ebook img

Tumor Angiogenesis: A Key Target for Cancer Therapy PDF

304 Pages·6.337 MB·English
Save to my drive
Quick download
Download
Most books are stored in the elastic cloud where traffic is expensive. For this reason, we have a limit on daily download.

Preview Tumor Angiogenesis: A Key Target for Cancer Therapy

Mechanisms of Tumor Angiogenesis Iratxe Zuazo-Gaztelu and Oriol Casanovas Abstract activator of the angiogenic switch, its regula- Tumor angiogenesis, the process by which tion and a detailed description of normal and bloodvesselspenetrateandgrowinthetumor aberrant tumor vessels are also provided. The microenvironment,isessentialforoxygenand understanding of the foundations of these nutrient supply and hence constitutes a key mechanisms is crucial for an effective thera- playerforthesurvivalofsolidneoplasms.Dif- peutictargetingoftheangiogenicprocess. ferent mechanisms of angiogenesis are devel- oped during tumor progression such as Keywords vasculogenesis,sproutingangiogenesis,intus- Physiologicalangiogenesis(cid:129)Tumorangiogen- susception, and vasculogenic mimicry. The esis (cid:129) Sprouting angiogenesis (cid:129) transition from a quiescent vasculature to an Vasculogenesis (cid:129) Intussusception (cid:129) actively growing one follows a series of syn- Vasculogenicmimicry(cid:129)Vesselgrowing(cid:129)Ves- chronic events and is finely tuned by a wide sel guidance (cid:129) Vascular-remodeling factors (cid:129) array of molecules and positive and negative Vessel stabilization (cid:129) Proangiogenic factors (cid:129) regulators of angiogenesis. Beginning with Antiangiogenic factors (cid:129) Angiogenic switch (cid:129) blood vessel sprouting and endothelial cell Hypoxia(cid:129)Tumorvasculature proliferation, followed by vessel navigation, remodeling,stabilization, andmaturation,and Contents finishing with blood vessel regression, the Introduction.......................................... 2 main molecular factors involved in the pro- MolecularMechanismsofAngiogenesis........... 2 gression of each step are profoundly detailed. MechanismsInvolvedinBloodVesselFormation... 2 Whenthebalancebetweenpositiveandnega- BiologicalProcessesInvolvedinAngiogenesis...... 6 tive regulators of angiogenesis is shifted TheAngiogenicSwitchinTumorigenesis.......... 18 towardproangiogenicmolecules,thequiescent HypoxiaandTumorAngiogenesis................... 19 vasculaturebecomesactivatedandinitiatesthe DifferencesbetweenPhysiologicalandTumor angiogenic state of tumor development. The Neovascularization................................ 21 role of intratumoral hypoxia as a potent Conclusion............................................ 23 Cross-References..................................... 24 I.Zuazo-Gaztelu(cid:129)O.Casanovas(*) References............................................ 24 TumorAngiogenesisGroup,ProCURE,CatalanInstitute ofOncology-IDIBELL,Barcelona,Spain e-mail:[email protected]; [email protected] #SpringerInternationalPublishingAG2016 1 D.Marmé(ed.),TumorAngiogenesis, DOI10.1007/978-3-319-31215-6_1-1 2 I.Zuazo-GazteluandO.Casanovas Introduction Molecular Mechanisms of Angiogenesis Tumorangiogenesis,theprocessbywhichblood vesselspenetrateandgrowinthetumormicroen- MechanismsInvolvedinBloodVessel vironment, is essential for oxygen and nutrient Formation supply and, therefore, for survival of solid neo- plasms. Taking into account its role in tumor Vessels can be described as highly branched and progression and metastasis, angiogenesis is one ordered tubular networks that allow transport of of the widely acknowledged hallmarks of cancer gases, nutrients, signaling molecules, and cells. (Hanahan and Weinberg 2011). Since the Beyond their nutritive function, blood vessels pioneering studies by Folkman established the provide instructive trophic signals essential for initial foundations of tumor angiogenesis nearly organ morphogenesis and the development of 40 years ago, research in the field has reached a every complex organism (Carmeliet and Jain significant level of maturity, allowing detailed 2011). While the luminal side of all types of descriptionoftheintricateprocessesofpatholog- blood vessels, including arteries, veins, and cap- icalvesselproliferation(Folkmanetal.1971).In illaries, is formed by a lined monolayer of endo- this chapter, we compiled the basic mechanisms thelialcells,vesselsarecoveredintheoutsidebya ofbloodvesselformationandthebiologicalroles basementmembranefollowedbyalayerofmural of the main molecular regulators involved in accessory cells (pericytes and vascular smooth tumorangiogenesis.Furthermore,wedescribein musclecells(SMCs)). detailthe“angiogenicswitch”occurringinmalig- Prototypically, vasculogenesis and sprouting nantneoplasms,focusingontheroleofthecom- angiogenesis are the two main mechanisms plex tumor microenvironment, the differences responsible for neovascularization (Fig. 1a, b). between physiological and pathological angio- Sprouting angiogenesis is defined as the forma- genesis, and the abnormalities found in tumor tionofnewvascularstructuresfromapreexisting vasculature. vessel, while vasculogenesis refers to de novo Fig.1 Mechanismsinvolvedinbloodvesselformation. growth,aprocessthatinvolvesvesselsplitting(c).Addi- Innormaltissuesvesselscangrowbytherecruitmentof tionally, tumor cells use other mechanisms such as bonemarrow-derivedendothelialprogenitorcells(EPCs) vasculogenicmimicry,duringwhichvessel-likestructures that differentiate into endothelial cells (a), by sprouting arelinedbytumorcells(d) angiogenesis (b), or by intussusceptive microvascular MechanismsofTumorAngiogenesis 3 bloodvesselformationduetovascularprogenitor homingtoneo-angiogenicspots areyet notfully cell differentiation. Both mechanisms contribute understood. to the formation and remodeling of the vessel MobilizationofEPCsstartsfollowingtheacti- networkduringdevelopment,remainnearlyinac- vationofthematrixmetalloprotease9(MMP9)in tiveintheadultbody,andareonlyreactivatedto the osteoblastic zone by the tumor-derived pro- allowtissuerepairorintheeventofadisease. angiogenicfactors.TheactivationofMMP9trig- Beyond vasculogenesis and sprouting angio- gers the proteolytic processing of membrane- genesis, other less frequent mechanisms have bound Kit ligand to its soluble active form. The recently been reported in neoplasms, including soluble Kit ligand is a stem cell-active cytokine vessel co-option, intussusception, and that promotes migration of hematopoietic and vasculogenicmimicry(Fig.1c,d).Inmostcases, endothelial progenitor cells to the vascular zone mutualexclusivitybetweendifferentmechanisms ofthebonemarrowandtheirposteriorreleasein does not exist; indeed, they simultaneously par- thecirculation(Heissigetal.2002). ticipate both in physiological and pathological Whenhomed,endothelialprogenitorcellscan angiogenesis. beincorporatedintotheendothelialmonolayerof avesselorrecruitedtoangiogenicsprouts.Atthis Vasculogenesis step,P-selectin,E-selectin,andintegrinsarecrit- Vasculogenesis (Fig. 1a) has been extensively icalforthecorrectadhesionofEPCstothevessel describedintheearlystagesofvasculardevelop- walls (Deb et al. 2004). Differentiation into ment.Itwasnotuntil1997thatthegrowthofnew mature endothelial cells is mainly mediated by blood vessels in postembryonic tissues was con- VEGF and physically contributes to vessel size sidered to occur also through vasculogenesis since it increases the diameter of the vessel. In (Asaharaetal.1997).Moreover,compellingevi- additiontothephysicalcontributionoftheEPCs dence suggests that bone marrow-derived circu- tothenewlyformedvessels,EPCssupportangio- lating endothelial progenitor cells (EPCs) genesis by a paracrine mechanism that includes contribute to the induction and progression the release of proangiogenic factors in neo- of postnatal neovascularization (Kopp et al. vascularizationsitesofthetumorstromaorische- 2006). EPCs, also known as angioblasts, are cir- mictissues(UrbichandDimmeler2004). culating cells that express several endothelial Formal demonstration of the contribution of lineage-specific markers such as CD34, CD31, vasculogenesis to tumor angiogenesis has been VEGFR-2,andTie-2. achieved through the use of knockout mice for TogetherwithEPCs,maturecirculatingendo- inhibitorsofdifferentiationfactors.Thesefactors thelial cells derived from blood vessel renewal initiate the mobilization of bone marrow-derived alsotakepartinadultvasculogenesis. Moreover, dendritic cells to angiogenic sites in the tumor. inordertofacilitateincorporationofthosecircu- Genetic ablation of inhibitors of differentiation latingendothelialadultandprogenitorcellsandto factors disrupted tumor vascularization and sustain the stability of the nascent vasculature, blocked tumor growing as a consequence hematopoietic stem and progenitor cells are of an impaired angiogenesis (Benezra et al. recruited(Koppetal.2005).Severalchemokines, 2001). The restoration of the mobilization cytokines,andgrowthfactorsthatareproducedin capabilityofbonemarrow-derivedcellsbytrans- response to tissue ischemia and tumor growth plantation of wild-type bone marrow rescued promote mobilization and recruitment of EPCs. tumor neovascularization and growth in these For instance, tumor cells produce proangiogenic knockoutmice. factors, such as VEGF, and cytokines (i.e., The contribution of vasculogenesis to tumor stromal-derived factor-1) that recruit bone vessel formation ranges from 0.1 to 50% marrow-deriveddendriticcellsandpromotetheir depending on the experimental cancer model proliferation and differentiation. The precise and tumor type. Lymphomas and hematological mechanismsgoverningmobilizationofprecursor tumors are more dependent on bone marrow- cells from the bone marrow and their posterior derived dendritic cells in comparison to other 4 I.Zuazo-GazteluandO.Casanovas tumors. The knowledge obtained from the (Ferrara et al. 2003). At this step, an immature study of tumor vasculogenesis has sparkled the blood vessel is formed, and the opposite development of new applications at the clinical mesenchymal-endothelial transition directs the setting. The potential existing correlation reversal of the proliferative state of endothelial between the levels of endothelial progenitor and cells to the previous resting state. In detail, the circulating cells in blood and the outcome in return to quiescence is retrieved by synthesis of patients undergoing an antiangiogenic treatment newbasementmembraneandpericyteandmural couldbeusedasacellularbiomarkerformonitor- cell recruitment (Jain 2003). This latter step is ing the response to antitumor therapy (Bertolini known as vessel maturation and is characterized etal.2003). byalackoftumorangiogenesis. Recently, the study of tumor vasculogenesis Recent studies have remarked the specializa- hasshiftedfromitsroleinprimarytumorgrowth tion that endothelial cells undergo in order to toward the study of its implication in dissemina- enrolltheangiogenic process.Toachievelocally tion and metastasis. In addition to angiogenesis neededvascularpatterns,themultistepprocessof activation, EPCs are able to promote metastatic sprouting angiogenesis requires functional spe- growth by homing into metastatic sites prior to cialization of endothelial cells in the angiogenic tumorcellarrival(Koppetal.2005).Thesponta- sprout together with vascular guidance cues that neous secretion of SDF-1 by EPCs generates a allow regulation of the topological extension of gradient that could promote the extravasation the forming vessel. The main group of signaling and development of the pre-metastatic niche (Jin pathways essential for the initial morphogenetic etal.2012). events includes VEGF and Notch (Iruela-Arispe and Dvorak 1997). Attracted by proangiogenic SproutingAngiogenesis signals, the phenotype of the sprouting endothe- Sprouting angiogenesis is the best described lial cells gains an invasive and motile behavior, mechanismusedbytumorstopromotetheirown protease activation, cell-cell contact remodeling, vascularizationbyinducingnewcapillarysprouts andapical-basalpolarityreversal.Theendothelial from preexisting host capillaries (Fig. 1b). The cells that are selected to guide the sprouting are mechanisminvolvesseveralwell-definedsequen- locatedatthetipoftheangiogenicsproutandare, tialstepsandanextensiveinterplaybetweensol- therefore,commonlyknownas“tipcells.”These uble factors, extracellular matrix (ECM) leading cells respond to VEGF signaling by components, and cells (Paku and Paweletz dynamicallyextendinglargefilopodiainorderto 1991). At the onset of sprouting angiogenesis, sense and guide the forming vessel along the there is a destabilization of the endothelial- forthcoming vascular bed. Recent studies show pericyte contacts which are essential for vessel similarities between the molecular regulation of integrity and quiescence maintenance. Endothe- guidance cues of neural and endothelial cells. lial and mural cells share a complex basement These specialized nonproliferative endothelial membrane that forms a protective coat around cells also release different molecular signals that endothelial tubules, preventing resident endothe- promote the recruitment of mural cells like peri- lial cells from leaving their location. Once cytes, SMCs, and fibroblasts, guaranteeing the destabilized, endothelial cells undergo an stabilizationofemergingvessels. endothelial-mesenchymaltransitionthatenhances Apartfromtipcells,endothelialcellscanspe- their migratory, invasive, and proliferative prop- cialize to highly proliferating cells located in the erties. These activated cells are then able to stalk of the angiogenic sprout. The proliferative degrade the surrounding ECM and the basement potential of the “stalk cells,” capable of forming membrane by activated proteases (such as tubes and branches, assures the expansion of MMPs), opening the path for guided migration the structure. In contrast to tip cells, stalk and proliferation. Vessel lumen is then formed cellsextendfewerfilopodiabutproliferatetosup- by polarization of themigrating endothelial cells port vessel elongation in response to VEGF-A MechanismsofTumorAngiogenesis 5 (Gerhardtetal.2003).Tipcellsaremigratoryand tumor angiogenesis, there are alternative pro- polarized, whereas stalk cells proliferate during cesses such as intussusceptive microvascular the extension of the sprout and form the new growth(IMG;Fig.1c)andvasculogenicmimicry vascular lumen cell population. Stalk cells also (VM; Fig. 1d). These nonconventional pathways produce components of the basement membrane introduceanadditionallevelofcomplexitytothe and establish adherens and tight junctions with understanding of tumor vascularization neighboringcells,thusstrengtheningtheintegrity mechanisms. ofthenewsproutandluminal-abluminalpolarity (Dejanaetal.2009). Intussusception Inordertoformanewvascularconnection,the Intussusception(IMG)isa variant ofangiogene- tip cell phenotype must be switched off after sisthatwasfirstobservedinpostnatalremodeling connecting with the tips of other sprouts or of lung capillaries (Caduff et al. 1986; Fig. 1c). existing vessels. Tip cells build vessel loops by This developmental intravasculargrowthmecha- anastomosing with cells from neighboring nism is based on the splitting of preexisting ves- sprouts. The sprouting process is rehearsed until selsintotwonewvesselsaftertheformationofa proangiogenic signals decrease, a new basement transvascularconnectivetissuecolumn,calledtis- membraneisformed,quiescenceisreestablished, suepillar,intothelumenofthevessel. andVEGFlevelsdampen(Leslieetal.2007).In Incontrasttosproutingangiogenesis,IMGisa thetransitionfromactivesproutingtoquiescence, fast process that can occur within hours, or even endothelial tip cells adopt a “phalanx”-like phe- minutes,sinceitdoesnotrequireproliferationof notype, with features of lumenized, non- endothelial cells. Even though sprouting has the proliferative, and immobile cells (Bautch 2009). advantage of being invasive and permits joining Eventually, maturity and stabilization are vascular gaps, it is a slow process that highly achieved through the generation of a lumen and relies on endothelial cell proliferation and basal the migration of pericytes along the basement membrane degradation. In IMG, the remodeling membrane until vessels are covered, initiating ofendothelialcellsisaconsequenceoftheirvol- bloodflowandallowingperfusion. ume increase and narrowing. It is believed that The correct extension and morphology of the IMG happens after vasculogenesis or sprouting nascentvesselsisregulatedbytheprecariousbal- angiogenesis in order to expand the capillary ance between tip cell navigation and stalk cell plexus without a high metabolic demand (Burri proliferation. The phenotypic specialization to etal.2004). tip or stalk cell depends on the balance between TheonsetofIMGisthe“touchingcontact”of proangiogenic factors andendothelialcellprolif- endothelial cells from opposite walls. Following erationsuppressors(GeudensandGerhardt2011). the transendothelial cell bridge created from the Theabnormalvascularstructuresgenerallyfound touching spot, interendothelial junctions are in cancer are a consequence of the imbalance reorganized, and the endothelial bilayer is between these two processes. The biological performed. Then, the interstitial pillar forms to nature of the molecules and signals that initiate reinforcethebridges,andmuralcellsarerecruited the angiogenic cascade from the initial destabili- to cover this new interstitial wall. Due to their zation to the formation of mature and functional contractile features, pericytes are believed to be vasculaturehasbeenprofoundlystudiedandchar- themaintriggersofthisphase.Finally,theinter- acterizedandisfurtherdescribedinthefollowing stitial pillars widen, endothelial cells retract, and sectionsofthischapter. two independent vessels are created (Burri et al. 2004). By IMG, a large vessel can split into AlternativeWaysofBloodSupply smallervessels. inTumors Even though the precise mechanism of intus- Although sprouting angiogenesis is regarded as susception is poorly understood, there are some the most important contributing mechanism to key mediators that influence pillar formation. 6 I.Zuazo-GazteluandO.Casanovas Alterations in blood flow dynamics, changes in components, and low levels of oxygen are shearstressonendothelialcellssensedandtrans- known to promote VM (Seftor et al. 2005). ducedbymoleculessuchasCD31,modifications Although the exact mechanism remains to be of wall stress on the pericytes, and absences of unraveled, it involves deregulation of the VEGFaresomeamongthepossiblefactorsdriv- lineage-specific phenotype and the concomitant ingbiochemicalcascadesthatresultincytoskele- transdifferentiationtoendothelial-likecells. tal rearrangements and intussusception initiation VM occurs mainly in aggressive tumors such (DjonovandMakanya2005).Forinstance,itwas asmelanomas,and even though theiroccurrence observedthathumanmelanomasbareahighnum- is relatively rare within tumors, the presence of berofintraluminaltissuefoldstogetherandthata VM-associatedpatternednetworksintumortissue correlation between VEGF and intussusceptive correlateswithanincreasedriskofmetastasisand angiogenesisexistsinthesetumors(Ribattietal. poor clinical outcome (Sun et al. 2004). Until 2005). In this context, sprouting angiogenesis now, VM has been described in melanomas, inhibitionmightstimulatetheprocessofintussus- breastcarcinoma,prostaticcarcinoma,hepatocel- ceptive angiogenesis. Since IMG can only occur lular carcinoma, bladder carcinoma, and other on existing vessel networks, its most important aggressive tumors. Increasing evidence demon- contribution is its ability to increase the density strates that tumor cell-dominant VM has a key and complexity of tumor microvessel networks roleintumorprogressionandmetastasis. already established by sprouting angiogenesis. Moreover, IMG also provides additional surface forfurthersproutingangiogenesis.IMGhasbeen BiologicalProcessesInvolved observedincolorectal,melanoma,andmammary inAngiogenesis tumors(Domeetal.2007). As stated above, in order to build new and fully VasculogenicMimicry functional vascular structures, several biological Vasculogenic mimicry (VM; Fig. 1d) describes processesmustbeaccuratelyregulated.Different the ability of some tumor cells to dedifferentiate pro- and antiangiogenic regulators are needed to into multiple cellular phenotypes, obtaining perform and control each specific step of the endothelial-likeproperties(Maniotisetal.1999). angiogenic cascade. Multifunctionality among This process leads to the creation of de novo those factors is one of their relatively common vasculogenic-like matrix embedded networks. features, empowering some molecules with the The new perfusable vascular-like structures are outstanding ability to either activate or inhibit composedofredbloodcellsandplasmaandcon- vascularization.Onaccountofsimplification,we tribute to blood circulation (Frenkel et al. 2008). willdescribethemolecularregulationofthemain Endothelial cells undergoing VM mimic the pat- biologicalprocessesinvolvedinvesselformation: ternofembryonicvascularnetwork,possiblypro- sprouting and proliferation (Fig. 2a), guidance viding tumor cells with a secondary circulation and navigation (Fig. 2b), stabilization, matura- system,independentfromangiogenesis. tion,andremodeling(Fig.2c),andregression. Molecularanalysiscomparinghighlyinvasive andnoninvasivemelanomacellsderivedfromthe SproutingofBloodVessels same patient suggests a genetic reversion of andEndothelialCellProliferation the aggressive cells to an embryonic-like cell Endothelial cells in the adult organism remain fate and increased cell plasticity. The quiescent and are protected against external undifferentiated phenotype includes the expres- insults by autocrine maintenance signals. These sion of endothelium-associated genes such as cells form a monolayer of phalanx cells and VE-cadherin and Ephrin-A2, among others are interconnected by junctional molecules like (Hendrixetal.2003).Theactivationoftransmem- VE-cadherin and claudins. The surface of brane metalloproteinases, release of ECM the endothelium monolayer is covered by MechanismsofTumorAngiogenesis 7 Fig. 2 Molecular basis of angiogenesis. Sequential pericyte detachment. A provisional matrix layer is steps of blood vessel formation and their features are depositedbyextravasationofplasmaproteins(e.g.,fibrin- depicted.Themostimportantmolecularplayersinvolved ogen) due to increased permeability. Cell migration is ineachprocessaredenotedinparentheses.(a)Uponangio- favoredbyprotease-mediatedmatrixremodeling.(b)Tip genicstimulationbyproangiogenicfactors,thequiescent cells sense the environment and navigate in response vessel dilates and an endothelial tip cell is selected. to guidance cues (e.g., semaphorins and ephrins) while Tipcellgenerationrequiresbasementmembranedegrada- adhering to the extracellular matrix (ECM) in order to tion, loosening of endothelial cell-cell junctions, and migrate.Stalkcellsbehindtheleadingtipcellproliferate 8 I.Zuazo-GazteluandO.Casanovas pericytes, which suppress endothelial cell prolif- hypoxia,oncogeneactivation,lossoftumorsup- eration, and releases pro-survival signals such as pressors, cytokines, and growth factors levels. VEGFandAng-1.Whenquiescentvesselssense There is also an autocrine production of VEGF an angiogenic signal such as VEGF, fibroblast by endothelial cells which is critical for vascular growth factors (FGFs), or chemokines, released homeostasisandearlystagesofvasculardevelop- by hypoxic, inflammatory, or tumor cells, ment(Leeetal.2007).Ingeneralterms,paracrine sproutingangiogenesisistriggered(Fig.2a). VEGF, released by tumor or stromal cells, VEGF-VEGFR signaling pathway is increases vessel branching and promotes tumor established as the master regulator of the forma- vessel abnormalities (Stockmann et al. 2008), tion and remodeling of vasculature. VEGF whereasautocrineVEGF,releasedbyendothelial ligands are the prototypical, multifunctional pro- cells, empowers vascular homeostasis. The dele- angiogenic factors that control endothelial cell tion of a single allele of VEGF-A causes embry- proliferation and migration and regulate cardio- oniclethality,reinforcingthebelievedkeyroleof vascularsystemhomeostasis(Carmeliet andJain thisfamilyindevelopmentalvascularphysiology. 2011).Untilnow,VEGFmoleculesareallegedly There are three different tyrosine kinase-type themostpotentvascularpermeabilityfactorsand receptorsfortheVEGFfamily:VEGFR-1(orFlt1 vasodilatationinductors.Besides,endothelialpre- inmouse),VEGFR-2(alsoknownaskinaseinsert cursor cell differentiation and vascular guidance domain-containing receptor in humans or Flk1), oftipcellsarealsocontrolledbyVEGFfamily. andVEGFR-3(orFlt4).Thesereceptorsareprin- TheVEGFfamilyiscomposedofsixdifferent cipally expressed in endothelial cells (Roskoski members: VEGF-A (referred herein as VEGF), 2008). Furthermore, certain VEGF isoforms are VEGF-B,VEGF-C,VEGF-D,VEGF-E,andpla- also able to bind to non-tyrosine kinase cental growth factor (PlGF). Contrary to other coreceptorssuchasneuropilins1and2toenhance angiogenic superfamilies, the VEGF family dis- VEGFR-2 activity. Neuropilins are best known tinguishes itself by the nonredundant role of its for their interaction with semaphorin and their members. At least five different isoforms of angiogenesis-independent function in axonal VEGF are generated by alternative splicing of a guidance(Gluzman-Poltoraketal.2000). single gene: VEGF , VEGF , VEGF , VEGFR-1hasbeenidentifiedasthehighaffin- 121 145 165 VEGF , and VEGF . These isoforms differ ityreceptor,whereasVEGFR-2isthelow-affinity 189 206 intheirbindingaffinityforheparin,whichmight receptor. While placental growth factor and affect their diffusion rates in the extracellular VEGF-B bind to VEGFR-1, VEGF-A can bind space. The predominant splicing variant of both VEGFR-1 and VEGFR-2, VEGF-C and VEGF both in normal and tumor cells is the VEGF-D bind to VEGFR-2 and VEGFR-3, and VEGF isoform.AllthemembersoftheVEGF VEGF-E binds to VEGFR-2. Binding of the 165 family and their corresponding receptors usually ligand leads to the dimerization of the receptors; work as homodimers, although heterodimers this in turn initiates the autophosphorylation of between different members have also been several intracellularly located tyrosine residues. reported(DiSalvoetal.1995). The activated dimers now expose new docking During tumor progression, VEGF levels are sitesfortherecruitmentofdifferenttypesofinter- controlled through diverse mechanisms such as mediary signaling molecules by protein-protein (cid:1)(cid:1)(cid:1) Fig.2 (continued)andelongate,attractingpericytesinthe posterior lumen formation drive neovessel perfusion, process.Stalkcellsarefurtherstabilizedbythedeposition whichconcludeswithquiescencebyaphalanxphenotype of the new basement membrane. Immune myeloid cell promotion, deposition of the new basement membrane, recruitment of tumor-associated macrophages (TAMs) maturationofpericytes,reestablishmentofcell-celljunc- andTIE-2-expressingmonocytes(TEMs)producesaddi- tions,andreleaseofvascularmaintenancemolecules tional proangiogenic factors and triggers the release of ECM-boundfactors.(c)Fusionofadjacentbranchesand MechanismsofTumorAngiogenesis 9 interactions through specific SH2 and SH3 shown that VEGFR-1 acts as a decoy receptor, domains. These large signaling complexes are since it is able to sequester VEGF, implying a known as signalosomes and can be different negativeroleforthisreceptorinangiogenesis.In depending on the combination of VEGF ligand addition,micelackingVEGFR-1presentahigher and receptor. In fact, differences between number of endothelial cells than wild-type mice, signalosomes allow a broad range of biological whereas the decreased expression of VEGFR-1 effects to VEGF stimulation that include increases VEGF availability and VEGFR-2 increasedendothelialcellproliferation,migration, activity. In fact, a soluble isoform of VEGFR-1 survival,permeability,andECMdegradation. (sVEGFR-1), which encodes the extracellular Several studiesindicate that VEGF stimulates ligand-binding domain, can be produced by sur- both physiological and tumor angiogenesis by rounding cells. While soluble VEGF isoforms signalingthroughVEGFR-2inadose-dependent promote vessel enlargement, matrix-bound manner (Carmeliet and Jain 2011). In fact, isoforms stimulate the branching pattern. VEGFR-2nullmicedieduringembryonicdevel- sVEGFR-1 inhibits angiogenesis by acting as a opment by defects in the vascular system, molecular trap for VEGF ligand, assisting the reinforcing its role in proliferation, survival, and guidance of emerging branches or inhibiting the migration of vascular endothelial cells. The acti- sprouting. The imbalance between the functions vation of VEGFR-2 promotes differentiation of ofVEGFR-1andVEGFR-2causeshemangiomas progenitors, mitogenesis, chemotaxis, survival, andbenigntumorswithincreasedaberrantangio- and vascular permeability. It also increases the genesis(Jinninetal.2008). expressionofmatrixmetalloproteinasesandplas- Inpathologicalconditions,PlGFcontributesto minogenactivatorsforECMdegradationandfur- the angiogenic switch by affecting multiple cell ther endothelial cell migration. In detail, VEGF type directly and indirectly and also activates release causes plasma proteins extravasation and bone marrow-derived endothelial progenitor the deposition of a provisional ECM scaffold cells. PlGF can induce its own signaling and toward which endothelial cells migrate in amplify VEGF-driven angiogenesis through responsetointegrinsignaling.Theactivatedpro- direct effects on endothelial cells (Autiero et al. teasesliberatetheangiogenicmoleculesstoredin 2003).ThesynergismbetweenVEGFandPlGFis theECMsuchasVEGFandFGF,andtheECMis also stated by the fact that PlGF upregulates the remodeledintoanangio-competentmilieu.Once expression of VEGF. PlGF might also indirectly VEGFisreleased,itbindstotheVEGFR-2recep- influence SMC proliferation and migration tors oftheendothelial cells.Tipcellmigration is throughactivatedendothelialcellcytokinerelease regulated by VEGF gradient, whereas stalk cell (Luttunetal.2002). proliferation depends on VEGF concentration Other prototypical proangiogenic signaling (Gerhardtetal.2003).VEGF/VEGFR-2signaling pathway involves the fibroblast growth factor axis induces the formation and extension of (FGF) family, comprised by 23 different ligands filopodia and the expression of delta-like ligand and four tyrosine kinase-type receptors (FGFR- 4(Dll4)proteinintipcells,whichactivatesNotch 1–4) expressed widely in the organism (Presta in stalk cells. Notch, in turn, downregulates et al. 2005). FGFR-1 and FGFR-2 are expressed VEGFR-2 expression in stalk cells, rendering by endothelial cells, and their binding to ligands them less responsive to VEGF and ensuring tip FGF-1, FGF-2, FGF-4, and FGF-5 leads to the cellleading.TheblockadeofVEGFR2signaling induction of critical stages of angiogenesis isassociatedwithsproutingdefects(Bentleyetal. in vivo. Among the four pleiotropic pro- 2009). angiogenic ligands, at least FGF-1 and FGF-2 On the other hand, VEGFR-1 is only slightly directly stimulate endothelial cell proliferation, activatedbyproangiogenicfactors,anditsprecise detachment, migration, and ultimate differentia- role in angiogenesis is poorly understood tion into a functional capillary vessel. For the (Schwartz et al. 2010). Nevertheless, it has been maintenance of vascular integrity, endothelial 10 I.Zuazo-GazteluandO.Casanovas quiescent cells require low levels of FGF, since like TGF-β induce the autocrine production of vesseldisintegrationhasbeenobservedasacon- EGF-likemoleculessuchasTGF-α,thuspromot- sequence of FGFR signaling inhibition ing endothelial cell survival through PI3K-Akt (Murakami et al. 2008). FGF ligands exert their signaling(ViñalsandPouyssegur2001). functions in endothelial cells after paracrine The ECM itself provides a link between vas- release by stromal or tumor cells, or by endoge- cular cells and their surrounding environment. nousFGFinanautocrinefashion. Proteolytic degradation of the basement mem- Even though VEGF has a pivotal role during brane and the surrounding ECM is an integral angiogenesis, an important cross talk takes place partofangiogenesis.Inthisstep,severalprotein- between FGF and VEGF. For instance, VEGF ase families are involved, including matrix systemactivationisrequiredforlaterFGFinduc- metalloproteinases (MMP and their tissue-type tion and in vivo angiogenesis promotion. The inhibitors or TIMP), plasminogen activators opposite cross regulation has also been demon- (uPA and its inhibitor PAI-1), heparanases, strated, and FGF also seems able to stimulate tryptases, chymases, cathepsins, etc. Besides tumor angiogenesis under certain experimental breaking down ECM components and clearing a conditions.TheeffectsofFGFareduetoitsdual pathforendothelialcellmigration,proteinasesare action including a direct effect over endothelial abletoswitchonangiogenesisbytheliberationof cellsandanindirecteffectconcerningtheregula- matrix-bound angiogenic activators (bFGF, tion of the production of other proangiogenic VEGF, TGF-β, HGH, etc.) and proteolytically molecules like VEGF, angiopoietin-2 (Ang-2), activatingangiogenicchemokinessuchasIL-1β. or interleukin-8 (IL-8) by tumor or stromal cells WhereasVEGFisoformscleavedbyMMPspref- (BeenkenandMohammadi2009).Bothinmouse erentiallyenlargevessels,MMP-resistantmatrix- and human tumors, the role of FGF in tumor bound VEGF is involved in vessel branching growthandneovascularizationhasbeendescribed (Iruela-Arispe and Davis 2009). Moreover, pro- (Prestaetal.2005). teasessuchasMMP9participateinthemobiliza- Thethirdproangiogenicpathwayincludesthe tionofbonemarrowprogenitorsbytheliberation epidermal growth factor (EGF)-like family of ofcytokinessuchastheKitligand(Heissigetal. growth factors and their receptors (ErbB). The 2002)andbytheestablishmentofapre-metastatic integrants of this family play various functions niche (Kaplan et al. 2005). The proteolytic indifferenttissues,buttheyarebasicallyinvolved remodeling of the ECM occurs in a sharply con- in cell proliferation and survival stimulation. trolledmode,andthepleiotropicactivitiesofpro- SomeoftheligandsincludedareEGFandtrans- teinasesarecontextandconcentrationdependent. forminggrowthfactoralpha(TGF-α),whichbind In fact, excessive breakdown removes guidance to the ErbB family of tyrosine kinase receptors. cuesforendothelialcellmigration,thusinhibiting The ErbB family of receptors is composed angiogenesis, while insufficient degradation pre- of ErbB1/HER1/EGFR, ErbB2/HER2/Neu, vents vascular cell mobility (Carmeliet and Jain ErbB3/HER3,andErbB4/HER418. 2011). Bothintumorandphysiologicalsettings,EGF Moreover, proteinases can switch off angio- family members display proangiogenic activity. genesis,astheyliberatematrix-boundangiogenic However, whether the effects of EGF are direct inhibitorssuchasarrestin,angiostatin,TSP-1,and orindirectremainsunclear.EGFstimulationpro- inactivate angiogenic cytokines like SDF-1. The ducesthereleaseofproangiogenicfactorsuchas basementmembraneofquiescentvesselsiscom- VEGF,IL-8,andFGFbytumorandstromalcells. posedmainlyofcollagenIVandlaminin,whereas The ErbB family receptors in endothelial cell the interstitial matrix of collagen I and elastin membranes enable these cells to respond to betweenvascularcellsprovidesfurtherviscoelas- EGF-like factors by increased proliferation and ticity and strength to the endothelial cell wall. survival. Furthermore, other angiogenic factors Proteinases expose novel epitopes of these ECM

See more

The list of books you might like

Most books are stored in the elastic cloud where traffic is expensive. For this reason, we have a limit on daily download.