Open access

MicroRNAs in Invasion and Metastasis in Lung Cancer

Written By

Lili Jiang and Xueshan Qiu

Submitted: 18 February 2012 Published: 13 March 2013

DOI: 10.5772/55624

From the Edited Volume

Oncogenomics and Cancer Proteomics - Novel Approaches in Biomarkers Discovery and Therapeutic Targets in Cancer

Edited by César López-Camarillo and Elena Aréchaga-Ocampo

Chapter metrics overview

4,025 Chapter Downloads

View Full Metrics

1. Introduction

Despite advances in diagnosis and treatment, the morbidity and mortality of lung cancer remains to mount up. The key factor of cancer associated morbidity and mortality is principally attributable to the development of metastases. Cancer cells depart their normal microenvironment from the primary tumor site through complicated and multistep processes disseminate and colonize distant organs [1]. However, the cellular and molecular machinery underlying metastasis is relatively poorly understood so far. In order to resist cancer dissemination, more effective therapeutic strategies are clearly required.

Cellular migration and invasion mechanism are commonly thought to be associated with Rho family GTPases [2-4], JAK-STAT [5-7], MAPK [8-10], Wnt [11-13], Notch pathway [14-16]. Recently, epithelial–mesenchymal transition (EMT) programs have become the focus of the mechanism of metastasis [1, 17-20]. EMT is an embryologically conserved genetic program by which epithelial cells down regulate intercellular tight junctions, loose polarity, express mesenchymal markers, and manifest a migratory phenotype [1]. In the EMT process, Rho family GTPases [21], JAK-STAT [22], MAPK [23], Wnt [24] and Notch [25] pathways may also play an important role. In recent years, emerging studies have highlighted the critical role of these pathways and their regulation by microRNAs (miRNAs) in cancer invasion and metastasis.

MiRNAs, short (18-24 nucleotides) non-coding RNAs, are derived from long transcripts pri-miRNAs and pre-miRNAs [26-30].By targeting 3’ untranslated regions (3’UTRs) of cognate mRNAs, miRNAs post-transcriptionally regulate gene expression and induce translational repression [29, 30]. Their specificity is determined by nucleotides 2–8 at the 5′ end, termed the miRNA “seed sequence”. To date, 1527 human miRNAs have been identified (Sanger miRBase 18 http://www.miRbase.org/index.shtml), forming less than 1% of all human genes, potentially regulating more than 10% of all protein coding genes [1]. Recently, miRNAs have been discovered to play important roles in the invasion and metastasis of malignant tumors. [31-33]. Understanding specific characteristics of miRNAs would probably serve as predictive markers and as therapeutic strategies for patients with metastasis.

In light of these recent discoveries, the present article discusses how invasion and EMT pathways are regulated by miRNAs. We have classified invasion programs and key proteins involved in EMT according to the signaling pathway showed above and point out validated miRNAs regulating their expression and highlight critical knowledge gaps that remain to be addressed to enable improved understanding of the molecular mechanisms behind EMT and metastasis. A list of experimentally validated miRNAs regulating key proteins involved in invasion–metastasis programs or participating in some principal pathways can be found in Figure 1.

Figure 1.

The experimentally validated miRNAs regulate key proteins involved in invasion–metastasis programs or participating in some principal pathways.

Advertisement

2. Rho family of GTPases

The Rho family of GTPases, a family of small (~21 kDa) signalingG protein, is a subfamily of the Ras superfamily [34]. In mammals, the Rho family is made of 20 members distributed into eight subfamilies: Rho, Rac, Cdc42, Rnd, RhoU/V, RhoBTB, RhoH and RhoD/F. Almost all research involves the three most common members of the Rho family: Cdc42, Rac1 and RhoA [35]. Over expression of Rho GTPases is associated with reorganization of actin cytoskeleton, which plays an important role in cell migration, invasion and metastasis that are important aspects of cancer progression [36].

Emerging studies have indicated that miRNAs participate in the Rho GTPasessignaling pathway. Among the tested miRNAs,the present articles demonstrated that miR-155, miR-185, miR-31 and miR-133a are associated with RhoA in cell migration and invasion. MiR-155 may play an important role in TGF-β-induced EMT and cell migration and invasion by targeting RhoA [37]. MiR-185 is a negative regulator of RhoA and Cdc42, and could inhibit proliferation and invasion of colorectal cancer cells [38]. The Effects of miR-31 on metastasis may be associated with concurrent suppression of integrin alpha5, radixin, and RhoAphenocopies [39]. Chiba and his colleagues reported that RhoA expression is negatively regulated by miR-133a in bronchial smooth muscle cells [40].

Moreover, some studies discussed the regulation of cell migration and invasion by miRNA may be attribute to Rho-associated serine-threonine protein kinase (ROCK), one of the best characterized downstream effectors of Rho, that is activated when it selectively binds to the active GTP-bound form of Rho [41, 42]. As with Rho, ROCK has been implicated in altering cell migration and invasion during tumor cell metastasis [43, 44]. Yu and his colleagues indicate that downregulation of miR-205 resulted in an increase in Rho-ROCKI activity, phosphorylation of the actin severing protein cofilin, and a corresponding diminution of filamentous actin [45].

A number of articles reported that some miRNAs regulate cell migration and invasion by targeting Rac and Cdc42. Recently,microRNA-142-3p, a new regulator of Rac1, suppresses the migration and invasion of hepatocellular carcinoma cells [46]. The regulation of cancer cell migration by MiR-10bmay be attribute to activate Rac by targets Tiam1 [47]. MiR-151 exerts this function by directly targeting RhoGDIA, a putative metastasis suppressor in hepatocellular carcinoma (HCC), thus leading to the activation of Rac1, Cdc42 and Rho GTPases [48]. Liu and his colleagues have found that miR-137 may have a tumor suppressor function by directly targeting Cdc42 to inhibit the proliferation and invasion activities of colorectal cancer cells [49, 50]. MiR-206 may suppress invasion and migration of MDA-MB-231 cells in vitro partly via regulating actin cytoskeleton remodelling by downregulating Cdc42 [51]. MiR-29 activates p53 by targeting p85 alpha and Cdc42 [52].

In addition, MiR-21 targets the tumor suppressor Rho B and regulates proliferation, invasion positively in colorectal cancer cells [53, 54]. Jiang and his colleagues have indicated that miR-138 plays an important role in tongue squamous cell carcinoma cell migration and invasion by concurrently targeting Rho C and ROCK2 [36]. Studies on the association of Rho with miRNAs highlight the importance of miRNAs in invasion and metastasis of malignant tumors.

Advertisement

3. JAK-STAT

The JAK-STAT signaling pathway transmits information from chemical signals outside the cell, through the cell membrane, and into gene promoters on the DNA in the cell nucleus, which causes DNA transcription and activity in the cell. JAK, short for Janus Kinase, is a family of intracellular, nonreceptor tyrosine kinases that transduce cytokine-mediated signals via the JAK-STAT pathway. As a key component of the JAK/STAT pathway, Signal Transducer and Activator of Transcription, an important transcription factors, is activated by JAK [55, 56]. In JAK and STAT family, emerging studies have indicated that JAK2/STAT3 pathway is well-established regulators of cell migration, and has been implicated in the process of tumor cell invasion and metastasis [57].

Some studies have indicated that miRNAs participate in the JAK-STAT signaling pathway.MiR-375 may function as a tumor suppressor to regulate gastric cancer cell proliferation potentially by targeting the JAK2 oncogene [58]. MiR-125b suppresses the proliferation and migration of osteosarcoma cells through downregulation of STAT3 [59]. Transfection of precursor miR-199a-3p into osteosarcoma cell lines significantly decreased cell growth and migration. Duan and his colleagues observed decreased mTOR and STAT3 expression in miR-199a-3p transfected cells [60]. Yan and his colleagues indicated that miR-20a regulates STAT3 at the post-transcriptional level, resulting in inhibition of cell proliferation and invasion of pancreatic carcinoma [61].

Advertisement

4. MAPK pathway

The Mitogen Activated Protein Kinase (MAPK) pathway is a frequent event in tumorigenesis. MAPKs have been implicated in cell migration, proteinase induction, apoptosis, and angiogenesis, events that are essential for successful completion of metastasis [8]. The presence of at least six MAPK in yeast suggests that there are more in mammals: extracellular signal-regulated kinases (ERK1, ERK2), c-Jun N-terminal kinases (JNKs), p38 isoforms, ERK5, ERK3/4, ERK7/8. In vivo and in vitro studies have confirmed that three major subgroups of MAPK including ERK1/2, JNK, and p38, are specifically involved in invasion and metastasis [9, 10, 62].

Mounting studies have indicated that miRNAs participate in the MAPK signaling pathway.MiR-143 plays an important role in prostate cancer proliferation, migration and chemosensitivity by suppressing KRAS and subsequent inactivation of MAPK pathway [63]. MiR-17-5p significantly activates the p38 kinase pathway [64]. Raf kinase inhibitory protein suppresses a cascade of metastasis signalling involving LIN28 and let-7 [65]. Zhu and his colleagues found that miR-101 targets MAPK phosphatase 1 to regulate the activation of MAPKs in macrophages [66]. MiR-146a suppresses tumor growth and progression by targeting EGFR pathway and in a p-ERK-dependent manner in castration-resistant prostate cancer [67]. Liu and his colleagues indicated that miR-21 induces tumor angiogenesis through targeting PTEN, leading to activate AKT and ERK1/2 signaling pathways [68,69].EGFR promotes lung tumorigenesis by activating miR-7 through a Ras/ERK/Myc pathway that targets the ETS2 transcriptional repressor ERF [70].

Advertisement

5. Wntsignaling pathway

Wntsignaling pathway controls tissue polarity and cell movement through the activation of RhoA, JNK, and nemo-like kinase (NLK) signaling cascades. The Wnt gene family is a group of developmental genes that encode cysteine-rich glycosylated proteins [71]. Aberrant activation of Wntsignaling pathway in human cancer leads to more malignant phenotypes, such as abnormal tissue polarity, invasion, and metastasis [72].

A number of studies have indicated that miRNAs participate in the Wntsignaling pathway.MiR-200a is a new tumor suppressor that can regulate the activity of the Wnt/β-catenin signaling pathway [73]. MiR-371-373 expression is induced by lithium chloride and is positively correlated with Wnt/β-catenin-signaling activity in several human cancer cell lines [74].MiR-27 directly targeted and inhibited adenomatous polyposis coli (APC) gene expression, and activated Wntsignaling through accumulation of β-catenin [75].Kapinas and his colleagues reported that miR-29 modulates Wntsignaling in human osteoblasts through a positive feedback loop [76].MiR-17-5p plays an important role in breast cancer cell invasion and migration by suppressing HBP1 and subsequent activation of Wnt/β-catenin [77]. Kennell and his colleagues demonstrated that miR-8 family members play an evolutionarily conserved role in regulating the Wntsignaling pathway [78].

Advertisement

6. Notch signaling pathway

The Notch signaling pathway is a conserved ligand–receptor signaling pathway. Notch genes encode single-pass transmembrane proteins that can be activated by interacting with a family of its ligands. To date, four Notch receptors have been identified in mammals, including human, such as Notch-1-4. It has been well known that Notch signaling plays important roles in maintaining the balance involved in cell proliferation, survival, apoptosis, and differentiation which affects the development and function of many organs [79]. Therefore, dysfunction of Notch prevents differentiation, ultimately guiding undifferentiated cells toward malignant transformation. Indeed, many observations suggest that alterations in Notch signaling are associated with invasion and metastasis in many human cancers [14-16].

Mounting studies have indicated that miRNAs participate in the Notch signaling pathway. MicroRNA-23b is capable of inducingtolerogenic properties of dendritic cells in vitro through the inhibition of the Notch1 and NF-κB signalling pathways [80]. MicroRNA-181 promotes natural killer(NK) cell development by regulating Notch signaling [81]. MiR-124a mediates stroke-induced neurogenesis by targeting the JAG-Notch signaling pathway [82]. Pang and his colleagues demonstrated that miR-34a affected cell invasion by regulating expression of urokinase plasminogen activator through Notch [83].MiR-206 targets Notch 3, activates apoptosis, and inhibits tumor cell migration and focus formation [84]. MiR-1 influences cardiac differentiation in Drosophila and regulates Notch signaling [85]. Some studies indicated that the ZEB1/miR-200 feedback loop controls Notch signalling in cancer cells [86, 87].

Advertisement

7. EMT

Several oncogenic pathways (Rho GTPases, JAK-STAT, MAPK, Wnt and Notch) may induce EMT [21-25]. In particular, the association of those pathways with EMT has been shown to activate EMT-inducing transcriptional regulators such as the members of the Snail family, the zinc finger transcription factors (ZEB), Transforming growth factor beta (TGF-β), Twist and Slug.

Members of the Snail family of transcriptional regulators, namely Snail 1 and Snail 2, have emerged as a key regulatory factor of EMT. The zinc finger transcription factors ZEB1 and ZEB2 also make a pivotal contribution to this regulation. TGF-β, a major inducer of EMT, exists in at least three isoforms called TGF-β1, TGF-β2 and TGF-β3. It cooperates with stem cell pathways like Wnt, Ras and Notch to induce EMT [88, 89]. Twist, a basic helix-loop-helix transcription factor, exists in at least two isoforms called Twist 1 and Twist 2. Twist proteins promote EMT by turning-down the expression of epithelial specific proteins, such as the E-cadherin and by up-regulating the expression of mesenchymal markers such as the N-cadherin, the vimentin and the smooth-muscle actin [90]. Slug, a zinc finger transcription factor, whose product belongs to the Snail family of developmental regulatory proteins, is transcriptional repressors of E-cadherin and induces EMT [1].

Emerging studies have indicated that miRNAs participate in the EMT. The miR-106b-25 cluster targets Smad7, activates TGF-β signaling, and induces EMT in human breast cancer [91]. MiR-27 promoted EMT by activating the Wnt pathway [92]. MiR-221/222 targeting of trichorhinophalangeal 1 (TRPS1) promotes EMT in breast cancer [93]. MiR-194 inhibits EMT of endometrial cancer cells by targeting oncogene BMI-1 [94]. Let-7d negatively modulates EMT expression and also plays a role in regulating chemo-resistant ability in oral cancer [95]. MiR-200b and miR-15b regulate chemotherapy-induced EMT in human tongue cancer cells by targeting BMI-1 [96]. Kumarswamy and his colleagues found that miR-30a targets Snai1, inhibits invasion and metastasis, and is downregulated in non-small cell lung cancer (NSCLC) [97]. Vetter and his colleagues indicated that miR-661 expression in Snail 1-induced EMT contributes to breast cancer cell invasion by targeting Nectin-1 and StarD10 messengers [98]. Some studies indicated that the miR-200 family and miR-205 regulate EMT by targeting ZEB1 and SIP1 [99, 100].

Advertisement

8. MicroRNAs in invasion and metastasis in lung cancer

Lung cancer is the leading cause of death among the malignant tumors worldwide, and the incidence of lung cancer is increasing. Tumor invasion and metastasis are the critical steps in determining the aggressive phenotype of human cancers. Mortality of tumor patients results mainly from cancer cells spreading to distant organs. In order to resist cancer dissemination, more effective therapeutic strategies are clearly required. However, the cellular and molecular machinery, underlying invasion and metastasis by miRNA in lung cancer, is relatively poorly understood. In light of these recent discoveries, we have classified the experimentally validated miRNAs regulating the invasion and metastasis of lung cancer and showed in Figure 2.

In light of these recent discoveries, the present article indicated that miRNAs participate in invasion and metastasis in lung cancer.Zhu and his colleagues indicated that MTA1 functions in regulating the invasive phenotype of lung cancer cells and this regulation may be through altered miRNA expression, such as miR-125b, miR-210, miR-103, miR-194 and miR-500 [101]. Huand his colleagues reported that MiR-193b modulated proliferation, migration, and invasion of NSCLC [102].A p53/miR-34 axis has been found that it regulates Snail1-dependent cancer cell EMT[103]. MiR-378 is associated with NSCLC brain metastasis by promoting cell migration, invasion and tumor angiogenesis [104].MiR-30a targets Snai1, inhibits invasion and metastasis, and is downregulated in NSCLC [105].Expression level of miR-206 was inversely correlated with metastatic potential of lung cancer [106].Roybal and his colleagues demonstrated that miR-200 Inhibits lung adenocarcinoma cell invasion and metastasis by targeting Flt1 [107]. Loss of miR-200c expression induces an aggressive, invasive, and chemoresistant phenotype in NSCLC [108]. In our previous studies, we found that hsa-miR-125a-3p and hsa-miR-125a-5p are downregulated in NSCLC and have inverse effects on invasion and migration of lung cancer cells [109]. Zhang and his colleagues reported that miR-21 post-transcriptionally downregulates the expression of tumor suppressor PTEN and stimulates growth and invasion in NSCLC [110].Crawford and his colleagues indicated that MiR-126 alters lung cancer cell phenotype by inhibiting adhesion, migration, and invasion and the effects on invasion may be partially mediated through Crk regulation [111]. The deep mechanisms of miRNAs in invasion and metastasis which contribute to lung cancer are worthy of further investigation.

Figure 2.

The experimentally validated miRNAs regulate the invasion and metastasis in lung cancer.

Advertisement

9. Conclusion and future perspective

Despite recent advances in diagnosis and treatment, lung cancer remains a leading cause of death among the malignant tumors worldwide, and the incidence of lung cancer is increasing. Even so, no improvement in prognosis has been observed if the patient presents with metastases at diagnosis. A better understanding of the mechanism of tumor cell invasion is critical for the development of more effective treatments for metastatic cancer. In recent years, emerging studies have attested to the association between miRNAs and the mechanism in critical processes during cancer dissemination, and we have summarized many of these in the present manuscript. Here, we have condensed much of this early work, and highlight key deregulated miRNAs targeting molecules involved in Rho family GTPases, JAK-STAT, MAPK, Wnt, Notch pathway and transcriptional control of EMT. In the future, a more complete dissection of the pathways controlled by miRNAs may offer new insights on metastasis, and highlight promising areas for the development of novel anti-cancer therapies.

Acknowledgement

Lili Jiang collectedinformation andwrote the manuscript.XueShanQiu helped with the manuscript design and gave critical review of the manuscript.We are grateful to the members of our laboratory for useful suggestions.

References

  1. 1. SreekumarRSayanB. SMirnezamiA. HSayanA. EMicroRNA Control of Invasion and Metastasis Pathways.Front Genet 201158 EOF
  2. 2. BaranwalSAlahariS. KRho GTPase effector functions in tumor cell invasion and metastasis.Curr Drug Targets 20111281194201
  3. 3. Struckhoff AP, Rana MK, WorthylakeRA. RhoA can lead the way in tumor cell invasion and metastasis. Front Biosci 2011; 16: 1915-26.
  4. 4. NarumiyaSTanjiMIshizakiTRho signaling, ROCK and mDia1, in transformation, metastasis and invasion. Cancer Metastasis Rev 2009
  5. 5. LaiS. YChildsE. EXiSCoppelliF. MGoodingW. EWellsAFerrisR. LGrandisJ. RErythropoietin-mediated activation of JAK-STAT signaling contributes to cellular invasion in head and neck squamous cell carcinoma.Oncogene2005242744429
  6. 6. ZhaoYZhangJXiaHZhangBJiangTWangJChenXWangYStat3 is involved in the motility, metastasis and prognosis in lingual squamous cell carcinoma.Cell BiochemFunct. 20123043406
  7. 7. DevarajanEHuangSSTAT3 as a central regulator of tumor metastases.CurrMol Med 20099562633
  8. 8. ReddyK. BNabhaS. MAtanaskovaNGomesL. RTerraL. FWailemannR. ALabriolaLSogayarM. CRole of MAP kinase in tumor progression and invasion.Cancer Metastasis Rev 2003224395403
  9. 9. Del Barco Barrantes INebreda AR. Roles of 38MAPKs in invasion and metastasis.BiochemSoc Trans 2012
  10. 10. ZhangSGuoWRenT. TLuX. CTangG. QZhaoF. LArsenic trioxide inhibits Ewing’s sarcoma cell invasiveness by targeting 38MAPK) and c-Jun N-terminal kinase. Anticancer Drugs 2012
  11. 11. SarkarF. HLiYWangZKongDThe role of nutraceuticals in the regulation of Wnt and Hedgehog signaling in cancer.Cancer Metastasis Rev 201029338394
  12. 12. HuangDDuXCrosstalk between tumor cells and microenvironment via Wnt pathway in colorectal cancer disseminationWorld J Gastroenterol 2008141218237
  13. 13. NethPRiesCKarowMEgeaVIlmerMJochumMThe Wnt signal transduction pathway in stem cells and cancer cells: influence on cellular invasionStem Cell Rev 2007311829
  14. 14. AsnaghiLEbrahimiK. BSchreckK. CBarE. ECoonfieldM. LBellW. RHandaJMerbsS. LHarbourJ. WEberhartC. GNotch signaling promotes growth and invasion in uveal melanoma.Clin Cancer Res 201218365465
  15. 15. BaileyJ. MSinghP. KHollingsworthM. ACancer metastasis facilitated by developmental pathways: Sonic hedgehog, Notch, and bone morphogenic proteins.J Cell Biochem 2007102482939
  16. 16. WangX. QZhangWLuiE. LZhuYLuPYuXSunJYangSPoonR. TFanS. TNotch1Snail1-E-cadherin pathway in metastatic hepatocellular carcinoma.Int J Cancer 2012 1E163-72.
  17. 17. ShihJ. YYangP. CThe EMT regulator slug and lung carcinogenesis.201120113291299304
  18. 18. WellsAChaoY. LGrahovacJWuQLauffenburgerD. AEpithelial and mesenchymal phenotypic switchings modulate cell motility in metastasisFront Biosci 20111681537
  19. 19. SavagnerPThe epithelial-mesenchymal transition (EMT) phenomenon.Ann Oncol 2010Suppl 7:vii8992
  20. 20. CanoC. EMotooYIovannaJ. LEpithelial-to-mesenchymal transition in pancreatic adenocarcinoma.ScientificWorldJournal 201010194757
  21. 21. SavagnerPLeaving the neighborhood: molecular mechanisms involved during epithelial-mesenchymal transition.Bioessays 2001231091223
  22. 22. YadavAKumarBDattaJTeknosT. NKumarPIL-6 promotes head and neck tumor metastasis by inducing epithelial-mesenchymal transition via the JAK-STAT3-SNAIL signaling pathway.Mol Cancer Res 2011912165867
  23. 23. WuW. SThe signaling mechanism of ROS in tumor progressionCancer Metastasis Rev 2006254695705
  24. 24. NethPRiesCKarowMEgeaVIlmerMJochumMThe Wnt signal transduction pathway in stem cells and cancer cells: influence on cellular invasionStem Cell Rev 2007311829
  25. 25. WangZLiYKongDSarkarF. HThe role of Notch signaling pathway in epithelial-mesenchymal transition (EMT) during development and tumor aggressiveness.Curr Drug Targets 201011674551
  26. 26. LeeR. CFeinbaumR. LAmbrosVTheCelegansheterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993
  27. 27. LeeYAhnCHanJChoiHKimJYimJLeeJProvostPRadmarkOKimSKimV. NThe nuclear RNase III Drosha initiates microRNA processing.Nature2003415 EOF9 EOF
  28. 28. KongWZhaoJ. JHe L&Cheng JQ. Strategies for profiling microRNA expression.J Cell Physiol 2009
  29. 29. HutvágnerGMclachlanJPasquinelliA. EBálintETuschlTZamoreP. DA cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA.Science 2001
  30. 30. GrishokAPasquinelliA. EConteDLiNParrishSHaIBaillieD. LFireARuvkunGMelloC. CGenes and mechanisms related to RNA interference regulate expression of the small temporal RNAs that control C. elegans developmental timing. Cell 2001
  31. 31. CrawfordMBrawnerEBatteKYuLHunterM. GOttersonG. ANuovoGMarshC. BNana-sinkamS. PMicroRNA-126 inhibits invasion in non-small cell lung carcinoma cell lines.BiochemBiophys Res Commun 2008
  32. 32. ZhuSWuHWuFNieDShengSMoY. YMicroRNA-21 targets tumor suppressor genes in invasion and metastasis.Cell Res 2008350 EOF9 EOF
  33. 33. TavazoieS. FAlarcónCOskarssonTPaduaDWangQBosP. DGeraldW. LMassaguéJEndogenous human microRNAs that suppress breast cancer metastasis.Nature2008147 EOF52 EOF
  34. 34. BusteloX. RSauzeauVBerenjenoI. MGTP-binding proteins of the Rho/Rac family: regulation, effectors and functions in vivo.Bioessays2007294356370
  35. 35. BoureuxAVignalEFaureSFortPEvolution of the Rho family of ras-like GTPases in eukaryotes.MolBiolEvol200724120316
  36. 36. RathinamRBerrierAAlahariS. KRole of Rho GTPases and their regulators in cancer progression.Front Biosci 201116256171
  37. 37. KongWYangHHeLZhaoJ. JCoppolaDDaltonW. SChengJ. QMicroRNA-155 is regulated by the transforming growth factor beta/Smad pathway and contributes to epithelial cell plasticity by targeting RhoA.Mol Cell Biol 20082822677384
  38. 38. JiangLLiuXKolokythasAYuJWangAHeidbrederC. EShiFZhouXDownregulation of the Rho GTPasesignaling pathway is involved in the microRNA-138-mediated inhibition of cell migration and invasion in tongue squamous cell carcinoma.Int J Cancer 2010127350512
  39. 39. ValastyanSChangABenaichNReinhardtFWeinbergR. AConcurrent suppression of integrin alpha5, radixin, and RhoAphenocopies the effects of miR-31 on metastasis.Cancer Res 20107012514754
  40. 40. ChibaYTanabeMGotoKSakaiHMisawaMDown-regulation of miR-133a contributes to up-regulation of Rhoa in bronchial smooth muscle cells.Am J RespirCrit Care Med 200918087139
  41. 41. KamaiTTsujiiTAraiKTakagiKAsamiHItoYOshimaHSignificant association of Rho/ROCK pathway with invasion and metastasis of bladder cancer.Clin Cancer Res 200397263241
  42. 42. BishopA. LHallARho GTPases and their effector proteins.Biochem J 2000Pt 224155
  43. 43. RientoKRidleyA. JRocks: multifunctional kinases in cell behaviour.Nat Rev Mol Cell Biol 2003446 EOF56 EOF
  44. 44. SalhiaBRuttenFNakadaMBeaudryCBerensMKwanARutkaJ. TInhibition of Rho-kinase affects astrocytoma morphology, motility, and invasion through activation of Rac1.Cancer Res 20058792 EOF800 EOF
  45. 45. YuJPengHRuanQFatimaAGetsiosSLavkerR. MMicroRNA-205 promotes keratinocyte migration via the lipid phosphatase SHIP2.FASEB J 2010241039509
  46. 46. WuLCaiCWangXLiuMLiXTangHMicroRNA-142-3p, a new regulator of RAC1, suppresses the migration and invasion of hepatocellular carcinoma cellsFEBS Lett 20115859132230
  47. 47. Moriarty CH, Pursell B, Mercurio AM.miR-10b targets Tiam1: implications for Rac activation and carcinoma migration. J BiolChem 2010; 285 (27): 20541-6.
  48. 48. LueddeTMicroRNA-151 and its hosting gene FAK (focal adhesion kinase) regulate tumor cell migration and spreading of hepatocellular carcinoma.Hepatology 201052311646
  49. 49. Liu M, Lang N, Qiu M, Xu F, Li Q, Tang Q, Chen J, Chen X, Zhang S, Liu Z, Zhou J, Zhu Y, Deng Y, Zheng Y, Bi F. miR-137 targets Cdc42 expression, induces cell cycle G1 arrest and inhibits invasion in colorectal cancer cells.Int J Cancer 2011; 128 (6): 1269-79.
  50. 50. Chen Q, Chen X, Zhang M, Fan Q, Luo S, Cao X. miR-137 is frequently down-regulated in gastric cancer and is a negative regulator of Cdc42. Dig Dis Sci. 2011; 56 (7): 2009-16.
  51. 51. LiuHCaoY. DYeW. XSunY. YEffect of microRNA-206 on cytoskeleton remodelling by downregulating Cdc42 in MDA-MB-231 cells.Tumori 20109657515
  52. 52. Park SY, Lee JH, Ha M, Nam JW, Kim VN. miR-29miRNAs activate p53 by targeting p85 alpha and CDC42. Nat StructMolBiol 2009; 16 (1): 23-9.
  53. 53. Liu M, Tang Q, Qiu M, Lang N, Li M, Zheng Y, Bi F. miR-21 targets the tumor suppressor RhoB and regulates proliferation, invasion and apoptosis in colorectal cancer cells. FEBS Lett 2011; 585 (19): 2998-3005.
  54. 54. ConnollyE. CVan DoorslaerKRoglerL. ERoglerC. EOverexpression of miR-21 promotes an in vitro metastatic phenotype by targeting the tumor suppressor RHOB.Mol Cancer Res 201085691700
  55. 55. AaronsonD. SHorvathC. MA road map for those who don’t know JAK-STATScience2002296557316535
  56. 56. LooyengaB. DHutchingsDCherniIKingsleyCWeissG. JMackeiganJ. PSTAT3 Is Activated by JAK2 Independent of Key Oncogenic Driver Mutations in Non-Small Cell Lung Carcinoma.PLoS One2012e30820 EOF
  57. 57. DevarajanEHuangSSTAT3 as a central regulator of tumor metastases.CurrMol Med. 20099562633
  58. 58. Ding L, Xu Y, Zhang W, Deng Y, Si M, Du Y, Yao H, Liu X, Ke Y, Si J, Zhou T. MiR-375 frequently downregulated in gastric cancer inhibits cell proliferation by targeting JAK2. Cell Res 2010; 20 (7): 784-93.
  59. 59. Liu LH, Li H, Li JP, Zhong H, Zhang HC, Chen J, Xiao T. miR-125b suppresses the proliferation and migration of osteosarcoma cells through down-regulation of STAT3. BiochemBiophys Res Commun 2011; 416 (1-2): 31-8.
  60. 60. DuanZChoyEHarmonDLiuXSusaMMankinHHornicekFMicroRNA-199a-3p is downregulated in human osteosarcoma and regulates cell proliferation and migration.Mol Cancer Ther 2011108133745
  61. 61. YanHWuJLiuWZuoYChenSZhangSZengMHuangWMicroRNA-20a overexpression inhibited proliferation and metastasis of pancreatic carcinoma cells.Hum Gene Ther 20102112172334
  62. 62. GomesL. RTerraL. FWailemannR. ALabriolaLSogayarM. CTGF-ß1 modulates the homeostasis between MMPs and MMP inhibitors through 38MAPK and ERK1/2 in highly invasive breast cancer cells. BMC Cancer2012
  63. 63. Xu B, Niu X, Zhang X, Tao J, Wu D, Wang Z, Li P, Zhang W, Wu H, Feng N, Wang Z, Hua L, Wang X. miR-143 decreases prostate cancer cells proliferation and migration and enhances their sensitivity to docetaxel through suppression of KRAS.Mol Cell Biochem 2011; 350 (1-2): 207-13.
  64. 64. Yang F, Yin Y, Wang F, Wang Y, Zhang L, Tang Y, Sun S. miR-17-5p Promotes migration of human hepatocellular carcinoma cells through the p38 mitogen-activated protein kinase-heat shock protein 27 pathway.Hepatology 2010; 51 (5): 1614-23.
  65. 65. Dangi-garimellaSYunJEvesE. MNewmanMErkelandS. JHammondS. MMinnA. JRosnerM. RRaf kinase inhibitory protein suppresses a metastasis signalling cascade involving LIN28 and let-7.EMBO J 200928434758
  66. 66. ZhuQ. YLiuQChenJ. XLanKGeB. XMicroRNA-101 targets MAPK phosphatase-1 to regulate the activation of MAPKs in macrophages.J Immunol 201018512743542
  67. 67. Xu B, Wang N, Wang X, Tong N, Shao N, Tao J, Li P, Niu X, Feng N, Zhang L, Hua L, Wang Z, Chen M. MiR-146a suppresses tumor growth and progression by targeting EGFR pathway and in a p-ERK-dependent manner in castration-resistant prostate cancer. Prostate. 2012; 72 (11): 1171-8.
  68. 68. Liu LZ, Li C, Chen Q, Jing Y, Carpenter R, Jiang Y, Kung HF, Lai L, Jiang BH. MiR-21 induced angiogenesis through AKT and ERK activation and HIF-1a expression.PLoS One 2011; 6 (4): e19139.
  69. 69. HatleyM. EPatrickD. MGarciaM. RRichardsonJ. ABassel-dubyRVan RooijEOlsonE. NModulation of K-Ras-dependent lung tumorigenesis by MicroRNA-21.Cancer Cell201018328293
  70. 70. ChouY. TLinH. HLienY. CWangY. HHongC. FKaoY. RLinS. CChangY. CLinS. YChenS. JChenH. CYehS. DWuC. WEGFR promotes lung tumorigenesis by activating miR-7 through a Ras/ERK/Myc pathway that targets the Ets2 transcriptional repressor ERF.Cancer Res 20107021882231
  71. 71. DaleT. CSignal transduction by the Wnt family of ligands.Biochem J 1998Pt 2): 209-23.
  72. 72. KatohMWNT/PCP signaling pathway and human cancer.Oncol Rep 200514615838
  73. 73. SuJZhangAShiZMaFPuPWangTZhangJKangCZhangQMicroRNA-200a suppresses the Wnt/ß-catenin signaling pathway by interacting with ß-catenin.Int J Oncol 2012404116270
  74. 74. ZhouA. DDiaoL. TXuHXiaoZ. DLiJ. HZhouHQuL. HCatenin/LEF1transactivates the microRNA-371-373 cluster that modulates the Wnt/ß-catenin-signaling pathway. Oncogene. Oncogene. 20123124296878
  75. 75. Wang T, Xu Z. miR-27 promotes osteoblast differentiation by modulating Wntsignaling. BiochemBiophys Res Commun 2010; 402 (2): 186-9.
  76. 76. Kapinas K, Kessler C, Ricks T, Gronowicz G, Delany AM. miR-29 modulates Wntsignaling in human osteoblasts through a positive feedback loop. J BiolChem 2010; 285 (33): 25221-31.
  77. 77. Li H, Bian C, Liao L, Li J, Zhao RC. miR-17-5p promotes human breast cancer cell migration and invasion through suppression of HBP1. Breast Cancer Res Treat 2011; 126 (3): 565-75.
  78. 78. KennellJ. AGerinIMacDougald OA, Cadigan KM. The microRNA miR-8 is a conserved negative regulator of Wntsignaling.ProcNatlAcadSci U S A 2008105401541722
  79. 79. WangZLiYAhmadAAzmiA. SBanerjeeSKongDSarkarF. HTargeting Notch signaling pathway to overcome drug resistance for cancer therapy.BiochimBiophysActa 20101806225867
  80. 80. ZhengJJiangH. YLiJTangH. CZhangX. MWangX. RDuJ. TLiH. BXuGMicroRNA-23b promotes tolerogenic properties of dendritic cells in vitro through inhibiting Notch1/NF-?B signalling pathways. Allergy 201267336270
  81. 81. CichockiFFelicesMMccullarVPresnellS. RAl-attarALutzC. TMillerJ. SCutting edge: microRNA-181 promotes human NK cell development by regulating Notch signaling.J Immunol 20111871261715
  82. 82. LiuX. SChoppMZhangR. LTaoTWangX. LKassisHHozeska-solgotAZhangLChenCZhangZ. GMicroRNA profiling in subventricular zone after stroke: MiR-124a regulates proliferation of neural progenitor cells through Notch signaling pathway.PLoS One 2011e23461.
  83. 83. PangR. TLeungC. OYeT. MLiuWChiuP. CLamK. KLeeK. FYeungW. SMicroRNA-34a suppresses invasion through downregulation of Notch1 and Jagged1 in cervical carcinoma and choriocarcinoma cells.Carcinogenesis2010316103744
  84. 84. SongGZhangYWangLMicroRNA-206 targets notch3, activates apoptosis, and inhibits tumor cell migration and focus formation.J BiolChem 200928446319217
  85. 85. KwonCHanZOlsonE. NSrivastavaDMicroRNA1 influences cardiac differentiation in Drosophila and regulates Notch signalingProcNatlAcadSci U S A 2005102521898691
  86. 86. BrabletzSBajdakKMeidhofSBurkUNiedermannGFiratEWellnerUDimmlerAFallerGSchubertJBrabletzTThe ZEB1/miR-200 feedback loop controls Notch signalling in cancer cells.EMBO J 201130477082
  87. 87. VallejoD. MCaparrosEDominguezMTargeting Notch signalling by the conserved miR-8/200 microRNA family in development and cancer cells.EMBO J 201130475669
  88. 88. FuxeJVincentTGarcia de Herreros A. Transcriptional crosstalk between TGF-ß and stem cell pathways in tumor cell invasion: role of EMT promoting Smad complexes. Cell Cycle 2010912236374
  89. 89. WendtM. KAllingtonT. MSchiemannW. PMechanisms of the epithelial-mesenchymal transition by TGF-beta.Future Oncol 200958114568
  90. 90. OnderT. TGuptaP. BManiS. AYangJLanderE. SWeinbergR. ALoss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways.Cancer Res 20086810364554
  91. 91. SmithA. LIwanagaRDrasinD. JMicalizziD. SVartuliR. LTanA. CFordH. LThe miR-106b-25 cluster targets Smad7, activates TGF-ß signaling, and induces EMT and tumor initiating cell characteristics downstream of Six1 in human breast cancer.Oncogene20123150516271
  92. 92. Zhang Z, Liu S, Shi R, Zhao G. miR-27 promotes human gastric cancer cell metastasis by inducing epithelial-to-mesenchymal transition. Cancer Genet 2011; 204 (9): 486-91.
  93. 93. StinsonSLacknerM. RAdaiA. TYuNKimH. JOBrienCSpoerkeJJhunjhunwalaSBoydZJanuarioTNewmanR. JYuePBourgonRModrusanZSternH. MWarmingSDe SauvageF. JAmlerLYehR. FDornanDTrpstargeting by miR-221/222 promotes the epithelial-to-mesenchymal transition in breast cancer.Sci Signal 2011ra41.
  94. 94. DongPKaneuchiMWatariHHamadaJSudoSJuJSakuragiNMicroRNA-194 inhibits epithelial to mesenchymal transition of endometrial cancer cells by targeting oncogene BMI-1.Mol Cancer 201199 EOF
  95. 95. ChangC. JHsuC. CChangC. HTsaiL. LChangY. CLuS. WYuC. HHuangH. SWangJ. JTsaiC. HChouM. YYuC. CHuF. WLet-7d functions as novel regulator of epithelial-mesenchymal transition and chemoresistant property in oral cancer.Oncol Rep 2011264100310
  96. 96. Sun L, Yao Y, Liu B, Lin Z, Lin L, Yang M, Zhang W, Chen W, Pan C, Liu Q, Song E, Li J. MiR-200b and miR-15b regulate chemotherapy-induced epithelial-mesenchymal transition in human tongue cancer cells by targeting BMI1. Oncogene 2012; 31 (4): 432-45.
  97. 97. KumarswamyRMudduluruGCeppiPMuppalaSKozlowskiMNiklinskiJPapottiMAllgayerHMicroRNA-30a inhibits epithelial-to-mesenchymal transition by targeting Snai1 and is downregulated in non-small cell lung cancer.Int J Cancer. 20121309204453
  98. 98. VetterGSaumetAMoesMVallarLLe Béchec A, Laurini C, Sabbah M, Arar K, Theillet C, Lecellier CH, Friederich E. miR-661 expression in SNAI1-induced epithelial to mesenchymal transition contributes to breast cancer cell invasion by targeting Nectin-1 and StarD10 messengers.Oncogene20102931443648
  99. 99. GregoryP. ABertA. GPatersonE. LBarryS. CTsykinAFarshidGVadasM. AKhew-goodallYGoodallG. JThe miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1.Nat Cell Biol 2008105593601
  100. 100. XiongMJiangLZhouYQiuWFangLTanRWenPYangJThe miR-200 family regulates TGF-ß1induced renal tubular epithelial to mesenchymal transition through Smad pathway by targeting ZEB1 and ZEB2 expressionAm J Physiol Renal Physiol 2012F369-79.
  101. 101. ZhuXZhangXWangHSongQZhangGYangLGengJLiXYuanYChenLMTA1 gene silencing inhibits invasion and alters the microRNA expression profile of human lung cancer cells.Oncol Rep. 201228121824
  102. 102. HuHLiSLiuJNiBMicroRNA-193b modulates proliferation, migration, and invasion of non-small cell lung cancer cells.ActaBiochimBiophys Sin. 201244542430
  103. 103. KimN. HKimH. SLiX. YLeeIChoiH. SKangS. EChaS. YRyuJ. KYoonDFearonE. RRoweR. GLeeSMaherC. AWeissS. JYookJ. IA 53miRNA-34 axis regulates Snail1-dependent cancer cell epithelial-mesenchymal transition.J Cell Biol. 2011
  104. 104. ChenL. TXuS. DXuHZhangJ. FNingJ. FWangS. FMicroRNA-378 is associated with non-small cell lung cancer brain metastasis by promoting cell migration, invasion and tumor angiogenesis.Med Oncol 2012293167380
  105. 105. KumarswamyRMudduluruGCeppiPMuppalaSKozlowskiMNiklinskiJPapottiMAllgayerHMicroRNA-30a inhibits epithelial-to-mesenchymal transition by targeting Snai1 and is downregulated in non-small cell lung cancer.Int J Cancer. 20121309204453
  106. 106. WangXLingCBaiYZhaoJMicroRNA-206 is associated with invasion and metastasis of lung cancer.Anat Rec 201129418892
  107. 107. Roybal JD, Zang Y, Ahn YH, Yang Y, Gibbons DL, Baird BN, Alvarez C, Thilaganathan N, Liu DD, Saintigny P, Heymach JV, Creighton CJ, Kurie JM.miR-200 Inhibits lung adenocarcinoma cell invasion and metastasis by targeting Flt1/VEGFR1. Mol Cancer Res 2011; 9 (1): 25-35.
  108. 108. CeppiPMudduluruGKumarswamyRRapaIScagliottiG. VPapottiMAllgayerHLoss of miR-200c expression induces an aggressive, invasive, and chemoresistant phenotype in non-small cell lung cancer.Mol Cancer Res 201089120716
  109. 109. Jiang L, Huang Q, Zhang S, Zhang Q, Chang J, Qiu X, Wang E. Hsa-miR-125a-3p and hsa-miR-125a-5p are downregulated in non-small cell lung cancer and have inverse effects on invasion and migration of lung cancer cells. BMC Cancer 2010; 10: 318.
  110. 110. ZhangJ. GWangJ. JZhaoFLiuQJiangKYangG. HMicroRNA-21 (miR-21) represses tumor suppressor PTEN and promotes growth and invasion in non-small cell lung cancer (NSCLC)ClinChimActa 2010846 EOF852 EOF
  111. 111. CrawfordMBrawnerEBatteKYuLHunterM. GOttersonG. ANuovoGMarshC. BNana-sinkamS. PMicroRNA-126 inhibits invasion in non-small cell lung carcinoma cell lines.BiochemBiophys Res Commun. 2008373460712

Written By

Lili Jiang and Xueshan Qiu

Submitted: 18 February 2012 Published: 13 March 2013