期刊论文详细信息
Respiratory Research
Arsenic trioxide inhibits transforming growth factor-β1-induced fibroblast to myofibroblast differentiation in vitro and bleomycin induced lung fibrosis in vivo
Joseph A Lasky2  Eric S White1  Bin Shan2  Cecilia G Sanchez2  Mark D Sides2  Yan Zhuang2  Fayong Luo2 
[1] Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, MI, USA;Department of Medicine, Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Tulane University Health Science Center, New Orleans, LA 70112, USA
关键词: Bleomycin;    PML;    Pulmonary fibrosis;    TGF-β1;    IPF;    Arsenic trioxide;   
Others  :  790294
DOI  :  10.1186/1465-9921-15-51
 received in 2013-09-11, accepted in 2014-02-10,  发布年份 2014
PDF
【 摘 要 】

Background

Idiopathic pulmonary fibrosis (IPF) is a progressive disease of insidious onset, and is responsible for up to 30,000 deaths per year in the U.S. Excessive production of extracellular matrix by myofibroblasts has been shown to be an important pathological feature in IPF. TGF-β1 is expressed in fibrotic lung and promotes fibroblast to myofibroblast differentiation (FMD) as well as matrix deposition.

Methods

To identify the mechanism of Arsenic trioxide’s (ATO)’s anti-fibrotic effect in vitro, normal human lung fibroblasts (NHLFs) were treated with ATO for 24 hours and were then exposed to TGF-β1 (1 ng/ml) before harvesting at multiple time points. To investigate whether ATO is able to alleviate lung fibrosis in vivo, C57BL/6 mice were administered bleomycin by oropharyngeal aspiration and ATO was injected intraperitoneally daily for 14 days. Quantitative real-time PCR, western blotting, and immunofluorescent staining were used to assess the expression of fibrotic markers such as α-smooth muscle actin (α-SMA) and α-1 type I collagen.

Results

Treatment of NHLFs with ATO at very low concentrations (10-20nM) inhibits TGF-β1-induced α-smooth muscle actin (α-SMA) and α-1 type I collagen mRNA and protein expression. ATO also diminishes the TGF-β1-mediated contractile response in NHLFs. ATO’s down-regulation of profibrotic molecules is associated with inhibition of Akt, as well as Smad2/Smad3 phosphorylation. TGF-β1-induced H2O2 and NOX-4 mRNA expression are also blocked by ATO. ATO-mediated reduction in Smad3 phosphorylation correlated with a reduction of promyelocytic leukemia (PML) nuclear bodies and PML protein expression. PML-/- mouse embryonic fibroblasts (MEFs) showed decreased fibronectin and PAI-1 expression in response to TGF-β1. Daily intraperitoneal injection of ATO (1 mg/kg) in C57BL/6 mice inhibits bleomycin induced lung α-1 type I collagen mRNA and protein expression.

Conclusions

In summary, these data indicate that low concentrations of ATO inhibit TGF-β1-induced fibroblast to myofibroblast differentiation and decreases bleomycin induced pulmonary fibrosis.

【 授权许可】

   
2014 Luo et al.; licensee BioMed Central Ltd.

【 预 览 】
附件列表
Files Size Format View
20140704232534480.pdf 3523KB PDF download
Figure 7. 84KB Image download
Figure 6. 102KB Image download
Figure 5. 62KB Image download
Figure 4. 53KB Image download
Figure 3. 101KB Image download
Figure 2. 47KB Image download
Figure 1. 85KB Image download
【 图 表 】

Figure 1.

Figure 2.

Figure 3.

Figure 4.

Figure 5.

Figure 6.

Figure 7.

【 参考文献 】
  • [1]King TE Jr, Schwarz MI, Brown K, Tooze JA, Colby TV, Waldron JA Jr, Flint A, Thurlbeck W, Cherniack RM: Idiopathic Pulmonary Fibrosis Relationship between Histopathologic Features and Mortality. Am J Respir Crit Care Med 2001, 164:1025-1032.
  • [2]Raghu G, Weycker D, Edelsberg J, Bradford WZ, Oster G: Incidence and prevalence of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2006, 174:810Y816.
  • [3]Hinz B, Phan SH, Thannickal VJ, Galli A, Bochaton-Piallat ML, Gabbiani G: The myofibroblast: one function, multiple origins. Am J Pathol 2007, 170:1807-1816.
  • [4]Sime PJ, Xing Z, Graham FL, Csaky KG, Gauldie J: Adenovector-mediated gene transfer of active transforming growth factor-β1 induces prolonged severe fibrosis in rat lung. J Clin Invest 1997, 100:768-776.
  • [5]Guo W, Shan B, Klingsberg RC, Qin X, Lasky JA: Abrogation of TGF-β1-induced fibroblast-myofibroblast differentiation by histone deacetylase inhibition. Am J Physiol Lung Cell Mol Physiol 2009, 297(5):L864-L870. Epub 2009 Aug 21
  • [6]Hu B, Wu Z, Phan SH: Smad3 mediates transforming growth factor-β induced alpha-smooth muscle actin expression. Am J Respir Cell Mol Biol 2003, 29:397-404.
  • [7]Li M, Krishnaveni MS, Li C, Zhou B, Xing Y, Banfalvi A, Li A, Lombardi V, Akbari O, Borok Z, Minoo P: Epithelium-specific deletion of TGF-β receptor type II protects mice from bleomycin-induced pulmonary fibrosis. J Clin Invest 2011, 121(1):277-287. doi:10.1172/JCI42090. Epub 2010 Dec 6
  • [8]Waxman S, Anderson KC: Oncologist. History of the development of arsenic derivatives in cancer therapy 2001, 6(Suppl 2):3-10.
  • [9]Sun HD, Ma L, Hu XC: Ai-Lin 1 treated 32 cases of acute promyelocytic leukemia. Chin J Integrated Tradit West Med 1992, 1:170-171.
  • [10]Wang ZY, Chen Z: Acute promyelocytic leukemia: from highly fatal to highly curable. Blood 2008, 111:2505-2515.
  • [11]Smith DM, Patel S, Raffoul F, Haller E, Mills GB, Nanjundan M: Arsenic trioxide induces a beclin-1-independent autophagic pathway via modulation of SnoN/SkiL expression in ovarian carcinoma cells. Cell Death Differ 2010, 17(12):1867-1881.
  • [12]Zhu J, Lallemand-Breitenbach V, de Thé H: Pathways of retinoic acid or arsenic trioxide-induced PML/RARalpha catabolism, role of oncogene degradation in disease remission. Oncogene 2001, 20(49):7257-7265.
  • [13]De Thé H, Chomienne C, Lanotte M, Degos L, Dejean A: The t(15;17) translocation of acute promyelocytic leukaemia fuses the retinoic acid receptor alpha gene to a novel transcribed locus. Nature 1990, 347(6293):558-561.
  • [14]Bernardi R, Pandolfi PP: Structure, dynamics and functions of promyelocytic leukaemia nuclear bodies. Nature Rev. Mol. Cell Biol 2007, 8:1006-1016.
  • [15]Hofmann TG, Will H: Body language: the function of PML nuclear bodies in apoptosis regulation. Cell Death Differ 2003, 10:1290-1299.
  • [16]Lin HK, Bergmann S, Pandolfi PP: Cytoplasmic PML function in TGF-beta signalling. Nature 2004, 431(7005):205-211.
  • [17]Huet E, Vallée B, Szul D, Verrecchia F, Mourah S, Jester JV, Hoang-Xuan T, Menashi S, Gabison EE: Extracellular matrix metalloproteinase inducerCD147 promotes myofibroblast differentiation by inducing alpha-smooth muscle actin expression and collagen gel contraction implications in tissue remodeling. FASEB J 2008, 22(4):1144-1154. Epub 2007 Oct 26
  • [18]Li WQ, Qureshi HY, Liacini A, Dehnade F, Zafarullah M: Transforming growth factor Beta1 induction of tissue inhibitor of metalloproteinases 3 in articular chondrocytes is mediated by reactive oxygen species. Free Radic Biol Med 2004, 37(2):196-207.
  • [19]Rojewski MT, Baldus C, Knauf W, Thiel E, Schrezenmeier H: Dual effects of arsenic trioxide (As2O3) on non-acute promyelocytic leukaemia myeloid cell lines: induction of apoptosis and inhibition of proliferation. Br J Haematol 2002, 116(3):555-563.
  • [20]Kanzawa T, Kondo Y, Ito H, Kondo S, Germano I: Induction of autophagic cell death in malignant glioma cells by arsenic trioxide. Cancer Res 2003, 63(9):2103-2108.
  • [21]Redondo-Muñoz J, Escobar-Díaz E, Hernández Del Cerro M, Pandiella A, Terol MJ, García-Marco JA, García-Pardo A: Induction of B-chronic lymphocytic leukemia cell apoptosis by arsenic trioxide involves suppression of the phosphoinositide 3-kinase/Akt survival pathway via c-jun-NH2 terminal kinase activation and PTEN upregulation. Clin Cancer Res 2010, 16(17):4382-4391. Epub 2010 Jun 9
  • [22]You BR, Park WH: Arsenic trioxide induces human pulmonary fibroblast cell death via increasing ROS levels and GSH depletion. Oncol Rep 2012. doi: 10.3892/or.2012.1852
  • [23]Wipff PJ, Rifkin DB, Meister JJ, Hinz B: Myofibroblast contraction activates latent TGF-beta1 from the extracellular matrix. J Cell Biol 2007, 179(6):1311-1323.
  • [24]Derynck R, Zhang YE: Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature 2003, 425(6958):577-584.
  • [25]Amara N, Goven D, Prost F, Muloway R, Crestani B, Boczkowski J: NOX4/NADPH oxidase expression is increased in pulmonary fibroblasts from patients with idiopathic pulmonary fibrosis and mediates TGFbeta1-induced fibroblast differentiation into myofibroblasts. Thorax 2010, 65(8):733-738.
  • [26]Hecker L, Vittal R, Jones T, Jagirdar R, Luckhardt TR, Horowitz JC, Pennathur S, Martinez FJ, Thannickal VJ: NADPH oxidase-4 mediates myofibroblast activation and fibrogenic responses to lung injury. Nat Med 2009, 15(9):1077-1081. Epub 2009 Aug 23
  • [27]Bonniaud P, Kolb M, Galt T, Robertson J, Robbins C, Stampfli M, Lavery C, Margetts PJ, Roberts AB, Gauldie J, Bonniaud P, Kolb M, Galt T, Robertson J, Robbins C, Stampfli M, Lavery C, Margetts PJ, Roberts AB, Gauldie J: Smad3 null mice develop airspace enlargement and are resistant to TGF-beta-mediated pulmonary fibrosis. J Immunol 2004, 173(3):2099-2108.
  • [28]Moore BB, Hogaboam CM: Murine models of pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2008, 294(2):L152-L160. Epub 2007 Nov 9
  • [29]Kim HR, Kim EJ, Yang SH, Jeong ET, Park C, Kim SJ, Youn MJ, So HS, Park R: Combination treatment with arsenic trioxide and sulindac augments their apoptotic potential in lung cancer cells through activation of caspase cascade and mitochondrial dysfunction. Int J Oncol 2006, 28:1401-1408.
  • [30]Schrier DJ, Kunkel RG, Phan SH: The role of strain variation in murine bleomycin-induced pulmonary fibrosis. Am Rev Respir Dis 1983, 127(1):63-66.
  • [31]Ghatak S, Biswas A, Dhali GK, Chowdhury A, Boyer JL, Santra A: Oxidative stress and hepatic stellate cell activation are key events in arsenic induced liver fibrosis in mice. Toxicol Appl Pharmacol 2011, 251(1):59-69. Epub 2010 Dec 4
  • [32]Pan X, Dai Y, Li X, Niu N, Li W, Liu F, Zhao Y, Yu Z: Inhibition of arsenic-induced rat liver injury by grape seed exact through suppression of NADPH oxidase and TGF-β/Smad activation. Toxicol Appl Pharmacol 2011, 254(3):323-331. Epub 2011 May 13
  • [33]Kavian N, Marut W, Servettaz A, Nicco C, Chéreau C, Lemaréchal H, Borderie D, Dupin N, Weill B, Batteux F: Reactive oxygen species-mediated killing of activated fibroblasts by arsenic trioxide ameliorates fibrosis in a murine model of systemic sclerosis. Arthritis Rheum 2012, 64(10):3430-3440.
  • [34]Sherwood CL, Lantz RC, Burgess JL, Boitano S: Arsenic alters ATP-dependent Ca²+ signaling in human airway epithelial cell wound response. Toxicol Sci 2011, 121(1):191-206. Epub 2011 Feb 25
  • [35]Chu W, Li C, Qu X, Zhao D, Wang X, Yu X, Cai F, Liang H, Zhang Y, Zhao X, Li B, Qiao G, Dong D, Lu Y, Du Z, Yang B: Arsenic-induced interstitial myocardial fibrosis reveals a new insight into drug-induced long QT syndrome. Cardiovasc Res 2012, 96(1):90-98. Epub 2012 Aug 1
  • [36]Breccia M, Lo-Coco F: Arsenic trioxide for management of acute promyelocytic leukemia: current evidence on its role in front-line therapy and recurrent disease. Expert Opin Pharmacother 2012, 13(7):1031-1043. Epub 2012 Apr 3
  • [37]Shan B, Yao TP, Nguyen HT, Zhuo Y, Levy DR, Klingsberg RC, Tao H, Palmer ML, Holder KN, Lasky JA: Requirement of HDAC6 for transforming growth factor-beta1-induced epithelial-mesenchymal transition. J Biol Chem 2008, 283(30):21065-21073. Epub 2008 May 21
  • [38]Waghray M, Cui Z, Horowitz JC, Subramanian IM, Martinez FJ, Toews GB, Thannickal VJ: Hydrogen peroxide is a diffusible paracrine signal for the induction of epithelial cell death by activated myofibroblasts. FASEB J 2005, 19(7):854-856. Epub 2005 Feb 16
  • [39]Zhang XW, Yan XJ, Zhou ZR, Yang FF, Wu ZY, Sun HB, Liang WX, Song AX, Lallemand-Breitenbach V, Jeanne M, Zhang QY, Yang HY, Huang QH, Zhou GB, Tong JH, Zhang Y, Wu JH, Hu HY, de Thé H, Chen SJ, Chen Z: Arsenic trioxide controls the fate of the PML-RARalpha oncoprotein by directly binding PML. Science 2010, 328(5975):240-243.
  • [40]De Stanchina E, Querido E, Narita M, Davuluri RV, Pandolfi PP, Ferbeyre G, Lowe SW: PML is a direct p53 target that modulates p53 effector functions. Mol Cell 2004, 13(4):523-535.
  • [41]Oh W, Ghim J, Lee EW, Yang MR, Kim ET, Ahn JH, Song J: PML-IV functions as a negative regulator of telomerase by interacting with TERT. J Cell Sci 2009, 122(Pt 15):2613-2622. Epub 2009 Jun 30
  • [42]Alder JK, Chen JJ, Lancaster L, Danoff S, Su SC, Cogan JD, Vulto I, Xie M, Qi X, Tuder RM, Lansdorp PM, Loyd JE, Armanios MY, Phillips JA 3rd: Short telomeres are a risk factor for idiopathic pulmonary fibrosis. Proc Natl Acad Sci USA 2008, 105(35):13051-13056. Epub 2008 Aug 27
  文献评价指标  
  下载次数:16次 浏览次数:7次