#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Thrombospondin-I is a critical modulator in non-alcoholic steatohepatitis (NASH)


Autoři: Jessica Min-DeBartolo aff001;  Franklin Schlerman aff003;  Sandeep Akare aff004;  Ju Wang aff003;  James McMahon aff003;  Yutian Zhan aff004;  Jameel Syed aff004;  Wen He aff005;  Baohong Zhang aff005;  Robert V. Martinez aff003
Působiště autorů: BioMedicine Design, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, United States of America aff001;  Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, United States of America aff002;  Inflammation and Immunology Research Unit, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, United States of America aff003;  Drug Safety Research and Development, Pfizer Worldwide Research and Development, Groton, Connecticut, United States of America aff004;  Early Clinical Development, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, United States of America aff005
Vyšlo v časopise: PLoS ONE 14(12)
Kategorie: Research Article
doi: https://doi.org/10.1371/journal.pone.0226854

Souhrn

Non-alcoholic fatty liver disease (NAFLD) is a progressive liver disease characterized by dysregulated lipid metabolism and chronic inflammation ultimately resulting in fibrosis. Untreated, NAFLD may progress to non-alcoholic steatohepatitis (NASH), cirrhosis and death. However, currently there are no FDA approved therapies that treat NAFLD/NASH. Thrombospondin-I (TSP-1) is a large glycoprotein in the extracellular matrix that regulates numerous cellular pathways including transforming growth factor beta 1 (TGF-β1) activation, angiogenesis, inflammation and cellular adhesion. Increased expression of TSP-1 has been reported in various liver diseases; however, its role in NAFLD/NASH is not well understood. We first examined TSP-1 modulation in hepatic stellate cell activation, a critical initiating step in hepatic fibrosis. Knockdown or inhibition of TSP-1 attenuated HSC activation measured by alpha smooth muscle actin (α-SMA) and Collagen I expression. To investigate the impact of TSP-1 modulation in context of NAFLD/NASH, we examined the effect of TSP-1 deficiency in the choline deficient L-amino acid defined high fat diet (CDAHFD) model of NASH in mice by assessing total body and liver weight, serum liver enzyme levels, serum lipid levels, liver steatosis, liver fibrosis and liver gene expression in wild type (WT) and TSP-1 null mice. CDAHFD fed mice, regardless of genotype, developed phenotypes of NASH, including significant increase in liver weight and liver enzymes, steatosis and fibrosis. However, in comparison to WT, CDAHFD-fed TSP-1 deficient mice were protected against numerous NASH phenotypes. TSP-1 null mice exhibited a decrease in serum lipid levels, inflammation markers and hepatic fibrosis. RNA-seq based transcriptomic profiles from the liver of CDAHFD fed mice determined that both WT and TSP-1 null mice exhibited similar gene expression signatures following CDAHFD, similar to biophysical and histological assessment comparison. Comparison of transcriptomic profiles based on genotype suggested that peroxisome proliferator activated receptor alpha (PPARα) pathway and amino acid metabolism pathways are differentially expressed in TSP-1 null mice. Activation of PPARα pathway was supported by observed decrease in serum lipid levels. Our findings provide important insights into the role of TSP-1 in context of NAFLD/NASH and TSP-1 may be a target of interest to develop anti-fibrotic therapeutics for NAFLD/NASH.

Klíčová slova:

Amino acid metabolism – Collagens – Diet – Fatty liver – Fibrosis – Liver fibrosis – Mouse models – Trophic interactions


Zdroje

1. Mortality GBD, Causes of Death C. Global, regional, and national age-sex specific all-cause and cause-specific mortality for 240 causes of death, 1990–2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet. 2015;385(9963):117–71. Epub 2014/12/23. doi: 10.1016/S0140-6736(14)61682-2 25530442

2. Pellicoro A, Ramachandran P, Iredale JP, Fallowfield JA. Liver fibrosis and repair: immune regulation of wound healing in a solid organ. Nat Rev Immunol. 2014;14(3):181–94. Epub 2014/02/26. doi: 10.1038/nri3623 24566915.

3. Ellis EL, Mann DA. Clinical evidence for the regression of liver fibrosis. J Hepatol. 2012;56(5):1171–80. Epub 2012/01/17. doi: 10.1016/j.jhep.2011.09.024 22245903.

4. Williams CD, Stengel J, Asike MI, Torres DM, Shaw J, Contreras M, et al. Prevalence of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis among a largely middle-aged population utilizing ultrasound and liver biopsy: a prospective study. Gastroenterology. 2011;140(1):124–31. Epub 2010/09/23. doi: 10.1053/j.gastro.2010.09.038 20858492.

5. Rinella ME. Nonalcoholic fatty liver disease: a systematic review. JAMA. 2015;313(22):2263–73. Epub 2015/06/10. doi: 10.1001/jama.2015.5370 26057287.

6. Idowu MO, Chhatrala R, Siddiqui MB, Driscoll C, Stravitz RT, Sanyal AJ, et al. De novo hepatic steatosis drives atherogenic risk in liver transplantation recipients. Liver Transpl. 2015;21(11):1395–402. Epub 2015/08/01. doi: 10.1002/lt.24223 26228654.

7. Hannah WN Jr., Harrison SA. Nonalcoholic fatty liver disease and elastography: Incremental advances but work still to be done. Hepatology. 2016;63(6):1762–4. Epub 2016/02/19. doi: 10.1002/hep.28504 26891023.

8. Puche JE, Lee YA, Jiao J, Aloman C, Fiel MI, Munoz U, et al. A novel murine model to deplete hepatic stellate cells uncovers their role in amplifying liver damage in mice. Hepatology. 2013;57(1):339–50. doi: 10.1002/hep.26053 22961591

9. Resovi A, Pinessi D, Chiorino G, Taraboletti G. Current understanding of the thrombospondin-1 interactome. Matrix Biol. 2014;37:83–91. Epub 2014/01/31. doi: 10.1016/j.matbio.2014.01.012 24476925.

10. Ribeiro SM, Poczatek M, Schultz-Cherry S, Villain M, Murphy-Ullrich JE. The activation sequence of thrombospondin-1 interacts with the latency-associated peptide to regulate activation of latent transforming growth factor-beta. J Biol Chem. 1999;274(19):13586–93. Epub 1999/05/01. doi: 10.1074/jbc.274.19.13586 10224129.

11. Schultz-Cherry S, Chen H, Mosher DF, Misenheimer TM, Krutzsch HC, Roberts DD, et al. Regulation of transforming growth factor-beta activation by discrete sequences of thrombospondin 1. J Biol Chem. 1995;270(13):7304–10. Epub 1995/03/31. doi: 10.1074/jbc.270.13.7304 7706271.

12. Crawford SE, Stellmach V, Murphy-Ullrich JE, Ribeiro SM, Lawler J, Hynes RO, et al. Thrombospondin-1 is a major activator of TGF-beta1 in vivo. Cell. 1998;93(7):1159–70. Epub 1998/07/10. doi: 10.1016/s0092-8674(00)81460-9 9657149.

13. Rosini S, Pugh N, Bonna AM, Hulmes DJS, Farndale RW, Adams JC. Thrombospondin-1 promotes matrix homeostasis by interacting with collagen and lysyl oxidase precursors and collagen cross-linking sites. Sci Signal. 2018;11(532). Epub 2018/05/31. doi: 10.1126/scisignal.aar2566 29844053.

14. Smalling RL, Delker DA, Zhang Y, Nieto N, McGuiness MS, Liu S, et al. Genome-wide transcriptome analysis identifies novel gene signatures implicated in human chronic liver disease. Am J Physiol Gastrointest Liver Physiol. 2013;305(5):G364–74. Epub 2013/07/03. doi: 10.1152/ajpgi.00077.2013 23812039

15. El-Youssef M, Mu Y, Huang L, Stellmach V, Crawford SE. Increased expression of transforming growth factor-beta1 and thrombospondin-1 in congenital hepatic fibrosis: possible role of the hepatic stellate cell. J Pediatr Gastroenterol Nutr. 1999;28(4):386–92. Epub 1999/04/16. doi: 10.1097/00005176-199904000-00008 10204502.

16. Kondou H, Mushiake S, Etani Y, Miyoshi Y, Michigami T, Ozono K. A blocking peptide for transforming growth factor-beta1 activation prevents hepatic fibrosis in vivo. J Hepatol. 2003;39(5):742–8. doi: 10.1016/s0168-8278(03)00377-5 14568256.

17. Liao F, Li G, Yuan W, Chen Y, Zuo Y, Rashid K, et al. LSKL peptide alleviates subarachnoid fibrosis and hydrocephalus by inhibiting TSP1-mediated TGF-beta1 signaling activity following subarachnoid hemorrhage in rats. Exp Ther Med. 2016;12(4):2537–43. doi: 10.3892/etm.2016.3640 27698755

18. Inoue M, Jiang Y, Barnes RH 2nd, Tokunaga M, Martinez-Santibanez G, Geletka L, et al. Thrombospondin 1 mediates high-fat diet-induced muscle fibrosis and insulin resistance in male mice. Endocrinology. 2013;154(12):4548–59. doi: 10.1210/en.2013-1587 24140711

19. Cui W, Maimaitiyiming H, Qi X, Norman H, Wang S. Thrombospondin 1 mediates renal dysfunction in a mouse model of high-fat diet-induced obesity. Am J Physiol Renal Physiol. 2013;305(6):F871–80. doi: 10.1152/ajprenal.00209.2013 23863467

20. Kuroki H, Hayashi H, Nakagawa S, Sakamoto K, Higashi T, Nitta H, et al. Effect of LSKL peptide on thrombospondin 1-mediated transforming growth factor beta signal activation and liver regeneration after hepatectomy in an experimental model. Br J Surg. 2015;102(7):813–25. Epub 2015/04/14. doi: 10.1002/bjs.9765 25866938

21. Voros G, Maquoi E, Demeulemeester D, Clerx N, Collen D, Lijnen HR. Modulation of angiogenesis during adipose tissue development in murine models of obesity. Endocrinology. 2005;146(10):4545–54. Epub 2005/07/16. doi: 10.1210/en.2005-0532 16020476.

22. Varma V, Yao-Borengasser A, Bodles AM, Rasouli N, Phanavanh B, Nolen GT, et al. Thrombospondin-1 is an adipokine associated with obesity, adipose inflammation, and insulin resistance. Diabetes. 2008;57(2):432–9. Epub 2007/12/07. doi: 10.2337/db07-0840 18057090

23. Kong P, Gonzalez-Quesada C, Li N, Cavalera M, Lee DW, Frangogiannis NG. Thrombospondin-1 regulates adiposity and metabolic dysfunction in diet-induced obesity enhancing adipose inflammation and stimulating adipocyte proliferation. Am J Physiol Endocrinol Metab. 2013;305(3):E439–50. Epub 2013/06/13. doi: 10.1152/ajpendo.00006.2013 23757408

24. Belmadani S, Bernal J, Wei CC, Pallero MA, Dell’italia L, Murphy-Ullrich JE, et al. A thrombospondin-1 antagonist of transforming growth factor-beta activation blocks cardiomyopathy in rats with diabetes and elevated angiotensin II. Am J Pathol. 2007;171(3):777–89. Epub 2007/07/21. doi: 10.2353/ajpath.2007.070056 17640965

25. Lu A, Miao M, Schoeb TR, Agarwal A, Murphy-Ullrich JE. Blockade of TSP1-dependent TGF-beta activity reduces renal injury and proteinuria in a murine model of diabetic nephropathy. Am J Pathol. 2011;178(6):2573–86. Epub 2011/06/07. doi: 10.1016/j.ajpath.2011.02.039 21641382

26. Caballero F, Fernandez A, Matias N, Martinez L, Fucho R, Elena M, et al. Specific contribution of methionine and choline in nutritional nonalcoholic steatohepatitis: impact on mitochondrial S-adenosyl-L-methionine and glutathione. J Biol Chem. 2010;285(24):18528–36. doi: 10.1074/jbc.M109.099333 20395294

27. Zhao S, Xi L, Quan J, Xi H, Zhang Y, von Schack D, et al. QuickRNASeq lifts large-scale RNA-seq data analyses to the next level of automation and interactive visualization. BMC Genomics. 2016;17:39. doi: 10.1186/s12864-015-2356-9 26747388

28. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47. Epub 2015/01/22. doi: 10.1093/nar/gkv007 25605792

29. Matsumoto M, Hada N, Sakamaki Y, Uno A, Shiga T, Tanaka C, et al. An improved mouse model that rapidly develops fibrosis in non-alcoholic steatohepatitis. Int J Exp Pathol. 2013;94(2):93–103. doi: 10.1111/iep.12008 23305254

30. Kelder T, Pico AR, Hanspers K, van Iersel MP, Evelo C, Conklin BR. Mining biological pathways using WikiPathways web services. PLoS One. 2009;4(7):e6447. Epub 2009/08/04. doi: 10.1371/journal.pone.0006447 19649250

31. WikiPathways 2018. wikipathways.org.

32. McPherson S, Stewart SF, Henderson E, Burt AD, Day CP. Simple non-invasive fibrosis scoring systems can reliably exclude advanced fibrosis in patients with non-alcoholic fatty liver disease. Gut. 2010;59(9):1265–9. Epub 2010/08/31. doi: 10.1136/gut.2010.216077 20801772.

33. Machado MV, Michelotti GA, Xie G, Almeida Pereira T, Boursier J, Bohnic B, et al. Mouse models of diet-induced nonalcoholic steatohepatitis reproduce the heterogeneity of the human disease. PLoS One. 2015;10(5):e0127991. Epub 2015/05/29. doi: 10.1371/journal.pone.0127991 26017539

34. Sunny NE, Kalavalapalli S, Bril F, Garrett TJ, Nautiyal M, Mathew JT, et al. Cross-talk between branched-chain amino acids and hepatic mitochondria is compromised in nonalcoholic fatty liver disease. Am J Physiol Endocrinol Metab. 2015;309(4):E311–9. Epub 2015/06/11. doi: 10.1152/ajpendo.00161.2015 26058864

35. Lynch CJ, Adams SH. Branched-chain amino acids in metabolic signalling and insulin resistance. Nat Rev Endocrinol. 2014;10(12):723–36. Epub 2014/10/08. doi: 10.1038/nrendo.2014.171 25287287

36. Yamakado M, Tanaka T, Nagao K, Imaizumi A, Komatsu M, Daimon T, et al. Plasma amino acid profile associated with fatty liver disease and co-occurrence of metabolic risk factors. Sci Rep. 2017;7(1):14485. Epub 2017/11/05. doi: 10.1038/s41598-017-14974-w 29101348

37. Cheng S, Wiklund P, Autio R, Borra R, Ojanen X, Xu L, et al. Adipose Tissue Dysfunction and Altered Systemic Amino Acid Metabolism Are Associated with Non-Alcoholic Fatty Liver Disease. PLoS One. 2015;10(10):e0138889. Epub 2015/10/07. doi: 10.1371/journal.pone.0138889 26439744

38. Soto-Pantoja DR, Sipes JM, Martin-Manso G, Westwood B, Morris NL, Ghosh A, et al. Dietary fat overcomes the protective activity of thrombospondin-1 signaling in the Apc(Min/+) model of colon cancer. Oncogenesis. 2016;5(5):e230. Epub 2016/05/31. doi: 10.1038/oncsis.2016.37 27239962

39. O’Brien PJ, Slaughter MR, Polley SR, Kramer K. Advantages of glutamate dehydrogenase as a blood biomarker of acute hepatic injury in rats. Lab Anim. 2002;36(3):313–21. Epub 2002/07/30. doi: 10.1258/002367702320162414 12144742.

40. McGill MR, Sharpe MR, Williams CD, Taha M, Curry SC, Jaeschke H. The mechanism underlying acetaminophen-induced hepatotoxicity in humans and mice involves mitochondrial damage and nuclear DNA fragmentation. J Clin Invest. 2012;122(4):1574–83. Epub 2012/03/02. doi: 10.1172/JCI59755 22378043

41. Jaeschke H, McGill MR. Serum glutamate dehydrogenase—biomarker for liver cell death or mitochondrial dysfunction? Toxicol Sci. 2013;134(1):221–2. Epub 2013/04/10. doi: 10.1093/toxsci/kft087 23568080.

42. Staels B, Rubenstrunk A, Noel B, Rigou G, Delataille P, Millatt LJ, et al. Hepatoprotective effects of the dual peroxisome proliferator-activated receptor alpha/delta agonist, GFT505, in rodent models of nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. Hepatology. 2013;58(6):1941–52. Epub 2013/05/25. doi: 10.1002/hep.26461 23703580.

43. Fernandez-Miranda C, Perez-Carreras M, Colina F, Lopez-Alonso G, Vargas C, Solis-Herruzo JA. A pilot trial of fenofibrate for the treatment of non-alcoholic fatty liver disease. Dig Liver Dis. 2008;40(3):200–5. Epub 2008/02/12. doi: 10.1016/j.dld.2007.10.002 18261709.

44. Basaranoglu M, Acbay O, Sonsuz A. A controlled trial of gemfibrozil in the treatment of patients with nonalcoholic steatohepatitis. J Hepatol. 1999;31(2):384. Epub 1999/08/24. doi: 10.1016/s0168-8278(99)80243-8 10453959.

45. Laurin J, Lindor KD, Crippin JS, Gossard A, Gores GJ, Ludwig J, et al. Ursodeoxycholic acid or clofibrate in the treatment of non-alcohol-induced steatohepatitis: a pilot study. Hepatology. 1996;23(6):1464–7. Epub 1996/06/01. doi: 10.1002/hep.510230624 8675165.

46. Belfort R, Harrison SA, Brown K, Darland C, Finch J, Hardies J, et al. A placebo-controlled trial of pioglitazone in subjects with nonalcoholic steatohepatitis. N Engl J Med. 2006;355(22):2297–307. Epub 2006/12/01. doi: 10.1056/NEJMoa060326 17135584.

47. Bril F, Kalavalapalli S, Clark VC, Lomonaco R, Soldevila-Pico C, Liu IC, et al. Response to Pioglitazone in Patients With Nonalcoholic Steatohepatitis With vs Without Type 2 Diabetes. Clin Gastroenterol Hepatol. 2018;16(4):558–66 e2. Epub 2017/12/11. doi: 10.1016/j.cgh.2017.12.001 29223443.

48. Satapati S, Kucejova B, Duarte JA, Fletcher JA, Reynolds L, Sunny NE, et al. Mitochondrial metabolism mediates oxidative stress and inflammation in fatty liver. J Clin Invest. 2015;125(12):4447–62. Epub 2015/11/17. doi: 10.1172/JCI82204 26571396

49. Perry RJ, Zhang D, Zhang XM, Boyer JL, Shulman GI. Controlled-release mitochondrial protonophore reverses diabetes and steatohepatitis in rats. Science. 2015;347(6227):1253–6. Epub 2015/02/28. doi: 10.1126/science.aaa0672 25721504


Článek vyšel v časopise

PLOS One


2019 Číslo 12
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

KOST
Koncepce osteologické péče pro gynekology a praktické lékaře
nový kurz
Autoři: MUDr. František Šenk

Sekvenční léčba schizofrenie
Autoři: MUDr. Jana Hořínková

Hypertenze a hypercholesterolémie – synergický efekt léčby
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Svět praktické medicíny 5/2023 (znalostní test z časopisu)

Imunopatologie? … a co my s tím???
Autoři: doc. MUDr. Helena Lahoda Brodská, Ph.D.

Všechny kurzy
Kurzy Podcasty Doporučená témata Časopisy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#