Abstract
Platelet-derived growth factor-D (PDGF-D) signaling pathway has been reported to be involved in regulating various cellular processes, such as cell growth, apoptotic cell death, migration, invasion, angiogenesis and metastasis. Recently, multiple studies have shown that PDGF-D plays a critical role in governing Epithelial-to-Mesenchymal Transition (EMT), although the underlying mechanism of PDGF-D-mediated acquisition of EMT is largely unclear. Therefore, this mini review will discuss recent advances in our understanding of the role of PDGF-D in the acquisition of EMT during tumorigenesis. Furthermore, we will summarize the function of chemical inhibitors and natural compounds that are known to inactivate PDGF-D signaling pathway, which leads to the reversal of EMT. In summary, inactivation of PDGF-D could be a novel strategy for achieving better treatment outcome of patients inflicted with cancers.
Keywords: PDGF-D, EMT, migration, invasion, miRNA, natural compounds, stem cells
Introduction
Cancer is a major public health problem and one of the most common causes of deaths in the world. It has been estimated that 1,638,910 new cancer cases will be diagnosed and 577,190 cancer-related deaths will occur worldwide in 2012 1. Currently, 33% women and 50% men will develop cancer in their lifetime in the United States 2. Although progress has been made in reducing cancer incidence, the number of individuals diagnosed with cancer each year is still increasing which is in part due to aging and population growth in some countries 2. Moreover, five-year relative survival rate is approximately 65% for all human cancers. One of the reasons for this high mortality rate is due to the lack of understanding of the exact mechanism underlying cancer development and progression, and thus resulting in the shortage of effective treatment strategies 1. Therefore, it is pivotal to explore the molecular mechanisms how cancer is developed and discover novel drugs for the treatment of human cancer patients.
In recent years, although the causes of carcinogenesis are largely unknown, studies indicated that many factors and co-factors such as family history, smoking, obesity, and diet are correlated with increased incidence of cancer 3, 4. Furthermore, it has been demonstrated that many genes and cellular signaling pathways such as Notch 5–7, PI3K (phosphatidylinositol 3-Kinase)/Akt 8, 9, Hedgehog 10, 11, Wnt 5, 12, NF-κB (nuclear factor-kappa B) 13–15 have been found to play critical roles in human tumorigenesis. Among many of these signaling pathways, PDGF-D has recently gained tremendous amount of attention due to its involvement in carcinogenesis, which is mediated through regulating many cellular functions 16–21. More importantly, PDGF-D has been found to regulate the process of Epithelial-to-Mesenchymal Transition (EMT) that is important for tumor metastasis 22–27. Therefore, in this review article, we will briefly introduce the role of PDGF-D signaling pathway and further discuss its role in the acquisition of EMT phenotype. Lastly, we will describe potential application of PDGF-D inhibitors for the prevention and/or treatment of human cancers in the near future.
PDGF-D signaling pathway
PDGF-D belongs to PDGF family that is comprised of four polypeptide chains encoded by four genes, known as PDGF-A, B, C, and D 28. It is clear that in order to exert its function, PDGF-D needs to be assembled into dimmers via homodimerization and also requires to activate its full-length PDGF-D into a biologically active PDGF-D form 28. It is worth mentioning that PDGF-D exerts its cellular effects through specifically binding to and activating its cognate receptor PDGFR-β, resulting in phosphorylation of PDGFR-β and subsequent triggering of a number of downstream signaling pathways 29. In recent years, studies have shown that multiple cellular signaling pathways such as matriptase, PTEN (phosphatase and tensin homologue deleted on chromosome 10), ROS (reactive oxygen species), and uPA (urokinase-type plasminogen activator) govern the PDGF-D activity 16, 18, 19, 30–33. Recently, we found that activated K-ras and INK4a/Arf deficiency led to high expression of PDGF-D pathway 27. On the other hand, PDGF-D could regulate various cellular pathways including PI3K/Akt, NF-κB, Notch, ERK (extracellular-regulated kinase), mTOR (mammalian target of rapamycin), MAPK (mitogen-activated protein kinase), VEGF (vascular endothelial growth factor), MMPs (matrix metalloproteinases), Cyclin D1, and Bcl-2 to facilitate its oncogenic functions 22–24, 34–37.
In agreement with the oncogenic fuction of PDGF-D in human malignancies, over-expression of PDGF-D has been observed in a variety of cancers including prostate, lung, renal, ovarian, brain, and pancreatic cancer 38, 39. For example, we found that PDGF-D is highly expressed in pancreatic adenocarcinoma specimens as well as in chronic pancreatitis associated with pancreatic adenocarcinoma 36. In line with this notion, we also observed over-expression of PDGF-D in breast cancer tissues 26. Similarly, PDGF-D is upregulated in primary prostate cancer and bone metastases 16, 18, 30. To further support the role of PDGF-D in tumorigenesis, Yang et al. reported that PDGF-D is overexpressed in gastric cancer patient tissue samples compared with normal tissues 40. Moreover, PDGF-D was found to be elevated in the sera of ovarian, renal, lung and brain cancer patients, indicating that PDGF-D plays a crucial role in certain human cancers 41. Because several excellent published reviews have discussed the roles of PDGF-D in cell growth, apoptosis, and angiogenesis 28, 29, 38, 39, 42, 43, in the following paragraph we will describe the novel functions of PDGF-D in regulating EMT in human cancer, which have recently burst onto the scene.
The Epithelial-to-Mesenchymal Transition (EMT)
It is known that EMT is essential for numerous developmental processes during embryogenesis 44. Morphologically, epithelial cells lose their apical-basal polarization and adopt front-to-back polarization. Moreover, cells acquire mesenchymal characteristics such as increased invasive and motility characteristics 44. After completion of embryogenesis, it has been believed that EMT is switched off and put to rest. In specific physiological condition such as wound healing, EMT might be reactivated 44. Various molecular markers have been used to distinguish mesenchymal cells from epithelial cells which have helped in the identification of the EMT phenotypic cells 45, 46. E-cadherin, the best-characterized member of epithelial markers, is lost upon EMT, leading to migration 47. The mesenchymal markers such as Snail, Slug, ZEB1, and Twist have been found to repress the expression of E-cadherin. Vimentin and fibronectin as mesenchymal molecules provide a flexible structure and promote cells to move 44, 47.
Recently, accumulating evidence suggests that cancer cells reactivate the processes of EMT in order to migrate to distant tissue sites 44. Due to increased cell migratory capacity after EMT, EMT is believed to be involved in tumor metastasis 44. So far, many growth factors and cytokines as well as cellular signaling pathways have been found to trigger EMT program such as TGF-β (transforming growth factor beta), Wnt, FoxM1 (forkhead box protein M1), NF-κB, EGF (epidermal growth factor), HGF (hepatocyte growth factor) and Notch 44, 47–49. For example, we found that over-expression of FoxM1 in pancreatic cancer cells induced EMT 50. In addition, we also revealed that up-regulation of sonic hedgehog contributes to TGF-β-induced EMT in NSCLC cells 51. Recently, the growing body of literature strongly suggests that PDGF-D plays a critical role in governing EMT 22–27. Therefore, we will highlight the function of PDGF-D in triggering EMT in a variety of human cancers.
The role of PDGF-D in the processes of EMT
Several lines of evidence have revealed that PDGF-D could facilitate EMT. The group led by Dr. Sarkar demonstrated that overexpression of PDGF-D in prostate cancer cells caused changes in the morphology with elongated and irregular fibroblastoid appearance 24. Consistent with these changes, PDGF-D-overexpressing cells showed loss or relocation of E-cadherin at cell-cell junction, and disrupted ZO-1 (zonual occludens-1) in tight junction 24. Concomitantly, β-catenin was relocated from cell membrane to the nuclear compartment by PDGF-D overexpression 24. In addition, increased expression levels of vimentin and nestin were also observed in PDGF-D-overexpressing cells 24. Recently, we found that PDGF-D knockdown in Rink cells led to down-regulation of mesenchymal marker vimentin 27. Taken together, PDGF-D overexpression contributes to EMT in human cancers.
Although the molecular mechanisms by which PDGF-D triggers EMT are largely unknown, multiple studies have indicated that PDGF-D-mediated EMT could be mediated through several cellular signaling pathways such as mTOR, Notch, NF-κB, CXCR4 (C-X-C chemokine receptor type 4), and Bcl-2 22–27. Strikingly, microRNAs (miRNAs) have been reported to regulate PDGF-D-mediated EMT 23. Furthermore, PDGF-D-induced EMT phenotypic cells have cancer stem-like cell characteristic 22. These findings provide compelling evidence for the role of PDGF-D in regulating EMT via multiple mechanisms.
PDGF-D-mediated EMT is mediated through multiple signaling pathways
mTOR signaling pathway
mTOR, an evolutionarily conserved Serine/Theonine kinase, belongs to the PI3K-related protein kinase subfamily 52. It is known that mTOR regulates multiple cellular processes including cell proliferation, apoptotic cell death, cell metabolism and autophagy 53. To exert its functions, mTOR assembles into two sub-complexes, known as mTORC1 (mTOR complex 1) and mTORC2 (mTOR complex 2). mTORC1 is comprised of five components, such as DEPTOR (DEP-domain-containing mTOR-interacting protein), mTOR, mLST8/GβL (mammalian lethal with Sec13 protein 8/G protein β-subunit-like protein), PRAS40 (proline-rich Akt substrate of 40 kilodaltons), and Raptor (regulatory-associated protein of mTOR) 53. mTORC2 consists of six components, DEPTOR, MAPKAP1 (mitogen-activated protein kinase associated protein 1; also known as mSIN1), mLST8/GβL, mTOR, PROTOR (protein observed with Rictor-1)/PRR5 (proline-rich protein 5) and Rictor (Rapamycin-insensitive companion of mTOR) 54.
It has been well documented that deregulation of the mTOR pathway occurs in many human cancers 55. Moreover, mTORC1 facilitates its oncogenic function through regulating its downstream targets such as S6K (p70 S6 ribosomal kinase) and 4E-BP1 (the eukaryotic translation initiation factor 4E-binding protein 1), while mTORC2 phosphorylates Akt and SGK1 (serum/glucocorticoid regulated kinase 1) to active their kinase activation 56. Furthermore, mTORC2 mediates the actin cytoskeletal organization, leading to increased tumor cell invasion and metastasis 54. Consistent with this notion, studies from different groups have suggested that mTOR signaling is associated with cancer progression and metastasis 56. Accordingly, mTOR signaling has been found to be significantly elevated in a variety of human cancers. Moreover, inhibiting mTOR signaling by its inhibitor rapamycin attenuated tumor cell migration and invasion 57.
mTOR has been reported to drive cancer-associated EMT 58, 59. For example, inhibition of mTORC1 and mTORC2 induced the reversal of EMT to MET (mesenchymal-to-epithelial transition) with increased cell-cell contact and decreased actin cytoskeletal remodeling, suggesting that mTORC1 and mTORC2 could regulate EMT in human cancers 58. In supporting the role of mTOR signaling in the regulation of EMT, it has been observed that TGF-β-induced EMT is mediated through induction of mTORC2 kinase activity, demonstrating that mTORC2 is an essential downstream molecule of TGF-β signaling, and represents a novel target for the inhibition of EMT 59. Recently, our studies have shown that over-expression of PDGF-D in prostate cancer cells promoted cell proliferation and tumor growth in vitro and in vivo through activation of mTOR downstream targets S6K and 4E-BP1 24. More importantly, we found that activation of mTOR pathway is involved in EMT induced by PDGF-D 24. To support this concept, we observed an increased activation of mTOR concomitant with loss of E-cadherin and gain of increased vimentin and nestin in PDGF-D-overexpressing cells 24. To further confirm this notion, knockdown of mTOR increased the expression of E-cadherin and reduced the level of vimentin in PDGF-D-overexpressing cells 24. These results provided the direct evidence to support that the activation of mTOR is associated with the acquisition of EMT induced by PDGF-D.
Notch signaling pathway
The Notch signaling pathway is known as a conserved ligand–receptor pathway, which is involved in various cellular processes such as cell proliferation, differentiation, apoptosis, migration, invasion and metastasis 60. So far, four Notch receptors (Notch-1, Notch-2, Notch-3, and Notch-4) and five ligands (Dll-1, Dll-3, Dll-4, Jagged-1 and Jagged-2) have been identified 60. The activation of Notch signaling is triggered after Notch ligand binds to its receptor, thus leading to Notch proteolytic cleavage and releasing the active Notch intracellular domain (NICD). The NICD then translocates into the nucleus and activates its target genes 61. It has been known that Notch downstream targets include the Hes-1 (Hairy enhance of split-1), COX-2 (cyclooxygenase-2), VEGF, MMP-9 (matrix metalloproteinase-9), ERK, Akt, Cyclin D1, c-Myc, p21, p27, p53, etc 62.
Notch signaling network is frequently deregulated in human malignancies 6, 63–65. Recently, Notch signaling pathway has been observed to induce EMT 66. For example, we showed that forced over-expression of Notch-1 led to the induction of EMT by activation of mesenchymal cell markers in pancreatic cancer cells 67. In addition, our study demonstrated that over-expression of PDGF-D caused up-regulation of Notch-1 and Jagged-1 in breast cancer cells 34. Consistent with this notion, we found that down-regulation of PDGF-D inhibited Notch-1 expression whereas up-regulation of PDGF-D increased Notch-1 activation in pancreatic cancer cells 36. Furthermore, PDGF-D overexpression increased the expression of mesenchymal markers vimentin and ZEB2 and decreased the expression of epithelial marker E-cadherin in breast cancer cells 34. Taken together, since Notch-1 plays a critical role in regulating EMT, PDGF-D-mediated EMT could indeed be mediated through up-regulation of Notch-1 signaling pathway.
NF-κB signaling pathway
It has been well documented that NF-κB plays pivotal roles in multiple cellular processes such as cell growth, cellular apoptotic death, inflammation, differentiation, and stress response 13. NF-κB is sequestered in the cytoplasm via association with its inhibitory protein IκB. Under stimulation conditions, IKKα (IκB kinase α) and IKKβ (IκB kinase β) phosphorylate IκB, leading to the degradation of IκB by E3 ubiquitin ligase, subsequently allowing the NF-κB to translocate into the nucleus 68. Then, NF-κB binds to the DNA and recruits additional transcription factors and regulates the expression of its target genes such as Bcl-2, MMP-9, Cyclin D1 and VEGF 14, 69.
NF-κB has been found to be involved in the development and progression of human cancers 13. Moreover, NF-κB was recently found to regulate EMT in cancers 70. Our studies have demonstrated that PDGF-D controlled NF-κB activity in breast cancer, prostate cancer, and pancreatic cancer cell lines 24, 34, 36. Specifically, over-expression of PDGF-D enhanced NF-κB expression and its nuclear abundance as well as its DNA-binding activity, leading to the activation of its target genes including VEGF and MMP-9 34, 36. On the contrary, down-regulation of PDGF-D led to the inactivation of NF-κB DNA-binding activity and, in turn, down-regulated the expression of its target genes, such as VEGF and MMP-9 34, 36. Since both PDGF-D and its downstream gene NF-κB induced EMT, it is possible that PDGF-D governs EMT through NF-κB pathway. It is worth mentioning that NF-κB could be a Notch-1 target gene in various cancer cells 71. Therefore, we assumed that PDGF-D triggers EMT partly mediated through the regulation of Notch/NF-κB pathways. However, further in-depth investigation is required to validate this hypothesis in the future.
Bcl-2 signaling pathway
It has been well accepted that Bcl-2 protein family plays a critical role in the apoptotic signaling pathway 72. The Bcl-2 family of proteins is classified into anti-apoptotic group and pro-apoptotic group 72. The anti-apoptotic members have Bcl-2, Bcl-xL, Bcl-w and Mcl-1, while the pro-apoptotic Bcl-2 members include Bax, Bak, and BH3-only proteins such as Bim 73, 74. Recently, the pro-apoptotic protein Bcl-2 has attracted increasing attention due to its oncogenic functions in addition to being a pivotal regulator of apoptosis. For example, Choi et al. reported that Bcl-2 promotes invasion and lung metastasis via induction of MMP-2 75. Similarly, another study from different group showed that Bcl-2 over-expression enhanced in vitro invasion and in vivo tumor growth through regulating several MMPs such as MMP-2, MMP-7, MT1-MMP, and TIMP1 (tissue inhibitors of metalloproteases-1) and TIMP-2 in melanoma 76. Furthermore, it has been found that under hypoxia, Bcl-2 protein promotes HIF-1 (hypoxia-inducible factor-1)/VEGF-mediated tumor angiogenesis 77. Moreover, Bcl-2 regulates HIF-1α protein stabilization involving the molecular chaperone Hsp90 in hypoxic melanoma cells 78.
Recently, Bcl-2 was found to be involved in EMT. For example, Wang et al. reported that Bcl-2 plays a critical role in oestrogen-mediated EMT 79. Specifically, oestrogen signaling inhibits EMT through repressing Bcl-2 expression 79. To further support the role of Bcl-2 in EMT, one study has shown that Bcl-2 could bind to the EMT-regulating transcription factor Twist1 in vitro and in vivo, and facilitate the nuclear transport of Twist, leading to transcriptional activation of multiple genes that can promote tumor cell metastasis 80. This finding indicates a new function of Bcl-2 in the induction of EMT 80. Consistent with this observation, we demonstrated that over-expression of Bcl-2 in prostate cancer cells decreased the expression of E-cadherin 24. Moreover, we revealed that Bcl-2 contributes to EMT induced by PDGF-D overexpression 24. First, up-regulation of Bcl-2 was observed in PDGF-D-overexpressing cells with EMT phenotype 24. Second, knockdown of Bcl-2 in PDGF-D-overexpressing cells decreased the expression of vimentin, which is the key marker of EMT cells 24. Taken together, Bcl-2 could partly contribute to EMT induced by PDGF-D over-expression. It is important to note that Bcl-2 is one of many targets of NF-κB. Therefore, it is possible that PDGF-D controls EMT partly through the regulation of NF-κB/Bcl-2 pathways.
CXCR4 signaling pathway
CXCR4 is a G-protein-coupled receptor for SDF-1/CXCL12 (stromal cell-derived factor-1/CXC chemokine ligand 12) that is a small pro-inflammatory chemoattractant cytokine 81, 82. SDF-1 binds to CXCR4 and subsequently activates several downstream signaling pathways such as ERK2, protein kinase B, PI3K, MAPK and NF-κB, which in turn govern cell proliferation, survival and chemotaxis 83. In line with the notion that CXCR4 positively regulates multiple oncogenic pathways, over-expression of CXCR4 has been reported in various human cancers including prostate, breast and pancreatic cancer 83. Moreover, activation of CXCR4 induced cancer cell migration and adhesion to stromal cells, thus activated growth factors to the tumor cells. Accordingly, inactivation of SDF-1/CXCR4 signaling blocked cancer cell invasion and migration 84.
CXCR4 has been found to play a critical role in EMT 84. SDF-1/CXCR4 induced EMT processes, which was in part mediated through the activation of hedgehog signaling pathway in pancreatic cancer cells. This EMT process was inhibited by Smoothened inhibitor cyclopamine, and knockdown of hedgehog targeted Gli-1 84. Recently, PDGF-D has been found to induce CXCR4 expression in breast cancer cells 85. Specifically, over-expression of PDGF-D promotes tumor growth and lymph node metastasis which was partly mediated through up-regulation of CXCR4 expression 85. In support of this, inhibition of CXCR4 abolished PDGF-D-mediated lymph node metastasis 85. Taken together, these results suggest that the SDF-1/CXCR4 axis is involved in PDGF-D-induced EMT, suggesting that modulating SDF-1/CXCR4 may have clinical applications in the inhibition of EMT and metastatic potential of cancer cells.
miRNA regulates PDGF-D-mediated EMT
It became clear that miRNAs are small regulatory non-coding RNA molecules that negatively regulate the expression of the target genes through interacting with the 3′ untranslated regions (UTRs) of specific mRNAs 86. Therefore, miRNA represses gene expression at a post-transcriptional level by blocking mRNA translation or promoting their degradation 87. Notably, miRNAs have been found to be involved in the development and progression of human cancers. Without a doubt, miRNAs can serve as tumor suppressor genes or oncogenes depending on the function of their targets 88. It has been established that miRNAs are involved in cancer pathogenesis including cell growth, migration, invasion, metastasis, and drug resistance 89.
Increasing evidence indicates that miRNAs play important roles in EMT. For example, miR-150 targets the EMT inducer ZEB1 in esophageal squamous cell carcinoma, leading to the induction of MET-like changes with up-regulation of E-cadherin and down-regulation of vimentin 90. Han et al. reported that miR-21 regulates EMT via over-expression of HIF-1α 91, while inhibition of miR-21 reverses EMT phenotype through inactivation of Akt/ERK by targeting PTEN 92. Cao et al. found that miR-23 regulates TGF-β-induced EMT by targeting E-cadherin in lung cancer cells 93. Recently, we found that miR-200 regulates PDGF-D-mediated EMT in prostate cancer cells 23. Specifically, miR-200 contributes to the regulation of EMT partly through down-regulation of ZEB1, ZEB2 as well as Snail2, and up-regulation of epithelial marker genes such as E-cadherin, stratifin, EpCAM, and connexin 26 23. Re-expression of miR-200b reversed EMT to MET in PDGF-D-overexpressing prostate cancer cells, leading to inhibition of cell adhesion and invasion 23. More importantly, purified PDGF-D protein treatment led to the repression of miR-200 family and increased expression of ZEB2, Snail2 as well as down-regulation of E-cadherin 23. Taken together, these results suggest that miR-200 governs PDGF-D-mediated EMT in human cancers.
PDGF-D-induced EMT-type cells show cancer stem cell features
Accumulating evidence suggests that EMT-type cells have CSCs (cancer stem cells) characteristics in various human malignancies 94, 95. It is believed that CSCs possess self-renewal activity and generate various cell populations 96. More importantly, CSCs have been identified and isolated using different sets of stem cell surface markers in a wide variety of human cancers including breast, lung, prostate, colon, brain, head and neck, and pancreatic cancers 97. Recently, the positive relationship between EMT and CSCs has been observed in human cancers 48, 98. To support this concept, studies have shown that epithelial cancer stem cells express a wide array of mesenchymal markers. On the other hand, the activation of EMT program has been associated with the acquisition of stem cell traits 99. For example, Mani et al. reported that the EMT transition generates cells with properties of stem cells in breast cancer 96. Additionally, this group showed that normal and neoplastic breast stem-like cells express mesenchymal markers 96. At the same time, another group confirmed that generation of breast cancer stem cells is mediated through EMT induction by the activation of the Ras-MAPK pathway 100. Similarly, Shah et al. revealed that EMT-type pancreatic cancer cells have up-regulation of the stem cell markers CD24, CD44, and ESA 101. Consistent with this finding, our studies have demonstrated that pancreatic cancer cells with EMT phenotype showed increased sphere-forming capacity and overexpression of CSC surface markers 50, 67. Therefore, the connection between EMT and CSCs demonstrated that the EMT process is involved in tumorigenesis through its cellular traits associated with motility, invasiveness and metastasis, as well as its stem cell traits with self-renewal capacity.
Although EMT is associated with CSCs, the mechanism by which EMT-type cells acquire CSCs features remains largely elusive. Recently, PDGF-D was found to play a critical role in governing EMT that also showed connection with CSCs 22. In concordance with this notion, we observed that prostate cancer cells with EMT phenotype induced by PDGF-D displayed CSCs features including up-regulation of Sox2, Nanog, Oct4, Lin28B and Notch-1, which is consistent with enhanced sphere-forming ability as well as rapid tumor growth in vivo 22. Moreover, reversal of EMT decreased prostasphere-forming ability of EMT-type cells and down-regulated the expression of Lin28B and Notch-1, leading to the repression of self-renewal capability 22. Recently, we found that activated K-Ras and INK4a/Arf deficiency promote aggressiveness of pancreatic cancer by the acquisition of EMT phenotype and induction of CSC characteristics partly through the activation of PDGF-D 27. Specifically, the deletion of Ink4a/Arf in K-Ras (G12D) expressing mice showed high expression of PDGF-D in the pancreatic tumor and tumor-derived cell line (Rink-1 cells) 27. Depletion of PDGF-D in Rink-1 cells led to reduced pancreatosphere formation and decreased expression of EZH2, CD44, EPCAM, and vimentin, indicating that PDGF-D plays a critical role in the induction of EMT consistent with CSCs phenotype 27. More recently, it has also been reported that tissue-resident stem cells induce EMT through interaction with cancer microenvironment via PDGF-D, thereby leading to increased numbers of CSCs and promoting tumor growth 25. However, further investigation is required to investigate how PDGF-D-induced EMT-type cells acquire CSCs characteristics in human cancers.
PDGF-D as a cancer therapeutic target
Since PDGF-D plays oncogenic activity through the regulation of tumor cell growth, invasion, and metastasis that are associated with EMT and CSCs, targeting PDGF-D signaling pathway could be a novel strategy for the treatment of human cancers. However, to our knowledge, small chemical inhibitors of PDGF-D have not been developed so far with proven anti-tumor efficacy in humans. Because PDGF-D exerts its function via activation of its receptor PDGFR-β, multiple small molecule tyrosine kinase inhibitors that inactivate PDGFR have been reported 102–105. For example, imatinib, a tyrosine kinase inhibitor that interferes with the phosphorylation and activation of PDGFRβ, has been reported to inhibit cell growth and invasion in various cancer cell lines 106. Sunitinib, a multi-tyrosine kinase inhibitor, reduces survival and migration of human meningioma cells by targeting PDGFR 107. Moreover, Sunitinib could serve as radiosensitizer for human prostate cancer partly through the inhibition of PDGFR and VEGFR 102. ABT-348 exerts its anti-tumor activity via targeting the VEGFR, PDGFR, Aurora, and SRC kinase 103. AL3810 exhibits potential anti-angiogenic and anti-tumor activity partly through the inhibition of PDGFR in human cancer cells 105. Cediranib was also found to reduce intraosseous growth of prostate tumor and associated bone reaction through the suppression of PDGFR and VEGFR 108. TKI258, a tyrosine kinase inhibitor of FGFR, PDGFR, and VEGFR, has also been reported to inhibit tumor growth, angiogenesis, and metastasis in pancreatic cancer 104. Tricin, a naturally occurring flavone, exhibited anti-oncogenic activity through blocking tyrosine phosphorylation of PDGFRβ and its downstream signaling molecules ERK1/2 109. It is important to note that most small molecule tyrosine kinase inhibitors target multiple tyrosine kinases.
Although there is no specific inhibitor for PDGF-D, a fully-humanized monoclonal antibody against PDGF-D, known as CR002, has been developed and shown to be safe in a phase I study 110. Interestingly, we found that 3,3′-Diindolylmethane (DIM) significantly reduced the expression and activation of PDGF-D in prostate cancer cells, leading to the inhibition of cell proliferation, invasion and angiogenesis 35. Consistent with this finding, DIM significantly decreased the expression of PDGF-D target genes including mTOR, S6K, and 4E-BP1 phosphoylation 35. Recently, we also observed that DIM treatment led to the regulation of EMT marker expression and the reversal of EMT 111. For example, DIM treatment caused up-regulation of epithelial marker E-cadherin and down-regulation of mesenchymal markers ZEB1, vimentin and Slug 111. Accordingly, the morphology of cancer cells after DIM treatment changed from elongated fibroblasoid to epithelial cobblestone-like appearance, suggesting that DIM could reverse EMT to MET 111. Since PDGF-D plays a critical role in induction of EMT, the reversal of EMT by DIM is possibly mediated through the down-regulation of PDGF-D. However, further investigation is required to experimentally address whether DIM could control EMT mainly via regulation of PDGF-D.
Conclusion and overall perspectives
In conclusion, PDGF-D plays a pivotal role in the development and progression of human malignancies, which is mediated through deregulation of various cellular processes including cell proliferation, cellular apoptotic death, migration, invasion, angiogenesis and metastasis. Moreover, PDGF-D has been demonstrated to exert its oncogenic function via regulation of multiple cellular signaling pathways, leading to governing EMT that is associated with CSCs characteristics (Figure 1). Therefore, targeting PDGF-D could be a promising strategy for the treatment of cancers. Since there is no specific inhibitor for PDGF-D, it could be useful to target several signaling pathways such as PTEN, uPA that regulate PDGF-D expression as an alternative strategy. More importantly, due to non-toxic nature, inactivation of PDGF-D by natural agent DIM could be a safer approach to achieve better treatment outcome of patients diagnosed with cancers. However, further in-depth investigation is necessary to explore the mechanism(s) how PDGF-D affects tumorigenesis using transgenic mouse model. More importantly, we hope that specific PDGF-D inhibitor could be developed which will serve as an efficient chemopreventive or chemotherapeutic agent for most human malignancies that over-express PDGF-D in the future.
Figure 1.
A proposed model for PDGF-D signaling pathway that controls the processes of EMT. PDGF-D triggers EMT through regulation of mTOR, Notch, NF-κB, CXCR4, Bcl-2 and microRNAs. Bcl-2: B-cell lymphoma 2; CXCR4: C-X-C chemokine receptor type 4; EMT: Epithelial to mesenchymal transition; mTOR: Mammalian target of rapamycin; NF-κB: Nuclear factor kappa B. PDGF-D: Platelet-derived growth factor-D; S6K: p70 S6 ribosomal kinase; 4E-BP1: the eukaryotic translation initiation factor 4E-binding protein 1.
Acknowledgments
This work was supported by funding from the NSFC (81172087), Anhui Provincial Natural Science Foundation (1208085MH170), and the Natural Science Research key Project of Education Office of Anhui Province (KJ2012A196). This work was also funded by grants from the National Cancer Institute, NIH CA131151 awarded to F.H.S.
Footnotes
Conflict of Interest
The authors declare that they have no conflict of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer J Clin. 2012;62:10–29. doi: 10.3322/caac.20138. [DOI] [PubMed] [Google Scholar]
- 2.Siegel R, DeSantis C, Virgo K, Stein K, Mariotto A, Smith T, et al. Cancer treatment and survivorship statistics, 2012. CA Cancer J Clin. 2012;62:220–41. doi: 10.3322/caac.21149. [DOI] [PubMed] [Google Scholar]
- 3.Gatenby RA, Gillies RJ, Brown JS. Evolutionary dynamics of cancer prevention. Nat Rev Cancer. 2010;10:526–7. doi: 10.1038/nrc2892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Luch A. Nature and nurture - lessons from chemical carcinogenesis. Nat Rev Cancer. 2005;5:113–25. doi: 10.1038/nrc1546. [DOI] [PubMed] [Google Scholar]
- 5.Bertrand FE, Angus CW, Partis WJ, Sigounas G. Developmental pathways in colon cancer: Crosstalk between WNT, BMP, Hedgehog and Notch. Cell Cycle. 2012:11. doi: 10.4161/cc.22134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Piazzi G, Bazzoli F, Ricciardiello L. Epigenetic silencing of Notch signaling in gastrointestinal cancers. Cell Cycle. 2012:11. doi: 10.4161/cc.22388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wang Z, Li Y, Ahmad A, Azmi AS, Banerjee S, Kong D, et al. Targeting Notch signaling pathway to overcome drug resistance for cancer therapy. Biochim Biophys Acta. 2010;1806:258–67. doi: 10.1016/j.bbcan.2010.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Gonzalez E, McGraw TE. The Akt kinases: isoform specificity in metabolism and cancer. Cell Cycle. 2009;8:2502–8. doi: 10.4161/cc.8.16.9335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Engelman JA. Targeting PI3K signalling in cancer: opportunities, challenges and limitations. Nat Rev Cancer. 2009;9:550–62. doi: 10.1038/nrc2664. [DOI] [PubMed] [Google Scholar]
- 10.Ng JM, Curran T. The Hedgehog’s tale: developing strategies for targeting cancer. Nat Rev Cancer. 2011;11:493–501. doi: 10.1038/nrc3079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Jagani Z, Dorsch M, Warmuth M. Hedgehog pathway activation in chronic myeloid leukemia. Cell Cycle. 2010;9:3449–56. doi: 10.4161/cc.9.17.12945. [DOI] [PubMed] [Google Scholar]
- 12.Klaus A, Birchmeier W. Wnt signalling and its impact on development and cancer. Nat Rev Cancer. 2008;8:387–98. doi: 10.1038/nrc2389. [DOI] [PubMed] [Google Scholar]
- 13.Perkins ND. The diverse and complex roles of NF-kappaB subunits in cancer. Nat Rev Cancer. 2012;12:121–32. doi: 10.1038/nrc3204. [DOI] [PubMed] [Google Scholar]
- 14.Nogueira L, Ruiz-Ontanon P, Vazquez-Barquero A, Moris F, Fernandez-Luna JL. The NFkappaB pathway: a therapeutic target in glioblastoma. Oncotarget. 2011;2:646–53. doi: 10.18632/oncotarget.322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Demchenko YN, Kuehl WM. A critical role for the NFkB pathway in multiple myeloma. Oncotarget. 2010;1:59–68. doi: 10.18632/oncotarget.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Huang W, Fridman Y, Bonfil RD, Ustach CV, Conley-Lacomb MK, Wiesner C, et al. A novel function for platelet-derived growth factor D: induction of osteoclastic differentiation for intraosseous tumor growth. Oncogene. 2012;31:4527–35. doi: 10.1038/onc.2011.573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Lokker NA, Sullivan CM, Hollenbach SJ, Israel MA, Giese NA. Platelet-derived growth factor (PDGF) autocrine signaling regulates survival and mitogenic pathways in glioblastoma cells: evidence that the novel PDGF-C and PDGF-D ligands may play a role in the development of brain tumors. Cancer Res. 2002;62:3729–35. [PubMed] [Google Scholar]
- 18.Ustach CV, Huang W, Conley-LaComb MK, Lin CY, Che M, Abrams J, et al. A novel signaling axis of matriptase/PDGF-D/ss-PDGFR in human prostate cancer. Cancer Res. 2010;70:9631–40. doi: 10.1158/0008-5472.CAN-10-0511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ustach CV, Kim HR. Platelet-derived growth factor D is activated by urokinase plasminogen activator in prostate carcinoma cells. Mol Cell Biol. 2005;25:6279–88. doi: 10.1128/MCB.25.14.6279-6288.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Xu L, Tong R, Cochran DM, Jain RK. Blocking platelet-derived growth factor-D/platelet-derived growth factor receptor beta signaling inhibits human renal cell carcinoma progression in an orthotopic mouse model. Cancer Res. 2005;65:5711–9. doi: 10.1158/0008-5472.CAN-04-4313. [DOI] [PubMed] [Google Scholar]
- 21.Zhao L, Zhang C, Liao G, Long J. RNAi-mediated inhibition of PDGF-D leads to decreased cell growth, invasion and angiogenesis in the SGC-7901 gastric cancer xenograft model. Cancer Biol Ther. 2010;9:42–8. doi: 10.4161/cbt.9.1.10282. [DOI] [PubMed] [Google Scholar]
- 22.Kong D, Banerjee S, Ahmad A, Li Y, Wang Z, Sethi S, et al. Epithelial to mesenchymal transition is mechanistically linked with stem cell signatures in prostate cancer cells. PLoS One. 2010;5:e12445. doi: 10.1371/journal.pone.0012445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kong D, Li Y, Wang Z, Banerjee S, Ahmad A, Kim HR, et al. miR-200 regulates PDGF-D-mediated epithelial-mesenchymal transition, adhesion, and invasion of prostate cancer cells. Stem Cells. 2009;27:1712–21. doi: 10.1002/stem.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kong D, Wang Z, Sarkar SH, Li Y, Banerjee S, Saliganan A, et al. Platelet-derived growth factor-D overexpression contributes to epithelial-mesenchymal transition of PC3 prostate cancer cells. Stem Cells. 2008;26:1425–35. doi: 10.1634/stemcells.2007-1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Devarajan E, Song YH, Krishnappa S, Alt E. Epithelial-mesenchymal transition in breast cancer lines is mediated through PDGF-D released by tissue-resident stem cells. Int J Cancer. 2012;131:1023–31. doi: 10.1002/ijc.26493. [DOI] [PubMed] [Google Scholar]
- 26.Sethi S, Sarkar FH, Ahmed Q, Bandyopadhyay S, Nahleh ZA, Semaan A, et al. Molecular markers of epithelial-to-mesenchymal transition are associated with tumor aggressiveness in breast carcinoma. Transl Oncol. 2011;4:222–6. doi: 10.1593/tlo.10244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Wang Z, Ali S, Banerjee S, Bao B, Li Y, Azmi AS, et al. Activated K-Ras and INK4a/Arf deficiency promote aggressiveness of pancreatic cancer by induction of EMT consistent with cancer stem cell phenotype. J Cell Physiol. 2012 doi: 10.1002/jcp.24162. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 28.Reigstad LJ, Varhaug JE, Lillehaug JR. Structural and functional specificities of PDGF-C and PDGF-D, the novel members of the platelet-derived growth factors family. FEBS J. 2005;272:5723–41. doi: 10.1111/j.1742-4658.2005.04989.x. [DOI] [PubMed] [Google Scholar]
- 29.Li X, Eriksson U. Novel PDGF family members: PDGF-C and PDGF-D. Cytokine Growth Factor Rev. 2003;14:91–8. doi: 10.1016/s1359-6101(02)00090-4. [DOI] [PubMed] [Google Scholar]
- 30.Conley-LaComb MK, Huang W, Wang S, Shi D, Jung YS, Najy A, et al. PTEN regulates PDGF ligand switch for beta-PDGFR signaling in prostate cancer. Am J Pathol. 2012;180:1017–27. doi: 10.1016/j.ajpath.2011.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Ustach CV, Taube ME, Hurst NJ, Jr, Bhagat S, Bonfil RD, Cher ML, et al. A potential oncogenic activity of platelet-derived growth factor d in prostate cancer progression. Cancer Res. 2004;64:1722–9. doi: 10.1158/0008-5472.can-03-3047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Liu MY, Eyries M, Zhang C, Santiago FS, Khachigian LM. Inducible platelet-derived growth factor D-chain expression by angiotensin II and hydrogen peroxide involves transcriptional regulation by Ets-1 and Sp1. Blood. 2006;107:2322–9. doi: 10.1182/blood-2005-06-2377. [DOI] [PubMed] [Google Scholar]
- 33.Najy AJ, Won JJ, Movilla LS, Kim HR. Differential tumorigenic potential and matriptase activation between PDGF B versus PDGF D in prostate cancer. Mol Cancer Res. 2012;10:1087–97. doi: 10.1158/1541-7786.MCR-12-0071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ahmad A, Wang Z, Kong D, Ali R, Ali S, Banerjee S, et al. Platelet-derived growth factor-D contributes to aggressiveness of breast cancer cells by up-regulating Notch and NF-kappaB signaling pathways. Breast Cancer Res Treat. 2011;126:15–25. doi: 10.1007/s10549-010-0883-2. [DOI] [PubMed] [Google Scholar]
- 35.Kong D, Banerjee S, Huang W, Li Y, Wang Z, Kim HR, et al. Mammalian target of rapamycin repression by 3,3′-diindolylmethane inhibits invasion and angiogenesis in platelet-derived growth factor-D-overexpressing PC3 cells. Cancer Res. 2008;68:1927–34. doi: 10.1158/0008-5472.CAN-07-3241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Wang Z, Kong D, Banerjee S, Li Y, Adsay NV, Abbruzzese J, et al. Down-regulation of platelet-derived growth factor-D inhibits cell growth and angiogenesis through inactivation of Notch-1 and nuclear factor-kappaB signaling. Cancer Res. 2007;67:11377–85. doi: 10.1158/0008-5472.CAN-07-2803. [DOI] [PubMed] [Google Scholar]
- 37.Okada A, Yaguchi T, Kanno T, Gotoh A, Nakano T, Nishizaki T. PDGF-D/PDGF-betabeta receptor-regulated chemotaxis of malignant mesothelioma cells. Cell Physiol Biochem. 2012;29:241–50. doi: 10.1159/000337605. [DOI] [PubMed] [Google Scholar]
- 38.Wang Z, Ahmad A, Li Y, Kong D, Azmi AS, Banerjee S, et al. Emerging roles of PDGF-D signaling pathway in tumor development and progression. Biochim Biophys Acta. 2010;1806:122–30. doi: 10.1016/j.bbcan.2010.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Wang Z, Kong D, Li Y, Sarkar FH. PDGF-D signaling: a novel target in cancer therapy. Curr Drug Targets. 2009;10:38–41. doi: 10.2174/138945009787122914. [DOI] [PubMed] [Google Scholar]
- 40.Yang S, Shin J, Park KH, Jeung HC, Rha SY, Noh SH, et al. Molecular basis of the differences between normal and tumor tissues of gastric cancer. Biochim Biophys Acta. 2007;1772:1033–40. doi: 10.1016/j.bbadis.2007.05.005. [DOI] [PubMed] [Google Scholar]
- 41.LaRochelle WJ, Jeffers M, Corvalan JR, Jia XC, Feng X, Vanegas S, et al. Platelet-derived growth factor D: tumorigenicity in mice and dysregulated expression in human cancer. Cancer Res. 2002;62:2468–73. [PubMed] [Google Scholar]
- 42.Floege J, Eitner F, Van Roeyen C, Ostendorf T. PDGF-D and renal disease: yet another one of those growth factors? J Am Soc Nephrol. 2003;14:2690–1. doi: 10.1097/01.asn.0000090831.40856.69. [DOI] [PubMed] [Google Scholar]
- 43.Fredriksson L, Li H, Eriksson U. The PDGF family: four gene products form five dimeric isoforms. Cytokine Growth Factor Rev. 2004;15:197–204. doi: 10.1016/j.cytogfr.2004.03.007. [DOI] [PubMed] [Google Scholar]
- 44.Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871–90. doi: 10.1016/j.cell.2009.11.007. [DOI] [PubMed] [Google Scholar]
- 45.Zlobec I, Lugli A. Epithelial mesenchymal transition and tumor budding in aggressive colorectal cancer: tumor budding as oncotarget. Oncotarget. 2010;1:651–61. doi: 10.18632/oncotarget.199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Wang Z, Li Y, Ahmad A, Azmi AS, Kong D, Banerjee S, et al. Targeting miRNAs involved in cancer stem cell and EMT regulation: An emerging concept in overcoming drug resistance. Drug Resist Updat. 2010;13:109–18. doi: 10.1016/j.drup.2010.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kang Y, Massague J. Epithelial-mesenchymal transitions: twist in development and metastasis. Cell. 2004;118:277–9. doi: 10.1016/j.cell.2004.07.011. [DOI] [PubMed] [Google Scholar]
- 48.Fuxe J, Vincent T, Garcia de Herreros A. Transcriptional crosstalk between TGF-beta and stem cell pathways in tumor cell invasion: role of EMT promoting Smad complexes. Cell Cycle. 2010;9:2363–74. doi: 10.4161/cc.9.12.12050. [DOI] [PubMed] [Google Scholar]
- 49.Grego-Bessa J, Diez J, Timmerman L, de la Pompa JL. Notch and epithelial-mesenchyme transition in development and tumor progression: another turn of the screw. Cell Cycle. 2004;3:718–21. [PubMed] [Google Scholar]
- 50.Bao B, Wang Z, Ali S, Kong D, Banerjee S, Ahmad A, et al. Over-expression of FoxM1 leads to epithelial-mesenchymal transition and cancer stem cell phenotype in pancreatic cancer cells. J Cell Biochem. 2011;112:2296–306. doi: 10.1002/jcb.23150. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 51.Maitah MY, Ali S, Ahmad A, Gadgeel S, Sarkar FH. Up-regulation of sonic hedgehog contributes to TGF-beta1-induced epithelial to mesenchymal transition in NSCLC cells. PLoS One. 2011;6:e16068. doi: 10.1371/journal.pone.0016068. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 52.Bjornsti MA, Houghton PJ. The TOR pathway: a target for cancer therapy. Nat Rev Cancer. 2004;4:335–48. doi: 10.1038/nrc1362. [DOI] [PubMed] [Google Scholar]
- 53.Sabatini DM. mTOR and cancer: insights into a complex relationship. Nat Rev Cancer. 2006;6:729–34. doi: 10.1038/nrc1974. [DOI] [PubMed] [Google Scholar]
- 54.Oh WJ, Jacinto E. mTOR complex 2 signaling and functions. Cell Cycle. 2011;10:2305–16. doi: 10.4161/cc.10.14.16586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Shor B, Gibbons JJ, Abraham RT, Yu K. Targeting mTOR globally in cancer: thinking beyond rapamycin. Cell Cycle. 2009;8:3831–7. doi: 10.4161/cc.8.23.10070. [DOI] [PubMed] [Google Scholar]
- 56.Zoncu R, Efeyan A, Sabatini DM. mTOR: from growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol. 2011;12:21–35. doi: 10.1038/nrm3025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Foster DA, Toschi A. Targeting mTOR with rapamycin: one dose does not fit all. Cell Cycle. 2009;8:1026–9. doi: 10.4161/cc.8.7.8044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Gulhati P, Bowen KA, Liu J, Stevens PD, Rychahou PG, Chen M, et al. mTORC1 and mTORC2 regulate EMT, motility, and metastasis of colorectal cancer via RhoA and Rac1 signaling pathways. Cancer Res. 2011;71:3246–56. doi: 10.1158/0008-5472.CAN-10-4058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Lamouille S, Connolly E, Smyth JW, Akhurst RJ, Derynck R. TGF-beta-induced activation of mTOR complex 2 drives epithelial-mesenchymal transition and cell invasion. J Cell Sci. 2012;125:1259–73. doi: 10.1242/jcs.095299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Radtke F, Raj K. The role of Notch in tumorigenesis: oncogene or tumour suppressor? Nat Rev Cancer. 2003;3:756–67. doi: 10.1038/nrc1186. [DOI] [PubMed] [Google Scholar]
- 61.Wang Z, Li Y, Sarkar FH. Notch signaling proteins: legitimate targets for cancer therapy. Curr Protein Pept Sci. 2010;11:398–408. doi: 10.2174/138920310791824039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ranganathan P, Weaver KL, Capobianco AJ. Notch signalling in solid tumours: a little bit of everything but not all the time. Nat Rev Cancer. 2011;11:338–51. doi: 10.1038/nrc3035. [DOI] [PubMed] [Google Scholar]
- 63.Efstratiadis A, Szabolcs M, Klinakis A. Notch, Myc and breast cancer. Cell Cycle. 2007;6:418–29. doi: 10.4161/cc.6.4.3838. [DOI] [PubMed] [Google Scholar]
- 64.Chen X, Stoeck A, Lee SJ, Shih Ie M, Wang MM, Wang TL. Jagged1 expression regulated by Notch3 and Wnt/beta-catenin signaling pathways in ovarian cancer. Oncotarget. 2010;1:210–8. doi: 10.18632/oncotarget.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Piazzi G, Fini L, Selgrad M, Garcia M, Daoud Y, Wex T, et al. Epigenetic regulation of Delta-Like1 controls Notch1 activation in gastric cancer. Oncotarget. 2011;2:1291–301. doi: 10.18632/oncotarget.414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Wang Z, Li Y, Kong D, Sarkar FH. The role of Notch signaling pathway in epithelial-mesenchymal transition (EMT) during development and tumor aggressiveness. Curr Drug Targets. 2010;11:745–51. doi: 10.2174/138945010791170860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Bao B, Wang Z, Ali S, Kong D, Li Y, Ahmad A, et al. Notch-1 induces epithelial-mesenchymal transition consistent with cancer stem cell phenotype in pancreatic cancer cells. Cancer Lett. 2011;307:26–36. doi: 10.1016/j.canlet.2011.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 68.Lu T, Stark GR. Cytokine overexpression and constitutive NFkappaB in cancer. Cell Cycle. 2004;3:1114–7. [PubMed] [Google Scholar]
- 69.Fraser CC. Exploring the positive and negative consequences of NF-kappaB inhibition for the treatment of human disease. Cell Cycle. 2006;5:1160–3. doi: 10.4161/cc.5.11.2773. [DOI] [PubMed] [Google Scholar]
- 70.Min C, Eddy SF, Sherr DH, Sonenshein GE. NF-kappaB and epithelial to mesenchymal transition of cancer. J Cell Biochem. 2008;104:733–44. doi: 10.1002/jcb.21695. [DOI] [PubMed] [Google Scholar]
- 71.Osipo C, Golde TE, Osborne BA, Miele LA. Off the beaten pathway: the complex cross talk between Notch and NF-kappaB. Lab Invest. 2008;88:11–7. doi: 10.1038/labinvest.3700700. [DOI] [PubMed] [Google Scholar]
- 72.Cory S, Adams JM. The Bcl2 family: regulators of the cellular life-or-death switch. Nat Rev Cancer. 2002;2:647–56. doi: 10.1038/nrc883. [DOI] [PubMed] [Google Scholar]
- 73.Quinn LM, Richardson H. Bcl-2 in cell cycle regulation. Cell Cycle. 2004;3:7–9. [PubMed] [Google Scholar]
- 74.Shroff EH, Snyder C, Chandel NS. Role of Bcl-2 family members in anoxia induced cell death. Cell Cycle. 2007;6:807–9. doi: 10.4161/cc.6.7.4044. [DOI] [PubMed] [Google Scholar]
- 75.Choi J, Choi K, Benveniste EN, Rho SB, Hong YS, Lee JH, et al. Bcl-2 promotes invasion and lung metastasis by inducing matrix metalloproteinase-2. Cancer Res. 2005;65:5554–60. doi: 10.1158/0008-5472.CAN-04-4570. [DOI] [PubMed] [Google Scholar]
- 76.Trisciuoglio D, Desideri M, Ciuffreda L, Mottolese M, Ribatti D, Vacca A, et al. Bcl-2 overexpression in melanoma cells increases tumor progression-associated properties and in vivo tumor growth. J Cell Physiol. 2005;205:414–21. doi: 10.1002/jcp.20413. [DOI] [PubMed] [Google Scholar]
- 77.Trisciuoglio D, Gabellini C, Desideri M, Ragazzoni Y, De Luca T, Ziparo E, et al. Involvement of BH4 domain of bcl-2 in the regulation of HIF-1-mediated VEGF expression in hypoxic tumor cells. Cell Death Differ. 2011;18:1024–35. doi: 10.1038/cdd.2010.175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Trisciuoglio D, Gabellini C, Desideri M, Ziparo E, Zupi G, Del Bufalo D. Bcl-2 regulates HIF-1alpha protein stabilization in hypoxic melanoma cells via the molecular chaperone HSP90. PLoS One. 2010;5:e11772. doi: 10.1371/journal.pone.0011772. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 79.Wang X, Belguise K, Kersual N, Kirsch KH, Mineva ND, Galtier F, et al. Oestrogen signalling inhibits invasive phenotype by repressing RelB and its target BCL2. Nat Cell Biol. 2007;9:470–8. doi: 10.1038/ncb1559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Sun T, Sun BC, Zhao XL, Zhao N, Dong XY, Che N, et al. Promotion of tumor cell metastasis and vasculogenic mimicry by way of transcription coactivation by Bcl-2 and Twist1: a study of hepatocellular carcinoma. Hepatology. 2011;54:1690–706. doi: 10.1002/hep.24543. [DOI] [PubMed] [Google Scholar]
- 81.Epstein RJ. The CXCL12-CXCR4 chemotactic pathway as a target of adjuvant breast cancer therapies. Nat Rev Cancer. 2004;4:901–9. doi: 10.1038/nrc1473. [DOI] [PubMed] [Google Scholar]
- 82.Orimo A, Weinberg RA. Stromal fibroblasts in cancer: a novel tumor-promoting cell type. Cell Cycle. 2006;5:1597–601. doi: 10.4161/cc.5.15.3112. [DOI] [PubMed] [Google Scholar]
- 83.Domanska UM, Kruizinga RC, Nagengast WB, Timmer-Bosscha H, Huls G, de Vries EG, et al. A review on CXCR4/CXCL12 axis in oncology: No place to hide. Eur J Cancer. 2012 doi: 10.1016/j.ejca.2012.05.005. [DOI] [PubMed] [Google Scholar]
- 84.Li X, Ma Q, Xu Q, Liu H, Lei J, Duan W, et al. SDF-1/CXCR4 signaling induces pancreatic cancer cell invasion and epithelial-mesenchymal transition in vitro through non-canonical activation of Hedgehog pathway. Cancer Lett. 2012;322:169–76. doi: 10.1016/j.canlet.2012.02.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Liu J, Liao S, Huang Y, Samuel R, Shi T, Naxerova K, et al. PDGF-D improves drug delivery and efficacy via vascular normalization, but promotes lymphatic metastasis by activating CXCR4 in breast cancer. Clin Cancer Res. 2011;17:3638–48. doi: 10.1158/1078-0432.CCR-10-2456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.van Kouwenhove M, Kedde M, Agami R. MicroRNA regulation by RNA-binding proteins and its implications for cancer. Nat Rev Cancer. 2011;11:644–56. doi: 10.1038/nrc3107. [DOI] [PubMed] [Google Scholar]
- 87.Medina PP, Slack FJ. microRNAs and cancer: an overview. Cell Cycle. 2008;7:2485–92. doi: 10.4161/cc.7.16.6453. [DOI] [PubMed] [Google Scholar]
- 88.Deng S, Calin GA, Croce CM, Coukos G, Zhang L. Mechanisms of microRNA deregulation in human cancer. Cell Cycle. 2008;7:2643–6. doi: 10.4161/cc.7.17.6597. [DOI] [PubMed] [Google Scholar]
- 89.Kasinski AL, Slack FJ. Epigenetics and genetics. MicroRNAs en route to the clinic: progress in validating and targeting microRNAs for cancer therapy. Nat Rev Cancer. 2011;11:849–64. doi: 10.1038/nrc3166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Yokobori T, Suzuki S, Tanaka N, Inose T, Sohda M, Sano A, et al. MiR-150 is associated with poor prognosis in esophageal squamous cell carcinoma via targeting the EMT inducers ZEB1. Cancer Sci. 2012 doi: 10.1111/cas.12030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Han M, Wang Y, Liu M, Bi X, Bao J, Zeng N, et al. MiR-21 regulates epithelial-mesenchymal transition phenotype and hypoxia-inducible factor-1alpha expression in third-sphere forming breast cancer stem cell-like cells. Cancer Sci. 2012;103:1058–64. doi: 10.1111/j.1349-7006.2012.02281.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Han M, Liu M, Wang Y, Chen X, Xu J, Sun Y, et al. Antagonism of miR-21 reverses epithelial-mesenchymal transition and cancer stem cell phenotype through AKT/ERK1/2 inactivation by targeting PTEN. PLoS One. 2012;7:e39520. doi: 10.1371/journal.pone.0039520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Cao M, Seike M, Soeno C, Mizutani H, Kitamura K, Minegishi Y, et al. MiR-23a regulates TGF-beta-induced epithelial-mesenchymal transition by targeting E-cadherin in lung cancer cells. Int J Oncol. 2012;41:869–75. doi: 10.3892/ijo.2012.1535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Scheel C, Weinberg RA. Cancer stem cells and epithelial-mesenchymal transition: Concepts and molecular links. Semin Cancer Biol. 2012;22:396–403. doi: 10.1016/j.semcancer.2012.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kong D, Li Y, Wang Z, Sarkar FH. Cancer Stem Cells and Epithelial-to-Mesenchymal Transition (EMT)-Phenotypic Cells: Are They Cousins or Twins? Cancers (Basel) 2011;3:716–29. doi: 10.3390/cancers30100716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133:704–15. doi: 10.1016/j.cell.2008.03.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Nguyen LV, Vanner R, Dirks P, Eaves CJ. Cancer stem cells: an evolving concept. Nat Rev Cancer. 2012;12:133–43. doi: 10.1038/nrc3184. [DOI] [PubMed] [Google Scholar]
- 98.Scheel C, Weinberg RA. Phenotypic plasticity and epithelial-mesenchymal transitions in cancer and normal stem cells? Int J Cancer. 2011;129:2310–4. doi: 10.1002/ijc.26311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Brabletz T. To differentiate or not--routes towards metastasis. Nat Rev Cancer. 2012;12:425–36. doi: 10.1038/nrc3265. [DOI] [PubMed] [Google Scholar]
- 100.Morel AP, Lievre M, Thomas C, Hinkal G, Ansieau S, Puisieux A. Generation of breast cancer stem cells through epithelial-mesenchymal transition. PLoS One. 2008;3:e2888. doi: 10.1371/journal.pone.0002888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Shah AN, Summy JM, Zhang J, Park SI, Parikh NU, Gallick GE. Development and characterization of gemcitabine-resistant pancreatic tumor cells. Ann Surg Oncol. 2007;14:3629–37. doi: 10.1245/s10434-007-9583-5. [DOI] [PubMed] [Google Scholar]
- 102.Brooks C, Sheu T, Bridges K, Mason K, Kuban D, Mathew P, et al. Preclinical evaluation of sunitinib, a multi-tyrosine kinase inhibitor, as a radiosensitizer for human prostate cancer. Radiat Oncol. 2012;7:154. doi: 10.1186/1748-717X-7-154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Glaser KB, Li J, Marcotte PA, Magoc TJ, Guo J, Reuter DR, et al. Preclinical characterization of ABT-348, a kinase inhibitor targeting the Aurora, VEGFR/PDGFR, and SRC kinase families. J Pharmacol Exp Ther. 2012 doi: 10.1124/jpet.112.197087. [DOI] [PubMed] [Google Scholar]
- 104.Taeger J, Moser C, Hellerbrand C, Mycielska ME, Glockzin G, Schlitt HJ, et al. Targeting FGFR/PDGFR/VEGFR impairs tumor growth, angiogenesis, and metastasis by effects on tumor cells, endothelial cells, and pericytes in pancreatic cancer. Mol Cancer Ther. 2011;10:2157–67. doi: 10.1158/1535-7163.MCT-11-0312. [DOI] [PubMed] [Google Scholar]
- 105.Zhou Y, Chen Y, Tong L, Xie H, Wen W, Zhang J, et al. AL3810, a multi-tyrosine kinase inhibitor, exhibits potent anti-angiogenic and anti-tumour activity via targeting VEGFR, FGFR and PDGFR. J Cell Mol Med. 2012;16:2321–30. doi: 10.1111/j.1582-4934.2012.01541.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Sanford M, Scott LJ. Imatinib: as adjuvant therapy for gastrointestinal stromal tumour. Drugs. 2010;70:1963–72. doi: 10.2165/11205000-000000000-00000. [DOI] [PubMed] [Google Scholar]
- 107.Andrae N, Kirches E, Hartig R, Haase D, Keilhoff G, Kalinski T, et al. Sunitinib targets PDGF-receptor and Flt3 and reduces survival and migration of human meningioma cells. Eur J Cancer. 2012;48:1831–41. doi: 10.1016/j.ejca.2012.01.032. [DOI] [PubMed] [Google Scholar]
- 108.Najy AJ, Jung YS, Won JJ, Conley-LaComb MK, Saliganan A, Kim CJ, et al. Cediranib inhibits both the intraosseous growth of PDGF D-positive prostate cancer cells and the associated bone reaction. Prostate. 2012;72:1328–38. doi: 10.1002/pros.22481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Seki N, Toh U, Kawaguchi K, Ninomiya M, Koketsu M, Watanabe K, et al. Tricin inhibits proliferation of human hepatic stellate cells in vitro by blocking tyrosine phosphorylation of PDGF receptor and its signaling pathways. J Cell Biochem. 2012;113:2346–55. doi: 10.1002/jcb.24107. [DOI] [PubMed] [Google Scholar]
- 110.Hawthorne T, Giot L, Blake L, Kuang B, Gerwien R, Smithson G, et al. A phase I study of CR002, a fully-human monoclonal antibody against platelet-derived growth factor-D. Int J Clin Pharmacol Ther. 2008;46:236–44. doi: 10.5414/cpp46236. [DOI] [PubMed] [Google Scholar]
- 111.Li Y, VandenBoom TG, 2nd, Kong D, Wang Z, Ali S, Philip PA, et al. Up-regulation of miR-200 and let-7 by natural agents leads to the reversal of epithelial-to-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells. Cancer Res. 2009;69:6704–12. doi: 10.1158/0008-5472.CAN-09-1298. [DOI] [PMC free article] [PubMed] [Google Scholar]