期刊论文详细信息
BMC Immunology
Immunoexpression of TGF-β/Smad and VEGF-A proteins in serum and BAL fluid of sarcoidosis patients
Ewa Brzeziańska-Lasota2  Zofia Kurmanowska2  Karolina H. Czarnecka2  Ewa Nawrot2  Daria Domańska-Senderowska2  Monika Migdalska-Sęk2  Dorota Pastuszak-Lewandoska2  Witold Górski1  Adam Antczak3  Paweł Górski1  Justyna Kiszałkiewicz2  Wojciech J. Piotrowski1 
[1] Department of Pneumonology and Allergy, 1st Chair of Internal Medicine, Medical University of Lodz, Lodz, Poland;Department of Molecular Bases of Medicine, 1st Chair of Internal Medicine, Medical University of Lodz, 251 Pomorska St., Lodz, 92-213, Poland;Department of General and Oncological Pulmonology, 1st Chair of Internal Medicine, Medical University of Lodz, Lodz, Poland
关键词: Angiogenesis;    Growth factors;    Prognosis;    VEGF-A;    TGF-β1;    Sarcoidosis;   
Others  :  1228736
DOI  :  10.1186/s12865-015-0123-y
 received in 2015-01-28, accepted in 2015-09-28,  发布年份 2015
PDF
【 摘 要 】

Background

The chronic course of pulmonary sarcoidosis can lead to lung dysfunction due to fibrosis, in which the signalling pathways TGF-β/Smad and VEGF-A may play a key role.

Methods

We evaluated immunoexpression of TGF-β 1 , Smad2, 3, and 7, and VEGF-A in serum and bronchoalveolar lavage (BAL) fluid of patients (n = 57) classified according to the presence of lung parenchymal involvement (radiological stage I vs. II-III), acute vs. insidious onset, lung function test (LFT) results, calcium metabolism parameters, percentage of BAL lymphocytes (BAL-L%), BAL CD4 + /CD8 +ratio, age, and gender. Immunoexpression analysis of proteins was performed by ELISA.

Results

The immunoexpression of all studied proteins were higher in serum than in BAL fluid of patients (p >0.05). The serum levels of TGF-β 1(p = 0.03), Smad2 (p = 0.01), and VEGF-A (p = 0.0002) were significantly higher in sarcoidosis patients compared to healthy controls. There were no differences within the sarcoidosis group between patients with vs. without parenchymal involvement, acute vs. insidious onset, or patients with normal vs. abnormal spirometry results. In patients with abnormal spirometry results a negative correlation was found between forced vital capacity (FVC) % predicted value and TGF-β 1immunoexpression in BAL fluid, and positive correlations were observed between the intensity of lung parenchymal changes estimated by high-resolution computed tomography (HRCT scores) and Smad 2 level in serum.

Conclusions

TGF-β/Smad signalling pathway and VEGF-A participate in the pathogenesis of sarcoidosis. BAL TGF-β 1 , and Smad 2 in serum seem to be promising biomarkers with negative prognostic value, but further studies are required to confirmed our observations.

【 授权许可】

   
2015 Piotrowski et al.

【 预 览 】
附件列表
Files Size Format View
20151019021134327.pdf 563KB PDF download
Fig. 3. 13KB Image download
Fig. 2. 10KB Image download
Fig. 1. 8KB Image download
【 图 表 】

Fig. 1.

Fig. 2.

Fig. 3.

【 参考文献 】
  • [1]European Respiratory Society, World Association of Sarcoidosis and Other Granulomatous Disorders: Statement on sarcoidosis. Am J Respir Crit Care Med. 1999; 160:736-755.
  • [2]Ianuzzi MC, Rybicki BA, Teirstein AS. Medical Progress. Sarcoidosis. N Eng J Med. 2007; 357:2153-2165.
  • [3]Piotrowski WJ. Prognostic factors in sarcoidosis. In: Sarcoidosis Diagnosis and Management. Motamedi HKM, editor. InTech, Rijeka; 2011: p.137-162.
  • [4]Kim KK, Wei Y, Szekeres C, Kugler MC, Wolters PJ, Hill ML, Frank JA, Brumwell AN, Wheeler SE, Kreidberg JA, Chapman HA. Epithelial cell alpha3beta1 integrin links beta-catenin and Smad signaling to promote myofibroblast formation and pulmonary fibrosis. J Clin Invest. 2009; 119:213-224.
  • [5]Khalil N, O’Connor RN, Flanders KC, Unruh H. TGF-beta 1, but not TGF-beta 2 or TGF-beta 3, is differentially present in epithelial cells of advanced pulmonary fibrosis: an immunohistochemical study. Am J Respir Cell Mol Biol. 1996; 14:131-138.
  • [6]Penn JW, Grobbelaar AO, Rolfe KJ. The role of the TGF-β family in would healing, burns and scarring: a review. Int J Burn Trauma. 2012; 2:18-28.
  • [7]Farkas L, Farkas D, Ask K, Möller A, Gauldie J, Margetts P, Inman M, Kolb M. VEGF ameliorates pulmonary hypertension through inhibition of endothelial apoptosis in experimental lung fibrosis in rats. J Clin Invest. 2009; 119:1298-1311.
  • [8]Smadja DM, Nunes H, Juvin K, Bertil S, Valeyre D, Gaussem P, Israel-Biet D. Increase in both angiogenic and angiostatic mediators in patients with idiopathic pulmonary fibrosis. Pathol Biol (Paris). 2014; 62:391-394.
  • [9]Yamauchi K, Nishimura Y, Shigematsu S, Takeuchi Y, Nakamura J, Aizawa T, Hashizume K. Vascular Endothelial Cell Growth Factor attenuates actions of Transforming Growth Factor-β in human endothelial cells. J Biol Chem. 2004; 279:55104-55108.
  • [10]Cui Y, Osorio JC, Risquez C, Wang H, Shi Y, Gochuico BR, Morse D, Rosas IO, El-Chemaly S. Transforming Growth Factor-β1 downregulates Vascular Endothelial Growth Factor-D expression in human lung fibroblasts via the Jun NH2-Terminal Kinase signaling pathway. Mol Med. 2013; 20:120-134.
  • [11]Salez F, Gosset P, Copin MC, Lamblin Degros C, Tonnel AB, Wallaert B. Transforming growth factor-β1 in sarcoidosis. Eur Respir J. 1998; 12:913-919.
  • [12]Jonth AC, Silveira L, Fingerlin TE, Sato H, Luby JC, Welsh KI, Rose CS, Newman LS, du Bois RM, Maier LA. TGF-β1 variants in chronic beryllium disease and sarcoidosis. J Immunol. 2007; 179:4255-4262.
  • [13]Kruit A, Grutters JC, Ruven HJT, van Moorsel CHM, Weiskirchen R, Mengsteab S, van den Bosch JMM. Transforming growth factor-β gene polymorphisms in sarcoidosis patients with and without fibrosis. Chest. 2006; 129:1584-1591.
  • [14]Pabst S, Fränken T, Schönau J, Stier S, Nickening G, Meyer R, Skowasch D, Grohé C. Transforming growth factor-b gene polymorphisms in different phenotypes of sarcoidosis. Eur Respir J. 2011; 38:169-175.
  • [15]Antoniou KM, Soufla G, Proklou A, Margaritopoulos G, Choulaki C, Samara R, Spandidos DA, Siafakas NM. Different activity of the biological axis VEGF-Flt-1 (fms-like tyrosine kinase 1) and CXC chemokines between pulmonary sarcoidosis and idiopathic pulmonary fibrosis: a bronchoalveolar lavage study. Clin Dev Immunol. 2009; 2009:537929.
  • [16]Vasakova M, Sterclova M, Kolesar L, Slavcev A, Pohunek P, Sulc J, Skibova J, Striz I. Bronchoalveolar lavage fluid cellular characteristics, functional parameters and cytokine and chemokine levels in interstitial lung diseases. Scand J Immunol. 2009; 69:268-274.
  • [17]Fireman E, Gilburd D, Marmor S. Angiogenic cytokines in induced sputum of patients with sarcoidosis. Respirology. 2009; 14:117-123.
  • [18]Yamashita M, Mouri T, Nilsato M, Kowada K, Kobayashi H, Chiba R, Satoh T, Sugai T, Sawai T, Takahashi T, Yamauchi K. Heterogeneous characteristics of lymphatic microvasculatures associated with pulmonary sarcoid granulomas. Ann Am Thorac Soc. 2013; 10:90-97.
  • [19]Chciałowski A, Chorostowska-Wynimko J, Fal A, Pawłowicz R, Domagała-Kulawik J. Recommendation of the Polish Respiratory Society for bronchoalveolar lavage (BAL) sampling processing and analysis methods. Pneumonol Alergol Pol. 2011; 79:75-89.
  • [20]Standardized lung function testing. Official statement of the European Respiratory Society. Eur Respir J. 1993; 16(suppl):1-100.
  • [21]Tomalak W, Antczak A, Boros P, Czajkowska-Malinowska M, Franczuk M, Gondorowicz K et al.. Zalecenia Polskiego Towarzystwa Ftyzjopneumonologicznego dotyczące wykonywania badań spirometrycznych. Pneumonol Alergol Pol. 2006; 74(suppl 1):1-32.
  • [22]Drent M, De Vries J, Lenters M, Lamers RJ, Rothkranz-Kos S, Wouters EF, van Dieijen-Visser MP, Verschakelen JA. Sarcoidosis: assessment of disease severity using HRCT. Eur Radiol. 2003; 13:2462-2471.
  • [23]Miyara M, Amoura Z, Prizot C, Badoual C, Dorgham K, Trad S, Kambouchner M, Valeyre D, Chapelon-Abric C, Debré P, Piette JC, Gorochov G. The immune paradox of sarcoidosis and regulatory T cells. J Exp Med. 2006; 203:359-370.
  • [24]Idali F, Wahlstrom J, Muller-Suur C, Eklund A, Grunewald J. Analysis of regulatory T cell associated forkhead box P3 expression in the lungs of patients with sarcoidosis. Clin Exp Immunol. 2008; 152:127-137.
  • [25]Prior C, Knight RA, Herold M, Ott G, Spiteri MA. Pulmonary sarcoidosis: patterns of cytokine release in vitro. Eur Respir J. 1996; 9:47-53.
  • [26]Brown KA, Pietenpol JA, Moses HL. Tale of Two Proteins: Differential Roles and Regulation of Smad2 and Smad3 in TGF-b Signaling. J Cell Biochem. 2007; 101:9-33.
  • [27]Krafft E, Lybaert P, Roels E, Laurila HP, Rajamäki MM, Farnir F, Myllärniemi M, Day MJ, Mc Entee K, Clercx C. Transforming growth factor Beta 1 activation, storage, and signaling pathways in idiopathic pulmonary fibrosis in dogs. J Vet Intern Med. 2014; 28:1666-1675.
  • [28]Tzouvelekis A, Ntolios P, Karameris A, Koutsopoulos A, Boglou P, Koulelidis A, Archontogeorgis K, Zacharis G, Drakopanagiotakis F, Steiropoulos P, Anevlavis S, Polychronopoulos V, Mikroulis D, Bouros D. Expression of hypoxia-inducible factor (HIF)-1a-vascular endothelial growth factor (VEGF)-inhibitory growth factor (ING)-4- axis in sarcoidosis patients. BMC Research Notes. 2012; 5:654. BioMed Central Full Text
  • [29]Porębska I, Jankowska R, Piesiak P, Dyła T, Szeliga W. VEGF and IL-8 analysis in BALF and sera of patients with sarcoidosis. Adv Clin Exp Med. 2004; 13:555-560.
  • [30]Zielonka TM, Demkow U, Radzikowska E, Białas B, Filewska M, Życińska K, Obrowski MH, Kowalski J, Wardyn KA, Skopińska-Różewska E. Angiogenic activity of sera from interstitial lung disease patients in relations to pulmonary function. Eur J Med Res. 2010; 15:229-234.
  • [31]Ziora D, Dworniczak S, Niepsuj G, Niepsuj K, Jarosz W, Sielska-Sytek E, Ciekalska K, Oklek K. Proangiogenic cytokines (bFGF and VEGF) in BALF from two different lung segments examined by high resolution computed tomography (HRCT) in patients with sarcoidosis. Pneumonol Alergol Pol. 2000; 68:120-130.
  文献评价指标  
  下载次数:16次 浏览次数:5次