Thromb Haemost 2017; 117(04): 769-783
DOI: 10.1160/TH16-10-0790
Stroke, Systemic or Venous Thromboembolism
Schattauer GmbH

From thrombosis to fibrosis in chronic thromboembolic pulmonary hypertension

Magdalena L. Bochenek
1   Center for Cardiology, Cardiology I, University Medical Center Mainz, Germany
2   Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany
3   Deutsches Zentrum für Herz-Kreislauf-Forschung e. V., partner site RheinMain (Mainz), Germany
,
Nico S. Rosinus
1   Center for Cardiology, Cardiology I, University Medical Center Mainz, Germany
3   Deutsches Zentrum für Herz-Kreislauf-Forschung e. V., partner site RheinMain (Mainz), Germany
,
Mareike Lankeit
2   Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany
,
Lukas Hobohm
1   Center for Cardiology, Cardiology I, University Medical Center Mainz, Germany
2   Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany
,
Felix Bremmer
4   Department of Pathology, Medical University of Göttingen, Germany
,
Eva Schütz
1   Center for Cardiology, Cardiology I, University Medical Center Mainz, Germany
,
Frederikus A. Klok
2   Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany
,
Sven Horke
2   Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany
5   Department of Pharmacology, University Medical Center Mainz, Germany
,
Christoph B. Wiedenroth
6   Thoracic Surgery, Kerckhoff-Clinic, Bad Nauheim, Germany
,
Thomas Münzel
1   Center for Cardiology, Cardiology I, University Medical Center Mainz, Germany
3   Deutsches Zentrum für Herz-Kreislauf-Forschung e. V., partner site RheinMain (Mainz), Germany
,
Irene M. Lang
7   Department of Internal Medicine, Division of Cardiology, Medical University of Vienna, Vienna, Austria
,
Eckhard Mayer
6   Thoracic Surgery, Kerckhoff-Clinic, Bad Nauheim, Germany
,
Stavros Konstantinides
2   Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany
8   Department of Cardiology, Democritus University of Thrace, Greece
,
Katrin Schäfer
1   Center for Cardiology, Cardiology I, University Medical Center Mainz, Germany
3   Deutsches Zentrum für Herz-Kreislauf-Forschung e. V., partner site RheinMain (Mainz), Germany
› Author Affiliations
Financial support: This study was supported by the Bundesministerium für Bildung und Forschung (BMBF 01E01003; Virchow fellowship to MLB) and the Deutsches Zentrum für Herz-Kreislaufforschung (DZHK Doktorandenstipendium to NSR).
Further Information

Publication History

Received: 17 October 2016

Accepted after major revision: 29 February 2016

Publication Date:
13 November 2017 (online)

Summary

The pathomechanisms underlying the development of thrombofibrotic pulmonary artery occlusions in Chronic Thromboembolic Pulmonary Hypertension (CTEPH) are largely unknown. The aim of this study was to allocate distinct cellular processes playing a role in thrombus resolution, such as inflammation, hypoxia, proliferation, apoptosis and angiogenesis, to different stages of thrombofibrotic remodelling. A total of 182 pulmonary endarterectomy (PEA) specimens were collected from 31 CTEPH patients. To facilitate co-localisation, Tissue MicroArrays were prepared and processed for (immuno)-histochemistry and confocal fluorescence microscopy. Murine venous thrombus formation and resolution was examined after inferior vena cava ligation. PEA tissues exhibited five morphologically distinct regions predominantly consisting of either fibrin-, erythrocyte- or extracellular matrix-rich thrombus, myofibroblasts, vessels or fibrotic tissue, and were found to resemble chronological stages of thrombus resolution in mice. Cellularity was highest in vessel-rich regions, and numerous cells were strongly positive for HIF1α or HIF2α as well as markers of activated VEGF signalling, including endothelial nitric oxide synthase. On the other hand, negative regulators of angiogenic growth factor signalling and reactive oxygen species were also highly expressed. Immune cells, primarily macrophages of the M2 subtype and CD117 haematopoietic progenitors were detected and highest in vascularised regions. Our findings demonstrate the simultaneous presence of different stages of thrombus organisation and suggest that hypoxia-induced endothelial, mesenchymal and immune cell activation may contribute to thrombofibrosis in CTEPH. This systematic histological characterisation of the material obstructing pulmonary vessels in CTEPH may provide a valuable basis for further studies aimed at determining causal factors underlying this disease.

Supplementary Material to this article is available online at www.thrombosis-online.com.

 
  • References

  • 1 Galie N, Humbert M, Vachiery JL. et al. 2015 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension: The Joint Task Force for the Diagnosis and Treatment of Pulmonary Hypertension of the European Society of Cardiology (ESC) and the European Respiratory Society (ERS): Endorsed by: Association for European Paediatric and Congenital Cardiology (AEPC), International Society for Heart and Lung Transplantation (ISHLT). Eur Heart J 2016; 37: 67-119
  • 2 Kim NH, Lang IM. Risk factors for chronic thromboembolic pulmonary hypertension. Eur Respir Rev 2012; 21: 27-31
  • 3 Lang IM, Pesavento R, Bonderman D. et al. Risk factors and basic mechanisms of chronic thromboembolic pulmonary hypertension: a current understanding. Eur Respir J 2013; 41: 462-468
  • 4 Wong CL, Szydlo R, Gibbs S. et al. Hereditary and acquired thrombotic risk factors for chronic thromboembolic pulmonary hypertension. Blood Coagul Fibrinolysis 2010; 21: 201-206
  • 5 Hosokawa K, Ishibashi-Ueda H, Kishi T. et al. Histopathological multiple recanalized lesion is critical element of outcome after pulmonary thromboendarterectomy. Int Heart J 2011; 52: 377-381
  • 6 Yamaki S, Ando M, Fukumoto Y. et al. Histopathological examination by lung biopsy for the evaluation of operability and postoperative prognosis in patients with chronic thromboembolic pulmonary hypertension. Circ J 2014; 78: 476-482
  • 7 Pengo V, Lensing AW, Prins MH. et al. Incidence of chronic thromboembolic pulmonary hypertension after pulmonary embolism. N Engl J Med 2004; 350: 2257-2264
  • 8 Guerin L, Couturaud F, Parent F. et al. Prevalence of chronic thromboembolic pulmonary hypertension after acute pulmonary embolism. Prevalence of CTEPH after pulmonary embolism. Thromb Haemost 2014; 112: 598-605
  • 9 Lang I. Chronic thromboembolic pulmonary hypertension: a distinct disease entity. Eur Respir Rev 2015; 24: 246-252
  • 10 Morris TA, Marsh JJ, Chiles PG. et al. Fibrin derived from patients with chronic thromboembolic pulmonary hypertension is resistant to lysis. Am J Respir Crit Care Med 2006; 173: 1270-1275
  • 11 Maruoka M, Sakao S, Kantake M. et al. Characterization of myofibroblasts in chronic thromboembolic pulmonary hypertension. Int J Cardiol 2012; 159: 119-127
  • 12 Masri FA, Xu W, Comhair SA. et al. Hyperproliferative apoptosis-resistant endothelial cells in idiopathic pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2007; 293: L548-554
  • 13 von Bruhl ML, Stark K, Steinhart A. et al. Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med 2012; 209: 819-835
  • 14 Zhou J, May L, Liao P. et al. Inferior vena cava ligation rapidly induces tissue factor expression and venous thrombosis in rats. Arterioscler Thromb Vasc Biol 2009; 29: 863-869
  • 15 Jamieson SW, Kapelanski DP. Pulmonary endarterectomy. Curr Probl Surg 2000; 37: 165-252
  • 16 Saha P, Humphries J, Modarai B. et al. Leukocytes and the natural history of deep vein thrombosis: current concepts and future directions. Arterioscler Thromb Vasc Biol 2011; 31: 506-512
  • 17 Ogawa A, Firth AL, Ariyasu S. et al. Thrombin-mediated activation of Akt signaling contributes to pulmonary vascular remodeling in pulmonary hypertension. Physiol Rep 2013; 1: e00190
  • 18 Firth AL, Yao W, Ogawa A. et al. Multipotent mesenchymal progenitor cells are present in endarterectomized tissues from patients with chronic thromboembolic pulmonary hypertension. Am J Physiol Cell Physiol 2010; 298: C1217-225
  • 19 Sakao S, Taraseviciene-Stewart L, Lee JD. et al. Initial apoptosis is followed by increased proliferation of apoptosis-resistant endothelial cells. FASEB J 2005; 19: 1178-1780
  • 20 Jujo T, Sakao S, Kantake M. et al. Characterization of sarcoma-like cells derived from endarterectomized tissues from patients with CTEPH and establishment of a mouse model of pulmonary artery intimal sarcoma. Int J Oncol 2012; 41: 701-711
  • 21 Wang L, Gan HL, Liu Y. et al. The distinguishing cellular features of pulmonary artery smooth muscle cells from chronic thromboembolic pulmonary hypertension patients. Exp Lung Res 2013; 39: 349-358
  • 22 Tuder RM, Groves B, Badesch DB. et al. Exuberant endothelial cell growth and elements of inflammation are present in plexiform lesions of pulmonary hypertension. Am J Pathol 1994; 144: 275-285
  • 23 Zabini D, Heinemann A, Foris V. et al. Comprehensive analysis of inflammatory markers in chronic thromboembolic pulmonary hypertension patients. Eur Respir J 2014; 44: 951-962
  • 24 Stewart GJ. Neutrophils and deep venous thrombosis. Haemostasis 1993; 23 Suppl 1 127-140
  • 25 Singh I, Burnand KG, Collins M. et al. Failure of thrombus to resolve in urokinase-type plasminogen activator gene-knockout mice: rescue by normal bone marrow-derived cells. Circulation 2003; 107: 869-875
  • 26 Quarck R, Wynants M, Verbeken E. et al. Contribution of inflammation and impaired angiogenesis to the pathobiology of chronic thromboembolic pulmonary hypertension. Eur Respir J 2015; 46: 431-443
  • 27 Groth A, Vrugt B, Brock M. et al. Inflammatory cytokines in pulmonary hypertension. Respir Res 2014; 15: 47
  • 28 Wynn TA, Barron L. Macrophages: master regulators of inflammation and fibrosis. Semin Liver Dis 2010; 30: 245-257
  • 29 Kodelja V, Muller C, Tenorio S. et al. Differences in angiogenic potential of classically vs alternatively activated macrophages. Immunobiology 1997; 197: 478-493
  • 30 Arbustini E, Morbini P, D’Armini AM. et al. Plaque composition in plexogenic and thromboembolic pulmonary hypertension: the critical role of thrombotic material in pultaceous core formation. Heart 2002; 88: 177-182
  • 31 Evans CE, Humphries J, Mattock K. et al. Hypoxia and upregulation of hypoxia-inducible factor 1{alpha} stimulate venous thrombus recanalization. Arterioscler Thromb Vasc Biol 2010; 30: 2443-2451
  • 32 Skuli N, Liu L, Runge A. et al. Endothelial deletion of hypoxia-inducible factor-2alpha (HIF-2alpha) alters vascular function and tumor angiogenesis. Blood 2009; 114: 469-477
  • 33 Lawson CA, Yan SD, Yan SF. et al. Monocytes and tissue factor promote thrombosis in a murine model of oxygen deprivation. J Clin Invest 1997; 99: 1729-1738
  • 34 Uchiyama T, Kurabayashi M, Ohyama Y. et al. Hypoxia induces transcription of the plasminogen activator inhibitor-1 gene through genistein-sensitive tyrosine kinase pathways in vascular endothelial cells. Arterioscler Thromb Vasc Biol 2000; 20: 1155-1561
  • 35 Brill A, Suidan GL, Wagner DD. Hypoxia, such as encountered at high altitude, promotes deep vein thrombosis in mice. J Thromb Haemost 2013; 11: 1773-1775
  • 36 Modarai B, Burnand KG, Humphries J. et al. The role of neovascularisation in the resolution of venous thrombus. Thromb Haemost 2005; 93: 801-809
  • 37 Alias S, Redwan B, Panzenbock A. et al. Defective angiogenesis delays thrombus resolution: a potential pathogenetic mechanism underlying chronic thromboembolic pulmonary hypertension. Arterioscler Thromb Vasc Biol 2014; 34: 810-819
  • 38 Waltham M, Burnand KG, Collins M. et al. Vascular endothelial growth factor enhances venous thrombus recanalisation and organisation. Thromb Haemost 2003; 89: 169-176
  • 39 Evans CE, Grover SP, Humphries J. et al. Antiangiogenic therapy inhibits venous thrombus resolution. Arterioscler Thromb Vasc Biol 2014; 34: 565-570
  • 40 Cool CD, Stewart JS, Werahera P. et al. Three-dimensional reconstruction of pulmonary arteries in plexiform pulmonary hypertension using cell-specific markers. Evidence for a dynamic and heterogeneous process of pulmonary endothelial cell growth Am J Pathol 1999; 155: 411-419
  • 41 Chua CC, Hamdy RC, Chua BH. Upregulation of vascular endothelial growth factor by H2O2 in rat heart endothelial cells. Free Radic Biol Med 1998; 25: 891-897
  • 42 Knighton DR, Hunt TK, Scheuenstuhl H. et al. Oxygen tension regulates the expression of angiogenesis factor by macrophages. Science 1983; 221: 1283-1285
  • 43 Waltham M, Burnand KG, Collins M. et al. Vascular endothelial growth factor and basic fibroblast growth factor are found in resolving venous thrombi. J Vasc Surg 2000; 32: 988-996
  • 44 Lanahan AA, Lech D, Dubrac A. et al. PTP1b is a physiologic regulator of vascular endothelial growth factor signaling in endothelial cells. Circulation 2014; 130: 902-909
  • 45 Davies MH, Stempel AJ, Hubert KE. et al. Altered vascular expression of EphrinB2 and EphB4 in a model of oxygen-induced retinopathy. Dev Dyn 2010; 239: 1695-1707
  • 46 Eriksson O, Ramstrom M, Hornaeus K. et al. The Eph tyrosine kinase receptors EphB2 and EphA2 are novel proteolytic substrates of tissue factor/coagulation factor VIIa. J Biol Chem 2014; 289: 32379-3291
  • 47 Zeisberg EM, Tarnavski O, Zeisberg M. et al. Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat Med 2007; 13: 952-961
  • 48 Ranchoux B, Antigny F, Rucker-Martin C. et al. Endothelial-to-Mesenchymal Transition in Pulmonary Hypertension. Circulation 2015; 131: 1006-1018
  • 49 Xu X, Tan X, Tampe B. et al. Snail Is a Direct Target of Hypoxia-inducible Factor 1alpha (HIF1alpha) in Hypoxia-induced Endothelial to Mesenchymal Transition of Human Coronary Endothelial Cells. J Biol Chem 2015; 290: 16653-16664
  • 50 Arciniegas E, Sutton AB, Allen TD. et al. Transforming growth factor beta 1 promotes the differentiation of endothelial cells into smooth muscle-like cells in vitro. J Cell Sci 1992; 103: 521-529
  • 51 Smith P, Heath D. Electron microscopy of the plexiform lesion. Thorax 1979; 34: 177-186