期刊论文详细信息
Vascular Cell
Eribulin mesylate exerts specific gene expression changes in pericytes and shortens pericyte-driven capillary network in vitro
Yasuhiro Funahashi3  Yoshiya Oda3  Jesse Chow3  Junji Matsui1  Judith Oestreicher2  Noel Taylor3  Satoshi Kawano1  Sergei I Agoulnik3 
[1] Eisai Co., Ltd, Tokodai 5-1-3, Tsukuba, Ibaraki 300-2635, Japan;Present address: Infinity Pharmaceuticals, 780 Memorial Drive, Cambridge, MA 02139, USA;Eisai Inc, 4 Corporate Drive, Andover, MA 01810, USA
关键词: Angiogenesis;    Co-culture assay;    Gene expression profiling;    Pericytes;    Endothelial cells;    Eribulin;   
Others  :  801486
DOI  :  10.1186/2045-824X-6-3
 received in 2013-09-21, accepted in 2014-02-24,  发布年份 2014
PDF
【 摘 要 】

Background

Eribulin mesylate is a synthetic macrocyclic ketone analog of the marine sponge natural product halichondrin B. Eribulin is a tubulin-binding drug and approved in many countries worldwide for treatment of certain patients with advanced breast cancer. Here we investigated antiproliferative and antiangiogenic effects of eribulin on vascular cells, human umbilical vein endothelial cells (HUVECs) and human brain vascular pericytes (HBVPs), in vitro in comparison with another tubulin-binding drug, paclitaxel.

Methods

HUVECs and HBVPs were treated with either eribulin or paclitaxel and their antiproliferative effects were evaluated. Global gene expression profiling changes caused by drug treatments were studied using Affymetrix microarray platform and custom TaqMan Low Density Cards. To examine effects of the drugs on pericyte-driven in vitro angiogenesis, we compared lengths of capillary networks in co-cultures of HUVECs with HBVPs.

Results

Both eribulin and paclitaxel showed potent activities in in vitro proliferation of HUVECs and HBVPs, with the half-maximal inhibitory concentrations (IC50) in low- to sub-nmol/L concentrations. When gene expression changes were assessed in HUVECs, the majority of affected genes overlapped for both treatments (59%), while in HBVPs, altered gene signatures were drug-dependent and the overlap was limited to just 12%. In HBVPs, eribulin selectively affected 11 pathways (p < 0.01) such as Cell Cycle Control of Chromosomal Replication. In contrast, paclitaxel was tended to regulate 27 pathways such as PI3K/AKT. Only 5 pathways were commonly affected by both treatments. In in vitro pericyte-driven angiogenesis model, paclitaxel showed limited activity while eribulin shortened the formed capillary networks of HUVECs driven by HBVPs at low nmol/L concentrations starting at day 3 after treatments.

Conclusions

Our findings suggest that pericytes, but not endothelial cells, responded differently, to two mechanistically-distinct tubulin-binding drugs, eribulin and paclitaxel. While eribulin and paclitaxel induced similar changes in gene expression in endothelial cells, in pericytes their altered gene expression was unique and drug-specific. In the functional endothelial-pericyte co-culture assay, eribulin, but not paclitaxel showed strong efficacy not only as a cytotoxic drug but also as a potent antivascular agent that affected pericyte-driven in vitro angiogenesis.

【 授权许可】

   
2014 Agoulnik et al.; licensee BioMed Central Ltd.

【 预 览 】
附件列表
Files Size Format View
20140708010342597.pdf 1059KB PDF download
Figure 4. 20KB Image download
Figure 3. 75KB Image download
Figure 2. 69KB Image download
Figure 1. 46KB Image download
【 图 表 】

Figure 1.

Figure 2.

Figure 3.

Figure 4.

【 参考文献 】
  • [1]Gerhardt H, Betsholtz C: Endothelial-pericyte interactions in angiogenesis. Cell Tissue Res 2003, 314:15-23.
  • [2]Dulmovits BM, Herman IM: Microvascular remodeling and wound healing: a role for pericytes. Int J Biochem Cell Biol 2012, 44(11):1800-1812.
  • [3]Ding Y, Song N, Luo Y: Role of bone marrow-derived cells in angiogenesis: focus on macrophages and pericytes. Cancer Microenviron 2012, 5(3):225-236.
  • [4]Darland DC, Massingham LJ, Smith SR, Piek E, Saint-Geniez M, D’Amore PA: Pericyte production of cell-associated VEGF is differentiation-dependent and is associated with endothelial survival. Dev Biol 2003, 264(1):275-288.
  • [5]Guo P, Hu B, Gu W, Xu L, Wang D, Huang HJ, Cavenee WK, Cheng SY: Platelet-derived growth factor-B enhances glioma angiogenesis by stimulating vascular endothelial growth factor expression in tumor endothelia and by promoting pericyte recruitment. Am J Pathol 2003, 162(4):1083-1093.
  • [6]Metheny-Barlow LJ, Li LY: The enigmatic role of angiopoietin-1 in tumor angiogenesis. Cell Res 2003, 13(5):309-317.
  • [7]Saharinen P, Bry M, Alitalo K: How do angiopoietins Tie in with vascular endothelial growth factors? Curr Opin Hematol 2010, 17(3):198-205.
  • [8]Franco M, Roswall P, Cortez E, Hanahan D, Pietras K: Pericytes promote endothelial cell survival through induction of autocrine VEGF-A signaling and Bcl-w expression. Blood 2011, 118:2906-2917.
  • [9]Rapisarda A, Melillo G: Role of the VEGF/VEGFR axis in cancer biology and therapy. Adv Cancer Res 2012, 114:237-267.
  • [10]Weisshardt P, Trarbach T, Dürig J, Paul A, Reis H, Tilki D, Miroschnik I, Ergün S, Klein D: Tumor vessel stabilization and remodeling by anti-angiogenic therapy with bevacizumab. Histochem Cell Biol 2012, 137(3):391-401.
  • [11]Carmeliet P, Jain RK: Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat Rev Drug Discov 2011, 10(6):417-427.
  • [12]Goel S, Duda DG, Xu L, Munn LL, Boucher Y, Fukumura D, Jain RK: Normalization of the vasculature for treatment of cancer and other diseases. Physiol Rev 2011, 91(3):1071-1121.
  • [13]Bottsford-Miller JN, Coleman RL, Sood AK: Resistance and escape from antiangiogenesis therapy: clinical implications and future strategies. J Clin Oncol 2012, 30(32):4026-4034.
  • [14]Spear MA, LoRusso P, Mita A, Mita M: Vascular disrupting agents (VDA) in oncology: advancing towards new therapeutic paradigms in the clinic. Curr Drug Targets 2011, 12(14):2009-2015.
  • [15]McKeage MJ, Baguley BC: Disrupting established tumor blood vessels: an emerging therapeutic strategy for cancer. Cancer 2010, 116(8):1859-1871.
  • [16]Strieth S, Eichhorn ME, Sauer B, Schulze B, Teifel M, Michaelis U, Dellian M: Neovascular targeting chemotherapy: encapsulation of paclitaxel in cationic liposomes impairs functional tumor microvasculature. Int J Cancer 2004, 110(1):117-124.
  • [17]Tozer GM, Kanthou C, Baguley BC: Disrupting tumour blood vessels. Nat Rev Cancer 2005, 5(6):423-435.
  • [18]Towle MJ, Salvato KA, Budrow J, Wels BF, Kuznetsov G, Aalfs KK, Welsh S, Zheng W, Seletsky BM, Palme MH, Habgood GJ, Singer LA, Dipietro LV, Wang Y, Chen JJ, Quincy DA, Davis A, Yoshimatsu K, Kishi Y, Yu MJ, Littlefield BA: In vitro and in vivo anticancer activities of synthetic macrocyclic ketone analogues of halichondrin B. Cancer Res 2001, 61(3):1013-1021.
  • [19]Jordan MA, Kamath K, Manna T, Okouneva T, Miller HP, Davis C, Littlefield BA, Wilson L: The primary antimitotic mechanism of action of the synthetic halichondrin E7389 is suppression of microtubule growth. Mol Cancer Ther 2005, 4(7):1086-1095.
  • [20]Kuznetsov G, Towle MJ, Cheng H, Kawamura T, TenDyke K, Liu D, Kishi Y, Yu MJ, Littlefield BA: Induction of morphological and biochemical apoptosis following prolonged mitotic blockage by halichondrin B macrocyclic ketone analog E7389. Cancer Res 2004, 64(16):5760-5766.
  • [21]Towle MJ, Salvato KA, Wels BF, Aalfs KK, Zheng W, Seletsky BM, Zhu X, Lewis BM, Kishi Y, Yu MJ, Littlefield BA: Eribulin induces irreversible mitotic blockade: implications of cell-based pharmacodynamics for in vivo efficacy under intermittent dosing conditions. Cancer Res 2011, 71(2):496-505.
  • [22]Smith JA, Wilson L, Azarenko O, Zhu X, Lewis BM, Littlefield BA, Jordan MA: Eribulin binds at microtubule ends to a single site on tubulin to suppress dynamic instability. Biochemistry 2010, 49(6):1331-1337.
  • [23]Matsui J, Wakabayashi T, Asada M, Yoshimatsu K, Okada M: Stem cell factor/c-kit signaling promotes the survival, migration, and capillary tube formation of human umbilical vein endothelial cells. J Biol Chem 2004, 279(18):18600-18607.
  • [24]Ishiyama M, Tominaga H, Shiga M, Sasamoto K, Ohkura Y, Ueno K: A combined assay of cell viability and in vitro cytotoxicity with a highly water-soluble tetrazolium salt, neutral red and crystal violet. Biol Pharm Bull 1996, 19(11):1518-20.
  • [25]Folkman J: Tumor angiogenesis: therapeutic implications. N Engl J Med 1971, 285:1182-1186.
  • [26]Hammes HP, Lin J, Renner O, Shani M, Lundqvist A, Betsholtz C, Brownlee M, Deutsch U: Pericytes and the pathogenesis of diabetic retinopathy. Diabetes 2002, 51:3107-3112.
  • [27]Collett GDM, Canfield AE: Angiogenesis and pericytes in the initiation of ectopic calcification. Circ Res 2005, 96:930-938.
  • [28]Liu S, Agalliu D, Chuanhui Y, Fisher M: The roles of pericytes in blood–brain function and stroke. Curr Pharm Des 2012, 25:3653-62.
  • [29]Dziewulska D, Lewandowska E: Pericytes as a new target for pathological processes in CADASIL. Neuropathology 2012, 32:515-21.
  • [30]Sitohy B, Nagy JA, Dvorak HF: Anti-VEGF/VEGFR therapy for cancer: reassessing the target. Cancer Res 1909–1914, 2012:72.
  • [31]Kowalski A, Pałyga J: Linker histone subtypes and their allelic variants. Cell Biol Int 2012, 36(11):981-996.
  • [32]Li JL, Harris AL: Notch signaling from tumor cells: a new mechanism of angiogenesis. Cancer Cell 2005, 8(1):1-3.
  • [33]Wolf FW, Marks RM, Sarma V, Byers MG, Katz RW, Shows TB, Dixit VM: Characterization of a novel tumor necrosis factor-alpha-induced endothelial primary response gene. J Biol Chem 1992, 267(2):1317-1326.
  • [34]Chen MH, Yeh YC, Shyr YM, Jan YH, Chao Y, Li CP, Wang SE, Tzeng CH, Chang PM, Liu CY, Chen MH, Hsiao M, Huang CY: Expression of gremlin 1 correlates with increased angiogenesis and progression-free survival in patients with pancreatic neuroendocrine tumors. J Gastroenterol 2013, 48(1):101-108.
  • [35]Sun L, Ishida T, Okada T, Yasuda T, Hara T, Toh R, Shinohara M, Yamashita T, Rikitake Y, Hirata K: Expression of endothelial lipase correlates with the size of neointima in a murine model of vascular remodeling. J Atheroscler Thromb 2012, 19(12):1110-1127.
  • [36]Christoforou N, Chellappan M, Adler AF, Kirkton RD, Wu T, Addis RC, Bursac N, Leong KW: Transcription factors MYOCD, SRF, Mesp1 and SMARCD3 enhance the cardio-inducing effect of GATA4, TBX5, and MEF2C during direct cellular reprogramming. PLoS One 2013, 8(5):e63577.
  • [37]Joutel A, Corpechot C, Ducros A, Vahedi K, Chabriat H, Mouton P, Alamowitch S, Domenga V, Cécillion M, Marechal E, Maciazek J, Vayssiere C, Cruaud C, Cabanis EA, Ruchoux MM, Weissenbach J, Bach JF, Bousser MG, Tournier-Lasserve E: Notch3 mutations in CADASIL, a hereditary adult-onset condition causing stroke and dementia. Nature 1996, 383:707-710.
  • [38]Meng H, Zhang X, Yu G, Lee SJ, Chen YE, Prudovsky I, Wang MM: Biochemical characterization and cellular effects of CADASIL mutants of NOTCH3. PLoS One 2012, 7(9):e44964.
  • [39]Gridley T: Notch signaling in the vasculature. Curr Top Dev Biol 2010, 92:277-309.
  • [40]Hofmann JJ, Iruela-Arispe ML: Notch signaling in blood vessels: who is talking to whom about what? Circ Res 2007, 100(11):1556-1568.
  文献评价指标  
  下载次数:15次 浏览次数:11次