#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

PARP-1 Regulates Metastatic Melanoma through Modulation of Vimentin-induced Malignant Transformation


PARP inhibition can induce anti-neoplastic effects when used as monotherapy or in combination with chemo- or radiotherapy in various tumor settings; however, the basis for the anti-metastasic activities resulting from PARP inhibition remains unknown. PARP inhibitors may also act as modulators of tumor angiogenesis. Proteomic analysis of endothelial cells revealed that vimentin, an intermediary filament involved in angiogenesis and a specific hallmark of EndoMT (endothelial to mesenchymal transition) transformation, was down-regulated following loss of PARP-1 function in endothelial cells. VE-cadherin, an endothelial marker of vascular normalization, was up-regulated in HUVEC treated with PARP inhibitors or following PARP-1 silencing; vimentin over-expression was sufficient to drive to an EndoMT phenotype. In melanoma cells, PARP inhibition reduced pro-metastatic markers, including vasculogenic mimicry. We also demonstrated that vimentin expression was sufficient to induce increased mesenchymal/pro-metastasic phenotypic changes in melanoma cells, including ILK/GSK3-β-dependent E-cadherin down-regulation, Snail1 activation and increased cell motility and migration. In a murine model of metastatic melanoma, PARP inhibition counteracted the ability of melanoma cells to metastasize to the lung. These results suggest that inhibition of PARP interferes with key metastasis-promoting processes, leading to suppression of invasion and colonization of distal organs by aggressive metastatic cells.


Vyšlo v časopise: PARP-1 Regulates Metastatic Melanoma through Modulation of Vimentin-induced Malignant Transformation. PLoS Genet 9(6): e32767. doi:10.1371/journal.pgen.1003531
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003531

Souhrn

PARP inhibition can induce anti-neoplastic effects when used as monotherapy or in combination with chemo- or radiotherapy in various tumor settings; however, the basis for the anti-metastasic activities resulting from PARP inhibition remains unknown. PARP inhibitors may also act as modulators of tumor angiogenesis. Proteomic analysis of endothelial cells revealed that vimentin, an intermediary filament involved in angiogenesis and a specific hallmark of EndoMT (endothelial to mesenchymal transition) transformation, was down-regulated following loss of PARP-1 function in endothelial cells. VE-cadherin, an endothelial marker of vascular normalization, was up-regulated in HUVEC treated with PARP inhibitors or following PARP-1 silencing; vimentin over-expression was sufficient to drive to an EndoMT phenotype. In melanoma cells, PARP inhibition reduced pro-metastatic markers, including vasculogenic mimicry. We also demonstrated that vimentin expression was sufficient to induce increased mesenchymal/pro-metastasic phenotypic changes in melanoma cells, including ILK/GSK3-β-dependent E-cadherin down-regulation, Snail1 activation and increased cell motility and migration. In a murine model of metastatic melanoma, PARP inhibition counteracted the ability of melanoma cells to metastasize to the lung. These results suggest that inhibition of PARP interferes with key metastasis-promoting processes, leading to suppression of invasion and colonization of distal organs by aggressive metastatic cells.


Zdroje

1. BaumB, SettlemanJ, QuinlanMP (2008) Transitions between epithelial and mesenchymal states in development and disease. Semin Cell Dev Biol 19: 294–308.

2. CarmelietP, JainRK (2000) Angiogenesis in cancer and other diseases. Nature 407: 249–257.

3. VestweberD, WinderlichM, CagnaG, NottebaumAF (2009) Cell adhesion dynamics at endothelial junctions: VE-cadherin as a major player. Trends Cell Biol 19: 8–15.

4. RadiskyDC (2005) Epithelial-mesenchymal transition. J Cell Sci 118: 4325–4326.

5. HugoH, AcklandML, BlickT, LawrenceMG, ClementsJA, et al. (2007) Epithelial–mesenchymal and mesenchymal–epithelial transitions in carcinoma progression. J Cell Physiol 213: 374–383.

6. ThieryJP, SleemanJP (2006) Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol 7: 131–142.

7. SchreiberV, DantzerF, AmeJC, de MurciaG (2006) Poly(ADP-ribose): novel functions for an old molecule. Nat Rev Mol Cell Biol 7: 517–528.

8. BryantHE, SchultzN, ThomasHD, ParkerKM, FlowerD, et al. (2005) Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 434: 913–917.

9. FarmerH, McCabeN, LordCJ, TuttAN, JohnsonDA, et al. (2005) Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434: 917–921.

10. McCabeN, TurnerNC, LordCJ, KluzekK, BialkowskaA, et al. (2006) Deficiency in the repair of DNA damage by homologous recombination and sensitivity to poly(ADP-ribose) polymerase inhibition. Cancer Res 66: 8109–8115.

11. FongPC, BossDS, YapTA, TuttA, WuP, et al. (2009) Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med 361: 123–134.

12. Quiles-PerezR, Munoz-GamezJA, Ruiz-ExtremeraA, O'ValleF, Sanjuan-NunezL, et al. (2010) Inhibition of poly adenosine diphosphate-ribose polymerase decreases hepatocellular carcinoma growth by modulation of tumor-related gene expression. Hepatology 51: 255–266.

13. Martin-OlivaD, Aguilar-QuesadaR, O'ValleF, Munoz-GamezJA, Martinez-RomeroR, et al. (2006) Inhibition of poly(ADP-ribose) polymerase modulates tumor-related gene expression, including hypoxia-inducible factor-1 activation, during skin carcinogenesis. Cancer Res 66: 5744–5756.

14. LacalPM, TentoriL, MuziA, RuffiniF, DorioAS, et al. (2009) Pharmacological inhibition of poly(ADP-ribose) polymerase activity down-regulates the expression of syndecan-4 and Id-1 in endothelial cells. Int J Oncol 34: 861–872.

15. PyriochouA, OlahG, DeitchEA, SzaboC, PapapetropoulosA (2008) Inhibition of angiogenesis by the poly(ADP-ribose) polymerase inhibitor PJ-34. Int J Mol Med 22: 113–118.

16. RodriguezMI, Gonzalez-FloresA, DantzerF, CollardJ, de HerrerosAG, et al. (2011) Poly(ADP-ribose)-dependent regulation of Snail1 protein stability. Oncogene 30: 4365–4372.

17. TentoriL, MuziA, DorioAS, BultriniS, MazzonE, et al. (2008) Stable depletion of poly (ADP-ribose) polymerase-1 reduces in vivo melanoma growth and increases chemosensitivity. Eur J Cancer 44: 1302–1314.

18. Bargagna-MohanP, HamzaA, KimYE, Khuan Abby HoY, Mor-VakninN, et al. (2007) The tumor inhibitor and antiangiogenic agent withaferin A targets the intermediate filament protein vimentin. Chem Biol 14: 623–634.

19. GillesC, PoletteM, MestdagtM, Nawrocki-RabyB, RuggeriP, et al. (2003) Transactivation of vimentin by beta-catenin in human breast cancer cells. Cancer Res 63: 2658–2664.

20. KalluriR, ZeisbergM (2006) Fibroblasts in cancer. Nat Rev Cancer 6: 392–401.

21. VuoriluotoK, HaugenH, KiviluotoS, MpindiJP, NevoJ, et al. (2011) Vimentin regulates EMT induction by Slug and oncogenic H-Ras and migration by governing Axl expression in breast cancer. Oncogene 30: 1436–1448.

22. LiY, BhargavaMM, JosephA, JinL, RosenEM, et al. (1994) Effect of hepatocyte growth factor/scatter factor and other growth factors on motility and morphology of non-tumorigenic and tumor cells. In Vitro Cell Dev Biol Anim 30A: 105–110.

23. HessAR, SeftorEA, GrumanLM, KinchMS, SeftorRE, et al. (2006) VE-cadherin regulates EphA2 in aggressive melanoma cells through a novel signaling pathway: implications for vasculogenic mimicry. Cancer Biol Ther 5: 228–233.

24. OrsenigoF, GiampietroC, FerrariA, CoradaM, GalaupA, et al. (2012) Phosphorylation of VE-cadherin is modulated by haemodynamic forces and contributes to the regulation of vascular permeability in vivo. Nat Commun 3: 1208.

25. MaedaM, SugaT, TakasukaN, HoshiA, SasakiT (1990) Effect of bis(bilato)-1,2-cyclohexanediammineplatinum(II) complexes on lung metastasis of B16-F10 melanoma cells in mice. Cancer Lett 55: 143–147.

26. KhannaC, HunterK (2005) Modeling metastasis in vivo. Carcinogenesis 26: 513–523.

27. PenaC, GarciaJM, LarribaMJ, BarderasR, GomezI, et al. (2009) SNAI1 expression in colon cancer related with CDH1 and VDR downregulation in normal adjacent tissue. Oncogene 28: 4375–4385.

28. SilyeR, KarayiannakisAJ, SyrigosKN, PooleS, van NoordenS, et al. (1998) E-cadherin/catenin complex in benign and malignant melanocytic lesions. J Pathol 186: 350–355.

29. HsuM, AndlT, LiG, MeinkothJL, HerlynM (2000) Cadherin repertoire determines partner-specific gap junctional communication during melanoma progression. J Cell Sci 113(Pt 9): 1535–1542.

30. IvaskaJ, PallariHM, NevoJ, ErikssonJE (2007) Novel functions of vimentin in cell adhesion, migration, and signaling. Exp Cell Res 313: 2050–2062.

31. van BeijnumJR, DingsRP, van der LindenE, ZwaansBM, RamaekersFC, et al. (2006) Gene expression of tumor angiogenesis dissected: specific targeting of colon cancer angiogenic vasculature. Blood 108: 2339–2348.

32. MinC, EddySF, SherrDH, SonensheinGE (2008) NF-kappaB and epithelial to mesenchymal transition of cancer. J Cell Biochem 104: 733–744.

33. WuY, ZhangX, SalmonM, LinX, ZehnerZE (2007) TGFbeta1 regulation of vimentin gene expression during differentiation of the C2C12 skeletal myogenic cell line requires Smads, AP-1 and Sp1 family members. Biochim Biophys Acta 1773: 427–439.

34. LonnP, van der HeideLP, DahlM, HellmanU, HeldinCH, et al. (2010) PARP-1 attenuates Smad-mediated transcription. Mol Cell 40: 521–532.

35. BertaudJ, QinZ, BuehlerMJ (2010) Intermediate filament-deficient cells are mechanically softer at large deformation: a multi-scale simulation study. Acta Biomater 6: 2457–2466.

36. TapodiA, DebreceniB, HantoK, BognarZ, WittmannI, et al. (2005) Pivotal role of Akt activation in mitochondrial protection and cell survival by poly(ADP-ribose)polymerase-1 inhibition in oxidative stress. J Biol Chem 280: 35767–35775.

37. KrengelS, GroteluschenF, BartschS, TronnierM (2004) Cadherin expression pattern in melanocytic tumors more likely depends on the melanocyte environment than on tumor cell progression. J Cutan Pathol 31: 1–7.

38. LichtAH, Muller-HoltkampF, FlammeI, BreierG (2006) Inhibition of hypoxia-inducible factor activity in endothelial cells disrupts embryonic cardiovascular development. Blood 107: 584–590.

39. WeisSM, ChereshDA (2011) Tumor angiogenesis: molecular pathways and therapeutic targets. Nat Med 17: 1359–1370.

40. FidlerIJ (1973) Selection of successive tumour lines for metastasis. Nat New Biol 242: 148–149.

41. MelnikovaVO, BolshakovSV, WalkerC, AnanthaswamyHN (2004) Genomic alterations in spontaneous and carcinogen-induced murine melanoma cell lines. Oncogene 23: 2347–2356.

42. ChevanneM, ZampieriM, CaldiniR, RizzoA, CiccaroneF, et al. (2010) Inhibition of PARP activity by PJ-34 leads to growth impairment and cell death associated with aberrant mitotic pattern and nucleolar actin accumulation in M14 melanoma cell line. J Cell Physiol 222: 401–410.

43. BarberaMJ, PuigI, DominguezD, Julien-GrilleS, Guaita-EsteruelasS, et al. (2004) Regulation of Snail transcription during epithelial to mesenchymal transition of tumor cells. Oncogene 23: 7345–7354.

44. SinghSK, HawkinsC, ClarkeID, SquireJA, BayaniJ, et al. (2004) Identification of human brain tumour initiating cells. Nature 432: 396–401.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 6
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#