1
|
Staples SCR, Yin H, Sutherland FSK, Prescott EK, Tinney D, Hamilton DW, Goldman D, Poepping TL, Ellis CG, Pickering JG. Intussusceptive angiogenesis-on-a-chip: Evidence for transluminal vascular bridging by endothelial delamination. Proc Natl Acad Sci U S A 2025; 122:e2423700122. [PMID: 40244661 PMCID: PMC12036988 DOI: 10.1073/pnas.2423700122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Intussusceptive angiogenesis is an increasingly recognized vessel duplication process that generates and reshapes microvascular beds. However, the mechanism by which a vessel splits into two is poorly understood. Particularly vexing is formation of the hallmark transluminal endothelial cell bridge. How an endothelial cell comes to cross a flowing lumen rather than line it is enigmatic. To elucidate this, we used a microvessel-on-a-chip strategy, creating a microconduit coherently lined with flow-sensitive endothelial cells but in which transluminal bridges also formed. Bridge morphologies ranged from filamentous strand to multicellular columns with a central extracellular matrix-containing core. These bridge architectures were found to recapitulate those in microvessels in embryos, tumors, diseased organs, and the dermis of patients with limb-threatening ischemia. Time-lapse, multiplane, three-dimensional (3D) microscopy of the microphysiologic conduit revealed that bridges arose from endothelial cells oriented orthogonal to flow that partially released from the wall while retaining attachments at the ends. This delamination process was blocked by hyperactivation of Rho and augmented by interventions that weaken cell-substrate interactions, including inhibiting nonmuscle myosin II and blocking α5ß1 integrin. Thus, endothelial cells can leave their monolayer and transect a flowing lumen through controlled delamination. This previously unrecognized lumen entry program could explain the launch of intussusceptive angiogenesis and opens a framework for intervening.
Collapse
Affiliation(s)
- Sabrina C. R. Staples
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Hao Yin
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Frances S. K. Sutherland
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Emma K. Prescott
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Dylan Tinney
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Douglas W. Hamilton
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Daniel Goldman
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - Tamie L. Poepping
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Physics and Astronomy, Western University, London, ONN6A 3K7, Canada
| | - Christopher G. Ellis
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
| | - J. Geoffrey Pickering
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ONN6A 3K7, Canada
- London Health Sciences Centre, London, ONN6A 5A5, Canada
| |
Collapse
|
2
|
Grego A, Fernandes C, Fonseca I, Dias-Neto M, Costa R, Leite-Moreira A, Oliveira SM, Trindade F, Nogueira-Ferreira R. Endothelial dysfunction in cardiovascular diseases: mechanisms and in vitro models. Mol Cell Biochem 2025:10.1007/s11010-025-05289-w. [PMID: 40259179 DOI: 10.1007/s11010-025-05289-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 04/08/2025] [Indexed: 04/23/2025]
Abstract
Endothelial cells (ECs) are arranged side-by-side to create a semi-permeable monolayer, forming the inner lining of every blood vessel (micro and macrocirculation). Serving as the first barrier for circulating molecules and cells, ECs represent the main regulators of vascular homeostasis being able to respond to environmental changes, either physical or chemical signals, by producing several factors that regulate vascular tone and cellular adhesion. Healthy endothelium has anticoagulant properties that prevent the adhesion of leukocytes and platelets to the vessel walls, contributing to resistance to thrombus formation, and regulating inflammation, and vascular smooth muscle cell proliferation. Many risk factors of cardiovascular diseases (CVDs) promote the endothelial expression of chemokines, cytokines, and adhesion molecules. The resultant endothelial activation can lead to endothelial cell dysfunction (ECD). In vitro models of ECD allow the study of cellular and molecular mechanisms of disease and provide a research platform for screening potential therapeutic agents. Even though alternative models are available, such as animal models or ex vivo models, in vitro models offer higher experimental flexibility and reproducibility, making them a valuable tool for the understanding of pathophysiological mechanisms of several diseases, such as CVDs. Therefore, this review aims to synthesize the currently available in vitro models regarding ECD, emphasizing CVDs. This work will focus on 2D cell culture models (endothelial cell lines and primary ECs), 3D cell culture systems (scaffold-free and scaffold-based), and 3D cell culture models (such as organ-on-a-chip). We will dissect the role of external stimuli-chemical and mechanical-in triggering ECD.
Collapse
Affiliation(s)
- Ana Grego
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Cristiana Fernandes
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Ivo Fonseca
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Marina Dias-Neto
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- Department of Angiology and Vascular Surgery, Unidade Local de Saúde de São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Raquel Costa
- Universidade Católica Portuguesa, CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005, Porto, Portugal
| | - Adelino Leite-Moreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- Department of Cardiothoracic Surgery, Unidade Local de Saúde de São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Sandra Marisa Oliveira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Fábio Trindade
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Rita Nogueira-Ferreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.
| |
Collapse
|
3
|
Andrews TGR, Priya R. The Mechanics of Building Functional Organs. Cold Spring Harb Perspect Biol 2025; 17:a041520. [PMID: 38886066 PMCID: PMC7616527 DOI: 10.1101/cshperspect.a041520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Organ morphogenesis is multifaceted, multiscale, and fundamentally a robust process. Despite the complex and dynamic nature of embryonic development, organs are built with reproducible size, shape, and function, allowing them to support organismal growth and life. This striking reproducibility of tissue form exists because morphogenesis is not entirely hardwired. Instead, it is an emergent product of mechanochemical information flow, operating across spatial and temporal scales-from local cellular deformations to organ-scale form and function, and back. In this review, we address the mechanical basis of organ morphogenesis, as understood by observations and experiments in living embryos. To this end, we discuss how mechanical information controls the emergence of a highly conserved set of structural motifs that shape organ architectures across the animal kingdom: folds and loops, tubes and lumens, buds, branches, and networks. Moving forward, we advocate for a holistic conceptual framework for the study of organ morphogenesis, which rests on an interdisciplinary toolkit and brings the embryo center stage.
Collapse
Affiliation(s)
| | - Rashmi Priya
- The Francis Crick Institute, London NW1 1AT, United Kingdom
| |
Collapse
|
4
|
Kowalski WJ, Vatti S, Sakamoto T, Li W, Odutola SR, Liu C, Chen G, Boehm M, Mukouyama YS. In vivo transplantation of mammalian vascular organoids onto the chick chorioallantoic membrane reveals the formation of a hierarchical vascular network. Sci Rep 2025; 15:7150. [PMID: 40021912 PMCID: PMC11871353 DOI: 10.1038/s41598-025-91826-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/24/2025] [Indexed: 03/03/2025] Open
Abstract
The dynamic remodeling of the nascent vascular network into a mature hierarchy is essential for embryo survival. Cell behaviors and signaling mechanisms are often investigated with animal models and perfused microchannels, giving insights into this process. To support these studies and enrich our understanding, we demonstrate a complementary approach using vascular organoids. Organoids initially form a primitive endothelial plexus lined with NG2+/PDGFRβ+ mural cell progenitors containing immature pericytes, but there is no formation of large-diameter vessels covered with αSMA+ cells containing immature vascular smooth muscle cells (vSMCs). After transplantation to the chick chorioallantoic membrane, the network reorganizes into a branched architecture with large-diameter vessels covered by αSMA+ cells. We additionally show that blood flow from the host circulation perfuses the organoid. Compared with the developing skin vasculature in mouse embryos, organoids successfully recapitulate vascular morphogenesis, both in vitro and after transplantation. The model described here presents a further approach to enhance the study of vascular remodeling.
Collapse
Affiliation(s)
- William J Kowalski
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shravani Vatti
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Tyler Sakamoto
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Harvard College, Cambridge, MA, USA
| | - Wenling Li
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sarah Rose Odutola
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Harvard College, Cambridge, MA, USA
| | - Chengyu Liu
- Transgenic Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Guibin Chen
- Laboratory of Cardiovascular Regenerative Medicine, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Manfred Boehm
- Laboratory of Cardiovascular Regenerative Medicine, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yoh-Suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
5
|
Ferner K. Skin development in the gray short-tailed opossum (Monodelphis domestica)-From skin respiration to thermoregulation. J Anat 2025. [PMID: 39980182 DOI: 10.1111/joa.14236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025] Open
Abstract
Marsupials are born at an early stage of development, and compared to eutherians, skin development is slow, and a functional change during skin ontogenesis occurs. The skin development in 36 gray short-tailed opossums (Monodelphis domestica) has been examined using histological, morphometric, and μCT methods during postnatal development from neonate to adult. The aim of the study is to follow the structural and functional transition of the skin in this immature marsupial species. Additionally, the postnatal development of the external appearance and the cardiac and respiratory systems is looked at to assess skin development in relation to the general development. The skin of the newborn gray short-tailed opossum is thin and undifferentiated (no hair follicles, no sebaceous and sweat glands). Numerous subepidermal capillaries allow for gaseous exchange via the skin. A dense cutaneous capillary net with a high capillary volume density (0.25 ± 0.04) is present at term, indicating significant cutaneous gas exchange in the neonate. The capillary volume density decreases markedly during the first postnatal week (0.08 ± 0.01). In the same time period, the skin diffusion barrier increases from 27 ± 4 to 87 ± 1 μm. From this age on, the skin development is characterized by thickening of the different cutaneous layers and beginning formation of hair follicles. First, hair covering the skin, sweat glands, and subcutaneous fat are observed by day 28, indicating the onset of thermoregulation. The total skin thickness in the gray short-tailed opossum increases from 58 μm at birth to 726 μm by day 35, when the pelage is fully developed. The cardiac and respiratory systems are immature at birth. A fenestrated interatrial septum is present for the first 4 days, allowing skin respiration. Between day 4 and day 7, the lung enters the saccular stage of lung development and is mature enough to meet the respiratory needs of the growing organism. During a long period of postnatal development, the structural differentiation of the skin results in a functional shift from transcutaneous gas exchange to thermoregulation in later life.
Collapse
Affiliation(s)
- Kirsten Ferner
- Museum für Naturkunde, Leibniz-Institut für Evolutions- und Biodiversitätsforschung, Berlin, Germany
| |
Collapse
|
6
|
Giese W, Albrecht JP, Oppenheim O, Akmeriç EB, Kraxner J, Schmidt D, Harrington K, Gerhardt H. Polarity-JaM: an image analysis toolbox for cell polarity, junction and morphology quantification. Nat Commun 2025; 16:1474. [PMID: 39922822 PMCID: PMC11807127 DOI: 10.1038/s41467-025-56643-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 01/24/2025] [Indexed: 02/10/2025] Open
Abstract
Cell polarity involves the asymmetric distribution of cellular components such as signalling molecules and organelles within a cell, alterations in cell morphology and cell-cell contacts. Advances in fluorescence microscopy and deep learning algorithms open up a wealth of unprecedented opportunities to characterise various aspects of cell polarity, but also create new challenges for comprehensible and interpretable image data analysis workflows to fully exploit these new opportunities. Here we present Polarity-JaM, an open source package for reproducible exploratory image analysis that provides versatile methods for single cell segmentation, feature extraction and statistical analysis. We demonstrate our analysis using fluorescence image data of endothelial cells and their collective behaviour, which has been shown to be essential for vascular development and disease. The general architecture of the software allows its application to other cell types and imaging modalities, as well as seamless integration into common image analysis workflows, see https://polarityjam.readthedocs.io . We also provide a web application for circular statistics and data visualisation, available at www.polarityjam.com , and a Napari plug-in, each with a graphical user interface to facilitate exploratory analysis. We propose a holistic image analysis workflow that is accessible to the end user in bench science, enabling comprehensive analysis of image data.
Collapse
Affiliation(s)
- Wolfgang Giese
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- DZHK (German Center for Cardiovascular Research), Berlin, Germany.
| | - Jan Philipp Albrecht
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- HELMHOLTZ IMAGING, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Faculty of Mathematics and Natural Sciences, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Olya Oppenheim
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Emir Bora Akmeriç
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Kraxner
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Berlin, Germany
| | - Deborah Schmidt
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- HELMHOLTZ IMAGING, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Kyle Harrington
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Chan Zuckerberg Institute for Advanced Biological Imaging, Redwood City, CA, USA
| | - Holger Gerhardt
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
7
|
Oppenheim O, Giese W, Park H, Baumann E, Ivanov A, Beule D, Eichmann A, Gerhardt H. Divergent endothelial mechanisms drive arteriovenous malformations in Alk1 and SMAD4 loss-of-function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.03.631070. [PMID: 39829872 PMCID: PMC11741317 DOI: 10.1101/2025.01.03.631070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Hereditary hemorrhagic telangiectasia is an autosomal dominant disorder caused by mutations in the bone morphogenetic protein signaling pathway, leading to arteriovenous malformations. While previously thought to share molecular and cellular dysregulation, this study reveals highly distinct mechanisms depending on whether mutations occur in Alk1 or SMAD4. Loss of SMAD4 enhances endothelial cell responses to flow, including flow-regulated transcription and cell migration against blood flow, causing excessive pruning of capillaries and the formation of single large shunts. Conversely, Alk1 deficiency disrupts endothelial flow responses, including cell polarization and directional migration, leading to a dense vascular network and the persistence of a malformation nidus. In vivo cell population tracking of mutant cells validates unique endothelial cell migration defects. Mosaic cell culture models further illustrate that mutant cells co-opt wild-type cells driving distinct Alk1 or SMAD4 mutant-like behavioral defects. These findings demonstrate that arteriovenous malformations develop through fundamentally different cellular mechanisms based on the specific genetic mutation emphasizing the need for tailored diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Olya Oppenheim
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Charité Universitätsmedizin Berlin, Germany
| | - Wolfgang Giese
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
| | - Hyojin Park
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Elisabeth Baumann
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Charité Universitätsmedizin Berlin, Germany
| | - Andranik Ivanov
- Charité Universitätsmedizin Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Dieter Beule
- Charité Universitätsmedizin Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Anne Eichmann
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT, USA
- PARCC, INSERM, Université de Paris, Paris, France
| | - Holger Gerhardt
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Charité Universitätsmedizin Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
8
|
Lupu IE, Grainger DE, Kirschnick N, Weischer S, Zhao E, Martinez-Corral I, Schoofs H, Vanhollebeke M, Jones G, Godwin J, Forrow A, Lahmann I, Riley PR, Zobel T, Alitalo K, Mäkinen T, Kiefer F, Stone OA. Direct specification of lymphatic endothelium from mesenchymal progenitors. NATURE CARDIOVASCULAR RESEARCH 2025; 4:45-63. [PMID: 39747454 PMCID: PMC11738995 DOI: 10.1038/s44161-024-00570-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 10/25/2024] [Indexed: 01/04/2025]
Abstract
During embryogenesis, endothelial cells (ECs) are generally described to arise from a common pool of progenitors termed angioblasts, which diversify through iterative steps of differentiation to form functionally distinct subtypes of ECs. A key example is the formation of lymphatic ECs (LECs), which are thought to arise largely through transdifferentiation from venous endothelium. Opposing this model, here we show that the initial expansion of mammalian LECs is primarily driven by the in situ differentiation of mesenchymal progenitors and does not require transition through an intermediate venous state. Single-cell genomics and lineage-tracing experiments revealed a population of paraxial mesoderm-derived Etv2+Prox1+ progenitors that directly give rise to LECs. Morphometric analyses of early LEC proliferation and migration, and mutants that disrupt lymphatic development supported these findings. Collectively, this work establishes a cellular blueprint for LEC specification and indicates that discrete pools of mesenchymal progenitors can give rise to specialized subtypes of ECs.
Collapse
Affiliation(s)
- Irina-Elena Lupu
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK
| | - David E Grainger
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK
| | - Nils Kirschnick
- University of Münster, European Institute for Molecular Imaging, Münster, Germany
- Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Sarah Weischer
- University of Münster, Cells in Motion Interfaculty Centre, Imaging Network, Münster, Germany
| | - Erica Zhao
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK
| | - Ines Martinez-Corral
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Hans Schoofs
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Marie Vanhollebeke
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK
| | - Grace Jones
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK
| | - Jonathan Godwin
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK
| | - Aden Forrow
- Mathematical Institute, University of Oxford, Oxford, UK
| | - Ines Lahmann
- Developmental Biology/Signal Transduction, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Neurowissenschaftliches Forschungzentrum, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Paul R Riley
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK
| | - Thomas Zobel
- University of Münster, Cells in Motion Interfaculty Centre, Imaging Network, Münster, Germany
| | - Kari Alitalo
- Wihuri Research Institute, Helsinki, Finland
- Translational Cancer Medicine Program, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Taija Mäkinen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Wihuri Research Institute, Helsinki, Finland
| | - Friedemann Kiefer
- University of Münster, European Institute for Molecular Imaging, Münster, Germany.
- Max Planck Institute for Molecular Biomedicine, Münster, Germany.
| | - Oliver A Stone
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, UK.
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
9
|
Ouarné M, Pena A, Ramalho D, Conchinha NV, Costa T, Enjalbert R, Figueiredo AM, Saraiva MP, Carvalho Y, Bernabeu MO, Henao Misikova L, Oh SP, Franco CA. A non-genetic model of vascular shunts informs on the cellular mechanisms of formation and resolution of arteriovenous malformations. Cardiovasc Res 2024; 120:1967-1984. [PMID: 39308243 PMCID: PMC11629978 DOI: 10.1093/cvr/cvae160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 04/11/2024] [Accepted: 05/23/2024] [Indexed: 12/11/2024] Open
Abstract
AIMS Arteriovenous malformations (AVMs), a disorder characterized by direct shunts between arteries and veins, are associated with genetic mutations. However, the mechanisms leading to AV shunt formation and how shunts can be reverted are poorly understood. METHODS AND RESULTS Here, we report that oxygen-induced retinopathy (OIR) protocol leads to the consistent and stereotypical formation of AV shunts in non-genetically altered mice. OIR-induced AV shunts show all the canonical markers of AVMs. Genetic and pharmacological interventions demonstrated that changes in the volume of venous endothelial cells (EC)-hypertrophic venous cells-are the initiating step promoting AV shunt formation, whilst EC proliferation or migration played minor roles. Inhibition of the mTOR pathway prevents pathological increases in EC volume and significantly reduces the formation of AV shunts. Importantly, we demonstrate that ALK1 signalling cell-autonomously regulates EC volume in pro-angiogenic conditions, establishing a link with hereditary haemorrhagic telangiectasia-related AVMs. Finally, we demonstrate that a combination of EC volume control and EC migration is associated with the regression of AV shunts. CONCLUSION Our findings highlight that an increase in the EC volume is the key mechanism driving the initial stages of AV shunt formation, leading to asymmetric capillary diameters. Based on our results, we propose a coherent and unifying timeline leading to the fast conversion of a capillary vessel into an AV shunt. Our data advocate for further investigation into the mechanisms regulating EC volume in health and disease as a way to identify therapeutic approaches to prevent and revert AVMs.
Collapse
Affiliation(s)
- Marie Ouarné
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon 1649-028, Portugal
| | - Andreia Pena
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon 1649-028, Portugal
- Católica Biomedical Research Centre, Universidade Católica Portuguesa, Católica Medical School, Lisbon 1649-023, Portugal
| | - Daniela Ramalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon 1649-028, Portugal
- Católica Biomedical Research Centre, Universidade Católica Portuguesa, Católica Medical School, Lisbon 1649-023, Portugal
| | - Nadine V Conchinha
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon 1649-028, Portugal
| | - Tiago Costa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon 1649-028, Portugal
| | - Romain Enjalbert
- Centre for Medical Informatics, Usher Institute, The University of Edinburgh, Edinburgh EH16 4UX, UK
| | - Ana M Figueiredo
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon 1649-028, Portugal
| | - Marta Pimentel Saraiva
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon 1649-028, Portugal
| | - Yulia Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon 1649-028, Portugal
| | - Miguel O Bernabeu
- Centre for Medical Informatics, Usher Institute, The University of Edinburgh, Edinburgh EH16 4UX, UK
- The Bayes Centre, The University of Edinburgh, Edinburgh EH8 9BT, UK
| | - Lenka Henao Misikova
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon 1649-028, Portugal
- Católica Biomedical Research Centre, Universidade Católica Portuguesa, Católica Medical School, Lisbon 1649-023, Portugal
| | - S Paul Oh
- Barrow Aneurysm & AVM Research Center, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Cláudio A Franco
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon 1649-028, Portugal
- Católica Biomedical Research Centre, Universidade Católica Portuguesa, Católica Medical School, Lisbon 1649-023, Portugal
| |
Collapse
|
10
|
Shrestha S, Shrestha BK, Tettey-Engmann F, Auniq RBZ, Subedi K, Ghimire S, Desai S, Bhattarai N. Zein-Coated Zn Metal Particles-Incorporated Nanofibers: A Potent Fibrous Platform for Loading and Release of Zn Ions for Wound Healing Application. ACS APPLIED MATERIALS & INTERFACES 2024; 16:49197-49217. [PMID: 39235841 DOI: 10.1021/acsami.4c13458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
Metal particles incorporated into polymer matrices in various forms and geometries are attractive material platforms for promoting wound healing and preventing infections. However, the fate of these metal particles and their degraded products in the tissue environment are still unknown, as both can produce cytotoxic effects and promote unwanted wound reactions. In this study, we develop biodegradable fibrous biomaterials embedded with metal particles that have an immune activation functions. Initially, biodegradable zinc (Zn) nanoparticles were modified with zein (G), a protein derived from corn. The zein-coated zinc particles (Z-G) were then embedded in polycaprolactone (P) fibers at different weight ratios to create fibrous biomaterials via electrospinning, which were subsequently analyzed for potential wound healing applications. We performed multimodal evaluations of the fibrous scaffolds, examining physicochemical properties such as fiber morphology, mechanical strength, hydrophilicity, degradation, and release of zinc ions (Zn2+), as well as biological properties, including in vitro cell culture studies. We provide evidence that the integration of 2.4 wt % of Z-G particles in polycaprolactone (PCL) nanofibrous scaffolds improved its physicochemical and biological functions. The in vitro cellular response of the scaffolds was evaluated using a series of cytotoxicity assays and immunocytochemistry analyses with three different cell types: mouse-derived fibroblast cell lines (NIH/3T3), human dermal fibroblasts (HDFn), and human umbilical vein endothelial cells (HUVECs). The composite fibrous scaffold exhibited robust activation and proliferation of NIH/3T3 and HDFn cells, along with a significant angiogenic potential in HUVECs. Immunocytochemistry confirmed elevated expression of vimentin and α-smooth muscle actin (α-SMA), suggesting that NIH/3T3 and Haden cells were highly differentiated into myofibroblasts. Additionally, the increased expression of CD31 and VE-cadherin in HUVECs suggests that the scaffold supports tube formation, thereby enhancing neovascularization and promoting an effective immune response. Overall, our findings demonstrate the regenerative potential of the self-enhanced Zn hemostatic bioscaffolds, which deliver both Zn2+ ions and zein proteins to nourish cells. This capability not only modulates cellular activities but also contributes to tissue repair and remodeling, making the scaffolds suitable for wound repair and various bioengineering applications.
Collapse
Affiliation(s)
- Sita Shrestha
- Department of Chemical, Biological and Bioengineering, North Carolina A&T State University, Greensboro, North Carolina 27411, United States
| | - Bishnu Kumar Shrestha
- Department of Chemical, Biological and Bioengineering, North Carolina A&T State University, Greensboro, North Carolina 27411, United States
- Department of Chemistry, North Carolina A&T State University, 1601 E Market St, Greensboro, North Carolina 27411, United States
| | - Felix Tettey-Engmann
- Department of Chemical, Biological and Bioengineering, North Carolina A&T State University, Greensboro, North Carolina 27411, United States
- Department of Industrial and Systems Engineering, North Carolina A&T State University, Greensboro, North Carolina 27411, United States
| | - Reedwan Bin Zafar Auniq
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering (JSNN), North Carolina A&T State University, Greensboro, North Carolina 27401, United States
| | - Kiran Subedi
- Analytical Services Laboratory, College of Agriculture and Environmental Sciences, North Carolina A&T State University, Greensboro, North Carolina 27411, United States
| | - Sanjaya Ghimire
- Department of Chemical, Biological and Bioengineering, North Carolina A&T State University, Greensboro, North Carolina 27411, United States
| | - Salil Desai
- Department of Industrial and Systems Engineering, North Carolina A&T State University, Greensboro, North Carolina 27411, United States
| | - Narayan Bhattarai
- Department of Chemical, Biological and Bioengineering, North Carolina A&T State University, Greensboro, North Carolina 27411, United States
| |
Collapse
|
11
|
Nicolas N, de Tilly A, Roux E. Blood shear stress during the cardiac cycle and endothelial cell orientation and polarity in the carotid artery of male and female mice. Front Physiol 2024; 15:1386151. [PMID: 39072218 PMCID: PMC11272658 DOI: 10.3389/fphys.2024.1386151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 06/17/2024] [Indexed: 07/30/2024] Open
Abstract
Introduction: Blood flow produces fluid shear stress (SS), a frictional force parallel to the blood flow, on the endothelial cell (EC) layer of the lumen of the vessels. ECs themselves are sensitive to this frictional force in terms of directionality and intensity. The aim of this study was to determine the physiological shear stress value during the cardiac cycle and EC polarity and orientation from blood flow in healthy male and female mouse carotid artery. Methods: Experimentation is done on anesthetized male and female 8-week-old C5BL/6J mice. In vivo measurements of maximum blood velocity and vessel diameter in diastole and systole were performed on the right common carotid artery by Doppler ultrasound imaging. Blood viscosity (total and plasmatic) and hematocrit were determined on blood samples. For SS calculation, we developed a new method assuming heterogenous blood flow, i.e., a red cell central plug flow surrounded by a peripheral plasma sheath flow, and computing SS from vessel diameter and hemodynamical measurements (maximal blood velocity, hematocrit and plasmatic viscosity). Results: Results were compared with the classical method assuming a homogenous blood flow with constant apparent total blood viscosity. EC polarity and orientation were determined ex vivo on the carotid endothelium by confocal imaging after labeling of the EC nucleus and Golgi apparatus. Diastolic and systolic SS were 6 ± 2.5 Pa and 30 ± 6.5 Pa, respectively. Total blood and plasmatic viscosity was 4 ± 0.5 cP and 1.27 cP, respectively. ECs were polarized and significantly oriented against blood flow. No sex difference was identified.
Collapse
Affiliation(s)
- Nabil Nicolas
- Biologie des Maladies Cardiovasculaires, INSERM, U1034, University of Bordeaux, Pessac, France
| | | | - Etienne Roux
- Biologie des Maladies Cardiovasculaires, INSERM, U1034, University of Bordeaux, Pessac, France
| |
Collapse
|
12
|
Bougaran P, Bautch VL. Life at the crossroads: the nuclear LINC complex and vascular mechanotransduction. Front Physiol 2024; 15:1411995. [PMID: 38831796 PMCID: PMC11144885 DOI: 10.3389/fphys.2024.1411995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/02/2024] [Indexed: 06/05/2024] Open
Abstract
Vascular endothelial cells line the inner surface of all blood vessels, where they are exposed to polarized mechanical forces throughout their lifespan. Both basal substrate interactions and apical blood flow-induced shear stress regulate blood vessel development, remodeling, and maintenance of vascular homeostasis. Disruption of these interactions leads to dysfunction and vascular pathologies, although how forces are sensed and integrated to affect endothelial cell behaviors is incompletely understood. Recently the endothelial cell nucleus has emerged as a prominent force-transducing organelle that participates in vascular mechanotransduction, via communication to and from cell-cell and cell-matrix junctions. The LINC complex, composed of SUN and nesprin proteins, spans the nuclear membranes and connects the nuclear lamina, the nuclear envelope, and the cytoskeleton. Here we review LINC complex involvement in endothelial cell mechanotransduction, describe unique and overlapping functions of each LINC complex component, and consider emerging evidence that two major SUN proteins, SUN1 and SUN2, orchestrate a complex interplay that extends outward to cell-cell and cell-matrix junctions and inward to interactions within the nucleus and chromatin. We discuss these findings in relation to vascular pathologies such as Hutchinson-Gilford progeria syndrome, a premature aging disorder with cardiovascular impairment. More knowledge of LINC complex regulation and function will help to understand how the nucleus participates in endothelial cell force sensing and how dysfunction leads to cardiovascular disease.
Collapse
Affiliation(s)
- Pauline Bougaran
- Department of Biology, The University of North Carolina, Chapel Hill, NC, United States
| | - Victoria L. Bautch
- Department of Biology, The University of North Carolina, Chapel Hill, NC, United States
- McAllister Heart Institute, The University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
13
|
Abdelilah-Seyfried S, Ola R. Shear stress and pathophysiological PI3K involvement in vascular malformations. J Clin Invest 2024; 134:e172843. [PMID: 38747293 PMCID: PMC11093608 DOI: 10.1172/jci172843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024] Open
Abstract
Molecular characterization of vascular anomalies has revealed that affected endothelial cells (ECs) harbor gain-of-function (GOF) mutations in the gene encoding the catalytic α subunit of PI3Kα (PIK3CA). These PIK3CA mutations are known to cause solid cancers when occurring in other tissues. PIK3CA-related vascular anomalies, or "PIKopathies," range from simple, i.e., restricted to a particular form of malformation, to complex, i.e., presenting with a range of hyperplasia phenotypes, including the PIK3CA-related overgrowth spectrum. Interestingly, development of PIKopathies is affected by fluid shear stress (FSS), a physiological stimulus caused by blood or lymph flow. These findings implicate PI3K in mediating physiological EC responses to FSS conditions characteristic of lymphatic and capillary vessel beds. Consistent with this hypothesis, increased PI3K signaling also contributes to cerebral cavernous malformations, a vascular disorder that affects low-perfused brain venous capillaries. Because the GOF activity of PI3K and its signaling partners are excellent drug targets, understanding PIK3CA's role in the development of vascular anomalies may inform therapeutic strategies to normalize EC responses in the diseased state. This Review focuses on PIK3CA's role in mediating EC responses to FSS and discusses current understanding of PIK3CA dysregulation in a range of vascular anomalies that particularly affect low-perfused regions of the vasculature. We also discuss recent surprising findings linking increased PI3K signaling to fast-flow arteriovenous malformations in hereditary hemorrhagic telangiectasias.
Collapse
Affiliation(s)
| | - Roxana Ola
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
14
|
Lin Y, Gahn J, Banerjee K, Dobreva G, Singhal M, Dubrac A, Ola R. Role of endothelial PDGFB in arterio-venous malformations pathogenesis. Angiogenesis 2024; 27:193-209. [PMID: 38070064 PMCID: PMC11021264 DOI: 10.1007/s10456-023-09900-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/05/2023] [Indexed: 04/17/2024]
Abstract
Arterial-venous malformations (AVMs) are direct connections between arteries and veins without an intervening capillary bed. Either familial inherited or sporadically occurring, localized pericytes (PCs) drop is among the AVMs' hallmarks. Whether impaired PC coverage triggers AVMs or it is a secondary event is unclear. Here we evaluated the role of the master regulator of PC recruitment, Platelet derived growth factor B (PDGFB) in AVM pathogenesis. Using tamoxifen-inducible deletion of Pdgfb in endothelial cells (ECs), we show that disruption of EC Pdgfb-mediated PC recruitment and maintenance leads to capillary enlargement and organotypic AVM-like structures. These vascular lesions contain non-proliferative hyperplastic, hypertrophic and miss-oriented capillary ECs with an altered capillary EC fate identity. Mechanistically, we propose that PDGFB maintains capillary EC size and caliber to limit hemodynamic changes, thus restricting expression of Krüppel like factor 4 and activation of Bone morphogenic protein, Transforming growth factor β and NOTCH signaling in ECs. Furthermore, our study emphasizes that inducing or activating PDGFB signaling may be a viable therapeutic approach for treating vascular malformations.
Collapse
Affiliation(s)
- Yanzhu Lin
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Johannes Gahn
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kuheli Banerjee
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gergana Dobreva
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), Heidelberg, Germany
| | - Mahak Singhal
- Laboratory of AngioRhythms, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alexandre Dubrac
- Centre de Recherche, CHU St. Justine, Montreal, QC, H3T 1C5, Canada
- Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montreal, QC, H3T 1J4, Canada
| | - Roxana Ola
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
15
|
Genet G, Genet N, Paila U, Cain SR, Cwiek A, Chavkin NW, Serbulea V, Figueras A, Cerdà P, McDonnell SP, Sankaranarayanan D, Huba M, Nelson EA, Riera-Mestre A, Hirschi KK. Induced Endothelial Cell Cycle Arrest Prevents Arteriovenous Malformations in Hereditary Hemorrhagic Telangiectasia. Circulation 2024; 149:944-962. [PMID: 38126211 PMCID: PMC10954087 DOI: 10.1161/circulationaha.122.062952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/27/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Distinct endothelial cell cycle states (early G1 versus late G1) provide different "windows of opportunity" to enable the differential expression of genes that regulate venous versus arterial specification, respectively. Endothelial cell cycle control and arteriovenous identities are disrupted in vascular malformations including arteriovenous shunts, the hallmark of hereditary hemorrhagic telangiectasia (HHT). To date, the mechanistic link between endothelial cell cycle regulation and the development of arteriovenous malformations (AVMs) in HHT is not known. METHODS We used BMP (bone morphogenetic protein) 9/10 blocking antibodies and endothelial-specific deletion of activin A receptor like type 1 (Alk1) to induce HHT in Fucci (fluorescent ubiquitination-based cell cycle indicator) 2 mice to assess endothelial cell cycle states in AVMs. We also assessed the therapeutic potential of inducing endothelial cell cycle G1 state in HHT to prevent AVMs by repurposing the Food and Drug Administration-approved CDK (cyclin-dependent kinase) 4/6 inhibitor (CDK4/6i) palbociclib. RESULTS We found that endothelial cell cycle state and associated gene expressions are dysregulated during the pathogenesis of vascular malformations in HHT. We also showed that palbociclib treatment prevented AVM development induced by BMP9/10 inhibition and Alk1 genetic deletion. Mechanistically, endothelial cell late G1 state induced by palbociclib modulates the expression of genes regulating arteriovenous identity, endothelial cell migration, metabolism, and VEGF-A (vascular endothelial growth factor A) and BMP9 signaling that collectively contribute to the prevention of vascular malformations. CONCLUSIONS This study provides new insights into molecular mechanisms leading to HHT by defining how endothelial cell cycle is dysregulated in AVMs because of BMP9/10 and Alk1 signaling deficiencies, and how restoration of endothelial cell cycle control may be used to treat AVMs in patients with HHT.
Collapse
Affiliation(s)
- Gael Genet
- Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville
| | - Nafiisha Genet
- Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville
| | - Umadevi Paila
- Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville
| | - Shelby R Cain
- Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville
| | - Aleksandra Cwiek
- Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville
| | - Nicholas W Chavkin
- Robert M. Berne Cardiovascular Research Center (N.W.C., V.S., K.K.H.), School of Medicine, University of Virginia, Charlottesville
| | - Vlad Serbulea
- Robert M. Berne Cardiovascular Research Center (N.W.C., V.S., K.K.H.), School of Medicine, University of Virginia, Charlottesville
| | - Agnès Figueras
- Program Against Cancer Therapeutic Resistance, Institut Catala d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain (A.F.)
- Oncobell Program (A.F.), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Pau Cerdà
- (P.C., A.R.-M.), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
- HHT Unit, Internal Medicine Department, Hospital Universitari Bellvitge, Barcelona, Spain (P.C., A.R.-M.)
| | - Stephanie P McDonnell
- Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville
| | - Danya Sankaranarayanan
- Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville
| | - Mahalia Huba
- Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville
| | - Elizabeth A Nelson
- Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville
| | - Antoni Riera-Mestre
- (P.C., A.R.-M.), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
- HHT Unit, Internal Medicine Department, Hospital Universitari Bellvitge, Barcelona, Spain (P.C., A.R.-M.)
- Department of Clinical Science, Faculty of Medicine and Health Sciences, Universitat de Barcelona, Spain (A.R.-M.)
| | - Karen K Hirschi
- Department of Cell Biology (G.G., N.G., U.P., S.R.C., A.C., S.P.M., D.S., M.H., E.A.N., K.K.H.), School of Medicine, University of Virginia, Charlottesville
- Robert M. Berne Cardiovascular Research Center (N.W.C., V.S., K.K.H.), School of Medicine, University of Virginia, Charlottesville
- Department of Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT (K.K.H.)
| |
Collapse
|
16
|
Stepanova D, Byrne HM, Maini PK, Alarcón T. Computational modeling of angiogenesis: The importance of cell rearrangements during vascular growth. WIREs Mech Dis 2024; 16:e1634. [PMID: 38084799 DOI: 10.1002/wsbm.1634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 03/16/2024]
Abstract
Angiogenesis is the process wherein endothelial cells (ECs) form sprouts that elongate from the pre-existing vasculature to create new vascular networks. In addition to its essential role in normal development, angiogenesis plays a vital role in pathologies such as cancer, diabetes and atherosclerosis. Mathematical and computational modeling has contributed to unraveling its complexity. Many existing theoretical models of angiogenic sprouting are based on the "snail-trail" hypothesis. This framework assumes that leading ECs positioned at sprout tips migrate toward low-oxygen regions while other ECs in the sprout passively follow the leaders' trails and proliferate to maintain sprout integrity. However, experimental results indicate that, contrary to the snail-trail assumption, ECs exchange positions within developing vessels, and the elongation of sprouts is primarily driven by directed migration of ECs. The functional role of cell rearrangements remains unclear. This review of the theoretical modeling of angiogenesis is the first to focus on the phenomenon of cell mixing during early sprouting. We start by describing the biological processes that occur during early angiogenesis, such as phenotype specification, cell rearrangements and cell interactions with the microenvironment. Next, we provide an overview of various theoretical approaches that have been employed to model angiogenesis, with particular emphasis on recent in silico models that account for the phenomenon of cell mixing. Finally, we discuss when cell mixing should be incorporated into theoretical models and what essential modeling components such models should include in order to investigate its functional role. This article is categorized under: Cardiovascular Diseases > Computational Models Cancer > Computational Models.
Collapse
Affiliation(s)
- Daria Stepanova
- Laboratorio Subterráneo de Canfranc, Canfranc-Estación, Huesca, Spain
| | - Helen M Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Philip K Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
| | - Tomás Alarcón
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
- Centre de Recerca Matemàtica, Bellaterra, Barcelona, Spain
- Departament de Matemàtiques, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
17
|
Orozco-García E, Getova V, Calderón JC, Harmsen MC, Narvaez-Sanchez R. Angiogenesis is promoted by hypoxic cervical carcinoma-derived extracellular vesicles depending on the endothelial cell environment. Vascul Pharmacol 2024; 154:107276. [PMID: 38242295 DOI: 10.1016/j.vph.2024.107276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/04/2024] [Accepted: 01/15/2024] [Indexed: 01/21/2024]
Abstract
INTRODUCTION Cancer needs perfusion for its growth and metastasis. Cancer cell-derived extracellular vesicles (CA-EVs) alter the tumor microenvironment (TME), potentially promoting angiogenesis. We hypothesize that conditions in the tumor, e.g., hypoxia, and in the target cells of the TME, e.g., nutrient deprivation or extracellular matrix, can affect the angiogenic potential of CA-EVs, which would contribute to explaining the regulation of tumor vascularization and its influence on cancer growth and metastasis. METHODS CA-EVs were isolated and characterized from cervical carcinoma cell lines HeLa and SiHa cultured under normoxia and hypoxia, and their angiogenic potential was evaluated in vitro in three endothelial cells (ECs) lines and aortic rings, cultured in basal (growth factor-reduced) or complete medium. RESULTS Hypoxia increased EV production 10-100 times and protein content 2-4 times compared to normoxic CA-EVs. HeLa-EVs contained six times more RNA than SiHa-EVs, and this concentration was not affected by hypoxia. Treatment with CA-EVs increased tube formation and sprouting in ECs and aortic rings cultured in basal medium and long-term stabilized the stablished vascular networks formed by ECs cultured in complete medium. CONCLUSION Hypoxia differentially affects CA-EVs in a cell line-dependent manner. The cellular environment (nutrient availability and extracellular matrix scaffold) influences the effect of CA-EV on the angiogenic potential of ECs.
Collapse
Affiliation(s)
- E Orozco-García
- Physiology and Biochemistry Research Group - PHYSIS, Faculty of Medicine, University of Antioquia, Medellin, Colombia; University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Hanzeplein 1 (EA11), Groningen 9713 GZ, the Netherlands; University of Groningen, University Medical Center Groningen, W.J. Kolff Research Institute, Groningen, the Netherlands
| | - V Getova
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Hanzeplein 1 (EA11), Groningen 9713 GZ, the Netherlands; University of Groningen, University Medical Center Groningen, W.J. Kolff Research Institute, Groningen, the Netherlands
| | - J C Calderón
- Physiology and Biochemistry Research Group - PHYSIS, Faculty of Medicine, University of Antioquia, Medellin, Colombia
| | - M C Harmsen
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Hanzeplein 1 (EA11), Groningen 9713 GZ, the Netherlands; University of Groningen, University Medical Center Groningen, W.J. Kolff Research Institute, Groningen, the Netherlands.
| | - R Narvaez-Sanchez
- Physiology and Biochemistry Research Group - PHYSIS, Faculty of Medicine, University of Antioquia, Medellin, Colombia
| |
Collapse
|
18
|
Qi Y, Chang SS, Wang Y, Chen C, Baek KI, Hsiai T, Roper M. Hemodynamic regulation allows stable growth of microvascular networks. Proc Natl Acad Sci U S A 2024; 121:e2310993121. [PMID: 38386707 PMCID: PMC10907248 DOI: 10.1073/pnas.2310993121] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 01/16/2024] [Indexed: 02/24/2024] Open
Abstract
How do vessels find optimal radii? Capillaries are known to adapt their radii to maintain the shear stress of blood flow at the vessel wall at a set point, yet models of adaptation purely based on average shear stress have not been able to produce complex loopy networks that resemble real microvascular systems. For narrow vessels where red blood cells travel in a single file, the shear stress on vessel endothelium peaks sharply when a red blood cell passes through. We show that stable shear-stress-based adaptation is possible if vessel shear stress set points are cued to the stress peaks. Model networks that respond to peak stresses alone can quantitatively reproduce the observed zebrafish trunk microcirculation, including its adaptive trajectory when hematocrit changes or parts of the network are amputated. Our work reveals the potential for mechanotransduction alone to generate stable hydraulically tuned microvascular networks.
Collapse
Affiliation(s)
- Yujia Qi
- Department of Mechanical Engineering, University of California, Los Angeles, CA90095
| | - Shyr-Shea Chang
- Department of Mathematics, University of California, Los Angeles, CA90095
| | - Yixuan Wang
- Department of Mathematics, University of California, Los Angeles, CA90095
| | - Cynthia Chen
- Department of Bioengineering, University of California, Los Angeles, CA90095
| | - Kyung In Baek
- Department of Bioengineering, University of California, Los Angeles, CA90095
| | - Tzung Hsiai
- Department of Bioengineering, University of California, Los Angeles, CA90095
| | - Marcus Roper
- Department of Mathematics, University of California, Los Angeles, CA90095
- Department of Computational Medicine, University of California, Los Angeles, CA90095
| |
Collapse
|
19
|
Köry J, Narain V, Stolz BJ, Kaeppler J, Markelc B, Muschel RJ, Maini PK, Pitt-Francis JM, Byrne HM. Enhanced perfusion following exposure to radiotherapy: A theoretical investigation. PLoS Comput Biol 2024; 20:e1011252. [PMID: 38363799 PMCID: PMC10903964 DOI: 10.1371/journal.pcbi.1011252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 02/29/2024] [Accepted: 01/23/2024] [Indexed: 02/18/2024] Open
Abstract
Tumour angiogenesis leads to the formation of blood vessels that are structurally and spatially heterogeneous. Poor blood perfusion, in conjunction with increased hypoxia and oxygen heterogeneity, impairs a tumour's response to radiotherapy. The optimal strategy for enhancing tumour perfusion remains unclear, preventing its regular deployment in combination therapies. In this work, we first identify vascular architectural features that correlate with enhanced perfusion following radiotherapy, using in vivo imaging data from vascular tumours. Then, we present a novel computational model to determine the relationship between these architectural features and blood perfusion in silico. If perfusion is defined to be the proportion of vessels that support blood flow, we find that vascular networks with small mean diameters and large numbers of angiogenic sprouts show the largest increases in perfusion post-irradiation for both biological and synthetic tumours. We also identify cases where perfusion increases due to the pruning of hypoperfused vessels, rather than blood being rerouted. These results indicate the importance of considering network composition when determining the optimal irradiation strategy. In the future, we aim to use our findings to identify tumours that are good candidates for perfusion enhancement and to improve the efficacy of combination therapies.
Collapse
Affiliation(s)
- Jakub Köry
- School of Mathematics and Statistics, University of Glasgow, Glasgow, United Kingdom
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Vedang Narain
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Bernadette J. Stolz
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
- Laboratory for Topology and Neuroscience, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Jakob Kaeppler
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Bostjan Markelc
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Ruth J. Muschel
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Philip K. Maini
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Joe M. Pitt-Francis
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Helen M. Byrne
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
20
|
Hirth E, Cao W, Peltonen M, Kapetanovic E, Dietsche C, Svanberg S, Filippova M, Reddy S, Dittrich PS. Self-assembled and perfusable microvasculature-on-chip for modeling leukocyte trafficking. LAB ON A CHIP 2024; 24:292-304. [PMID: 38086670 PMCID: PMC10793075 DOI: 10.1039/d3lc00719g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/20/2023] [Indexed: 01/18/2024]
Abstract
Leukocyte recruitment from blood to tissue is a process that occurs at the level of capillary vessels during both physiological and pathological conditions. This process is also relevant for evaluating novel adoptive cell therapies, in which the trafficking of therapeutic cells such as chimeric antigen receptor (CAR)-T cells throughout the capillaries of solid tumors is important. Local variations in blood flow, mural cell concentration, and tissue stiffness contribute to the regulation of capillary vascular permeability and leukocyte trafficking throughout the capillary microvasculature. We developed a platform to mimic a biologically functional human arteriole-venule microcirculation system consisting of pericytes (PCs) and arterial and venous primary endothelial cells (ECs) embedded within a hydrogel, which self-assembles into a perfusable, heterogeneous microvasculature. Our device shows a preferential association of PCs with arterial ECs that drives the flow-dependent formation of microvasculature networks. We show that PCs stimulate basement membrane matrix synthesis, which affects both vessel diameter and permeability in a manner correlating with the ratio of ECs to PCs. Moreover, we demonstrate that hydrogel concentration can affect capillary morphology but has no observed effect on vascular permeability. The biological function of our capillary network was demonstrated using an inflammation model, where significantly higher expression of cytokines, chemokines, and adhesion molecules was observed after tumor necrosis factor-alpha (TNF-α) treatment. Accordingly, T cell adherence and transendothelial migration were significantly increased in the immune-activated state. Taken together, our platform allows the generation of a perfusable microvasculature that recapitulates the structure and function of an in vivo capillary bed that can be used as a model for developing potential immunotherapies.
Collapse
Affiliation(s)
- Elisabeth Hirth
- Department of Biosystems Science and Engineering, ETH Zurich, 4056, Basel, Switzerland.
| | - Wuji Cao
- Department of Biosystems Science and Engineering, ETH Zurich, 4056, Basel, Switzerland.
| | - Marina Peltonen
- Department of Biosystems Science and Engineering, ETH Zurich, 4056, Basel, Switzerland.
| | - Edo Kapetanovic
- Department of Biosystems Science and Engineering, ETH Zurich, 4056, Basel, Switzerland.
| | - Claudius Dietsche
- Department of Biosystems Science and Engineering, ETH Zurich, 4056, Basel, Switzerland.
| | - Sara Svanberg
- Department of Biosystems Science and Engineering, ETH Zurich, 4056, Basel, Switzerland.
| | - Maria Filippova
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Sai Reddy
- Department of Biosystems Science and Engineering, ETH Zurich, 4056, Basel, Switzerland.
| | - Petra S Dittrich
- Department of Biosystems Science and Engineering, ETH Zurich, 4056, Basel, Switzerland.
| |
Collapse
|
21
|
Kobialka P, Llena J, Deleyto-Seldas N, Munar-Gelabert M, Dengra JA, Villacampa P, Albinyà-Pedrós A, Muixi L, Andrade J, van Splunder H, Angulo-Urarte A, Potente M, Grego-Bessa J, Castillo SD, Vanhaesebroeck B, Efeyan A, Graupera M. PI3K-C2β limits mTORC1 signaling and angiogenic growth. Sci Signal 2023; 16:eadg1913. [PMID: 38015911 DOI: 10.1126/scisignal.adg1913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 11/07/2023] [Indexed: 11/30/2023]
Abstract
Phosphoinositide 3-kinases (PI3Ks) phosphorylate intracellular inositol lipids to regulate signaling and intracellular vesicular trafficking. Mammals have eight PI3K isoforms, of which class I PI3Kα and class II PI3K-C2α are essential for vascular development. The class II PI3K-C2β is also abundant in endothelial cells. Using in vivo and in vitro approaches, we found that PI3K-C2β was a critical regulator of blood vessel growth by restricting endothelial mTORC1 signaling. Mice expressing a kinase-inactive form of PI3K-C2β displayed enlarged blood vessels without corresponding changes in endothelial cell proliferation or migration. Instead, inactivation of PI3K-C2β resulted in an increase in the size of endothelial cells, particularly in the sprouting zone of angiogenesis. Mechanistically, we showed that the aberrantly large size of PI3K-C2β mutant endothelial cells was caused by mTORC1 activation, which sustained growth in these cells. Consistently, pharmacological inhibition of mTORC1 with rapamycin normalized vascular morphogenesis in PI3K-C2β mutant mice. Together, these results identify PI3K-C2β as a crucial determinant of endothelial signaling and illustrate the importance of mTORC1 regulation during angiogenic growth.
Collapse
Affiliation(s)
- Piotr Kobialka
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Judith Llena
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Nerea Deleyto-Seldas
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Center (CNIO), Melchor Fernandez Almagro 3, Madrid 28029, Spain
| | - Margalida Munar-Gelabert
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Jose A Dengra
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Pilar Villacampa
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Alba Albinyà-Pedrós
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Laia Muixi
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Jorge Andrade
- Angiogenesis & Metabolism Laboratory, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 10178 Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Hielke van Splunder
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Ana Angulo-Urarte
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Michael Potente
- Angiogenesis & Metabolism Laboratory, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 10178 Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Joaquim Grego-Bessa
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Sandra D Castillo
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Bart Vanhaesebroeck
- Cancer Institute, Paul O'Gorman Building, University College London, WC1N 1EH London, UK
| | - Alejo Efeyan
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Center (CNIO), Melchor Fernandez Almagro 3, Madrid 28029, Spain
| | - Mariona Graupera
- Endothelial Pathobiology and Microenvironment Group, Josep Carreras Leukaemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
- ICREA, Institució Catalana de Recerca i Estudis Avançats, Pg. Lluís Companys 23, 08010 Barcelona, Spain
- CIBERONC, Instituto de Salud Carlos III, Av. de Monforte de Lemos, 5, 28029 Madrid, Spain
| |
Collapse
|
22
|
Shi Z, Yao C, Shui Y, Li S, Yan H. Research progress on the mechanism of angiogenesis in wound repair and regeneration. Front Physiol 2023; 14:1284981. [PMID: 38089479 PMCID: PMC10711283 DOI: 10.3389/fphys.2023.1284981] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/13/2023] [Indexed: 12/10/2024] Open
Abstract
Poor wound healing and pathological healing have been pressing issues in recent years, as they impact human quality of life and pose risks of long-term complications. The study of neovascularization has emerged as a prominent research focus to address these problems. During the process of repair and regeneration, the establishment of a new vascular system is an indispensable stage for complete healing. It provides favorable conditions for nutrient delivery, oxygen supply, and creates an inflammatory environment. Moreover, it is a key manifestation of the proliferative phase of wound healing, bridging the inflammatory and remodeling phases. These three stages are closely interconnected and inseparable. This paper comprehensively integrates the regulatory mechanisms of new blood vessel formation in wound healing, focusing on the proliferation and migration of endothelial cells and the release of angiogenesis-related factors under different healing outcomes. Additionally, the hidden link between the inflammatory environment and angiogenesis in wound healing is explored.
Collapse
Affiliation(s)
- Zhuojun Shi
- Department of Plastic and Burns Surgery, The Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair and Regeneration Laboratory, Luzhou, Sichuan, China
| | - Chong Yao
- Department of Plastic and Burns Surgery, The Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair and Regeneration Laboratory, Luzhou, Sichuan, China
| | - Yujie Shui
- Department of Plastic and Burns Surgery, The Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair and Regeneration Laboratory, Luzhou, Sichuan, China
| | - Site Li
- Department of Plastic and Burns Surgery, The Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair and Regeneration Laboratory, Luzhou, Sichuan, China
| | - Hong Yan
- Laboratory of Plastic Surgery, Department of Plastic Surgery and Reconstruction, Second Hospital of West China, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
23
|
Santamaría R, Cruz-Caballero J, Gkontra P, Jiménez-Montiel A, Clemente C, López JA, Villalba-Orero M, Vázquez J, Hutloff A, Lara-Pezzi E, Arroyo AG. Capillary pruning couples tissue perfusion and oxygenation with cardiomyocyte maturation in the postnatal mouse heart. Front Cell Dev Biol 2023; 11:1256127. [PMID: 38020883 PMCID: PMC10661946 DOI: 10.3389/fcell.2023.1256127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction: Removal of poorly perfused capillaries by pruning contributes to remodeling the microvasculature to optimize oxygen and nutrient delivery. Blood flow drives this process by promoting the intravascular migration of endothelial cells in developing networks, such as in the yolk sac, zebrafish brain or postnatal mouse retina. Methods: In this study, we have implemented innovative tools to recognize capillary pruning in the complex 3D coronary microvasculature of the postnatal mouse heart. We have also experimentally tested the impact of decreasing pruning on the structure and function of this network by altering blood flow with two different vasodilators: losartan and prazosin. Results: Although both drugs reduced capillary pruning, a combination of experiments based on ex vivo imaging, proteomics, electron microscopy and in vivo functional approaches showed that losartan treatment resulted in an inefficient coronary network, reduced myocardial oxygenation and metabolic changes that delayed the arrest of cardiomyocyte proliferation, in contrast to the effects of prazosin, probably due to its concomitant promotion of capillary expansion. Discussion: Our work demonstrates that capillary pruning contributes to proper maturation and function of the heart and that manipulation of blood flow may be a novel strategy to refine the microvasculature and improve tissue perfusion after damage.
Collapse
Affiliation(s)
- Ricardo Santamaría
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Polyxeni Gkontra
- Artificial Intelligence in Medicine Lab (BCN-AIM), Departament de Matemàtiques i Informàtica, Universitat de Barcelona, Barcelona, Spain
| | | | - Cristina Clemente
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Juan A. López
- Cardiovascular Proteomics Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - María Villalba-Orero
- Myocardial Pathology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jesús Vázquez
- Cardiovascular Proteomics Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Andreas Hutloff
- Institute of Immunology, University Hospital Schleswig-Holstein, Kiel, Germany
- German Rheumatism Research Centre, A Leibniz Institute, Berlin, Germany
| | - Enrique Lara-Pezzi
- Myocardial Pathology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Alicia G. Arroyo
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| |
Collapse
|
24
|
Blazeski A, Floryan MA, Fajardo-Ramírez OR, Meibalan E, Ortiz-Urbina J, Angelidakis E, Shelton SE, Kamm RD, García-Cardeña G. Engineering microvascular networks using a KLF2 reporter to probe flow-dependent endothelial cell function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.31.565021. [PMID: 37961543 PMCID: PMC10635035 DOI: 10.1101/2023.10.31.565021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Shear stress generated by the flow of blood in the vasculature is a potent regulator of endothelial cell phenotype and vascular structure. While vascular responses to flow are complex and context-dependent, endothelial cell signaling in response to shear stress induced by laminar flows is coordinated by the transcription factor KLF2. The expression of KLF2 in endothelial cells is associated with a quiescent, anti-inflammatory phenotype and has been well characterized in two-dimensional systems, but has not been studied in three-dimensional in vitro systems. Here we develop engineered microvascular networks (MVNs) with a KLF2-based endothelial cell sensor within a microfluidic chip, apply continuous flow using an attached microfluidic pump, and study the effects of this flow on vascular structure and function. We found that culture of MVNs exposed to flow for 48 hours that resulted in increased expression of the KLF2-GFP-reporter display larger vessel diameters and decreased vascular branching and resistance. Additionally, vessel diameters after the application of flow were independent of initial MVN morphologies. Finally, we found that MVNs exposed to flow have improved vascular barrier function and decreased platelet adhesion. The MVNs with KLF2-based flow sensors represent a powerful tool for evaluating the structural and functional effects of flow on engineered three-dimensional vascular systems.
Collapse
Affiliation(s)
- Adriana Blazeski
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA and Harvard Medical School, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marie A. Floryan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Oscar R. Fajardo-Ramírez
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA and Harvard Medical School, Boston, MA, USA
| | - Elamaran Meibalan
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA and Harvard Medical School, Boston, MA, USA
| | - Jesús Ortiz-Urbina
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA and Harvard Medical School, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Emmanouil Angelidakis
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sarah E. Shelton
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Roger D. Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Guillermo García-Cardeña
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA and Harvard Medical School, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
25
|
Choi DH, Oh D, Na K, Kim H, Choi D, Jung YH, Ahn J, Kim J, Kim CH, Chung S. Radiation induces acute and subacute vascular regression in a three-dimensional microvasculature model. Front Oncol 2023; 13:1252014. [PMID: 37909014 PMCID: PMC10613678 DOI: 10.3389/fonc.2023.1252014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/28/2023] [Indexed: 11/02/2023] Open
Abstract
Radiation treatment is one of the most frequently used therapies in patients with cancer, employed in approximately half of all patients. However, the use of radiation therapy is limited by acute or chronic adverse effects and the failure to consider the tumor microenvironment. Blood vessels substantially contribute to radiation responses in both normal and tumor tissues. The present study employed a three-dimensional (3D) microvasculature-on-a-chip that mimics physiological blood vessels to determine the effect of radiation on blood vessels. This model represents radiation-induced pathophysiological effects on blood vessels in terms of cellular damage and structural and functional changes. DNA double-strand breaks (DSBs), apoptosis, and cell viability indicate cellular damage. Radiation-induced damage leads to a reduction in vascular structures, such as vascular area, branch length, branch number, junction number, and branch diameter; this phenomenon occurs in the mature vascular network and during neovascularization. Additionally, vasculature regression was demonstrated by staining the basement membrane and microfilaments. Radiation exposure could increase the blockage and permeability of the vascular network, indicating that radiation alters the function of blood vessels. Radiation suppressed blood vessel recovery and induced a loss of angiogenic ability, resulting in a network of irradiated vessels that failed to recover, deteriorating gradually. These findings demonstrate that this model is valuable for assessing radiation-induced vascular dysfunction and acute and chronic effects and can potentially improve radiotherapy efficiency.
Collapse
Affiliation(s)
- Dong-Hee Choi
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
- R&D Research Center, Next&Bio Inc, Seoul, Republic of Korea
| | - Dongwoo Oh
- Korea University-Korea institute of Science and Technology (KU-KIST) Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Kyuhwan Na
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
- R&D Research Center, Next&Bio Inc, Seoul, Republic of Korea
| | - Hyunho Kim
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, United States
| | - Dongjin Choi
- Laboratory of Tissue Engineering, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Yong Hun Jung
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
- R&D Research Center, Next&Bio Inc, Seoul, Republic of Korea
| | - Jinchul Ahn
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
- R&D Research Center, Next&Bio Inc, Seoul, Republic of Korea
| | - Jaehoon Kim
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Chun-Ho Kim
- Laboratory of Tissue Engineering, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Seok Chung
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
- Korea University-Korea institute of Science and Technology (KU-KIST) Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| |
Collapse
|
26
|
Zhang P, Yan X, Zhang X, Liu Y, Feng X, Yang Z, Zhang J, Xu X, Zheng Q, Liang L, Han H. TMEM215 Prevents Endothelial Cell Apoptosis in Vessel Regression by Blunting BIK-Regulated ER-to-Mitochondrial Ca Influx. Circ Res 2023; 133:739-757. [PMID: 37750320 DOI: 10.1161/circresaha.123.322686] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 09/14/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND In developmental and pathological tissues, nascent vessel networks generated by angiogenesis require further pruning/regression to delete nonfunctional endothelial cells (ECs) by apoptosis and migration. Mechanisms underlying EC apoptosis during vessel pruning remain elusive. TMEM215 (transmembrane protein 215) is an endoplasmic reticulum-located, 2-pass transmembrane protein. We have previously demonstrated that TMEM215 knockdown in ECs leads to cell death, but its physiological function and mechanism are unclear. METHODS We characterized the role and mechanism of TMEM215 in EC apoptosis using human umbilical vein endothelial cells by identifying its interacting proteins with immunoprecipitation-mass spectrometry. The physiological function of TMEM215 in ECs was assessed by establishing a conditional knockout mouse strain. The role of TMEM215 in pathological angiogenesis was evaluated by tumor and choroidal neovascularization models. We also tried to evaluate its translational value by delivering a Tmem215 small interfering RNA (siRNA) using nanoparticles in vivo. RESULTS TMEM215 knockdown in ECs induced apoptotic cell death. We identified the chaperone BiP as a binding partner of TMEM215, and TMEM215 forms a complex with and facilitates the interaction of BiP (binding immunoglobin protein) with the BH (BCL-2 [B-cell lymphoma 2] homology) 3-only proapoptotic protein BIK (BCL-2 interacting killer). TMEM215 knockdown triggered apoptosis in a BIK-dependent way and was abrogated by BCL-2. Notably, TMEM215 knockdown increased the number and diminished the distance of mitochondria-associated endoplasmic reticulum membranes and increased mitochondrial calcium influx. Inhibiting mitochondrial calcium influx by blocking the IP3R (inositol 1,4,5-trisphosphate receptor) or MCU (mitochondrial calcium uniporter) abrogated TMEM215 knockdown-induced apoptosis. TMEM215 expression in ECs was induced by physiological laminar shear stress via EZH2 downregulation. In EC-specific Tmem215 knockout mice, induced Tmem215 depletion impaired the regression of retinal vasculature characterized by reduced vessel density, increased empty basement membrane sleeves, and increased EC apoptosis. Moreover, EC-specific Tmem215 ablation inhibited tumor growth with disrupted vasculature. However, Tmem215 ablation in adult mice attenuated lung metastasis, consistent with reduced Vcam1 expression. Administration of nanoparticles carrying Tmem215 siRNA also inhibited tumor growth and choroidal neovascularization injury. CONCLUSIONS TMEM215, which is induced by blood flow-derived shear stress via downregulating EZH2, protects ECs from BIK-triggered mitochondrial apoptosis mediated by calcium influx through mitochondria-associated ER membranes during vessel pruning, thus providing a novel target for antiangiogenic therapy.
Collapse
Affiliation(s)
- Peiran Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology (P.Z., X.Y., X.Z., Y.L., X.F., Z.Y., J.Z., X.X., L.L., H.H.), Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Xianchun Yan
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology (P.Z., X.Y., X.Z., Y.L., X.F., Z.Y., J.Z., X.X., L.L., H.H.), Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaoyan Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology (P.Z., X.Y., X.Z., Y.L., X.F., Z.Y., J.Z., X.X., L.L., H.H.), Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yuan Liu
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology (P.Z., X.Y., X.Z., Y.L., X.F., Z.Y., J.Z., X.X., L.L., H.H.), Tangdu Hospital, Fourth Military Medical University, Xi'an, China
- The Affiliated Northwest Women's and Children's Hospital of Xi'an Jiaotong University Health Science Center, China (Y.L.)
| | - Xingxing Feng
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology (P.Z., X.Y., X.Z., Y.L., X.F., Z.Y., J.Z., X.X., L.L., H.H.), Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Ziyan Yang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology (P.Z., X.Y., X.Z., Y.L., X.F., Z.Y., J.Z., X.X., L.L., H.H.), Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jiayulin Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology (P.Z., X.Y., X.Z., Y.L., X.F., Z.Y., J.Z., X.X., L.L., H.H.), Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Xinyuan Xu
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology (P.Z., X.Y., X.Z., Y.L., X.F., Z.Y., J.Z., X.X., L.L., H.H.), Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Qijun Zheng
- Department of Cardiovascular Surgery, Shenzhen People's Hospital, China (Q.Z.)
| | - Liang Liang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology (P.Z., X.Y., X.Z., Y.L., X.F., Z.Y., J.Z., X.X., L.L., H.H.), Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Hua Han
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology (P.Z., X.Y., X.Z., Y.L., X.F., Z.Y., J.Z., X.X., L.L., H.H.), Tangdu Hospital, Fourth Military Medical University, Xi'an, China
- Department of Gastroenterology (H.H.), Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
27
|
Uçar MC, Hannezo E, Tiilikainen E, Liaqat I, Jakobsson E, Nurmi H, Vaahtomeri K. Self-organized and directed branching results in optimal coverage in developing dermal lymphatic networks. Nat Commun 2023; 14:5878. [PMID: 37735168 PMCID: PMC10514270 DOI: 10.1038/s41467-023-41456-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/05/2023] [Indexed: 09/23/2023] Open
Abstract
Branching morphogenesis is a ubiquitous process that gives rise to high exchange surfaces in the vasculature and epithelial organs. Lymphatic capillaries form branched networks, which play a key role in the circulation of tissue fluid and immune cells. Although mouse models and correlative patient data indicate that the lymphatic capillary density directly correlates with functional output, i.e., tissue fluid drainage and trafficking efficiency of dendritic cells, the mechanisms ensuring efficient tissue coverage remain poorly understood. Here, we use the mouse ear pinna lymphatic vessel network as a model system and combine lineage-tracing, genetic perturbations, whole-organ reconstructions and theoretical modeling to show that the dermal lymphatic capillaries tile space in an optimal, space-filling manner. This coverage is achieved by two complementary mechanisms: initial tissue invasion provides a non-optimal global scaffold via self-organized branching morphogenesis, while VEGF-C dependent side-branching from existing capillaries rapidly optimizes local coverage by directionally targeting low-density regions. With these two ingredients, we show that a minimal biophysical model can reproduce quantitatively whole-network reconstructions, across development and perturbations. Our results show that lymphatic capillary networks can exploit local self-organizing mechanisms to achieve tissue-scale optimization.
Collapse
Affiliation(s)
- Mehmet Can Uçar
- Institute of Science and Technology Austria (IST Austria), Am Campus 1, 3400, Klosterneuburg, Austria
| | - Edouard Hannezo
- Institute of Science and Technology Austria (IST Austria), Am Campus 1, 3400, Klosterneuburg, Austria.
| | - Emmi Tiilikainen
- Translational Cancer Medicine Research Program, University of Helsinki, Biomedicum Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
| | - Inam Liaqat
- Translational Cancer Medicine Research Program, University of Helsinki, Biomedicum Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
| | - Emma Jakobsson
- Translational Cancer Medicine Research Program, University of Helsinki, Biomedicum Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
| | - Harri Nurmi
- Translational Cancer Medicine Research Program, University of Helsinki, Biomedicum Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
- Wihuri Research Institute, Biomedicum Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
| | - Kari Vaahtomeri
- Translational Cancer Medicine Research Program, University of Helsinki, Biomedicum Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland.
- Wihuri Research Institute, Biomedicum Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland.
| |
Collapse
|
28
|
Zhao ZA, Yan L, Wen J, Satyanarayanan SK, Yu F, Lu J, Liu YU, Su H. Cellular and molecular mechanisms in vascular repair after traumatic brain injury: a narrative review. BURNS & TRAUMA 2023; 11:tkad033. [PMID: 37675267 PMCID: PMC10478165 DOI: 10.1093/burnst/tkad033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 02/01/2023] [Accepted: 05/26/2023] [Indexed: 09/08/2023]
Abstract
Traumatic brain injury (TBI) disrupts normal brain function and is associated with high morbidity and fatality rates. TBI is characterized as mild, moderate or severe depending on its severity. The damage may be transient and limited to the dura matter, with only subtle changes in cerebral parenchyma, or life-threatening with obvious focal contusions, hematomas and edema. Blood vessels are often injured in TBI. Even in mild TBI, dysfunctional cerebral vascular repair may result in prolonged symptoms and poor outcomes. Various distinct types of cells participate in vascular repair after TBI. A better understanding of the cellular response and function in vascular repair can facilitate the development of new therapeutic strategies. In this review, we analyzed the mechanism of cerebrovascular impairment and the repercussions following various forms of TBI. We then discussed the role of distinct cell types in the repair of meningeal and parenchyma vasculature following TBI, including endothelial cells, endothelial progenitor cells, pericytes, glial cells (astrocytes and microglia), neurons, myeloid cells (macrophages and monocytes) and meningeal lymphatic endothelial cells. Finally, possible treatment techniques targeting these unique cell types for vascular repair after TBI are discussed.
Collapse
Affiliation(s)
- Zi-Ai Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
- Department of Neurology, General Hospital of Northern Theater Command, 83# Wen-Hua Road, Shenyang 110840, China
| | - Lingli Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Jing Wen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Senthil Kumaran Satyanarayanan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Feng Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Jiahong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Yong U Liu
- Laboratory of Neuroimmunology in Health and Disease Institute, Guangzhou First People’s Hospital School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Center, Panyu District, Guangzhou 511400, China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| |
Collapse
|
29
|
Hossain MMN, Hu NW, Abdelhamid M, Singh S, Murfee WL, Balogh P. Angiogenic Microvascular Wall Shear Stress Patterns Revealed Through Three-dimensional Red Blood Cell Resolved Modeling. FUNCTION 2023; 4:zqad046. [PMID: 37753184 PMCID: PMC10519277 DOI: 10.1093/function/zqad046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/28/2023] Open
Abstract
The wall shear stress (WSS) exerted by blood flowing through microvascular capillaries is an established driver of new blood vessel growth, or angiogenesis. Such adaptations are central to many physiological processes in both health and disease, yet three-dimensional (3D) WSS characteristics in real angiogenic microvascular networks are largely unknown. This marks a major knowledge gap because angiogenesis, naturally, is a 3D process. To advance current understanding, we model 3D red blood cells (RBCs) flowing through rat angiogenic microvascular networks using state-of-the-art simulation. The high-resolution fluid dynamics reveal 3D WSS patterns occurring at sub-endothelial cell (EC) scales that derive from distinct angiogenic morphologies, including microvascular loops and vessel tortuosity. We identify the existence of WSS hot and cold spots caused by angiogenic surface shapes and RBCs, and notably enhancement of low WSS regions by RBCs. Spatiotemporal characteristics further reveal how fluctuations follow timescales of RBC "footprints." Altogether, this work provides a new conceptual framework for understanding how shear stress might regulate EC dynamics in vivo.
Collapse
Affiliation(s)
- Mir Md Nasim Hossain
- Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ 07114, USA
| | - Nien-Wen Hu
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Maram Abdelhamid
- Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ 07114, USA
| | - Simerpreet Singh
- Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ 07114, USA
| | - Walter L Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Peter Balogh
- Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ 07114, USA
| |
Collapse
|
30
|
Crawshaw JR, Flegg JA, Bernabeu MO, Osborne JM. Mathematical models of developmental vascular remodelling: A review. PLoS Comput Biol 2023; 19:e1011130. [PMID: 37535698 PMCID: PMC10399886 DOI: 10.1371/journal.pcbi.1011130] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023] Open
Abstract
Over the past 40 years, there has been a strong focus on the development of mathematical models of angiogenesis, while developmental remodelling has received little such attention from the mathematical community. Sprouting angiogenesis can be seen as a very crude way of laying out a primitive vessel network (the raw material), while remodelling (understood as pruning of redundant vessels, diameter control, and the establishment of vessel identity and hierarchy) is the key to turning that primitive network into a functional network. This multiscale problem is of prime importance in the development of a functional vasculature. In addition, defective remodelling (either during developmental remodelling or due to a reactivation of the remodelling programme caused by an injury) is associated with a significant number of diseases. In this review, we discuss existing mathematical models of developmental remodelling and explore the important contributions that these models have made to the field of vascular development. These mathematical models are effectively used to investigate and predict vascular development and are able to reproduce experimentally observable results. Moreover, these models provide a useful means of hypothesis generation and can explain the underlying mechanisms driving the observed structural and functional network development. However, developmental vascular remodelling is still a relatively new area in mathematical biology, and many biological questions remain unanswered. In this review, we present the existing modelling paradigms and define the key challenges for the field.
Collapse
Affiliation(s)
- Jessica R. Crawshaw
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
- School of Mathematics and Statistics, The University of Melbourne, Melbourne, Australia
| | - Jennifer A. Flegg
- School of Mathematics and Statistics, The University of Melbourne, Melbourne, Australia
| | - Miguel O. Bernabeu
- Centre for Medical Informatics, The Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
- The Bayes Centre, The University of Edinburgh, Edinburgh, United Kingdom
| | - James M. Osborne
- School of Mathematics and Statistics, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
31
|
Leonard-Duke J, Bruce AC, Peirce SM, Taite LJ. Variations in mechanical stiffness alter microvascular sprouting and stability in a PEG hydrogel model of idiopathic pulmonary fibrosis. Microcirculation 2023; 30:e12817. [PMID: 37248193 PMCID: PMC10524245 DOI: 10.1111/micc.12817] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 04/07/2023] [Accepted: 05/16/2023] [Indexed: 05/31/2023]
Abstract
OBJECTIVE Microvascular remodeling is governed by biomechanical and biochemical cues which are dysregulated in idiopathic pulmonary fibrosis. Understanding how these cues impact endothelial cell-pericyte interactions necessitates a model system in which both variables can be independently and reproducibly modulated. In this study we develop a tunable hydrogel-based angiogenesis assay to study how varying angiogenic growth factors and environmental stiffness affect sprouting and vessel organization. METHODS Lungs harvested from mice were cut into 1 mm long segments then cultured on hydrogels having one of seven possible stiffness and growth factor combinations. Time course, brightfield, and immunofluorescence imaging were used to observe and quantify sprout formation. RESULTS Our assay was able to support angiogenesis in a comparable manner to Matrigel in soft 2 kPa gels while enabling tunability to study the effects of stiffness on sprout formation. Matrigel and 2 kPa groups contained significantly more samples with sprouts when compared to the stiffer 10 and 20 kPa gels. Growth factor treatment did not have as obvious an effect, although the 20 kPa PDGF + FGF-treated group had significantly longer vessels than the vascular endothelial growth factor-treated group. CONCLUSIONS We have developed a novel, tunable hydrogel assay for the creation of lung explant vessel organoids which can be modulated to study the impact of specific environmental cues on vessel formation and maturation.
Collapse
Affiliation(s)
- Julie Leonard-Duke
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Anthony C Bruce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Lakeshia J Taite
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
32
|
Travasso RDM, Coelho-Santos V. Image-based angio-adaptation modelling: a playground to study cerebrovascular development. Front Physiol 2023; 14:1223308. [PMID: 37565149 PMCID: PMC10411953 DOI: 10.3389/fphys.2023.1223308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/12/2023] [Indexed: 08/12/2023] Open
Affiliation(s)
- Rui D. M. Travasso
- Department of Physics, Center for Physics of the University of Coimbra (CFisUC), University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, Portugal
| | - Vanessa Coelho-Santos
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, Portugal
- Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
33
|
Kam CY, Singh ID, Gonzalez DG, Matte-Martone C, Solá P, Solanas G, Bonjoch J, Marsh E, Hirschi KK, Greco V. Mechanisms of skin vascular maturation and maintenance captured by longitudinal imaging of live mice. Cell 2023; 186:2345-2360.e16. [PMID: 37167971 PMCID: PMC10225355 DOI: 10.1016/j.cell.2023.04.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/03/2023] [Accepted: 04/11/2023] [Indexed: 05/13/2023]
Abstract
A functional network of blood vessels is essential for organ growth and homeostasis, yet how the vasculature matures and maintains homeostasis remains elusive in live mice. By longitudinally tracking the same neonatal endothelial cells (ECs) over days to weeks, we found that capillary plexus expansion is driven by vessel regression to optimize network perfusion. Neonatal ECs rearrange positions to evenly distribute throughout the developing plexus and become positionally stable in adulthood. Upon local ablation, adult ECs survive through a plasmalemmal self-repair response, while neonatal ECs are predisposed to die. Furthermore, adult ECs reactivate migration to assist vessel repair. Global ablation reveals coordinated maintenance of the adult vascular architecture that allows for eventual network recovery. Lastly, neonatal remodeling and adult maintenance of the skin vascular plexus are orchestrated by temporally restricted, neonatal VEGFR2 signaling. Our work sheds light on fundamental mechanisms that underlie both vascular maturation and adult homeostasis in vivo.
Collapse
Affiliation(s)
- Chen Yuan Kam
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Ishani D Singh
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - David G Gonzalez
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Paloma Solá
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Guiomar Solanas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Júlia Bonjoch
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Edward Marsh
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Karen K Hirschi
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| | - Valentina Greco
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; Departments of Cell Biology and Dermatology, Yale Stem Cell Center, Yale Cancer Center, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
34
|
Kemmler CL, Moran HR, Murray BF, Scoresby A, Klem JR, Eckert RL, Lepovsky E, Bertho S, Nieuwenhuize S, Burger S, D'Agati G, Betz C, Puller AC, Felker A, Ditrychova K, Bötschi S, Affolter M, Rohner N, Lovely CB, Kwan KM, Burger A, Mosimann C. Next-generation plasmids for transgenesis in zebrafish and beyond. Development 2023; 150:dev201531. [PMID: 36975217 PMCID: PMC10263156 DOI: 10.1242/dev.201531] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/10/2023] [Indexed: 03/29/2023]
Abstract
Transgenesis is an essential technique for any genetic model. Tol2-based transgenesis paired with Gateway-compatible vector collections has transformed zebrafish transgenesis with an accessible modular system. Here, we establish several next-generation transgenesis tools for zebrafish and other species to expand and enhance transgenic applications. To facilitate gene regulatory element testing, we generated Gateway middle entry vectors harboring the small mouse beta-globin minimal promoter coupled to several fluorophores, CreERT2 and Gal4. To extend the color spectrum for transgenic applications, we established middle entry vectors encoding the bright, blue-fluorescent protein mCerulean and mApple as an alternative red fluorophore. We present a series of p2A peptide-based 3' vectors with different fluorophores and subcellular localizations to co-label cells expressing proteins of interest. Finally, we established Tol2 destination vectors carrying the zebrafish exorh promoter driving different fluorophores as a pineal gland-specific transgenesis marker that is active before hatching and through adulthood. exorh-based reporters and transgenesis markers also drive specific pineal gland expression in the eye-less cavefish (Astyanax). Together, our vectors provide versatile reagents for transgenesis applications in zebrafish, cavefish and other models.
Collapse
Affiliation(s)
- Cassie L. Kemmler
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Hannah R. Moran
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Brooke F. Murray
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Aaron Scoresby
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - John R. Klem
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Rachel L. Eckert
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Elizabeth Lepovsky
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Sylvain Bertho
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Susan Nieuwenhuize
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Sibylle Burger
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Gianluca D'Agati
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Charles Betz
- Growth & Development, Biozentrum, Spitalstrasse 41, University of Basel, 4056 Basel, Switzerland
| | - Ann-Christin Puller
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Anastasia Felker
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Karolina Ditrychova
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Seraina Bötschi
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Markus Affolter
- Growth & Development, Biozentrum, Spitalstrasse 41, University of Basel, 4056 Basel, Switzerland
| | - Nicolas Rohner
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - C. Ben Lovely
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Kristen M. Kwan
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Alexa Burger
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Christian Mosimann
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| |
Collapse
|
35
|
Abstract
Vascular endothelial cells form the inner layer of blood vessels where they have a key role in the development and maintenance of the functional circulatory system and provide paracrine support to surrounding non-vascular cells. Technical advances in the past 5 years in single-cell genomics and in in vivo genetic labelling have facilitated greater insights into endothelial cell development, plasticity and heterogeneity. These advances have also contributed to a new understanding of the timing of endothelial cell subtype differentiation and its relationship to the cell cycle. Identification of novel tissue-specific gene expression patterns in endothelial cells has led to the discovery of crucial signalling pathways and new interactions with other cell types that have key roles in both tissue maintenance and disease pathology. In this Review, we describe the latest findings in vascular endothelial cell development and diversity, which are often supported by large-scale, single-cell studies, and discuss the implications of these findings for vascular medicine. In addition, we highlight how techniques such as single-cell multimodal omics, which have become increasingly sophisticated over the past 2 years, are being utilized to study normal vascular physiology as well as functional perturbations in disease.
Collapse
Affiliation(s)
- Emily Trimm
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Biophysics Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
36
|
Como CN, Cervantes C, Pawlikowski B, Siegenthaler J. Retinoic acid signaling in mouse retina endothelial cells is required for early angiogenic growth. Differentiation 2023; 130:16-27. [PMID: 36528974 PMCID: PMC10006372 DOI: 10.1016/j.diff.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
The development of the retinal vasculature is essential to maintain health of the tissue, but the developmental mechanisms are not completely understood. The aim of this study was to investigate the cell-autonomous role of retinoic acid signaling in endothelial cells during retina vascular development. Using a temporal and cell-specific mouse model to disrupt retinoic acid signaling in endothelial cells in the postnatal retina (Pdgfbicre/+dnRAR403fl/fl mutants), we discovered that angiogenesis in the retina is significantly decreased with a reduction in retina vascularization, endothelial tip cell number and filipodia, and endothelial 'crowding' of stalk cells. Interestingly, by P15, the vasculature can overcome the early angiogenic defect and fully vascularized the retina. At P60, the vasculature is intact with no evidence of retina cell death or altered blood retinal barrier integrity. Further, we identified that the angiogenic defect seen in mutants at P6 correlates with decreased Vegfr3 expression in endothelial cells. Collectively, our work identified a previously unappreciated function for endothelial retinoic acid signaling in early retinal angiogenesis.
Collapse
Affiliation(s)
- Christina N Como
- University of Colorado, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO, 80045, USA; University of Colorado, Anschutz Medical Campus, Neuroscience Graduate Program, Aurora, CO, 80045, USA; University of Colorado, Anschutz Medical Campus, Summer Research Training Program, Aurora, CO, 80045, USA
| | - Cesar Cervantes
- University of Colorado, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO, 80045, USA; University of Colorado, Anschutz Medical Campus, Summer Research Training Program, Aurora, CO, 80045, USA
| | - Brad Pawlikowski
- University of Colorado, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO, 80045, USA
| | - Julie Siegenthaler
- University of Colorado, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO, 80045, USA; University of Colorado, Anschutz Medical Campus, Neuroscience Graduate Program, Aurora, CO, 80045, USA; University of Colorado, Anschutz Medical Campus, Summer Research Training Program, Aurora, CO, 80045, USA.
| |
Collapse
|
37
|
Xia S, Vila Ellis L, Winkley K, Menden H, Mabry SM, Venkatraman A, Louiselle D, Gibson M, Grundberg E, Chen J, Sampath V. Neonatal hyperoxia induces activated pulmonary cellular states and sex-dependent transcriptomic changes in a model of experimental bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2023; 324:L123-L140. [PMID: 36537711 PMCID: PMC9902224 DOI: 10.1152/ajplung.00252.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/08/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022] Open
Abstract
Hyperoxia disrupts lung development in mice and causes bronchopulmonary dysplasia (BPD) in neonates. To investigate sex-dependent molecular and cellular programming involved in hyperoxia, we surveyed the mouse lung using single cell RNA sequencing (scRNA-seq), and validated our findings in human neonatal lung cells in vitro. Hyperoxia-induced inflammation in alveolar type (AT) 2 cells gave rise to damage-associated transient progenitors (DATPs). It also induced a new subpopulation of AT1 cells with reduced expression of growth factors normally secreted by AT1 cells, but increased mitochondrial gene expression. Female alveolar epithelial cells had less EMT and pulmonary fibrosis signaling in hyperoxia. In the endothelium, expansion of Car4+ EC (Cap2) was seen in hyperoxia along with an emergent subpopulation of Cap2 with repressed VEGF signaling. This regenerative response was increased in females exposed to hyperoxia. Mesenchymal cells had inflammatory signatures in hyperoxia, with a new distal interstitial fibroblast subcluster characterized by repressed lipid biosynthesis and a transcriptomic signature resembling myofibroblasts. Hyperoxia-induced gene expression signatures in human neonatal fibroblasts and alveolar epithelial cells in vitro resembled mouse scRNA-seq data. These findings suggest that neonatal exposure to hyperoxia programs distinct sex-specific stem cell progenitor and cellular reparative responses that underpin lung remodeling in BPD.
Collapse
Affiliation(s)
- Sheng Xia
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Lisandra Vila Ellis
- Department of Pulmonary Medicine, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Konner Winkley
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
| | - Heather Menden
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Sherry M Mabry
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Aparna Venkatraman
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Daniel Louiselle
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
| | - Margaret Gibson
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
| | - Elin Grundberg
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
- Children's Mercy Research Institute, Kansas City, Missouri
| | - Jichao Chen
- Department of Pulmonary Medicine, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Venkatesh Sampath
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
- Children's Mercy Research Institute, Kansas City, Missouri
| |
Collapse
|
38
|
Apeldoorn C, Safaei S, Paton J, Maso Talou GD. Computational models for generating microvascular structures: Investigations beyond medical imaging resolution. WIREs Mech Dis 2023; 15:e1579. [PMID: 35880683 PMCID: PMC10077909 DOI: 10.1002/wsbm.1579] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/22/2022] [Accepted: 06/29/2022] [Indexed: 01/31/2023]
Abstract
Angiogenesis, arteriogenesis, and pruning are revascularization processes essential to our natural vascular development and adaptation, as well as central players in the onset and development of pathologies such as tumoral growth and stroke recovery. Computational modeling allows for repeatable experimentation and exploration of these complex biological processes. In this review, we provide an introduction to the biological understanding of the vascular adaptation processes of sprouting angiogenesis, intussusceptive angiogenesis, anastomosis, pruning, and arteriogenesis, discussing some of the more significant contributions made to the computational modeling of these processes. Each computational model represents a theoretical framework for how biology functions, and with rises in computing power and study of the problem these frameworks become more accurate and complete. We highlight physiological, pathological, and technological applications that can be benefit from the advances performed by these models, and we also identify which elements of the biology are underexplored in the current state-of-the-art computational models. This article is categorized under: Cancer > Computational Models Cardiovascular Diseases > Computational Models.
Collapse
Affiliation(s)
- Cameron Apeldoorn
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Soroush Safaei
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Julian Paton
- Cardiovascular Autonomic Research Cluster, Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Gonzalo D Maso Talou
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
39
|
Bradbury JJ, Lovegrove HE, Giralt-Pujol M, Herbert SP. Analysis of mRNA Subcellular Distribution in Collective Cell Migration. Methods Mol Biol 2023; 2608:389-407. [PMID: 36653719 DOI: 10.1007/978-1-0716-2887-4_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The movement of groups of cells by collective cell migration requires division of labor between group members. Therefore, distinct cell identities, unique cell behaviors, and specific cellular roles are acquired by cells undergoing collective movement. A key driving force behind the acquisition of discrete cell states is the precise control of where, when, and how genes are expressed, both at the subcellular and supracellular level. Unraveling the mechanisms underpinning the spatiotemporal control of gene expression in collective cell migration requires not only suitable experimental models but also high-resolution imaging of messenger RNA and protein localization during this process. In recent times, the highly stereotyped growth of new blood vessels by sprouting angiogenesis has become a paradigm for understanding collective cell migration, and consequently this has led to the development of numerous user-friendly in vitro models of angiogenesis. In parallel, single-molecule fluorescent in situ hybridization (smFISH) has come to the fore as a powerful technique that allows quantification of both RNA number and RNA spatial distribution in cells and tissues. Moreover, smFISH can be combined with immunofluorescence to understand the precise interrelationship between RNA and protein distribution. Here, we describe methods for use of smFISH and immunofluorescence microscopy in in vitro angiogenesis models to enable the investigation of RNA and protein expression and localization during endothelial collective cell migration.
Collapse
Affiliation(s)
- Joshua J Bradbury
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Holly E Lovegrove
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Marta Giralt-Pujol
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Shane P Herbert
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
40
|
Endothelial mechanosensing: A forgotten target to treat vascular remodeling in hypertension? Biochem Pharmacol 2022; 206:115290. [DOI: 10.1016/j.bcp.2022.115290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/23/2022]
|
41
|
Barrasa-Ramos S, Dessalles CA, Hautefeuille M, Barakat AI. Mechanical regulation of the early stages of angiogenesis. J R Soc Interface 2022; 19:20220360. [PMID: 36475392 PMCID: PMC9727679 DOI: 10.1098/rsif.2022.0360] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Favouring or thwarting the development of a vascular network is essential in fields as diverse as oncology, cardiovascular disease or tissue engineering. As a result, understanding and controlling angiogenesis has become a major scientific challenge. Mechanical factors play a fundamental role in angiogenesis and can potentially be exploited for optimizing the architecture of the resulting vascular network. Largely focusing on in vitro systems but also supported by some in vivo evidence, the aim of this Highlight Review is dual. First, we describe the current knowledge with particular focus on the effects of fluid and solid mechanical stimuli on the early stages of the angiogenic process, most notably the destabilization of existing vessels and the initiation and elongation of new vessels. Second, we explore inherent difficulties in the field and propose future perspectives on the use of in vitro and physics-based modelling to overcome these difficulties.
Collapse
Affiliation(s)
- Sara Barrasa-Ramos
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Claire A. Dessalles
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR7622), Institut de Biologie Paris Seine, Sorbonne Université, Paris, France,Facultad de Ciencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Abdul I. Barakat
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| |
Collapse
|
42
|
Bautch VL, Mukouyama YS. The Beauty and Complexity of Blood Vessel Patterning. Cold Spring Harb Perspect Med 2022; 12:a041167. [PMID: 35379659 PMCID: PMC9619359 DOI: 10.1101/cshperspect.a041167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
This review highlights new concepts in vascular patterning in the last 10 years, with emphasis on its beauty and complexity. Endothelial cell signaling pathways that respond to molecular or mechanical signals are described, and examples of vascular patterning that use these pathways in brain, skin, heart, and kidney are highlighted. The pathological consequences of patterning loss are discussed in the context of arteriovenous malformations (AVMs), and prospects for the next 10 years presented.
Collapse
Affiliation(s)
- Victoria L Bautch
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Yoh-Suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Development Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
43
|
Tregub PP, Averchuk AS, Baranich TI, Ryazanova MV, Salmina AB. Physiological and Pathological Remodeling of Cerebral Microvessels. Int J Mol Sci 2022; 23:12683. [PMID: 36293539 PMCID: PMC9603917 DOI: 10.3390/ijms232012683] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/10/2022] [Accepted: 10/18/2022] [Indexed: 11/13/2022] Open
Abstract
There is growing evidence that the remodeling of cerebral microvessels plays an important role in plastic changes in the brain associated with development, experience, learning, and memory consolidation. At the same time, abnormal neoangiogenesis, and deregulated regulation of microvascular regression, or pruning, could contribute to the pathogenesis of neurodevelopmental diseases, stroke, and neurodegeneration. Aberrant remodeling of microvesselsis associated with blood-brain barrier breakdown, development of neuroinflammation, inadequate microcirculation in active brain regions, and leads to the dysfunction of the neurovascular unit and progressive neurological deficits. In this review, we summarize current data on the mechanisms of blood vessel regression and pruning in brain plasticity and in Alzheimer's-type neurodegeneration. We discuss some novel approaches to modulating cerebral remodeling and preventing degeneration-coupled aberrant microvascular activity in chronic neurodegeneration.
Collapse
Affiliation(s)
- Pavel P. Tregub
- Federal State Budgetary Scientific Institution Research Center of Neurology, 125367 Moscow, Russia
| | | | | | | | | |
Collapse
|
44
|
Barbacena P, Dominguez-Cejudo M, Fonseca CG, Gómez-González M, Faure LM, Zarkada G, Pena A, Pezzarossa A, Ramalho D, Giarratano Y, Ouarné M, Barata D, Fortunato IC, Misikova LH, Mauldin I, Carvalho Y, Trepat X, Roca-Cusachs P, Eichmann A, Bernabeu MO, Franco CA. Competition for endothelial cell polarity drives vascular morphogenesis in the mouse retina. Dev Cell 2022; 57:2321-2333.e9. [PMID: 36220082 PMCID: PMC9552591 DOI: 10.1016/j.devcel.2022.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 07/15/2022] [Accepted: 09/07/2022] [Indexed: 12/02/2022]
Abstract
Blood-vessel formation generates unique vascular patterns in each individual. The principles governing the apparent stochasticity of this process remain to be elucidated. Using mathematical methods, we find that the transition between two fundamental vascular morphogenetic programs-sprouting angiogenesis and vascular remodeling-is established by a shift of collective front-to-rear polarity of endothelial cells in the mouse retina. We demonstrate that the competition between biochemical (VEGFA) and mechanical (blood-flow-induced shear stress) cues controls this collective polarity shift. Shear stress increases tension at focal adhesions overriding VEGFA-driven collective polarization, which relies on tension at adherens junctions. We propose that vascular morphogenetic cues compete to regulate individual cell polarity and migration through tension shifts that translates into tissue-level emergent behaviors, ultimately leading to uniquely organized vascular patterns.
Collapse
Affiliation(s)
- Pedro Barbacena
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Dominguez-Cejudo
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Catarina G Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Manuel Gómez-González
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
| | - Laura M Faure
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
| | - Georgia Zarkada
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Andreia Pena
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Anna Pezzarossa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; Champalimaud Foundation, Champalimaud Research, Lisbon, Portugal
| | - Daniela Ramalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ylenia Giarratano
- Centre for Medical Informatics, Usher Institute, The University of Edinburgh, Edinburgh, UK
| | - Marie Ouarné
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - David Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Isabela C Fortunato
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Lenka Henao Misikova
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ian Mauldin
- Centre for Medical Informatics, Usher Institute, The University of Edinburgh, Edinburgh, UK; School of Informatics, The University of Edinburgh, Edinburgh, UK
| | - Yulia Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain; Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain; Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Anne Eichmann
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA; Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA; Université de Paris, PARCC, INSERM, 75006 Paris, France
| | - Miguel O Bernabeu
- Centre for Medical Informatics, Usher Institute, The University of Edinburgh, Edinburgh, UK; The Bayes Centre, The University of Edinburgh, Edinburgh, UK
| | - Cláudio A Franco
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; Universidade Católica Portuguesa, Católica Medical School, Católica Biomedical Research Centre, Lisbon, Portugal.
| |
Collapse
|
45
|
Arthur HM, Roman BL. An update on preclinical models of hereditary haemorrhagic telangiectasia: Insights into disease mechanisms. Front Med (Lausanne) 2022; 9:973964. [PMID: 36250069 PMCID: PMC9556665 DOI: 10.3389/fmed.2022.973964] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
Endoglin (ENG) is expressed on the surface of endothelial cells (ECs) where it efficiently binds circulating BMP9 and BMP10 ligands to initiate activin A receptor like type 1 (ALK1) protein signalling to protect the vascular architecture. Patients heterozygous for ENG or ALK1 mutations develop the vascular disorder known as hereditary haemorrhagic telangiectasia (HHT). Many patients with this disorder suffer from anaemia, and are also at increased risk of stroke and high output heart failure. Recent work using animal models of HHT has revealed new insights into cellular and molecular mechanisms causing this disease. Loss of the ENG (HHT1) or ALK1 (HHT2) gene in ECs leads to aberrant arteriovenous connections or malformations (AVMs) in developing blood vessels. Similar phenotypes develop following combined EC specific loss of SMAD1 and 5, or EC loss of SMAD4. Taken together these data point to the essential role of the BMP9/10-ENG-ALK1-SMAD1/5-SMAD4 pathway in protecting the vasculature from AVMs. Altered directional migration of ECs in response to shear stress and increased EC proliferation are now recognised as critical factors driving AVM formation. Disruption of the ENG/ALK1 signalling pathway also affects EC responses to vascular endothelial growth factor (VEGF) and crosstalk between ECs and vascular smooth muscle cells. It is striking that the vascular lesions in HHT are both localised and tissue specific. Increasing evidence points to the importance of a second genetic hit to generate biallelic mutations, and the sporadic nature of such somatic mutations would explain the localised formation of vascular lesions. In addition, different pro-angiogenic drivers of AVM formation are likely to be at play during the patient’s life course. For example, inflammation is a key driver of vessel remodelling in postnatal life, and may turn out to be an important driver of HHT disease. The current wealth of preclinical models of HHT has led to increased understanding of AVM development and revealed new therapeutic approaches to treat AVMs, and form the topic of this review.
Collapse
Affiliation(s)
- Helen M. Arthur
- Biosciences Institute, Centre for Life, University of Newcastle, Newcastle, United Kingdom
- *Correspondence: Helen M. Arthur,
| | - Beth L. Roman
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
46
|
Pibouin-Fragner L, Eichmann A, Pardanaud L. Environmental and intrinsic modulations of venous differentiation. Cell Mol Life Sci 2022; 79:491. [PMID: 35987946 PMCID: PMC11072674 DOI: 10.1007/s00018-022-04470-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 06/16/2022] [Accepted: 07/05/2022] [Indexed: 11/03/2022]
Abstract
Endothelial cells in veins differ in morphology, function and gene expression from those in arteries and lymphatics. Understanding how venous and arterial identities are induced during development is required to understand how arterio-venous malformations occur, and to improve the outcome of vein grafts in surgery by promoting arterialization of veins. To identify factors that promote venous endothelial cell fate in vivo, we isolated veins from quail embryos, at different developmental stages, that were grafted into the coelom of chick embryos. Endothelial cells migrated out from the grafted vein and their colonization of host veins and/or arteries was quantified. We show that venous fate is promoted by sympathetic vessel innervation at embryonic day 11. Removal of sympathetic innervation decreased vein colonization, while norepinephrine enhanced venous colonization. BMP treatment or inhibition of ERK enhanced venous fate, revealing environmental neurotransmitter and BMP signaling and intrinsic ERK inhibition as actors in venous fate acquisition. We also identify the BMP antagonist Noggin as a potent mediator of venous arterialization.
Collapse
Affiliation(s)
| | - Anne Eichmann
- Université de Paris Cité, Inserm, PARCC, 75015, Paris, France.
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT, USA.
| | - Luc Pardanaud
- Université de Paris Cité, Inserm, PARCC, 75015, Paris, France.
| |
Collapse
|
47
|
Orlova VV, Nahon DM, Cochrane A, Cao X, Freund C, van den Hil F, Westermann CJJ, Snijder RJ, Ploos van Amstel JK, Ten Dijke P, Lebrin F, Mager HJ, Mummery CL. Vascular defects associated with hereditary hemorrhagic telangiectasia revealed in patient-derived isogenic iPSCs in 3D vessels on chip. Stem Cell Reports 2022; 17:1536-1545. [PMID: 35777360 PMCID: PMC9287680 DOI: 10.1016/j.stemcr.2022.05.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/31/2022] [Accepted: 05/31/2022] [Indexed: 11/30/2022] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is a genetic disease characterized by weak blood vessels. HHT1 is caused by mutations in the ENDOGLIN (ENG) gene. Here, we generated induced pluripotent stem cells (hiPSCs) from a patient with rare mosaic HHT1 with tissues containing both mutant (ENGc.1678C>T) and normal cells, enabling derivation of isogenic diseased and healthy hiPSCs, respectively. We showed reduced ENG expression in HHT1 endothelial cells (HHT1-hiPSC-ECs), reflecting haploinsufficiency. HHT1c.1678C>T-hiPSC-ECs and the healthy isogenic control behaved similarly in two-dimensional (2D) culture, forming functionally indistinguishable vascular networks. However, when grown in 3D organ-on-chip devices under microfluidic flow, lumenized vessels formed in which defective vascular organization was evident: interaction between inner ECs and surrounding pericytes was decreased, and there was evidence for vascular leakage. Organs on chip thus revealed features of HHT in hiPSC-derived blood vessels that were not evident in conventional 2D assays. Vessels from isogenic hiPSCs from HHT1 patients compared HHT1-hiPSC-ECs show defective vascular organization in 3D microfluidic chips HHT1-hiPSC-ECs show defective EC-pericyte interaction
Collapse
Affiliation(s)
- Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333ZA, the Netherlands.
| | - Dennis M Nahon
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333ZA, the Netherlands
| | - Amy Cochrane
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333ZA, the Netherlands
| | - Xu Cao
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333ZA, the Netherlands
| | - Christian Freund
- Department of Anatomy and Embryology and Human iPSC Hotel, Leiden University Medical Center, Leiden 2333ZA, the Netherlands
| | - Francijna van den Hil
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333ZA, the Netherlands
| | | | | | | | - Peter Ten Dijke
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden 2333ZA, the Netherlands
| | - Franck Lebrin
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands; INSERM U1273, ESPCI, CNRS FRE 2031, Paris, France
| | | | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden 2333ZA, the Netherlands; Department of Anatomy and Embryology and Human iPSC Hotel, Leiden University Medical Center, Leiden 2333ZA, the Netherlands.
| |
Collapse
|
48
|
D'Amico G, Fernandez I, Gómez-Escudero J, Kim H, Maniati E, Azman MS, Mardakheh FK, Serrels B, Serrels A, Parsons M, Squire A, Birdsey GM, Randi AM, Bolado-Carrancio A, Gangeswaran R, Reynolds LE, Bodrug N, Wang Y, Wang J, Meier P, Hodivala-Dilke KM. ERG activity is regulated by endothelial FAK coupling with TRIM25/USP9x in vascular patterning. Development 2022; 149:dev200528. [PMID: 35723257 PMCID: PMC9340553 DOI: 10.1242/dev.200528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/29/2022] [Indexed: 11/20/2022]
Abstract
Precise vascular patterning is crucial for normal growth and development. The ERG transcription factor drives Delta-like ligand 4 (DLL4)/Notch signalling and is thought to act as a pivotal regulator of endothelial cell (EC) dynamics and developmental angiogenesis. However, molecular regulation of ERG activity remains obscure. Using a series of EC-specific focal adhesion kinase (FAK)-knockout (KO) and point-mutant FAK-knock-in mice, we show that loss of ECFAK, its kinase activity or phosphorylation at FAK-Y397, but not FAK-Y861, reduces ERG and DLL4 expression levels together with concomitant aberrations in vascular patterning. Rapid immunoprecipitation mass spectrometry of endogenous proteins identified that endothelial nuclear-FAK interacts with the deubiquitinase USP9x and the ubiquitin ligase TRIM25. Further in silico analysis confirms that ERG interacts with USP9x and TRIM25. Moreover, ERG levels are reduced in FAKKO ECs via a ubiquitin-mediated post-translational modification programme involving USP9x and TRIM25. Re-expression of ERG in vivo and in vitro rescues the aberrant vessel-sprouting defects observed in the absence of ECFAK. Our findings identify ECFAK as a regulator of retinal vascular patterning by controlling ERG protein degradation via TRIM25/USP9x.
Collapse
Affiliation(s)
- Gabriela D'Amico
- Centre for Tumour Microenvironment, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Isabelle Fernandez
- Centre for Tumour Microenvironment, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Jesús Gómez-Escudero
- Centre for Tumour Microenvironment, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Hyojin Kim
- The Breakthrough Toby Robins Breast Cancer Research Centre, Institute of Cancer Research, Chester Beatty Laboratories, Fulham Road, London SW3 6JB, UK
| | - Eleni Maniati
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Muhammad Syahmi Azman
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Faraz K. Mardakheh
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Bryan Serrels
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden G61 1QH, UK
| | - Alan Serrels
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh BioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Maddy Parsons
- Kings College London, Randall Centre of Cell and Molecular Biophysics, Room 3.22B, New Hunts House, Guys Campus, London SE1 1UL, UK
| | - Anthony Squire
- IMCES - Imaging Centre Essen, Institute for Experimental Immunology and Imaging, University Clinic Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Graeme M. Birdsey
- National Heart & Lung Institute, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Anna M. Randi
- National Heart & Lung Institute, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | | | - Rathi Gangeswaran
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Louise E. Reynolds
- Centre for Tumour Microenvironment, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Natalia Bodrug
- Centre for Tumour Microenvironment, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Yaohe Wang
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Jun Wang
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Pascal Meier
- The Breakthrough Toby Robins Breast Cancer Research Centre, Institute of Cancer Research, Chester Beatty Laboratories, Fulham Road, London SW3 6JB, UK
| | - Kairbaan M. Hodivala-Dilke
- Centre for Tumour Microenvironment, Barts Cancer Institute – a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
49
|
Edgar LT, Park H, Crawshaw JR, Osborne JM, Eichmann A, Bernabeu MO. Traffic Patterns of the Migrating Endothelium: How Force Transmission Regulates Vascular Malformation and Functional Shunting During Angiogenic Remodelling. Front Cell Dev Biol 2022; 10:840066. [PMID: 35663401 PMCID: PMC9160721 DOI: 10.3389/fcell.2022.840066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
Angiogenesis occurs in distinct phases: initial spouting is followed by remodelling in which endothelial cells (ECs) composing blood vessels rearrange by migrating against the direction of flow. Abnormal remodelling can result in vascular malformation. Such is the case in mutation of the Alk1 receptor within the mouse retina which disrupts flow-migration coupling, creating mixed populations of ECs polarised with/against flow which aggregate into arteriovenous malformations (AVMs). The lack of live imaging options in vivo means that the collective EC dynamics that drive AVM and the consequences of mixed populations of polarity remain a mystery. Therefore, our goal is to present a novel agent-based model to provide theoretical insight into EC force transmission and collective dynamics during angiogenic remodelling. Force transmission between neighbouring agents consists of extrusive forces which maintain spacing and cohesive forces which maintain the collective. We performed migration simulations within uniformly polarised populations (against flow) and mixed polarity (with/against flow). Within uniformly polarised populations, extrusive forces stabilised the plexus by facilitating EC intercalation which ensures that cells remained evenly distributed. Excess cohesion disrupts intercalation, resulting in aggregations of cells and functional shunting. Excess cohesion between ECs prevents them from resolving diameter balances within the plexus, leading to prolonged flow reversals which exert a critical behaviour change within the system as they switch the direction of cell migration and traffic patterns at bifurcations. Introducing mixtures of cell polarity dramatically changed the role of extrusive forces within the system. At low extrusion, opposing ECs were able to move past each other; however, at high extrusion the pushing between cells resulted in migration speeds close to zero, forming traffic jams and disrupting migration. In our study, we produced vascular malformations and functional shunting with either excess cohesion between ECs or mixtures of cell polarity. At the centre of both these mechanisms are cell-cell adherens junctions, which are involved in flow sensing/polarity and must remodelling dynamically to allow rearrangements of cells during vascular patterning. Thus, our findings implicate junctional dysfunction as a new target in the treatment and prevention of vascular disease and AVMs.
Collapse
Affiliation(s)
- Lowell T. Edgar
- Centre for Medical Informatics, Usher Institute, The University of Edinburgh, Edinburgh, United Kingdom
- *Correspondence: Lowell T. Edgar, ; Miguel O. Bernabeu,
| | - Hyojin Park
- Cardiovascular Research Center Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Jessica R. Crawshaw
- School of Mathematics and Statistics, University of Melbourne, Melbourne, VIC, Australia
| | - James M. Osborne
- School of Mathematics and Statistics, University of Melbourne, Melbourne, VIC, Australia
| | - Anne Eichmann
- Cardiovascular Research Center Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
- Yale University School of Medicine, Department of Cellular and Molecular Physiology, New Haven, CT, United States
- Université de Paris, PARCC, INSERM, Paris, France
| | - Miguel O. Bernabeu
- Centre for Medical Informatics, Usher Institute, The University of Edinburgh, Edinburgh, United Kingdom
- The Bayes Centre, The University of Edinburgh, Edinburgh, United Kingdom
- *Correspondence: Lowell T. Edgar, ; Miguel O. Bernabeu,
| |
Collapse
|
50
|
Bats ML, Peghaire C, Delobel V, Dufourcq P, Couffinhal T, Duplàa C. Wnt/frizzled Signaling in Endothelium: A Major Player in Blood-Retinal- and Blood-Brain-Barrier Integrity. Cold Spring Harb Perspect Med 2022; 12:a041219. [PMID: 35074794 PMCID: PMC9121893 DOI: 10.1101/cshperspect.a041219] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The Wnt/frizzled signaling pathway is one of the major regulators of endothelial biology, controlling key cellular activities. Many secreted Wnt ligands have been identified and can initiate diverse signaling via binding to a complex set of Frizzled (Fzd) transmembrane receptors and coreceptors. Roughly, Wnt signaling is subdivided into two pathways: the canonical Wnt/β-catenin signaling pathway whose main downstream effector is the transcriptional coactivator β-catenin, and the noncanonical Wnt signaling pathway, which is subdivided into the Wnt/Ca2+ pathway and the planar cell polarity pathway. Here, we will focus on its cross talk with other angiogenic pathways and on its role in blood-retinal- and blood-brain-barrier formation and its maintenance in a differentiated state. We will unravel how retinal vascular pathologies and neurovascular degenerative diseases result from disruption of the Wnt pathway related to vascular instability, and highlight current research into therapeutic options.
Collapse
Affiliation(s)
- Marie-Lise Bats
- Univ. Bordeaux, Inserm, UMR1034, Biology of Cardiovascular Diseases, F-33600 Pessac, France
- Department of Biochemistry, Pellegrin Hospital, University Hospital of Bordeaux, 33076 Bordeaux Cedex, France
| | - Claire Peghaire
- Univ. Bordeaux, Inserm, UMR1034, Biology of Cardiovascular Diseases, F-33600 Pessac, France
| | - Valentin Delobel
- Univ. Bordeaux, Inserm, UMR1034, Biology of Cardiovascular Diseases, F-33600 Pessac, France
| | - Pascale Dufourcq
- Univ. Bordeaux, Inserm, UMR1034, Biology of Cardiovascular Diseases, F-33600 Pessac, France
| | - Thierry Couffinhal
- Univ. Bordeaux, Inserm, UMR1034, Biology of Cardiovascular Diseases, F-33600 Pessac, France
- Centre d'exploration, de prévention et de traitement de l'athérosclérose (CEPTA), CHU Bordeaux, 33000 Bordeaux, France
| | - Cécile Duplàa
- Univ. Bordeaux, Inserm, UMR1034, Biology of Cardiovascular Diseases, F-33600 Pessac, France
| |
Collapse
|