Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Circ Res ; 131(4): 308-327, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35862101

ABSTRACT

BACKGROUND: Pericytes and vascular smooth muscle cells, collectively known as mural cells, are recruited through PDGFB (platelet-derived growth factor B)-PDGFRB (platelet-derived growth factor receptor beta) signaling. MCs are essential for vascular integrity, and their loss has been associated with numerous diseases. Most of this knowledge is based on studies in which MCs are insufficiently recruited or fully absent upon inducible ablation. In contrast, little is known about the physiological consequences that result from impairment of specific MC functions. Here, we characterize the role of the transcription factor SRF (serum response factor) in MCs and study its function in developmental and pathological contexts. METHODS: We generated a mouse model of MC-specific inducible Srf gene deletion and studied its consequences during retinal angiogenesis using RNA-sequencing, immunohistology, in vivo live imaging, and in vitro techniques. RESULTS: By postnatal day 6, pericytes lacking SRF were morphologically abnormal and failed to properly comigrate with angiogenic sprouts. As a consequence, pericyte-deficient vessels at the retinal sprouting front became dilated and leaky. By postnatal day 12, also the vascular smooth muscle cells had lost SRF, which coincided with the formation of pathological arteriovenous shunts. Mechanistically, we show that PDGFB-dependent SRF activation is mediated via MRTF (myocardin-related transcription factor) cofactors. We further show that MRTF-SRF signaling promotes pathological pericyte activation during ischemic retinopathy. RNA-sequencing, immunohistology, in vivo live imaging, and in vitro experiments demonstrated that SRF regulates expression of contractile SMC proteins essential to maintain the vascular tone. CONCLUSIONS: SRF is crucial for distinct functions in pericytes and vascular smooth muscle cells. SRF directs pericyte migration downstream of PDGFRB signaling and mediates pathological pericyte activation during ischemic retinopathy. In vascular smooth muscle cells, SRF is essential for expression of the contractile machinery, and its deletion triggers formation of arteriovenous shunts. These essential roles in physiological and pathological contexts provide a rationale for novel therapeutic approaches through targeting SRF activity in MCs.


Subject(s)
Pericytes , Retinal Diseases , Animals , Mice , Pericytes/metabolism , Proto-Oncogene Proteins c-sis/metabolism , RNA/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Retinal Diseases/metabolism , Serum Response Factor/genetics , Serum Response Factor/metabolism
2.
Development ; 145(13)2018 07 02.
Article in English | MEDLINE | ID: mdl-29853619

ABSTRACT

Formation and homeostasis of the vascular system requires several coordinated cellular functions, but their precise interplay during development and their relative importance for vascular pathologies remain poorly understood. Here, we investigated the endothelial functions regulated by Cdc42 and their in vivo relevance during angiogenic sprouting and vascular morphogenesis in the postnatal mouse retina. We found that Cdc42 is required for endothelial tip cell selection, directed cell migration and filopodia formation, but dispensable for cell proliferation or apoptosis. Although the loss of Cdc42 seems generally compatible with apical-basal polarization and lumen formation in retinal blood vessels, it leads to defective endothelial axial polarization and to the formation of severe vascular malformations in capillaries and veins. Tracking of Cdc42-depleted endothelial cells in mosaic retinas suggests that these capillary-venous malformations arise as a consequence of defective cell migration, when endothelial cells that proliferate at normal rates are unable to re-distribute within the vascular network.


Subject(s)
Capillaries/abnormalities , Cell Movement , Endothelial Cells/metabolism , Retinal Vein/abnormalities , Vascular Malformations/embryology , cdc42 GTP-Binding Protein/deficiency , Animals , Capillaries/embryology , Cell Polarity/genetics , Endothelial Cells/pathology , Mice , Mice, Knockout , Pseudopodia/genetics , Pseudopodia/metabolism , Retinal Vein/embryology , Vascular Malformations/genetics , Vascular Malformations/pathology
3.
Circ Res ; 124(8): 1240-1252, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30732528

ABSTRACT

RATIONALE: Aberrant formation of blood vessels precedes a broad spectrum of vascular complications; however, the cellular and molecular events governing vascular malformations are not yet fully understood. OBJECTIVE: Here, we investigated the role of CDC42 (cell division cycle 42) during vascular morphogenesis and its relative importance for the development of cerebrovascular malformations. METHODS AND RESULTS: To avoid secondary systemic effects often associated with embryonic gene deletion, we generated an endothelial-specific and inducible knockout approach to study postnatal vascularization of the mouse brain. Postnatal endothelial-specific deletion of Cdc42 elicits cerebrovascular malformations reminiscent of cerebral cavernous malformations (CCMs). At the cellular level, loss of CDC42 function in brain endothelial cells (ECs) impairs their sprouting, branching morphogenesis, axial polarity, and normal dispersion within the brain tissue. Disruption of CDC42 does not alter EC proliferation, but malformations occur where EC proliferation is the most pronounced during brain development-the postnatal cerebellum-indicating that a high, naturally occurring EC proliferation provides a permissive state for the appearance of these malformations. Mechanistically, CDC42 depletion in ECs elicited increased MEKK3 (mitogen-activated protein kinase kinase kinase 3)-MEK5 (mitogen-activated protein kinase kinase 5)-ERK5 (extracellular signal-regulated kinase 5) signaling and consequent detrimental overexpression of KLF (Kruppel-like factor) 2 and KLF4, recapitulating the hallmark mechanism for CCM pathogenesis. Through genetic approaches, we demonstrate that the coinactivation of Klf4 reduces the severity of vascular malformations in Cdc42 mutant mice. Moreover, we show that CDC42 interacts with CCMs and that CCM3 promotes CDC42 activity in ECs. CONCLUSIONS: We show that endothelial-specific deletion of Cdc42 elicits CCM-like cerebrovascular malformations and that CDC42 is engaged in the CCM signaling network to restrain the MEKK3-MEK5-ERK5-KLF2/4 pathway.


Subject(s)
Blood Vessels/abnormalities , Cell Proliferation , Endothelial Cells/physiology , Gene Deletion , Hemangioma, Cavernous, Central Nervous System/etiology , cdc42 GTP-Binding Protein/genetics , Animals , Animals, Newborn , Apoptosis Regulatory Proteins/genetics , Brain/blood supply , Cell Cycle/physiology , KRIT1 Protein/genetics , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , MAP Kinase Kinase 5/metabolism , MAP Kinase Kinase Kinase 3/metabolism , Mice , Microfilament Proteins/genetics , cdc42 GTP-Binding Protein/metabolism
4.
PLoS Genet ; 14(5): e1007391, 2018 05.
Article in English | MEDLINE | ID: mdl-29758044

ABSTRACT

Planar cell polarity (PCP) instructs tissue patterning in a wide range of organisms from fruit flies to humans. PCP signaling coordinates cell behavior across tissues and is integrated by cells to couple cell fate identity with position in a developing tissue. In the fly eye, PCP signaling is required for the specification of R3 and R4 photoreceptors based upon their positioning relative to the dorso-ventral axis. The 'core' PCP pathway involves the asymmetric localization of two distinct membrane-bound complexes, one containing Frizzled (Fz, required in R3) and the other Van Gogh (Vang, required in R4). Inhibitory interactions between the cytosolic components of each complex reinforce asymmetric localization. Prickle (Pk) and Spiny-legs (Pk-Sple) are two antagonistic isoforms of the prickle (pk) gene and are cytoplasmic components of the Vang complex. The balance between their levels is critical for tissue patterning, with Pk-Sple being the major functional isoform in the eye. Here we uncover a post-translational role for Nemo kinase in limiting the amount of the minor isoform Pk. We identified Pk as a Nemo substrate in a genome-wide in vitro band-shift screen. In vivo, nemo genetically interacts with pkpk but not pksple and enhances PCP defects in the eye and leg. Nemo phosphorylation limits Pk levels and is required specifically in the R4 photoreceptor like the major isoform, Pk-Sple. Genetic interaction and biochemical data suggest that Nemo phosphorylation of Pk leads to its proteasomal degradation via the Cullin1/SkpA/Slmb complex. dTAK and Homeodomain interacting protein kinase (Hipk) may also act together with Nemo to target Pk for degradation, consistent with similar observations in mammalian studies. Our results therefore demonstrate a mechanism to maintain low levels of the minor Pk isoform, allowing PCP complexes to form correctly and specify cell fate.


Subject(s)
Cell Polarity/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , LIM Domain Proteins/genetics , Mitogen-Activated Protein Kinases/genetics , Animals , Animals, Genetically Modified , Cell Line , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Eye/cytology , Eye/metabolism , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , LIM Domain Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proteolysis , Signal Transduction/genetics , Substrate Specificity , Wings, Animal/cytology , Wings, Animal/metabolism
5.
N Engl J Med ; 386(13): 1291, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35353972
6.
Development ; 141(23): 4489-99, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25406396

ABSTRACT

Vascular development of the central nervous system and blood-brain barrier (BBB) induction are closely linked processes. The role of factors that promote endothelial sprouting and vascular leak, such as vascular endothelial growth factor A, are well described, but the factors that suppress angiogenic sprouting and their impact on the BBB are poorly understood. Here, we show that integrin αVß8 activates angiosuppressive TGFß gradients in the brain, which inhibit endothelial cell sprouting. Loss of αVß8 in the brain or downstream TGFß1-TGFBR2-ALK5-Smad3 signaling in endothelial cells increases vascular sprouting, branching and proliferation, leading to vascular dysplasia and hemorrhage. Importantly, BBB function in Itgb8 mutants is intact during early stages of vascular dysgenesis before hemorrhage. By contrast, Pdgfb(ret/ret) mice, which exhibit severe BBB disruption and vascular leak due to pericyte deficiency, have comparatively normal vascular morphogenesis and do not exhibit brain hemorrhage. Our data therefore suggest that abnormal vascular sprouting and patterning, not BBB dysfunction, underlie developmental cerebral hemorrhage.


Subject(s)
Blood-Brain Barrier/physiology , Brain/blood supply , Cerebral Hemorrhage/etiology , Neovascularization, Pathologic/complications , Signal Transduction/physiology , Analysis of Variance , Animals , Brain/metabolism , Cell Count , Endothelial Cells/physiology , Immunohistochemistry , Integrins/metabolism , Mice , Microscopy, Confocal , Transforming Growth Factor beta/metabolism
7.
Curr Opin Hematol ; 22(3): 258-66, 2015 May.
Article in English | MEDLINE | ID: mdl-25767958

ABSTRACT

PURPOSE OF REVIEW: Throughout history, development of novel microscopy techniques has been of fundamental importance to advance the vascular biology field.This review offers a concise summary of the most recently developed imaging techniques and discusses how they can be applied to vascular biology. In addition, we reflect upon the most important fluorescent reporters for vascular research that are currently available. RECENT FINDINGS: Recent advances in light sheet-based imaging techniques now offer the ability to live image the vascular system in whole organs or even in whole animals during development and in pathological conditions with a satisfactory spatial and temporal resolution. Conversely, super resolution microscopy now allows studying cellular processes at a near-molecular resolution. SUMMARY: Major recent improvements in a number of imaging techniques now allow study of vascular biology in ways that could not be considered previously. Researchers now have well-developed tools to specifically examine the dynamic nature of vascular development during angiogenic sprouting, remodeling and regression as well as the vascular responses in disease situations in vivo. In addition, open questions in endothelial and lymphatic cell biology that require subcellular resolution such as actin dynamics, junctional complex formation and stability, vascular permeability and receptor trafficking can now be approached with high resolution.


Subject(s)
Blood Vessels/metabolism , Image Processing, Computer-Assisted/methods , Humans
8.
Cell Rep ; 43(3): 113911, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38446668

ABSTRACT

Claudin-5 (CLDN5) is an endothelial tight junction protein essential for blood-brain barrier (BBB) formation. Abnormal CLDN5 expression is common in brain disease, and knockdown of Cldn5 at the BBB has been proposed to facilitate drug delivery to the brain. To study the consequences of CLDN5 loss in the mature brain, we induced mosaic endothelial-specific Cldn5 gene ablation in adult mice (Cldn5iECKO). These mice displayed increased BBB permeability to tracers up to 10 kDa in size from 6 days post induction (dpi) and ensuing lethality from 10 dpi. Single-cell RNA sequencing at 11 dpi revealed profound transcriptomic differences in brain endothelial cells regardless of their Cldn5 status in mosaic mice, suggesting major non-cell-autonomous responses. Reactive microglia and astrocytes suggested rapid cellular responses to BBB leakage. Our study demonstrates a critical role for CLDN5 in the adult BBB and provides molecular insight into the consequences and risks associated with CLDN5 inhibition.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Animals , Mice , Biological Transport , Blood-Brain Barrier/metabolism , Brain/metabolism , Claudin-5/genetics , Claudin-5/metabolism , Endothelial Cells/metabolism
9.
Nat Cardiovasc Res ; 2: 2023530-549, 2023 May 29.
Article in English | MEDLINE | ID: mdl-37745941

ABSTRACT

The Notch pathway is a major regulator of endothelial transcriptional specification. Targeting the Notch receptors or Delta-like ligand 4 (Dll4) dysregulates angiogenesis. Here, by analyzing single and compound genetic mutants for all Notch signaling members, we find significant differences in the way ligands and receptors regulate liver vascular homeostasis. Loss of Notch receptors caused endothelial hypermitogenic cell-cycle arrest and senescence. Conversely, Dll4 loss triggered a strong Myc-driven transcriptional switch inducing endothelial proliferation and the tip-cell state. Myc loss suppressed the induction of angiogenesis in the absence of Dll4, without preventing the vascular enlargement and organ pathology. Similarly, inhibition of other pro-angiogenic pathways, including MAPK/ERK and mTOR, had no effect on the vascular expansion induced by Dll4 loss; however, anti-VEGFA treatment prevented it without fully suppressing the transcriptional and metabolic programs. This study shows incongruence between single-cell transcriptional states, vascular phenotypes and related pathophysiology. Our findings also suggest that the vascular structure abnormalization, rather than neoplasms, causes the reported anti-Dll4 antibody toxicity.

10.
Arterioscler Thromb Vasc Biol ; 29(5): 630-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19164813

ABSTRACT

Mural cells are essential components of blood vessels and are necessary for normal development, homeostasis, and organ function. Alterations in mural cell density or the stable attachment of mural cells to the endothelium is associated with several human diseases such as diabetic retinopathy, venous malformation, and hereditary stroke. In addition mural cells are implicated in regulating tumor growth and have thus been suggested as potential antiangiogenic targets in tumor therapy. In recent years our knowledge of mural cell function and endothelial-mural cell signaling has increased dramatically, and we now begin to understand the mechanistic basis of the key signaling pathways involved. This is mainly thanks to sophisticated in vivo experiments using a broad repertoire of genetic technologies. In this review, we summarize the five currently best understood signaling pathways implicated in mural cell biology. We discuss PDGFB/PDGFRbeta- dependent pericyte recruitment, as well as the role of angiopoietins and Tie receptors in vascular maturation. In addition, we highlight the effects of sphingosine-1-phosphate signaling on adherens junction assembly and vascular stability, as well as the role of TGF-beta-signaling in mural cell differentiation. We further reflect recent data suggesting an important function for Notch3 signaling in mural cell maturation.


Subject(s)
Neovascularization, Physiologic/physiology , Pericytes/physiology , Signal Transduction/physiology , Adherens Junctions/physiology , Angiopoietins/physiology , Animals , Lysophospholipids/physiology , Mice , Mice, Knockout , Neovascularization, Pathologic/physiopathology , Receptor, Notch3 , Receptor, Platelet-Derived Growth Factor beta/physiology , Receptors, Notch/physiology , Receptors, TIE/physiology , Sphingosine/analogs & derivatives , Sphingosine/physiology
11.
Dev Cell ; 52(6): 779-793.e7, 2020 03 23.
Article in English | MEDLINE | ID: mdl-32059774

ABSTRACT

Transcriptional mechanisms that drive angiogenesis and organotypic vascular endothelial cell specialization are poorly understood. Here, we show that retinal endothelial sphingosine 1-phosphate receptors (S1PRs), which restrain vascular endothelial growth factor (VEGF)-induced angiogenesis, spatially restrict expression of JunB, a member of the activator protein 1 (AP-1) family of transcription factors (TFs). Mechanistically, VEGF induces JunB expression at the sprouting vascular front while S1PR-dependent vascular endothelial (VE)-cadherin assembly suppresses JunB expression in the nascent vascular network, thus creating a gradient of this TF. Endothelial-specific JunB knockout mice showed diminished expression of neurovascular guidance genes and attenuated retinal vascular network progression. In addition, endothelial S1PR signaling is required for normal expression of ß-catenin-dependent genes such as TCF/LEF1 and ZIC3 TFs, transporters, and junctional proteins. These results show that S1PR signaling restricts JunB function to the expanding vascular front, thus creating an AP-1 gradient and enabling organotypic endothelial cell specialization of the vascular network.


Subject(s)
Endothelial Cells/metabolism , Neovascularization, Physiologic , Retinal Vessels/metabolism , Signal Transduction , Sphingosine-1-Phosphate Receptors/metabolism , Transcription Factor AP-1/metabolism , Animals , Cells, Cultured , Chromatin Assembly and Disassembly , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Retinal Vessels/cytology , Retinal Vessels/embryology , Transcription Factor AP-1/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
12.
Open Biol ; 9(8): 190148, 2019 08 30.
Article in English | MEDLINE | ID: mdl-31409231

ABSTRACT

Integrins mediate the anchorage between cells and their environment, the extracellular matrix (ECM), and form transmembrane links between the ECM and the cytoskeleton, a conserved feature throughout development and morphogenesis of epithelial organs. Here, we demonstrate that integrins and components of the ECM are required during the planar cell polarity (PCP) signalling-regulated cell movement of ommatidial rotation in the Drosophila eye. The loss-of-function mutations of integrins or ECM components cause defects in rotation, with mutant clusters rotating asynchronously compared to wild-type clusters. Initially, mutant clusters tend to rotate faster, and at later stages they fail to be synchronous with their neighbours, leading to aberrant rotation angles and resulting in a disorganized ommatidial arrangement in adult eyes. We further demonstrate that integrin localization changes dynamically during the rotation process. Our data suggest that core Frizzled/PCP factors, acting through RhoA and Rho kinase, regulate the function/activity of integrins and that integrins thus contribute to the complex interaction network of PCP signalling, cell adhesion and cytoskeletal elements required for a precise and synchronous 90° rotation movement.


Subject(s)
Drosophila/embryology , Drosophila/physiology , Extracellular Matrix/metabolism , Eye/embryology , Eye/metabolism , Integrins/genetics , Signal Transduction , Animals , Body Patterning , Cell Polarity , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Eye/cytology , Gene Deletion , Gene Expression Regulation, Developmental , Immunohistochemistry , Integrins/metabolism , Models, Biological , Mutation , Protein Transport
13.
Methods Mol Biol ; 420: 277-87, 2008.
Article in English | MEDLINE | ID: mdl-18641954

ABSTRACT

The regular appearance and the repetitive nature of the Drosophila eye, consisting of several hundred identical multicellular units, the ommatidia, has long served as an invaluable experimental system to study cell-cell interactions, inductive signaling events, cell proliferation, programmed cell death, cell differentiation, cell organization, and planar cell polarity among others. Importantly, the eye is dispensable for viability and fertility of the fly and thus, it can easily be manipulated, making it an ideal target for genetic screens. This chapter described an essential technique in the analysis of different genotypes in the adult fly eye, and allows detailed analyses with single cell resolution.


Subject(s)
Drosophila melanogaster/embryology , Genetic Techniques , Photoreceptor Cells, Invertebrate/embryology , Retina/embryology , Animals , Body Patterning , Cell Differentiation , Cell Polarity , Cell Proliferation , Cell Survival , Drosophila Proteins/metabolism , Genotype , Image Processing, Computer-Assisted , Microscopy/methods , Signal Transduction
14.
J Cereb Blood Flow Metab ; 38(3): 456-468, 2018 03.
Article in English | MEDLINE | ID: mdl-28276839

ABSTRACT

The establishment of a fully functional blood vascular system requires elaborate angiogenic and vascular maturation events in order to fulfill organ-specific anatomical and physiological needs. Although vascular mural cells, i.e. pericytes and vascular smooth muscle cells, are known to play fundamental roles during these processes, their characteristics during vascular development remain incompletely understood. In this report, we utilized transgenic reporter mice in which mural cells are genetically labeled to examine developing vascular mural cells in the central nervous system (CNS). We found platelet-derived growth factor receptor ß gene ( Pdgfrb)-driven EGFP reporter expression as a suitable marker for vascular mural cells at the earliest stages of mouse brain vascularization. Furthermore, the combination of Pdgfrb and NG2 gene (Cspg4) driven reporter expression increased the specificity of brain vascular mural cell labeling at later stages. The expression of other known pericyte markers revealed time-, region- and marker-specific patterns, suggesting heterogeneity in mural cell maturation. We conclude that transgenic reporter mice provide an important tool to explore the development of CNS pericytes in health and disease.


Subject(s)
Blood Vessels/ultrastructure , Brain/cytology , Brain/growth & development , Genes, Reporter/genetics , Animals , Antigens/genetics , Blood Vessels/growth & development , Brain/ultrastructure , Cerebral Cortex/growth & development , Cerebral Cortex/ultrastructure , Embryonic Development , Female , Mice , Mice, Transgenic , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle/ultrastructure , Pericytes/ultrastructure , Proteoglycans/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics
15.
Curr Top Dev Biol ; 123: 433-482, 2017.
Article in English | MEDLINE | ID: mdl-28236974

ABSTRACT

Vascular endothelial growth factor receptor (VEGFR) tyrosine kinases are key regulators of vascular development in vertebrates. Their activation is regulated through a family of secreted glycoproteins, the vascular endothelial growth factors (VEGFs). Expression, proteolytic processing, and diffusion range of VEGF proteins need to be tightly regulated, due to their crucial roles in development. While some VEGFs form concentration gradients across developing tissues and act as morphogenes, others function as inhibitors of receptor activation and downstream signaling. Ligand-induced receptor dimerization leads to activation of the intrinsic tyrosine kinase activity, which results in autophosphorylation of the receptors and in turn triggers the recruitment of interacting proteins as well as the initiation of downstream signaling. Although many biochemical details of VEGFR signaling have been revealed, the in vivo relevance of certain signaling aspects still remains to be demonstrated. Here, we highlight basic principles of VEGFR signaling and discuss its crucial role during development of the vascular system in mammals.


Subject(s)
Blood Vessels/physiology , Receptors, Vascular Endothelial Growth Factor/metabolism , Animals , Evolution, Molecular , Humans , Ligands , Neuropilins/metabolism , Receptors, Vascular Endothelial Growth Factor/chemistry , Signal Transduction
16.
PLoS One ; 10(9): e0137949, 2015.
Article in English | MEDLINE | ID: mdl-26394398

ABSTRACT

Despite its known expression in both the vascular endothelium and the lung epithelium, until recently the physiological role of the adhesion receptor Gpr116/ADGRF5 has remained elusive. We generated a new mouse model of constitutive Gpr116 inactivation, with a large genetic deletion encompassing exon 4 to exon 21 of the Gpr116 gene. This model allowed us to confirm recent results defining Gpr116 as necessary regulator of surfactant homeostasis. The loss of Gpr116 provokes an early accumulation of surfactant in the lungs, followed by a massive infiltration of macrophages, and eventually progresses into an emphysema-like pathology. Further analysis of this knockout model revealed cerebral vascular leakage, beginning at around 1.5 months of age. Additionally, endothelial-specific deletion of Gpr116 resulted in a significant increase of the brain vascular leakage. Mice devoid of Gpr116 developed an anatomically normal and largely functional vascular network, surprisingly exhibited an attenuated pathological retinal vascular response in a model of oxygen-induced retinopathy. These data suggest that Gpr116 modulates endothelial properties, a previously unappreciated function despite the pan-vascular expression of this receptor. Our results support the key pulmonary function of Gpr116 and describe a new role in the central nervous system vasculature.


Subject(s)
Alveolar Epithelial Cells/metabolism , Endothelium, Vascular/metabolism , Pulmonary Surfactants/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Blood-Brain Barrier/metabolism , Blotting, Western , Bronchoalveolar Lavage Fluid/chemistry , Capillary Permeability/genetics , Female , Gene Expression , Homeostasis/genetics , Lung/metabolism , Lung/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Models, Biological , Myocardium/metabolism , Myocardium/pathology , Receptors, G-Protein-Coupled/genetics , Retinal Neovascularization/genetics , Retinal Neovascularization/metabolism , Spleen/metabolism , Spleen/pathology
17.
Nat Cell Biol ; 15(3): 233-5, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23449144

ABSTRACT

Endocytosis has proved to be a versatile mechanism regulating diverse cellular processes, ranging from nutrient uptake to intracellular signal transduction. New work reinforces the importance of endocytosis for VEGF receptor signalling and angiogenesis in the developing eye, and describes a mechanism for its differential regulation in angiogenic versus quiescent endothelial cells.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Cell Adhesion Molecules/metabolism , Endocytosis/physiology , Endothelium, Vascular/cytology , Ephrin-B2/metabolism , Neovascularization, Physiologic , Protein Kinase C/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Adaptor Proteins, Signal Transducing , Animals , Apoptosis Regulatory Proteins , Cell Cycle Proteins , Female , Humans , Male
18.
Dev Cell ; 23(3): 587-99, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22975327

ABSTRACT

Angiogenesis, the process by which new blood vessels arise from preexisting ones, is critical for embryonic development and is an integral part of many disease processes. Recent studies have provided detailed information on how angiogenic sprouts initiate, elongate, and branch, but less is known about how these processes cease. Here, we show that S1PR1, a receptor for the blood-borne bioactive lipid sphingosine-1-phosphate (S1P), is critical for inhibition of angiogenesis and acquisition of vascular stability. Loss of S1PR1 leads to increased endothelial cell sprouting and the formation of ectopic vessel branches. Conversely, S1PR1 signaling inhibits angiogenic sprouting and enhances cell-to-cell adhesion. This correlates with inhibition of vascular endothelial growth factor-A (VEGF-A)-induced signaling and stabilization of vascular endothelial (VE)-cadherin localization at endothelial junctions. Our data suggest that S1PR1 signaling acts as a vascular-intrinsic stabilization mechanism, protecting developing blood vessels against aberrant angiogenic responses.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Neovascularization, Physiologic , Receptors, Lysosphingolipid/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cells, Cultured , Endothelial Cells/metabolism , Humans , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Lysosphingolipid/deficiency , Sphingosine-1-Phosphate Receptors , Zebrafish
19.
Nat Struct Mol Biol ; 18(6): 665-72, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21552260

ABSTRACT

Frizzled planar cell polarity (PCP) signaling regulates cell motility in several tissues, including ommatidial rotation in Drosophila melanogaster. The Nemo kinase (Nlk in vertebrates) has also been linked to cell-motility regulation and ommatidial rotation but its mechanistic role(s) during rotation remain obscure. We show that nemo functions throughout the entire rotation movement, increasing the rotation rate. Genetic and molecular studies indicate that Nemo binds both the core PCP factor complex of Strabismus-Prickle, as well as the E-cadherin-ß-catenin (E-cadherin-Armadillo in Drosophila) complex. These two complexes colocalize and, like Nemo, also promote rotation. Strabismus (also called Vang) binds and stabilizes Nemo asymmetrically within the ommatidial precluster; Nemo and ß-catenin then act synergistically to promote rotation, which is mediated in vivo by Nemo's phosphorylation of ß-catenin. Our data suggest that Nemo serves as a conserved molecular link between core PCP factors and E-cadherin-ß-catenin complexes, promoting cell motility.


Subject(s)
Cadherins/metabolism , Cell Movement , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , beta Catenin/metabolism , Animals , Drosophila melanogaster/physiology , LIM Domain Proteins , Ocular Physiological Phenomena , Phosphorylation , Protein Binding
20.
Hum Mol Genet ; 15(4): 543-53, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16399795

ABSTRACT

Mutations in the PTPN11 gene, which encodes the protein tyrosine phosphatase SHP-2, causes Noonan syndrome (NS), an autosomal dominant disorder with pleomorphic developmental abnormalities. Certain germline and somatic PTPN11 mutations cause leukemias. Mutations have gain-of-function (GOF) effects with the commonest NS allele, N308D, being weaker than the leukemia-causing mutations. To study the effects of disease-associated PTPN11 alleles, we generated transgenic fruitflies with GAL4-inducible expression of wild-type or mutant csw, the Drosophila orthologue of PTPN11. All three transgenic mutant CSWs rescued a hypomorphic csw allele's eye phenotype, documenting activity. Ubiquitous expression of two strong csw mutant alleles were lethal, but did not perturb development from some CSW-dependent receptor tyrosine kinase pathways. Ubiquitous expression of the weaker N308D allele caused ectopic wing veins, identical to the EGFR GOF phenotype. Epistatic analyses established that csw(N308D)'s ectopic wing vein phenotype required intact EGF ligand and receptor, and that this transgene interacted genetically with Notch, DPP and JAK/STAT signaling. Expression of the mutant csw transgenes increased RAS-MAP kinase activation, which was necessary but not sufficient for transducing their phenotypes. The findings from these fly models provided hypotheses testable in mammalian models, in which these signaling cassettes are largely conserved. In addition, these fly models can be used for sensitized screens to identify novel interacting genes as well as for high-throughput screening of therapeutic compounds for NS and PTPN11-related cancers.


Subject(s)
Drosophila Proteins/genetics , Genes, Dominant/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Noonan Syndrome/genetics , Protein Tyrosine Phosphatases/genetics , Signal Transduction/genetics , Animals , Animals, Genetically Modified , Disease Models, Animal , Drosophila , Drosophila Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Noonan Syndrome/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL