Botulinum toxin type A prevents the phenotypic transformation of fibroblasts induced by TGF‑β1 via the PTEN/PI3K/Akt signaling pathway

Xue Zhang, Dong Lan, Shuhua Ning, Hongxia Jia, Sisi Yu, Xue Zhang, Dong Lan, Shuhua Ning, Hongxia Jia, Sisi Yu

Abstract

Hypertrophic scar (HS) is a common type of dermatosis. Botulinum toxin type A (BTXA) can exert an anti‑HS effect; however, the regulatory mechanisms underlying this effect remain unclear. Thus, the aim of this study was to examine the effects of BTXA on phosphatase and tensin homolog deleted on chromosome ten (PTEN) expression and the fibroblast phenotypic transformation induced by transforming growth factor (TGF)‑β1, which is an important regulatory factor involved in the process of HS. For this purpose, fibroblasts were treated with various concentrations of BTXA and then treated with 10 ng/ml of TGF‑β1 with gradient concentrations of BTXA. The proliferation and apoptosis of fibroblasts were measured by cell counting kit‑8 assay (CCK‑8) and flow cytometry, respectively. PTEN methylation was analyzed by methylation‑specific PCR (MSP) and DNA methyltransferase (DNMT) activity was determined using a corresponding kit. RT‑qPCR and western blot analysis were performed to detect the transcription and translation levels. The results revealed that BTXA suppressed the proliferation and increased the apoptosis of fibroblasts treated with TGF‑β1 in a dose‑dependent manner. BTXA in combination with TGF‑β1 suppressed the expression of molecules related to the extracellular matrix (ECM), epithelial‑mesenchymal transition (EMT) and apoptosis. BTXA reduced the PTEN methylation level and downregulated the expression levels of methylation‑associated genes. BTXA also inhibited the phosphorylation of phosphoinositide 3‑kinase (PI3K) and Akt. On the whole, the findings of this study indicate that BTXA may inhibit fibroblast phenotypic transformation by regulating PTEN methylation and the phosphorylation of related pathways. The findings of this study can provide a theoretical basis for HS treatment.

Figures

Figure 1
Figure 1
Effect of BTXA on the viability of mouse L929 fibroblasts. (A) The morphological changes of fibroblasts observed under a microscope. (B) The vimentin identification of fibroblasts with Hoechst 33258 was determined by immunofluorescence assay. (C) BTXA inhibited fibroblast viability, which was detected by cell counting kit-8 assay in a dose- (0, 0.125, 0.25, 0.5, 1 and 2 IU/ml) and time- (12, 24 and 48 h) dependent manner. **P<0.01 vs. control. BTXA, botulinum toxin type A.
Figure 2
Figure 2
BTXA suppresses fibroblast viability and promotes apoptosis. (A) BTXA suppressed the high fibroblast viability induced by TGF-β1. (B) BTXA increased the apoptosis of fibroblasts induced by 10 ng/ml of TGF-β1. Dotted line separation represents whether or not fibroblasts were treated with TGF-β1. Data are shown as the means ± SD, n=3. *P<0.05 and **P<0.01 vs. control without TGF-β1; ^P<0.05 and ^^P<0.01 vs. control with TGF-β1. BTXA, botulinum toxin type A; TGF-β1, transforming growth factor-β1.
Figure 3
Figure 3
BTXA prevents extracellular matrix (ECM) over-deposition in fibroblasts. BTXA decreased the high expression levels of collagen I, collagen III and α-SMA induced by 10 ng/ml of TGF-β1 in fibroblasts. The mRNA and protein levels of collagen I, collagen III and α-SMA were detected by (A) RT-qPCR and (B) western blot analysis, respectively. (C) α-SMA expression was determined by immunofluorescence. β-actin was used as an internal control. Dotted line separation represents whether or not fibroblast were treated with TGF-β1. Data were shown as the means ± SD, n=3. *P<0.05 and **P<0.01 vs. control without TGF-β1; ^P<0.05 and ^^P<0.01 vs. control with TGF-β1. BTXA, botulinum toxin type A; TGF-β1, transforming growth factor-β1.
Figure 4
Figure 4
BTXA suppresses the expression levels of MMP-2 and MMP-9. BTXA decreased the high expression levels of MMP-2 and MMP-9 induced by 10 ng/ml of TGF-β1 in fibroblasts. The mRNA and protein levels of MMP-2 and MMP-9 were respectively detected by (A) RT-qPCR and (B) western blot analysis, respectively. β-actin was used as an internal control. Dotted line separation represents whether or not fibroblast were treated with TGF-β1. Data are shown as the means ± SD, n=3. *P<0.05 and **P<0.01 vs. control without TGF-β1; ^P<0.05 and ^^P<0.01 vs. control with TGF-β1. BTXA, botulinum toxin type A; TGF-β1, transforming growth factor-β1; MMP, matrix metalloproteinase.
Figure 5
Figure 5
Effect of BTXA on PTEN and caspase-3 expression, as well as PTEN methylation. (A) Effect of treatment of fibroblasts, which were cultured with or without 10 ng/ml of TGF-β1, with various concentrations (0.25, 0.5 and 1 UI/ml) of BTXA on the mRNA expression of PTEN, as determined by RT-qPCR. (B) Effects of BTXA on the protein levels of PTEN, pro-caspase-3 and cleaved-caspase-3, as detected by western blot analysis. (C) PTEN methylation was measured by methylation-specific PCR (MSP). M, methylated; U, unmethylated; MC, methylated control; UC, unmethylated control. β-actin was used as an internal control. Dotted line separation represents whether or not fibroblast were treated with TGF-β1. Data are shown as the means ± SD, n=3. *P<0.05 and **P<0.01 vs. control without TGF-β1; ^P<0.05 and ^^P<0.01 vs. control with TGF-β1. BTXA, botulinum toxin type A; TGF-β1, transforming growth factor-β1; PTEN, phosphatase and tensin homolog deleted on chromosome ten.
Figure 6
Figure 6
BTXA inhibits the activities of DNMTs. (A) The high DNMT activity induced by 10 ng/ml of TGF-β1 was decreased by BTXA. (B and C) The mRNA and protein levels of DNMT1, DNMT3a and DNMT3b were determined by RT-qPCR and western blot analysis, respectively. β-actin was used as an internal control. Dotted line separation represents whether or not fibroblasts were treated with TGF-β1. Data are shown as the means ± SD, n=3. *P<0.05 and **P<0.01 vs. control without TGF-β1; ^P<0.05 and ^^P<0.01 vs. control with TGF-β1. BTXA, botulinum toxin type A; TGF-β1, transforming growth factor-β1; DNMT, DNA methyltransferase.
Figure 7
Figure 7
BTXA inactivates the phosphorylation of PI3K and Akt. (A) The protein expression of p-PI3K, PI3K, p-Akt and Akt was examined by western blot analysis. (B) BTXA notably suppressed the ratio of p-PI3K/PI3K. (C) BTXA notably suppressed the ratio of p-Akt/Akt. β-actin was used as an internal control. Dotted line separation represents whether or not fibroblasts were treated with TGF-β1. Data are shown as the means ± SD, n=3. *P<0.05 and **P<0.01 vs. control without TGF-β1; ^^P<0.01 vs. control with TGF-β1.

References

    1. Xiao Z, Zhang F, Lin W, Zhang M, Liu Y. Effect of botulinum toxin type A on transforming growth factor beta1 in fibroblasts derived from hypertrophic scar: A preliminary report. Aesthetic Plast Surg. 2010;34:424–427. doi: 10.1007/s00266-009-9423-z.
    1. Butzelaar L, Ulrich MM, Mink van der Molen AB, Niessen FB, Beelen RH. Currently known risk factors for hypertrophic skin scarring: A review. J Plast Reconstr Aesthet Surg. 2016;69:163–169. doi: 10.1016/j.bjps.2015.11.015.
    1. Xue M, Jackson CJ. Extracellular matrix reorganization during wound healing and its impact on abnormal scarring. Adv Wound Care (New Rochelle) 2015;4:119–136. doi: 10.1089/wound.2013.0485.
    1. Liu J, Wang Y, Pan Q, Su Y, Zhang Z, Han J, Zhu X, Tang C, Hu D. Wnt/β-catenin pathway forms a negative feedback loop during TGF- β1 induced human normal skin fibroblast-to-myofibroblast transition. J Dermatol Sci. 2012;65:38–49. doi: 10.1016/j.jdermsci.2011.09.012.
    1. Chun Q, ZhiYong W, Fei S, XiQiao W. Dynamic biological changes in fibroblasts during hypertrophic scar formation and regression. Int Wound J. 2016;13:257–262. doi: 10.1111/iwj.12283.
    1. Sarrazy V, Billet F, Micallef L, Coulomb B, Desmoulière A. Mechanisms of pathological scarring: Role of myofibroblasts and current developments. Wound Repair Regen. 2011;19(Suppl 1):s10–s15. doi: 10.1111/j.1524-475X.2011.00708.x.
    1. Curran TA, Ghahary A. Evidence of a role for fibrocyte and keratinocyte-like cells in the formation of hypertrophic scars. J Burn Care Res. 2013;34:227–231. doi: 10.1097/BCR.0b013e318254d1f9.
    1. Ding J, Ma Z, Shankowsky HA, Medina A, Tredget EE. Deep dermal fibroblast profibrotic characteristics are enhanced by bone marrow-derived mesenchymal stem cells. Wound Repair Regen. 2013;21:448–455. doi: 10.1111/wrr.12046.
    1. Gökçinar-Yagci B, Uçkan-Çetinkaya D, Çelebi-Saltik B. Pericytes: Properties, functions and applications in tissue engineering. Stem Cell Rev. 2015;11:549–559. doi: 10.1007/s12015-015-9590-z.
    1. Yan C, Grimm WA, Garner WL, Qin L, Travis T, Tan N, Han YP. Epithelial to mesenchymal transition in human skin wound healing is induced by tumor necrosis factor-alpha through bone morphogenic protein-2. Am J Pathol. 2010;176:2247–2258. doi: 10.2353/ajpath.2010.090048.
    1. Wang X, Chu J, Wen CJ, Fu SB, Qian YL, Wo Y, Wang C, Wang DR. Functional characterization of TRAP1-like protein involved in modulating fibrotic processes mediated by TGF-β/Smad signaling in hypertrophic scar fibroblasts. Exp Cell Res. 2015;332:202–211. doi: 10.1016/j.yexcr.2015.01.015.
    1. Lu L, Saulis AS, Liu WR, Roy NK, Chao JD, Ledbetter S, Mustoe TA. The temporal effects of anti-TGF-beta1, 2, and 3 monoclonal antibody on wound healing and hypertrophic scar formation. J Am Coll Surg. 2005;201:391–397. doi: 10.1016/j.jamcollsurg.2005.03.032.
    1. Pakyari M, Farrokhi A, Maharlooei MK, Ghahary A. Critical role of transforming growth factor beta in different phases of wound healing. Adv Wound Care (New Rochelle) 2013;2:215–224. doi: 10.1089/wound.2012.0406.
    1. Yin L, Zhao X, Ji S, He C, Wang G, Tang C, Gu S, Yin C. The use of gene activated matrix to mediate effective SMAD2 gene silencing against hypertrophic scar. Biomaterials. 2014;35:2488–2498. doi: 10.1016/j.biomaterials.2013.12.015.
    1. Zhang YF, Zhou SZ, Cheng XY, Yi B, Shan SZ, Wang J, Li QF. Baicalein attenuates hypertrophic scar formation via inhibition of the transforming growth factor-β/Smad2/3 signalling pathway. Br J Dermatol. 2016;174:120–130. doi: 10.1111/bjd.14108.
    1. Bai X, He T, Liu J, Wang Y, Fan L, Tao K, Shi J, Tang C, Su L, Hu D. Loureirin B inhibits fibroblast proliferation and extracellular matrix deposition in hypertrophic scar via TGF-β/Smad pathway. Exp Dermatol. 2015;24:355–360. doi: 10.1111/exd.12665.
    1. Zunwen L, Shizhen Z, Dewu L, Yungui M, Pu N. Effect of tetrandrine on the TGF-β-induced smad signal transduction pathway in human hypertrophic scar fibroblasts in vitro. Burns. 2012;38:404–413. doi: 10.1016/j.burns.2011.08.013.
    1. Omranifard M, Heidari M, Farajzadegan Z, Niktabar MR, Motamedi N. Botulinum toxin and burn induces contracture. Arch Plast Surg. 2016;43:609–611. doi: 10.5999/aps.2016.43.6.609.
    1. Jeong HS, Lee BH, Sung HM, Park SY, Ahn DK, Jung MS, Suh IS. Effect of botulinum toxin type A on differentiation of fibroblasts derived from scar tissue. Plast Reconstr Surg. 2015;136:171e–178e. doi: 10.1097/PRS.0000000000001438.
    1. Gauglitz GG, Bureik D, Dombrowski Y, Pavicic T, Ruzicka T, Schauber J. Botulinum toxin A for the treatment of keloids. Skin Pharmacol Physiol. 2012;25:313–318. doi: 10.1159/000342125.
    1. Rasaii S, Sohrabian N, Gianfaldoni S, Hadibarhaghtalab M, Pazyar N, Bakhshaeekia A, Lotti T, Ramirez-Pacheco LA, Lange CS, Matta J, et al. Intralesional triamcinolone alone or in combination with botulinium toxin A is ineffective for the treatment of formed keloid scar: A double blind controlled pilot study. Dermatol Ther. 2019;32:e12781. doi: 10.1111/dth.12781.
    1. Xiao Z, Zhang F, Cui Z. Treatment of hypertrophic scars with intralesional botulinum toxin type A injections: A preliminary report. Aesthetic Plast Surg. 2009;33:409–412. doi: 10.1007/s00266-009-9334-z.
    1. Xiao Z, Qu G. Effects of botulinum toxin type a on collagen deposition in hypertrophic scars. Molecules. 2012;17:2169–2177. doi: 10.3390/molecules17022169.
    1. Lee SD, Yi MH, Kim DW, Lee Y, Choi Y, Oh SH. The effect of botulinum neurotoxin type A on capsule formation around silicone implants: The in vivo and in vitro study. Int Wound J. 2016;13:65–71. doi: 10.1111/iwj.12228.
    1. Chen M, Yan T, Ma K, Lai L, Liu C, Liang L, Fu X. Botulinum toxin type A inhibits α-smooth muscle actin and myosin II expression in fibroblasts derived from scar contracture. Ann Plast Surg. 2016;77:e46–e49. doi: 10.1097/SAP.0000000000000268.
    1. Xiao Z, Zhang M, Liu Y, Ren L. Botulinum toxin type a inhibits connective tissue growth factor expression in fibroblasts derived from hypertrophic scar. Aesthetic Plast Surg. 2011;35:802–807. doi: 10.1007/s00266-011-9690-3.
    1. Kim S, Ahn M, Piao Y, Ha Y, Choi DK, Yi MH, Shin N, Kim DW, Oh SH. Effect of botulinum toxin type A on TGF-beta/Smad pathway signaling: Implications for silicone-induced capsule formation. Plast Reconstr Surg. 2016;138:821e–829e. doi: 10.1097/PRS.0000000000002625.
    1. Li Y, Zhang J, Zhou Q, Wang H, Xie S, Yang X, Ji P, Zhang W, He T, Liu Y, et al. Linagliptin inhibits high glucose-induced transdifferentiation of hypertrophic scar-derived fibroblasts to myofibroblasts via IGF/Akt/mTOR signalling pathway. Exp Dermatol. 2019;28:19–27. doi: 10.1111/exd.13800.
    1. Liu B, Guo Z, Gao W. miR-181b-5p promotes proliferation and inhibits apoptosis of hypertrophic scar fibroblasts through regulating the MEK/ERK/p21 pathway. Exp Ther Med. 2019;17:1537–1544.
    1. Zhang Q, Guo B, Hui Q, Chang P, Tao K. miR-137 inhibits proliferation and metastasis of hypertrophic scar fibroblasts via targeting pleiotrophin. Cell Physiol Biochem. 2018;49:985–995. doi: 10.1159/000493236.
    1. Lim CP, Phan TT, Lim IJ, Cao X. Stat3 contributes to keloid pathogenesis via promoting collagen production, cell proliferation and migration. Oncogene. 2006;25:5416–5425. doi: 10.1038/sj.onc.1209531.
    1. Liao WT, Yu HS, Arbiser JL, Hong CH, Govindarajan B, Chai CY, Shan WJ, Lin YF, Chen GS, Lee CH. Enhanced MCP-1 release by keloid CD14+ cells augments fibroblast proliferation: Role of MCP-1 and Akt pathway in keloids. Exp Dermatol. 2010;19:e142–e150. doi: 10.1111/j.1600-0625.2009.01021.x.
    1. Kulkarni AA, Thatcher TH, Olsen KC, Maggirwar SB, Phipps RP, Sime PJ. PPAR-γ ligands repress TGFβ-induced myofibroblast differentiation by targeting the PI3K/Akt pathway: Implications for therapy of fibrosis. PLoS One. 2011;6:e15909. doi: 10.1371/journal.pone.0015909.
    1. Paterno J, Vial IN, Wong VW, Rustad KC, Sorkin M, Shi Y, Bhatt KA, Thangarajah H, Glotzbach JP, Gurtner GC. Akt-mediated mechanotransduction in murine fibroblasts during hypertrophic scar formation. Wound Repair Regen. 2011;19:49–58. doi: 10.1111/j.1524-475X.2010.00643.x.
    1. Dey JH, Bianchi F, Voshol J, Bonenfant D, Oakeley EJ, Hynes NE. Targeting fibroblast growth factor receptors blocks PI3K/AKT signaling, induces apoptosis, and impairs mammary tumor outgrowth and metastasis. Cancer Res. 2010;70:4151–4162. doi: 10.1158/0008-5472.CAN-09-4479.
    1. Deng B, Yang X, Liu J, He F, Zhu Z, Zhang C. Focal adhesion kinase mediates TGF-beta1-induced renal tubular epithelial-to-mesenchymal transition in vitro. Mol Cell Biochem. 2010;340:21–29. doi: 10.1007/s11010-010-0396-7.
    1. Guo L, Chen L, Bi S, Chai L, Wang Z, Cao C, Tao L, Li S. PTEN inhibits proliferation and functions of hypertrophic scar fibroblasts. Mol Cell Biochem. 2012;361:161–168. doi: 10.1007/s11010-011-1100-2.
    1. Takashima M, Parsons CJ, Ikejima K, Watanabe S, White ES, Rippe RA. The tumor suppressor protein PTEN inhibits rat hepatic stellate cell activation. J Gastroenterol. 2009;44:847–855. doi: 10.1007/s00535-009-0073-3.
    1. White ES, Thannickal VJ, Carskadon SL, Dickie EG, Livant DL, Markwart S, Toews GB, Arenberg DA. Integrin alpha4beta1 regulates migration across basement membranes by lung fibroblasts: A role for phosphatase and tensin homologue deleted on chromosome 10. Am J Respir Crit Care Med. 2003;168:436–442. doi: 10.1164/rccm.200301-041OC.
    1. Larsson O, Diebold D, Fan D, Peterson M, Nho RS, Bitterman PB, Henke CA. Fibrotic myofibroblasts manifest genome-wide derangements of translational control. PLoS One. 2008;3:e3220. doi: 10.1371/journal.pone.0003220.
    1. Mori T, Okumura M, Matsuura M, Ueno K, Tokura S, Okamoto Y, Minami S, Fujinaga T. Effects of chitin and its derivatives on the proliferation and cytokine production of fibroblasts in vitro. Biomaterials. 1997;18:947–951. doi: 10.1016/S0142-9612(97)00017-3.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 2001;25:402–408. doi: 10.1006/meth.2001.1262.
    1. Jiang B, Zu W, Xu J, Xiong Z, Zhang Y, Gao S, Ge S, Zhang L. Botulinum toxin type A relieves sternocleidomastoid muscle fibrosis in congenital muscular torticollis. Int J Biol Macromol. 2018;112:1014–1020. doi: 10.1016/j.ijbiomac.2018.02.077.
    1. Zhao JC, Zhang BR, Hong L, Shi K, Wu WW, Yu JA. Extracorporeal shock wave therapy with low-energy flux density inhibits hypertrophic scar formation in an animal model. Int J Mol Med. 2018;41:1931–1938.
    1. Qiu SS, Dotor J, Hontanilla B. Effect of P144® (Anti-TGF-β) in an 'In Vivo' human hypertrophic scar model in nude mice. PLoS One. 2015;10:e0144489. doi: 10.1371/journal.pone.0144489.
    1. Wang X, Gao Z, Wu X, Zhang W, Zhou G, Liu W. Inhibitory effect of TGF-β peptide antagonist on the fibrotic phenotype of human hypertrophic scar fibroblasts. Pharm Biol. 2016;54:1189–1197.
    1. Oliveira GV, Hawkins HK, Chinkes D, Burke A, Tavares AL, Ramos-e-Silva M, Albrecht TB, Kitten GT, Herndon DN. Hypertrophic versus non hypertrophic scars compared by immu-nohistochemistry and laser confocal microscopy: Type I and III collagens. Int Wound J. 2009;6:445–452. doi: 10.1111/j.1742-481X.2009.00638.x.
    1. Wang YW, Liou NH, Cherng JH, Chang SJ, Ma KH, Fu E, Liu JC, Dai NT. siRNA-targeting transforming growth factor-β type I receptor reduces wound scarring and extracellular matrix deposition of scar tissue. J Invest Dermatol. 2014;134:2016–2025. doi: 10.1038/jid.2014.84.
    1. Ulrich D, Ulrich F, Unglaub F, Piatkowski A, Pallua N. Matrix metalloproteinases and tissue inhibitors of metalloproteinases in patients with different types of scars and keloids. J Plast Reconstr Aesthet Surg. 2010;63:1015–1021. doi: 10.1016/j.bjps.2009.04.021.
    1. Huang D, Liu Y, Huang Y, Xie Y, Shen K, Zhang D, Mou Y. Mechanical compression upregulates MMP9 through SMAD3 but not SMAD2 modulation in hypertrophic scar fibroblasts. Connect Tissue Res. 2014;55:391–396. doi: 10.3109/03008207.2014.959118.
    1. Luan Y, Chen M, Zhou L. MiR-17 targets PTEN and facilitates glial scar formation after spinal cord injuries via the PI3K/Akt/mTOR pathway. Brain Res Bull. 2017;128:68–75. doi: 10.1016/j.brainresbull.2016.09.017.
    1. Pi WF, Guo XJ, Su LP, Xu WG. Troglitazone upregulates PTEN expression and induces the apoptosis of pulmonary artery smooth muscle cells under hypoxic conditions. Int J Mol Med. 2013;32:1101–1109. doi: 10.3892/ijmm.2013.1487.
    1. Yan YT, Li SD, Li C, Xiong YX, Lu XH, Zhou XF, Yang LQ, Pu LJ, Luo HY. Panax notoginsenoside saponins Rb1 regulates the expressions of Akt mTOR/PTEN signals in the hippocampus after focal cerebral ischemia in rats. Behav Brain Res. 2018;345:83–92. doi: 10.1016/j.bbr.2018.02.037.
    1. Sanders YY, Pardo A, Selman M, Nuovo GJ, Tollefsbol TO, Siegal GP, Hagood JS. Thy-1 promoter hypermethylation: A novel epigenetic pathogenic mechanism in pulmonary fibrosis. Am J Respir Cell Mol Biol. 2008;39:610–618. doi: 10.1165/rcmb.2007-0322OC.
    1. Bian EB, Huang C, Ma TT, Tao H, Zhang H, Cheng C, Lv XW, Li J. DNMT1-mediated PTEN hypermethylation confers hepatic stellate cell activation and liver fibrogenesis in rats. Toxicol Appl Pharmacol. 2012;264:13–22. doi: 10.1016/j.taap.2012.06.022.
    1. Wang X, Wang Z, Wang Q, Wang H, Liang H, Liu D. Epigenetic modification differences between fetal fibroblast cells and mesenchymal stem cells of the Arbas Cashmere goat. Res Vet Sci. 2017;114:363–369. doi: 10.1016/j.rvsc.2017.07.007.
    1. Zhu G, Chai J, Ma L, Duan H, Zhang H. Downregulated microRNA-32 expression induced by high glucose inhibits cell cycle progression via PTEN upregulation and Akt inactivation in bone marrow-derived mesenchymal stem cells. Biochem Biophys Res Commun. 2013;433:526–531. doi: 10.1016/j.bbrc.2013.03.018.
    1. Kitagishi Y, Matsuda S. Diets involved in PPAR and PI3K/AKT/PTEN pathway may contribute to neuroprotection in a traumatic brain injury. Alzheimers Res Ther. 2013;5:42. doi: 10.1186/alzrt208.
    1. Zhang Y, Yao X, Jiang C, Yue J, Guan J, Cheng H, Hajirashid M, Wang Y, Fan L. Expression of PI3K, PTEN and Akt in small intestinal adenocarcinoma detected by quantum dots-based immunofluorescence technology. Cancer Biomark. 2013;13:299–305. doi: 10.3233/CBM-130352.
    1. Pericacho M, Velasco S, Prieto M, Llano E, López-Novoa JM, Rodríguez-Barbero A. Endoglin haploinsufficiency promotes fibroblast accumulation during wound healing through Akt activation. PLoS One. 2013;8:e54687. doi: 10.1371/journal.pone.0054687.
    1. Seront E, Pinto A, Bouzin C, Bertrand L, Machiels JP, Feron O. PTEN deficiency is associated with reduced sensitivity to mTOR inhibitor in human bladder cancer through the unhampered feedback loop driving PI3K/Akt activation. Br J Cancer. 2013;109:1586–1592. doi: 10.1038/bjc.2013.505.

Source: PubMed

3
Abonnieren