogenic gene expression, and serum ketones. Cell Metab 9: 428–439. doi: 10.1016/j.cmet.2009.04.001 19416713

47. Furuta E, Pai SK, Zhan R, Bandyopadhyay S, Watabe M, et al. (2008) Fatty acid synthase gene is up-regulated by hypoxia via activation of Akt and sterol regulatory element binding protein-1. Cancer Res 68: 1003–1011. doi: 10.1158/0008-5472.CAN-07-2489 18281474

48. Gao GP, Alvira MR, Wang L, Calcedo R, Johnston J, et al. (2002) Novel adeno-associated viruses from rhesus monkeys as vectors for human gene therapy. Proc Natl Acad Sci U S A 99: 11854–11859. 12192090

49. Grimm D, Kern A, Rittner K, Kleinschmidt JA (1998) Novel tools for production and purification of recombinant adenoassociated virus vectors. Hum Gene Ther 9: 2745–2760. 9874273

50. Klingmuller U, Bauer A, Bohl S, Nickel PJ, Breitkopf K, et al. (2006) Primary mouse hepatocytes for systems biology approaches: a standardized in vitro system for modelling of signal transduction pathways. Syst Biol (Stevenage) 153: 433–447.

51. Folch J, Lees M, Sloane Stanley GH (1957) A simple method for the isolation and purification of total lipides from animal tissues. J Biol Chem 226: 497–509. 13428781

52. Bluher M, Unger R, Rassoul F, Richter V, Paschke R (2002) Relation between glycaemic control, hyperinsulinaemia and plasma concentrations of soluble adhesion molecules in patients with impaired glucose tolerance or Type II diabetes. Diabetologia 45: 210–216. 11935152

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2015 Číslo 10
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#