1
|
Broer T, Tsintolas N, Purkey K, Hammond S, DeLuca S, Wu T, Gupta I, Khodabukus A, Bursac N. Engineered myovascular tissues for studies of endothelial/satellite cell interactions. Acta Biomater 2024; 188:65-78. [PMID: 39299621 PMCID: PMC11486565 DOI: 10.1016/j.actbio.2024.09.020] [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: 04/15/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
In native skeletal muscle, capillaries reside in close proximity to muscle stem cells (satellite cells, SCs) and regulate SC numbers and quiescence through partially understood mechanisms that are difficult to study in vivo. This challenge could be addressed by the development of a 3-dimensional (3D) in vitro model of vascularized skeletal muscle harboring both a pool of quiescent SCs and a robust network of capillaries. Still, studying interactions between SCs and endothelial cells (ECs) within a tissue-engineered muscle environment has been hampered by the incompatibility of commercially available EC media with skeletal muscle differentiation. In this study, we first optimized co-culture media and cellular ratios to generate highly functional vascularized human skeletal muscle tissues ("myovascular bundles") with contractile properties (∼10 mN/mm2) equaling those of avascular, muscle-only tissues ("myobundles"). Within one week of muscle differentiation, ECs in these tissues formed a dense network of capillaries that co-aligned with muscle fibers and underwent initial lumenization. Incorporating vasculature within myobundles increased the total SC number by 82%, with SC density and quiescent signature being increased proximal (≤20μm) to EC networks. In vivo, at two weeks post-implantation into dorsal window chambers in nude mice, vascularized myobundles exhibited improved calcium handling compared to avascular implants. In summary, we engineered highly functional myovascular tissues that enable studies of the roles of EC-SC crosstalk in human muscle development, physiology, and disease. STATEMENT OF SIGNIFICANCE: In native skeletal muscle, intricate relationships between vascular cells and muscle stem cells ("satellite cells") play critical roles in muscle growth and regeneration. Current methods for in vitro engineering of contractile skeletal muscle do not recreate capillary networks present in vivo. Our study for the first time generates in vitro robustly vascularized, highly functional engineered human skeletal muscle tissues. Within these tissues, satellite cells are more abundant and, similar to in vivo, they are more dense and less proliferative proximal to endothelial cells. Upon implantation in mice, vascularized engineered muscles show improved calcium handling compared to muscle-only implants. We expect that this versatile in vitro system will enable studies of muscle-vasculature crosstalk in human development and disease.
Collapse
Affiliation(s)
- Torie Broer
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA
| | - Nick Tsintolas
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA
| | - Karly Purkey
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA
| | - Stewart Hammond
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA
| | - Sophia DeLuca
- Department of Cell Biology, Duke University, Durham, NC 27708, USA
| | - Tianyu Wu
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA
| | - Ishika Gupta
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA
| | - Alastair Khodabukus
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA.
| |
Collapse
|
2
|
Kartal B, Alimogullari E, Elçi P, Fatsa T, Ören S. The effects of Quercetin on wound healing in the human umbilical vein endothelial cells. Cell Tissue Bank 2024; 25:851-860. [PMID: 38944663 DOI: 10.1007/s10561-024-10144-1] [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: 12/10/2022] [Accepted: 06/12/2024] [Indexed: 07/01/2024]
Abstract
An injury that affects the integrity of the skin, either inside or externally, is called a wound. Damaged tissue is repaired by a set of cellular and molecular mechanisms known as wound healing. Quercetin, a naturally occurring flavonoid, may hasten the healing of wounds. The study's objective was to investigate any potential impacts of quercetin on the wound-healing process. Human umbilical vein endothelial cells (HUVECs) were treated to varying dose ranges of quercetin (5-320 nM) for 24 and 48 h. Cultured cells were evaluated by using the MTT analysis, wound scratch assay and vascular tube formation. Furthermore the gene expression of VEGF and FGF were evaluated by qRT-PCR to determine the effects of quercetin on angiogenezis and wound repair. Positive effects of quercetin on cellular viability were demonstrated by the MTT experiment. In HUVECs quercetin promoted tube formation, migration, and proliferation while also averting wound breakage. Moreover, quercetin increased the expression of the FGF and VEGF genes, which aid in the healing of wounds in HUVECs. Quercetin may be bioactive molecule that successfully speeds up wound healing by regulating the vasculogenezis and healing cells.
Collapse
Affiliation(s)
- Bahar Kartal
- Ankara Yıldırım Beyazıt Üniversitesi: Ankara Yildirim Beyazit Universitesi Ankara, Çankaya, Turkey.
| | - Ebru Alimogullari
- Ankara Yıldırım Beyazıt Üniversitesi: Ankara Yildirim Beyazit Universitesi Ankara, Çankaya, Turkey
| | - Pınar Elçi
- Ankara Yıldırım Beyazıt Üniversitesi: Ankara Yildirim Beyazit Universitesi Ankara, Çankaya, Turkey
| | - Tugba Fatsa
- Ankara Yıldırım Beyazıt Üniversitesi: Ankara Yildirim Beyazit Universitesi Ankara, Çankaya, Turkey
| | - Sema Ören
- Ankara Yıldırım Beyazıt Üniversitesi: Ankara Yildirim Beyazit Universitesi Ankara, Çankaya, Turkey
| |
Collapse
|
3
|
Zhong K, Luo W, Li N, Tan X, Li Y, Yin S, Huang Y, Fang L, Ma W, Cai Y, Yin Y. CDK12 regulates angiogenesis of advanced prostate cancer by IGFBP3. Int J Oncol 2024; 64:20. [PMID: 38186306 PMCID: PMC10783938 DOI: 10.3892/ijo.2024.5608] [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: 06/10/2023] [Accepted: 11/16/2023] [Indexed: 01/09/2024] Open
Abstract
Prostate cancer (PCa) is a prevalent malignancy among men, with a majority of patients presenting with distant metastases at the time of initial diagnosis. These patients are at a heightened risk of developing more aggressive castration‑resistant PCa following androgen deprivation therapy, which poses a greater challenge for treatment. Notably, the inhibition of tumor angiogenesis should not be considered an ineffective treatment strategy. The regulatory role of CDK12 in transcriptional and post‑transcriptional processes is essential for the proper functioning of various cellular processes. In the present study, the expression of CDK12 was first knocked down in cells using CRISPR or siRNA technology. Subsequently, RNA‑seq analysis, co‑immunoprecipitation, western blotting, reverse transcription‑quantitative polymerase chain reaction and the LinkedOmics database were employed to reveal that CDK12 inhibits insulin like growth factor binding protein 3 (IGFBP3). Western blot analysis also demonstrated that CDK12 promoted VEGFA expression by inhibiting IGFBP3, which involves the Akt signaling pathway. Then, CDK12 was found to promote PCa cell proliferation, cell migration and angiogenesis by inhibiting IGFBP3 through cell proliferation assays, cell migration assays and tube formation assays, respectively. Finally, animal experiments were performed for in vivo validation. It was concluded that CDK12 promoted PCa and its angiogenesis by inhibiting IGFBP3.
Collapse
Affiliation(s)
- Kun Zhong
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Wenwu Luo
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Nan Li
- Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Xin Tan
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yuqing Li
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Shiyuan Yin
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yuhang Huang
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Linna Fang
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Wei Ma
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yongping Cai
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yu Yin
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
4
|
Chayaburakul K, Ong WY, Herr DR, Kobutree P, Chantra K. Differences in the ultrastructure of neurons in the spinal ganglion and dorsal rootlet between rats treated with cisplatin only versus co-administration with a sphingosine 1-phosphate receptor 2 agonist in attenuating neuropathy and allodynia. J Peripher Nerv Syst 2023; 28:476-489. [PMID: 37483146 DOI: 10.1111/jns.12582] [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: 09/19/2022] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/25/2023]
Abstract
BACKGROUND AND AIMS Cisplatin is a chemotherapeutic agent for many types of cancer. The neurotoxicity of cisplatin includes neuropathy and allodynia. We aimed to study structural changes by using CYM54-78, attenuating cisplatin-induced neuropathy and blocking the pathogenesis in neurons, and promoting axonal regeneration. METHODS TEM (transmission electron microscopy) was used to distinguish ultrastructural changes in dorsal root ganglion (DRG) and dorsal rootlets (DR) between rats treated with cisplatin alone and rats co-treated with cisplatin and sphingosine -1-phosphate receptor2 (S1P2) agonist, CYM-5478. RESULTS In DRG of rats treated with cisplatin alone, TEM micrographs showed necrosis and apoptotic cells. Neuronal cytoplasm showed numerous vacuole (stage C) and swelling (stage B➔C) mitochondrial degeneration. Neurons in DRG from cisplatin+CYM-5478 group showed a higher percentage of healthy mitochondria (from 5.3% to 75.6%) than those treated with cisplatin alone. DR of cisplatin only group showed abnormal axoplasm, axolemma, and focal detached myelin sheaths, especially in Aδ (fast pain) and Aβ (touch) fibers, and revealed collateral branches that sprouted from Aβ fibers, which is characteristic of allodynia. Moreover, vasoconstriction was observed in DRG and DR. Rats in cisplatin+CYM-5478 group showed not only fewer abnormal structures than those in cisplatin only group, but also showed Bands of Büngner and onion bulb-like structures, which are characteristic of nerve regeneration. INTERPRETATION Together with our previous study, showed that CYM-5478 attenuated neuropathy and allodynia in a rat model of cisplatin-induced neuropathy, these results suggest S1P2 agonists as a potential approach the for treatment of cancer due to the reduction of side effects of cisplatin.
Collapse
Affiliation(s)
| | - Wei Yi Ong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore, Singapore
| | - Deron R Herr
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore, Singapore
| | - Phetnarin Kobutree
- Anatomy Unit, Faculty of Science, Rangsit University, Meung, Pathum Tani, Thailand
| | - Kraisri Chantra
- Department of Neurosurgery, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
5
|
Abstract
The vasculature consists of vessels of different sizes that are arranged in a hierarchical pattern. Two cell populations work in concert to establish this pattern during embryonic development and adopt it to changes in blood flow demand later in life: endothelial cells that line the inner surface of blood vessels, and adjacent vascular mural cells, including smooth muscle cells and pericytes. Despite recent progress in elucidating the signalling pathways controlling their crosstalk, much debate remains with regard to how mural cells influence endothelial cell biology and thereby contribute to the regulation of blood vessel formation and diameters. In this Review, I discuss mural cell functions and their interactions with endothelial cells, focusing on how these interactions ensure optimal blood flow patterns. Subsequently, I introduce the signalling pathways controlling mural cell development followed by an overview of mural cell ontogeny with an emphasis on the distinguishing features of mural cells located on different types of blood vessels. Ultimately, I explore therapeutic strategies involving mural cells to alleviate tissue ischemia and improve vascular efficiency in a variety of diseases.
Collapse
Affiliation(s)
- Arndt F. Siekmann
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 1114 Biomedical Research Building, 421 Curie Boulevard, Philadelphia, PA 19104, USA
| |
Collapse
|
6
|
Kartal B, Alimogulları E, Elçi P, Fatsa T, Ören S. RETRACTED ARTICLE: The effects of quercetin on wound healing in the human umbilical vein endothelial cells. Cell Tissue Bank 2023; 24:387. [PMID: 36138273 DOI: 10.1007/s10561-022-10041-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/13/2022] [Indexed: 11/02/2022]
Affiliation(s)
- Bahar Kartal
- Ankara Yıldırım Beyazıt Üniversitesi, Çankaya, Ankara, Turkey.
| | | | - Pınar Elçi
- Ankara Yıldırım Beyazıt Üniversitesi, Çankaya, Ankara, Turkey
| | - Tugba Fatsa
- Ankara Yıldırım Beyazıt Üniversitesi, Çankaya, Ankara, Turkey
| | - Sema Ören
- Ankara Yıldırım Beyazıt Üniversitesi, Çankaya, Ankara, Turkey
| |
Collapse
|
7
|
PCSK9 Promotes Endothelial Dysfunction During Sepsis Via the TLR4/MyD88/NF-κB and NLRP3 Pathways. Inflammation 2023; 46:115-128. [PMID: 35930089 DOI: 10.1007/s10753-022-01715-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/23/2022] [Accepted: 07/08/2022] [Indexed: 11/05/2022]
Abstract
Endothelial dysfunction often accompanies sepsis. We aimed to explore the role of PCSK9 in septic endothelial dysfunction. Sepsis was induced by lipopolysaccharide (LPS) treatment of human umbilical vein endothelial cells (HUVECs) in vitro and cecal ligation and puncture (CLP) surgery in mice in vivo. Evolocumab (EVC) and Pep 2-8, PCSK9 inhibitors, were subsequently used to determine the role of PCSK9 in sepsis-induced endothelial dysfunction in vitro and in vivo, respectively. In addition, the TLR4 agonist, Kdo2-Lipid A ammonium (KLA), was used to determine the related mechanism. Protein expression of eNOS, VE-cadherin, PCSK9, TLR4, MyD88, p-p65, p65, NLRP3, ASC, and caspase-1 p20 in mice aortas and HUVECs was measured by western blotting, while mRNA expression of TNFα, IL-1β, and IL-18 was determined by qRT-PCR. The level of inflammatory cytokines of mouse aortas was visualized by immunohistochemistry. Vasodilation of the aorta was detected by vascular reactivity experiments. The 96-h survival rate after CLP was assessed. Our findings showed that the expression of eNOS and VE-cadherin decreased, and PCSK9 expression increased, in septic HUVECs or mice. Inhibition of PCSK9 increased eNOS and VE-cadherin expression. Activation of the TLR4/MyD88/NF-κB and NLRP3 pathways may be responsible for PCSK9-induced endothelial dysfunction in sepsis. Vascular reactivity tests and survival studies showed that inhibition of PCSK9 could prevent the decrease in endothelium-dependent vasodilation function and improve the survival rates of septic mice. In summary, our results suggested that increased PCSK9 expression during sepsis activated the TLR4/MyD88/NF-κB and NLRP3 pathways to induce inflammation, which resulted in vascular endothelial dysfunction and decreased survival rates. Thus, inhibition of PCSK9 may be a potential clinical therapeutic target to improve vascular endothelial function in sepsis.
Collapse
|
8
|
Cabral KA, Srivastava V, Graham AJ, Coyle MC, Stashko C, Weaver V, Gartner ZJ. Programming the Self-Organization of Endothelial Cells into Perfusable Microvasculature. Tissue Eng Part A 2023; 29:80-92. [PMID: 36181350 PMCID: PMC10266707 DOI: 10.1089/ten.tea.2022.0072] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 09/15/2022] [Indexed: 11/12/2022] Open
Abstract
The construction of three-dimensional (3D) microvascular networks with defined structures remains challenging. Emerging bioprinting strategies provide a means of patterning endothelial cells (ECs) into the geometry of 3D microvascular networks, but the microenvironmental cues necessary to promote their self-organization into cohesive and perfusable microvessels are not well known. To this end, we reconstituted microvessel formation in vitro by patterning thin lines of closely packed ECs fully embedded within a 3D extracellular matrix (ECM) and observed how different microenvironmental parameters influenced EC behaviors and their self-organization into microvessels. We found that the inclusion of fibrillar matrices, such as collagen I, into the ECM positively influenced cell condensation into extended geometries such as cords. We also identified the presence of a high-molecular-weight protein(s) in fetal bovine serum that negatively influenced EC condensation. This component destabilized cord structure by promoting cell protrusions and destabilizing cell-cell adhesions. Endothelial cords cultured in the presence of fibrillar collagen and in the absence of this protein activity were able to polarize, lumenize, incorporate mural cells, and support fluid flow. These optimized conditions allowed for the construction of branched and perfusable microvascular networks directly from patterned cells in as little as 3 days. These findings reveal important design principles for future microvascular engineering efforts based on bioprinting and micropatterning techniques. Impact statement Bioprinting is a potential strategy to achieve microvascularization in engineered tissues. However, the controlled self-organization of patterned endothelial cells into perfusable microvasculature remains challenging. We used DNA Programmed Assembly of Cells to create cell-dense, capillary-sized cords of endothelial cells with complete control over their structure. We optimized the matrix and media conditions to promote self-organization and maturation of these endothelial cords into stable and perfusable microvascular networks.
Collapse
Affiliation(s)
- Katelyn A. Cabral
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, Berkeley, California, USA
| | - Vasudha Srivastava
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| | - Austin J. Graham
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
- Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, California, USA
| | - Maxwell C. Coyle
- Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, California, USA
| | - Connor Stashko
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, Berkeley, California, USA
| | - Valerie Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Zev J. Gartner
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
- Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, California, USA
- Center for Cellular Construction, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
9
|
Yavvari P, Laporte A, Elomaa L, Schraufstetter F, Pacharzina I, Daberkow AD, Hoppensack A, Weinhart M. 3D-Cultured Vascular-Like Networks Enable Validation of Vascular Disruption Properties of Drugs In Vitro. Front Bioeng Biotechnol 2022; 10:888492. [PMID: 35769106 PMCID: PMC9234334 DOI: 10.3389/fbioe.2022.888492] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/13/2022] [Indexed: 02/06/2023] Open
Abstract
Vascular-disrupting agents are an interesting class of anticancer compounds because of their combined mode of action in preventing new blood vessel formation and disruption of already existing vasculature in the immediate microenvironment of solid tumors. The validation of vascular disruption properties of these drugs in vitro is rarely addressed due to the lack of proper in vitro angiogenesis models comprising mature and long-lived vascular-like networks. We herein report an indirect coculture model of human umbilical vein endothelial cells (HUVECs) and human dermal fibroblasts (HDFs) to form three-dimensional profuse vascular-like networks. HUVECs embedded and sandwiched in the collagen scaffold were cocultured with HDFs located outside the scaffold. The indirect coculture approach with the vascular endothelial growth factor (VEGF) producing HDFs triggered the formation of progressively maturing lumenized vascular-like networks of endothelial cells within less than 7 days, which have proven to be viably maintained in culture beyond day 21. Molecular weight-dependent Texas red-dextran permeability studies indicated high vascular barrier function of the generated networks. Their longevity allowed us to study the dose-dependent response upon treatment with the three known antiangiogenic and/or vascular disrupting agents brivanib, combretastatin A4 phosphate (CA4P), and 6´-sialylgalactose (SG) via semi-quantitative brightfield and qualitative confocal laser scanning microscopic (CLSM) image analysis. Compared to the reported data on in vivo efficacy of these drugs in terms of antiangiogenic and vascular disrupting effects, we observed similar trends with our 3D model, which are not reflected in conventional in vitro angiogenesis assays. High-vascular disruption under continuous treatment of the matured vascular-like network was observed at concentrations ≥3.5 ng·ml−1 for CA4P and ≥300 nM for brivanib. In contrast, SG failed to induce any significant vascular disruption in vitro. This advanced model of a 3D vascular-like network allows for testing single and combinational antiangiogenic and vascular disrupting effects with optimized dosing and may thus bridge the gap between the in vitro and in vivo experiments in validating hits from high-throughput screening. Moreover, the physiological 3D environment mimicking in vitro assay is not only highly relevant to in vivo studies linked to cancer but also to the field of tissue regeneration.
Collapse
Affiliation(s)
| | - Anna Laporte
- Institute of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, Hannover, Germany
| | - Laura Elomaa
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | | | - Inga Pacharzina
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | | | - Anke Hoppensack
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Marie Weinhart
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
- Institute of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, Hannover, Germany
- *Correspondence: Marie Weinhart, ,
| |
Collapse
|
10
|
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
|
11
|
Nawrot DA, Ozer LY, Al Haj Zen A. A Novel High Content Angiogenesis Assay Reveals That Lacidipine, L-Type Calcium Channel Blocker, Induces In Vitro Vascular Lumen Expansion. Int J Mol Sci 2022; 23:ijms23094891. [PMID: 35563280 PMCID: PMC9100973 DOI: 10.3390/ijms23094891] [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: 04/01/2022] [Revised: 04/23/2022] [Accepted: 04/26/2022] [Indexed: 11/21/2022] Open
Abstract
Angiogenesis is a critical cellular process toward establishing a functional circulatory system capable of delivering oxygen and nutrients to the tissue in demand. In vitro angiogenesis assays represent an important tool for elucidating the biology of blood vessel formation and for drug discovery applications. Herein, we developed a novel, high content 2D angiogenesis assay that captures endothelial morphogenesis’s cellular processes, including lumen formation. In this assay, endothelial cells form luminized vascular-like structures in 48 h. The assay was validated for its specificity and performance. Using the optimized assay, we conducted a phenotypic screen of a library containing 150 FDA-approved cardiovascular drugs to identify modulators of lumen formation. The screening resulted in several L-type calcium channel blockers being able to expand the lumen space compared to controls. Among these blockers, Lacidipine was selected for follow-up studies. We found that the endothelial cells treated with Lacidipine showed enhanced activity of caspase-3 in the luminal space. Pharmacological inhibition of caspase activity abolished the Lacidipine-enhancing effect on lumen formation, suggesting the involvement of apoptosis. Using a Ca2+ biosensor, we found that Lacipidine reduces the intracellular Ca2+ oscillations amplitude in the endothelial cells at the early stage, whereas Lacidipine blocks these Ca2+ oscillations completely at the late stage. The inhibition of MLCK exhibits a phenotype of lumen expansion similar to that of Lacidipine. In conclusion, this study describes a novel high-throughput phenotypic assay to study angiogenesis. Our findings suggest that calcium signalling plays an essential role during lumen morphogenesis. L-type Ca2+ channel blockers could be used for more efficient angiogenesis-mediated therapies.
Collapse
Affiliation(s)
- Dorota A. Nawrot
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK;
- Alzheimer’s Research UK, Oxford Drug Discovery Institute, Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK
| | - Lutfiye Yildiz Ozer
- College of Health and Life Sciences, Hamad Bin Khalifa University, Education City, Doha P.O. Box 34110, Qatar;
| | - Ayman Al Haj Zen
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK;
- College of Health and Life Sciences, Hamad Bin Khalifa University, Education City, Doha P.O. Box 34110, Qatar;
- Correspondence: ; Tel.: +974-4454-6352
| |
Collapse
|
12
|
The Evolution of Biomineralization through the Co-Option of Organic Scaffold Forming Networks. Cells 2022; 11:cells11040595. [PMID: 35203246 PMCID: PMC8870065 DOI: 10.3390/cells11040595] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/05/2022] Open
Abstract
Biomineralization is the process in which organisms use minerals to generate hard structures like teeth, skeletons and shells. Biomineralization is proposed to have evolved independently in different phyla through the co-option of pre-existing developmental programs. Comparing the gene regulatory networks (GRNs) that drive biomineralization in different species could illuminate the molecular evolution of biomineralization. Skeletogenesis in the sea urchin embryo was extensively studied and the underlying GRN shows high conservation within echinoderms, larval and adult skeletogenesis. The organic scaffold in which the calcite skeletal elements form in echinoderms is a tubular compartment generated by the syncytial skeletogenic cells. This is strictly different than the organic cartilaginous scaffold that vertebrates mineralize with hydroxyapatite to make their bones. Here I compare the GRNs that drive biomineralization and tubulogenesis in echinoderms and in vertebrates. The GRN that drives skeletogenesis in the sea urchin embryo shows little similarity to the GRN that drives bone formation and high resemblance to the GRN that drives vertebrates’ vascular tubulogenesis. On the other hand, vertebrates’ bone-GRNs show high similarity to the GRNs that operate in the cells that generate the cartilage-like tissues of basal chordate and invertebrates that do not produce mineralized tissue. These comparisons suggest that biomineralization in deuterostomes evolved through the phylum specific co-option of GRNs that control distinct organic scaffolds to mineralization.
Collapse
|
13
|
Ismail AA, Shaker BT, Bajou K. The Plasminogen-Activator Plasmin System in Physiological and Pathophysiological Angiogenesis. Int J Mol Sci 2021; 23:ijms23010337. [PMID: 35008762 PMCID: PMC8745544 DOI: 10.3390/ijms23010337] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis is a process associated with the migration and proliferation of endothelial cells (EC) to form new blood vessels. It is involved in various physiological and pathophysiological conditions and is controlled by a wide range of proangiogenic and antiangiogenic molecules. The plasminogen activator–plasmin system plays a major role in the extracellular matrix remodeling process necessary for angiogenesis. Urokinase/tissue-type plasminogen activators (uPA/tPA) convert plasminogen into the active enzyme plasmin, which in turn activates matrix metalloproteinases and degrades the extracellular matrix releasing growth factors and proangiogenic molecules such as the vascular endothelial growth factor (VEGF-A). The plasminogen activator inhibitor-1 (PAI-1) is the main inhibitor of uPA and tPA, thereby an inhibitor of pericellular proteolysis and intravascular fibrinolysis, respectively. Paradoxically, PAI-1, which is expressed by EC during angiogenesis, is elevated in several cancers and is found to promote angiogenesis by regulating plasmin-mediated proteolysis and by promoting cellular migration through vitronectin. The urokinase-type plasminogen activator receptor (uPAR) also induces EC cellular migration during angiogenesis via interacting with signaling partners. Understanding the molecular functions of the plasminogen activator plasmin system and targeting angiogenesis via blocking serine proteases or their interactions with other molecules is one of the major therapeutic strategies scientists have been attracted to in controlling tumor growth and other pathological conditions characterized by neovascularization.
Collapse
Affiliation(s)
- Asmaa Anwar Ismail
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (A.A.I.); (B.T.S.)
- Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Baraah Tariq Shaker
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (A.A.I.); (B.T.S.)
- Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Khalid Bajou
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (A.A.I.); (B.T.S.)
- Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
- Correspondence:
| |
Collapse
|
14
|
Kumar N, Saraber P, Ding Z, Kusumbe AP. Diversity of Vascular Niches in Bones and Joints During Homeostasis, Ageing, and Diseases. Front Immunol 2021; 12:798211. [PMID: 34975909 PMCID: PMC8718446 DOI: 10.3389/fimmu.2021.798211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/19/2021] [Indexed: 12/29/2022] Open
Abstract
The bones and joints in the skeletal system are composed of diverse cell types, including vascular niches, bone cells, connective tissue cells and mineral deposits and regulate whole-body homeostasis. The capacity of maintaining strength and generation of blood lineages lies within the skeletal system. Bone harbours blood and immune cells and their progenitors, and vascular cells provide several immune cell type niches. Blood vessels in bone are phenotypically and functionally diverse, with distinct capillary subtypes exhibiting striking changes with age. The bone vasculature has a special impact on osteogenesis and haematopoiesis, and dysregulation of the vasculature is associated with diverse blood and bone diseases. Ageing is associated with perturbed haematopoiesis, loss of osteogenesis, increased adipogenesis and diminished immune response and immune cell production. Endothelial and perivascular cells impact immune cell production and play a crucial role during inflammation. Here, we discuss normal and maladapted vascular niches in bone during development, homeostasis, ageing and bone diseases such as rheumatoid arthritis and osteoarthritis. Further, we discuss the role of vascular niches during bone malignancy.
Collapse
Affiliation(s)
| | | | | | - Anjali P. Kusumbe
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), Tissue and Tumor Microenvironments Group, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
15
|
Wälchli T, Bisschop J, Miettinen A, Ulmann-Schuler A, Hintermüller C, Meyer EP, Krucker T, Wälchli R, Monnier PP, Carmeliet P, Vogel J, Stampanoni M. Hierarchical imaging and computational analysis of three-dimensional vascular network architecture in the entire postnatal and adult mouse brain. Nat Protoc 2021; 16:4564-4610. [PMID: 34480130 DOI: 10.1038/s41596-021-00587-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 06/08/2021] [Indexed: 02/08/2023]
Abstract
The formation of new blood vessels and the establishment of vascular networks are crucial during brain development, in the adult healthy brain, as well as in various diseases of the central nervous system. Here, we describe a step-by-step protocol for our recently developed method that enables hierarchical imaging and computational analysis of vascular networks in postnatal and adult mouse brains. The different stages of the procedure include resin-based vascular corrosion casting, scanning electron microscopy, synchrotron radiation and desktop microcomputed tomography imaging, and computational network analysis. Combining these methods enables detailed visualization and quantification of the 3D brain vasculature. Network features such as vascular volume fraction, branch point density, vessel diameter, length, tortuosity and directionality as well as extravascular distance can be obtained at any developmental stage from the early postnatal to the adult brain. This approach can be used to provide a detailed morphological atlas of the entire mouse brain vasculature at both the postnatal and the adult stage of development. Our protocol allows the characterization of brain vascular networks separately for capillaries and noncapillaries. The entire protocol, from mouse perfusion to vessel network analysis, takes ~10 d.
Collapse
Affiliation(s)
- Thomas Wälchli
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, Zurich, Switzerland.
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland.
- Group Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada.
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada.
| | - Jeroen Bisschop
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, Zurich, Switzerland
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
- Group Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Arttu Miettinen
- Swiss Light Source, Paul Scherrer Institute, Villigen, Switzerland
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
- Department of Physics, University of Jyväskylä, Jyväskylä, Finland
| | | | | | - Eric P Meyer
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Thomas Krucker
- Novartis Institutes for BioMedical Research Inc, Emeryville, CA, USA
| | - Regula Wälchli
- Department of Dermatology, Pediatric Skin Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Philippe P Monnier
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Krembil Research Institute, Vision Division, Krembil Discovery Tower, Toronto, Ontario, Canada
- Department of Ophthalmology and Vision Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Johannes Vogel
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Marco Stampanoni
- Swiss Light Source, Paul Scherrer Institute, Villigen, Switzerland
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
16
|
The endothelium-bone axis in development, homeostasis and bone and joint disease. Nat Rev Rheumatol 2021; 17:608-620. [PMID: 34480164 DOI: 10.1038/s41584-021-00682-3] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2021] [Indexed: 01/20/2023]
Abstract
Blood vessels form a versatile transport network that is best known for its critical roles in processes such as tissue oxygenation, metabolism and immune surveillance. The vasculature also provides local, often organ-specific, molecular signals that control the behaviour of other cell types in their vicinity during development, homeostasis and regeneration, and also in disease processes. In the skeletal system, the local vasculature is actively involved in both bone formation and resorption. In addition, blood vessels participate in inflammatory processes and contribute to the pathogenesis of diseases that affect the joints, such as rheumatoid arthritis and osteoarthritis. This Review summarizes the current understanding of the architecture, angiogenic growth and functional properties of the bone vasculature. The effects of ageing and pathological conditions, including arthritis and osteoporosis, are also discussed.
Collapse
|
17
|
Hu Z, Liu W, Hua X, Chen X, Chang Y, Hu Y, Xu Z, Song J. Single-Cell Transcriptomic Atlas of Different Human Cardiac Arteries Identifies Cell Types Associated With Vascular Physiology. Arterioscler Thromb Vasc Biol 2021; 41:1408-1427. [PMID: 33626908 DOI: 10.1161/atvbaha.120.315373] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Zhan Hu
- Department of Cardiovascular Surgery (Z.H., X.H., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wendao Liu
- Department of Cardiovascular Surgery (Z.H., X.H., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,The Cardiomyopathy Research Group at Fuwai Hospital (W.L., X.H., X.C., Y.C., Y.H., J.S.)
| | - Xiumeng Hua
- Department of Cardiovascular Surgery (Z.H., X.H., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Cardiovascular Disease (W.L., X.H., X.C., Y.C., Y.H., Z.X., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,The Cardiomyopathy Research Group at Fuwai Hospital (W.L., X.H., X.C., Y.C., Y.H., J.S.)
| | - Xiao Chen
- State Key Laboratory of Cardiovascular Disease (W.L., X.H., X.C., Y.C., Y.H., Z.X., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,The Cardiomyopathy Research Group at Fuwai Hospital (W.L., X.H., X.C., Y.C., Y.H., J.S.)
| | - Yuan Chang
- State Key Laboratory of Cardiovascular Disease (W.L., X.H., X.C., Y.C., Y.H., Z.X., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,The Cardiomyopathy Research Group at Fuwai Hospital (W.L., X.H., X.C., Y.C., Y.H., J.S.).,Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (Y.C.)
| | - Yiqing Hu
- State Key Laboratory of Cardiovascular Disease (W.L., X.H., X.C., Y.C., Y.H., Z.X., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,The Cardiomyopathy Research Group at Fuwai Hospital (W.L., X.H., X.C., Y.C., Y.H., J.S.)
| | - Zhenyu Xu
- State Key Laboratory of Cardiovascular Disease (W.L., X.H., X.C., Y.C., Y.H., Z.X., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Pathology Center, State Key Laboratory of Cardiovascular Disease (Z.X.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangping Song
- Department of Cardiovascular Surgery (Z.H., X.H., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Cardiovascular Disease (W.L., X.H., X.C., Y.C., Y.H., Z.X., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,The Cardiomyopathy Research Group at Fuwai Hospital (W.L., X.H., X.C., Y.C., Y.H., J.S.)
| |
Collapse
|
18
|
Huang X, Liang P, Jiang B, Zhang P, Yu W, Duan M, Guo L, Cui X, Huang M, Huang X. Hyperbaric oxygen potentiates diabetic wound healing by promoting fibroblast cell proliferation and endothelial cell angiogenesis. Life Sci 2020; 259:118246. [DOI: 10.1016/j.lfs.2020.118246] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/31/2020] [Accepted: 08/06/2020] [Indexed: 12/31/2022]
|
19
|
Blatchley MR, Gerecht S. Reconstructing the Vascular Developmental Milieu In Vitro. Trends Cell Biol 2020; 30:15-31. [DOI: 10.1016/j.tcb.2019.10.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 10/14/2019] [Indexed: 12/25/2022]
|
20
|
Jusoh N, Ko J, Jeon NL. Microfluidics-based skin irritation test using in vitro 3D angiogenesis platform. APL Bioeng 2019; 3:036101. [PMID: 31431937 PMCID: PMC6697035 DOI: 10.1063/1.5093975] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/26/2019] [Indexed: 01/19/2023] Open
Abstract
A global ban on animal experiments has been proposed. Hence, it is imperative to develop alternative models. Artificial skin models should reflect the responses of subcutaneous blood vessels and the immune system to elucidate disease and identify cosmetics' base materials. Notably, in vivo skin-irritation cascades involve disruption of the epidermal barrier and the release of proinflammatory mediators in response to chemical stimuli. Such proinflammatory factors promote angiogenesis and blood vessel permeability, as observed in irritant contact dermatitis. As an alternative to animal models, we propose a novel skin-irritation model based on a three-dimensional in vitro angiogenesis platform, in which irritated keratinocytes biochemically stimulate vascular endothelial growth factors. Our microfluidic platform hosts interactions between keratinocytes and dermal fibroblasts, which promote angiogenic sprouting. We use sodium lauryl sulfate (SLS) and steartrimonium chloride (SC) as chemical irritants. The irritative effects of SLS and SC are of particular interest due to the ubiquity of both SLS and SC in cosmetics. SLS was observed to significantly affect angiogenic performance, with increasing sprout length. Further promotion of vessel sprouting and lumen formation was observed with 10, 20, and 60 μM of SC, despite its classification as nonirritating and use in supposedly safe formulations. This platform provides an alternative to animal testing as a basis for testing cosmetics and pharmaceutical substances, in addition to serving as a disease model for irritant contact dermatitis.
Collapse
Affiliation(s)
| | - Jihoon Ko
- Department of Mechanical and Aerospace Engineering, Seoul National University, Seoul 08826, South Korea
| | | |
Collapse
|
21
|
Fearnley GW, Abdul-Zani I, Latham AM, Hollstein MC, Ladbury JE, Wheatcroft SB, Odell AF, Ponnambalam S. Tpl2 is required for VEGF-A-stimulated signal transduction and endothelial cell function. Biol Open 2019; 8:bio.034215. [PMID: 31072823 PMCID: PMC6550078 DOI: 10.1242/bio.034215] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
New blood vessel sprouting (angiogenesis) and vascular physiology are fundamental features of metazoan species but we do not fully understand how signal transduction pathways regulate diverse vascular responses. The vascular endothelial growth factor (VEGF) family bind membrane-bound receptor tyrosine kinases (VEGFRs), which trigger multiple signal transduction pathways and diverse cellular responses. We evaluated whether the MAP3K family member and proto-oncoprotein Tpl2 (MAP3K8) regulates basal and VEGF-A-stimulated signal transduction in endothelial cells. Notably, stimulation with exogenous VEGF-A increased Tpl2 mRNA levels and consequently de novo protein synthesis. Depletion of Tpl2 levels reveals a role in both basal and VEGF-A-stimulated endothelial cell responses, including endothelial-leukocyte interactions, monolayer permeability and new blood vessel formation. Under basal conditions, Tpl2 modulates a signal transduction cascade resulting in phosphorylation of a nuclear transcription factor (ATF-2) and altered endothelial gene expression, a pathway previously identified as crucial in VEGF-dependent vascular responses. Loss of Tpl2 expression or activity impairs signal transduction through Akt, eNOS and ATF-2, broadly impacting on endothelial function. Our study now provides a mechanism for Tpl2 as a central component of signal transduction pathways in the endothelium. Summary: Our study shows that the growth factor VEGF-A stimulates synthesis in endothelial cells of a proto-oncoprotein and protein kinase, Tpl2, and this is required for signal transduction and angiogenesis.
Collapse
Affiliation(s)
- Gareth W Fearnley
- School of Molecular & Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Izma Abdul-Zani
- School of Molecular & Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Antony M Latham
- School of Molecular & Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Monica C Hollstein
- Leeds Institute of Cardiovascular & Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - John E Ladbury
- School of Molecular & Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular & Metabolic Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Adam F Odell
- Leeds Institute for Medical Research at St. James's, University of Leeds, Leeds, LS2 9JT, UK
| | | |
Collapse
|
22
|
Combination of Ligusticum Chuanxiong and Radix Paeonia Promotes Angiogenesis in Ischemic Myocardium through Notch Signalling and Mobilization of Stem Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:7912402. [PMID: 30906416 PMCID: PMC6398078 DOI: 10.1155/2019/7912402] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/10/2019] [Accepted: 02/03/2019] [Indexed: 01/07/2023]
Abstract
Objective To study the cardioprotective mechanism by which the combination of Chuanxiong (CX) and Chishao (CS) promotes angiogenesis. Methods Myocardial infarction (MI) mouse models were induced by ligation of the left anterior descending coronary artery. The effects on cardiac function were evaluated in the perindopril tert-butylamine group (PB group) (3 mg/kg/d), CX group (55 mg/kg/d), CS group (55 mg/kg/d), and CX and CS combination (CX-CS) group (27.5 mg/kg/d CX plus 27.5 mg/kg/d CS). RO4929097, an inhibitor of Notch γ secretase, was used (10 mg/kg/d) to explore the role of Notch signalling in the CX-CS-induced promotion of angiogenesis in the myocardial infarcted border zone (IBZ). The left ventricular ejection fraction (LVEF) and percentage of MI area were evaluated with animal ultrasound and Masson staining. The average optical densities (AODs) of CD31 and vWF in the myocardial IBZ were detected by immunofluorescence. Angiogenesis-related proteins including hypoxia-inducible factor 1-alpha (HIF-1α), fibroblast growth factor receptor 1 (FGFR-1), Notch1 and Notch intracellular domain (NICD), and stem cell mobilization-related proteins including stromal cell-derived factor 1 (SDF-1), C-X-C chemokine receptor type 4 (CXCR-4), and cardiotrophin1 were detected by western blot analysis. Results Compared with the model group, the CX-CS and PB groups both showed markedly improved LVEF and decreased percentage of MI area after 21 days of treatment. Although the CX group and CS group showed increased LVEF and decreased MI areas compared with the model group, the difference was not significant. The AOD of CD31 in the IBZ in both the model and the CX-CS-I group was markedly reduced compared with that in the sham group. CX-CS significantly increased the CD31 AOD in the IBZ and decreased the AODs of CD31 and vWF in the infarct zone compared with those in the model group. The expression of HIF-1α in both the model group and the CX-CS group was higher than that in the sham group. Compared with the model group, the expression of FGFR-1, SDF-1, cardiotrophin1, Notch1, and NICD was increased in the CX-CS group. Notch1 and NICD expression in the CX-CS-I group was reduced compared with that in the CX-CS group. Conclusions The combination of CX and CS protected cardiomyocytes in the IBZ better than CX or CS alone. The mechanism by which CX-CS protects ischemic myocardium may be related to the proangiogenesis effect of CX-CS exerted through Notch signalling and the mobilization of stem cells to the IBZ.
Collapse
|
23
|
Rajkovic I, Wong R, Lemarchand E, Rivers-Auty J, Rajkovic O, Garlanda C, Allan SM, Pinteaux E. Pentraxin 3 promotes long-term cerebral blood flow recovery, angiogenesis, and neuronal survival after stroke. J Mol Med (Berl) 2018; 96:1319-1332. [PMID: 30315331 PMCID: PMC6245246 DOI: 10.1007/s00109-018-1698-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 09/24/2018] [Accepted: 09/25/2018] [Indexed: 01/08/2023]
Abstract
Restoration of cerebral blood flow (CBF) and upregulation of angiogenesis are crucial for brain repair and functional recovery after cerebral ischaemia. Pentraxin 3 (PTX3) is a key regulator of angiogenesis and is emerging as a promising target for cerebrovascular repair after stroke. Here, we investigated for the first time the role of PTX3 in long-term CBF, angiogenesis, and neuronal viability after ischaemic stroke induced by transient middle cerebral artery occlusion (MCAo). Lack of PTX3 had no effect on early brain damage, but significantly impaired restoration of CBF, 14 and 28 days after MCAo, compared to wild-type (WT) mice. Immunohistochemical analysis revealed that PTX3 KO mice have significantly greater neuronal loss, significantly decreased vessel diameter, vessel proliferation, vascular density, and reactive astrocytes and decreased expression of vascular endothelial growth factor receptor 2 (VEGR2), vascular extracellular matrix (ECM)-proteins (collagen IV, laminin), and integrin-β, in the ipsilateral (stroke) hemisphere compared to WT mice, 28 days after MCAo. Therefore, PTX3 promotes sustained long-term recovery of CBF, angiogenesis, and neuronal viability after cerebral ischaemia. Collectively, these findings demonstrate the potential and clinical relevance of PTX3 as a promising therapeutic target, providing sustained long-term post-stroke neurovascular repair and reducing the loss of neurons. KEY MESSAGES: Pentraxin 3 (PTX3) is a key regulator of angiogenesis and is emerging as a promising target for cerebrovascular repair after stroke. Restoration of cerebral blood flow (CBF) and angiogenesis are crucial for brain repair and functional recovery after cerebral ischaemia. PTX3 promotes sustained long-term recovery of CBF, angiogenesis, and neuronal viability after cerebral ischaemia.
Collapse
Affiliation(s)
- Ivana Rajkovic
- Faculty of Biology, Medicine and Health, AV Hill Building, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Raymond Wong
- Faculty of Biology, Medicine and Health, AV Hill Building, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Eloise Lemarchand
- Faculty of Biology, Medicine and Health, AV Hill Building, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Jack Rivers-Auty
- Faculty of Biology, Medicine and Health, AV Hill Building, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Olivera Rajkovic
- Faculty of Biology, Medicine and Health, AV Hill Building, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Cecilia Garlanda
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center, 20089, Rozzano, MI, Italy
| | - Stuart M Allan
- Faculty of Biology, Medicine and Health, AV Hill Building, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, AV Hill Building, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK.
| |
Collapse
|
24
|
Rhodocetin-αβ selectively breaks the endothelial barrier of the tumor vasculature in HT1080 fibrosarcoma and A431 epidermoid carcinoma tumor models. Oncotarget 2018; 9:22406-22422. [PMID: 29854288 PMCID: PMC5976474 DOI: 10.18632/oncotarget.25032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 03/19/2018] [Indexed: 11/29/2022] Open
Abstract
The tumor vasculature differs from normal blood vessels in morphology, composition and stability. Here, we describe a novel tumor vessel-disrupting mechanism. In an HT1080/mouse xenograft tumor model rhodocetin-αβ was highly effective in disrupting the tumor endothelial barrier. Mechanistically, rhodocetin-αβ triggered MET signaling via neuropilin-1. As both neuropilin-1 and MET were only lumen-exposed in a subset of abnormal tumor vessels, but not in normal vessels, the prime target of rhodocetin-αβ were these abnormal tumor vessels. Consequently, cells lining such tumor vessels became increasingly motile which compromised the vessel wall tightness. After this initial leakage, rhodocetin-αβ could leave the bloodstream and reach the as yet inaccessible neuropilin-1 on the basolateral side of endothelial cells and thus disrupt nearby vessels. Due to the specific neuropilin-1/MET co-distribution on cells lining such abnormal tumor vessels in contrast to normal endothelial cells, rhodocetin-αβ formed the necessary trimeric signaling complex of rhodocetin-αβ-MET-neuropilin-1 only in these abnormal tumor vessels. This selective attack of tumor vessels, sparing endothelial cell-lined vessels of normal tissues, suggests that the neuropilin-1-MET signaling axis may be a promising drugable target for anti-tumor therapy, and that rhodocetin-αβ may serve as a lead structure to develop novel anti-tumor drugs that target such vessels.
Collapse
|
25
|
Cheng X, Zheng J, Li G, Göbel V, Zhang H. Degradation for better survival? Role of ubiquitination in epithelial morphogenesis. Biol Rev Camb Philos Soc 2018; 93:1438-1460. [PMID: 29493067 DOI: 10.1111/brv.12404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/31/2018] [Accepted: 02/05/2018] [Indexed: 02/06/2023]
Abstract
As a prevalent post-translational modification, ubiquitination is essential for many developmental processes. Once covalently attached to the small and conserved polypeptide ubiquitin (Ub), a substrate protein can be directed to perform specific biological functions via its Ub-modified form. Three sequential catalytic reactions contribute to this process, among which E3 ligases serve to identify target substrates and promote the activated Ub to conjugate to substrate proteins. Ubiquitination has great plasticity, with diverse numbers, topologies and modifications of Ub chains conjugated at different substrate residues adding a layer of complexity that facilitates a huge range of cellular functions. Herein, we highlight key advances in the understanding of ubiquitination in epithelial morphogenesis, with an emphasis on the latest insights into its roles in cellular events involved in polarized epithelial tissue, including cell adhesion, asymmetric localization of polarity determinants and cytoskeletal organization. In addition, the physiological roles of ubiquitination are discussed for typical examples of epithelial morphogenesis, such as lung branching, vascular development and synaptic formation and plasticity. Our increased understanding of ubiquitination in epithelial morphogenesis may provide novel insights into the molecular mechanisms underlying epithelial regeneration and maintenance.
Collapse
Affiliation(s)
- Xiaoxiang Cheng
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Jun Zheng
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Gang Li
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Verena Göbel
- Department of Pediatrics, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114,, U.S.A
| | - Hongjie Zhang
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| |
Collapse
|
26
|
Patra C, Boccaccini A, Engel F. Vascularisation for cardiac tissue engineering: the extracellular matrix. Thromb Haemost 2017; 113:532-47. [DOI: 10.1160/th14-05-0480] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 09/03/2014] [Indexed: 02/07/2023]
Abstract
SummaryCardiovascular diseases present a major socio-economic burden. One major problem underlying most cardiovascular and congenital heart diseases is the irreversible loss of contractile heart muscle cells, the cardiomyocytes. To reverse damage incurred by myocardial infarction or by surgical correction of cardiac malformations, the loss of cardiac tissue with a thickness of a few millimetres needs to be compensated. A promising approach to this issue is cardiac tissue engineering. In this review we focus on the problem of in vitro vascularisation as implantation of cardiac patches consisting of more than three layers of cardiomyocytes (> 100 μm thick) already results in necrosis. We explain the need for vascularisation and elaborate on the importance to include non-myocytes in order to generate functional vascularised cardiac tissue. We discuss the potential of extracellular matrix molecules in promoting vascularisation and introduce nephronectin as an example of a new promising candidate. Finally, we discuss current biomaterial- based approaches including micropatterning, electrospinning, 3D micro-manufacturing technology and porogens. Collectively, the current literature supports the notion that cardiac tissue engineering is a realistic option for future treatment of paediatric and adult patients with cardiac disease.
Collapse
|
27
|
Collateral Damage Intended-Cancer-Associated Fibroblasts and Vasculature Are Potential Targets in Cancer Therapy. Int J Mol Sci 2017; 18:ijms18112355. [PMID: 29112161 PMCID: PMC5713324 DOI: 10.3390/ijms18112355] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/25/2017] [Accepted: 11/02/2017] [Indexed: 02/07/2023] Open
Abstract
After oncogenic transformation, tumor cells rewire their metabolism to obtain sufficient energy and biochemical building blocks for cell proliferation, even under hypoxic conditions. Glucose and glutamine become their major limiting nutritional demands. Instead of being autonomous, tumor cells change their immediate environment not only by their metabolites but also by mediators, such as juxtacrine cell contacts, chemokines and other cytokines. Thus, the tumor cells shape their microenvironment as well as induce resident cells, such as fibroblasts and endothelial cells (ECs), to support them. Fibroblasts differentiate into cancer-associated fibroblasts (CAFs), which produce a qualitatively and quantitatively different extracellular matrix (ECM). By their contractile power, they exert tensile forces onto this ECM, leading to increased intratumoral pressure. Moreover, along with enhanced cross-linkage of the ECM components, CAFs thus stiffen the ECM. Attracted by tumor cell- and CAF-secreted vascular endothelial growth factor (VEGF), ECs sprout from pre-existing blood vessels during tumor-induced angiogenesis. Tumor vessels are distinct from EC-lined vessels, because tumor cells integrate into the endothelium or even mimic and replace it in vasculogenic mimicry (VM) vessels. Not only the VM vessels but also the characteristically malformed EC-lined tumor vessels are typical for tumor tissue and may represent promising targets in cancer therapy.
Collapse
|
28
|
Prakash Parthiban S, Rana D, Jabbari E, Benkirane-Jessel N, Ramalingam M. Covalently immobilized VEGF-mimicking peptide with gelatin methacrylate enhances microvascularization of endothelial cells. Acta Biomater 2017; 51:330-340. [PMID: 28110074 DOI: 10.1016/j.actbio.2017.01.046] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 12/16/2016] [Accepted: 01/16/2017] [Indexed: 01/11/2023]
Abstract
Clinically usable tissue-engineered constructs are currently limited due to their inability of forming microvascular networks necessary for adequate cellular oxygen and nutrient supply upon implantation. The aim of this study is to investigate the conditions necessary for microvascularization in a tissue-engineered construct using vascular endothelial growth factor (VEGF). The construct was made of gelatin methacrylate (GelMA) based cell-laden hydrogel system, which was then covalently linked with VEGF-mimicking peptide (AcQK), using human umbilical vein endothelial cells (HUVECs) as the model cell. The results of the mechanics and gene expression analysis indicated significant changes in mechanical properties and upregulation of vascular-specific genes. The major finding of this study is that the increased expression of vascular-specific genes could be achieved by employing AcQK in the GelMA based hydrogel system, leading to accelerated microvascularization. We conclude that GelMA with covalently-linked angiogenic peptide is a useful tissue engineered construct suitable for microvascularization. STATEMENT OF SIGNIFICANCE: (1) This study reports the conditions necessary for microvascularization in a tissue-engineered construct using vascular endothelial growth factor (VEGF). (2) The construct was made of gelatin methacrylate based cell-laden hydrogel system. (3) There is a significant change observed in mechanical properties and upregulation of vascular-specific genes, in particular CD34, when AcQK is used. (4) The major finding of this study is that the increased expression of vascular-specific genes, i.e., CD34 could be achieved by employing AcQK in the GelMA based hydrogel system, leading to accelerated microvascularization.
Collapse
|
29
|
Barnett FH, Rosenfeld M, Wood M, Kiosses WB, Usui Y, Marchetti V, Aguilar E, Friedlander M. Macrophages form functional vascular mimicry channels in vivo. Sci Rep 2016; 6:36659. [PMID: 27834402 PMCID: PMC5105153 DOI: 10.1038/srep36659] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/18/2016] [Indexed: 12/02/2022] Open
Abstract
Macrophages, key cells of the innate immune system, are known to support angiogenesis but are not believed to directly form vessel walls. Here we show that macrophages structurally form primitive, NON-ENDOTHELIAL “vessels” or vascular mimicry (VM) channels in both tumor and angiogenesis in vivo models. These channels are functionally connected to the systemic vasculature as they are perfused by intravenously injected dye. Since both models share hypoxic micro-environments, we hypothesized that hypoxia may be an important mediator of VM formation. Indeed, conditional genetic depletion of myeloid-specific HIF-1α results in decreased VM network formation, dye perfusion and tumor size. Although the macrophage VM network shares some features with an endothelial vasculature, it is ultrastructurally different. Cancer stem cells have been shown to form vascular mimicry channels. Our data demonstrates that tumor-associated macrophages also form them. The identification of this novel type of vascular mimicry may help in the development of targeted cancer therapeutics.
Collapse
Affiliation(s)
- Faith H Barnett
- Department of Cell and Molecular Biology, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Mauricio Rosenfeld
- Department of Cell and Molecular Biology, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Malcolm Wood
- Department of Cell and Molecular Biology, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - William B Kiosses
- Department of Cell and Molecular Biology, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Yoshihiko Usui
- Department of Cell and Molecular Biology, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Valentina Marchetti
- Department of Cell and Molecular Biology, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Edith Aguilar
- Department of Cell and Molecular Biology, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Martin Friedlander
- Department of Cell and Molecular Biology, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| |
Collapse
|
30
|
Hensley MR, Cui Z, Chua RFM, Simpson S, Shammas NL, Yang JY, Leung YF, Zhang G. Evolutionary and developmental analysis reveals KANK genes were co-opted for vertebrate vascular development. Sci Rep 2016; 6:27816. [PMID: 27292017 PMCID: PMC4904190 DOI: 10.1038/srep27816] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 05/25/2016] [Indexed: 11/28/2022] Open
Abstract
Gene co-option, usually after gene duplication, in the evolution of development is found to contribute to vertebrate morphological innovations, including the endothelium-based vascular system. Recently, a zebrafish kank gene was found expressed in the vascular vessel primordium, suggesting KANK genes are a component of the developmental tool kit for the vertebrate vascular system. However, how the KANK gene family is involved in vascular vessel development during evolution remains largely unknown. First, we analyzed the molecular evolution of the KANK genes in metazoan, and found that KANK1, KANK2, KANK3 and KANK4 emerged in the lineage of vertebrate, consistent with the two rounds of vertebrate whole-genome duplications (WGD). Moreover, KANK genes were further duplicated in teleosts through the bony-fish specific WGD, while only kank1 and kank4 duplicates were retained in some of the examined fish species. We also found all zebrafish kank genes, except kank1b, are primarily expressed during embryonic vascular development. Compared to invertebrate KANK gene expression in the central nervous system, the vascular expression of zebrafish kank genes suggested KANK genes were co-opted for vertebrate vascular development. Given the cellular roles of KANK genes, our results suggest that this co-option may facilitate the evolutionary origin of vertebrate vascular vessels.
Collapse
Affiliation(s)
- Monica R Hensley
- Department of Comparative Pathobiology;.725 Harrison Street, West Lafayette, IN 47907, USA
| | - Zhibin Cui
- Department of Comparative Pathobiology;.725 Harrison Street, West Lafayette, IN 47907, USA
| | - Rhys F M Chua
- Department of Comparative Pathobiology;.725 Harrison Street, West Lafayette, IN 47907, USA
| | - Stefanie Simpson
- Department of Comparative Pathobiology;.725 Harrison Street, West Lafayette, IN 47907, USA
| | - Nicole L Shammas
- Department of Comparative Pathobiology;.725 Harrison Street, West Lafayette, IN 47907, USA
| | - Jer-Yen Yang
- Purdue University Center for Cancer Research; 725 Harrison Street, West Lafayette, IN 47907, USA.,Department of Basic Medical Sciences; 725 Harrison Street, West Lafayette, IN 47907,USA
| | - Yuk Fai Leung
- Department of Biological Sciences, 725 Harrison Street, West Lafayette, IN 47907,USA.,Integrative Neuroscience Center; 725 Harrison Street, West Lafayette, IN 47907, USA
| | - GuangJun Zhang
- Department of Comparative Pathobiology;.725 Harrison Street, West Lafayette, IN 47907, USA.,Purdue University Center for Cancer Research; 725 Harrison Street, West Lafayette, IN 47907, USA.,Integrative Neuroscience Center; 725 Harrison Street, West Lafayette, IN 47907, USA.,Purdue Institute for Inflammation, Immunology and Infectious Diseases (PI4D), 725 Harrison Street, West Lafayette, IN 47907, USA
| |
Collapse
|
31
|
|
32
|
Jusoh N, Oh S, Kim S, Kim J, Jeon NL. Microfluidic vascularized bone tissue model with hydroxyapatite-incorporated extracellular matrix. LAB ON A CHIP 2015; 15:3984-8. [PMID: 26288174 DOI: 10.1039/c5lc00698h] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Current in vitro systems mimicking bone tissues fail to fully integrate the three-dimensional (3D) microvasculature and bone tissue microenvironments, decreasing their similarity to in vivo conditions. Here, we propose 3D microvascular networks in a hydroxyapatite (HA)-incorporated extracellular matrix (ECM) for designing and manipulating a vascularized bone tissue model in a microfluidic device. Incorporation of HA of various concentrations resulted in ECM with varying mechanical properties. Sprouting angiogenesis was affected by mechanically modulated HA-extracellular matrix interactions, generating a model of vascularized bone microenvironment. Using this platform, we observed that hydroxyapatite enhanced angiogenic properties such as sprout length, sprouting speed, sprout number, and lumen diameter. This new platform integrates fibrin ECM with the synthetic bone mineral HA to provide in vivo-like microenvironments for bone vessel sprouting.
Collapse
Affiliation(s)
- Norhana Jusoh
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, 151-744, South Korea.
| | | | | | | | | |
Collapse
|
33
|
Charpentier MS, Tandon P, Trincot CE, Koutleva EK, Conlon FL. A distinct mechanism of vascular lumen formation in Xenopus requires EGFL7. PLoS One 2015; 10:e0116086. [PMID: 25705891 PMCID: PMC4338030 DOI: 10.1371/journal.pone.0116086] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 12/04/2014] [Indexed: 01/03/2023] Open
Abstract
During vertebrate blood vessel development, lumen formation is the critical process by which cords of endothelial cells transition into functional tubular vessels. Here, we use Xenopus embryos to explore the cellular and molecular mechanisms underlying lumen formation of the dorsal aorta and the posterior cardinal veins, the primary major vessels that arise via vasculogenesis within the first 48 hours of life. We demonstrate that endothelial cells are initially found in close association with one another through the formation of tight junctions expressing ZO-1. The emergence of vascular lumens is characterized by elongation of endothelial cell shape, reorganization of junctions away from the cord center to the periphery of the vessel, and onset of Claudin-5 expression within tight junctions. Furthermore, unlike most vertebrate vessels that exhibit specialized apical and basal domains, we show that early Xenopus vessels are not polarized. Moreover, we demonstrate that in embryos depleted of the extracellular matrix factor Epidermal Growth Factor-Like Domain 7 (EGFL7), an evolutionarily conserved factor associated with vertebrate vessel development, vascular lumens fail to form. While Claudin-5 localizes to endothelial tight junctions of EGFL7-depleted embryos in a timely manner, endothelial cells of the aorta and veins fail to undergo appropriate cell shape changes or clear junctions from the cell-cell contact. Taken together, we demonstrate for the first time the mechanisms by which lumens are generated within the major vessels in Xenopus and implicate EGFL7 in modulating cell shape and cell-cell junctions to drive proper lumen morphogenesis.
Collapse
Affiliation(s)
- Marta S. Charpentier
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Panna Tandon
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Claire E. Trincot
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Elitza K. Koutleva
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Frank L. Conlon
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, UNC-CH, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
34
|
Sumida GM, Yamada S. Rho GTPases and the downstream effectors actin-related protein 2/3 (Arp2/3) complex and myosin II induce membrane fusion at self-contacts. J Biol Chem 2014; 290:3238-47. [PMID: 25527498 DOI: 10.1074/jbc.m114.612168] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Actin regulation is required for membrane activities that drive cell adhesion and migration. The Rho GTPase family plays critical roles in actin and membrane dynamics; however, the roles of the Rho GTPase family are not limited to cell adhesion and migration. Using micron-sized obstacles to induce the formation of self-contacts in epithelial cells, we previously showed that self-adhesion is distinct from cell-to-cell adhesion in that self-contacts are eliminated by membrane fusion. In the current study, we identified Rho GTPases, RhoA, Rac1, and Cdc42, as potential upstream regulators of membrane fusion. The RhoA downstream effector myosin II is required for fusion as the expression of mutant myosin light chain reduced membrane fusion. Furthermore, an inhibitor of the Arp2/3 complex, a downstream effector of Rac1 and Cdc42, also reduced self-contact-induced membrane fusion. At self-contacts, while the concentration of E-cadherin diminished, the intensity of GFP-tagged Arp3 rapidly fluctuated then decreased and stabilized after membrane fusion. Taken together, these data suggest that the Arp2/3 complex-mediated actin polymerization brings two opposing membranes into close apposition by possibly excluding E-cadherin from contact sites, thus promoting membrane fusion at self-contacts.
Collapse
Affiliation(s)
- Grant M Sumida
- From the Department of Biomedical Engineering, University of California, Davis, California 95616
| | - Soichiro Yamada
- From the Department of Biomedical Engineering, University of California, Davis, California 95616
| |
Collapse
|
35
|
Quantitative assessment of angiogenesis, perfused blood vessels and endothelial tip cells in the postnatal mouse brain. Nat Protoc 2014; 10:53-74. [PMID: 25502884 DOI: 10.1038/nprot.2015.002] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
During development and in various diseases of the CNS, new blood vessel formation starts with endothelial tip cell selection and vascular sprout migration, followed by the establishment of functional, perfused blood vessels. Here we describe a method that allows the assessment of these distinct angiogenic steps together with antibody-based protein detection in the postnatal mouse brain. Intravascular and perivascular markers such as Evans blue (EB), isolectin B4 (IB4) or laminin (LN) are used alongside simultaneous immunofluorescence on the same sections. By using confocal laser-scanning microscopy and stereological methods for analysis, detailed quantification of the 3D postnatal brain vasculature for perfused and nonperfused vessels (e.g., vascular volume fraction, vessel length and number, number of branch points and perfusion status of the newly formed vessels) and characterization of sprouting activity (e.g., endothelial tip cell density, filopodia number) can be obtained. The entire protocol, from mouse perfusion to vessel analysis, takes ∼10 d.
Collapse
|
36
|
Rai A, Cross JC. Three-dimensional cultures of trophoblast stem cells autonomously develop vascular-like spaces lined by trophoblast giant cells. Dev Biol 2014; 398:110-9. [PMID: 25499676 DOI: 10.1016/j.ydbio.2014.11.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 11/12/2014] [Accepted: 11/26/2014] [Indexed: 10/24/2022]
Abstract
The maternal blood space in the mouse placenta is lined not by endothelial cells but rather by various subtypes of trophoblast giant cells (TGCs), defined by their location and different patterns of gene expression. While TGCs invade the spiral arteries to displace the maternal endothelium, the rest of the vascular space is created de novo but the mechanisms are not well understood. We cultured mouse trophoblast stem (TS) cells in suspension and found that they readily form spheroids (trophospheres). Compared to cells grown in monolayer, differentiating trophospheres showed accelerated expression of TGC-specific genes. Morphological and gene expression studies showed that cavities form within the trophospheres that are primarily lined by Prl3d1/Pl1α-positive cells analogous to parietal-TGCs (P-TGCs) which line the maternal venous blood within the placenta. Lumen formation in trophospheres and in vivo was associated with cell polarization including CD34 sialomucin deposition on the apical side and cytoskeletal rearrangement. While P-TGCs preferentially formed in trophospheres at atmospheric oxygen levels (19%), decreasing oxygen to 3% shifted differentiation towards Ctsq-positive sinusoidal and/or channel TGCs. These studies show that trophoblast cells have the intrinsic ability to form vascular channels in ways analogous to endothelial cells. The trophosphere system will be valuable for assessing mechanisms that regulate specification of different TGC subtypes and their morphogenesis into vascular spaces.
Collapse
Affiliation(s)
- Anshita Rai
- Departments of Biochemistry and Molecular Biology, Comparative Biology and Experimental Medicine, Obstetrics and Gynecology, and Medical Genetics, University of Calgary, HSC Room 2279, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1
| | - James C Cross
- Departments of Biochemistry and Molecular Biology, Comparative Biology and Experimental Medicine, Obstetrics and Gynecology, and Medical Genetics, University of Calgary, HSC Room 2279, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1.
| |
Collapse
|
37
|
Abstract
Chronic progressive renal fibrosis leads to end-stage renal failure many patients with chronic kidney disease (CKD). Loss of the rich peritubular capillary network is a prominent feature, and seems independent of the specific underlying disease. The mechanisms that contribute to peritubular capillary regression include the loss of glomerular perfusion, as flow-dependent shear forces are required to provide the survival signal for endothelial cells. Also, reduced endothelial cell survival signals from sclerotic glomeruli and atrophic or injured tubule epithelial cells contribute to peritubular capillary regression. In response to direct tubular epithelial cell injury, and the inflammatory reaction that ensues, capillary pericytes dissociate from their blood vessels, also reducing endothelial cell survival. In addition, direct inflammatory injury of capillary endothelial cells, for instance in chronic allograft nephropathy, also contributes to capillary dropout. Chronic tissue hypoxia, which ensues from the rarefaction of the peritubular capillary network, can generate both an angiogenic and a fibrogenic response. However, in CKD, the balance is strongly tipped toward fibrogenesis. Understanding the underlying mechanisms for failed angiogenesis in CKD and harnessing endothelial-specific survival and pro-angiogenic mechanisms for therapy should be our goal if we are to reduce the disease burden from CKD.
Collapse
Affiliation(s)
| | - Marya Obeidat
- Department of Medicine, University of Alberta , Edmonton, Alberta, Canada
| |
Collapse
|
38
|
Newey SE, Tsaknakis G, Khoo CP, Athanassopoulos T, Camicia R, Zhang Y, Grabowska R, Harris AL, Roubelakis MG, Watt SM. The hematopoietic chemokine CXCL12 promotes integration of human endothelial colony forming cell-derived cells into immature vessel networks. Stem Cells Dev 2014; 23:2730-43. [PMID: 24940843 DOI: 10.1089/scd.2014.0005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Proangiogenic factors, vascular endothelial growth factor (VEGF), and fibroblast growth factor-2 (FGF-2) prime endothelial cells to respond to "hematopoietic" chemokines and cytokines by inducing/upregulating expression of the respective chemokine/cytokine receptors. Coculture of human endothelial colony forming cell (ECFC)-derived cells with human stromal cells in the presence of VEGF and FGF-2 for 14 days resulted in upregulation of the "hematopoietic" chemokine CXCL12 and its CXCR4 receptor by day 3 of coculture. Chronic exposure to the CXCR4 antagonist AMD3100 in this vasculo/angiogenesis assay significantly reduced vascular tubule formation, an observation recapitulated by delayed AMD3100 addition. While AMD3100 did not affect ECFC-derived cell proliferation, it did demonstrate a dual action. First, over the later stages of the 14-day cocultures, AMD3100 delayed tubule organization into maturing vessel networks, resulting in enhanced endothelial cell retraction and loss of complexity as defined by live cell imaging. Second, at earlier stages of cocultures, we observed that AMD3100 significantly inhibited the integration of exogenous ECFC-derived cells into established, but immature, vascular networks. Comparative proteome profiler array analyses of ECFC-derived cells treated with AMD3100 identified changes in expression of potential candidate molecules involved in adhesion and/or migration. Blocking antibodies to CD31, but not CD146 or CD166, reduced the ECFC-derived cell integration into these extant vascular networks. Thus, CXCL12 plays a key role not only in endothelial cell sensing and guidance, but also in promoting the integration of ECFC-derived cells into developing vascular networks.
Collapse
Affiliation(s)
- Sarah E Newey
- 1 Stem Cell Research Laboratory , Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, and NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Wang Y, Qian DEJ, Zhong WY, Lu JH, Guo XK, Cao YL, Liu J. TGF-β1 induces the formation of vascular-like structures in embryoid bodies derived from human embryonic stem cells. Exp Ther Med 2014; 8:52-58. [PMID: 24944596 PMCID: PMC4061233 DOI: 10.3892/etm.2014.1721] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 05/14/2014] [Indexed: 12/17/2022] Open
Abstract
Human embryonic stem cells (ESCs) can differentiate into endothelial cells in response to stimuli from extracellular cytokines. Transforming growth factor (TGF)-β1 signaling is involved in stem cell renewal and vascular development. Previously, human ESCs were isolated from inner cell mass and a stable ESC line was developed. In the present study, the effects of extracellular TGF-β1 were investigated on human ESC-derived embryoid bodies (EB) in suspension. The structures of the EBs were analyzed with light and electron microscopy, while the cellular composition of the EBs was examined via the expression levels of specific markers. Vascular-like tubular structures and cardiomyocyte-like beating cells were observed in the EBs at day 3 and 8, respectively. The frequencies of vascular-like structures and beating cells in the TGF-β1 treated group were significantly higher compared with the control group (84.31 vs. 12.77%; P<0.001; 37.25 vs. 8.51%; P<0.001, respectively). Electron microscopy revealed the presence of lumens and gap junctions in the sections of the tubular structures. Semiquantitative polymerase chain reaction revealed elevated expression levels of CD31 and fetal liver kinase-1 in EBs cultured with TGF-β1. In addition, extensive staining of von Willebrand factor was observed in the vascular-like structures of TGF-β1-treated EBs. Therefore, the results of the present study may aid the understanding of the underlying mechanisms of human ESC differentiation and improve the methods of propagating specific cell types for the clinical therapy of cardiovascular diseases.
Collapse
Affiliation(s)
- Yan Wang
- Department of Plastic Surgery, Provincial Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| | - DE-Jian Qian
- Department of Plastic Surgery, Provincial Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| | - Wen-Yu Zhong
- Department of Gynecology and Obstetrics, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
| | - Jun-Hong Lu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai 200011, P.R. China
| | - Xiang-Kai Guo
- Department of Plastic Surgery, Provincial Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| | - Yi-Lin Cao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai 200011, P.R. China
| | - Ju Liu
- Department of Plastic Surgery, Provincial Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China ; Medical Research Center, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
40
|
Neufeld S, Planas-Paz L, Lammert E. Blood and lymphatic vascular tube formation in mouse. Semin Cell Dev Biol 2014; 31:115-23. [PMID: 24631829 DOI: 10.1016/j.semcdb.2014.02.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 02/26/2014] [Indexed: 12/30/2022]
Abstract
The blood and lymphatic vasculatures are essential for nutrient delivery, gas exchange and fluid homeostasis in all tissues of higher vertebrates. They are composed of a hierarchical network of vessels, which are lined by vascular or lymphatic endothelial cells. For blood vascular lumen formation to occur, endothelial cell cords polarize creating apposing apical cell surfaces, which repulse each other and give rise to a small intercellular lumen. Following cell shape changes, the vascular lumen expands. Various junctional proteins, polarity complexes, extracellular matrix binding and actin remodelling molecules are required for blood vascular lumen formation. In contrast, little is known regarding the molecular mechanisms leading to lymphatic vascular tube formation. Current models agree that lymphatic vessels share a blood vessel origin, but they differ in identifying the mechanism by which a lymphatic lumen is formed. A ballooning mechanism was proposed, in which lymph sacs are connected via their lumen to the cardinal veins. Alternatively, a mechanism involving budding of streams of lymphatic endothelial cells from either the cardinal veins or both the cardinal veins and the intersomitic vessels, and subsequent assembly and lumenisation was recently described. Here, we discuss what is currently known about the molecular and cellular machinery that guides blood and lymphatic vascular tube formation in mouse.
Collapse
Affiliation(s)
- Sofia Neufeld
- Institute of Metabolic Physiology, Heinrich-Heine Universität Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Lara Planas-Paz
- Institute of Metabolic Physiology, Heinrich-Heine Universität Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Eckhard Lammert
- Institute of Metabolic Physiology, Heinrich-Heine Universität Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany; Institute for Beta Cell Biology, German Diabetes Center, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany.
| |
Collapse
|
41
|
Rai A, Cross JC. Development of the hemochorial maternal vascular spaces in the placenta through endothelial and vasculogenic mimicry. Dev Biol 2014; 387:131-41. [PMID: 24485853 DOI: 10.1016/j.ydbio.2014.01.015] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 01/14/2014] [Accepted: 01/19/2014] [Indexed: 11/27/2022]
Abstract
The maternal vasculature within the placenta in primates and rodents is unique because it is lined by fetal cells of the trophoblast lineage and not by maternal endothelial cells. In addition to trophoblast cells that invade the uterine spiral arteries that bring blood into the placenta, other trophoblast subtypes sit at different levels of the vascular space. In mice, at least five distinct subtypes of trophoblast cells have been identified which engage maternal endothelial cells on the arterial and venous frontiers of the placenta, but which also form the channel-like spaces within it through a process analogous to formation of blood vessels (vasculogenic mimicry). These cells are all large, post-mitotic trophoblast giant cells. In addition to assuming endothelial cell-like characteristics (endothelial mimicry), they produce dozens of different hormones that are thought to regulate local and systemic maternal adaptations to pregnancy. Recent work has identified distinct molecular pathways in mice that regulate the morphogenesis of trophoblast cells on the arterial and venous sides of the vascular circuit that may be analogous to specification of arterial and venous endothelial cells.
Collapse
Affiliation(s)
- Anshita Rai
- Department of Biochemistry and Molecular Biology, University of Calgary, HSC Room 2279, 3330 Hospital Drive NW, Calgary, Alta., Canada; Department of Comparative Biology and Experimental Medicine, Obstetrics and Gynecology, and Medical Genetics, University of Calgary, HSC Room 2279, 3330 Hospital Drive NW, Calgary, Alta., Canada T2N 4N1
| | - James C Cross
- Department of Biochemistry and Molecular Biology, University of Calgary, HSC Room 2279, 3330 Hospital Drive NW, Calgary, Alta., Canada; Department of Comparative Biology and Experimental Medicine, Obstetrics and Gynecology, and Medical Genetics, University of Calgary, HSC Room 2279, 3330 Hospital Drive NW, Calgary, Alta., Canada T2N 4N1.
| |
Collapse
|
42
|
Wittko-Schneider IM, Schneider FT, Plate KH. Cerebral angiogenesis during development: who is conducting the orchestra? Methods Mol Biol 2014; 1135:3-20. [PMID: 24510850 DOI: 10.1007/978-1-4939-0320-7_1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Blood vessels provide the brain with the oxygen and the nutrients it requires to develop and function. Endothelial cells (ECs) are the principal cell type forming the vascular system and driving its development and remodeling. All vessels are lined by a single EC layer. Larger blood vessels are additionally enveloped by vascular smooth muscle cells (VSMCs) and pericytes, which increase their stability and regulate their perfusion and form the blood-brain barrier (BBB). The development of the vascular system occurs by two processes: (1) vasculogenesis, the de novo assembly of the first blood vessels, and (2) angiogenesis, the creation of new blood vessels from preexisting ones by sprouting from or by division of the original vessel. The walls of maturing vessels produce a basal lamina and recruit pericytes and vascular smooth muscle cells for structural support. Whereas the process of vasculogenesis seems to be genetically programmed, angiogenesis is induced mainly by hypoxia in development and disease. Both processes and the subsequent vessel maturation are further orchestrated by a complex interplay of inhibiting and stimulating growth factors and their respective receptors, many of which are hypoxia-inducible. This chapter intends to give an overview about the array of factors directing the development and maintenance of the brain vasculature and their interdependent actions.
Collapse
Affiliation(s)
- Ina M Wittko-Schneider
- Institute for Stroke and Dementia Research, Klinikum der Universität München, München, Germany
| | | | | |
Collapse
|
43
|
Abstract
Mutual, homophilic cell-cell adhesion between epithelial cells is required for proper maintenance of epithelial barrier function. Whereas opposing membranes from neighboring cells rapidly assemble junctional complexes, self-contacting membranes curiously do not, suggesting that cells have the ability to prevent the maturation of self-junctions. Using a self-contact-inducing microfabricated substrate, we show that self-contacts of normal epithelial cells are rapidly eliminated by membrane fusion between two opposing plasma membranes of a single cell. This membrane fusion is most frequently observed in E-cadherin-expressing epithelial cells, but not in fibroblasts. The efficiency of self-contact elimination depends on extracellular calcium concentration and the level of E-cadherin, suggesting that E-cadherin, although not required, enhances membrane fusion efficiency by bringing opposing membranes into close apposition to one another. Additionally, Rho-associated protein kinase inhibition decreases self-contact-induced membrane fusion of epithelial cells, suggesting that this fusion may be mechanically regulated through the actin-myosin network. This self-contact-induced membrane fusion is a key elimination mechanism for unwanted self-junctions and may be a feature of cell self-recognition.
Collapse
|
44
|
Chaki SP, Rivera GM. Integration of signaling and cytoskeletal remodeling by Nck in directional cell migration. BIOARCHITECTURE 2013; 3:57-63. [PMID: 23887203 PMCID: PMC3782540 DOI: 10.4161/bioa.25744] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Planar and apical-basal cellular polarization of epithelia and endothelia are crucial during morphogenesis. The establishment of these distinct polarity states and their transitions are regulated by signaling networks that include polarity complexes, Rho GTPases, and phosphoinositides. The spatiotemporal coordination of signaling by these molecules modulates cytoskeletal remodeling and vesicle trafficking to specify membrane domains, a prerequisite for the organization of tissues and organs. Here we present an overview of how activation of the WASp/Arp2/3 pathway of actin remodeling by Nck coordinates directional cell migration and speculate on its role as a signaling integrator in the coordination of cellular processes involved in endothelial cell polarity and vascular lumen formation.
Collapse
Affiliation(s)
- Sankar P Chaki
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, USA
| | | |
Collapse
|