83
Views
10
CrossRef citations to date
0
Altmetric
Review

The potential for targeting extracellular LOX proteins in human malignancy

&
Pages 1729-1735 | Published online: 25 Nov 2013

Abstract

The extracellular matrix (ECM) is the physical scaffold where cells are organized into tissues and organs. The ECM may be modified during cancer to allow and promote proliferation, invasion, and metastasis. The family of lysyl oxidase (LOX) enzymes cross-links collagens and elastin and, therefore, is a central player in ECM deposition and maturation. Extensive research has revealed how the LOX proteins participate in every stage of cancer progression, and two family members, LOX and LOX-like 2, have been linked to metastasis, the final stage of cancer responsible for over 90% of cancer patient deaths. However, LOX biosynthesis results in by-product with antiproliferative properties in certain cancers, and LOX enzymes may have different effects depending on the molecular network in which they are active. Therefore, the design of therapies targeting the LOX family needs to be guided by the molecular makeup of the individual disease and will probably require other agents to act on both the LOX enzymes and their associated network.

Extracellular matrix in cancer

Cancer cells in a tumor proliferate amongst stromal cells and vessels supported by the interstitial extracellular matrix (ECM). The ECM is a material formed by macromolecules that gives structure and anatomy to every tissue, but during cancer it also supplies proinflammatory signals associated with invasion, intravasation, and metastasis. These biological functions seem to be related to the physical properties of the ECM; cancer-associated ECM is stiffer and denser and suffers simultaneous processes of deposition and degradation that liberate growth factors embedded in it, while trapping more stromal and circulating cancer cells.Citation1,Citation2 Lysyl oxidase (LOX) has been proven to be a crucial mediator in remodeling of the cancer-associated ECM, metastasis, and the premetastatic niche.Citation3 This evidence points to the LOX family as a potential target in the prevention and treatment of metastatic disease. However, extracellular LOX has been shown to have antitumor activity in certain cancers,Citation4 whereas the by-product of LOX biosynthesis has antioncogenic activity. This suggests that: 1) an anti-LOX treatment for cancer must be limited to tumors where LOX enhances progression, which possibly implies the need for molecular diagnostic tools; and 2) there may be a role for LOX or LOX by-products as anticancer agents. Here we will review the current knowledge on the extracellular LOX enzymes and their potential role in cancer therapy.

The LOX family

LOX is an enzyme that mediates the cross-linking of collagens and elastin; two basic components of the ECM. LOX was first isolated from bovine aorta and some insight into function was obtained.Citation5 Initial research of LOX in cancer found it was downregulated in epithelial tumorsCitation6 and had oncogene-suppressing actions.Citation7 The LOX family has five members; LOX and LOX-like 1 to 4 (LOXL2, LOXL3, LOXL4, respectively) (). The family shares a highly conserved amino acid sequence that includes a cytokine receptor-like domain, residues for carbonyl cofactor formation, and a copper-binding site. An atom of copper tightly bound to the active site is essential for protein conformation and, therefore, for the catalytic activity of the enzymes. The N-terminal of every member differs, but, in LOX-like 2, 3, and 4, it is formed of scavenger receptor cysteine-rich domains.

Figure 1 The lysyl oxidase family.

Notes: The lysyl oxidase family shares a highly conserved catalytic domain that includes a copper-binding motif, a lysyl-tyrosine-quinone cofactor and a cytokine receptor-like domain. These are necessary for protein conformation and enzymatic activity. LOX and LOXL1 contain pro-sequences which are cleaved by bone morphogenetic protein 1 before their secretion into the extracellular space. LOXL2 to 4 form a subfamily defined by its scavenger receptor cysteine rich domains thought to mediate cell adhesion and protein-protein interactions.
Abbreviations: LOX, lysyl oxidase; LOXL, lysyl oxidase-like; LTQ, lysyl-tyrosine-quinone; SRCR, scavenger receptor cysteine rich; CRL, cytokine receptor-like.
Figure 1 The lysyl oxidase family.

LOX is synthesized as a preproenzyme that is first cleaved in the endoplasmic reticulum before the N-terminal propeptide is glycosylated and the C-terminal is folded to acquire three disulphide bonds. The catalytic site incorporates copper and the proenzyme is then released to the extracellular space. The glycosylated N-terminal is cleaved by bone morphogenetic protein 1 (BMP1) (or procollagen C proteinase [PCP])Citation8 resulting in release of the active form of the enzyme and of the LOX propeptide (LOX-PP). LOXL1 is also synthesized as a proenzyme; however, less is known about its secretion and activation, and nothing is known about that of the other family members. While LOX is necessary for ECM maintenance, LOX-PP is biologically active and suppresses gene expression.Citation4

The first substrates described for LOX and later for the other LOXL proteins were collagens and elastin.Citation5 LOX cross-links fibers of collagen and elastin by oxidizing lysine residues into an aldehyde that spontaneously condenses with other aldehydes or peptidyl lysines to form covalent unions among fibers. These unions insolubilize, stabilize, and harden the ECM. The other LOX family members are thought to act in the same way, which is highly likely given the close homology of their catalytic domains.Citation9,Citation10

The LOX family’s role in ECM building and maintenance implies its involvement in pathological conditions. LOX has been showed to be an agent of chronic hepatic fibrosis, arterial fibrosis, and Adriamycin-induced kidney fibrosis in human patients,Citation11 and liver and lung fibrosis in mouse models.Citation12 Lack of LOX is also related to vascular, respiratory, and cutaneous fragility; LOX−/− mice have defective collagen and elastin cross-linking, which translates into impaired development of the upper and lower airways, lungs, vessels, and skin.Citation13 These mice die at birth; thus, LOX function cannot be substituted by any of the other family members. Reduced LOX activity has been detected in two inherited human syndromes: Menkes disease and occipital horn syndrome.Citation11 Both are caused by mutations that limit copper availability, therefore affecting LOX-mediated catalysis. Patients diagnosed with these syndromes have a shortened lifespan, defective connective tissues, and neurological impairment. LOX overexpression as a result of copper deficiency is associated with Ehlers Danlos syndrome (EDS), where patients have over-elasticated skin,Citation14 and a mutation of the LOX gene is related to EDS type VI, which has as its main clinical characteristics neonatal kyphoscoliosis, joint laxity, skin fragility, and abnormal muscle tone.Citation15 LOX has been also linked to neurodegenerative disorders in animal models of amyotrophic lateral sclerosis, Alzheimer’s disease, and dementia.Citation16,Citation17 This suggests that LOX may contribute to diseases not only related to abnormal connective tissue.

LOX−/− mice are viable, but the females have defective elastin renewal in the pelvic floor, which leads to postpartum pelvic prolapse.Citation18 In humans, the lack of LOXL1 is associated with pseudoexfoliation syndrome; a pathology of elastic fibres that degenerates in fragile vascularity, including brain vessels. Currently, a clinical trial for the detection of LOXL1 polymorphism is ongoing.Citation19 Other mutations of LOXL1 produce abnormal elastic fibers in the eyes, heart, lungs, liver, kidneys, and other tissues.Citation20

LOXL2 acts by oxidizing collagens and elastin.Citation21 It promotes chondrocyte differentiation and cartilage ECM maturation.Citation22 It is overexpressed in the stroma of pathologically fibrotic tissues, such as cirrhotic liver and Wilson’s disease, and confers susceptibility to brain aneurysms.Citation23 Antibody targeting of LOXL2 decreases lung and liver fibrosis in mouse models and prevents the activation of ECM-deposing cells such as fibroblasts and endothelial cells.Citation24 Use of a LOXL2-targeting monoclonal antibodyCitation24 is currently in Phase II clinical trials for fibrosis patients.

LOXL3 has been less studied than the other family members. Its anatomical pattern of expression overlaps partially with other members of the LOX family and shares substrates (collagens and elastin).Citation25 It has an enzymatically active subvariant that lacks the scavenger domains 1, 2, and 3. LOXL3 overexpression is related to ventricular stiffness and T lymphocyte function in cardiac remodeling.Citation26

LOXL4 was first described as necessary for cartilage maturation and cartilage ECM deposition.Citation27 It is widely expressed in human tissues, notably by endothelial cells;Citation10 Transforming growth factor beta 1-activated LOXL4 contributes to vascular basement membrane and vascular ECM.Citation28 LOXL4 is also overexpressed during the proliferative phase of the human reproductive cycle and has been associated, by one population study, to endometriosis.Citation29

The LOX family as cancer therapy targets

The discovery of the prominent role played by the LOX family in the ECM coincided with the growing interest in the tumor microenvironment, and today the LOX family is considered a potential cancer therapy target; however, early research proposed LOX as a tumor suppressor. LOX was identified as a Ras rescission gene with reduced expression in Ras transformed fibroblasts, but highly expressed in spontaneous revertants.Citation7,Citation30 The case for the tumor suppressor role of LOX was reinforced by studies showing its epigenetic inactivation in a wide diversity of human gastric cancer cell lines and in gastric tract tumors.Citation31 Later research attributed the antitumor activity of LOX to the peptide cleaved by BMP1 from preproLOX.Citation32 This inhibits the activation of NF-kB and may also repress the transcription of the BCL2 oncogene. On the other hand, LOX and other family members (notably LOXL2) induce epithelial-to-mesenchymal transition (EMT) and enhance invasion.Citation33 Hypoxia, a common condition in tumors, is associated with EMT. Hypoxia induces tumor expression of LOX through hypoxia-inducible factor-1 (HIF-1) to enhance cell-matrix adhesion, migration, invasion, and metastasis.Citation34 Hypoxia also induces stromal expression of LOX, which produces a linearization of collagen I, increases the stiffness of the ECM, and induces a loss of epithelial phenotype in cancer cells, enhancing tumor cell invasion through ECM remodeling, intravasation and metastasis.Citation35,Citation39 In addition, tumor-derived LOX stabilizes Snail1 through a regulatory loop with Notch and HIF-1alpha and is, therefore, directly involved in the regulatory mechanisms of EMTCitation36 and likely participates in the opposite mesenchymal-to-epithelial transition; a step thought to be necessary for target organ colonization.

Head and neck cancer patients with high LOX have a higher probability of metastatic invasion and shorter survival.Citation34 Similarly, LOX expression is associated with increased staging and metastasis in colon cancer and renal cancer patients.Citation37,Citation38 LOX-mediated ECM remodeling seems to be essential for the creation of the “cancer niche”; a location that provides proliferative signals, protection from immune attack, and vascular supply ().Citation39 This mediation seems to be triggered by hypoxia and HIF-1.Citation40 A provocative result obtained along with these findings is the elimination of metastasis by LOX inhibition.Citation34

Figure 2 The cancer niche.

Notes: Just as bone marrow cells require a “niche” to maintain their character and complete their development, cancer cells require a specialized microenvironment to thrive. The cancer niche typically is backed-up by a densely woven ECM formed mainly of collagen I, collagen IV, fibronectin, laminin, and tenascin. This ECM serves as a physical scaffold for the tumor and as a path for stromal cells, such as immune cells and fibroblasts, to migrate along to the tumor mass and, inversely, as a way for tumor cells to escape from the tumor and enter circulation. LOX and LOXL2 expression is induced by tumor hypoxia and mediates the cross-linking of collagen. The LOX family is a central player in cancer related fibrosis, which results in the constant recruitment of protumor stromal cells (ie, cancer-associated inflammation). Additionally, LOXL2 induces epithelial mesenchymal transition, a process that allows tumor cells to acquire fibroblast-like mobility.
Abbreviations: ECM, extracellular matrix; EMT, epithelial mesenchymal transition; LOX, lysyl oxidase; LOXL2, lysyl oxidase-like 2.
Figure 2 The cancer niche.

If a cancer niche is necessary for primary tumor success, then it follows that another is even more necessary to shield metastatic cells conquering a foreign organ. An elegant series of experiments later pushed this idea further by showing the “pre-metastatic niche” ().Citation41,Citation42 These studies demonstrate that primary tumors are able to modify there targets at a distance to foster progression. Both the metastatic and pre-metastatic niches use the mediation of LOX to cross-link collagen I and IV, components of cancer-associated ECM, and targets of the LOX family.Citation43 A dense ECM buttressed by collagens attracts and traps myeloid cells that conform a chemoattractive area located in places usually invaded by circulating cancer cells.Citation44

Figure 3 The premetastatic niche.

Notes: Hypoxic cancer cells produce high levels of secreted LOX that mediates the remodeling of ECM at distant sites of future metastasis, resulting in the attraction of CD11b+ myeloid cells. These stromal cells produce matrix metalloproteinases, urokinase, kallikrein-related peptidases, and cathepsins, which can cleave collagen and other ECM proteins initiating a typical cancer-associated process of ECM destruction and deposition. These alterations create a chemoattractive niche for circulating cancer cells and the ECM remodeling enhances metastatic efficiency by enhancing cell-matrix adhesion and tumor cell proliferation. One unresolved question is the anatomic specificity of LOX mediated premetastatic niche formation. This may be explained by different extracellular matrix components present in organs and different tumor tissues; cancers of the lung, breast, ovary, colon, and liver, among others, are all different regarding ECM components such as fibrin, collagen (different types), vitronectin, fibronectin, laminin, etc.
Abbreviations: ECM, extracellular matrix; KLK, kallikrein-related peptidases; LOX, lysyl oxidase; VEGFR, vascular endothelial growth factor receptors.
Figure 3 The premetastatic niche.

LOXL2 maintains the tumor stroma and the stroma of fibrotic conditions, activating cancer-associated fibroblasts and vasculature in mice and humans.Citation24 Antibody blocking of LOXL2 in mice decreases tumor-induced and chemically-induced fibrosis, improving survival and hepatic function.

There are currently no clinical trials testing for LOX family members against cancer. However, it has been proven that these are inhibitable and “druggable” factors as active site antagonist beta-aminopropionitrile or copper chelator D-penicillamine are able to block the catalytic activity of LOX and LOXL2, albeit nonselectively, and function-blocking antibodies against LOX and LOXL2 have been made.Citation24,Citation35 It must be remembered, however, that extracellular LOX is composed of two parts: the enzyme and its propeptide (PP; LOX-PP). It has been shown that LOX-PP inhibits the extracellular signal-regulated kinase (ERK)-mitogen-activated protein kinase (MAPK) pathway and other pathways associated with cell division during Ewing’s sarcomaCitation4 and has antiproliferative properties, while the active enzyme simultaneously fulfils its protumor functions, as described above. Therefore, anti-LOX therapies must target the cleaved, active enzyme (not its pro-form proLOX or LOX-PP) or, if this is not feasible, then LOX-PP may be used as an additional antitumor agent.

The LOX family as part of a network

One important message from the vast literature on the LOX family is that the lysyl oxidases may induce contradictory effects depending on the tissue studied and on the pathways to which they are associated. Another message is that the same catalytic action, such as the cross-linking of collagen, may not be associated to pathological conditions, but be part of a reaction needed to repair or strengthen a tissue.

A LOX (PP)-Harvey rat sarcoma viral oncogene homolog (HRAS)-nuclear factor kappa B (NFKB) pathway in gastric cancers is associated with tumor suppression,Citation30 but a LOX-FAK-SRC increases cell motility and cell-ECM adhesion,Citation35 which is necessary for local invasion and metastasis. Data from our group suggests that the inhibition of extracellular LOXL2 may hamper or enhance metastasis, depending on the tumor type treated. We are only beginning to scratch at the surface of understanding LOX family function, and further research about the usefulness of the LOX family in aneurysms and vascular defects is needed.

A targeted attack on LOX family synthesis will conceivably affect its antitumoral as well as its protumoral characteristics, and, given the importance of the ECM for cellular behavior, altering LOX family function is potentially fraught with secondary effects. Using LOX as an anti-HRAS agent could slow primary tumor growth, but boost metastasis (a situation similar to that found with other targeted drugs in humans). However, as the tumor suppressor functions of LOX have been shown to be independent of its enzymatic function, selective inhibitors of LOX catalytic activity should leave any tumor suppressor function intact. However, development of such compounds is complicated by the identical nature of the LOX family active sites and by the lack of available crystal structures. Nevertheless, the overexpression of LOX early during in situ breast cancer followed by its repression during invasive carcinoma suggests that it may rewire its molecular connections over time, pushed by dynamic microenvironmental changes.Citation44,Citation45 Thus, much more needs to be learnt about LOX function and dynamics, in both healthy and disease contexts, before effective targeting can begin.

Network medicine may provide a way to tackle these problems.Citation46 It has been demonstrated that a systems biology simulation of epidermal growth factor receptor signaling variation in vitro will produce a combined therapy cycle that maximizes effect and minimizes the emergence of resistance.Citation47 The simulation predicts the rewiring of molecular networks associated with cancer proliferation, allowing for therapy that prevents resistance. However, predicting and attacking networks involved in the running of the cancer microenvironment is an unmet challenge. Knowledge may be gained by using omics approaches to understand how LOX connects to molecules produced locally and how molecules in the primary tumor interact with those in metastatic target organs. Targeting molecular networks involving LOX family members may allow for dissection and attack of their protumoral activities, while keeping side-effects and resistance at a minimum.

Conclusion

While it is clear from the extensive research that the LOX family are potent anticancer targets, strategies for combination therapies and for preventing resistance have not been investigated. Despite this, clinical trials in cancer patients are due to begin with the LOXL2 monoclonal antibody, and several LOX/LOXL2 drug development programs are underway, quickly approaching clinical trials in patients.

Acknowledgments

AMG is supported by funding from the Association for International Cancer Research and the Danish Cancer Society. JTE is supported by a Hallas Møller Stipend from the Novo Nordisk Foundation, and Biotech Research and Innovation Centre (BRIC).

Disclosure

The authors report no conflicts of interest in this work.

References

  • LuPWeaverVMWerbZThe extracellular matrix: a dynamic niche in cancer progressionJ Cell Biol2012196439540622351925
  • GritsenkoPGIlinaOFriedlPInterstitial guidance of cancer invasionJ Pathol2012226218519922006671
  • BarkerHECoxTRErlerJTThe rationale for targeting the LOX family in cancerNature Cancer Rev2012128540552
  • AgraNCidreFGarcía-GarcíaLde la ParraJAlonsoJLysysl oxidase is downregulated by the EWS/FLI1 oncoprotein and its propeptide domain displys tumor suppressor activities in ewing sarcoma cellsPLoS One201386e6628123750284
  • PinnellSRMartinGRThe cross-linking of collagen and elastin: enzymatic conversion of lysine in peptide lynkage to alpha-aminoadipic-delta-semialdehyde (allysine) by an extract from boneProc Natl Acad Sci U S A19686127087165246001
  • HamalainenERKemppainenRKuivaniemiHQuatitative polymerase chain reaction of lysil oxidase mRNA in malignantly transformed human cell liness demonstrates that their low lysyl oxidase activity is due to low quantities of its mRNA and low levels of transcription of the respective geneJ Biol Chem19952703721590215937665572
  • ContenteSKenyonKRimoldiDFriedmanRMExpression of gene rrg is associated with reversion of NIH 3T3 transformed by LTR-c-H-rasScience199024949707967981697103
  • UzelMIScottICBabakhanlou-ChaseHMultiple bone morphogenetic protein 1-related mammalian metalloproteinases process pro-lysyl oxidase at the correct physiological site and control lysyl oxidase activation in mouse embryo fibroblast culturesJ Biol Chem200127625225372254311313359
  • KaganHMLiWLysyl oxidase: properties, specificity, and biological roles inside and outside of the cellJ Cell Biochem200388466067212577300
  • MolnarJFongKSHeQPStructural and functional diversity of lysyl oxidase and the LOX-like proteinsBiochim Biophys Acta200316471–222022412686136
  • MäkiJMLysyl oxidases in mammalian development and certain pathological conditionsHistol Histopathol200924565166019283672
  • CoxTRBirdDBakerAMLOX-mediated collagen crosslinking is responsible for fibrosis-enhanced metastasisCancer Res20137361721173223345161
  • MäkiJMSormunenRLippoSKaarteenaho-WiikRSoininenRMyllyharjuJLysyl oxidase is essential for normal development and function of the respiratory system and for the integrity of elastic and collagen fibres in various tissuesAm J Pathol2005167492793616192629
  • KuivaniemiHPeltonenLKivirikkoKIType IX Ehlers-Danlos syndrome and Menkes syndrome: the decrease in lysyl oxidase activity is associated with a corresponding deficiency in the enzyme proteinAm J Human Genet19853747988089556668
  • YeowellHNWalkerLCMutations in the lysyl hydroxylase 1 gene that result in enzyme deficiency and the clinical phenotype of Ehlers–Danlos syndrome type VIMol Genet Metab2000711–221222411001813
  • LiPAHeQCaoTUp-regulation and altered distribution of lysyl oxidase in the centrak nervous system of mutant SOD1 transgenic mouse model of amyotrophic lateral sclerosisBrain Res Mol Brain Res2004120211512214741400
  • GiladGMKaganHMGiladVHEvidence for increased lysyl oxidase, the extracellular matrix-forming enzyme, in Alzheimer’s disease brainNeurosci Lett2005376321021415721223
  • LiuXZhaoYGaoJElastic fiber homeostasis leads to pelvic floor disordersNat Genet200436217818214745449
  • Samsung Medical CenterLOXL1 polymorphism in pseudoexfoliation syndrome Available from: http://clinicaltrials.gov/show/NCT01515735. NLM identifier: NCT01515735Accessed July 1, 2013
  • TarkanenAReunanenAKveläTFrequency of systemic vascular diseases in patients with primary open angle glaucoma and exfoliation glaucomaActa Ophtamol2008866598602
  • KimYMKimEKimYThe human lysyl oxidase-like 2 protein functions as an amine oxidase toward collagen and elastinMol Bio Rep201138114514920306300
  • IftikharMHurtadoPBaisMVlysyl oxidase-like-2 (loxl2) is a major isoform in chondrocytes and is critically required for differentiationJ Biol Chem201128690991821071451
  • VadaszZKesslerOAkiriGAbnormal deposition of collagen around hepatocytes in Wilson’s disease is associated with hepatocyte specific expression of lysyl oxidase and lysyl oxidase like 2J Hepatol200543349950716023247
  • Barry-HamiltonVSpanglerRMarshallDAllosteric inhibition of lysyl oxidase-like-2 impedes the development of a pathologic microenvironmentNat Med20101691009101720818376
  • LeeJEKimYA Tissue-specific variant of the human lysyl oxidase-like protein 3 (LOXL3) functions as an amine oxidase with substrate specificityJ Biol Chem200628149372823729017018530
  • YuQHorakKLarsonDFRole of T lymphocytes in hypertension-induced cardiac extracellular matrix remodellingHypertension20064819810416735642
  • ItoHAkiyamaHIguchiHMolecular cloning and biological activity of a novel lysyl oxidase-related gene expressed in cartilageJ Biol Chem200127626240232402911292829
  • BusnadiegoOGonzález-SantamaríaJLagaresDLOXL4 is induced by transforming growth factor β1 through Smad and JunB/Fra2 and contributes to vascular matrix remodelingMol Cell Biol201333122388240123572561
  • LaRuizDutilJRuizASingle-nucleotide polymorphisms in the lysyl oxidase-like protein 4 and complement component 3 genes are associated with increased risk for endometriosis and endometriosis-associated infertilityFertil Steril201196251251521733505
  • PalamakumburaAHJeaySGuoYThe propeptide domain of lysyl oxidase induces phenotypic reversion of ras-transformed cellsJ Biol Chem200427939405934060015277520
  • KanedaAWakazonoKTsukamotoTLysyl oxidase is a tumor suppressor gene inactivated by methylation and loss of heterozygosity in human gastric cancersCancer Res200464186410641515374948
  • WuMMinCWangXRepression of BCL2 by the tumor suppressor activity of the lysyl oxidase propeptide inhibits transformed phenotype of lung and pancreatic cancer cellsCancer Res200767136278628517616686
  • CanoASantamaríaPGMoreno-BuenoGLOXL2 in epithelial cell plasticity and tumor progressionFuture Oncol2012891095110823030485
  • ErlerJTBennewithKLNicolauMLysyl oxidase is essential for hypoxia-induced metastasisNature200644070881222122616642001
  • PickupMWLaklaiHAcerbiIStromally derived lysyl oxidase promotes metastasis of transforming growth factor-β-deficient mouse mammary carcinomasCancer Res201373175336534623856251
  • SahlgrenCGustafssonMVJinSPoellingerLLendahlUNotch signaling mediates hypoxia-induced tumor cell migration and invasionProc Natl Acad Sci U S A2008105176392639718427106
  • BakerAMCoxTRBirdDThe role of lysyl oxidase in SRC-dependent proliferation and metastasis of colorectal cancerJ Natl Cancer Inst2011103540742421282564
  • KirschmannDASeftorEAFongSFA molecular role for lysyl oxidase in breast cancer invasionCancer Res200262154478448312154058
  • LeventalKRYuHKassLMatrix crosslinking forces tumor progression by enhancing integrin signallingCell2009139589190619931152
  • SchietkeRWarneckeCWackerIThe lysyl oxidases LOX and LOXL2 are necessary and sufficient to repress E-cadherin in hypoxia: insights into cellular transformation processes mediated by HIF-1J Biol Chem201028596658666920026874
  • KaplanRNRibaRDZacharoulisSVEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic nicheNature2005438706982082716341007
  • ErlerJTBennewithKLCoxTRHypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic nicheCancer Cell2009151354419111879
  • WongCCGilkesDMZhangHHypoxia-inducible factor 1 is a master regulator of breast cancer metastatic niche formationProc Natl Acad Sci U S A201110839163691637421911388
  • DecitreMGleyzalCRaccurtMLysyl oxidase-like protein localizes to sites of de novo fibrinogenesis in fibrosis and in the early stroma reaction of ductal breast carcinomasLab Invest19987821431519484712
  • PeyrolSRaccurtMGerardFGleyzalCGrimaudJASommerPLysyl oxidase gene expression in the stromal reaction to in situ and invasive ductal breast carcinomaAm J Pathol199715024975079033266
  • PawsonTLindingRNetwork MedicineFEBS Lett200858281266127018282479
  • LeeMJYeASGardinoAKSequential et alCell201249478079422579283