REVIEW ARTICLE

 

JOP. J Pancreas (Online) 2014 July 28; 15(4):289-285.

 

 

Microenvironmental Factors and Extracellular Matrix Degradation in Pancreatic Cancer

 

 

Marcelo G Binker1,2, Makena J Binker-Cosen2, Andres A Binker-Cosen2, Laura I Cosen-Binker1,2

 

 

1Department of Molecular and Cell Biology, School of Dental Medicine, Boston University, Boston, MA, USA

2CBRHC Research Center, Buenos Aires, Argentina

 

 

ABSTRACT

Pancreatic cancer is a devastating malady with proclivity for early metastasis, accounting for its poor prognosis. Pancreatic ductal adenocarcinoma, the most common type of pancreatic malignancy, exhibits an over-expression of several growth factors such as epidermal growth factor and transforming growth factor beta, which correlate with a decrease in patient survival. These growth factors as well as hypoxia-reoxygenation conditions have been shown to increase pancreatic tumor cell invasiveness. This review will focus on the signaling pathways used by these distinct microenvironmental factors to promote extracellular matrix degradation and invasion by pancreatic tumor cells.

 

 

Introduction

Pancreatic cancer is a devastating malady associated with poor survival and ranks as the fourth most common cause of cancer mortality. Currently, surgical resection with negative margins offers the only hope for cure, but a mere 10-20% of patients are suitable for resection at the time of diagnosis, which often occurs at advanced stages due to: a) the indolent growth of the tumors within the pancreas, b) the non-existence of truly effective conventional radiological tests to identify the disease at an early phase, c) the absence of specific and sensitive diagnostic serum markers, and d) the proclivity for early metastasis [1-3].

Interactions between cells of the stromal tissue and cancer cells are bidirectional, dynamic and complex. The non-malignant cells of stromal tissue produce a particular microenvironment that can modify the neoplastic properties of tumor cells, promoting tumor progression by providing growth factors, pro-angiogenic factors, proteases, and adhesion molecules that facilitate tumor cell proliferation, angiogenesis, invasion, and metastasis [4].

Pancreatic ductal adenocarcinoma (PDAC), the most common type of pancreatic malignancy, is characterized by a desmoplastic reaction consisting of an extensive and dense fibrotic stroma that surrounds and infiltrates clusters of malignant epithelial cells, together with the loss of basement membrane integrity and an abnormal vasculature. This fibrotic stroma is composed of connective tissue rich in collagen (types I and III) and fibronectin produced mainly by activated pancreatic stellate cells (PSC). Pancreatic cancer cells have been shown to activate PSC by paracrine mechanisms involving transforming growth factor-beta (TGF-β). However, after recurrent injury, PSC can retain its activated state by autocrine mechanisms as they can secrete cytokines and growth factors that stimulate receptors on their own cell surfaces [5].

Infiltrating immune cells are regularly present in tumors. The tumor-promoting inflammatory cells include macrophages, mast cells and neutrophils, as well as T and B lymphocytes [6, 7]. Amongst these infiltrating cells, mast cells can secrete several signaling molecules, including epidermal growth factor (EGF), which serve as effectors of their tumor-promoting actions [8]. Interaction between EGF and its receptor (EGFR) stimulates intrinsic activity of tyrosine kinase [9]. The EGFR signaling plays important roles in human cancers, enhancing tumor growth, invasion, motility, tumor spreading and metastasis. Over-expression of both EGF and EGFR is observed in pancreatic cancer and is linked with its development and poor prognosis [10-15].

Other growth factors, such as TGF-βs, derive from tumor cells and become sequestered within the stroma, which thus acts as a storage site. The invading cancer cells produce matrix metalloproteinases (MMPs) that release these growth factors [3]. The binding of TGF-β to its receptors activates many non-canonical signaling pathways, such as the phosphoinositol-3 kinase (PI3K), mitogen-activated protein kinase, and small guanosine triphosphatase (GTPase) pathways [16]. Remarkably, PDAC is a solid tumor that exhibits a notorious up-regulation of TGF-β isoforms, which correlates with a decrease in patient survival [17].

Solid tumors are complex structures presenting an abnormal vasculature that greatly contributes to the heterogeneous oxygenation of tumor cells. The irregular blood flow in these tumors during ischemia-reperfusion events is responsible for hypoxia and reoxygenation phases, also called intermittent hypoxia, which has been associated with both invasion and metastasis [18-21]. The periodicity in the detention of the blood flow is highly variable from one tumor to another as well as within the same tumor, depending on the architectural complexity and maturation level of the tumor vascular network [22, 23]. Studies of murine and human tumors have shown that the blood flow fluctuations observed in these tumors could vary from several minutes to more than one hour [22-24]. Hypoxia produces several changes in gene expression in normal and tumor cells [25]. This adaptive response to hypoxia is orchestrated by transcription factors such as hypoxia-inducible factor-1 (HIF-1), which induces the transcription of target genes involved in glycolysis and angiogenesis, thus allowing cells to adapt to hypoxia [26, 27]. Moreover, HIF-1alpha (HIF-1 α) also targets fascin, an actin-bundling protein that is overexpressed in PDAC and enhances motility, scattering, and invasiveness of pancreatic cancer cells [28, 29]. Interestingly, HIF-1 α has been shown to induce the conversion of non-stem pancreatic cancer cells into pancreatic cancer stem-like cells (CSLCs) [30]. The importance of CSLCs lies in their ability to self-renew, differentiate into heterogeneous types of tumor cells, and sustain tumor growth in vivo [31]. Consequently, CSLCs are responsible for tumor formation, progression, drug resistance, metastasis, and recurrence [31]. Patients, whose tumors express high levels of CSLCs markers such as CD44, have a significantly reduced median survival rate. Noteworthy, it was recently shown that the treatment of pancreatic cancer cell lines with an antibody anti-CD44 down-regulates the stem cell self-renewal genes Nanog, Sox-2, and Rex-1 and inhibits STAT-3-mediated cell proliferation and survival signaling [32]. Furthermore, anti-CD44 treatment also reduces growth, metastasis, and post-radiation recurrence of pancreatic xenograft tumors in mice [32]. Subpopulations of CSLCs have been identified from most tumors including pancreatic cancer, and targeting differentially expressed CSLC-related genes could result in better treatment outcomes of aggressive tumors. Indeed, FoxQ1 knock-down inhibits CSLCs aggressiveness and attenuates tumor formation and growth, and expression of CSLCs markers in the xenograft tumor derived from CSLCs of the moderately-to-poorly differentiated pancreatic adenocarcinoma cells MIA PaCa-2 [33].

The reactive oxygen species (ROS) generated during the reoxygenation periods are also important, since they modify gene expression through the regulation of the activity of some transcription factors, such as activator protein-1 (AP-1) or nuclear factor kappa B (NF-κB). While AP-1 is known to fulfill a pivotal role in tumorigenesis, regulating the expression and function of cell cycle regulators [34], NF-κB is a key player in tumor development through its ability to induce the transcription of genes coding for apoptosis inhibitor factors, proliferation molecules, pro-angiogenic factors, and extracellular matrix degrading enzymes [35].

Cancer metastasis involves tumor cells invading the surrounding tissue. Cell invasion is a complex process that can be defined as the migration of cells within a tissue in response to chemical signals, physical cues, and physicochemical processes. Invasive cells must traverse tissue barriers such as the basement membrane, comprised largely of type IV collagen. Remodeling of these tissue barriers depends on the ability of tumor cells to degrade the surrounding collagen matrix and then migrate through the matrix defects. Several proteolytic enzymes, including MMPs, seem to be implicated in this process. Among the MMP family, MMP-2 (gelatinase A, 72kDa type-IV collagenase) is a main character in the proteolytic degradation of basement membranes [36], being secreted by cells as a pro-enzyme and subsequently activated in the extracellular milieu to execute its proteolytic activity [36]. Although mmp-2 deletion produces mice with a relatively mild phenotype, exhibiting minor defects in developmental angiogenesis, tumor angiogenesis and tumor growth are highly reduced [37]. In humans, high expression and activation levels of this extracellular matrix (ECM)-degrading proteinase have been found in various cancer tissues [38-44]. In particular, high levels of activated MMP-2 were shown to correlate with tumor invasion and metastasis in pancreatic carcinoma [41, 42]. Although pre-clinical data supports the use of MMP inhibitors in cancer treatment, clinical trials involving these agents have rendered disappointing results, suggesting that the manipulation of MMPs to achieve tumor stasis may require altering the signal-transduction pathways that regulate the activity of MMPs rather than global inhibition [36]. Other proteolytic enzymes include urokinase-type plasminogen activator (u-PA), which is secreted as an inactive pro-enzyme and is activated by proteolytic cleavage. u-PA converts plasminogen to plasmin, which facilitates matrix degradation, and activates several MMPs [45, 46]. Activation of u-PA receptor (u-PAR) by u-PA binding results in activation of the Ras/extracellular signal regulated kinase (ERK) pathway, leading to cell proliferation, migration, and invasion [47, 48]. As for MMP-2, high levels of u-PA and u-PAR have been shown in pancreatic cancers [49], correlating with poor prognosis [50, 51].

A Common Signaling Pathway for Invasion

The small GTPases of the Rho family are molecular switches that control a broad variety of signal transduction pathways, regulating actin cytoskeleton, cell polarity, microtubule dynamics, membrane transport pathways and transcription factor activity [52]. Activation of the Ras-related C3 botulinum toxin substrate 1 (Rac1), a member of this family, has been shown to be detrimental not only during the reoxygenation posterior to hypoxia in non-tumor cells [53, 54], but also during the reperfusion that follows ischemia of tissues [55-57]. However, ‘beneficial’ effects of Rac1 activation were found in various tumor cells [58]. In the well characterized KrasG12D-induced murine model of pancreatic ductal adenocarcinoma, the pancreatic epithelial metaplasia is accompanied by apical-basolateral redistribution of F-actin, along with basal expression of Rac1 [59]. Notably, deletion of rac1 reduces formation of acinar-ductal metaplasia, pancreatic intraepithelial neoplasia and tumors [59].

The above mentioned growth factors EGF and TGF-β1, as well as the hypoxia-reoxygenation conditions, have been shown to increase tumor cell invasion through MMP-2 in various tumor cell types [60-66]. Additionally, our group established the requirement of Rac1 activation for these signals to stimulate secretion and activation of MMP-2, and cell invasion by the poorly and the well-to-moderately differentiated pancreatic adenocarcinoma cells PANC-1 and SW1990, respectively [62, 65, 66]. Mediators of Rac1 activation seem to be stimuli- and cell type-dependent. Accordingly, while EGF activates Rac1 through PI3K and Src [62], hypoxia-reperfusion conditions only stimulate PI3K in PANC-1 cells [66]. In concordance, the glial cell line-derived neurotrophic factor was shown to stimulate invasion via PI3K in PANC-1 cells [67]. In the well-to-moderately differentiated human pancreatic cancer cells CAPAN-2, TGF-β1 stimulates Rac1 activation also through PI3K (unpublished data). Other cell types such as MIA PaCa-2 cells and the moderately differentiated pancreatic adenocarcinoma cells BxPC-3 exhibit reduced invasiveness upon Src inhibition [68].

It is well established that Rac is the upstream signal protein for nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-dependent ROS generation [69]. Growth factors including EGF, TGF-β1, insulin-like growth factor I (IGF-I) and fibroblast growth factor-2 as well as hypoxia-reperfusion, are responsible for stimulating the production of ROS in PANC-1, SW1990 and MIA PaCa-2 cells through non-mitochondrial NADPH oxidase, the major source of ROS induced by growth factors in these tumor cells [62, 65, 66, 70]. In addition to being a pro-survival, anti-apoptotic factor in pancreatic cancer cells [70], ROS mediate invasion by various pancreatic cancer cells, including PANC-1, SW1990 and BxPC-3 [62, 65, 66, 71, 72]. Moreover, our studies show NADPH oxidase-generated ROS as the downstream signal for Rac1-mediated pancreatic cancer cells’ invasion [62, 65, 66].

Pathways triggered by ROS in tumor invasion include MAPK signaling (JNK, ERK, p38) and regulation of transcription factors (AP-1, Ets-1, NF-κB) that lie upstream of MMPs and u-PA [28, 34, 35, 73, 74]. Transfection with an antibody against RelA (p65), a NF-κB subunit, not only decreases expression and bioactivity of MMP-9 and u-PA, but also suppresses invasion and angiogenesis in glioma cells [75]. SW1990 cells exhibit a similar behavior, since TGF-β1-induced MMP-2 secretion and activation, and cell invasion depend on NF-κB activity [65]. Interestingly, we observed that TGF-β1-stimulated CAPAN-2 cells increased the expression of u-PA and cell invasiveness, which were mediated by Rac1/ROS activation of NF-κB (unpublished data). Furthermore, TGF-β1 was shown to increase both u-PA and invasiveness of responsive pancreatic cancer cell lines PANC-1, IMIM-PC1 and SW1990 [63]. However, TGF-β1 failed to augment invasiveness in non-responsive cell lines CAPAN-1, MIA Paca-2 and IMIM-PC2 [63]. Other growth factors positively regulating invasiveness through the induction of u-PA and u-PAR include IGF-I and hepatocyte growth factor [49, 76, 77].

In summary, the experimental data available support a sequential model that explains increased invasiveness by pancreatic cancer cells ranging from poorly to well-to-moderately differentiated types (Figure 1). In this hypothetical mechanistic model, different signals such as growth factors and hypoxia-reoxygenation conditions promote invasion by pancreatic cancer cells through a common Rac1/ROS/NF-κB dependent pathway. Mediators of Rac1 activation as well as proteolytic enzyme effectors seem to be cell type dependent. While PI3K and Src appear as the intermediate molecules for Rac1activation, MMP-2 and u-PA emerge as the main ECM-degrading proteins in pancreatic cancer invasion.

 

 

Figure 1. Model for Rac1/ROS/NF-κB-mediated pancreatic cancer invasion. Distinct signals such as growth factors and hypoxia-reoxygenation conditions activate PI3K and/or Src, which alone or combined switch on Rac1 in pancreatic cancer cells. Through NADPH oxidase, activated Rac1 induces formation of ROS. Once generated, these intermediate molecules activate NF-κB, which mediates secretion and activation of MMP-2 and/or u-PA in the extracellular milieu. These proteolytic enzymes are responsible for degradation of the basement membrane, promoting invasion by pancreatic cancer cells. For this common signaling pathway, mediators of Rac1 activation as well as proteolytic enzyme effectors seem to be cell type dependent.

 

 

Future Directions

The exact cell type that gives origin to PDAC is not known. However, it is supposed to arise from a poorly differentiated ductal cell, a de-differentiated acinar or islet cell, a progenitor cell, or a stem cell [78]. While most of the studies on pancreatic cancer invasion are focused on single stimulus, the influence of concomitant multifactors on pancreatic cancer progression needs to be deciphered. Additionally, co-culture of tumor and non-tumor cells requires to be further explored in order to properly address microenvironment cell interactions.

 

 

Received March 28th, 2014 – Accepted May 29th, 2014

Key words Carcinoma, Pancreatic Ductal; Environmental Medicine; Extracellular Matrix; Matrix Metalloproteinases; Neoplasm Invasiveness; Neoplastic Stem Cells; NF-kappa B; RAC1 protein, human; Reactive Oxygen Species; Urokinase-Type Plasminogen Activator

Abbreviations AP-1, activator protein-1; CSLC, cancer stem-like cells; ECM, extracellular matrix; EGF, epidermal growth factor; ERK, extracellular signal regulated kinase; GTPase, guanosine triphosphatase; HIF-1, hypoxia-inducible factor-1; IGF-I, insulin-like growth factor I; MMP, matrix metalloproteinase; NADPH, nicotinamide adenine dinucleotide phosphate; NF-κB, nuclear factor-kappa beta; PDAC, pancreatic ductal adenocarcinoma; PSC, pancreatic stellate cells; PI3K, phosphatidylinositol 3-kinase; ROS, reactive oxygen species; TGF-β, transforming growth factor-beta; u-PA, urokinase-type plasminogen activator.

Acknowledgement Supported by KB & Associates Representing Certification International Limited (UK).

Conflict of Interest The authors hereby state that there is no conflict of interest to disclose.

Correspondence
Laura Cosen-Binker
72 East Concord Street
Evans Building - Room 408
Department of Molecular and Cell Biology
Henry M. Goldman School of Dental Medicine
Boston University
Boston, MA 02118
USA
Phone: +1-617.414.1043
E-mail: licb@bu.edu

 

 

References

1. Raimondi S, Maisonneuve P, Lowenfels AB. Epidemiology of pancreatic cancer: an overview. Nat Rev Gastroenterol Hepatol 2009; 6:699-708. [PMID:19806144].

2. Collins A, Bloomston M. Diagnosis and management of pancreatic cancer. Minerva Gastroenterol Dietol 2009; 55:445-54. [PMID:19942828].

3. Korc M. Pancreatic cancer-associated stroma production. Am J Surg 2007; 194:S84-6. [PMID:17903452].

4. Spano D, Zollo M. Tumor microenvironment: a main actor in the metastasis process. Clin Exp Metastasis 2012; 29:381-95. [PMID:22322279].

5. Wilson JS, Pirola RC, Apte MV. Stars and stripes in pancreatic cancer: role of stellate cells and stroma in cancer progression. Front Physiol 2014; 5:52. [PMID:24592240].

6. Coffelt SB, Lewis CE, Naldini L, Brown JM, Ferrara N, De Palma M. Elusive identities and overlapping phenotypes of proangiogenic myeloid cells in tumors. Am J Pathol 2010; 176:1564-76. [PMID:20167863].

7. DeNardo DG, Andreu P, Coussens LM. Interactions between lymphocytes and myeloid cells regulate pro- versus anti-tumor immunity. Cancer Metastasis Rev 2010; 29:309-16. [PMID:20405169].

8. Theoharides TC, Alysandratos KD, Angelidou A, Zhang B. Mast Cells and Tumor Microenvironment. In: Bagley RG, ed. The Tumor Microenvironment, Cancer Drug Discovery and Development. Chapter 17. Springer Science+Business Media, 2010:353-70.

9. Carpenter G, Cohen S. Epidermal growth factor. J Biol Chem 1990; 265:7709-12. [PMID:2186024].

10. Ciardiello F, Tortora G. EGFR antagonists in cancer treatment. N Engl J Med 2008; 358:1160-74. [PMID:18337605].

11. Mimeault M, Brand RE, Sasson AA, Batra SK. Recent advances on the molecular mechanisms involved in pancreatic cancer progression and therapies. Pancreas 2005; 31:301-16. [PMID:16258363].

12. Wang Z, Sengupta R, Banerjee S, Li Y, Zhang Y, Rahman KM, et al. Epidermal growth factor receptor-related protein inhibits cell growth and invasion in pancreatic cancer. Cancer Res 2006; 66:7653-60. [PMID:].

13. Uegaki K, Nio Y, Inoue Y, Minari Y, Sato Y, Song MM, et al. Clinicopathological significance of epidermal growth factor and its receptor in human pancreatic cancer. Anticancer Res 1997; 17:3841-7. [PMID:16885366].

14. Yeh TS, Jan YY, Chiu CT, Ho YB, Chen TC, Lee KF, et al. Characterisation of oestrogen receptor, progesterone receptor, trefoil factor 1, and epidermal growth factor and its receptor in pancreatic cystic neoplasms and pancreatic ductal adenocarcinoma. Gut 2002; 51:712-6. [PMID:12377812].

15. Pryczynicz A, Guzińska-Ustymowicz K, Kemona A, Czyzewska J. Expression of EGF and EGFR strongly correlates with metastasis of pancreatic ductal carcinoma. Anticancer Res 2008; 28:1399-404. [PMID:18505086].

16. Massague J. TGFbeta in Cancer. Cell 2008; 134:215-30. [PMID:18662538].

17. Friess H, Yamanaka Y, Büchler MW, Korc M. Enhanced expression of transforming growth factor-beta isoforms in human pancreatic cancer correlates with decreased survival. Gastroenterology 1993; 105:1846-56. [PMID:8253361].

18. Doi K, Horiuchi T, Uchinami M, Tabo T, Kimura N, Yokomachi J, et al. Hepatic ischemia-reperfusion promotes liver metastasis of colon cancer. J Surg Res 2002; 105:243-7. [PMID:12121713].

19. Man K, Ng KT, Lo CM, Ho JW, Sun BS, Sun CK, et al. Ischemia-reperfusion of small liver remnant promotes liver tumor growth and metastases--activation of cell invasion and migration pathways. Liver Transpl 2007; 13:1669-77. [PMID:18044786].

20. Nicoud IB, Jones CM, Pierce JM, Earl TM, Matrisian LM, Chari RS, Gorden DL. Warm hepatic ischemia-reperfusion promotes growth of colorectal carcinoma micrometastases in mouse liver via matrix metalloproteinase-9 induction. Cancer Res 2007; 67:2720-8. [PMID:17363593].

21. Rofstad EK. Microenvironment-induced cancer metastasis. Int J Radiat Biol 2000; 76:589-605. [PMID:10866281].

22. Chaplin DJ, Hill SA. Temporal heterogeneity in microregional erythrocyte flux in experimental solid tumours. Br J Cancer 1995; 71:1210-3. [PMID:].

23. Bennewith KL, Durand RE. Quantifying transient hypoxia in human tumor xenografts by flow cytometry. Cancer Res 2004; 64:6183-9. [PMID:7779713].

24. Braun RD, Lanzen JL, Dewhirst MW. Fourier analysis of fluctuations of oxygen tension and blood flow in R3230Ac tumors and muscle in rats. Am J Physiol 1999; 277:H551-68. [PMID:10444480].

25. Chi JT, Wang Z, Nuyten DS, Rodriguez EH, Schaner ME, Salim A, et al. Gene expression programs in response to hypoxia: cell type specificity and prognostic significance in human cancers. PLoS Med 2006; 3:e47. [PMID:16417408].

26. Jiang BH, Rue E, Wang GL, Roe R, Semenza GL. Dimerization, DNA binding, and transactivation properties of hypoxia-inducible factor 1. J Biol Chem 1996; 271:17771-8. [PMID:8663540].

27. Wenger RH. Mammalian oxygen sensing, signaling and gene regulation. J Exp Biol 2000; 203:1253-63. [PMID:10729275].

28. Zhao X, Gao S, Ren H, Sun W, Zhang H, Sun J, et al. Hypoxia-inducible factor-1 promotes pancreatic ductal adenocarcinoma invasion and metastasis by activating transcription of the actin-bundling protein fascin. Cancer Res 2014; 74:2455-64. [PMID:24599125].

29. Xu YF, Yu SN, Lu ZH, Liu JP, Chen J. Fascin promotes the motility and invasiveness of pancreatic cancer cells. World J Gastroenterol 2011; 17:4470-8. [PMID:22110277].

30. Zhu H, Wang D, Liu Y, Su Z, Zhang L, Chen F, et al. Role of the Hypoxia-inducible factor-1 alpha induced autophagy in the conversion of non-stem pancreatic cancer cells into CD133+ pancreatic cancer stem-like cells. Cancer Cell Int 2013; 13:119. [PMID:24505593].

31. Herreros-Villanueva M, Bujanda L, Billadeau DD, Zhang JS. Embryonic stem cell factors and pancreatic cancer. World J Gastroenterol 2014; 20:2247-54. [PMID:24605024].

32. Li L, Hao X, Qin J, Tang W, He F, Smith A, et al. Antibody Against CD44s Inhibits Pancreatic Tumor Initiation and Postradiation Recurrence in Mice. Gastroenterology 2014; 146:1108-18. [PMID:24397969].

33. Bao B, Azmi A, Aboukameel A, Ahmad A, Bolling-Fischer A, Sethi S, et al. Pancreatic cancer stem-like cells display aggressive behavior mediated via activation of FoxQ1. J Biol Chem 2014. [PMID:24719318].

34. Matthews CP, Colburn NH, Young MR. AP-1 a target for cancer prevention. Curr Cancer Drug Targets 2007; 7:317-24. [PMID:17979626].

35. Karin M, Cao Y, Greten FR, Li ZW. NF-kappaB in cancer: from innocent bystander to major culprit. Nat Rev Cancer 2002; 2:301-10. [PMID:12001991].

36. Björklund M, Koivunen . Gelatinase-mediated migration and invasion of cancer cells, Biochim Biophys Acta 2005; 1755:37-69. [PMID:15907591].

37. Itoh T, Tanioka M, Yoshida H, Yoshioka T, Nishimoto H, Itohara S. Reduced angiogenesis and tumor progression in gelatinase A-deficient mice. Cancer Res 1998; 58:1048-51. [PMID:9500469].

38. Yamagata S, Yoshii Y, Suh JG, Tanaka R, Shimizu S. Occurrence of an active form of gelatinase in human gastric and colorectal carcinoma tissues. Cancer Lett 1991; 59:51-5. [PMID:1652352].

39. Brown PD, Bloxidge RE, Stuart NS, Gatter KC, Carmichael J. Association between expression of activated 72-kilodalton gelatinase and tumor spread in non-small-cell lung carcinoma. J Natl Cancer Inst 1993; 85:574-8. [PMID:8384265].

40. Gress TM, Muller-Pillasch F, Lerch MM, Friess H, Büchler M, Adler G. Expression and in situ localization of genes coding for extracellular matrix proteins and extracellular matrix degrading proteases in pancreatic cancer. Int J Cancer 1995; 62:407-13. [PMID:7635566].

41. Koshiba T, Hosotani R, Wada M, Miyamoto Y, Fujimoto K, Lee JU, et al. Involvement of matrix metalloproteinase-2 activity in invasion and metastasis of pancreatic carcinoma. Cancer 1998; 82:642-50. [PMID:9477095].

42. Ellenrieder V, Alber B, Lacher U, Hendler SF, Menke A, Boeck W, et al. Role of MT-MMPs and MMP-2 in pancreatic cancer progression. Int J Cancer 2000; 85:14-20. [PMID:10585576].

43. Rauvala M, Aglund K, Puistola U, Turpeenniemi-Hujanen T, Horvath G, Willén R, Stendahl U. Matrix metalloproteinases-2 and -9 in cervical cancer: different roles in tumor progression. Int J Gynecol Cancer 2006; 16:1297-302. [PMID:16803520].

44. Jinga DC, Blidaru A, Condrea I, Ardeleanu C, Dragomir C, Szegli G, et al. MMP-9 and MMP-2 gelatinases and TIMP-1 and TIMP-2 inhibitors in breast cancer: correlations with prognostic factors. J Cell Mol Med 2006; 10:499-510. [PMID:16796815].

45. Nguyen DH, Hussaini IM, Gonias SL. Binding of urokinase-type plasminogen activator to its receptor in MCF-7 cells activates extracellular signal-regulated kinase 1 and 2 which is required for increased cellular motility. J Biol Chem 1998; 273:8502-7. [PMID:9525964].

46. Aguirre Ghiso JA, Kovalski K, Ossowski L. Tumor dormancy induced by down-regulation of urokinase receptor in human carcinoma involves integrin and MAPK signaling. J Cell Biol 1999; 147:89-104. [PMID:10508858].

47. Mignatti P, Rifkin DB. Plasminogen activators and matrix metalloproteinases in angiogenesis. Enzyme Protein 1996; 49:117-37. [PMID:8797002].

48. Carmeliet P, Moons L, Dewerchin M, Rosenberg S, Herbert JM, Lupu F, Collen D. Receptor-independent role of urokinase-type plasminogen activator in pericellular plasmin and matrix metalloproteinase proteolysis during vascular wound healing in mice. J Cell Biol 1998; 140:233-45. [PMID:9425170].

49. Paciucci R, Vila MR, Adell T, Díaz VM, Torà M, Nakamura T, Real FX. Activation of the urokinase plasminogen activator/urokinase plasminogen activator receptor system and redistribution of E-cadherin are associated with hepatocyte growth factor-induced motility of pancreas tumor cells overexpressing Met. Am J Pathol 1998; 153:201-12. [PMID:9665481].

50. Takeuchi Y, Nakao A, Harada A, Nonami T, Fukatsu T, Takagi H. Expression of plasminogen activators and their inhibitors in human pancreatic carcinoma: immunohistochemical study. Am J Gastroenterol 1993; 88:1928-33. [PMID:8237943].

51. Cantero D, Friess H, Deflorin J, Zimmermann A, Bründler MA, Riesle E, et al. Enhanced expression of urokinase plasminogen activator and its receptor in pancreatic carcinoma. Br J Cancer 1997; 75:388-95. [PMID:9020484].

52. Etienne-Manneville S, Hall A. Rho GTPases in cell biology. Nature 2002; 420:629-35. [PMID:12478284].

53. Kim KS, Takeda K, Sethi R, Pracyk JB, Tanaka K, Zhou YF, et al. Protection from reoxygenation injury by inhibition of rac1. J Clin Invest 1998; 101:1821-6. [PMID:9576744].

54. Ng CK, Deshpande SS, Irani K, Alevriadou BR. Adhesion of flowing monocytes to hypoxia-reoxygenation-exposed endothelial cells: role of Rac1, ROS, and VCAM-1. Am J Physiol Cell Physiol 2002; 283:93-102. [PMID:12055077].

55. Ozaki M, Deshpande SS, Angkeow P, Bellan J, Lowenstein CJ, Dinauer MC, et al. Inhibition of the Rac1 GTPase protects against nonlethal ischemia/reperfusion-induced necrosis and apoptosis in vivo. FASEB J 2000; 14:418-29. [PMID:10657998].

56. Ozaki M, Deshpande S, Angkeow P, Bellan J, Lowenstein CJ, Dinauer MC, et al. Targeted inhibition of the small GTPase protects against ischemia/reperfusion liver injury in mice, Transplant Proc 2001; 33:863-4. [PMID:11267105].

57. Harada N, Iimuro Y, Nitta T, Yoshida M, Uchinami H, Nishio T, et al. Inactivation of the small GTPase Rac1 protects the liver from ischemia/reperfusion injury in the rat. Surgery 2003; 134:480-91. [PMID:14555937].

58. Bosco EE, Mulloy JC, Zheng Y. Rac1 GTPase: a "Rac" of all trades. Cell Mol Life Sci 2009; 66:370-4. [PMID:19151919].

59. Heid I, Lubeseder-Martellato C, Sipos B, Mazur PK, Lesina M, Schmid RM, Siveke JT. Early requirement of Rac1 in a mouse model of pancreatic cancer. Gastroenterology 2001; 141:719-30. [PMID:21684285].

60. Yeh MW, Rougier JP, Park JW, Duh QY, Wong M, Werb Z, Clark OH. Differentiated thyroid cancer cell invasion is regulated through epidermal growth factor receptor-dependent activation of matrix metalloproteinase (MMP)-2/gelatinase A. Endocr Relat Cancer 2006; 13:1173-83. [PMID:17158762].

61. Staun-Ram E, Goldman S, Gabarin D, Shalev E. Expression and importance of matrix metalloproteinase 2 and 9 (MMP-2 and -9) in human trophoblast invasion. Reprod Biol Endocrinol 2004; 2:59-72. [PMID:15294019].

62. Binker MG, Binker-Cosen AA, Richards D, Oliver B, Cosen-Binker L. EGF promotes invasion by PANC-1 cells through Rac1/ROS-dependent secretion and activation of MMP-2. Biochem Biophys Res Commun 2009; 379:445-50. [PMID:19116140].

63. Ellenrieder V, Hendler SF, Ruhland C, Boeck W, Adler G, Gress TM. TGF-beta-induced invasiveness of pancreatic cancer cells is mediated by matrix metalloproteinase-2 and the urokinase plasminogen activator system. Int J Cancer. 2001; 93:204-11. [PMID:11410867].

64. Teraoka H, Sawada T, Yamashita Y, Nakata B, Ohira M, Ishikawa T, et al. TGF-beta1 promotes liver metastasis of pancreatic cancer by modulating the capacity of cellular invasion. Int J Oncol 2001; 19:709-15. [PMID:11562745].

65. Binker MG, Binker-Cosen AA, Gaisano HY, de Cosen RH, Cosen-Binker LI. TGF-β1 increases invasiveness of SW1990 cells through Rac1/ROS/NF-κB/IL-6/MMP-2. Biochem Biophys Res Commun 2011; 405:140-5. [PMID:21219858].

66. Binker MG, Binker-Cosen AA, Richards D, Gaisano HY, de Cosen RH, Cosen-Binker LI. Hypoxia-reoxygenation increase invasiveness of PANC-1 cells through Rac1/MMP-2. Biochem Biophys Res Commun 2010; 393:371-6. [PMID:20153729].

67. Veit C, Genze F, Menke A, Hoeffert S, Gress TM, Gierschik P, Giehl K. Activation of phosphatidylinositol 3-kinase and extracellular signal-regulated kinase is required for glial cell line-derived neurotrophic factor-induced migration and invasion of pancreatic carcinoma cells Cancer Res 2004; 64:5291-300. [PMID:15289335].

68. Ito H, Gardner-Thorpe J, Zinner MJ, Ashley SW, Whang EE. Inhibition of tyrosine kinase Src suppresses pancreatic cancer invasiveness. Surgery 2003; 134:221-6. [PMID:12947321].

69. Wu WS. The signaling mechanism of ROS in tumor progression. Cancer Metastasis Rev 2006; 25:695-705. [PMID:17160708].

70. Vaquero EC, Edderkaoui M, Pandol SJ, Gukovsky I, Gukovskaya AS Reactive oxygen species produced by NAD(P)H oxidase inhibit apoptosis in pancreatic cancer cells. J Biol Chem 2004; 279:34643-54. [PMID:15155719].

71. Liu J, Ben QW, Yao WY, Zhang JJ, Chen DF, He XY, et al. BMP2 induces PANC-1 cell invasion by MMP-2 overexpression through ROS and ERK. Front Biosci (Landmark Ed) 2012; 17:2541-9. [PMID:22652796].

72. Li W, Ma Q, Li J, Guo K, Liu H, Han L, Ma G. Hyperglycemia enhances the invasive and migratory activity of pancreatic cancer cells via hydrogen peroxide. Oncol Rep 2011; 25:1279-87. [PMID:21249318].

73. Kim MH, Yoo HS, Kim MY, Jang HJ, Baek MK, Kim HR, et al. Helicobacter pylori stimulates urokinase plasminogen activator receptor expression and cell invasiveness through reactive oxygen species and NF-kappaB signaling in human gastric carcinoma cells. Int J Mol Med 2007; 19:689-97. [PMID:17334646].

74. Tobar N, Villar V, Santibanez JF. ROS-NFkappaB mediates TGF-beta1-induced expression of urokinase-type plasminogen activator, matrix metalloproteinase-9 and cell invasion. Mol. Cell. Biochem 2010; 340:195-202. [PMID:20204677].

75. Li L, Gondi CS, Dinh DH, Olivero WC, Gujrati M, Rao JS. Transfection with anti p65 intrabody suppresses invasion and angiogenesis in glioma cells by blocking nuclear factor-kappaB transcriptional activity. Clin Cancer Res 2007; 13:2178-90. [PMID:17404102].

76. Tanno S, Mitsuuchi Y, Altomare DA, Xiao GH, Testa JR. AKT activation up-regulates insulin-like growth factor I receptor expression and promotes invasiveness of human pancreatic cancer cells. Cancer Res 2001; 61:589-93. [PMID:11212254].

77. Bauer TW, Liu W, Fan F, Camp ER, Yang A, Somcio RJ, Bucana et al. Targeting of urokinase plasminogen activator receptor in human pancreatic carcinoma cells inhibits c-Met- and insulin-like growth factor-I receptor-mediated migration and invasion and orthotopic tumor growth in mice. Cancer Res 2005; 65:7775-81. [PMID:16140945].

78. Hezel AF, Kimmelman AC, Stanger BZ, Bardeesy N, DePinho RA. Genetics and biology of pancreatic ductal adenocarcinoma. Genes & Dev 2006; 20:1218-49. [PMID:16702400].