Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Cell ; 181(6): 1346-1363.e21, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32473126

ABSTRACT

Enhanced blood vessel (BV) formation is thought to drive tumor growth through elevated nutrient delivery. However, this observation has overlooked potential roles for mural cells in directly affecting tumor growth independent of BV function. Here we provide clinical data correlating high percentages of mural-ß3-integrin-negative tumor BVs with increased tumor sizes but no effect on BV numbers. Mural-ß3-integrin loss also enhances tumor growth in implanted and autochthonous mouse tumor models with no detectable effects on BV numbers or function. At a molecular level, mural-cell ß3-integrin loss enhances signaling via FAK-p-HGFR-p-Akt-p-p65, driving CXCL1, CCL2, and TIMP-1 production. In particular, mural-cell-derived CCL2 stimulates tumor cell MEK1-ERK1/2-ROCK2-dependent signaling and enhances tumor cell survival and tumor growth. Overall, our data indicate that mural cells can control tumor growth via paracrine signals regulated by ß3-integrin, providing a previously unrecognized mechanism of cancer growth control.


Subject(s)
Integrin beta3/metabolism , Neoplasms/metabolism , Tumor Burden/physiology , Animals , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Female , Humans , Male , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
2.
Development ; 149(13)2022 07 01.
Article in English | MEDLINE | ID: mdl-35723257

ABSTRACT

Precise vascular patterning is crucial for normal growth and development. The ERG transcription factor drives Delta-like ligand 4 (DLL4)/Notch signalling and is thought to act as a pivotal regulator of endothelial cell (EC) dynamics and developmental angiogenesis. However, molecular regulation of ERG activity remains obscure. Using a series of EC-specific focal adhesion kinase (FAK)-knockout (KO) and point-mutant FAK-knock-in mice, we show that loss of ECFAK, its kinase activity or phosphorylation at FAK-Y397, but not FAK-Y861, reduces ERG and DLL4 expression levels together with concomitant aberrations in vascular patterning. Rapid immunoprecipitation mass spectrometry of endogenous proteins identified that endothelial nuclear-FAK interacts with the deubiquitinase USP9x and the ubiquitin ligase TRIM25. Further in silico analysis confirms that ERG interacts with USP9x and TRIM25. Moreover, ERG levels are reduced in FAKKO ECs via a ubiquitin-mediated post-translational modification programme involving USP9x and TRIM25. Re-expression of ERG in vivo and in vitro rescues the aberrant vessel-sprouting defects observed in the absence of ECFAK. Our findings identify ECFAK as a regulator of retinal vascular patterning by controlling ERG protein degradation via TRIM25/USP9x.


Subject(s)
Endothelial Cells , Transcription Factors , Animals , Endothelial Cells/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Mice , Neovascularization, Physiologic/genetics , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitins/metabolism
3.
Angiogenesis ; 24(3): 471-482, 2021 08.
Article in English | MEDLINE | ID: mdl-33730293

ABSTRACT

Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that is overexpressed in many cancer types and in vivo studies have shown that vascular endothelial cell FAK expression and FAK-phosphorylation at tyrosine (Y) 397, and subsequently FAK-Y861, are important in tumour angiogenesis. Pericytes also play a vital role in regulating tumour blood vessel stabilisation, but the specific involvement of pericyte FAK-Y397 and FAK-Y861 phosphorylation in tumour blood vessels is unknown. Using PdgfrßCre + ;FAKWT/WT, PdgfrßCre + ;FAKY397F/Y397F and PdgfrßCre + ;FAKY861F/Y861F mice, our data demonstrate that Lewis lung carcinoma tumour growth, tumour blood vessel density, blood vessel perfusion and pericyte coverage were affected only in late stage tumours in PdgfrßCre + ;FAKY861F/Y861F but not PdgfrßCre + ;FAKY397F/Y397F mice. Further examination indicates a dual role for pericyte FAK-Y861 phosphorylation in the regulation of tumour vessel regression and also in the control of pericyte derived signals that influence apoptosis in cancer cells. Overall this study identifies the role of pericyte FAK-Y861 in the regulation of tumour vessel regression and tumour growth control and that non-phosphorylatable FAK-Y861F in pericytes reduces tumour growth and blood vessel density.


Subject(s)
Apoptosis , Carcinoma, Lewis Lung , Focal Adhesion Kinase 1 , Mutation, Missense , Neoplasm Proteins , Neovascularization, Pathologic , Pericytes/enzymology , Amino Acid Substitution , Animals , Carcinoma, Lewis Lung/enzymology , Carcinoma, Lewis Lung/genetics , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/genetics , Phosphorylation
4.
Nature ; 514(7520): 112-6, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25079333

ABSTRACT

Chemoresistance is a serious limitation of cancer treatment. Until recently, almost all the work done to study this limitation has been restricted to tumour cells. Here we identify a novel molecular mechanism by which endothelial cells regulate chemosensitivity. We establish that specific targeting of focal adhesion kinase (FAK; also known as PTK2) in endothelial cells is sufficient to induce tumour-cell sensitization to DNA-damaging therapies and thus inhibit tumour growth in mice. The clinical relevance of this work is supported by our observations that low blood vessel FAK expression is associated with complete remission in human lymphoma. Our study shows that deletion of FAK in endothelial cells has no apparent effect on blood vessel function per se, but induces increased apoptosis and decreased proliferation within perivascular tumour-cell compartments of doxorubicin- and radiotherapy-treated mice. Mechanistically, we demonstrate that endothelial-cell FAK is required for DNA-damage-induced NF-κB activation in vivo and in vitro, and the production of cytokines from endothelial cells. Moreover, loss of endothelial-cell FAK reduces DNA-damage-induced cytokine production, thus enhancing chemosensitization of tumour cells to DNA-damaging therapies in vitro and in vivo. Overall, our data identify endothelial-cell FAK as a regulator of tumour chemosensitivity. Furthermore, we anticipate that this proof-of-principle data will be a starting point for the development of new possible strategies to regulate chemosensitization by targeting endothelial-cell FAK specifically.


Subject(s)
DNA Damage , Drug Resistance, Neoplasm/drug effects , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cytokines/biosynthesis , DNA Damage/drug effects , DNA Damage/genetics , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm/genetics , Endothelial Cells/metabolism , Focal Adhesion Protein-Tyrosine Kinases/deficiency , Focal Adhesion Protein-Tyrosine Kinases/genetics , Humans , Mice , NF-kappa B/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/radiotherapy , Phosphorylation/drug effects
5.
Eur Heart J ; 40(30): 2507-2520, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31162546

ABSTRACT

AIMS: A better understanding of the pathways that regulate regeneration of the coronary vasculature is of fundamental importance for the advancement of strategies to treat patients with heart disease. Here, we aimed to investigate the origin and clonal dynamics of endothelial cells (ECs) associated with neovascularization in the adult mouse heart following myocardial infarction (MI). Furthermore, we sought to define murine cardiac endothelial heterogeneity and to characterize the transcriptional profiles of pro-angiogenic resident ECs in the adult mouse heart, at single-cell resolution. METHODS AND RESULTS: An EC-specific multispectral lineage-tracing mouse (Pdgfb-iCreERT2-R26R-Brainbow2.1) was used to demonstrate that structural integrity of adult cardiac endothelium following MI was maintained through clonal proliferation by resident ECs in the infarct border region, without significant contributions from bone marrow cells or endothelial-to-mesenchymal transition. Ten transcriptionally discrete heterogeneous EC states, as well as the pathways through which each endothelial state is likely to enhance neovasculogenesis and tissue regeneration following ischaemic injury were defined. Plasmalemma vesicle-associated protein (Plvap) was selected for further study, which showed an endothelial-specific and increased expression in both the ischaemic mouse and human heart, and played a direct role in regulating human endothelial proliferation in vitro. CONCLUSION: We present a single-cell gene expression atlas of cardiac specific resident ECs, and the transcriptional hierarchy underpinning endogenous vascular repair following MI. These data provide a rich resource that could assist in the development of new therapeutic interventions to augment endogenous myocardial perfusion and enhance regeneration in the injured heart.


Subject(s)
Gene Expression Profiling/methods , Myocardial Infarction/metabolism , Neovascularization, Physiologic/genetics , Single-Cell Analysis/methods , Transcriptome/genetics , Animals , Cell Proliferation/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Mice , Mice, Transgenic , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology
6.
J Cell Sci ; 130(9): 1583-1595, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28289267

ABSTRACT

The α6ß1-integrin is a major laminin receptor, and formation of a laminin-rich basement membrane is a key feature in tumour blood vessel stabilisation and pericyte recruitment, processes that are important in the growth and maturation of tumour blood vessels. However, the role of pericyte α6ß1-integrin in angiogenesis is largely unknown. We developed mice where the α6-integrin subunit is deleted in pericytes and examined tumour angiogenesis and growth. These mice had: (1) reduced pericyte coverage of tumour blood vessels; (2) reduced tumour blood vessel stability; (3) increased blood vessel diameter; (4) enhanced blood vessel leakiness, and (5) abnormal blood vessel basement membrane architecture. Surprisingly, tumour growth, blood vessel density and metastasis were not altered. Analysis of retinas revealed that deletion of pericyte α6-integrin did not affect physiological angiogenesis. At the molecular level, we provide evidence that pericyte α6-integrin controls PDGFRß expression and AKT-mTOR signalling. Taken together, we show that pericyte α6ß1-integrin regulates tumour blood vessels by both controlling PDGFRß and basement membrane architecture. These data establish a novel dual role for pericyte α6-integrin as modulating the blood vessel phenotype during pathological angiogenesis.


Subject(s)
Blood Vessels/metabolism , Integrin alpha6beta1/metabolism , Neoplasms/blood supply , Pericytes/metabolism , Animals , Basement Membrane/drug effects , Basement Membrane/metabolism , Becaplermin , Blood Vessels/drug effects , Blood Vessels/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Integrases/metabolism , Mice , Neoplasm Metastasis , Neoplasms/metabolism , Neoplasms/pathology , Pericytes/drug effects , Proto-Oncogene Proteins c-sis/pharmacology , Receptor, Platelet-Derived Growth Factor beta/metabolism
8.
Circ Res ; 114(1): 79-91, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24103390

ABSTRACT

RATIONALE: The dramatic upregulation of αvß3-integrin that occurs in the vasculature during tumor growth has long suggested that the endothelial expression of this molecule is an ideal target for antiangiogenic therapy to treat cancer. This discovery led to the development of small-molecule inhibitors directed against αvß3-integrin that are currently in clinical trials. In 2002, we reported that ß3-integrin-knockout mice exhibit enhanced tumor growth and angiogenesis. However, as ß3-integrin is expressed by a wide variety of cells, endothelial cell-specific contributions to tumor angiogenesis are muddied by the use of a global knockout of ß3-integrin function. OBJECTIVE: Our aim was to examine the endothelial-specific contribution ß3-integrin makes to tumor growth and angiogenesis. METHODS AND RESULTS: We have crossed ß3-integrin-floxed (ß3-floxed) mice to 2 endothelial-specific Cre models and examined angiogenic responses in vivo, ex vivo, and in vitro. We show that acute depletion of endothelial ß3-integrin inhibits tumor growth and angiogenesis preventatively, but not in already established tumors. However, the effects are transient, and long-term depletion of the molecule is ineffective. Furthermore, long-term depletion of the molecule correlates with many molecular changes, such as reduced levels of focal adhesion kinase expression and a misbalance in focal adhesion kinase phosphorylation, which may lead to a release from the inhibitory effects of decreased endothelial ß3-integrin expression. CONCLUSIONS: Our findings imply that timing and length of inhibition are critical factors that need to be considered when targeting the endothelial expression of ß3-integrin to inhibit tumor growth and angiogenesis.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Integrin alphaVbeta3/genetics , Neoplasms, Experimental/blood supply , Neovascularization, Pathologic/metabolism , Animals , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Integrin alphaVbeta3/metabolism , Lung/blood supply , Lung/pathology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/pathology
9.
Nature ; 465(7299): 813-7, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20535211

ABSTRACT

Down's syndrome (DS) is a genetic disorder caused by full or partial trisomy of human chromosome 21 and presents with many clinical phenotypes including a reduced incidence of solid tumours. Recent work with the Ts65Dn model of DS, which has orthologues of about 50% of the genes on chromosome 21 (Hsa21), has indicated that three copies of the ETS2 (ref. 3) or DS candidate region 1 (DSCR1) genes (a previously known suppressor of angiogenesis) is sufficient to inhibit tumour growth. Here we use the Tc1 transchromosomic mouse model of DS to dissect the contribution of extra copies of genes on Hsa21 to tumour angiogenesis. This mouse expresses roughly 81% of Hsa21 genes but not the human DSCR1 region. We transplanted B16F0 and Lewis lung carcinoma tumour cells into Tc1 mice and showed that growth of these tumours was substantially reduced compared with wild-type littermate controls. Furthermore, tumour angiogenesis was significantly repressed in Tc1 mice. In particular, in vitro and in vivo angiogenic responses to vascular endothelial growth factor (VEGF) were inhibited. Examination of the genes on the segment of Hsa21 in Tc1 mice identified putative anti-angiogenic genes (ADAMTS1and ERG) and novel endothelial cell-specific genes, never previously shown to be involved in angiogenesis (JAM-B and PTTG1IP), that, when overexpressed, are responsible for inhibiting angiogenic responses to VEGF. Three copies of these genes within the stromal compartment reduced tumour angiogenesis, explaining the reduced tumour growth in DS. Furthermore, we expect that, in addition to the candidate genes that we show to be involved in the repression of angiogenesis, the Tc1 mouse model of DS will permit the identification of other endothelium-specific anti-angiogenic targets relevant to a broad spectrum of cancer patients.


Subject(s)
Carcinoma, Lewis Lung/blood supply , Disease Models, Animal , Down Syndrome/genetics , Gene Dosage/genetics , Melanoma, Experimental/blood supply , Neovascularization, Pathologic/genetics , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAMTS1 Protein , Animals , Carcinoma, Lewis Lung/complications , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Chromosomes, Mammalian/genetics , Down Syndrome/complications , Down Syndrome/physiopathology , Female , Humans , Immunoglobulins/genetics , Immunoglobulins/metabolism , Intracellular Signaling Peptides and Proteins , Male , Melanoma, Experimental/complications , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Proto-Oncogene Protein c-ets-2/genetics , Proto-Oncogene Protein c-ets-2/metabolism , Transcription Factors , Transcriptional Regulator ERG , Trisomy/genetics , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
10.
Int J Cancer ; 133(10): 2315-24, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23681936

ABSTRACT

The extracellular protease ADAMTS1 (A disintegrin and metalloprotease with thrombospondin repeats 1) has been described as an anti-angiogenic molecule and its role as a putative tumor protective molecule has also been suggested. Here, we have used a tumor xenograft model to determine the role of ADAMTS1 in tumor growth and angiogenesis. Increasing levels of the protease led to the complete inhibition of tumor growth. In an attempt to elucidate the mechanism of action of this protease, we focused our attention on its proteolytic activity on nidogens, one of the main components of the vascular basement membrane. The increased expression of ADAMTS1 was accompanied by increased proteolysis of nidogen-1 and -2 and their almost complete removal from vascular structures, together with major morphological alterations of tumor blood vessels and a decreased vessel density. The clinical relevance of this work is supported by our observations that ADAMTS1 expression is decreased in breast tumor specimens when compared with healthy tissue. Our studies also reveal that the cleavage of nidogen-1 and -2 is partially inhibited in human tumor samples. Moreover, the deposition of both nidogens surrounding vascular structures is drastically altered, implying a possible reduction in the maintenance of vessel integrity. Our studies reflect the requirement to explore the functional interactions between proteases and specific substrates in cancer biology.


Subject(s)
ADAM Proteins/metabolism , Breast Neoplasms/genetics , Cell Adhesion Molecules/metabolism , Genes, Tumor Suppressor , Membrane Glycoproteins/metabolism , Peptide Hydrolases/metabolism , ADAM Proteins/genetics , ADAMTS1 Protein , Animals , Basement Membrane/metabolism , Basement Membrane/pathology , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcium-Binding Proteins , Cell Adhesion Molecules/genetics , Cell Line , Down-Regulation , Glycosaminoglycans/genetics , Glycosaminoglycans/metabolism , Glycosaminoglycans/physiology , HEK293 Cells , Humans , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Proteolysis , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
11.
FEBS J ; 2023 Oct 24.
Article in English | MEDLINE | ID: mdl-37873605

ABSTRACT

Cellular senescence is a state of durable cell arrest that has been identified both in vitro and in vivo. It is associated with profound changes in gene expression and a specific secretory profile that includes pro-inflammatory cytokines, growth factors and matrix-remodelling enzymes, referred to as the senescence-associated secretory phenotype (SASP). In cancer, senescence can have anti- or pro-tumour effects. On one hand, it can inhibit tumour progression in a cell autonomous manner. On the other hand, senescence can also promote tumour initiation, progression, metastatic dissemination and resistance to therapy in a paracrine manner. Therefore, despite efforts to target senescence as a potential strategy to inhibit tumour growth, senescent cancer and microenvironmental cells can eventually lead to uncontrolled proliferation and aggressive tumour phenotypes. This can happen either through overcoming senescence growth arrest or through SASP-mediated effects in adjacent tumour cells. This review will discuss how senescence affects the tumour microenvironment, including extracellular matrix remodelling, the immune system and the vascular compartment, to promote tumourigenesis, metastasis and resistance to DNA-damaging therapies. It will also discuss current approaches used in the field to target senescence: senolytics, improving the immune clearance of senescent cells and targeting the SASP.

12.
J Exp Med ; 220(1)2023 01 02.
Article in English | MEDLINE | ID: mdl-36269299

ABSTRACT

Primary tumors and distant site metastases form a bidirectionally communicating system. Yet, the molecular mechanisms of this crosstalk are poorly understood. Here, we identified the proteolytically cleaved fragments of angiopoietin-like 4 (ANGPTL4) as contextually active protumorigenic and antitumorigenic contributors in this communication ecosystem. Preclinical studies in multiple tumor models revealed that the C-terminal fragment (cANGPTL4) promoted tumor growth and metastasis. In contrast, the N-terminal fragment of ANGPTL4 (nANGPTL4) inhibited metastasis and enhanced overall survival in a postsurgical metastasis model by inhibiting WNT signaling and reducing vascularity at the metastatic site. Tracing ANGPTL4 and its fragments in tumor patients detected full-length ANGPTL4 primarily in tumor tissues, whereas nANGPTL4 predominated in systemic circulation and correlated inversely with disease progression. The study highlights the spatial context of the proteolytic cleavage-dependent pro- and antitumorigenic functions of ANGPTL4 and identifies and validates nANGPTL4 as a novel biomarker of tumor progression and antimetastatic therapeutic agent.


Subject(s)
Angiopoietin-Like Protein 4 , Neoplasms , Humans , Angiopoietin-Like Protein 4/pharmacology , Angiopoietin-Like Protein 4/therapeutic use , Angiopoietins/pharmacology , Angiopoietins/therapeutic use , Biomarkers, Tumor , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/metabolism , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use
13.
Development ; 136(23): 4043-53, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19906871

ABSTRACT

Sprouting angiogenesis and lymphatic-blood vessel segregation both involve the migration of endothelial cells, but the precise migratory molecules that govern the decision of blood vascular endothelial cells to segregate into lymphatic vasculature are unknown. Here, we deleted endothelial Rac1 in mice (Tie1-Cre(+);Rac1(fl/fl)) and revealed, unexpectedly, that whereas blood vessel morphology appeared normal, lymphatic-blood vessel separation was impaired, with corresponding edema, haemorrhage and embryonic lethality. Importantly, normal levels of Rac1 were essential for directed endothelial cell migratory responses to lymphatic-inductive signals. Our studies identify Rac1 as a crucial part of the migratory machinery required for endothelial cells to separate and form lymphatic vasculature.


Subject(s)
Blood Vessels/metabolism , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Lymphatic Vessels/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Cell Separation/methods , Cells, Cultured , Embryo, Mammalian , Endothelium, Vascular/cytology , Endothelium, Vascular/embryology , Endothelium, Vascular/metabolism , Fluorescent Antibody Technique, Direct , Fluorescent Dyes/metabolism , Galactosides/metabolism , Gene Deletion , Immunohistochemistry , Indoles/metabolism , Mice , Mice, Transgenic , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , RNA, Small Interfering/metabolism , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Transfection , beta-Galactosidase/metabolism , rac1 GTP-Binding Protein/analysis , rac1 GTP-Binding Protein/genetics
14.
J Clin Invest ; 118(3): 965-74, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18246199

ABSTRACT

Effective reepithelialization after injury is essential for correct wound healing. The upregulation of keratinocyte alpha3beta1 integrin during reepithelialization suggests that this adhesion molecule is involved in wound healing; however, its precise role in this process is unknown. We have shown here that retarded reepithelialization in Itga3(-/-) mouse skin wounds is due predominantly to repressed TGF-beta1-mediated responses. Specifically, expression of the inhibitor of TGF-beta1-signaling Smad7 was elevated in Itga3(-/-) keratinocytes. Indeed, in vivo blockade of Smad7 increased the rate of reepithelialization in Itga3(-/-) and WT wounds to similar levels. Our data therefore indicate that the function of alpha3beta1 integrin as a mediator of keratinocyte migration is not essential for reepithelialization but suggest instead that alpha3beta1 integrin has a major new in vivo role as an inhibitor of Smad7 during wound healing. Moreover, our study may identify a previously undocumented function for Smad7 as a regulator of reepithelialization in vivo and implicates Smad7 as a potential novel target for the treatment of cutaneous wounds.


Subject(s)
Epithelium/physiology , Integrin alpha3beta1/physiology , Smad7 Protein/physiology , Wound Healing , Animals , Integrin alpha5beta1/physiology , Mice , Signal Transduction , Transforming Growth Factor beta1/physiology
15.
Am J Pathol ; 177(3): 1534-48, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20639457

ABSTRACT

Integrin alpha3beta1 is a major receptor for laminin. The expression levels of laminins-8 and -10 in the basement membrane surrounding blood vessels are known to change during tumor angiogenesis. Although some studies have suggested that certain ligands of alpha3beta1 can affect angiogenesis either positively or negatively, either a direct in vivo role for alpha3beta1 in this process or its mechanism of action in endothelial cells during angiogenesis is still unknown. Because the global genetic ablation of alpha3-integrin results in an early lethal phenotype, we have generated conditional-knockout mice where alpha3 is deleted specifically in endothelial cells (ec-alpha3-/-). Here we show that ec-alpha3-/- mice are viable, fertile, and display enhanced tumor growth, elevated tumor angiogenesis, augmented hypoxia-induced retinal angiogenesis, and increased vascular endothelial growth factor (VEGF)-mediated neovascularization ex vivo and in vivo. Furthermore, our data provide a novel method by which an integrin may regulate angiogenesis. We show that alpha3beta1 is a positive regulator of endothelial-VEGF and that, surprisingly, the VEGF produced by endothelial cells can actually repress VEGF-receptor 2 (Flk-1) expression. These data, therefore, identify directly that endothelial alpha3beta1 negatively regulates pathological angiogenesis and implicate an unexpected role for low levels of endothelial-VEGF as an activator of neovascularization.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Hypoxia/metabolism , Integrin alpha3beta1/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Blotting, Western , Endothelial Cells/pathology , Endothelium, Vascular/pathology , Female , Flow Cytometry , Hypoxia/genetics , Hypoxia/pathology , Immunohistochemistry , Integrin alpha3beta1/genetics , Male , Mice , Mice, Knockout , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Polymerase Chain Reaction , Vascular Endothelial Growth Factor Receptor-2/metabolism
16.
J Pathol ; 220(4): 435-45, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19967726

ABSTRACT

beta3-Integrin is a cell surface adhesion and signalling molecule important in the regulation of tumour angiogenesis. Mice with a global deficiency in beta3-integrin show increased pathological angiogenesis, most likely due to increased vascular endothelial growth factor receptor 2 expression on beta3-null endothelial cells. Here we transplanted beta3-null bone marrow (BM) into wild-type (WT) mice to dissect the role of BM beta3-integrin deficiency in pathological angiogenesis. Mice transplanted with beta3-null bone marrow show significantly enhanced angiogenesis in subcutaneous B16F0 melanoma and Lewis lung carcinoma (LLC) cell models and in B16F0 melanoma lung metastasis when compared with tumours grown in mice transplanted with WT bone marrow. The effect of bone marrow beta3-integrin deficiency was also assessed in the RIPTAg mouse model of pancreatic tumour growth. Again, angiogenesis in mice lacking BM beta3-integrin was enhanced. However, tumour weight between the groups was not significantly altered, suggesting that the enhanced blood vessel density in the mice transplanted with beta3-null bone marrow was not functional. Indeed, we demonstrate that in mice transplanted with beta3-null bone marrow a significant proportion of tumour blood vessels are non-functional when compared with tumour blood vessels in WT-transplanted controls. Furthermore, beta3-null-transplanted mice showed an increased angiogenic response to VEGF in vivo when compared with WT-transplanted animals. BM beta3-integrin deficiency affects the mobilization of progenitor cells to the peripheral circulation. We show that VEGF-induced mobilization of endothelial progenitor cells is enhanced in mice transplanted with beta3-null bone marrow when compared with WT-transplanted controls, suggesting a possible mechanism underlying the increased blood vessel density seen in beta3-null-transplanted mice. In conclusion, although BM beta3-integrin is not required for pathological angiogenesis, our studies demonstrate a role for BM beta3-integrin in VEGF-induced mobilization of bone marrow-derived cells to the peripheral circulation and for the functionality of those vessels in which BM-derived cells become incorporated.


Subject(s)
Bone Marrow/metabolism , Integrin beta3/physiology , Neoplasms, Experimental/blood supply , Neovascularization, Pathologic/metabolism , Animals , Bone Marrow Transplantation , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/metabolism , Cell Movement/physiology , Endothelial Cells/physiology , Female , Hematopoietic Stem Cells/physiology , Lung Neoplasms/blood supply , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Melanoma/blood supply , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/blood supply , Stem Cells/physiology , Vascular Endothelial Growth Factor A/toxicity
17.
Nat Med ; 8(1): 27-34, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11786903

ABSTRACT

Inhibition of alphavbeta3 or alphavbeta5 integrin function has been reported to suppress neovascularization and tumor growth, suggesting that these integrins are critical modulators of angiogenesis. Here we report that mice lacking beta3 integrins or both beta3 and beta5 integrins not only support tumorigenesis, but have enhanced tumor growth as well. Moreover, the tumors in these integrin-deficient mice display enhanced angiogenesis, strongly suggesting that neither beta3 nor beta5 integrins are essential for neovascularization. We also observed that angiogenic responses to hypoxia and vascular endothelial growth factor (VEGF) are augmented significantly in the absence of beta3 integrins. We found no evidence that the expression or functions of other integrins were altered as a consequence of the beta3 deficiency, but we did observe elevated levels of VEGF receptor-2 (also called Flk-1) in beta3-null endothelial cells. These data indicate that alphavbeta3 and alphavbeta5 integrins are not essential for vascular development or pathological angiogenesis and highlight the need for further evaluation of the mechanisms of action of alphav-integrin antagonists in anti-angiogenic therapeutics.


Subject(s)
Integrin beta Chains , Integrins/deficiency , Neovascularization, Pathologic/etiology , Platelet Membrane Glycoproteins/deficiency , Animals , Antigens, CD/genetics , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/drug effects , Integrin beta3 , Integrins/genetics , Integrins/metabolism , Lymphokines/pharmacology , Mice , Mice, Mutant Strains , Neoplasms, Experimental/blood supply , Platelet Membrane Glycoproteins/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Receptors, Vitronectin/metabolism , Retinal Neovascularization , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
18.
Cancers (Basel) ; 13(20)2021 Oct 17.
Article in English | MEDLINE | ID: mdl-34680355

ABSTRACT

Several strategies have been developed to modulate the tumour vasculature for cancer therapy including anti-angiogenesis and vascular normalisation. Vasculature modulation results in changes to the tumour microenvironment including oxygenation and immune cell infiltration, therefore lending itself to combination with cancer therapy. The development of immunotherapies has led to significant improvements in cancer treatment. Particularly promising are immune checkpoint blockade and CAR T cell therapies, which use antibodies against negative regulators of T cell activation and T cells reprogrammed to better target tumour antigens, respectively. However, while immunotherapy is successful in some patients, including those with advanced or metastatic cancers, only a subset of patients respond. Therefore, better predictors of patient response and methods to overcome resistance warrant investigation. Poor, or periphery-limited, T cell infiltration in the tumour is associated with poor responses to immunotherapy. Given that (1) lymphocyte recruitment requires leucocyte-endothelial cell adhesion and (2) the vasculature controls tumour oxygenation and plays a pivotal role in T cell infiltration and activation, vessel targeting strategies including anti-angiogenesis and vascular normalisation in combination with immunotherapy are providing possible new strategies to enhance therapy. Here, we review the progress of vessel modulation in enhancing immunotherapy efficacy.

19.
J Biol Chem ; 284(49): 33966-81, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19837659

ABSTRACT

Both vascular endothelial growth factor receptors (VEGFR) and integrins are major regulators of VEGF-induced angiogenesis. Previous work has shown that beta3 integrin can regulate negatively VEGFR2 expression. Here we show that beta3 integrin can regulate negatively VEGF-mediated angiogenesis by limiting the interaction of the co-receptor NRP1 (neuropilin-1) with VEGFR2. In the presence of alphav beta3 integrin, NRP1 contributed minimally to VEGF-induced angiogenic processes in vivo, ex vivo, and in vitro. Conversely, when beta3 integrin expression is absent or low or its function is blocked with RGD-mimetic inhibitors, VEGF-mediated responses became NRP1-dependent. Indeed, combined inhibition of beta3 integrin and NRP1 decreased VEGF-mediated angiogenic responses further than individual inhibition of these receptors. We also show that alphav beta3 integrin can associate with NRP1 in a VEGF-dependent fashion. Our data suggest that beta3 integrin may, in part, negatively regulate VEGF signaling by sequestering NRP1 and preventing it from interacting with VEGFR2.


Subject(s)
Integrin alphaVbeta3/metabolism , Neovascularization, Pathologic , Neuropilin-1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Aorta/cytology , Base Sequence , Endothelial Cells/cytology , Humans , Mice , Microcirculation , Molecular Sequence Data , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism , Wound Healing
20.
EMBO Mol Med ; 12(2): e11663, 2020 02 07.
Article in English | MEDLINE | ID: mdl-31858727

ABSTRACT

Pathological angiogenesis contributes to tumour progression as well as to chronic inflammatory diseases. In this issue of EMBO Molecular Medicine, Esteban and co-workers identify endothelial cell MT1-MMP as a key regulator of intussusceptive angiogenesis (IA) in inflammatory colitis. Thrombospondin 1 (TSP1) cleavage by MT1-MMP results in the binding of the c-terminal fragment of TSP1 to αvß3 integrin, which induces nitric oxide (NO) production, vasodilation and further initiation of IA. This novel control mechanism of inflammatory IA points towards promising new therapeutic targets for inflammatory bowel disease.


Subject(s)
Matrix Metalloproteinase 14 , Metalloendopeptidases , Endothelial Cells , Humans , Matrix Metalloproteinases, Membrane-Associated , Neovascularization, Pathologic
SELECTION OF CITATIONS
SEARCH DETAIL