Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
bioRxiv ; 2023 Sep 25.
Article in English | MEDLINE | ID: mdl-37808725

ABSTRACT

In brief: The mechanisms regulating the signaling pathways involved in angiogenesis are not fully known. Ristori et al. show that Lunatic Fringe (LFng) mediates the crosstalk between Bone Morphogenic Protein 9 (Bmp9) and Notch signaling, thereby regulating the endothelial cell behavior and temporal dynamics of their identity during sprouting angiogenesis. Highlights: Bmp9 upregulates the expression of LFng in endothelial cells.LFng regulates the temporal dynamics of tip/stalk selection and rearrangement.LFng indicated to play a role in hereditary hemorrhagic telangiectasia.Bmp9 and LFng mediate the endothelial cell-pericyte crosstalk.Bone Morphogenic Protein 9 (Bmp9), whose signaling through Activin receptor-like kinase 1 (Alk1) is involved in several diseases, has been shown to independently activate Notch target genes in an additive fashion with canonical Notch signaling. Here, by integrating predictive computational modeling validated with experiments, we uncover that Bmp9 upregulates Lunatic Fringe (LFng) in endothelial cells (ECs), and thereby also regulates Notch activity in an inter-dependent, multiplicative fashion. Specifically, the Bmp9-upregulated LFng enhances Notch receptor activity creating a much stronger effect when Dll4 ligands are also present. During sprouting, this LFng regulation alters vessel branching by modulating the timing of EC phenotype selection and rearrangement. Our results further indicate that LFng can play a role in Bmp9-related diseases and in pericyte-driven vessel stabilization, since we find LFng contributes to Jag1 upregulation in Bmp9-stimulated ECs; thus, Bmp9-upregulated LFng results in not only enhanced EC Dll4-Notch1 activation, but also Jag1-Notch3 activation in pericytes.

2.
iScience ; 25(5): 104306, 2022 May 20.
Article in English | MEDLINE | ID: mdl-35602952

ABSTRACT

Spatial regulation of angiogenesis is important for the generation of functional engineered vasculature in regenerative medicine. The Notch ligands Jag1 and Dll4 show distinct expression patterns in endothelial cells and, respectively, promote and inhibit endothelial sprouting. Therefore, patterns of Notch ligands may be utilized to spatially control sprouting, but their potential and the underlying mechanisms of action are unclear. Here, we coupled in vitro and in silico models to analyze the ability of micropatterned Jag1 and Dll4 ligands to spatially control endothelial sprouting. Dll4 patterns, but not Jag1 patterns, elicited spatial control. Computational simulations of the underlying signaling dynamics suggest that different timing of Notch activation by Jag1 and Dll4 underlie their distinct ability to spatially control sprouting. Hence, Dll4 patterns efficiently direct the sprouts, whereas longer exposure to Jag1 patterns is required to achieve spatial control. These insights in sprouting regulation offer therapeutic handles for spatial regulation of angiogenesis.

3.
Elife ; 112022 04 19.
Article in English | MEDLINE | ID: mdl-35438077

ABSTRACT

Coordination of cell proliferation and migration is fundamental for life, and its dysregulation has catastrophic consequences, such as cancer. How cell cycle progression affects migration, and vice versa, remains largely unknown. We address these questions by combining in silico modelling and in vivo experimentation in the zebrafish trunk neural crest (TNC). TNC migrate collectively, forming chains with a leader cell directing the movement of trailing followers. We show that the acquisition of migratory identity is autonomously controlled by Notch signalling in TNC. High Notch activity defines leaders, while low Notch determines followers. Moreover, cell cycle progression is required for TNC migration and is regulated by Notch. Cells with low Notch activity stay longer in G1 and become followers, while leaders with high Notch activity quickly undergo G1/S transition and remain in S-phase longer. In conclusion, TNC migratory identities are defined through the interaction of Notch signalling and cell cycle progression.


Subject(s)
Neural Crest , Zebrafish , Animals , Cell Division , Cell Movement/physiology , Signal Transduction , Zebrafish/physiology
4.
PLoS Comput Biol ; 17(7): e1009146, 2021 07.
Article in English | MEDLINE | ID: mdl-34252083

ABSTRACT

SARS-CoV-2 has spread across the world, causing high mortality and unprecedented restrictions on social and economic activity. Policymakers are assessing how best to navigate through the ongoing epidemic, with computational models being used to predict the spread of infection and assess the impact of public health measures. Here, we present OpenABM-Covid19: an agent-based simulation of the epidemic including detailed age-stratification and realistic social networks. By default the model is parameterised to UK demographics and calibrated to the UK epidemic, however, it can easily be re-parameterised for other countries. OpenABM-Covid19 can evaluate non-pharmaceutical interventions, including both manual and digital contact tracing, and vaccination programmes. It can simulate a population of 1 million people in seconds per day, allowing parameter sweeps and formal statistical model-based inference. The code is open-source and has been developed by teams both inside and outside academia, with an emphasis on formal testing, documentation, modularity and transparency. A key feature of OpenABM-Covid19 are its Python and R interfaces, which has allowed scientists and policymakers to simulate dynamic packages of interventions and help compare options to suppress the COVID-19 epidemic.


Subject(s)
COVID-19/prevention & control , Contact Tracing , Systems Analysis , COVID-19/epidemiology , COVID-19/transmission , COVID-19/virology , COVID-19 Testing , COVID-19 Vaccines/administration & dosage , Disease Outbreaks , Humans , Physical Distancing , Quarantine , SARS-CoV-2/isolation & purification
5.
Philos Trans R Soc Lond B Biol Sci ; 376(1821): 20190753, 2021 03 29.
Article in English | MEDLINE | ID: mdl-33550953

ABSTRACT

How do cells make efficient collective decisions during tissue morphogenesis? Humans and other organisms use feedback between movement and sensing known as 'sensorimotor coordination' or 'active perception' to inform behaviour, but active perception has not before been investigated at a cellular level within organs. Here we provide the first proof of concept in silico/in vivo study demonstrating that filopodia (actin-rich, dynamic, finger-like cell membrane protrusions) play an unexpected role in speeding up collective endothelial decisions during the time-constrained process of 'tip cell' selection during blood vessel formation (angiogenesis). We first validate simulation predictions in vivo with live imaging of zebrafish intersegmental vessel growth. Further simulation studies then indicate the effect is due to the coupled positive feedback between movement and sensing on filopodia conferring a bistable switch-like property to Notch lateral inhibition, ensuring tip selection is a rapid and robust process. We then employ measures from computational neuroscience to assess whether filopodia function as a primitive (basal) form of active perception and find evidence in support. By viewing cell behaviour through the 'basal cognitive lens' we acquire a fresh perspective on the tip cell selection process, revealing a hidden, yet vital time-keeping role for filopodia. Finally, we discuss a myriad of new and exciting research directions stemming from our conceptual approach to interpreting cell behaviour. This article is part of the theme issue 'Basal cognition: multicellularity, neurons and the cognitive lens'.


Subject(s)
Morphogenesis/physiology , Pseudopodia/physiology , Zebrafish/physiology , Actins/metabolism , Animals , Computer Simulation , Perception
6.
J Clin Invest ; 130(8): 4235-4251, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32427589

ABSTRACT

Aberrant, neovascular retinal blood vessel growth is a vision-threatening complication in ischemic retinal diseases. It is driven by retinal hypoxia frequently caused by capillary nonperfusion and endothelial cell (EC) loss. We investigated the role of EC apoptosis in this process using a mouse model of ischemic retinopathy, in which vessel closure and EC apoptosis cause capillary regression and retinal ischemia followed by neovascularization. Protecting ECs from apoptosis in this model did not prevent capillary closure or retinal ischemia. Nonetheless, it prevented the clearance of ECs from closed capillaries, delaying vessel regression and allowing ECs to persist in clusters throughout the ischemic zone. In response to hypoxia, these preserved ECs underwent a vessel sprouting response and rapidly reassembled into a functional vascular network. This alleviated retinal hypoxia, preventing subsequent pathogenic neovascularization. Vessel reassembly was not limited by VEGFA neutralization, suggesting it was not dependent on the excess VEGFA produced by the ischemic retina. Neutralization of ANG2 did not prevent vessel reassembly, but did impair subsequent angiogenic expansion of the reassembled vessels. Blockade of EC apoptosis may promote ischemic tissue revascularization by preserving ECs within ischemic tissue that retain the capacity to reassemble a functional network and rapidly restore blood supply.


Subject(s)
Apoptosis , Endothelial Cells/metabolism , Ischemia/metabolism , Retinal Vessels/metabolism , Ribonuclease, Pancreatic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Disease Models, Animal , Endothelial Cells/pathology , Ischemia/genetics , Ischemia/pathology , Mice , Mice, Knockout , Retinal Diseases , Retinal Vessels/pathology , Ribonuclease, Pancreatic/genetics , Vascular Endothelial Growth Factor A/genetics
7.
Elife ; 92020 02 19.
Article in English | MEDLINE | ID: mdl-32073398

ABSTRACT

As the general population ages, more people are affected by eye diseases, such as retinopathies. It is therefore critical to improve imaging of eye disease mouse models. Here, we demonstrate that 1) rapid, quantitative 3D and 4D (time lapse) imaging of cellular and subcellular processes in the mouse eye is feasible, with and without tissue clearing, using light-sheet fluorescent microscopy (LSFM); 2) flat-mounting retinas for confocal microscopy significantly distorts tissue morphology, confirmed by quantitative correlative LSFM-Confocal imaging of vessels; 3) LSFM readily reveals new features of even well-studied eye disease mouse models, such as the oxygen-induced retinopathy (OIR) model, including a previously unappreciated 'knotted' morphology to pathological vascular tufts, abnormal cell motility and altered filopodia dynamics when live-imaged. We conclude that quantitative 3D/4D LSFM imaging and analysis has the potential to advance our understanding of the eye, in particular pathological, neurovascular, degenerative processes.


Eye diseases affect millions of people worldwide and can have devasting effects on people's lives. To find new treatments, scientists need to understand more about how these diseases arise and how they progress. This is challenging and progress has been held back by limitations in current techniques for looking at the eye. Currently, the most commonly used method is called confocal imaging, which is slow and distorts the tissue. Distortion happens because confocal imaging requires that thin slices of eye tissue from mice used in experiments are flattened on slides; this makes it hard to accurately visualize three-dimensional structures in the eye. New methods are emerging that may help. One promising method is called light-sheet fluorescent microscopy (or LSFM for short). This method captures three-dimensional images of the blood vessels and cells in the eye. It is much faster than confocal imaging and allows scientists to image tissues without slicing or flattening them. This could lead to more accurate three-dimensional images of eye disease. Now, Prahst et al. show that LSFM can quickly produce highly detailed, three-dimensional images of mouse retinas, from the smallest parts of cells to the entire eye. The technique also identified new features in a well-studied model of retina damage caused by excessive oxygen exposure in young mice. Previous studies of this model suggested the disease caused blood vessels in the eye to balloon, hinting that drugs that shrink blood vessels would help. But using LSFM, Prahst et al. revealed that these blood vessels actually take on a twisted and knotted shape. This suggests that treatments that untangle the vessels rather than shrink them are needed. The experiments show that LSFM is a valuable tool for studying eye diseases, that may help scientists learn more about how these diseases arise and develop. These new insights may one day lead to better tests and treatments for eye diseases.


Subject(s)
Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Retina/physiology , Animals , Eye Diseases/diagnosis , Eye Diseases/therapy , Imaging, Three-Dimensional/methods , Mice , Retinal Vessels/diagnostic imaging
8.
Sci Rep ; 9(1): 15022, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31636306

ABSTRACT

Angiogenesis, the formation of new blood vessels from pre-existing ones, occurs in pathophysiological contexts such as wound healing, cancer, and chronic inflammatory disease. During sprouting angiogenesis, endothelial tip and stalk cells coordinately remodel their cell-cell junctions to allow collective migration and extension of the sprout while maintaining barrier integrity. All these processes require energy, and the predominant ATP generation route in endothelial cells is glycolysis. However, it remains unclear how ATP reaches the plasma membrane and intercellular junctions. In this study, we demonstrate that the glycolytic enzyme pyruvate kinase 2 (PKM2) is required for sprouting angiogenesis in vitro and in vivo through the regulation of endothelial cell-junction dynamics and collective migration. We show that PKM2-silencing decreases ATP required for proper VE-cadherin internalization/traffic at endothelial cell-cell junctions. Our study provides fresh insight into the role of ATP subcellular compartmentalization in endothelial cells during angiogenesis. Since manipulation of EC glycolysis constitutes a potential therapeutic intervention route, particularly in tumors and chronic inflammatory disease, these findings may help to refine the targeting of endothelial glycolytic activity in disease.


Subject(s)
Adenosine Triphosphate/biosynthesis , Carrier Proteins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Intercellular Junctions/metabolism , Membrane Proteins/metabolism , Neovascularization, Physiologic , Pyruvate Kinase/metabolism , Thyroid Hormones/metabolism , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Cell Movement , Endocytosis , Gene Silencing , Humans , Mice, Inbred C57BL , Pseudopodia/metabolism , Retina/metabolism , Thyroid Hormone-Binding Proteins
9.
Cell Rep ; 27(11): 3139-3151.e5, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31189101

ABSTRACT

Angiogenesis is driven by the coordinated collective branching of specialized leading "tip" and trailing "stalk" endothelial cells (ECs). While Notch-regulated negative feedback suppresses excessive tip selection, roles for positive feedback in EC identity decisions remain unexplored. Here, by integrating computational modeling with in vivo experimentation, we reveal that positive feedback critically modulates the magnitude, timing, and robustness of angiogenic responses. In silico modeling predicts that positive-feedback-mediated amplification of VEGF signaling generates an ultrasensitive bistable switch that underpins quick and robust tip-stalk decisions. In agreement, we define a positive-feedback loop exhibiting these properties in vivo, whereby Vegf-induced expression of the atypical tetraspanin, tm4sf18, amplifies Vegf signaling to dictate the speed and robustness of EC selection for angiogenesis. Consequently, tm4sf18 mutant zebrafish select fewer motile ECs and exhibit stunted hypocellular vessels with unstable tip identity that is severely perturbed by even subtle Vegfr attenuation. Hence, positive feedback spatiotemporally shapes the angiogenic switch to ultimately modulate vascular network topology.


Subject(s)
Feedback, Physiological , Neovascularization, Physiologic , Animals , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Receptors, Notch/metabolism , Tetraspanins/genetics , Tetraspanins/metabolism , Vascular Endothelial Growth Factor A/metabolism , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
10.
Proc Natl Acad Sci U S A ; 116(10): 4538-4547, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30787185

ABSTRACT

Diabetic macular edema is a major complication of diabetes resulting in loss of central vision. Although heightened vessel leakiness has been linked to glial and neuronal-derived factors, relatively little is known on the mechanisms by which mature endothelial cells exit from a quiescent state and compromise barrier function. Here we report that endothelial NOTCH1 signaling in mature diabetic retinas contributes to increased vascular permeability. By providing both human and mouse data, we show that NOTCH1 ligands JAGGED1 and DELTA LIKE-4 are up-regulated secondary to hyperglycemia and activate both canonical and rapid noncanonical NOTCH1 pathways that ultimately disrupt endothelial adherens junctions in diabetic retinas by causing dissociation of vascular endothelial-cadherin from ß-catenin. We further demonstrate that neutralization of NOTCH1 ligands prevents diabetes-induced retinal edema. Collectively, these results identify a fundamental process in diabetes-mediated vascular permeability and provide translational rational for targeting the NOTCH pathway (primarily JAGGED1) in conditions characterized by compromised vascular barrier function.


Subject(s)
Capillary Permeability , Diabetic Retinopathy/pathology , Receptor, Notch1/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Calcium-Binding Proteins/biosynthesis , Enzyme Activation , Hyperglycemia/metabolism , Jagged-1 Protein/biosynthesis , Mice , Nitric Oxide/biosynthesis , Retinal Vessels/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , src-Family Kinases/metabolism
11.
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
12.
J Am Acad Audiol ; 29(5): 378-388, 2018 05.
Article in English | MEDLINE | ID: mdl-29708488

ABSTRACT

BACKGROUND: There is a significant lack of evidence guiding our understanding of the needs of families of children who are deaf/hard of hearing (Deaf/HH) with an autism spectrum disorder (ASD). Much of our current knowledge is founded in case report studies with very small numbers of children with the dual diagnosis. PURPOSE: The purpose of this study was to gain an understanding of the factors relating to caregiver stress and needs (i.e., supports and interventions) in families of children who are Deaf/HH with ASD. RESEARCH DESIGN: Comparison groups of families of children who were Deaf/HH, families with a hearing child with ASD, and families of children who were Deaf/HH with ASD were administered standardized questionnaires of stress with brief qualitative questionnaires focusing on family-identified needs. STUDY SAMPLE: Six families of children with the dual diagnosis, four families of children who were Deaf/HH, and three families of children with ASD. DATA COLLECTION AND ANALYSIS: Surveys included demographic and support questionnaires, the Parenting Stress Index (PSI), the Pediatric Hearing Impairment Caregiver Experience, and a qualitative questionnaire. RESULTS: Families of children who were Deaf/HH with ASD had a higher median total stress score on the PSI as compared to families of children who were Deaf/HH only (58.5 versus 41.5, respectively; p = 0.02) and higher Child Domain scores (60 versus 43, respectively; p = 0.02), indicating higher levels of stress in families of children with the dual diagnosis. The families of children who were Deaf/HH with ASD reported similar levels of stress as families of children with ASD. CONCLUSIONS: Families of children who are Deaf/HH with an ASD experience stress and describe similar needs and priorities as families of hearing children with ASD. This suggests the needs related to having an autism spectrum disorder are of high priority in families of children with the dual diagnosis.


Subject(s)
Autism Spectrum Disorder/complications , Caregivers , Deafness/complications , Health Services Needs and Demand , Parents , Stress, Psychological/epidemiology , Adolescent , Caregivers/psychology , Child , Cost of Illness , Female , Humans , Male , Parents/psychology , Self Report , Young Adult
13.
Philos Trans R Soc Lond B Biol Sci ; 372(1720)2017 May 19.
Article in English | MEDLINE | ID: mdl-28348255

ABSTRACT

The process of new blood vessel growth (angiogenesis) is highly dynamic, involving complex coordination of multiple cell types. Though the process must carefully unfold over time to generate functional, well-adapted branching networks, we seldom hear about the time-based properties of angiogenesis, despite timing being central to other areas of biology. Here, we present a novel, time-based formulation of endothelial cell behaviour during angiogenesis and discuss a flurry of our recent, integrated in silico/in vivo studies, put in context to the wider literature, which demonstrate that tissue conditions can locally adapt the timing of collective cell behaviours/decisions to grow different vascular network architectures. A growing array of seemingly unrelated 'temporal regulators' have recently been uncovered, including tissue derived factors (e.g. semaphorins or the high levels of VEGF found in cancer) and cellular processes (e.g. asymmetric cell division or filopodia extension) that act to alter the speed of cellular decisions to migrate. We will argue that 'temporal adaptation' provides a novel account of organ/disease-specific vascular morphology and reveals 'timing' as a new target for therapeutics. We therefore propose and explain a conceptual shift towards a 'temporal adaptation' perspective in vascular biology, and indeed other areas of biology where timing remains elusive.This article is part of the themed issue 'Systems morphodynamics: understanding the development of tissue hardware'.


Subject(s)
Endothelial Cells/physiology , Neovascularization, Physiologic , Animals , Humans
14.
PLoS One ; 11(11): e0166489, 2016.
Article in English | MEDLINE | ID: mdl-27846305

ABSTRACT

Angiogenesis is a highly dynamic morphogenesis process; however, surprisingly little is known about the timing of the different molecular processes involved. Although the role of the VEGF-notch-DLL4 signaling pathway has been established as essential for tip/stalk cell competition during sprouting, the speed and dynamic properties of the underlying process at the individual cell level has not been fully elucidated. In this study, using mathematical modeling we investigate how specific, biologically meaningful, local conditions around and within an individual cell can influence their unique tip/stalk phenotype switching kinetics. To this end we constructed an ordinary differential equation model of VEGF-notch-DLL4 signaling in a system of two, coupled endothelial cells (EC). Our studies reveal that at any given point in an angiogenic vessel the time it takes a cell to decide to take on a tip or stalk phenotype may be drastically different, and this asynchrony of tip/stalk cell decisions along vessels itself acts to speed up later competitions. We unexpectedly uncover intermediate "partial" yet stable states lying between the tip and stalk cell fates, and identify that internal cellular factors, such as NAD-dependent deacetylase sirtuin-1 (Sirt1) and Lunatic fringe 1 (Lfng1), can specifically determine the length of time a cell spends in these newly identified partial tip/stalk states. Importantly, the model predicts that these partial EC states can arise during normal angiogenesis, in particular during cell rearrangement in sprouts, providing a novel two-stage mechanism for rapid adaptive behavior to the cells highly dynamic environment. Overall, this study demonstrates that different factors (both internal and external to EC) can be used to modulate the speed of tip/stalk decisions, opening up new opportunities and challenges for future biological experiments and therapeutic targeting to manipulate vascular network topology, and our basic understanding of developmental/pathological angiogenesis.


Subject(s)
Endothelial Cells/cytology , Feedback, Physiological , Gene Expression Regulation , Models, Biological , Neovascularization, Physiologic/genetics , Animals , Computer Simulation , Endothelial Cells/metabolism , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kinetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phenotype , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Sirtuin 1/genetics , Sirtuin 1/metabolism , Time Factors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
15.
Nat Cell Biol ; 18(12): 1292-1301, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27870831

ABSTRACT

The asymmetric division of stem or progenitor cells generates daughters with distinct fates and regulates cell diversity during tissue morphogenesis. However, roles for asymmetric division in other more dynamic morphogenetic processes, such as cell migration, have not previously been described. Here we combine zebrafish in vivo experimental and computational approaches to reveal that heterogeneity introduced by asymmetric division generates multicellular polarity that drives coordinated collective cell migration in angiogenesis. We find that asymmetric positioning of the mitotic spindle during endothelial tip cell division generates daughters of distinct size with discrete 'tip' or 'stalk' thresholds of pro-migratory Vegfr signalling. Consequently, post-mitotic Vegfr asymmetry drives Dll4/Notch-independent self-organization of daughters into leading tip or trailing stalk cells, and disruption of asymmetry randomizes daughter tip/stalk selection. Thus, asymmetric division seamlessly integrates cell proliferation with collective migration, and, as such, may facilitate growth of other collectively migrating tissues during development, regeneration and cancer invasion.


Subject(s)
Asymmetric Cell Division , Cell Movement , Neovascularization, Physiologic , Animals , Cell Polarity , Cell Size , Computer Simulation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Green Fluorescent Proteins/metabolism , Mitosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Notch , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction , Time-Lapse Imaging , Zebrafish , Zebrafish Proteins/metabolism
16.
Sci Signal ; 9(437): ra72, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27436360

ABSTRACT

Activation of vascular endothelial growth factor (VEGF) receptor 2 (VEGFR2) by VEGF binding is critical for vascular morphogenesis. In addition, VEGF disrupts the endothelial barrier by triggering the phosphorylation and turnover of the junctional molecule VE-cadherin, a process mediated by the VEGFR2 downstream effectors T cell-specific adaptor (TSAd) and the tyrosine kinase c-Src. We investigated whether the VEGFR2-TSAd-c-Src pathway was required for angiogenic sprouting. Indeed, Tsad-deficient embryoid bodies failed to sprout in response to VEGF. Tsad-deficient mice displayed impaired angiogenesis specifically during tracheal vessel development, but not during retinal vasculogenesis, and in VEGF-loaded Matrigel plugs, but not in those loaded with FGF. The SH2 and proline-rich domains of TSAd bridged VEGFR2 and c-Src, and this bridging was critical for the localization of activated c-Src to endothelial junctions and elongation of the growing sprout, but not for selection of the tip cell. These results revealed that vascular sprouting and permeability are both controlled through the VEGFR2-TSAd-c-Src signaling pathway in a subset of tissues, which may be useful in developing strategies to control tissue-specific pathological angiogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Endothelial Cells/metabolism , Neovascularization, Pathologic/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism , src-Family Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , CSK Tyrosine-Protein Kinase , Cell Line , Endothelial Cells/pathology , Mice , Mice, Knockout , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , src-Family Kinases/genetics
17.
Nat Commun ; 7: 12240, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27436424

ABSTRACT

During vessel sprouting, endothelial cells (ECs) dynamically rearrange positions in the sprout to compete for the tip position. We recently identified a key role for the glycolytic activator PFKFB3 in vessel sprouting by regulating cytoskeleton remodelling, migration and tip cell competitiveness. It is, however, unknown how glycolysis regulates EC rearrangement during vessel sprouting. Here we report that computational simulations, validated by experimentation, predict that glycolytic production of ATP drives EC rearrangement by promoting filopodia formation and reducing intercellular adhesion. Notably, the simulations correctly predicted that blocking PFKFB3 normalizes the disturbed EC rearrangement in high VEGF conditions, as occurs during pathological angiogenesis. This interdisciplinary study integrates EC metabolism in vessel sprouting, yielding mechanistic insight in the control of vessel sprouting by glycolysis, and suggesting anti-glycolytic therapy for vessel normalization in cancer and non-malignant diseases.


Subject(s)
Glycolysis , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic , Adenosine Triphosphate/metabolism , Antigens, CD/metabolism , Cadherins/antagonists & inhibitors , Cadherins/metabolism , Computer Simulation , Gene Knockdown Techniques , Glycolysis/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Indoles/pharmacology , Models, Biological , Neovascularization, Physiologic/drug effects , Phosphofructokinase-2/antagonists & inhibitors , Phosphofructokinase-2/metabolism , Pseudopodia/drug effects , Pseudopodia/metabolism , Pyridines/pharmacology , Pyrroles/pharmacology , Vascular Endothelial Growth Factor A/metabolism
18.
Elife ; 52016 04 13.
Article in English | MEDLINE | ID: mdl-27074663

ABSTRACT

Formation of a regularly branched blood vessel network is crucial in development and physiology. Here we show that the expression of the Notch ligand Dll4 fluctuates in individual endothelial cells within sprouting vessels in the mouse retina in vivo and in correlation with dynamic cell movement in mouse embryonic stem cell-derived sprouting assays. We also find that sprout elongation and branching associates with a highly differential phase pattern of Dll4 between endothelial cells. Stimulation with pathologically high levels of Vegf, or overexpression of Dll4, leads to Notch dependent synchronization of Dll4 fluctuations within clusters, both in vitro and in vivo. Our results demonstrate that the Vegf-Dll4/Notch feedback system normally operates to generate heterogeneity between endothelial cells driving branching, whilst synchronization drives vessel expansion. We propose that this sensitive phase transition in the behaviour of the Vegf-Dll4/Notch feedback loop underlies the morphogen function of Vegfa in vascular patterning.


Subject(s)
Brain Neoplasms/genetics , Endothelial Cells/metabolism , Glioblastoma/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neovascularization, Pathologic/genetics , Receptors, Notch/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adaptor Proteins, Signal Transducing , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Calcium-Binding Proteins , Cell Movement/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Feedback, Physiological , Gene Expression Regulation , Genes, Reporter , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , Neoplasm Transplantation , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Notch/genetics , Retina/cytology , Retina/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/pharmacology
19.
Nat Commun ; 7: 11017, 2016 Mar 23.
Article in English | MEDLINE | ID: mdl-27005951

ABSTRACT

The specific role of VEGFA-induced permeability and vascular leakage in physiology and pathology has remained unclear. Here we show that VEGFA-induced vascular leakage depends on signalling initiated via the VEGFR2 phosphosite Y949, regulating dynamic c-Src and VE-cadherin phosphorylation. Abolished Y949 signalling in the mouse mutant Vegfr2(Y949F/Y949F) leads to VEGFA-resistant endothelial adherens junctions and a block in molecular extravasation. Vessels in Vegfr2(Y949F/Y949F) mice remain sensitive to inflammatory cytokines, and vascular morphology, blood pressure and flow parameters are normal. Tumour-bearing Vegfr2(Y949F/Y949F) mice display reduced vascular leakage and oedema, improved response to chemotherapy and, importantly, reduced metastatic spread. The inflammatory infiltration in the tumour micro-environment is unaffected. Blocking VEGFA-induced disassembly of endothelial junctions, thereby suppressing tumour oedema and metastatic spread, may be preferable to full vascular suppression in the treatment of certain cancer forms.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Capillary Permeability/genetics , Endothelial Cells/metabolism , Glioma/pathology , Melanoma, Experimental/pathology , Neoplasm Metastasis/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Adherens Junctions , Animals , Edema , Endothelium, Vascular/metabolism , Mice , Microspheres , Mutation , Neoplasm Transplantation , Phosphorylation/genetics , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism
20.
Elife ; 52016 Feb 24.
Article in English | MEDLINE | ID: mdl-26910011

ABSTRACT

Vascular network density determines the amount of oxygen and nutrients delivered to host tissues, but how the vast diversity of densities is generated is unknown. Reiterations of endothelial-tip-cell selection, sprout extension and anastomosis are the basis for vascular network generation, a process governed by the VEGF/Notch feedback loop. Here, we find that temporal regulation of this feedback loop, a previously unexplored dimension, is the key mechanism to determine vascular density. Iterating between computational modeling and in vivo live imaging, we demonstrate that the rate of tip-cell selection determines the length of linear sprout extension at the expense of branching, dictating network density. We provide the first example of a host tissue-derived signal (Semaphorin3E-Plexin-D1) that accelerates tip cell selection rate, yielding a dense network. We propose that temporal regulation of this critical, iterative aspect of network formation could be a general mechanism, and additional temporal regulators may exist to sculpt vascular topology.


Subject(s)
Cell Proliferation , Endothelial Cells/physiology , Neovascularization, Physiologic , Receptors, Notch/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Computer Simulation , Mice, Inbred C57BL , Mice, Knockout , Optical Imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...