1
|
Lee JJ, Ng KY, Bakhtiar A. Extracellular matrix: unlocking new avenues in cancer treatment. Biomark Res 2025; 13:78. [PMID: 40426238 PMCID: PMC12117852 DOI: 10.1186/s40364-025-00757-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/05/2025] [Indexed: 05/29/2025] Open
Abstract
The extracellular matrix (ECM) plays a critical role in cancer progression by influencing tumor growth, invasion, and metastasis. This review explores the emerging therapeutic strategies that target the ECM as a novel approach in cancer treatment. By disrupting the structural and biochemical interactions within the tumor microenvironment, ECM-targeted therapies aim to inhibit cancer progression and overcome therapeutic resistance. We examine the current state of ECM research, focusing on key components such as collagen, laminin, fibronectin, periostin, and hyaluronic acid, and their roles in tumor biology. Additionally, we discuss the challenges associated with ECM-targeted therapies, including drug delivery, specificity, and potential side effects, while highlighting recent advancements and future directions. This review underscores the potential of ECM-focused strategies to enhance the efficacy of existing treatments and contribute to more effective cancer therapies.
Collapse
Affiliation(s)
- Jia Jing Lee
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| | - Athirah Bakhtiar
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia.
| |
Collapse
|
2
|
Payne S, Neal A, De Val S. Transcription factors regulating vasculogenesis and angiogenesis. Dev Dyn 2024; 253:28-58. [PMID: 36795082 PMCID: PMC10952167 DOI: 10.1002/dvdy.575] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/06/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Transcription factors (TFs) play a crucial role in regulating the dynamic and precise patterns of gene expression required for the initial specification of endothelial cells (ECs), and during endothelial growth and differentiation. While sharing many core features, ECs can be highly heterogeneous. Differential gene expression between ECs is essential to pattern the hierarchical vascular network into arteries, veins and capillaries, to drive angiogenic growth of new vessels, and to direct specialization in response to local signals. Unlike many other cell types, ECs have no single master regulator, instead relying on differing combinations of a necessarily limited repertoire of TFs to achieve tight spatial and temporal activation and repression of gene expression. Here, we will discuss the cohort of TFs known to be involved in directing gene expression during different stages of mammalian vasculogenesis and angiogenesis, with a primary focus on development.
Collapse
Affiliation(s)
- Sophie Payne
- Department of Physiology, Anatomy and GeneticsInstitute of Developmental and Regenerative Medicine, University of OxfordOxfordUK
| | - Alice Neal
- Department of Physiology, Anatomy and GeneticsInstitute of Developmental and Regenerative Medicine, University of OxfordOxfordUK
| | - Sarah De Val
- Department of Physiology, Anatomy and GeneticsInstitute of Developmental and Regenerative Medicine, University of OxfordOxfordUK
| |
Collapse
|
3
|
Shao M, Zhong MC, Wang Z, Ke Z, Zhong Z, Zhou J. Non-Invasive Dynamic Reperfusion of Microvessels In Vivo Controlled by Optical Tweezers. Front Bioeng Biotechnol 2022; 10:952537. [PMID: 35910027 PMCID: PMC9331193 DOI: 10.3389/fbioe.2022.952537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Distributive shock is considered to be a condition of microvascular hypoperfusion, which can be fatal in severe cases. However, traditional therapeutic methods to restore the macro blood flow are difficult to accurately control the blood perfusion of microvessels, and the currently developed manipulation techniques are inevitably incompatible with biological systems. In our approach, infrared optical tweezers are used to dynamically control the microvascular reperfusion within subdermal capillaries in the pinna of mice. Furthermore, we estimate the effect of different optical trap positions on reperfusion at branch and investigate the effect of the laser power on reperfusion. The results demonstrate the ability of optical tweezers to control microvascular reperfusion. This strategy allows near-noninvasive reperfusion of the microvascular hypoperfusion in vivo. Hence, our work is expected to provide unprecedented insights into the treatment of distributive shock.
Collapse
Affiliation(s)
- Meng Shao
- Anhui Province Key Laboratory of Measuring Theory and Precision Instrument, School of Instrument Science and Optoelectronics Engineering, Hefei University of Technology, Hefei, China
| | - Min-Cheng Zhong
- Anhui Province Key Laboratory of Measuring Theory and Precision Instrument, School of Instrument Science and Optoelectronics Engineering, Hefei University of Technology, Hefei, China
- *Correspondence: Min-Cheng Zhong, ; Jinhua Zhou,
| | - Zixin Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei, China
| | - Zeyu Ke
- School of Biomedical Engineering, Anhui Medical University, Hefei, China
| | - Zhensheng Zhong
- School of Biomedical Engineering, Anhui Medical University, Hefei, China
| | - Jinhua Zhou
- School of Biomedical Engineering, Anhui Medical University, Hefei, China
- *Correspondence: Min-Cheng Zhong, ; Jinhua Zhou,
| |
Collapse
|
4
|
Lin PK, Salvador J, Xie J, Aguera KN, Koller GM, Kemp SS, Griffin CT, Davis GE. Selective and Marked Blockade of Endothelial Sprouting Behavior Using Paclitaxel and Related Pharmacologic Agents. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:2245-2264. [PMID: 34563512 DOI: 10.1016/j.ajpath.2021.08.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/10/2021] [Accepted: 08/26/2021] [Indexed: 12/11/2022]
Abstract
Whether alterations in the microtubule cytoskeleton affect the ability of endothelial cells (ECs) to sprout and form branching networks of tubes was investigated in this study. Bioassays of human EC tubulogenesis, where both sprouting behavior and lumen formation can be rigorously evaluated, were used to demonstrate that addition of the microtubule-stabilizing drugs, paclitaxel, docetaxel, ixabepilone, and epothilone B, completely interferes with EC tip cells and sprouting behavior, while allowing for EC lumen formation. In bioassays mimicking vasculogenesis using single or aggregated ECs, these drugs induce ring-like lumens from single cells or cyst-like spherical lumens from multicellular aggregates with no evidence of EC sprouting behavior. Remarkably, treatment of these cultures with a low dose of the microtubule-destabilizing drug, vinblastine, led to an identical result, with complete blockade of EC sprouting, but allowing for EC lumen formation. Administration of paclitaxel in vivo markedly interfered with angiogenic sprouting behavior in developing mouse retina, providing corroboration. These findings reveal novel biological activities for pharmacologic agents that are widely utilized in multidrug chemotherapeutic regimens for the treatment of human malignant cancers. Overall, this work demonstrates that manipulation of microtubule stability selectively interferes with the ability of ECs to sprout, a necessary step to initiate and form branched capillary tube networks.
Collapse
Affiliation(s)
- Prisca K Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Jocelynda Salvador
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Jun Xie
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kalia N Aguera
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Gretchen M Koller
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Scott S Kemp
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Courtney T Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida.
| |
Collapse
|
5
|
Helker CS, Eberlein J, Wilhelm K, Sugino T, Malchow J, Schuermann A, Baumeister S, Kwon HB, Maischein HM, Potente M, Herzog W, Stainier DY. Apelin signaling drives vascular endothelial cells toward a pro-angiogenic state. eLife 2020; 9:55589. [PMID: 32955436 PMCID: PMC7567607 DOI: 10.7554/elife.55589] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 09/19/2020] [Indexed: 12/18/2022] Open
Abstract
To form new blood vessels (angiogenesis), endothelial cells (ECs) must be activated and acquire highly migratory and proliferative phenotypes. However, the molecular mechanisms that govern these processes are incompletely understood. Here, we show that Apelin signaling functions to drive ECs into such an angiogenic state. Zebrafish lacking Apelin signaling exhibit defects in endothelial tip cell morphology and sprouting. Using transplantation experiments, we find that in mosaic vessels, wild-type ECs leave the dorsal aorta (DA) and form new vessels while neighboring ECs defective in Apelin signaling remain in the DA. Mechanistically, Apelin signaling enhances glycolytic activity in ECs at least in part by increasing levels of the growth-promoting transcription factor c-Myc. Moreover, APELIN expression is regulated by Notch signaling in human ECs, and its function is required for the hypersprouting phenotype in Delta-like 4 (Dll4) knockdown zebrafish embryos. These data provide new insights into fundamental principles of blood vessel formation and Apelin signaling, enabling a better understanding of vascular growth in health and disease.
Collapse
Affiliation(s)
- Christian Sm Helker
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Philipps-University Marburg, Faculty of Biology, Cell Signaling and Dynamics, Marburg, Germany
| | - Jean Eberlein
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Philipps-University Marburg, Faculty of Biology, Cell Signaling and Dynamics, Marburg, Germany
| | - Kerstin Wilhelm
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Toshiya Sugino
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Julian Malchow
- Philipps-University Marburg, Faculty of Biology, Cell Signaling and Dynamics, Marburg, Germany
| | | | - Stefan Baumeister
- Philipps-University Marburg, Faculty of Biology, Cell Signaling and Dynamics, Marburg, Germany
| | - Hyouk-Bum Kwon
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Hans-Martin Maischein
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Michael Potente
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,DZHK (German Center for Cardiovascular Research), partner site Frankfurt Rhine-Main, Berlin, Germany
| | - Wiebke Herzog
- University of Muenster, Muenster, Germany.,Max Planck Institute for Molecular Biomedicine, Muenster, Germany
| | - Didier Yr Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,DZHK (German Center for Cardiovascular Research), partner site Frankfurt Rhine-Main, Berlin, Germany
| |
Collapse
|
6
|
Lu J, Liu R, Miao A, Chen X, Xiao W, Wang Y, Cao D, Pan J, Li L, Luo Y. The role of cldnh during the early retinal development in zebrafish. Exp Eye Res 2020; 200:108207. [PMID: 32866532 DOI: 10.1016/j.exer.2020.108207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/08/2020] [Accepted: 08/26/2020] [Indexed: 10/23/2022]
Abstract
Claudin-3, an integral component of tight junction, has recently been shown to be expressed in retinal ganglion cells, retinal pigment cells, and retinal vascular endothelial cells. However, the role of claudin-3 in the development of the neural retina and its vessels remains undefined. This study aimed to investigate the role of zebrafish claudin-h (cldnh), the closest ortholog of mouse and human claudin-3, in the development of the neural retina and its vessels. Cldnh levels in green fluorescent protein transgenic zebrafish were genetically manipulated by cldnh morpholino oligonucleotide (MO) and cldnh mRNA to investigate gene function. The expression of cldnh was analyzed using polymerase chain reaction and immunofluorescence staining. The altered morphological, cellular and molecular events in the cldnh MO-morphant eyes were detected using hematoxylin-eosin staining, fluorescent dye injection, confocal in vivo imaging, BrdU labeling, TUNEL assay, RNA sequencing, and Western blot. We demonstrated that the cldnh protein was expressed in the neural retina and the hyaloid vessel which is the predecessor of the retinal vessel in zebrafish. Cldnh knockdown delayed lamination of the neural retina and reduced its thickness, which might be associated with the downregulation of the retinal development-related genes of atoh7, pcdh17, crx, neurod1, insm1a, sox9b and cdh11, and the upregulation of the cell cycle and apoptosis-associated genes of tp53, cdkn1a and casp8. Cldnh knockdown also reduced the density and interrupted the lumenization of the hyaloid vessels, which might be owing to the downregulation of the vessel formation-related genes of hlx1 and myl7. In conclusion, cldnh was required for the normal development of the neural retina and its vessels in zebrafish, providing a basis for elucidating its role in the pathogenesis of retinal vascular or inflammatory diseases.
Collapse
Affiliation(s)
- Jing Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Ruyuan Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Aiwen Miao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xiaoyun Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Wei Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Yishen Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Di Cao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Jianying Pan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Lisha Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Yan Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
7
|
O'Brown NM, Megason SG, Gu C. Suppression of transcytosis regulates zebrafish blood-brain barrier function. eLife 2019; 8:e47326. [PMID: 31429822 PMCID: PMC6726461 DOI: 10.7554/elife.47326] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 08/19/2019] [Indexed: 12/15/2022] Open
Abstract
As an optically transparent model organism with an endothelial blood-brain barrier (BBB), zebrafish offer a powerful tool to study the vertebrate BBB. However, the precise developmental profile of functional zebrafish BBB acquisition and the subcellular and molecular mechanisms governing the zebrafish BBB remain poorly characterized. Here, we capture the dynamics of developmental BBB leakage using live imaging, revealing a combination of steady accumulation in the parenchyma and sporadic bursts of tracer leakage. Electron microscopy studies further reveal high levels of transcytosis in brain endothelium early in development that are suppressed later. The timing of this suppression of transcytosis coincides with the establishment of BBB function. Finally, we demonstrate a key mammalian BBB regulator Mfsd2a, which inhibits transcytosis, plays a conserved role in zebrafish, as mfsd2aa mutants display increased BBB permeability due to increased transcytosis. Our findings indicate a conserved developmental program of barrier acquisition between zebrafish and mice.
Collapse
Affiliation(s)
| | - Sean G Megason
- Department of Systems BiologyHarvard Medical SchoolBostonUnited States
| | - Chenghua Gu
- Department of NeurobiologyHarvard Medical SchoolBostonUnited States
| |
Collapse
|
8
|
Page DJ, Thuret R, Venkatraman L, Takahashi T, Bentley K, Herbert SP. Positive Feedback Defines the Timing, Magnitude, and Robustness of Angiogenesis. Cell Rep 2019; 27:3139-3151.e5. [PMID: 31189101 PMCID: PMC6581738 DOI: 10.1016/j.celrep.2019.05.052] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 04/01/2019] [Accepted: 05/15/2019] [Indexed: 12/20/2022] Open
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.
Collapse
Affiliation(s)
- Donna J Page
- Faculty of Biology, Medicine and Health, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK; School of Healthcare Science, Manchester Metropolitan University, Manchester M1 5GD, UK
| | - Raphael Thuret
- Faculty of Biology, Medicine and Health, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Lakshmi Venkatraman
- Biomedical Engineering Department, Boston University, 610 Commonwealth Avenue, Boston, MA 02215, USA; Immunology, Genetics and Pathology Department, University of Uppsala, 751 85 Uppsala, Sweden
| | - Tokiharu Takahashi
- Faculty of Biology, Medicine and Health, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Katie Bentley
- Biomedical Engineering Department, Boston University, 610 Commonwealth Avenue, Boston, MA 02215, USA; Immunology, Genetics and Pathology Department, University of Uppsala, 751 85 Uppsala, Sweden; Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Cellular Adaptive Behaviour Lab, The Francis Crick Institute, Midland Road, London NW1 1AT, UK; Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, Strand Campus, London WC2B 4BG, UK.
| | - Shane P Herbert
- Faculty of Biology, Medicine and Health, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK.
| |
Collapse
|
9
|
Tobia C, Chiodelli P, Barbieri A, Buraschi S, Ferrari E, Mitola S, Borsani G, Guerra J, Presta M. Atypical Chemokine Receptor 3 Generates Guidance Cues for CXCL12-Mediated Endothelial Cell Migration. Front Immunol 2019; 10:1092. [PMID: 31156639 PMCID: PMC6529557 DOI: 10.3389/fimmu.2019.01092] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/29/2019] [Indexed: 11/23/2022] Open
Abstract
Chemokine receptor CXCR4, its ligand stromal cell-derived factor-1 (CXCL12) and the decoy receptor atypical chemokine receptor 3 (ACKR3, also named CXCR7), are involved in the guidance of migrating cells in different anatomical districts. Here, we investigated the role of the ACKR3 zebrafish ortholog ackr3b in the vascularization process during embryonic development. Bioinformatics and functional analyses confirmed that ackr3b is a CXCL12-binding ortholog of human ACKR3. ackr3b is transcribed in the endoderm of zebrafish embryos during epiboly and is expressed in a wide range of tissues during somitogenesis, including central nervous system and somites. Between 18 somite and 26 h-post fertilization stages, the broad somitic expression of ackr3b becomes restricted to the basal part of the somites. After ackr3b knockdown, intersomitic vessels (ISVs) lose the correct direction of migration and are characterized by the presence of aberrant sprouts and ectopic filopodia protrusions, showing downregulation of the tip/stalk cell marker hlx1. In addition, ackr3b morphants show significant alterations of lateral dorsal aortae formation. In keeping with a role for ackr3b in endothelial cell guidance, CXCL12 gradient generated by ACKR3 expression in CHO cell transfectants guides human endothelial cell migration in an in vitro cell co-culture chemotaxis assay. Our results demonstrate that ackr3b plays a non-redundant role in the guidance of sprouting endothelial cells during vascular development in zebrafish. Moreover, ACKR3 scavenging activity generates guidance cues for the directional migration of CXCR4-expressing human endothelial cells in response to CXCL12.
Collapse
Affiliation(s)
- Chiara Tobia
- Unit of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Paola Chiodelli
- Unit of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Andrea Barbieri
- Unit of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Simone Buraschi
- Unit of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Elena Ferrari
- Unit of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Stefania Mitola
- Unit of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Giuseppe Borsani
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Jessica Guerra
- Unit of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Marco Presta
- Unit of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
10
|
Carretero-Ortega J, Chhangawala Z, Hunt S, Narvaez C, Menéndez-González J, Gay CM, Zygmunt T, Li X, Torres-Vázquez J. GIPC proteins negatively modulate Plexind1 signaling during vascular development. eLife 2019; 8:e30454. [PMID: 31050647 PMCID: PMC6499541 DOI: 10.7554/elife.30454] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 04/15/2019] [Indexed: 12/18/2022] Open
Abstract
Semaphorins (SEMAs) and their Plexin (PLXN) receptors are central regulators of metazoan cellular communication. SEMA-PLXND1 signaling plays important roles in cardiovascular, nervous, and immune system development, and cancer biology. However, little is known about the molecular mechanisms that modulate SEMA-PLXND1 signaling. As PLXND1 associates with GIPC family endocytic adaptors, we evaluated the requirement for the molecular determinants of their association and PLXND1's vascular role. Zebrafish that endogenously express a Plxnd1 receptor with a predicted impairment in GIPC binding exhibit low penetrance angiogenesis deficits and antiangiogenic drug hypersensitivity. Moreover, gipc mutant fish show angiogenic impairments that are ameliorated by reducing Plxnd1 signaling. Finally, GIPC depletion potentiates SEMA-PLXND1 signaling in cultured endothelial cells. These findings expand the vascular roles of GIPCs beyond those of the Vascular Endothelial Growth Factor (VEGF)-dependent, proangiogenic GIPC1-Neuropilin 1 complex, recasting GIPCs as negative modulators of antiangiogenic PLXND1 signaling and suggest that PLXND1 trafficking shapes vascular development.
Collapse
Affiliation(s)
- Jorge Carretero-Ortega
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Zinal Chhangawala
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Shane Hunt
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Carlos Narvaez
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Javier Menéndez-González
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Carl M Gay
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Tomasz Zygmunt
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| | - Xiaochun Li
- Department of Population HealthNew York University School of MedicineNew YorkUnited States
| | - Jesús Torres-Vázquez
- Department of Cell Biology, Skirball Institute of Biomolecular MedicineNew York University Langone Medical CenterNew YorkUnited States
| |
Collapse
|
11
|
A metabolic interplay coordinated by HLX regulates myeloid differentiation and AML through partly overlapping pathways. Nat Commun 2018; 9:3090. [PMID: 30082823 PMCID: PMC6078963 DOI: 10.1038/s41467-018-05311-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 06/29/2018] [Indexed: 02/07/2023] Open
Abstract
The H2.0-like homeobox transcription factor (HLX) regulates hematopoietic differentiation and is overexpressed in Acute Myeloid Leukemia (AML), but the mechanisms underlying these functions remain unclear. We demonstrate here that HLX overexpression leads to a myeloid differentiation block both in zebrafish and human hematopoietic stem and progenitor cells (HSPCs). We show that HLX overexpression leads to downregulation of genes encoding electron transport chain (ETC) components and upregulation of PPARδ gene expression in zebrafish and human HSPCs. HLX overexpression also results in AMPK activation. Pharmacological modulation of PPARδ signaling relieves the HLX-induced myeloid differentiation block and rescues HSPC loss upon HLX knockdown but it has no effect on AML cell lines. In contrast, AMPK inhibition results in reduced viability of AML cell lines, but minimally affects myeloid progenitors. This newly described role of HLX in regulating the metabolic state of hematopoietic cells may have important therapeutic implications. HLX transcription factor regulates haematopoietic stem and progenitor cell (HSPC) differentiation and is overexpressed in acute myeloid leukemia. Here the authors show that HLX overexpression leads to myeloid differentiation block in zebrafish and human HSPCs by direct regulation of metabolic pathways.
Collapse
|
12
|
Hogan BM, Schulte-Merker S. How to Plumb a Pisces: Understanding Vascular Development and Disease Using Zebrafish Embryos. Dev Cell 2017; 42:567-583. [PMID: 28950100 DOI: 10.1016/j.devcel.2017.08.015] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 08/01/2017] [Accepted: 08/21/2017] [Indexed: 01/09/2023]
Abstract
Our vasculature plays diverse and critical roles in homeostasis and disease. In recent decades, the use of zebrafish has driven our understanding of vascular development into new areas, identifying new genes and mechanisms controlling vessel formation and allowing unprecedented observation of the cellular and molecular events that shape the developing vasculature. Here, we highlight key mechanisms controlling formation of the zebrafish vasculature and investigate how knowledge from this highly tractable model system has informed our understanding of vascular disease in humans.
Collapse
Affiliation(s)
- Benjamin M Hogan
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Road, St Lucia, Brisbane, QLD 4072, Australia.
| | - Stefan Schulte-Merker
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Münster 48149, Germany; Cells-in-Motion Cluster of Excellence (EXC-1003), WWU Münster, 48149 Münster, Germany.
| |
Collapse
|
13
|
Fish JE, Cantu Gutierrez M, Dang LT, Khyzha N, Chen Z, Veitch S, Cheng HS, Khor M, Antounians L, Njock MS, Boudreau E, Herman AM, Rhyner AM, Ruiz OE, Eisenhoffer GT, Medina-Rivera A, Wilson MD, Wythe JD. Dynamic regulation of VEGF-inducible genes by an ERK/ERG/p300 transcriptional network. Development 2017; 144:2428-2444. [PMID: 28536097 DOI: 10.1242/dev.146050] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 05/15/2017] [Indexed: 12/20/2022]
Abstract
The transcriptional pathways activated downstream of vascular endothelial growth factor (VEGF) signaling during angiogenesis remain incompletely characterized. By assessing the signals responsible for induction of the Notch ligand delta-like 4 (DLL4) in endothelial cells, we find that activation of the MAPK/ERK pathway mirrors the rapid and dynamic induction of DLL4 transcription and that this pathway is required for DLL4 expression. Furthermore, VEGF/ERK signaling induces phosphorylation and activation of the ETS transcription factor ERG, a prerequisite for DLL4 induction. Transcription of DLL4 coincides with dynamic ERG-dependent recruitment of the transcriptional co-activator p300. Genome-wide gene expression profiling identified a network of VEGF-responsive and ERG-dependent genes, and ERG chromatin immunoprecipitation (ChIP)-seq revealed the presence of conserved ERG-bound putative enhancer elements near these target genes. Functional experiments performed in vitro and in vivo confirm that this network of genes requires ERK, ERG and p300 activity. Finally, genome-editing and transgenic approaches demonstrate that a highly conserved ERG-bound enhancer located upstream of HLX (which encodes a transcription factor implicated in sprouting angiogenesis) is required for its VEGF-mediated induction. Collectively, these findings elucidate a novel transcriptional pathway contributing to VEGF-dependent angiogenesis.
Collapse
Affiliation(s)
- Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, Canada .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada.,Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada
| | - Manuel Cantu Gutierrez
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.,Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lan T Dang
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada.,Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada
| | - Nadiya Khyzha
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada.,Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada
| | - Zhiqi Chen
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada.,Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada
| | - Shawn Veitch
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada.,Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada
| | - Henry S Cheng
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada.,Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada
| | - Melvin Khor
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada.,Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada
| | - Lina Antounians
- Genetics and Genome Biology, Hospital for Sick Children, Toronto M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto M5S 1A8, Canada
| | - Makon-Sébastien Njock
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada.,Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada
| | - Emilie Boudreau
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada.,Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada
| | - Alexander M Herman
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexander M Rhyner
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Oscar E Ruiz
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - George T Eisenhoffer
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Graduate School of Biomedical Sciences, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alejandra Medina-Rivera
- Genetics and Genome Biology, Hospital for Sick Children, Toronto M5G 0A4, Canada.,Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Querétaro 76230, México
| | - Michael D Wilson
- Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada.,Genetics and Genome Biology, Hospital for Sick Children, Toronto M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto M5S 1A8, Canada
| | - Joshua D Wythe
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA .,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.,Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
14
|
Asymmetric division coordinates collective cell migration in angiogenesis. Nat Cell Biol 2016; 18:1292-1301. [PMID: 27870831 DOI: 10.1038/ncb3443] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 10/21/2016] [Indexed: 12/18/2022]
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.
Collapse
|
15
|
Sacilotto N, Chouliaras KM, Nikitenko LL, Lu YW, Fritzsche M, Wallace MD, Nornes S, García-Moreno F, Payne S, Bridges E, Liu K, Biggs D, Ratnayaka I, Herbert SP, Molnár Z, Harris AL, Davies B, Bond GL, Bou-Gharios G, Schwarz JJ, De Val S. MEF2 transcription factors are key regulators of sprouting angiogenesis. Genes Dev 2016; 30:2297-2309. [PMID: 27898394 PMCID: PMC5110996 DOI: 10.1101/gad.290619.116] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 09/29/2016] [Indexed: 12/24/2022]
Abstract
Angiogenesis, the fundamental process by which new blood vessels form from existing ones, depends on precise spatial and temporal gene expression within specific compartments of the endothelium. However, the molecular links between proangiogenic signals and downstream gene expression remain unclear. During sprouting angiogenesis, the specification of endothelial cells into the tip cells that lead new blood vessel sprouts is coordinated by vascular endothelial growth factor A (VEGFA) and Delta-like ligand 4 (Dll4)/Notch signaling and requires high levels of Notch ligand DLL4. Here, we identify MEF2 transcription factors as crucial regulators of sprouting angiogenesis directly downstream from VEGFA. Through the characterization of a Dll4 enhancer directing expression to endothelial cells at the angiogenic front, we found that MEF2 factors directly transcriptionally activate the expression of Dll4 and many other key genes up-regulated during sprouting angiogenesis in both physiological and tumor vascularization. Unlike ETS-mediated regulation, MEF2-binding motifs are not ubiquitous to all endothelial gene enhancers and promoters but are instead overrepresented around genes associated with sprouting angiogenesis. MEF2 target gene activation is directly linked to VEGFA-induced release of repressive histone deacetylases and concurrent recruitment of the histone acetyltransferase EP300 to MEF2 target gene regulatory elements, thus establishing MEF2 factors as the transcriptional effectors of VEGFA signaling during angiogenesis.
Collapse
Affiliation(s)
- Natalia Sacilotto
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Kira M Chouliaras
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Leonid L Nikitenko
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Yao Wei Lu
- Center for Cardiovascular Sciences, Albany Medical College, Albany, New York 12208, USA
| | - Martin Fritzsche
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Marsha D Wallace
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Svanhild Nornes
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Fernando García-Moreno
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3QX, United Kingdom
| | - Sophie Payne
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Esther Bridges
- Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 7LJ, United Kingdom
| | - Ke Liu
- Institute of Aging and Chronic Disease, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Daniel Biggs
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Indrika Ratnayaka
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Shane P Herbert
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Zoltán Molnár
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3QX, United Kingdom
| | - Adrian L Harris
- Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 7LJ, United Kingdom
| | - Benjamin Davies
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Gareth L Bond
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - George Bou-Gharios
- Institute of Aging and Chronic Disease, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - John J Schwarz
- Center for Cardiovascular Sciences, Albany Medical College, Albany, New York 12208, USA
| | - Sarah De Val
- Ludwig Institute for Cancer Research Ltd., Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, United Kingdom
| |
Collapse
|
16
|
Mazurek R, Dave JM, Chandran RR, Misra A, Sheikh AQ, Greif DM. Vascular Cells in Blood Vessel Wall Development and Disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 78:323-350. [PMID: 28212800 DOI: 10.1016/bs.apha.2016.08.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The vessel wall is composed of distinct cellular layers, yet communication among individual cells within and between layers results in a dynamic and versatile structure. The morphogenesis of the normal vascular wall involves a highly regulated process of cell proliferation, migration, and differentiation. The use of modern developmental biological and genetic approaches has markedly enriched our understanding of the molecular and cellular mechanisms underlying these developmental events. Additionally, the application of similar approaches to study diverse vascular diseases has resulted in paradigm-shifting insights into pathogenesis. Further investigations into the biology of vascular cells in development and disease promise to have major ramifications on therapeutic strategies to combat pathologies of the vasculature.
Collapse
Affiliation(s)
- R Mazurek
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - J M Dave
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - R R Chandran
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - A Misra
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - A Q Sheikh
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - D M Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States.
| |
Collapse
|
17
|
Nagasawa-Masuda A, Terai K. ERK activation in endothelial cells is a novel marker during neovasculogenesis. Genes Cells 2016; 21:1164-1175. [PMID: 27696620 DOI: 10.1111/gtc.12438] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 08/16/2016] [Indexed: 01/01/2023]
Abstract
Vasculogenesis is essential during early development to construct networks transporting oxygen, blood and nutrients. Tip and stalk cells are specialized endothelial cells involved in novel vessel formation because of their behavior such as sprouting as a leading cell and following tip cell. However, the spatiotemporal details determining the emergence of these cells are unknown. Here, we first show that the ERK activity in endothelial cells represents the precursor of tip and stalk cells for vasculogenesis in zebrafish. We identified that tip and stalk cells for intersegmental vessel (ISV) formation were already specialized in the dorsal aorta (DA) before sprouting. Furthermore, similar specialization was observed in tip cells during parachordal vessel (PAV) formation in lymphangiogenesis. We also identified that the ERK activity was required for specialized cells to emerge from existing blood vessels. Our data show that the ERK activity is a novel marker for determining the emergence of cells in both angiogenesis and lymphangiogenesis.
Collapse
Affiliation(s)
- Ayumi Nagasawa-Masuda
- Laboratory of Function and Morphology, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Kenta Terai
- Laboratory of Function and Morphology, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo, 113-0032, Japan
| |
Collapse
|
18
|
Xu J, Vilanova G, Gomez H. A Mathematical Model Coupling Tumor Growth and Angiogenesis. PLoS One 2016; 11:e0149422. [PMID: 26891163 PMCID: PMC4758654 DOI: 10.1371/journal.pone.0149422] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 01/31/2016] [Indexed: 11/21/2022] Open
Abstract
We present a mathematical model for vascular tumor growth. We use phase fields to model cellular growth and reaction-diffusion equations for the dynamics of angiogenic factors and nutrients. The model naturally predicts the shift from avascular to vascular growth at realistic scales. Our computations indicate that the negative regulation of the Delta-like ligand 4 signaling pathway slows down tumor growth by producing a larger density of non-functional capillaries. Our results show good quantitative agreement with experiments.
Collapse
Affiliation(s)
- Jiangping Xu
- Applied Mathematics, University of A Coruña, A Coruña, Spain
| | | | - Hector Gomez
- Applied Mathematics, University of A Coruña, A Coruña, Spain
| |
Collapse
|
19
|
Liu T, Chen J, Xiao S, Lei X. H2.0-like homeobox 1 acts as a tumor suppressor in hepatocellular carcinoma. Tumour Biol 2015; 37:6419-28. [PMID: 26631039 DOI: 10.1007/s13277-015-4490-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 11/23/2015] [Indexed: 01/27/2023] Open
Abstract
H2.0-like homeobox 1 (HLX1) is a homeobox transcription factor gene expressed primarily in cytotrophoblast cell types in the early pregnancy human placenta and involved in the development of enteric nervous system. However, the biological function of HLX1 in hepatocellular carcinoma (HCC) remains unclear. In the present study, semiquantitative reverse transcription-polymerase chain reaction (RT-PCR), quantitative real-time RT-PCR, western blot, and immunohistochemical staining were used to examine the expression level of HLX1 in a total of 125 cases of HCC tissues and their matched adjacent nontumorous tissues (ANLTs), and its correlation with clinical features of HCC patients was analyzed. Our findings showed that the expression level of HLX1 was significantly reduced in HCCs compared to ANLTs. Besides, it was also remarkably downregulated in HCC cell lines compared to normal liver cell line. We further found that the HLX1 level was significantly associated with the tumor size (p = 0.016), tumor number (p = 0.004), vascular invasion (p = 0.031), Edmondson-Steiner grade (p = 0.041), tumor-node-metastasis (TNM) stage (p < 0.001), and Barcelona clinic liver cancer (BCLC) stage (p = 0.008). Moreover, HLX1 was an independent risk factor for overall survival (OS, p = 0.020) and disease-free survival (DFS, p = 0.024) of HCC patients. In vitro experiments showed that overexpression of HLX1 markedly suppressed the invasion, migration, proliferation, and colony formation of HCC cells; in contrast, downregulation of HLX1 significantly promoted the invasion, migration, proliferation, and colony formation of HCC cells. In vivo study indicated that overexpression of HLX1 significantly inhibited the tumorigenic capacity of HCC cells in nude mice. Based on these findings, we suggest that HLX1 acts as a tumor suppressor in HCC.
Collapse
Affiliation(s)
- Ting Liu
- Department of General Surgery, Second Xiangya Hospital, Central South University, Renmin Middle Road 139, Changsha, 410011, Hunan, China.
| | - Jing Chen
- Department of General Surgery, Peace Hospital Changzhi Medical College, Changzhi, 046000, Shanxi, China
| | - Shuai Xiao
- Department of General Surgery, Second Xiangya Hospital, Central South University, Renmin Middle Road 139, Changsha, 410011, Hunan, China
| | - Xiong Lei
- Department of Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| |
Collapse
|
20
|
Hernández-García R, Iruela-Arispe ML, Reyes-Cruz G, Vázquez-Prado J. Endothelial RhoGEFs: A systematic analysis of their expression profiles in VEGF-stimulated and tumor endothelial cells. Vascul Pharmacol 2015; 74:60-72. [PMID: 26471833 DOI: 10.1016/j.vph.2015.10.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 10/08/2015] [Accepted: 10/09/2015] [Indexed: 12/18/2022]
Abstract
Rho guanine nucleotide exchange factors (RhoGEFs) integrate cell signaling inputs into morphological and functional responses. However, little is known about the endothelial repertoire of RhoGEFs and their regulation. Thus, we assessed the expression of 81 RhoGEFs (70 homologous to Dbl and 11 of the DOCK family) in endothelial cells. Further, in the case of DH-RhoGEFs, we also determined their responses to VEGF exposure in vitro and in the context of tumors. A phylogenetic analysis revealed the existence of four groups of DH-RhoGEFs and two of the DOCK family. Among them, we found that the most abundant endothelial RhoGEFs were: Tuba, FGD5, Farp1, ARHGEF17, TRIO, P-Rex1, ARHGEF15, ARHGEF11, ABR, Farp2, ARHGEF40, ALS, DOCK1, DOCK7 and DOCK6. Expression of RASGRF2 and PREX2 increased significantly in response to VEGF, but most other RhoGEFs were unaffected. Interestingly murine endothelial cells isolated from tumors showed that all four phylogenetic subgroups of DH-RhoGEFs were altered when compared to non-tumor endothelial cells. In summary, our results provide a detailed assessment of RhoGEFs expression profiles in the endothelium and set the basis to systematically address their regulation in vascular signaling.
Collapse
Affiliation(s)
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell, and Developmental Biology and Molecular Biology Institute,University of California,Los Angeles, CA,USA
| | | | | |
Collapse
|
21
|
Scarlett K, Pattabiraman V, Barnett P, Liu D, Anderson LM. The proangiogenic effect of iroquois homeobox transcription factor Irx3 in human microvascular endothelial cells. J Biol Chem 2015; 290:6303-15. [PMID: 25512384 PMCID: PMC4358267 DOI: 10.1074/jbc.m114.601146] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 12/14/2014] [Indexed: 12/29/2022] Open
Abstract
Angiogenesis is a dynamic process required for embryonic development. However, postnatal vascular growth is characteristic of multiple disease states. Despite insights into the multistep process in which adhesion molecules, extracellular matrix proteins, growth factors, and their receptors work in concert to form new vessels from the preexisting vasculature, there remains a lack of insight of the nuclear transcriptional mechanisms that occur within endothelial cells (ECs) in response to VEGF. Iroquois homeobox gene 3 (Irx3) is a transcription factor of the Iroquois family of homeobox genes. Irx homeodomain transcription factors are involved in the patterning and development of several tissues. Irx3 is known for its role during embryogenesis in multiple organisms. However, the expression and function of Irx3 in human postnatal vasculature remains to be investigated. Here we show that Irx3 is expressed in human microvascular endothelial cells, and expression is elevated by VEGF stimulation. Genetic Irx3 gain and loss of function studies in human microvascular endothelial cells resulted in the modulation of EC migration during wound healing, chemotaxis and invasion, and tubulogenesis. Additionally, we observed increased delta-like ligand 4 (Dll4) expression, which suggests an increase in EC tip cell population. Finally, siRNA screening studies revealed that transient knockdown of Hey1, a downstream Notch signaling mediator, resulted in increased Irx3 expression in response to VEGF treatment. Strategies to pharmacologically regulate Irx3 function in adult endothelial cells may provide new therapies for angiogenesis.
Collapse
Affiliation(s)
| | | | - Petrina Barnett
- the Cancer Center for Therapeutic Development, Clark Atlanta University, Atlanta, Georgia 30314
| | - Dong Liu
- From the Cardiovascular Research Institute and Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia 30310 and
| | - Leonard M Anderson
- From the Cardiovascular Research Institute and Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia 30310 and
| |
Collapse
|
22
|
Tobia C, Gariano G, Guerra J, Presta M. Zebrafish embryo intersegmental vessels: a tool for investigating sprouting angiogenesis. Methods Mol Biol 2015; 1214:173-84. [PMID: 25468604 DOI: 10.1007/978-1-4939-1462-3_10] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Angiogenesis leads to the formation of the intersegmental vessels (ISVs) of the trunk in teleost zebrafish (Danio rerio) embryo. Here we describe experimental procedures, including in vivo observation of transgenic zebrafish embryo and whole mount in situ hybridization, to investigate ISV development in zebrafish embryos and assess the effect of antiangiogenic compounds on these vessels.
Collapse
Affiliation(s)
- Chiara Tobia
- Department of Molecular and Translational Medicine, Experimental Oncology and Immunology, School of Medicine, University of Brescia, Brescia, Italy
| | | | | | | |
Collapse
|
23
|
The H2.0-Like Homeobox Transcription Factor Modulates Yolk Sac Vascular Remodeling in Mouse Embryos. Arterioscler Thromb Vasc Biol 2014; 34:1468-76. [DOI: 10.1161/atvbaha.114.303626] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Objective—
The H2.0-like homeobox transcription factor (HLX) plays an essential role in visceral organogenesis in mice and has been shown to regulate angiogenic sprouting in vitro and in zebrafish embryos. We therefore examined the role of HLX in vascular development in mouse and avian embryos.
Approach and Results—
In situ hybridization showed that
Hlx
is expressed in a subset of sprouting blood vessels in postnatal mouse retinas and embryos.
Hlx
expression was conserved in quail embryos and upregulated in blood vessels at the onset of circulation. In vitro assays showed that
Hlx
is dynamically regulated by growth factors and shear stress alterations. Proangiogenic vascular endothelial growth factor induces
Hlx
expression in cultured endothelial cells, whereas signals that induce stalk cell identity lead to a reduction in
Hlx
expression. HLX was also downregulated in embryos in which flow was ablated, whereas injection of a starch solution, which increases blood viscosity and therefore shear stress, causes an upregulation in HLX. HLX knockdown in vitro resulted in a reduction in tip cell marker expression and in reduced angiogenic sprouting, but
Hlx
−/−
embryos showed no defect in vascular sprouting at E8.5, E9.5, or E11.5 in vivo. Vascular remodeling of the capillary plexus was altered in
Hlx
−/−
embryos, with a modestly enlarged venous plexus and reduction of the arterial plexus.
Conclusions—
Our findings indicate not only that
Hlx
regulates sprouting in vitro, but that its role in sprouting is nonessential in vivo. We find HLX is regulated by shear stress and a subtle defect in vascular remodeling is present in knockout embryos.
Collapse
|
24
|
Seidelmann SB, Lighthouse JK, Greif DM. Development and pathologies of the arterial wall. Cell Mol Life Sci 2014; 71:1977-99. [PMID: 24071897 PMCID: PMC11113178 DOI: 10.1007/s00018-013-1478-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 09/11/2013] [Accepted: 09/12/2013] [Indexed: 01/13/2023]
Abstract
Arteries consist of an inner single layer of endothelial cells surrounded by layers of smooth muscle and an outer adventitia. The majority of vascular developmental studies focus on the construction of endothelial networks through the process of angiogenesis. Although many devastating vascular diseases involve abnormalities in components of the smooth muscle and adventitia (i.e., the vascular wall), the morphogenesis of these layers has received relatively less attention. Here, we briefly review key elements underlying endothelial layer formation and then focus on vascular wall development, specifically on smooth muscle cell origins and differentiation, patterning of the vascular wall, and the role of extracellular matrix and adventitial progenitor cells. Finally, we discuss select human diseases characterized by marked vascular wall abnormalities. We propose that continuing to apply approaches from developmental biology to the study of vascular disease will stimulate important advancements in elucidating disease mechanism and devising novel therapeutic strategies.
Collapse
MESH Headings
- Angiogenic Proteins/genetics
- Angiogenic Proteins/metabolism
- Animals
- Arteries/growth & development
- Arteries/metabolism
- Arteries/pathology
- Cardiovascular Diseases/genetics
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/pathology
- Cell Differentiation
- Cell Lineage/genetics
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Endothelium, Vascular/growth & development
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Gene Expression Regulation, Developmental
- Humans
- Morphogenesis/genetics
- Muscle, Smooth, Vascular/growth & development
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neovascularization, Pathologic
- Neovascularization, Physiologic
Collapse
Affiliation(s)
- Sara B. Seidelmann
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, 300 George St., Rm 773J, New Haven, CT 06511 USA
| | - Janet K. Lighthouse
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, 300 George St., Rm 773J, New Haven, CT 06511 USA
| | - Daniel M. Greif
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, 300 George St., Rm 773J, New Haven, CT 06511 USA
| |
Collapse
|
25
|
Fang L, Liu C, Miller YI. Zebrafish models of dyslipidemia: relevance to atherosclerosis and angiogenesis. Transl Res 2014; 163:99-108. [PMID: 24095954 PMCID: PMC3946603 DOI: 10.1016/j.trsl.2013.09.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 09/07/2013] [Accepted: 09/10/2013] [Indexed: 01/07/2023]
Abstract
Lipid and lipoprotein metabolism in zebrafish and in humans are remarkably similar. Zebrafish express all major nuclear receptors, lipid transporters, apolipoproteins and enzymes involved in lipoprotein metabolism. Unlike mice, zebrafish express cetp and the Cetp activity is detected in zebrafish plasma. Feeding zebrafish a high cholesterol diet, without any genetic intervention, results in significant hypercholesterolemia and robust lipoprotein oxidation, making zebrafish an attractive animal model to study mechanisms relevant to early development of human atherosclerosis. These studies are facilitated by the optical transparency of zebrafish larvae and the availability of transgenic zebrafish expressing fluorescent proteins in endothelial cells and macrophages. Thus, vascular processes can be monitored in live animals. In this review article, we discuss recent advances in using dyslipidemic zebrafish in atherosclerosis-related studies. We also summarize recent work connecting lipid metabolism with regulation of angiogenesis, the work that considerably benefited from using the zebrafish model. These studies uncovered the role of aibp, abca1, abcg1, mtp, apoB, and apoC2 in regulation of angiogenesis in zebrafish and paved the way for future studies in mammals, which may suggest new therapeutic approaches to modulation of excessive or diminished angiogenesis contributing to the pathogenesis of human disease.
Collapse
Affiliation(s)
- Longhou Fang
- Department of Medicine, University of California, San Diego, La Jolla, Calif
| | - Chao Liu
- Department of Medicine, University of California, San Diego, La Jolla, Calif
| | - Yury I Miller
- Department of Medicine, University of California, San Diego, La Jolla, Calif.
| |
Collapse
|
26
|
Welti J, Loges S, Dimmeler S, Carmeliet P. Recent molecular discoveries in angiogenesis and antiangiogenic therapies in cancer. J Clin Invest 2013; 123:3190-200. [PMID: 23908119 DOI: 10.1172/jci70212] [Citation(s) in RCA: 456] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Four decades ago, angiogenesis was recognized as a therapeutic target for blocking cancer growth. Because of its importance, VEGF has been at the center stage of antiangiogenic therapy. Now, several years after FDA approval of an anti-VEGF antibody as the first antiangiogenic agent, many patients with cancer and ocular neovascularization have benefited from VEGF-targeted therapy; however, this anticancer strategy is challenged by insufficient efficacy, intrinsic refractoriness, and resistance. Here, we examine recent discoveries of new mechanisms underlying angiogenesis, discuss successes and challenges of current antiangiogenic therapy, and highlight emerging antiangiogenic paradigms.
Collapse
Affiliation(s)
- Jonathan Welti
- Vesalius Research Center, University of Leuven, Leuven, Belgium
| | | | | | | |
Collapse
|
27
|
Shirinifard A, McCollum CW, Bolin MB, Gustafsson JÅ, Glazier JA, Clendenon SG. 3D quantitative analyses of angiogenic sprout growth dynamics. Dev Dyn 2013; 242:518-26. [PMID: 23417958 DOI: 10.1002/dvdy.23946] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 01/16/2013] [Accepted: 02/03/2013] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Zebrafish intersegmental vessel (ISV) growth is widely used to study angiogenesis and to screen drugs and toxins that perturb angiogenesis. Most current ISV growth assays observe the presence or absence of ISVs or perturbation of ISV morphology but do not measure growth dynamics. We have developed a four-dimensional (4D, space plus time) quantitative analysis of angiogenic sprout growth dynamics for characterization of both normal and perturbed growth. RESULTS We tracked the positions of the ISV base and tip for each ISV sprout in 4D. Despite immobilization, zebrafish embryos translocated globally and non-uniformly during development. We used displacement of the ISV base and the angle between the ISV and the dorsal aorta to correct for displacement and rotation during development. From corrected tip cell coordinates, we computed average ISV trajectories. We fitted a quadratic curve to the average ISV trajectories to produce a canonical ISV trajectory for each experimental group, arsenic treated and untreated. From the canonical ISV trajectories, we computed curvature, average directed migration speed and directionality. Canonical trajectories from treated (arsenic exposed) and untreated groups differed in curvature, average directed migration speed and angle between the ISV and dorsal aorta. CONCLUSIONS 4D analysis of angiogenic sprout growth dynamics: (1) Allows quantitative assessment of ISV growth dynamics and perturbation, and (2) provides critical inputs for computational models of angiogenesis.
Collapse
Affiliation(s)
- Abbas Shirinifard
- Biocomplexity Institute and Department of Physics, Indiana University Bloomington, Bloomington, Indiana 47405-7003, USA.
| | | | | | | | | | | |
Collapse
|
28
|
Siekmann AF, Affolter M, Belting HG. The tip cell concept 10 years after: new players tune in for a common theme. Exp Cell Res 2013; 319:1255-63. [PMID: 23419245 DOI: 10.1016/j.yexcr.2013.01.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 01/31/2013] [Indexed: 01/24/2023]
Affiliation(s)
- Arndt F Siekmann
- Max Planck Institute for Molecular Biomedicine, D-48149 Muenster, Germany.
| | | | | |
Collapse
|