Skip to main content
Log in

The role of CD146 in renal disease: from experimental nephropathy to clinics

  • Review
  • Published:
Journal of Molecular Medicine Aims and scope Submit manuscript

Abstract

Vascular endothelial dysfunction is a major risk factor in the development of renal diseases. Recent studies pointed out a major interest for the inter-endothelial junction protein CD146, as its expression is modulated during renal injury. Indeed, some complex mechanisms involving this adhesion molecule and its multiple ligands are observed in a large number of renal diseases in fundamental or clinical research. The purpose of this review is to summarize the most recent literature on the role of CD146 in renal pathophysiology, from experimental nephropathy to clinical trials.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

Availability of data and material

No data availability.

References

  1. Marcu R, Choi YJ, Xue J et al (2018) Human organ-specific endothelial cell heterogeneity. IScience 4

  2. Herzlinger D, Hurtado R (2014) Patterning the renal vascular bed. Semin Cell Dev Biol 36

  3. Wong BW, Marsch E, Treps L, Baes M, Carmeliet P (2017) Endothelial cell metabolism in health and disease: impact of hypoxia. EMBO J 36

  4. Jourde-Chiche N, Fakhouri F, Dou L et al (2019) Endothelium structure and function in kidney health and disease. Nat Rev Nephrol 15

  5. Makris K, Spanou L (2016) Acute kidney injury: definition, pathophysiology and clinical phenotypes. Clin Biochem Rev 37

  6. Levey AS, Eckardt KU, Tsukamoto Y et al (2005) Definition and classification of chronic kidney disease: a position statement from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int 67

  7. Geevarghese A, Herman IM (2014) Pericyte-endothelial crosstalk: Implications and opportunities for advanced cellular therapies. J Transl Res 163

  8. Verma SK, Molitoris BA (2015) Renal endothelial injury and microvascular dysfunction in acute kidney injury. Semin Nephrol 35

  9. Goligorsky MS (2015) Pathogenesis of endothelial cell dysfunction in chronic kidney disease: a retrospective and what the future may hold. Kidney Res Clin Pract 34

  10. Kalucka J, de Rooij LPMH, Goveia J et al (2020) Single-cell transcriptome atlas of murine endothelial cells. Cell 180

  11. Molema G, Aird WC (2012) Vascular heterogeneity in the kidney. Semin Nephrol 32(2)

  12. Dumas SJ, Meta E, Borri M et al (2020) Single-cell RNA sequencing reveals renal endothelium heterogeneity and metabolic adaptation to water deprivation. J Am Soc Nephrol 31

  13. Rosivall L, Peti-Peterdi J (2006) Heterogeneity of the afferent arteriole--correlations between morphology and function. Nephrol Dial Transplant 21

  14. Guerci P, Ergin B, Ince C (2017) The macro- and microcirculation of the kidney. Best Pract Res Clin Anaesthesiol 31

  15. Wang K, Kestenbaum B (2018) Proximal tubular secretory clearance: a neglected partner of kidney function. Clin J Am Soc Nephrol 13

  16. Bobulescu IA, Moe OW (2006) Na+/H+ Exchangers in renal regulation of acid-base balance. Semin Nephrol 26(5)

  17. Stanton BA, Giebisch GH (1982) Potassium transport by the renal distal tubule: effects of potassium loading. Am J Physiol Renal Fluid Electrolyte Physiol 12

  18. Eisner C, Faulhaber-Walter R, Wang Y et al (2010) Major contribution of tubular secretion to creatinine clearance in mice. Kidney Int 77

  19. Berkhin EB, Humphreys MH (2001) Regulation of renal tubular secretion of organic compounds. Kidney Int 59

  20. Pannabecker TL, Layton AT (2014) Targeted delivery of solutes and oxygen in the renal medulla: role of microvessel architecture. Am J Physiol Renal Physiol 307

  21. Pober JS, Sessa WC (2007) Evolving functions of endothelial cells in inflammation. Nat Rev Immunol 7

  22. Sandoo A, Veldhuijzen van Zanten JJCS, Metsios GS, Carroll D, Kitas GD (2015) The endothelium and its role in regulating vascular tone. Open Cardiovasc Med J 4(1)

  23. Neubauer K, Zieger B (2022) Endothelial cells and coagulation. Cell Tissue Res 387

  24. Jin J, Fang F, Gao W, Chen H, Wen J, Wen X et al (2021) The structure and function of the glycocalyx and its connection with blood-brain barrier. Front Cell Neurosci 15

  25. Roumenina LT, Rayes J, Frimat M, Fremeaux-Bacchi V (2016) Endothelial cells: source, barrier, and target of defensive mediators. Immunol Rev 274

  26. Rezaie AR (2014) Protease-activated receptor signalling by coagulation proteases in endothelial cells. Thromb Haemost 112

  27. Nomura K, Liu N, Nagai K et al (2007) Roles of coagulation pathway and factor Xa in rat mesangioproliferative glomerulonephritis. Lab Invest 87

  28. Moussa L, Apostolopoulos J, Davenport P, Tchongue J, Tipping PG (2007) Protease-activated receptor-2 augments experimental crescentic glomerulonephritis. Am J Pathol 171

  29. Chung H, Ramachandran R, Hollenberg MD, Muruve DA (2013) Proteinase-activated receptor-2 transactivation of epidermal growth factor receptor and transforming growth factor-β receptor signaling pathways contributes to renal fibrosis. J Biol Chem 288

  30. Caveda L, Corada M, Padura IM et al (1994) Structural characteristics and functional role of endothelial cell to cell junctions. Endothelium 2

  31. Dejana E, Del Maschio A (1995) Molecular organization and functional regulation of cell to cell junctions in the endothelium. Thromb Haemost

  32. Lum H, Malik AB (1994) Regulation of vascular endothelial barrier function. Am J Physiol Lung Cell Mol Physiol 267

  33. Vestweber D (2000) Molecular mechanisms that control endothelial cell contacts. J Pathol 190

  34. Bardin N, George F, Mutin M et al (1996) S-Endo 1, a pan-endothelial monoclonal antibody recognizing a novel human endothelial antigen. Tissue Antigens 48

  35. Johnson JP, Rothbcher U, Sers C (1993) The progression associated antigen MUC18: a unique member of the immunoglobulin supergene family. Melanoma Res 3

  36. Wang Z, Yan X (2013) CD146, a multi-functional molecule beyond adhesion. Cancer Lett 330

  37. Dye DE, Karlen S, Rohrbach B et al (2009) hShroom1 links a membrane bound protein to the actin cytoskeleton. Cell Mol Life Sci 66

  38. Bu P, Zhuang J, Feng J, Yang D, Shen X, Yan X (2007) Visualization of CD146 dimerization and its regulation in living cells. Biochim Biophys Acta Mol Cell Res 1773

  39. Stowell SR, Cho M, Feasley CL et al (2009) Ligand reduces galectin-1 sensitivity to oxidative inactivation by enhancing dimer formation. J Biol Chem 284

  40. Leroyer AS, Blin MG, Bachelier R, Bardin N, Blot-Chabaud M, Dignat-George F (2019) CD146 (cluster of differentiation 146): an adhesion molecule involved in vessel homeostasis. Arterioscler Thromb Vasc Biol

  41. Bardin N, Moal V, Anfosso F et al (2003) Soluble CD146, a novel endothelial marker, is increased in physiopathological settings linked to endothelial junctional alteration. Thromb Haemost 90

  42. Bardin N, Francès V, Combes V, Sampol J, Dignat-George F (1998) CD146: biosynthesis and production of a soluble form in human cultured endothelial cells. FEBS Lett 421

  43. Bardin N, Blot-Chabaud M, Despoix N et al (2009) CD146 and its soluble form regulate monocyte transendothelial migration. Arterioscler Thromb Vasc Biol 29

  44. Anfosso F, Bardin N, Francès V et al (1998) Activation of human endothelial cells via S-Endo-1 antigen (CD146) stimulates the tyrosine phosphorylation of focal adhesion kinase p125 (FAK). J Biol Chem 273

  45. Wang D, Duan H, Feng J et al (2020) Soluble CD146, a cerebrospinal fluid marker for neuroinflammation, promotes blood-brain barrier dysfunction. Theranostics 10

  46. Kaspi E, Heim X, Granel B et al (2017) Identification of CD146 as a novel molecular actor involved in systemic sclerosis. J Allergy Clin Immunol 140

  47. Vainio O, Dunon D, Aïssi F, Dangy JP, McNagny KM, Imhof BA (1996) HEMCAM, an adhesion molecule expressed by c-kit+ hemopoietic progenitors. J Cell Biol 135

  48. Miner JH, Yurchenco PD (2004) Laminin functions in tissue morphogenesis. Annu Rev Cell Dev Biol 20

  49. Durbeej M (2010) Laminins. Cell Tissue Res 339

  50. Fujiwara H, Kikkawa Y, Sanzen N, Sekiguchi K (2001) Purification and characterization of human laminin-8. Laminin-8 stimulates cell adhesion and migration through α3β1 and α6β1 integrins. J Biol Chem 276

  51. Petäjäniemi N, Korhonen M, Kortesmaa J et al (2002) Localization of laminin α4-chain in developing and adult human tissues. J Histochem Cytochem 50

  52. Flanagan K, Fitzgerald K, Baker J et al (2012) Laminin-411 is a vascular ligand for MCAM and facilitates TH17 cell entry into the CNS. PLoS One 7

  53. Kawataki T, Yamane T, Naganuma H et al (2007) Laminin isoforms and their integrin receptors in glioma cell migration and invasiveness: evidence for a role of α5-laminin(s) and α3β1 integrin. Exp Cell Res 313

  54. Takkunen M, Ainola M, Vainionpää N et al (2008) Epithelial-mesenchymal transition downregulates laminin α5 chain and upregulates laminin α4 chain in oral squamous carcinoma cells. Histochem Cell Biol 130

  55. Thijssen VL, Rabinovich GA, Griffioen AW (2013) Vascular galectins: regulators of tumor progression and targets for cancer therapy. Cytokine Growth Factor Rev 24

  56. Barondes SH, Castronovo V, Cooper DNW et al (1994) Galectins: a family of animal β-galactoside-binding lectins. Cell 76

  57. Stowell SR, Arthur CM, Mehta P et al (2008) Galectin-1, -2, and -3 exhibit differential recognition of sialylated glycans and blood group antigens. J Biol Chem 283

  58. Croci DO, Cerliani JP, Dalotto-Moreno T et al (2014) Glycosylation-dependent lectin-receptor interactions preserve angiogenesis in anti-VEGF refractory tumors. Cell 156

  59. Burns JS, Kristiansen M, Kristensen LP et al (2011) Decellularized matrix from tumorigenic human mesenchymal stem cells promotes neovascularization with galectin-1 dependent endothelial interaction. PLoS One 6

  60. Jouve N, Despoix N, Espeli M et al (2013) The involvement of CD146 and its novel ligand galectin-1 in apoptotic regulation of endothelial cells. J Biol Chem 288

  61. Yazawa EM, Geddes-Sweeney JE, Cedeno-Laurent F et al (2015) Melanoma cell galectin-1 ligands functionally correlate with malignant potential. J Invest Dermatol 135

  62. Kaltner H, Gabius HJ (2012) A toolbox of lectins for translating the sugar code: the galectin network in phylogenesis and tumors. Histol Histopathol 27

  63. Smetana K, André S, Kaltner H, Kopitz J, Gabius HJ (2013) Context-dependent multifunctionality of galectin-1: a challenge for defining the lectin as therapeutic target. Expert Opin Ther Targets 17

  64. Colomb F, Wang W, Simpson D et al (2017) Galectin-3 interacts with the cell-surface glycoprotein CD146 (MCAM, MUC18) and induces secretion of metastasispromoting cytokines from vascular endothelial cells. J Biol Chem 292

  65. Zhang Z, Zheng Y, Wang H, Zhou Y, Tai G (2018) CD146 interacts with galectin-3 to mediate endothelial cell migration. FEBS Lett 592

  66. Zhang Z, Miller MC, Xu X et al (2019) NMR-based insight into galectin-3 binding to endothelial cell adhesion molecule CD146: evidence for noncanonical interactions with the lectin’s CRD β-sandwich F-face. Glycobiology 29

  67. Donato R, R. Cannon B, Sorci G et al (2012) Functions of S100 proteins. Curr Mol Med 13

  68. Ryckman C, Vandal K, Rouleau P, Talbot M, Tessier PA (2003) Proinflammatory activities of S100: proteins S100A8, S100A9, and S100A8/A9 induce neutrophil chemotaxis and adhesion. J Immunol 170

  69. Koch M, Chitayat S, Dattilo BM et al (2010) Structural basis for ligand recognition and activation of RAGE. Structure 18

  70. Ehrchen JM, Sunderkötter C, Foell D, Vogl T, Roth J (2009) The endogenous Toll–like receptor 4 agonist S100A8/S100A9 (calprotectin) as innate amplifier of infection, autoimmunity, and cancer. J Leukoc Biol 86

  71. Riuzzi F, Sorci G, Donato R (2011) S100B protein regulates myoblast proliferation and differentiation by activating FGFR1 in a bFGF-dependent manner. J Cell Sci 124

  72. Ruma IMW, Putranto EW, Kondo E et al (2016) MCAM, as a novel receptor for S100A8/A9, mediates progression of malignant melanoma through prominent activation of NF-κB and ROS formation upon ligand binding. Clin Exp Metastasis 33

  73. Chen Y, Sumardika IW, Tomonobu N et al (2019) Melanoma cell adhesion molecule is the driving force behind the dissemination of melanoma upon S100A8/A9 binding in the original skin lesion. Cancer Lett 452

  74. Ustach C V., Huang W, Conley-LaComb MK et al (2010) A novel signaling axis of matriptase/PDGF-D/β-PDGFR in human prostate cancer. Cancer Res 70

  75. List K (2009) Matriptase: a culprit in cancer? Future Oncol 5

  76. Tung HH, Lee SL (2017) Physical binding of endothelial MCAM and neural transmembrane protease matriptase - novel cell adhesion in neural stem cell vascular niche. Sci Rep 7

  77. Alexander SP, Fabbro D, Kelly E et al (2015) The concise guide to pharmacology in 2015, the catalytic receptors. Br Pharmacol 172

  78. Witmer AN, Dai J, Weich HA, Vrensen GFJM, Schlingemann RO (2002) Expression of vascular endothelial growth factor receptors 1, 2, and 3 in quiescent endothelia. J Histochem Cytochem 50

  79. Ishida A, Murray J, Saito Y et al (2001) Expression of vascular endothelial growth factor receptors in smooth muscle cells. J Cell Physiol 188

  80. Sweeney MD, Ayyadurai S, Zlokovic B V (2016) Pericytes of the neurovascular unit: key functions and signaling pathways. Nat Neurosci 19

  81. Armulik A, Genové G, Betsholtz C (2011) Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell 21

  82. Jiang T, Zhuang J, Duan H et al (2012) CD146 is a coreceptor for VEGFR-2 in tumor angiogenesis. Blood 120

  83. Folkman J, Kaipainen A (2004) Genes tell lymphatics to sprout or not. Nat Immunol 5

  84. Karkkainen MJ, Haiko P, Sainio K et al (2004) Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from embryonic veins. Nat Immunol 5

  85. Yan H, Zhang C, Wang Z et al (2017) CD146 is required for VEGF-C-induced lymphatic sprouting during lymphangiogenesis. Sci Rep 7

  86. Heldin CH (2013) Targeting the PDGF signaling pathway in tumor treatment. Cell Commun Signal 11

  87. Onimaru M, Yonemitsu Y, Fujii T et al (2009) VEGF-C regulates lymphangiogenesis and capillary stability by regulation of PDGF-B. Am J Physiol Heart Circ Physiol 297

  88. Shang Q, Zhao L, Wang X, Wang M, Sui SF, Mi LZ (2017) Expression and purification of functional PDGF receptor beta. Biochem Biophys Res Commun 489

  89. Ye Z, Zhang C, Tu T et al (2013) Wnt5a uses CD146 as a receptor to regulate cell motility and convergent extension. Nat Commun 4

  90. Zhang L, Luo Y, Teng X et al (2018) CD146: a potential therapeutic target for systemic sclerosis. Protein Cell 9

  91. Serafini T, Colamarino SA, Leonardo ED et al (1996) Netrin-1 is required for commissural axon guidance in the developing vertebrate nervous system. Cell 87

  92. Mehlen P, Guenebeaud C (2010) Netrin-1 and its dependence receptors as original targets for cancer therapy. Curr Opin Oncol 22

  93. Mehlen P, Mazelin L (2003) The dependence receptors DCC and UNC5H as a link between neuronal guidance and survival. Biol Cell 95

  94. Layne K, Ferro A, Passacquale G (2015) Netrin-1 as a novel therapeutic target in cardiovascular disease: to activate or inhibit? Cardiovasc Res 107

  95. Ramesh G, Kwon O, Ahn K (2010) Netrin-1: a novel universal biomarker of human kidney injury. Transplant Proc 42

  96. Aimi F, Georgiopoulou S, Kalus I et al (2015) Endothelial Rictor is crucial for midgestational development and sustained and extensive FGF2-induced neovascularization in the adult. Sci Rep 5

  97. Herriges JC, Verheyden JM, Zhang Z et al (2015) FGF-regulated ETV transcription factors control FGF-SHH feedback loop in lung branching. Dev Cell 35

  98. Fernandes-Freitas I, Owen BM (2020) Metabolic roles of endocrine fibroblast growth factors. Curr Opin Pharmacol 25

  99. Gao Q, Zhang J, Wang X et al (2017) The signalling receptor MCAM coordinates apical-basal polarity and planar cell polarity during morphogenesis. Nat Commun 8

  100. Allen BL, Filla MS, Rapraeger AC (2001) Role of heparan sulfate as a tissue-specific regulator of FGF-4 and FGF receptor recognition. J Cell Biol 155

  101. Stefanska A, Kenyon C, Christian HC et al (2016) Human kidney pericytes produce renin. Kidney Int 90

  102. Kwon O, Miller S, Li N, Khan A, Kadry Z, Uemura T (2010) Bone marrow-derived endothelial progenitor cells and endothelial cells may contribute to endothelial repair in the kidney immediately after ischemia-reperfusion. J Histochem Cytochem 58

  103. Bruno S, Bussolati B, Grange C et al (2009) Isolation and characterization of resident mesenchymal stem cells in human glomeruli. Stem Cells Dev 18

  104. Pippin JW, Kaverina N V., Eng DG et al (2015) Cells of Renin lineage are adult pluripotent progenitors in experimental glomerular disease. Am J Physiol Renal Physiol 309

  105. Abed A, Leroyer AS, Kavvadas P et al (2021) Endothelial-Specific Deletion of CD146 Protects Against Experimental Glomerulonephritis in Mice. Hypertension 77

  106. Li X, Wen J, Dong Y et al (2021) Wnt5a promotes renal tubular inflammation in diabetic nephropathy by binding to CD146 through noncanonical Wnt signaling. Cell Death Dis 12

  107. Wang F, Xing T, Wang N, Liu L (2012) Clinical significance of plasma CD146 and P-selectin in patients with type 2 diabetic nephropathy. Cytokine 57

  108. Briesemeister D, Sommermeyer D, Loddenkemper C et al (2011) Tumor rejection by local interferon gamma induction in established tumors is associated with blood vessel destruction and necrosis. Int J Cancer 128

  109. Roeder SS, Stefanska A, Eng DG et al (2015) Changes in glomerular parietal epithelial cells in mouse kidneys with advanced age. Am J Phys Renal Physiol 309

  110. Stasi A, Franzin R, Divella C, Gesualdo L, Stallone G, Castellano G (2020) Double labeling of PDGFR-β and α-SMA in swine models of acute kidney injury to detect pericyte-to-myofibroblast transdifferentation as early marker of fibrosis. Bio-protocol 10

  111. Zhao Y, Zhao H, Zhang Y et al (2014) Isolation and epithelial co-culture of mouse renal peritubular endothelial cells. BMC Cell Biol 15

  112. Fan Y, Fei Y, Zheng L et al (2018) Expression of endothelial cell injury marker Cd146 correlates with disease severity and predicts the renal outcomes in patients with diabetic nephropathy. Cell Physiolo Biochem Int J Exp Cell Physiol Biochem Pharmacol 48

  113. Ji L, Wu GM, Yang LC, Li L, Xu H (2009) Expression of adhension molecule CD146 in renal tubular epithelial cells and its clinical significance in IgA nephropathy. J Sichuan Univ 40

  114. Feng G, Fang F, Liu C, Zhang F, Huang H, Pu C (2012) CD146 gene expression in clear cell renal cell carcinoma: a potential marker for prediction of early recurrence after nephrectomy. Int Urol Nephrol 44

  115. Wragg J, Finnity JP, Anderson JA et al (2016) MCAM and LAMA4 are highly enriched in tumor blood vessels of renal cell carcinoma and predict patient outcome. Cancer Res 76

  116. Małyszko J, Małyszko JS, Brzosko S, Wołczynski S, Myśliwiec M (2005) Markers of endothelial cell activation/injury: CD146 and thrombomodulin are related to adiponectin in kidney allograft recipients. Am J Nephrol 25

  117. Karbowska A, Boratynska M, Kusztal M, Klinger M (2009) Hyperuricemia is a mediator of endothelial dysfunction and inflammation in renal allograft recipients. Transplant Proc 41

  118. Boratyńska M, Karbowska A, Klinger M (2010) The effect of hyperuricemia on endothelial biomarkers and renal function in kidney allograft recipients. Transplant Proc 42:4074–4077

    Article  PubMed  Google Scholar 

  119. Liao J, Fu Q, Chen W et al (2020) Plasma soluble CD146 as a potential diagnostic marker of acute rejection in kidney transplantation. Front Med 7

  120. Daniel L, Bardin N, Moal V, Dignat-George F, Berland Y, Figarella-Branger D (2005) Tubular CD146 expression in nephropathies is related to chronic renal failure. Nephron. Exp Nephrol 99

  121. Dursun I, Poyrazoglu HM, Gunduz Z et al (2009) The relationship between circulating endothelial microparticles and arterial stiffness and atherosclerosis in children with chronic kidney disease. Nephrology Dial Transplant 24

  122. Prud’homme M, Coutrot M, Michel T et al (2019) Acute kidney injury induces remote cardiac damage and dysfunction through the galectin-3 pathway. JACC Basic Transl Sci 4

Download references

Acknowledgements

Figures were made with Servier Medical Art by Servier, which is licensed under a Creative Commons Attribution 3.0 Unported License.

Funding

This work was supported by the French National Research Agency (ANR GalCAM-AKI grant to EG, MBC and CEC, ANR-22-CE14-0045), the French National Institute of Health and Medical Research (Inserm) and Sorbonne University. ER is a doctoral fellow of the French Ministry of National Education (“Ecole Doctorale de Physiologie and Physiopathologie,” ED 394). LB is a fellow of the SFAR (“Société Française d’Anesthésie et de Réanimation”).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, LB, CEC; writing—original draft preparation, LB, ER, MBC, and CEC; writing—review and editing, LB, ER, EG, FD, MCB, and CEC; supervision, CEC; project administration, CEC; funding acquisition, CEC. All authors have read and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Christos E. Chadjichristos.

Ethics declarations

Ethics approval and consent to participate

Not applicable. This study does not involve experiments.

Conflict of interest

The authors declare no competing interests.

Consent for publication

All authors approved the final version of the manuscript.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Boutin, L., Roger, E., Gayat, E. et al. The role of CD146 in renal disease: from experimental nephropathy to clinics. J Mol Med 102, 11–21 (2024). https://doi.org/10.1007/s00109-023-02392-7

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00109-023-02392-7

Keywords

Navigation