open access

Vol 74, No 1 (2023)
Original paper
Submitted: 2022-12-16
Accepted: 2022-12-17
Published online: 2023-02-02
Get Citation

Whole-exome sequencing as a tool for searching for genetic background modifiers in MEN1 patients with neuroendocrine pancreatic tumours, including insulinomas

Anna Skalniak1, Małgorzata Trofimiuk-Müldner1, Agata Jabrocka-Hybel1, Justyna Totoń-Żurańska2, Paweł Wołkow2, Alicja Hubalewska-Dydejczyk1
·
Pubmed: 36847722
·
Endokrynol Pol 2023;74(1):31-46.
Affiliations
  1. Chair and Department of Endocrinology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
  2. Center for Medical Genomics — OMICRON, Jagiellonian University Medical College, Krakow, Poland

open access

Vol 74, No 1 (2023)
Original Paper
Submitted: 2022-12-16
Accepted: 2022-12-17
Published online: 2023-02-02

Abstract

Introduction: Multiple endocrine neoplasia type 1 (MEN1) is a monogenic disease caused by inactivating variants in the MEN1 gene. Although the reason for its development is well-known, disease phenotypes are unpredictable and differ even among carriers of the same pathogenic driver mutation. Genetic, epigenetic, and environmental factors may play a role in driving the individual phenotype. Those factors, however, still mostly remain unidentified. In our work, we focused on the inherited genetic background in pancreatic neuroendocrine neoplasms (pNENs) in MEN1 patients, and the pancreatic tumour subgroup with insulinoma.

Material and methods: Whole exome sequencing was performed in MEN1 patients. The symptoms of interest were pancreatic neuroendocrine tumours in one analysis and insulinoma in the second. The study included families as well as unrelated cases. Genes with variants that are not neutral to the encoded gene product were defined in symptom-positive patients as compared to symptom-negative controls. The interpretation of the results was based on functional annotations and pathways shared between all patients with the given symptom in the course of MEN1.

Results: Whole-exome screening of family members and unrelated patients with and without pNENs revealed a number of pathways that are common for all the analysed cases with pNENs. Those included pathways crucial for morphogenesis and development, proper insulin signalling, and structural cellular organization. An additional analysis of insulinoma pNEN patients revealed additional pathways engaged in glucose and lipid homeostasis, and several non-canonical insulin-regulating mechanisms.

Conclusions: Our results show the existence of pathways that are identified in a non-literature-predefined manner, which might have a modifying function in MEN1, differentiating the specific clinical outcomes. Those results, although preliminary, provide evidence of the reasonableness of performing large-scale studies addressing the genetic background of MEN1 patients in determining their individual outcomes.

Abstract

Introduction: Multiple endocrine neoplasia type 1 (MEN1) is a monogenic disease caused by inactivating variants in the MEN1 gene. Although the reason for its development is well-known, disease phenotypes are unpredictable and differ even among carriers of the same pathogenic driver mutation. Genetic, epigenetic, and environmental factors may play a role in driving the individual phenotype. Those factors, however, still mostly remain unidentified. In our work, we focused on the inherited genetic background in pancreatic neuroendocrine neoplasms (pNENs) in MEN1 patients, and the pancreatic tumour subgroup with insulinoma.

Material and methods: Whole exome sequencing was performed in MEN1 patients. The symptoms of interest were pancreatic neuroendocrine tumours in one analysis and insulinoma in the second. The study included families as well as unrelated cases. Genes with variants that are not neutral to the encoded gene product were defined in symptom-positive patients as compared to symptom-negative controls. The interpretation of the results was based on functional annotations and pathways shared between all patients with the given symptom in the course of MEN1.

Results: Whole-exome screening of family members and unrelated patients with and without pNENs revealed a number of pathways that are common for all the analysed cases with pNENs. Those included pathways crucial for morphogenesis and development, proper insulin signalling, and structural cellular organization. An additional analysis of insulinoma pNEN patients revealed additional pathways engaged in glucose and lipid homeostasis, and several non-canonical insulin-regulating mechanisms.

Conclusions: Our results show the existence of pathways that are identified in a non-literature-predefined manner, which might have a modifying function in MEN1, differentiating the specific clinical outcomes. Those results, although preliminary, provide evidence of the reasonableness of performing large-scale studies addressing the genetic background of MEN1 patients in determining their individual outcomes.

Get Citation

Keywords

multiple endocrine neoplasia type 1; neuroendocrine tumours; pancreatic neoplasms; insulinoma; modifier; genetic background; exome sequencing

About this article
Title

Whole-exome sequencing as a tool for searching for genetic background modifiers in MEN1 patients with neuroendocrine pancreatic tumours, including insulinomas

Journal

Endokrynologia Polska

Issue

Vol 74, No 1 (2023)

Article type

Original paper

Pages

31-46

Published online

2023-02-02

Page views

2907

Article views/downloads

530

DOI

10.5603/EP.a2023.0009

Pubmed

36847722

Bibliographic record

Endokrynol Pol 2023;74(1):31-46.

Keywords

multiple endocrine neoplasia type 1
neuroendocrine tumours
pancreatic neoplasms
insulinoma
modifier
genetic background
exome sequencing

Authors

Anna Skalniak
Małgorzata Trofimiuk-Müldner
Agata Jabrocka-Hybel
Justyna Totoń-Żurańska
Paweł Wołkow
Alicja Hubalewska-Dydejczyk

References (89)
  1. Brandi ML, Agarwal SK, Perrier ND, et al. Multiple Endocrine Neoplasia Type 1: Latest Insights. Endocr Rev. 2021; 42(2): 133–170.
  2. Giusti F, Marini F, Brandi ML. Multiple Endocrine Neoplasia Type 1. In: Adam M, Ardinger H, Pagon R. ed. In GeneReviews® [Internet]. University of Washington, Seattle 2005.
  3. Singh G, Mulji NJ, Jialal I. Multiple Endocrine Neoplasia Type 1. In: StatPearls [Internet]. . StatPearls Publishing, Treasure Island 2022.
  4. Lemos MC, Thakker RV. Multiple endocrine neoplasia type 1 (MEN1): analysis of 1336 mutations reported in the first decade following identification of the gene. Hum Mutat. 2008; 29(1): 22–32.
  5. Lips CJ, Dreijerink KM, van der Luijt RB. Multiple Endocrine Neoplasia Type 1 (MEN1). In: Weiss RE, Refetoff S. ed. Genetic Diagnosis of Endocrine Disorders. Elsevier 2010: 261–270.
  6. Marini F, Giusti F, Brandi ML. Multiple endocrine neoplasia type 1: extensive analysis of a large database of Florentine patients. Orphanet J Rare Dis. 2018; 13(1): 205.
  7. Lemos MC, Thakker RV. Multiple endocrine neoplasia type 1 (MEN1): analysis of 1336 mutations reported in the first decade following identification of the gene. Hum Mutat. 2008; 29(1): 22–32.
  8. Pardi E, Borsari S, Saponaro F, et al. Mutational and large deletion study of genes implicated in hereditary forms of primary hyperparathyroidism and correlation with clinical features. PLoS One. 2017; 12(10): e0186485.
  9. Romanet P, Mohamed A, Giraud S, et al. UMD-MEN1 Database: An Overview of the 370 MEN1 Variants Present in 1676 Patients From the French Population. J Clin Endocrinol Metab. 2019; 104(3): 753–764.
  10. Palermo A, Capoluongo E, Del Toro R, et al. A novel germline mutation at exon 10 of MEN1 gene: a clinical survey and positive genotype-phenotype analysis of a MEN1 Italian family, including monozygotic twins. Hormones (Athens). 2018; 17(3): 427–435.
  11. Christakis I, Qiu W, Hyde SM, et al. Genotype-phenotype pancreatic neuroendocrine tumor relationship in multiple endocrine neoplasia type 1 patients: A 23-year experience at a single institution. Surgery. 2018; 163(1): 212–217.
  12. Stasiak M, Dedecjus M, Zawadzka-Starczewska K, et al. Novel Germline c.105_107dupGCT Mutation in a Family with Newly Diagnosed Multiple Endocrine Neoplasia Type 1. Genes (Basel). 2020; 11(9).
  13. Welsch C, Flügel AK, Rondot S, et al. Distinct clinical phenotypes in a family with a novel truncating MEN1 frameshift mutation. BMC Endocr Disord. 2022; 22(1): 64.
  14. Gilis-Januszewska A, Bogusławska A, Hasse-Lazar K, et al. Heterogeneity of the Clinical Presentation of the MEN1 LRG_509 c.781C>T (p.Leu261Phe) Variant Within a Three-Generation Family. Genes (Basel). 2021; 12(4).
  15. Burgess JR, Shepherd JJ, Parameswaran V, et al. Prolactinomas in a large kindred with multiple endocrine neoplasia type 1: clinical features and inheritance pattern. J Clin Endocrinol Metab. 1996; 81(5): 1841–1845.
  16. Thevenon J, Bourredjem A, Faivre L, et al. Unraveling the intrafamilial correlations and heritability of tumor types in MEN1: a Groupe d'étude des Tumeurs Endocrines study. Eur J Endocrinol. 2015; 173(6): 819–826.
  17. Nadeau JH. Modifier genes and protective alleles in humans and mice. Curr Opin Genet Dev. 2003; 13(3): 290–295.
  18. McCabe ERB. Modifier genes: Moving from pathogenesis to therapy. Mol Genet Metab. 2017; 122(1-2): 1–3.
  19. Grillo E, Lo Rizzo C, Bianciardi L, et al. Revealing the complexity of a monogenic disease: rett syndrome exome sequencing. PLoS One. 2013; 8(2): e56599.
  20. Andrews S. FastQC: A Quality Control Tool for High Throughput Sequence Data. 2010. Babraham bioinformatics 2010, http://www.bioinformatics.babraham.ac.uk/projects/.
  21. Li H, Durbin R. Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics. 2010; 26(5): 589–595.
  22. Li H, Handsaker B, Wysoker A, et al. 1000 Genome Project Data Processing Subgroup. The Sequence Alignment/Map format and SAMtools. Bioinformatics. 2009; 25(16): 2078–2079.
  23. Mi H, Ebert D, Muruganujan A, et al. PANTHER version 16: a revised family classification, tree-based classification tool, enhancer regions and extensive API. Nucleic Acids Res. 2021; 49(D1): D394–D403.
  24. Gene Ontology Consortium. The Gene Ontology resource: enriching a GOld mine. Nucleic Acids Res. 2021; 49(D1): D325–D334.
  25. Ashburner M, Ball CA, Blake JA, et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 2000; 25(1): 25–29.
  26. Szklarczyk D, Gable AL, Lyon D, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019; 47(D1): D607–D613.
  27. Karczewski KJ, Francioli LC, Tiao G, et al. Genome Aggregation Database Consortium, Genome Aggregation Database Consortium, Genome Aggregation Database Consortium. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature. 2020; 581(7809): 434–443.
  28. Domingo-Fernández D, Hoyt CT, Bobis-Álvarez C, et al. ComPath: an ecosystem for exploring, analyzing, and curating mappings across pathway databases. NPJ Syst Biol Appl. 2019; 5: 3.
  29. Pratt D, Chen J, Welker D, et al. NDEx, the Network Data Exchange. Cell Syst. 2015; 1(4): 302–305.
  30. Pillich RT, Chen J, Rynkov V, et al. NDEx: A Community Resource for Sharing and Publishing of Biological Networks. Methods Mol Biol. 2017; 1558: 271–301.
  31. Rahit KM, Tarailo-Graovac M. Genetic Modifiers and Rare Mendelian Disease. Genes (Basel). 2020; 11(3).
  32. Scarpa A, Chang DK, Nones K, et al. Australian Pancreatic Cancer Genome Initiative, Australian Pancreatic Cancer Genome Initiative. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature. 2017; 543(7643): 65–71.
  33. Jiao Y, Shi C, Edil BH, et al. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science. 2011; 331(6021): 1199–1203.
  34. Cartwright S, Karakesisoglou I. Nesprins in health and disease. Semin Cell Dev Biol. 2014; 29: 169–179.
  35. Seenappa V, Joshi MB, Satyamoorthy K. Intricate Regulation of Phosphoenolpyruvate Carboxykinase (PEPCK) Isoforms in Normal Physiology and Disease. Curr Mol Med. 2019; 19(4): 247–272.
  36. Dibble CC, Cantley LC. Regulation of mTORC1 by PI3K signaling. Trends Cell Biol. 2015; 25(9): 545–555.
  37. Koyanagi M, Asahara Si, Matsuda T, et al. Ablation of TSC2 enhances insulin secretion by increasing the number of mitochondria through activation of mTORC1. PLoS One. 2011; 6(8): e23238.
  38. White MF, Yenush L. The IRS-signaling system: a network of docking proteins that mediate insulin and cytokine action. Curr Top Microbiol Immunol. 1998; 228: 179–208.
  39. Pruett W, Yuan Y, Rose E, et al. Association between GRB2/Sos and insulin receptor substrate 1 is not sufficient for activation of extracellular signal-regulated kinases by interleukin-4: implications for Ras activation by insulin. Mol Cell Biol. 1995; 15(3): 1778–1785.
  40. Safran M, Rosen N, Twik M. The GeneCards Suite. Practical Guide to Life Science Databases. Springer , Singapore 2021: 27–56.
  41. Orlando RA, Rader K, Authier F, et al. Megalin is an endocytic receptor for insulin. J Am Soc Nephrol. 1998; 9(10): 1759–1766.
  42. Seo JiA, Kang MC, Yang WM, et al. Apolipoprotein J is a hepatokine regulating muscle glucose metabolism and insulin sensitivity. Nat Commun. 2020; 11(1): 2024.
  43. Bryniarski MA, Yee BM, Jaffri I, et al. Increased megalin expression in early type 2 diabetes: role of insulin-signaling pathways. Am J Physiol Renal Physiol. 2018; 315(5): F1191–F1207.
  44. Hosojima M, Sato H, Yamamoto K, et al. Regulation of megalin expression in cultured proximal tubule cells by angiotensin II type 1A receptor- and insulin-mediated signaling cross talk. Endocrinology. 2009; 150(2): 871–878.
  45. Williams AS, Kang Li, Wasserman DH. The extracellular matrix and insulin resistance. Trends Endocrinol Metab. 2015; 26(7): 357–366.
  46. Shiokawa M, Kodama Y, Sekiguchi K, et al. Laminin 511 is a target antigen in autoimmune pancreatitis. Sci Transl Med. 2018; 10(453).
  47. Missiaglia E, Dalai I, Barbi S, et al. Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. J Clin Oncol. 2010; 28(2): 245–255.
  48. Chan CS, Laddha SV, Lewis PW, et al. ATRX, DAXX or MEN1 mutant pancreatic neuroendocrine tumors are a distinct alpha-cell signature subgroup. Nat Commun. 2018; 9(1): 4158.
  49. Ciobanu OA, Martin SC, Herlea V, et al. Insights into Epigenetic Changes Related to Genetic Variants and Cells-of-Origin of Pancreatic Neuroendocrine Tumors: An Algorithm for Practical Workup. Cancers (Basel). 2022; 14(18).
  50. Park JK, Paik WH, Lee K, et al. DAXX/ATRX and MEN1 genes are strong prognostic markers in pancreatic neuroendocrine tumors. Oncotarget. 2017; 8(30): 49796–49806.
  51. Sun C, Estrella JS, Whitley EM, et al. Context matters - Daxx and Atrx are not robust tumor suppressors in the murine endocrine pancreas. Dis Model Mech. 2022; 15(8).
  52. Borbás T, Benko B, Dalmadi B, et al. Insulin in flavin-containing monooxygenase regulation. Flavin-containing monooxygenase and cytochrome P450 activities in experimental diabetes. Eur J Pharm Sci. 2006; 28(1-2): 51–58.
  53. Shih DM, Wang Z, Lee R, et al. Flavin containing monooxygenase 3 exerts broad effects on glucose and lipid metabolism and atherosclerosis. J Lipid Res. 2015; 56(1): 22–37.
  54. Scott F, Gonzalez Malagon SG, O'Brien BA, et al. Identification of Flavin-Containing Monooxygenase 5 (FMO5) as a Regulator of Glucose Homeostasis and a Potential Sensor of Gut Bacteria. Drug Metab Dispos. 2017; 45(9): 982–989.
  55. Schugar RC, Brown JM. Emerging roles of flavin monooxygenase 3 in cholesterol metabolism and atherosclerosis. Curr Opin Lipidol. 2015; 26(5): 426–431.
  56. Rodriguez-Diaz R, Dando R, Jacques-Silva MC, et al. Alpha cells secrete acetylcholine as a non-neuronal paracrine signal priming beta cell function in humans. Nat Med. 2011; 17(7): 888–892.
  57. Capaldo B, Napoli R, Di Marino L, et al. Epinephrine directly antagonizes insulin-mediated activation of glucose uptake and inhibition of free fatty acid release in forearm tissues. Metabolism. 1992; 41(10): 1146–1149.
  58. Burr IM, Slonim AE, Sharp R. Interactions of acetylcholine and epinephrine on the dynamics of insulin release in vitro. J Clin Invest. 1976; 58(1): 230–239.
  59. Arnold AC, Garland EM, Celedonio JE, et al. Hyperinsulinemia and Insulin Resistance in Dopamine β-Hydroxylase Deficiency. J Clin Endocrinol Metab. 2017; 102(1): 10–14.
  60. Gao N, Le Lay J, Qin W, et al. Foxa1 and Foxa2 maintain the metabolic and secretory features of the mature beta-cell. Mol Endocrinol. 2010; 24(8): 1594–1604.
  61. Wolfrum C, Besser D, Luca E, et al. Insulin regulates the activity of forkhead transcription factor Hnf-3beta/Foxa-2 by Akt-mediated phosphorylation and nuclear/cytosolic localization. Proc Natl Acad Sci U S A. 2003; 100(20): 11624–11629.
  62. Karakose E, Wang H, Inabnet W, et al. Aberrant methylation underlies insulin gene expression in human insulinoma. Nat Commun. 2020; 11(1): 5210.
  63. Wang H, Bender A, Wang P, et al. Insights into beta cell regeneration for diabetes via integration of molecular landscapes in human insulinomas. Nat Commun. 2017; 8(1): 767.
  64. Ramasamy I. Recent advances in physiological lipoprotein metabolism. Clin Chem Lab Med. 2014; 52(12): 1695–1727.
  65. Zanoni P, Khetarpal SA, Larach DB, et al. CHD Exome+ Consortium, CARDIoGRAM Exome Consortium, Global Lipids Genetics Consortium. Rare variant in scavenger receptor BI raises HDL cholesterol and increases risk of coronary heart disease. Science. 2016; 351(6278): 1166–1171.
  66. Gonzales JC, Gordts PL, Foley EM, et al. Apolipoproteins E and AV mediate lipoprotein clearance by hepatic proteoglycans. J Clin Invest. 2013; 123(6): 2742–2751.
  67. Kockx M, Traini M, Kritharides L. Cell-specific production, secretion, and function of apolipoprotein E. J Mol Med (Berl). 2018; 96(5): 361–371.
  68. Cruciani-Guglielmacci C, Meneyrol K, Denom J, et al. Homocysteine Metabolism Pathway Is Involved in the Control of Glucose Homeostasis: A Cystathionine Beta Synthase Deficiency Study in Mouse. Cells. 2022; 11(11).
  69. Li Y, Zhang H, Jiang C, et al. Hyperhomocysteinemia promotes insulin resistance by inducing endoplasmic reticulum stress in adipose tissue. J Biol Chem. 2013; 288(14): 9583–9592.
  70. Di Virgilio F. Purines, purinergic receptors, and cancer. Cancer Res. 2012; 72(21): 5441–5447.
  71. Hamze Z, Vercherat C, Bernigaud-Lacheretz A, et al. Altered MENIN expression disrupts the MAFA differentiation pathway in insulinoma. Endocr Relat Cancer. 2013; 20(6): 833–848.
  72. Henfling M, Perren A, Schmitt AM, et al. The IGF pathway is activated in insulinomas but downregulated in metastatic disease. Endocr Relat Cancer. 2018 [Epub ahead of print].
  73. Zhou W, Gong Li, Li X, et al. Screening key candidate genes and pathways involved in insulinoma by microarray analysis. Medicine (Baltimore). 2018; 97(22): e10826.
  74. Lemos MC, Harding B, Reed AAC, et al. Genetic background influences embryonic lethality and the occurrence of neural tube defects in Men1 null mice: relevance to genetic modifiers. J Endocrinol. 2009; 203(1): 133–142.
  75. Lines KE, Javid M, Reed AAC, et al. Genetic background influences tumour development in heterozygous Men1 knockout mice. Endocr Connect. 2020; 9(5): 426–437.
  76. Mohr H, Pellegata NS. Animal models of MEN1. Endocr Relat Cancer. 2017; 24(10): T161–T177.
  77. Brayton C. Spontaneous Diseases in Commonly Used Mouse Strains. In: The Mouse in Biomedical Research. Elsevier 2007: 623–717.
  78. Longuini VC, Lourenço DM, Sekiya T, et al. Association between the p27 rs2066827 variant and tumor multiplicity in patients harboring MEN1 germline mutations. Eur J Endocrinol. 2014; 171(3): 335–342.
  79. Circelli L, Ramundo V, Marotta V, et al. Multidisciplinary Group for NeuroEndocrine Tumours of Naples. Prognostic role of the CDNK1B V109G polymorphism in multiple endocrine neoplasia type 1. J Cell Mol Med. 2015; 19(7): 1735–1741.
  80. Tirosh A, Kebebew E. Genetic and epigenetic alterations in pancreatic neuroendocrine tumors. J Gastrointest Oncol. 2020; 11(3): 567–577.
  81. Soczomski P, Jurecka-Lubieniecka B, Rogozik N, et al. Multiple endocrine neoplasia type 1 in Poland: a two-centre experience. Endokrynol Pol. 2019; 70(5): 385–391.
  82. Cheng X, Yin H, Fu J, et al. Aggregate analysis based on TCGA: TTN missense mutation correlates with favorable prognosis in lung squamous cell carcinoma. J Cancer Res Clin Oncol. 2019; 145(4): 1027–1035.
  83. Parry ML, Ramsamooj M, Blanck G. Big genes are big mutagen targets: a connection to cancerous, spherical cells? Cancer Lett. 2015; 356(2 Pt B): 479–482.
  84. Parry ML, Blanck G. Flat cells come full sphere: Are mutant cytoskeletal-related proteins oncoprotein-monsters or useful immunogens? Hum Vaccin Immunother. 2016; 12(1): 120–123.
  85. Xiao H, Huang W, Li Y, et al. Targeting Long Non-Coding RNA TTN-AS1 Suppresses Bladder Cancer Progression. Front Genet. 2021; 12: 704712.
  86. Wang Y, Li D, Lu J, et al. Long noncoding RNA TTN-AS1 facilitates tumorigenesis and metastasis by maintaining TTN expression in skin cutaneous melanoma. Cell Death Dis. 2020; 11(8): 664.
  87. Fang J, Huang C, Ke J, et al. lncRNA TTN-AS1 facilitates proliferation, invasion, and epithelial-mesenchymal transition of breast cancer cells by regulating miR-139-5p/ZEB1 axis. J Cell Biochem. 2020 [Epub ahead of print].
  88. Cui Z, Han B, Wang X, et al. Long Non-Coding RNA TTN-AS1 Promotes the Proliferation and Invasion of Colorectal Cancer Cells by Activating miR-497-Mediated PI3K/Akt/mTOR Signaling. Onco Targets Ther. 2019; 12: 11531–11539.
  89. Zheng QX, Wang J, Gu XY, et al. TTN-AS1 as a potential diagnostic and prognostic biomarker for multiple cancers. Biomed Pharmacother. 2021; 135: 111169.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

Via MedicaWydawcą jest  VM Media Group sp. z o.o., Grupa Via Medica, ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail:  viamedica@viamedica.pl