Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Angiogenesis ; 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38955953

ABSTRACT

The proliferation of the endothelium is a highly coordinated process to ensure the emergence, expansion, and homeostasis of the vasculature. While Bone Morphogenetic Protein (BMP) signaling fine-tunes the behaviors of endothelium in health and disease, how BMP signaling influences the proliferation of endothelium and therefore, modulates angiogenesis remains largely unknown. Here, we evaluated the role of Activin A Type I Receptor (ACVR1/ALK2), a key BMP receptor in the endothelium, in modulating the proliferation of endothelial cells. We show that ACVR1/ALK2 is a key modulator for the proliferation of endothelium in the retinal vessels. Loss of endothelial ALK2 leads to a significant reduction in endothelial proliferation and results in fewer branches/endothelial cells in the retinal vessels. Interestingly, venous endothelium appears to be more susceptible to ALK2 deletion. Mechanistically, ACVR1/ALK2 inhibits the expression of CDKN1A/p21, a critical negative regulator of cell cycle progression, in a SMAD1/5-dependent manner, thereby enabling the venous endothelium to undergo active proliferation by suppressing CDKN1A/p21. Taken together, our findings show that BMP signaling mediated by ACVR1/ALK2 provides a critical yet previously underappreciated input to modulate the proliferation of venous endothelium, thereby fine-tuning the context of angiogenesis in health and disease.

2.
J Exp Med ; 221(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38442272

ABSTRACT

Meningeal lymphatic vessels (MLVs) promote tissue clearance and immune surveillance in the central nervous system (CNS). Vascular endothelial growth factor-C (VEGF-C) regulates MLV development and maintenance and has therapeutic potential for treating neurological disorders. Herein, we investigated the effects of VEGF-C overexpression on brain fluid drainage and ischemic stroke outcomes in mice. Intracerebrospinal administration of an adeno-associated virus expressing mouse full-length VEGF-C (AAV-mVEGF-C) increased CSF drainage to the deep cervical lymph nodes (dCLNs) by enhancing lymphatic growth and upregulated neuroprotective signaling pathways identified by single nuclei RNA sequencing of brain cells. In a mouse model of ischemic stroke, AAV-mVEGF-C pretreatment reduced stroke injury and ameliorated motor performances in the subacute stage, associated with mitigated microglia-mediated inflammation and increased BDNF signaling in brain cells. Neuroprotective effects of VEGF-C were lost upon cauterization of the dCLN afferent lymphatics and not mimicked by acute post-stroke VEGF-C injection. We conclude that VEGF-C prophylaxis promotes multiple vascular, immune, and neural responses that culminate in a protection against neurological damage in acute ischemic stroke.


Subject(s)
Ischemic Stroke , Stroke , Animals , Mice , Vascular Endothelial Growth Factor C , Neuroinflammatory Diseases , Drainage
3.
bioRxiv ; 2023 Jan 21.
Article in English | MEDLINE | ID: mdl-36711611

ABSTRACT

Background: The blood brain barrier (BBB) preserves neuronal function in the central nervous system (CNS) by tightly controlling metabolite exchanges with the blood. In the eye, the retina is likewise protected by the blood-retina barrier (BRB) to maintain phototransduction. We showed that the secreted guidance cue Netrin-1 regulated BBB integrity, by binding to endothelial Unc5B and regulating canonical ß-catenin dependent expression of BBB gene expression. Objective: Here, we investigated if Netrin-1-binding to endothelial Unc5B also controlled BRB integrity, and if this process involved Norrin/ß-catenin signaling, which is the major known driver of BRB development and maintenance. Methods: We analyzed Tamoxifen-inducible loss- and gain- of-function alleles of Unc5B, Ntn1 and Ctnnb1 in conjunction with tracer injections and biochemical signaling studies. Results: Inducible endothelial Unc5B deletion, and inducible global Ntn1 deletion in postnatal mice reduced phosphorylation of the Norrin receptor LRP5, leading to reduced ß-catenin and LEF1 expression, conversion of retina endothelial cells from a barrier-competent Claudin-5+/PLVAP- state to a Claudin-5-/PLVAP+ leaky phenotype, and extravasation of injected low molecular weight tracers. Inducible Ctnnb1 gain of function rescued vascular leak in Unc5B mutants, and Ntn1 overexpression induced BRB tightening. Unc5B expression in pericytes contributed to BRB permeability, via regulation of endothelial Unc5B. Mechanistically, Netrin-1-Unc5B signaling promoted ß-catenin dependent BRB signaling by enhancing phosphorylation of the Norrin receptor LRP5 via the Discs large homologue 1 (Dlg1) intracellular scaffolding protein. Conclusions: The data identify Netrin1-Unc5B as novel regulators of BRB integrity, with implications for diseases associated with BRB disruption.

4.
Nat Commun ; 13(1): 1169, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35246514

ABSTRACT

Blood-brain barrier (BBB) integrity is critical for proper function of the central nervous system (CNS). Here, we show that the endothelial Unc5B receptor controls BBB integrity by maintaining Wnt/ß-catenin signaling. Inducible endothelial-specific deletion of Unc5B in adult mice leads to BBB leak from brain capillaries that convert to a barrier-incompetent state with reduced Claudin-5 and increased PLVAP expression. Loss of Unc5B decreases BBB Wnt/ß-catenin signaling, and ß-catenin overexpression rescues Unc5B mutant BBB defects. Mechanistically, the Unc5B ligand Netrin-1 enhances Unc5B interaction with the Wnt co-receptor LRP6, induces its phosphorylation and activates Wnt/ß-catenin downstream signaling. Intravenous delivery of antibodies blocking Netrin-1 binding to Unc5B causes a transient BBB breakdown and disruption of Wnt signaling, followed by neurovascular barrier resealing. These data identify Netrin-1-Unc5B signaling as a ligand-receptor pathway that regulates BBB integrity, with implications for CNS diseases.


Subject(s)
Blood-Brain Barrier , Netrin Receptors , Animals , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Ligands , Mice , Netrin Receptors/genetics , Netrin Receptors/metabolism , Netrin-1/genetics , Netrin-1/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
5.
Commun Biol ; 3(1): 603, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33097786

ABSTRACT

Protein tyrosine phosphatases are essential modulators of angiogenesis and have been identified as novel therapeutic targets in cancer and anti-angiogenesis. The roles of atypical Phosphatase of Regenerative Liver (PRL) phosphatases in this context remain poorly understood. Here, we investigate the biological function of PRL phosphatases in developmental angiogenesis in the postnatal mouse retina and in cell culture. We show that endothelial cells in the retina express PRL-2 encoded by the Ptp4a2 gene, and that inducible endothelial and global Ptp4a2 mutant mice exhibit defective retinal vascular outgrowth, arteriovenous differentiation, and sprouting angiogenesis. Mechanistically, PTP4A2 deletion limits angiogenesis by inhibiting endothelial cell migration and the VEGF-A, DLL-4/NOTCH-1 signaling pathway. This study reveals the importance of PRL-2 as a modulator of vascular development.


Subject(s)
Immediate-Early Proteins , Neovascularization, Physiologic/genetics , Protein Tyrosine Phosphatases , Signal Transduction/genetics , Animals , Cell Movement/genetics , Cells, Cultured , Endothelial Cells/cytology , Female , Human Umbilical Vein Endothelial Cells , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Immediate-Early Proteins/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Protein Tyrosine Phosphatases/physiology , Retina/cytology , Retina/metabolism , Vascular Malformations/genetics , Vascular Malformations/pathology
6.
Sci Rep ; 10(1): 7557, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32372058

ABSTRACT

Generation of bispecific antibodies (BsAbs) having two unique Fab domains requires heterodimerization of the two heavy chains and pairing of each heavy chain with its cognate light chain. An alternative bispecific scaffold (Bipod) comprising an scFv and a Fab on a heterodimeric Fc eliminates the possibility of light chain mispairing. However, unpredictable levels of chain expression and scFv-induced aggregation can complicate purification and reduce the yield of desired Bipod. Here, we describe a high-throughput method for generation of Bipods based on protein A and CH1 domain affinity capture. This method exploits over-expression of the scFv chain to maximize heterodimer yield. Bipods purified by this method have purity suitable for cell-based functional assays and in vivo studies.


Subject(s)
Antibodies, Bispecific/chemistry , Immunoglobulin Fab Fragments/chemistry , Protein Engineering/methods , Single-Chain Antibodies/chemistry , Animals , Biological Products/therapeutic use , CHO Cells , Cricetulus , DNA/chemistry , Dimerization , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Epitopes/chemistry , Humans , Immunoglobulin G/genetics , Immunosuppressive Agents/therapeutic use , Mutation , Neoplasms/therapy , Plasmids , Protein Domains
7.
J Biol Chem ; 294(50): 19034-19047, 2019 12 13.
Article in English | MEDLINE | ID: mdl-31676684

ABSTRACT

Acyl-CoA thioesterases (Acots) hydrolyze fatty acyl-CoA esters. Acots in the mitochondrial matrix are poised to mitigate ß-oxidation overload and maintain CoA availability. Several Acots associate with mitochondria, but whether they all localize to the matrix, are redundant, or have different roles is unresolved. Here, we compared the suborganellar localization, activity, expression, and regulation among mitochondrial Acots (Acot2, -7, -9, and -13) in mitochondria from multiple mouse tissues and from a model of Acot2 depletion. Acot7, -9, and -13 localized to the matrix, joining Acot2 that was previously shown to localize there. Mitochondria from heart, skeletal muscle, brown adipose tissue, and kidney robustly expressed Acot2, -9, and -13; Acot9 levels were substantially higher in brown adipose tissue and kidney mitochondria, as was activity for C4:0-CoA, a unique Acot9 substrate. In all tissues, Acot2 accounted for about half of the thioesterase activity for C14:0-CoA and C16:0-CoA. In contrast, liver mitochondria from fed and fasted mice expressed little Acot activity, which was confined to long-chain CoAs and due mainly to Acot7 and Acot13 activities. Matrix Acots occupied different functional niches, based on substrate specificity (Acot9 versus Acot2 and -13) and strong CoA inhibition (Acot7, -9, and -13, but not Acot2). Interpreted in the context of ß-oxidation, CoA inhibition would prevent Acot-mediated suppression of ß-oxidation, while providing a release valve when CoA is limiting. In contrast, CoA-insensitive Acot2 could provide a constitutive siphon for long-chain fatty acyl-CoAs. These results reveal how the family of matrix Acots can mitigate ß-oxidation overload and prevent CoA limitation.


Subject(s)
Acyl Coenzyme A/metabolism , Mitochondria/enzymology , Palmitoyl-CoA Hydrolase/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Palmitoyl-CoA Hydrolase/deficiency , Palmitoyl-CoA Hydrolase/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Thiolester Hydrolases/metabolism
8.
Nat Commun ; 10(1): 2350, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31138815

ABSTRACT

Endothelial cell migration, proliferation and survival are triggered by VEGF-A activation of VEGFR2. However, how these cell behaviors are regulated individually is still unknown. Here we identify Endophilin-A2 (ENDOA2), a BAR-domain protein that orchestrates CLATHRIN-independent internalization, as a critical mediator of endothelial cell migration and sprouting angiogenesis. We show that EndoA2 knockout mice exhibit postnatal angiogenesis defects and impaired front-rear polarization of sprouting tip cells. ENDOA2 deficiency reduces VEGFR2 internalization and inhibits downstream activation of the signaling effector PAK but not ERK, thereby affecting front-rear polarity and migration but not proliferation or survival. Mechanistically, VEGFR2 is directed towards ENDOA2-mediated endocytosis by the SLIT2-ROBO pathway via SLIT-ROBO-GAP1 bridging of ENDOA2 and ROBO1. Blocking ENDOA2-mediated endothelial cell migration attenuates pathological angiogenesis in oxygen-induced retinopathy models. This work identifies a specific endocytic pathway controlling a subset of VEGFR2 mediated responses that could be targeted to prevent excessive sprouting angiogenesis in pathological conditions.


Subject(s)
Acyltransferases/genetics , Endothelial Cells/metabolism , Neovascularization, Physiologic/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cell Movement/genetics , Cell Polarity/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Endocytosis/genetics , Endothelial Cells/cytology , Intercellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/metabolism , Retinal Vessels/cytology , Retinal Vessels/growth & development , p21-Activated Kinases/metabolism , Roundabout Proteins
9.
Science ; 361(6402): 599-603, 2018 08 10.
Article in English | MEDLINE | ID: mdl-30093598

ABSTRACT

Excess dietary lipid uptake causes obesity, a major global health problem. Enterocyte-absorbed lipids are packaged into chylomicrons, which enter the bloodstream through intestinal lymphatic vessels called lacteals. Here, we show that preventing lacteal chylomicron uptake by inducible endothelial genetic deletion of Neuropilin1 (Nrp1) and Vascular endothelial growth factor receptor 1 (Vegfr1; also known as Flt1) renders mice resistant to diet-induced obesity. Absence of NRP1 and FLT1 receptors increased VEGF-A bioavailability and signaling through VEGFR2, inducing lacteal junction zippering and chylomicron malabsorption. Restoring permeable lacteal junctions by VEGFR2 and vascular endothelial (VE)-cadherin signaling inhibition rescued chylomicron transport in the mutant mice. Zippering of lacteal junctions by disassembly of cytoskeletal VE-cadherin anchors prevented chylomicron uptake in wild-type mice. These data suggest that lacteal junctions may be targets for preventing dietary fat uptake.


Subject(s)
Chylomicrons/metabolism , Diet, High-Fat/adverse effects , Dietary Fats/metabolism , Neuropilin-1/genetics , Obesity/etiology , Obesity/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Animals , Antigens, CD/metabolism , Cadherins/antagonists & inhibitors , Cadherins/metabolism , Chylomicrons/adverse effects , Dietary Fats/adverse effects , Enterocytes/metabolism , Gene Deletion , Intestinal Absorption/genetics , Intestinal Absorption/physiology , Male , Mice , Mice, Knockout , Signal Transduction , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
10.
Nat Commun ; 8(1): 1571, 2017 11 17.
Article in English | MEDLINE | ID: mdl-29146996

ABSTRACT

CXCR3 plays important roles in angiogenesis, inflammation, and cancer. However, the precise mechanism of regulation and activity in tumors is not well known. We focused on CXCR3-A conformation and on the mechanisms controlling its activity and trafficking and investigated the role of CXCR3/LRP1 cross talk in tumor cell invasion. Here we report that agonist stimulation induces an anisotropic response with conformational changes of CXCR3-A along its longitudinal axis. CXCR3-A is internalized via clathrin-coated vesicles and recycled by retrograde trafficking. We demonstrate that CXCR3-A interacts with LRP1. Silencing of LRP1 leads to an increase in the magnitude of ligand-induced conformational change with CXCR3-A focalized at the cell membrane, leading to a sustained receptor activity and an increase in tumor cell migration. This was validated in patient-derived glioma cells and patient samples. Our study defines LRP1 as a regulator of CXCR3, which may have important consequences for tumor biology.


Subject(s)
Brain Neoplasms/pathology , Cell Movement/physiology , Glioblastoma/pathology , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Receptors, CXCR3/metabolism , Animals , Cell Membrane/metabolism , Chick Embryo , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Male , Mice , Mice, Knockout , Neoplasm Invasiveness/pathology , Protein Binding , Protein Transport/physiology , Spheroids, Cellular , Tumor Cells, Cultured
11.
ACS Chem Biol ; 12(11): 2767-2778, 2017 11 17.
Article in English | MEDLINE | ID: mdl-28945356

ABSTRACT

CXCL4 chemokines have antiangiogenic properties, mediated by different mechanisms, including CXCR3 receptor activation. Chemokines have distinct oligomerization states that are correlated with their biological functions. CXCL4 exists as a stable tetramer under physiological conditions. It is unclear whether the oligomerization state impacts CXCL4-receptor interaction. We found that the CXCL4 tetramer is sensitive to pH and salt concentration. Residues Glu28 and Lys50 were important for tetramer formation, and the first ß-strand and the C-terminal helix are critical for dimerization. By mutating the critical residues responsible for oligomerization, we generated CXCL4 mutants that behave as dimers or monomers under neutral/physiological conditions. The CXCL4 monomer acts as the minimal active unit for interacting CXCR3A, and sulfation of N-terminal tyrosine residues on the receptor is important for binding. Noticeably, CXCL4L1, a CXCL4 variant that differs by three residues in the C-terminal helix, could activate CXCR3A. CXCL4L1 showed a higher tendency to dissociate into monomers, but native CXCL4 did not. This result indicates that monomeric CXCL4 behaves like CXCL4L1. Thus, in this chemokine family, being in the monomeric state seems critical for interaction with CXCR3A.


Subject(s)
Platelet Factor 4/metabolism , Receptors, CXCR3/metabolism , Cell Line , Humans , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , Platelet Factor 4/chemistry , Protein Binding , Protein Conformation , Protein Multimerization , Receptors, CXCR3/chemistry
12.
Eur J Cell Biol ; 95(11): 503-512, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27720259

ABSTRACT

Transforming growth factor-ß1 (TGF-ß1) is an important player in chronic liver diseases inducing fibrogenesis and hepatocellular carcinoma (HCC) development. TGF-ß1 promotes pleiotropic modifications at the cellular and matrix microenvironment levels. TGF-ß1 was described to enhance production of type I collagen and its associated cross-linking enzyme, the lysyl oxidase-like2 (LOXL2). In addition, TGF-ß1 and type I collagen are potent inducers of invadosomes. Indeed, type I collagen fibers induce the formation of active linear invadosomes through the discoidin domain receptor 1 (DDR1). The goal of our study was to address the role of TGF-ß1 in collagen cross-linking and its impact on the formation of linear invadosomes in liver cancer cells. We first report a significant correlation between expressions of TGF-ß1, and type I collagen, LOXL2, DDR1 and MT1-MMP in human HCCs. We demonstrate that TGF-ß1 promotes a Smad4-dependent up-regulation of DDR1, together with LOXL2, in cultured HCC cells. Moreover, we show that LOXL2-induced collagen cross-linking enhances linear invadosome formation. Altogether, our data demonstrate that TGF-ß1 favors linear invadosome formation through the expressions of both the inducers, such as collagen and LOXL2, and the components such as DDR1 and MT1-MMP of linear invadosomes in cancer cells. Meanwhile, our data uncover a new TGF-ß1-dependent regulation of DDR1 expression.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Collagen Type I/metabolism , Discoidin Domain Receptor 1/biosynthesis , Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , Neoplasm Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Up-Regulation , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Collagen Type I/genetics , Discoidin Domain Receptor 1/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Transforming Growth Factor beta1/genetics
13.
Cancer Res ; 76(22): 6507-6519, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27634764

ABSTRACT

The CXCL4 paralog CXCL4L1 is a less studied chemokine that has been suggested to exert an antiangiogenic function. However, CXCL4L1 is also expressed in patient tumors, tumor cell lines, and murine xenografts, prompting a more detailed analysis of its role in cancer pathogenesis. We used genetic and antibody-based approaches to attenuate CXCL4L1 in models of pancreatic ductal adenocarcinoma (PDAC). Mechanisms of expression were assessed in cell coculture experiments, murine, and avian xenotransplants, including through an evaluation of CpG methylation and mutation of critical CpG residues. CXCL4L1 gene expression was increased greatly in primary and metastatic PDAC. We found that myofibroblasts triggered cues in the tumor microenvironment, which led to induction of CXCL4L1 in tumor cells. CXCL4L1 expression was also controlled by epigenetic modifications at critical CpG islands, which were mapped. CXCL4L1 inhibited angiogenesis but also affected tumor development more directly, depending on the tumor cell type. In vivo administration of an mAb against CXCL4L1 demonstrated a blockade in the growth of tumors positive for CXCR3, a critical receptor for CXCL4 ligands. Our findings define a protumorigenic role in PDAC development for endogenous CXCL4L1, which is independent of its antiangiogenic function. Cancer Res; 76(22); 6507-19. ©2016 AACR.


Subject(s)
Angiogenesis Inhibitors/genetics , Pancreatic Neoplasms/genetics , Receptors, CXCR3/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Chemokines , Humans , Mice , Neovascularization, Pathologic , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Platelet Factor 4 , Survival Analysis , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...