Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
Add more filters

Publication year range
1.
Development ; 147(20)2020 10 27.
Article in English | MEDLINE | ID: mdl-32994164

ABSTRACT

Between embryonic days 10.5 and 14.5, active proliferation drives rapid elongation of the murine midgut epithelial tube. Within this pseudostratified epithelium, nuclei synthesize DNA near the basal surface and move apically to divide. After mitosis, the majority of daughter cells extend a long, basally oriented filopodial protrusion, building a de novo path along which their nuclei can return to the basal side. WNT5A, which is secreted by surrounding mesenchymal cells, acts as a guidance cue to orchestrate this epithelial pathfinding behavior, but how this signal is received by epithelial cells is unknown. Here, we have investigated two known WNT5A receptors: ROR2 and RYK. We found that epithelial ROR2 is dispensable for midgut elongation. However, loss of Ryk phenocopies the Wnt5a-/- phenotype, perturbing post-mitotic pathfinding and leading to apoptosis. These studies reveal that the ligand-receptor pair WNT5A-RYK acts as a navigation system to instruct filopodial pathfinding, a process that is crucial for continuous cell cycling to fuel rapid midgut elongation.


Subject(s)
Digestive System/growth & development , Digestive System/metabolism , Pseudopodia/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Apoptosis , Cell Nucleus/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium/metabolism , Female , Male , Mesoderm/metabolism , Mice, Inbred C57BL , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism
2.
Dev Dyn ; 251(2): 336-349, 2022 02.
Article in English | MEDLINE | ID: mdl-34174014

ABSTRACT

BACKGROUND: Lymphatic vascular development is regulated by well-characterized signaling and transcriptional pathways. These pathways regulate lymphatic endothelial cell (LEC) migration, motility, polarity, and morphogenesis. Canonical and non-canonical WNT signaling pathways are known to control LEC polarity and development of lymphatic vessels and valves. PKD1, encoding Polycystin-1, is the most commonly mutated gene in polycystic kidney disease but has also been shown to be essential in lymphatic vascular morphogenesis. The mechanism by which Pkd1 acts during lymphangiogenesis remains unclear. RESULTS: Here we find that loss of non-canonical WNT signaling components Wnt5a and Ryk phenocopy lymphatic defects seen in Pkd1 knockout mice. To investigate genetic interaction, we generated Pkd1;Wnt5a double knockout mice. Loss of Wnt5a suppressed phenotypes seen in the lymphatic vasculature of Pkd1-/- mice and Pkd1 deletion suppressed phenotypes observed in Wnt5a-/- mice. Thus, we report mutually suppressive roles for Pkd1 and Wnt5a, with developing lymphatic networks restored to a more wild type state in double mutant mice. This genetic interaction between Pkd1 and the non-canonical WNT signaling pathway ultimately controls LEC polarity and the morphogenesis of developing vessel networks. CONCLUSION: Our work suggests that Pkd1 acts at least in part by regulating non-canonical WNT signaling during the formation of lymphatic vascular networks.


Subject(s)
Lymphatic Vessels , Polycystic Kidney Diseases , Animals , Lymphatic Vessels/metabolism , Mice , Mice, Knockout , Morphogenesis/genetics , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism , Protein Kinase C , Receptor Protein-Tyrosine Kinases/metabolism , Wnt Signaling Pathway/genetics , Wnt-5a Protein/genetics , Wnt-5a Protein/metabolism
3.
Growth Factors ; 40(3-4): 119-152, 2022 08.
Article in English | MEDLINE | ID: mdl-35861197

ABSTRACT

Organ-specific metastasis to secondary organs is dependent on the formation of a supportive pre-metastatic niche. This tissue-specific microenvironmental response is thought to be mediated by mutational and epigenetic changes to primary tumour cells resulting in altered cross-talk between cell types. This response is augmented through the release of tumour and stromal signalling mediators including cytokines, chemokines, exosomes and growth factors. Although researchers have elucidated some of the cancer-promoting features that are bespoke to organotropic metastasis to the lungs, it remains unclear if these are organ-specific or generic between organs. Understanding the mechanisms that mediate the metastasis-promoting synergy between the host microenvironment, immunity, and pulmonary structures may elucidate predictive, prognostic and therapeutic markers that could be targeted to reduce the metastatic burden of disease. Herein, we give an updated summary of the known cellular and molecular mechanisms that contribute to the formation of the lung pre-metastatic niche and tissue-specific metastasis.


Subject(s)
Exosomes , Neoplasms , Exosomes/metabolism , Exosomes/pathology , Humans , Lung , Neoplasm Metastasis/pathology , Neoplasms/metabolism , Signal Transduction , Tumor Microenvironment
4.
Development ; 144(3): 507-518, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28087639

ABSTRACT

Vascular endothelial growth factors (VEGFs) control angiogenesis and lymphangiogenesis during development and in pathological conditions. In the zebrafish trunk, Vegfa controls the formation of intersegmental arteries by primary angiogenesis and Vegfc is essential for secondary angiogenesis, giving rise to veins and lymphatics. Vegfd has been largely thought of as dispensable for vascular development in vertebrates. Here, we generated a zebrafish vegfd mutant by genome editing. vegfd mutants display significant defects in facial lymphangiogenesis independent of vegfc function. Strikingly, we find that vegfc and vegfd cooperatively control lymphangiogenesis throughout the embryo, including during the formation of the trunk lymphatic vasculature. Interestingly, we find that vegfd and vegfc also redundantly drive artery hyperbranching phenotypes observed upon depletion of Flt1 or Dll4. Epistasis and biochemical binding assays suggest that, during primary angiogenesis, Vegfd influences these phenotypes through Kdr (Vegfr2) rather than Flt4 (Vegfr3). These data demonstrate that, rather than being dispensable during development, Vegfd plays context-specific indispensable and also compensatory roles during both blood vessel angiogenesis and lymphangiogenesis.


Subject(s)
Lymphangiogenesis/physiology , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor D/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , Zebrafish/physiology , Animals , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Lymphangiogenesis/genetics , Membrane Proteins/genetics , Membrane Proteins/physiology , Models, Biological , Mutagenesis , Neovascularization, Physiologic/genetics , Sequence Deletion , Signal Transduction , Up-Regulation , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/physiology , Vascular Endothelial Growth Factor D/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/physiology , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/physiology , Zebrafish/genetics , Zebrafish Proteins/genetics
5.
Growth Factors ; 36(1-2): 15-40, 2018 04.
Article in English | MEDLINE | ID: mdl-29806777

ABSTRACT

The receptor tyrosine kinases (RTKs) are a well-characterized family of growth factor receptors that have central roles in human disease and are frequently therapeutically targeted. The RYK, ROR, PTK7 and MuSK subfamilies make up an understudied subset of WNT-binding RTKs. Numerous developmental, stem cell and pathological roles of WNTs, in particular WNT5A, involve signalling via these WNT receptors. The WNT-binding RTKs have highly context-dependent signalling outputs and stimulate the ß-catenin-dependent, planar cell polarity and/or WNT/Ca2+ pathways. RYK, ROR and PTK7 members have a pseudokinase domain in their intracellular regions. Alternative signalling mechanisms, including proteolytic cleavage and protein scaffolding functions, have been identified for these receptors. This review explores the structure, signalling, physiological and pathological roles of RYK, with particular attention paid to cancer and the possibility of therapeutically targeting RYK. The other WNT-binding RTKs are compared with RYK throughout to highlight the similarities and differences within this subset of WNT receptors.


Subject(s)
Receptor Protein-Tyrosine Kinases/metabolism , Wnt Signaling Pathway , Animals , Humans , Ligands , Neoplasms/metabolism , Protein Domains , Receptor Protein-Tyrosine Kinases/chemistry
6.
Growth Factors ; 36(1-2): 58-68, 2018 04.
Article in English | MEDLINE | ID: mdl-30035654

ABSTRACT

Ryk is a member of the receptor tyrosine kinase (RTK) family of proteins that control and regulate cellular processes. It is distinguished by binding Wnt ligands and having no detectable intrinsic protein tyrosine kinase activity suggesting Ryk is a pseudokinase. Here, we show an essential role for Ryk in directing morphogenetic events required for normal cardiac development through the examination of Ryk-deficient mice. We employed vascular corrosion casting, vascular perfusion with contrast dye, and immunohistochemistry to characterize cardiovascular and pharyngeal defects in Ryk-/- embryos. Ryk-/- mice exhibit a variety of malformations of the heart and outflow tract that resemble human congenital heart defects. This included stenosis and interruption of the aortic arch, ventriculoarterial malalignment, ventricular septal defects and abnormal pharyngeal arch artery remodelling. This study therefore defines a key intersection between a subset of growth factor receptors involved in planar cell polarity signalling, the Wnt family and mammalian cardiovascular development.


Subject(s)
Heart Defects, Congenital/etiology , Pharynx/abnormalities , Receptor Protein-Tyrosine Kinases/physiology , Wnt Proteins/metabolism , Animals , Aorta, Thoracic/abnormalities , Female , Mice , Morphogenesis , Pregnancy
7.
J Biol Chem ; 291(53): 27265-27278, 2016 12 30.
Article in English | MEDLINE | ID: mdl-27852824

ABSTRACT

VEGF-C and VEGF-D are secreted glycoproteins that induce angiogenesis and lymphangiogenesis in cancer, thereby promoting tumor growth and spread. They exhibit structural homology and activate VEGFR-2 and VEGFR-3, receptors on endothelial cells that signal for growth of blood vessels and lymphatics. VEGF-C and VEGF-D were thought to exhibit similar bioactivities, yet recent studies indicated distinct signaling mechanisms (e.g. tumor-derived VEGF-C promoted expression of the prostaglandin biosynthetic enzyme COX-2 in lymphatics, a response thought to facilitate metastasis via the lymphatic vasculature, whereas VEGF-D did not). Here we explore the basis of the distinct bioactivities of VEGF-D using a neutralizing antibody, peptide mapping, and mutagenesis to demonstrate that the N-terminal α-helix of mature VEGF-D (Phe93-Arg108) is critical for binding VEGFR-2 and VEGFR-3. Importantly, the N-terminal part of this α-helix, from Phe93 to Thr98, is required for binding VEGFR-3 but not VEGFR-2. Surprisingly, the corresponding part of the α-helix in mature VEGF-C did not influence binding to either VEGFR-2 or VEGFR-3, indicating distinct determinants of receptor binding by these growth factors. A variant of mature VEGF-D harboring a mutation in the N-terminal α-helix, D103A, exhibited enhanced potency for activating VEGFR-3, was able to promote increased COX-2 mRNA levels in lymphatic endothelial cells, and had enhanced capacity to induce lymphatic sprouting in vivo This mutant may be useful for developing protein-based therapeutics to drive lymphangiogenesis in clinical settings, such as lymphedema. Our studies shed light on the VEGF-D structure/function relationship and provide a basis for understanding functional differences compared with VEGF-C.


Subject(s)
Endothelium, Vascular/pathology , Lymphangiogenesis , Lymphatic Vessels/pathology , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor D/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Animals , Antibodies, Neutralizing , Cells, Cultured , Dermis/metabolism , Dermis/pathology , Endothelium, Vascular/metabolism , Female , Humans , Lymphatic Vessels/metabolism , Mice, Inbred NOD , Mice, SCID , Mutagenesis, Site-Directed , Mutation/genetics , Neovascularization, Pathologic/metabolism , Signal Transduction , Vascular Endothelial Growth Factor C/chemistry , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor D/chemistry , Vascular Endothelial Growth Factor D/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics
8.
Growth Factors ; 35(2-3): 61-75, 2017 06.
Article in English | MEDLINE | ID: mdl-28697634

ABSTRACT

Remodelling of lymphatic vessels in tumours facilitates metastasis to lymph nodes. The growth factors VEGF-C and VEGF-D are well known inducers of lymphatic remodelling and metastasis in cancer. They are initially produced as full-length proteins requiring proteolytic processing in order to bind VEGF receptors with high affinity and thereby promote lymphatic remodelling. The fibrinolytic protease plasmin promotes processing of VEGF-C and VEGF-D in vitro, but its role in processing them in cancer was unknown. Here we explore plasmin's role in proteolytically activating VEGF-D in vivo, and promoting lymphatic remodelling and metastasis in cancer, by co-expressing the plasmin inhibitor α2-antiplasmin with VEGF-D in a mouse tumour model. We show that α2-antiplasmin restricts activation of VEGF-D, enlargement of intra-tumoural lymphatics and occurrence of lymph node metastasis. Our findings indicate that the fibrinolytic system influences lymphatic remodelling in tumours which is consistent with previous clinicopathological observations correlating fibrinolytic components with cancer metastasis.


Subject(s)
Antifibrinolytic Agents/therapeutic use , Neoplasms, Experimental/drug therapy , alpha-2-Antiplasmin/therapeutic use , Animals , Antifibrinolytic Agents/pharmacology , Cell Line , Cell Line, Tumor , Female , Humans , Lymph Nodes/drug effects , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphatic Metastasis , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Experimental/pathology , Receptors, Vascular Endothelial Growth Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism , alpha-2-Antiplasmin/pharmacology
9.
Development ; 141(6): 1239-49, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24523457

ABSTRACT

The VEGFC/VEGFR3 signaling pathway is essential for lymphangiogenesis (the formation of lymphatic vessels from pre-existing vasculature) during embryonic development, tissue regeneration and tumor progression. The recently identified secreted protein CCBE1 is indispensible for lymphangiogenesis during development. The role of CCBE1 orthologs is highly conserved in zebrafish, mice and humans with mutations in CCBE1 causing generalized lymphatic dysplasia and lymphedema (Hennekam syndrome). To date, the mechanism by which CCBE1 acts remains unknown. Here, we find that ccbe1 genetically interacts with both vegfc and vegfr3 in zebrafish. In the embryo, phenotypes driven by increased Vegfc are suppressed in the absence of Ccbe1, and Vegfc-driven sprouting is enhanced by local Ccbe1 overexpression. Moreover, Vegfc- and Vegfr3-dependent Erk signaling is impaired in the absence of Ccbe1. Finally, CCBE1 is capable of upregulating the levels of fully processed, mature VEGFC in vitro and the overexpression of mature VEGFC rescues ccbe1 loss-of-function phenotypes in zebrafish. Taken together, these data identify Ccbe1 as a crucial component of the Vegfc/Vegfr3 pathway in the embryo.


Subject(s)
Lymphangiogenesis/physiology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , DNA/genetics , Gene Expression Regulation, Developmental , Humans , Lymphangiogenesis/genetics , MAP Kinase Signaling System , Mice , Molecular Sequence Data , Point Mutation , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Signal Transduction , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
10.
PLoS Biol ; 12(6): e1001895, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24960609

ABSTRACT

The Wnt receptor Ryk is an evolutionary-conserved protein important during neuronal differentiation through several mechanisms, including γ-secretase cleavage and nuclear translocation of its intracellular domain (Ryk-ICD). Although the Wnt pathway may be neuroprotective, the role of Ryk in neurodegenerative disease remains unknown. We found that Ryk is up-regulated in neurons expressing mutant huntingtin (HTT) in several models of Huntington's disease (HD). Further investigation in Caenorhabditis elegans and mouse striatal cell models of HD provided a model in which the early-stage increase of Ryk promotes neuronal dysfunction by repressing the neuroprotective activity of the longevity-promoting factor FOXO through a noncanonical mechanism that implicates the Ryk-ICD fragment and its binding to the FOXO co-factor ß-catenin. The Ryk-ICD fragment suppressed neuroprotection by lin-18/Ryk loss-of-function in expanded-polyQ nematodes, repressed FOXO transcriptional activity, and abolished ß-catenin protection of mutant htt striatal cells against cell death vulnerability. Additionally, Ryk-ICD was increased in the nucleus of mutant htt cells, and reducing γ-secretase PS1 levels compensated for the cytotoxicity of full-length Ryk in these cells. These findings reveal that the Ryk-ICD pathway may impair FOXO protective activity in mutant polyglutamine neurons, suggesting that neurons are unable to efficiently maintain function and resist disease from the earliest phases of the pathogenic process in HD.


Subject(s)
Forkhead Transcription Factors/metabolism , Huntington Disease/etiology , Neurons/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Wnt/metabolism , Aged , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Line , Female , Humans , Huntington Disease/metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Oligonucleotide Array Sequence Analysis , Presenilin-1/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Wnt Signaling Pathway
11.
J Pathol ; 239(2): 152-61, 2016 06.
Article in English | MEDLINE | ID: mdl-26924464

ABSTRACT

Leakage of fluid from blood vessels, leading to oedema, is a key feature of many diseases including hyperoxic acute lung injury (HALI), which can occur when patients are ventilated with high concentrations of oxygen (hyperoxia). The molecular mechanisms driving vascular leak and oedema in HALI are poorly understood. VEGF-D is a protein that promotes blood vessel leak and oedema when overexpressed in tissues, but the role of endogenous VEGF-D in pathological oedema was unknown. To address these issues, we exposed Vegfd-deficient mice to hyperoxia. The resulting pulmonary oedema in Vegfd-deficient mice was substantially reduced compared to wild-type, as was the protein content of bronchoalveolar lavage fluid, consistent with reduced vascular leak. Vegf-d and its receptor Vegfr-3 were more highly expressed in lungs of hyperoxic, versus normoxic, wild-type mice, indicating that components of the Vegf-d signalling pathway are up-regulated in hyperoxia. Importantly, VEGF-D and its receptors were co-localized on blood vessels in clinical samples of human lungs exposed to hyperoxia; hence, VEGF-D may act directly on blood vessels to promote fluid leak. Our studies show that Vegf-d promotes oedema in response to hyperoxia in mice and support the hypothesis that VEGF-D signalling promotes vascular leak in human HALI. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Acute Lung Injury/complications , Hyperoxia/complications , Pulmonary Edema/etiology , Signal Transduction , Vascular Endothelial Growth Factor D/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Bronchoalveolar Lavage Fluid , Cell Line, Tumor , Female , Humans , Hyperoxia/metabolism , Hyperoxia/pathology , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Oxygen/metabolism , Pulmonary Edema/complications , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , Vascular Endothelial Growth Factor D/administration & dosage , Vascular Endothelial Growth Factor D/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics , Xenograft Model Antitumor Assays
12.
Hum Mol Genet ; 23(5): 1286-97, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24163130

ABSTRACT

Mutations in SOX18, VEGFC and Vascular Endothelial Growth Factor 3 underlie the hereditary lymphatic disorders hypotrichosis-lymphedema-telangiectasia (HLT), Milroy-like lymphedema and Milroy disease, respectively. Genes responsible for hereditary lymphedema are key regulators of lymphatic vascular development in the embryo. To identify novel modulators of lymphangiogenesis, we used a mouse model of HLT (Ragged Opossum) and performed gene expression profiling of aberrant dermal lymphatic vessels. Expression studies and functional analysis in zebrafish and mice revealed one candidate, ArfGAP with RhoGAP domain, Ankyrin repeat and PH domain 3 (ARAP3), which is down-regulated in HLT mouse lymphatic vessels and necessary for lymphatic vascular development in mice and zebrafish. We position this known regulator of cell behaviour during migration as a mediator of the cellular response to Vegfc signalling in lymphatic endothelial cells in vitro and in vivo. Our data refine common mechanisms that are likely to contribute during both development and the pathogenesis of lymphatic vascular disorders.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , GTPase-Activating Proteins/genetics , Gene Expression Regulation , Hypotrichosis/genetics , Lymphangiogenesis/genetics , Lymphedema/genetics , Telangiectasis/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Movement/genetics , Disease Models, Animal , Endothelial Cells/metabolism , Female , GTPase-Activating Proteins/metabolism , Lymphatic Vessels/metabolism , Mice , Mice, Knockout , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Syndrome , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism , Zebrafish
13.
Blood ; 123(7): 1102-12, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24269955

ABSTRACT

Vascular endothelial growth factor-D (VEGFD) is a potent pro-lymphangiogenic molecule during tumor growth and is considered a key therapeutic target to modulate metastasis. Despite roles in pathological neo-lymphangiogenesis, the characterization of an endogenous role for VEGFD in vascular development has remained elusive. Here, we used zebrafish to assay for genetic interactions between the Vegf/Vegf-receptor pathway and SoxF transcription factors and identified a specific interaction between Vegfd and Sox18. Double knockdown zebrafish embryos for Sox18/Vegfd and Sox7/Vegfd exhibit defects in arteriovenous differentiation. Supporting this observation, we found that Sox18/Vegfd double but not single knockout mice displayed dramatic vascular development defects. We find that VEGFD-mitogen-activated protein kinase kinase-extracellular signal-regulated kinase signaling modulates SOX18-mediated transcription, functioning at least in part by enhancing nuclear concentration and transcriptional activity in vascular endothelial cells. This work suggests that VEGFD-mediated pathologies include or involve an underlying dysregulation of SOXF-mediated transcriptional networks.


Subject(s)
Blood Vessels/embryology , Neovascularization, Physiologic/genetics , SOXF Transcription Factors/metabolism , Vascular Endothelial Growth Factor D/physiology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Animals, Genetically Modified , Embryo, Mammalian , Embryo, Nonmammalian , Female , Gene Expression Regulation, Developmental , Gene Regulatory Networks/genetics , Male , Mice , Mice, Inbred C57BL , SOXF Transcription Factors/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
15.
Mol Ther ; 22(1): 18-27, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24048441

ABSTRACT

The tumor microenvironment can promote tumor growth and reduce treatment efficacy. Tumors can occur in many sites in the body, but how surrounding normal tissues at different anatomical sites affect tumor microenvironments and their subsequent response to therapy is not known.We demonstrated that tumors from renal, colon, or prostate cell lines in orthotopic locations responded to immunotherapy consisting of three agonist antibodies, termed Tri-mAb, to a much lesser extent than the same tumor type located subcutaneously. A tissue-specific response to Tri-mAb was confirmed by ex vivo separation of subcutaneous (SC) or orthotopic tumor cells from stromal cells, followed by reinjection of tumor cells into the opposite site. Compared with SC tumors, orthotopic tumors had a microenvironment associated with a type 2 immune response, related to immunosuppression, and an involvement of alternatively activated macrophages in the kidney model. Orthotopic kidney tumors were more highly vascularized than SC tumors. Neutralizing the macrophage- and Th2-associated molecules chemokine (C-C motif) ligand 2 or interleukin-13 led to a significantly improved therapeutic effect. This study highlights the importance of the tissue of implantation in sculpting the tumor microenvironment. These are important fundamental issues in tumor biology and crucial factors to consider in the design of experimental models and treatment strategies.


Subject(s)
Immunotherapy , Neoplasms/immunology , Neoplasms/pathology , Tumor Microenvironment/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , CD40 Antigens/antagonists & inhibitors , CD40 Antigens/immunology , Cell Line, Tumor , Chemokine CCL2/immunology , Colonic Neoplasms/immunology , Disease Models, Animal , Gene Expression , Interleukin-13/immunology , Kidney Neoplasms/immunology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Neoplasms/mortality , Neoplasms/therapy , Neovascularization, Pathologic/immunology , Organ Specificity/immunology , Prostate/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Treatment Outcome , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
16.
J Biol Chem ; 288(12): 8176-8186, 2013 Mar 22.
Article in English | MEDLINE | ID: mdl-23404505

ABSTRACT

VEGF-D is an angiogenic and lymphangiogenic glycoprotein that can be proteolytically processed generating various forms differing in subunit composition due to the presence or absence of N- and C-terminal propeptides. These propeptides flank the central VEGF homology domain, that contains the binding sites for VEGF receptors (VEGFRs), but their biological functions were unclear. Characterization of propeptide function will be important to clarify which forms of VEGF-D are biologically active and therefore clinically relevant. Here we use VEGF-D mutants deficient in either propeptide, and in the capacity to process the remaining propeptide, to monitor the functions of these domains. We report for the first time that VEGF-D binds heparin, and that the C-terminal propeptide significantly enhances this interaction (removal of this propeptide from full-length VEGF-D completely prevents heparin binding). We also show that removal of either the N- or C-terminal propeptide is required for VEGF-D to drive formation of VEGFR-2/VEGFR-3 heterodimers which have recently been shown to positively regulate angiogenic sprouting. The mature form of VEGF-D, lacking both propeptides, can also promote formation of these receptor heterodimers. In a mouse tumor model, removal of only the C-terminal propeptide from full-length VEGF-D was sufficient to enhance angiogenesis and tumor growth. In contrast, removal of both propeptides is required for high rates of lymph node metastasis. The findings reported here show that the propeptides profoundly influence molecular interactions of VEGF-D with VEGF receptors, co-receptors, and heparin, and its effects on tumor biology.


Subject(s)
Heparin/chemistry , Vascular Endothelial Growth Factor D/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Animals , Cell Line , Chromatography, Affinity , Endothelial Cells/metabolism , Female , Humans , Lymphangiogenesis , Lymphatic Metastasis , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/metabolism , Neuropilins/metabolism , Protein Binding , Protein Multimerization , Protein Precursors/chemistry , Protein Precursors/genetics , Protein Precursors/metabolism , Protein Precursors/physiology , Protein Structure, Tertiary , Sequence Deletion , Vascular Endothelial Growth Factor D/chemistry , Vascular Endothelial Growth Factor D/genetics , Vascular Endothelial Growth Factor D/metabolism , Vascular Endothelial Growth Factor Receptor-2/chemistry , Vascular Endothelial Growth Factor Receptor-3/chemistry
17.
Growth Factors ; 32(1): 11-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24471468

ABSTRACT

The non-canonical Wnt receptor, Ryk, promotes chemorepulsive axon guidance in the developing mouse brain and spinal cord in response to Wnt5a. Ryk has also been identified as a major suppressor of axonal regrowth after spinal cord injury. Thus, a comprehensive understanding of how growing axons and dendrites respond to Wnt5a-mediated Ryk activation is required if we are to overcome this detrimental activity. Here we undertook a detailed analysis of the effect of Wnt5a/Ryk interactions on axonal and dendritic growth in dissociated embryonic mouse cortical neuron cultures, focusing on callosal neurons known to be responsive to Ryk-induced chemorepulsion. We show that Ryk inhibits axonal growth in response to Wnt5a. We also show that Wnt5a inhibits dendrite growth independently of Ryk. However, this inhibition is relieved when Ryk is present. Therefore, Wnt5a-mediated Ryk activation triggers divergent responses in callosal axons and dendrites in the in vitro context.


Subject(s)
Axons/metabolism , Dendrites/metabolism , Neurogenesis/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Wnt Proteins/metabolism , Animals , Brain/embryology , Brain/growth & development , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/genetics , Receptor Protein-Tyrosine Kinases/genetics , Spinal Cord/embryology , Spinal Cord/growth & development , Spinal Cord Injuries/metabolism , Wnt Proteins/genetics , Wnt Proteins/pharmacology , Wnt Signaling Pathway , Wnt-5a Protein
18.
Biochem Soc Trans ; 42(6): 1569-75, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25399572

ABSTRACT

A chronic hyperactivated angiogenic state in cancer plays an important role in tumour growth and metastasis and has been identified as one of the hallmarks of cancer. Inhibition of this process has been associated with tumour suppression in many pre-clinical contexts using different animal tumour models. Anti-angiogenic therapeutics were subsequently developed and used to treat several prevalent types of human cancer. However, recent clinical experience has revealed limitations of this approach in treating cancer as patient response varies over a wide range. Given that there are complex underlying molecular and cellular changes provoked by anti-angiogenic treatment within the tumour microenvironment (TME), it is not surprising that modest effectiveness and resistance have been observed in the clinical setting. This article discusses these issues in the context of VEGF-A-targeted anti-angiogenic treatment of cancer and provides insight into the importance of tumour endothelium for understanding the tumour response to anti-angiogenic therapy. Special consideration is also given to possible approaches for investigating how endothelium contributes to the tumour response to anti-angiogenic agents and for exploring the therapeutic and biomarker potential of targeting tumour endothelium.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Endothelium, Vascular/physiology , Angiogenesis Inhibitors/pharmacology , Endothelium, Vascular/drug effects , Humans , Tumor Microenvironment
19.
Nature ; 456(7222): 643-7, 2008 Dec 04.
Article in English | MEDLINE | ID: mdl-18931657

ABSTRACT

The lymphatic system plays a key role in tissue fluid regulation and tumour metastasis, and lymphatic defects underlie many pathological states including lymphoedema, lymphangiectasia, lymphangioma and lymphatic dysplasia. However, the origins of the lymphatic system in the embryo, and the mechanisms that direct growth of the network of lymphatic vessels, remain unclear. Lymphatic vessels are thought to arise from endothelial precursor cells budding from the cardinal vein under the influence of the lymphatic hallmark gene Prox1 (prospero homeobox 1; ref. 4). Defects in the transcription factor gene SOX18 (SRY (sex determining region Y) box 18) cause lymphatic dysfunction in the human syndrome hypotrichosis-lymphoedema-telangiectasia, suggesting that Sox18 may also play a role in lymphatic development or function. Here we use molecular, cellular and genetic assays in mice to show that Sox18 acts as a molecular switch to induce differentiation of lymphatic endothelial cells. Sox18 is expressed in a subset of cardinal vein cells that later co-express Prox1 and migrate to form lymphatic vessels. Sox18 directly activates Prox1 transcription by binding to its proximal promoter. Overexpression of Sox18 in blood vascular endothelial cells induces them to express Prox1 and other lymphatic endothelial markers, while Sox18-null embryos show a complete blockade of lymphatic endothelial cell differentiation from the cardinal vein. Our findings demonstrate a critical role for Sox18 in developmental lymphangiogenesis, and suggest new avenues to investigate for therapeutic management of human lymphangiopathies.


Subject(s)
Cell Differentiation , Lymphatic Vessels/cytology , Lymphatic Vessels/embryology , SOXF Transcription Factors/metabolism , Animals , Biomarkers/analysis , Cell Movement , Cells, Cultured , Edema/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Ephrin-B2/genetics , Female , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Hypotrichosis/genetics , Lymphangiogenesis , Lymphatic Vessels/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Promoter Regions, Genetic/genetics , SOXF Transcription Factors/deficiency , SOXF Transcription Factors/genetics , Telangiectasis/genetics , Tumor Suppressor Proteins/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics , Veins/cytology
20.
J Biol Chem ; 287(53): 44518-25, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23144463

ABSTRACT

The Wnt signaling pathways control many critical developmental and adult physiological processes. In vertebrates, one fundamentally important function of Wnts is to provide directional information by regulating the evolutionarily conserved planar cell polarity (PCP) pathway during embryonic morphogenesis. However, despite the critical roles of Wnts and PCP in vertebrate development and disease, little is known about the molecular mechanisms underlying Wnt regulation of PCP. Here, we have found that the receptor-like tyrosine kinase (Ryk), a Wnt5a-binding protein required in axon guidance, regulates PCP signaling. We show that Ryk interacts with Vangl2 genetically and biochemically, and such interaction is potentiated by Wnt5a. Loss of Ryk in a Vangl2(+/-) background results in classic PCP defects, including open neural tube, misalignment of sensory hair cells in the inner ear, and shortened long bones in the limbs. Complete loss of both Ryk and Vangl2 results in more severe phenotypes that resemble the Wnt5a(-/-) mutant in many aspects such as shortened anterior-posterior body axis, limb, and frontonasal process. Our data identify the Wnt5a-binding protein Ryk as a general regulator of the mammalian Wnt/PCP signaling pathway. We show that Ryk transduces Wnt5a signaling by forming a complex with Vangl2 and that Ryk regulates PCP by at least in part promoting Vangl2 stability. As human mutations in WNT5A and VANGL2 are found to cause Robinow syndrome and neural tube defects, respectively, our results further suggest that human mutations in RYK may also be involved in these diseases.


Subject(s)
Cell Polarity , Embryonic Development , Nerve Tissue Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Wnt Proteins/metabolism , Animals , Body Patterning , Bone and Bones/embryology , Bone and Bones/metabolism , Ear, Inner/embryology , Ear, Inner/metabolism , HEK293 Cells , Hair Cells, Auditory, Inner/metabolism , Humans , Mice , Mice, Knockout , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neural Tube/embryology , Neural Tube/metabolism , Protein Binding , Protein Stability , Proteolysis , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction , Wnt Proteins/genetics , Wnt-5a Protein
SELECTION OF CITATIONS
SEARCH DETAIL