201
|
Rustenhoven J, Smyth LC, Jansson D, Schweder P, Aalderink M, Scotter EL, Mee EW, Faull RLM, Park TIH, Dragunow M. Modelling physiological and pathological conditions to study pericyte biology in brain function and dysfunction. BMC Neurosci 2018; 19:6. [PMID: 29471788 PMCID: PMC5824614 DOI: 10.1186/s12868-018-0405-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/10/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Brain pericytes ensheathe the endothelium and contribute to formation and maintenance of the blood-brain-barrier. Additionally, pericytes are involved in several aspects of the CNS immune response including scarring, adhesion molecule expression, chemokine secretion, and phagocytosis. In vitro cultures are routinely used to investigate these functions of brain pericytes, however, these are highly plastic cells and can display differing phenotypes and functional responses depending on their culture conditions. Here we sought to investigate how two commonly used culture media, high serum containing DMEM/F12 and low serum containing Pericyte Medium (ScienCell), altered the phenotype of human brain pericytes and neuroinflammatory responses. METHODS Pericytes were isolated from adult human brain biopsy tissue and cultured in DMEM/F12 (D-pericytes) or Pericyte Medium (P-pericytes). Immunocytochemistry, qRT-PCR, and EdU incorporation were used to determine how this altered their basal phenotype, including the expression of pericyte markers, proliferation, and cell morphology. To determine whether culture media altered the inflammatory response in human brain pericytes, immunocytochemistry, qRT-PCR, cytometric bead arrays, and flow cytometry were used to investigate transcription factor induction, chemokine secretion, adhesion molecule expression, migration, phagocytosis, and response to inflammatory-related growth factors. RESULTS P-pericytes displayed elevated proliferation and a distinct bipolar morphology compared to D-pericytes. Additionally, P-pericytes displayed lower expression of pericyte-associated markers NG2, PDGFRβ, and fibronectin, with notably lower αSMA, CD146, P4H and desmin, and higher Col-IV expression. Nuclear NF-kB translocation in response to IL-1β stimulation was observed in both cultures, however, P-pericytes displayed elevated expression of the transcription factor C/EBPδ, and lower expression of the adhesion molecule ICAM-1. P-pericytes displayed elevated phagocytic and migratory ability. Both cultures responded similarly to stimulation by the growth factors TGFβ1 and PDGF-BB. CONCLUSIONS Despite differences in their phenotype and magnitude of response, both P-pericytes and D-pericytes responded similarly to all examined functions, indicating that the neuroinflammatory phenotype of these cells is robust to culture conditions.
Collapse
Affiliation(s)
- Justin Rustenhoven
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand.,Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand
| | - Leon C Smyth
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand.,Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand
| | - Deidre Jansson
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand.,Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand
| | - Patrick Schweder
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand.,Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand.,Auckland City Hospital, Auckland, New Zealand
| | - Miranda Aalderink
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand.,Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand
| | - Emma L Scotter
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand.,Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand
| | - Edward W Mee
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand.,Auckland City Hospital, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand.,Department of Anatomy and Medical Imagining, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand
| | - Thomas I-H Park
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand.,Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand
| | - Mike Dragunow
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand. .,Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Rd, Grafton, Auckland, 1023, New Zealand. .,Department of Pharmacology and Clinical Pharmacology, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
| |
Collapse
|
202
|
Yan M, Hu Y, Yao M, Bao S, Fang Y. GM-CSF ameliorates microvascular barrier integrity via pericyte-derived Ang-1 in wound healing. Wound Repair Regen 2018; 25:933-943. [PMID: 29328541 DOI: 10.1111/wrr.12608] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 12/04/2017] [Indexed: 12/20/2022]
Abstract
Skin wound healing involves complex coordinated interactions of cells, tissues, and mediators. Maintaining microvascular barrier integrity is one of the key events for endothelial homeostasis during wound healing. Vasodilation is observed after vasoconstriction, which causes blood vessels to become porous, facilitates leukocyte infiltration and aids angiogenesis at the wound-area, postinjury. Eventually, vessel integrity has to be reestablished for vascular maturation. Numerous studies have found that granulocyte macrophage colony-stimulating factor (GM-CSF) accelerates wound healing by inducing recruitment of repair cells into the injury area and releases of cytokines. However, whether GM-CSF is involving in the maintaining of microvascular barrier integrity and the underlying mechanism remain still unclear. Aim of this study was to investigate the effects of GM-CSF on modulation of microvascular permeability in wound healing and underlying mechanisms. Wound closure and microvascular leakage was investigated using a full-thickness skin wound mouse model after GM-CSF intervention. The endothelial permeability was measured by Evans blue assay in vivo and in vitro endothelium/pericyte co-culture system using a FITC-Dextran permeability assay. To identify the source of angiopoietin-1 (Ang-1), double staining is used in vivo and ELISA and qPCR are used in vitro. To determine the specific effect of Ang-1 on GM-CSF maintaining microvascular stabilization, Ang-1 siRNA was applied to inhibit Ang-1 production in vivo and in vitro. Wound closure was significantly accelerated and microvascular leakage was ameliorated after GM-CSF treatment in mouse wound sites. GM-CSF decreased endothelial permeability through tightening endothelial junctions and increased Ang-1 protein level that was derived by perictye. Furthermore, applications of siRNAAng-1 inhibited GM-CSF mediated protection of microvascular barrier integrity both in vivo and in vitro. Our data indicate that GM-CSF ameliorates microvascular barrier integrity via pericyte-derived Ang-1 during wound healing.
Collapse
Affiliation(s)
- Min Yan
- Department of Plastic Surgery, The Ninth People's Hospital, Shanghai Jiaotong University of Medicine, Shanghai, China
| | - Yange Hu
- Department of Plastic Surgery, The Ninth People's Hospital, Shanghai Jiaotong University of Medicine, Shanghai, China
| | - Min Yao
- Department of Plastic Surgery, The Ninth People's Hospital, Shanghai Jiaotong University of Medicine, Shanghai, China
| | - Shisan Bao
- Discipline of Pathology, Bosch Institute and School of Medical Sciences, The University of Sydney, Sydney, Australia
| | - Yong Fang
- Department of Plastic Surgery, The Ninth People's Hospital, Shanghai Jiaotong University of Medicine, Shanghai, China
| |
Collapse
|
203
|
Manikowski D, Andrée B, Samper E, Saint-Marc C, Olmer R, Vogt P, Strauß S, Haverich A, Hilfiker A. Human adipose tissue-derived stromal cells in combination with exogenous stimuli facilitate three-dimensional network formation of human endothelial cells derived from various sources. Vascul Pharmacol 2018; 106:28-36. [PMID: 29452238 DOI: 10.1016/j.vph.2018.02.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/16/2018] [Accepted: 02/11/2018] [Indexed: 01/22/2023]
Abstract
In natural tissues, the nutrition of cells and removal of waste products is facilitated by a dense capillary network which is generated during development. This perfusion system is also indispensable for tissue formation in vitro. Nutrition depending solely on diffusion is not sufficient to generate tissues of clinically relevant dimensions, which is a core aim in tissue engineering research. In this study, the establishment of a vascular network was investigated in a self-assembling approach employing endothelial and mural cells. The process of vascularization was analyzed in constructs based on a carrier matrix of decellularized porcine small intestinal submucosa (SIS). A three-dimensional hydrogel containing Matrigel™, collagen, and respective cells was casted on top of the SIS. Various types of human endothelial cells (hECs), e.g. HUVECs, cardiac tissue ECs (hCECs), pulmonary artery ECs (hPAECs), and iPSC-derived ECs, were co-cultured with human adipose tissue-derived stromal cells (hASCs) within the hydrogel. Analyzed hECs were able to self-assemble and form three-dimensional networks harboring small caliber lumens within the hydrogel constructs in the presence of hASCs as supporting cells. Additionally, microvessel assembling required exogenous growth factor supplementation. This study demonstrates the development of stable vascularized hydrogels applying hASCs as mural cells in combination with various types of hECs, paving the way for the generation of clinically applicable tissue engineered constructs.
Collapse
Affiliation(s)
- Dominique Manikowski
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany.
| | - Birgit Andrée
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany.
| | - Esther Samper
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany.
| | - Clémence Saint-Marc
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany.
| | - Ruth Olmer
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany; Hannover Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, Hannover, Germany.
| | - Peter Vogt
- Department of Plastic, Hand- and Reconstructive Surgery, Hannover Medical School, Hannover, Germany.
| | - Sarah Strauß
- Department of Plastic, Hand- and Reconstructive Surgery, Hannover Medical School, Hannover, Germany.
| | - Axel Haverich
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany.
| | - Andres Hilfiker
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany.
| |
Collapse
|
204
|
Salvador J, Davis GE. Evaluation and Characterization of Endothelial Cell Invasion and Sprouting Behavior. Methods Mol Biol 2018; 1846:249-259. [PMID: 30242764 DOI: 10.1007/978-1-4939-8712-2_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Here, we describe highly reproducible methods to investigate human EC invasion and sprouting behavior in 3D collagen matrices. Two assay models are presented whereby ECs are induced to sprout from a monolayer surface or from aggregated ECs suspended within a collagen gel matrix. In each case, the assays are performed using serum-free defined media containing a combination of five growth factors (Factors): FGF-2, SCF, IL-3, SDF-1α, and insulin. In both models, marked EC sprouting occurs with leading EC tip cells over a 12-24 h period. To illustrate their utility, we present data showing the influence of various pharmacologic inhibitors directed to membrane-type matrix metalloproteinases (MT-MMPs), protein kinase C alpha (PKCα), Src family kinases, and Notch-dependent signaling. Marked inhibition of sprouting is observed after blockade of MT-MMPs and PKCα, while strong increases in sprouting and EC tip cell number is observed following blockade of Src kinases, Notch signaling or both. Interestingly, the increased sprouting behavior observed following Src or Notch blockade directly correlates with a loss in the ability of ECs to form lumens. These defined in vitro assay models allow for a genetic and signaling dissection of EC tip cells vs. lumen forming ECs, which are both necessary for the formation of branching networks of tubes during vascular morphogenic events.
Collapse
Affiliation(s)
- Jocelynda Salvador
- Department of Medical Pharmacology and Physiology, Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO, USA
| | - George E Davis
- Department of Medical Pharmacology and Physiology, Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, MO, USA.
| |
Collapse
|
205
|
Abstract
The role of pericytes seems to extend beyond their known function in angiogenesis, fibrosis and wound healing, blood-brain barrier maintenance, and blood flow regulation. More and more data are currently accumulating indicating that pericytes, uniquely positioned at the interface between blood and parenchyma, secrete a large plethora of different molecules in response to microenvironmental changes. Their secretome is tissue-specific and stimulus-specific and includes pro- and anti-inflammatory factors, growth factors, and extracellular matrix as well as microvesicles suggesting the important role of pericytes in the regulation of immune response and immune evasion of tumors. However, the angiogenic and trophic secretome of pericytes indicates that their secretome plays a role in physiological homeostasis but possibly also in disease progression or could be exploited for regenerative processes in the future. This book chapter summarizes the current data on the secretory properties of pericytes from different tissues in response to certain pathological stimuli such as inflammatory stimuli, hypoxia, high glucose, and others and thereby aims to provide insights into the possible role of pericytes in these conditions.
Collapse
Affiliation(s)
- Abderahim Gaceb
- Translational Neurology Group, Department of Clinical Sciences and Wallenberg Center for Molecular Medicine, Department of Neurology, Lund University, Lund, Sweden
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Sciences and Wallenberg Center for Molecular Medicine, Department of Neurology, Lund University, Lund, Sweden. .,Skåne University Hospital, Lund, Sweden.
| |
Collapse
|
206
|
Abstract
Cell migration is an adaptive process that depends on and responds to physical and molecular triggers. Moving cells sense and respond to tissue mechanics and induce transient or permanent tissue modifications, including extracellular matrix stiffening, compression and deformation, protein unfolding, proteolytic remodelling and jamming transitions. Here we discuss how the bi-directional relationship of cell-tissue interactions (mechanoreciprocity) allows cells to change position and contributes to single-cell and collective movement, structural and molecular tissue organization, and cell fate decisions.
Collapse
|
207
|
Rayner SG, Zheng Y. Engineered Microvessels for the Study of Human Disease. J Biomech Eng 2017; 138:2545529. [PMID: 27537085 DOI: 10.1115/1.4034428] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Indexed: 12/22/2022]
Abstract
The microvasculature is an extensive, heterogeneous, and complex system that plays a critical role in human physiology and disease. It nourishes almost all living human cells and maintains a local microenvironment that is vital for tissue and organ function. Operating under a state of continuous flow, with an intricate architecture despite its small caliber, and subject to a multitude of biophysical and biochemical stimuli, the microvasculature can be a complex subject to study in the laboratory setting. Engineered microvessels provide an ideal platform that recapitulates essential elements of in vivo physiology and allows study of the microvasculature in a precise and reproducible way. Here, we review relevant structural and functional vascular biology, discuss different methods to engineer microvessels, and explore the applications of this exciting tool for the study of human disease.
Collapse
Affiliation(s)
- Samuel G Rayner
- Department of Pulmonary and Critical Care Medicine, University of Washington School of Medicine, Campus Box 356522, Seattle, WA 98195 e-mail:
| | - Ying Zheng
- Department of Bioengineering, University of Washington, 3720 15th Avenue NE, Seattle, WA 98105;Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109 e-mail:
| |
Collapse
|
208
|
Turner CJ, Badu-Nkansah K, Hynes RO. Endothelium-derived fibronectin regulates neonatal vascular morphogenesis in an autocrine fashion. Angiogenesis 2017; 20:519-531. [PMID: 28667352 PMCID: PMC5660148 DOI: 10.1007/s10456-017-9563-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 06/21/2017] [Indexed: 12/02/2022]
Abstract
Fibronectin containing alternatively spliced EIIIA and EIIIB domains is largely absent from mature quiescent vessels in adults, but is highly expressed around blood vessels during developmental and pathological angiogenesis. The precise functions of fibronectin and its splice variants during developmental angiogenesis however remain unclear due to the presence of cardiac, somitic, mesodermal and neural defects in existing global fibronectin KO mouse models. Using a rare family of surviving EIIIA EIIIB double KO mice, as well as inducible endothelial-specific fibronectin-deficient mutant mice, we show that vascular development in the neonatal retina is regulated in an autocrine manner by endothelium-derived fibronectin, and requires both EIIIA and EIIIB domains and the RGD-binding α5 and αv integrins for its function. Exogenous sources of fibronectin do not fully substitute for the autocrine function of endothelial fibronectin, demonstrating that fibronectins from different sources contribute differentially to specific aspects of angiogenesis.
Collapse
Affiliation(s)
- Christopher J Turner
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave, 76-361, Cambridge, MA, 02139, USA
- University of Suffolk, James Hehir Building, University Avenue, Ipswich, Suffolk, IP3 0FS, UK
| | - Kwabena Badu-Nkansah
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave, 76-361, Cambridge, MA, 02139, USA
- Duke University Medical Center, 307 Research Drive, Durham, NC, 27710, USA
| | - Richard O Hynes
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave, 76-361, Cambridge, MA, 02139, USA.
| |
Collapse
|
209
|
Lykkemark S, Mandrup OA, Jensen MB, Just J, Kristensen P. A novel excision selection method for isolation of antibodies binding antigens expressed specifically by rare cells in tissue sections. Nucleic Acids Res 2017; 45:e107. [PMID: 28369551 PMCID: PMC5499801 DOI: 10.1093/nar/gkx207] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 03/21/2017] [Indexed: 12/22/2022] Open
Abstract
There is a growing appreciation of single cell technologies to provide increased biological insight and allow development of improved therapeutics. The central dogma explains why single cell technologies is further advanced in studies targeting nucleic acids compared to proteins, as nucleic acid amplification makes experimental detection possible. Here we describe a novel method for single round phage display selection of antibody fragments from genetic libraries targeting antigens expressed by rare cells in tissue sections. We present and discuss the results of two selections of antibodies recognizing antigens expressed by perivascular cells surrounding capillaries located in a human brain section; with the aim of identifying biomarkers expressed by pericytes. The area targeted for selection was identified by a known biomarker and morphological appearance, however in situ hybridizations to nucleic acids can also be used for the identification of target cells. The antibody selections were performed directly on the tissue sections followed by excision of the target cells using a glass capillary attached to micromanipulation equipment. Antibodies bound to the target cells were characterized using ELISA, immunocytochemistry and immunohistochemistry. The described method will provide a valuable tool for the discovery of novel biomarkers on rare cells in all types of tissues.
Collapse
Affiliation(s)
- Simon Lykkemark
- Department of Clinical Medicine, Aarhus University, Nørrebrogade 44, 8000 Aarhus C, Denmark.,Sino-Danish Centre for Education and Research (SDC), Niels Jensens Vej 2, 8000 Aarhus C, Denmark
| | - Ole Aalund Mandrup
- Department of Engineering, Aarhus University, Gustav Wieds Vej 10, 8000 Aarhus C, Denmark
| | - Mads Bjørnkjær Jensen
- Department of Engineering, Aarhus University, Gustav Wieds Vej 10, 8000 Aarhus C, Denmark
| | - Jesper Just
- Department of Molecular Biology and Genetics, Gustav Wieds Vej 10, 8000 Aarhus C, Denmark
| | - Peter Kristensen
- Department of Engineering, Aarhus University, Gustav Wieds Vej 10, 8000 Aarhus C, Denmark
| |
Collapse
|
210
|
Rocca A, Tafuri D, Paccone M, Giuliani A, Zamboli AGI, Surfaro G, Paccone A, Compagna R, Amato M, Serra R, Amato B. Cell Based Therapeutic Approach in Vascular Surgery: Application and Review. Open Med (Wars) 2017; 12:308-322. [PMID: 29071303 PMCID: PMC5651406 DOI: 10.1515/med-2017-0045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 08/16/2017] [Indexed: 01/14/2023] Open
Abstract
Multipotent stem cells - such as mesenchymal stem/stromal cells and stem cells derived from different sources like vascular wall are intensely studied to try to rapidly translate their discovered features from bench to bedside. Vascular wall resident stem cells recruitment, differentiation, survival, proliferation, growth factor production, and signaling pathways transduced were analyzed. We studied biological properties of vascular resident stem cells and explored the relationship from several factors as Matrix Metalloproteinases (MMPs) and regulations of biological, translational and clinical features of these cells. In this review we described a translational and clinical approach to Adult Vascular Wall Resident Multipotent Vascular Stem Cells (VW-SCs) and reported their involvement in alternative clinical approach as cells based therapy in vascular disease like arterial aneurysms or peripheral arterial obstructive disease.
Collapse
Affiliation(s)
- Aldo Rocca
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, ItalyVia Sergio Pansini, 80131Naples, Italy
| | - Domenico Tafuri
- Department of Sport Sciences and Wellness, University of Naples “Parthenope”, Naples, Italy
| | - Marianna Paccone
- Department of Medicine and Health Sciences Vincenzo Tiberio, University of Molise, Campobasso, Italy
| | - Antonio Giuliani
- A.O.R.N. A. Cardarelli Hepatobiliary and Liver Transplatation Center, Naples, Italy
| | | | - Giuseppe Surfaro
- Antonio Cardarelli Hospital, General Surgery Unit, Campobasso, Italy
| | - Andrea Paccone
- Department of Medicine and Health Sciences Vincenzo Tiberio, University of Molise, Campobasso, Italy
| | - Rita Compagna
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Maurizo Amato
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Raffaele Serra
- Department of Medical and Surgical Sciences, University of Catanzaro, Catanzaro, Italy
| | - Bruno Amato
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
211
|
Zhang C, Hu K, Liu X, Reynolds MA, Bao C, Wang P, Zhao L, Xu HH. Novel hiPSC-based tri-culture for pre-vascularization of calcium phosphate scaffold to enhance bone and vessel formation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017. [DOI: 10.1016/j.msec.2017.05.035] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
212
|
Cho CS, Lee SM, Lee BJ, Jo DH, Kim JH, Kim JH, Yu YS. Chronological Changes in Tip Cells during Sprouting Angiogenesis of Development of the Retinal Vasculature in Newborn Mice. Curr Eye Res 2017; 42:1511-1517. [PMID: 28925736 DOI: 10.1080/02713683.2017.1347691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE To investigate a sequential chronological change in tip cells during the development of the retinal vasculature in newborn mice. MATERIALS AND METHODS Newborn C57BL/6 mice were used for this study. To elucidate the patterns in the developing retinal vasculature, histology, and immunohistochemistry-antiplatelet endothelial cell adhesion molecule-1, anticollagen type IV, isolectin IB4-were performed on sections of mouse retina on postnatal days (P)-4, -8, and -12. Staining patterns of isolectin IB4-stained arterial and venous tip cells were compared in retinal wholemounts, in which the numbers and characteristics of tip cells were compared between arteries and veins on P-4, -6, and -8. In addition, vascular densities and branching patterns were compared between arterial and venous vascular forefront areas. RESULTS Tip cells in the superficial vascular plexus were observed until P-8. The number of tip cells was highest on P-6, decreasing dramatically from P-6 to P-8 (P-4, 165.2 ± 10.1, n = 17; P-6, 183.8 ± 19.4, n = 15; P8, 21.4 ± 6.4, n = 15) (p < 0.05, respectively, t-test). There was a greater number of tip cells in veins versus arteries on P-4 and P-6 (P-4, 91.0 ± 9.2 veins versus 74.2 ± 10.4 arteries; P-6, 104.0 ± 10.2 veins versus 79.8 ± 11.3 arteries) (p < 0.05, respectively). Arterial tip cells had thinner and longer sprouts compared with venous tip cells (basal thickness: 15.7 ± 8.7 veins versus 9.9 ± 3.5 μm arteries) (length, 20.3 ± 9.1 veins versus 37.1 ± 13.2 μm arteries on P-4) (p < 0.05, respectively). Vessel areas and densities of vascular branch points were significantly higher around veins compared to arteries (vessel areas: 58.9 ± 1.2% veins versus 40.8 ± 1.9% arteries; vascular branch points, 1371.9 ± 136.7/mm2 veins versus 1046.7 ± 175.5/mm2 arteries) (p < 0.05, respectively). CONCLUSION The number of tip cells increased to a greater extent in the superficial vascular plexus of veins versus arteries until P-6. Consequently, there are more vessel areas and vascular branch points near retinal veins versus arteries. Arterial tip cells are longer and thinner than the shorter and thicker venous tip cells.
Collapse
Affiliation(s)
- Chang Sik Cho
- a Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute , Seoul National University Hospital , Seoul , South Korea
| | - Sang-Mok Lee
- b Department of Ophthalmology , Hallym University Sacred Heart Hospital , Gyeonggi-do , South Korea
| | - Byung Joo Lee
- a Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute , Seoul National University Hospital , Seoul , South Korea
| | - Dong Hyun Jo
- a Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute , Seoul National University Hospital , Seoul , South Korea
| | - Jin Hyoung Kim
- a Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute , Seoul National University Hospital , Seoul , South Korea
| | - Jeong Hun Kim
- a Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute , Seoul National University Hospital , Seoul , South Korea.,c Department of Ophthalmology , Seoul National University College of Medicine , Seoul , South Korea.,d Seoul Artificial Eye Center , Clinical Research Institute, Seoul National University Hospital , Seoul , South Korea
| | - Young Suk Yu
- a Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute , Seoul National University Hospital , Seoul , South Korea.,c Department of Ophthalmology , Seoul National University College of Medicine , Seoul , South Korea.,d Seoul Artificial Eye Center , Clinical Research Institute, Seoul National University Hospital , Seoul , South Korea
| |
Collapse
|
213
|
Kim DJ, Norden PR, Salvador J, Barry DM, Bowers SLK, Cleaver O, Davis GE. Src- and Fyn-dependent apical membrane trafficking events control endothelial lumen formation during vascular tube morphogenesis. PLoS One 2017; 12:e0184461. [PMID: 28910325 PMCID: PMC5598984 DOI: 10.1371/journal.pone.0184461] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 08/24/2017] [Indexed: 01/10/2023] Open
Abstract
Here we examine the question of how endothelial cells (ECs) develop their apical membrane surface domain during lumen and tube formation. We demonstrate marked apical membrane targeting of activated Src kinases to this apical domain during early and late stages of this process. Immunostaining for phosphotyrosine or phospho-Src reveals apical membrane staining in intracellular vacuoles initially. This is then followed by vacuole to vacuole fusion events to generate an apical luminal membrane, which is similarly decorated with activated phospho-Src kinases. Functional blockade of Src kinases completely blocks EC lumen and tube formation, whether this occurs during vasculogenic tube assembly or angiogenic sprouting events. Multiple Src kinases participate in this apical membrane formation process and siRNA suppression of Src, Fyn and Yes, but not Lyn, blocks EC lumen formation. We also demonstrate strong apical targeting of Src-GFP and Fyn-GFP fusion proteins and increasing their expression enhances lumen formation. Finally, we show that Src- and Fyn-associated vacuoles track and fuse along a subapically polarized microtubule cytoskeleton, which is highly acetylated. These vacuoles generate the apical luminal membrane in a stereotypically polarized, perinuclear position. Overall, our study identifies a critical role for Src kinases in creating and decorating the EC apical membrane surface during early and late stages of lumen and tube formation, a central event in the molecular control of vascular morphogenesis.
Collapse
Affiliation(s)
- Dae Joong Kim
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Dalton Cardiovascular Research Center, Columbia, MO, United States of America
| | - Pieter R Norden
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Dalton Cardiovascular Research Center, Columbia, MO, United States of America
| | - Jocelynda Salvador
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Dalton Cardiovascular Research Center, Columbia, MO, United States of America
| | - David M Barry
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas TX, United States of America
| | - Stephanie L K Bowers
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Dalton Cardiovascular Research Center, Columbia, MO, United States of America
| | - Ondine Cleaver
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas TX, United States of America
| | - George E Davis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Dalton Cardiovascular Research Center, Columbia, MO, United States of America
| |
Collapse
|
214
|
Ma Z, Li Z, Shou K, Jian C, Li P, Niu Y, Qi B, Yu A. Negative pressure wound therapy: Regulating blood flow perfusion and microvessel maturation through microvascular pericytes. Int J Mol Med 2017; 40:1415-1425. [PMID: 28901392 PMCID: PMC5627868 DOI: 10.3892/ijmm.2017.3131] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 07/11/2017] [Indexed: 01/18/2023] Open
Abstract
Negative pressure wound therapy (NPWT) has been demonstrated to accelerate wound healing by promoting angiogenesis. However, whether blood flow perfusion is regulated by microvessel maturation and pericytes following NPWT remains unclear, as well as the exact association between pericytes and collagen type IV. The aim of this study was to investigate the relevant association between blood flow perfusion and microvessel maturation and pericytes following NPWT, and to further explore the underlying molecular mechanisms. We also aimed to investigate the association between pericytes and collagen type IV. For this purpose, we created a rat model of diabetic wounds and microvascular blood flow perfusion was detected using a laser Doppler blood perfusion imager. The expression levels of angiogenin-1, tyrosine phosphorylation of tyrosine kinase receptor-2 (Tie-2), α-smooth muscle actin (α-SMA) and collagen type IV were detected and analyzed through immunohistochemistry, immunofluorescence, RT-qPCR and western blot analysis. The results revealed that NPWT promoted the overexpression of angiogenin-1, Tie-2, α-SMA and collagen type IV, and significantly increased blood flow perfusion coupled with microvessel maturation in the NPWT group at the later stages (7–10 days) of wound healing. Our results suggested that NPWT can preferentially enhance vessel maturation and increase the number of pericytes, thus regulating blood flow perfusion. On the other hand, pericytes and collagen type IV had a mutual interaction, promoting microvessel maturation.
Collapse
Affiliation(s)
- Zhanjun Ma
- Zhongnan Hospital of Wuhan University, Department of Orthopedics, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zonghuan Li
- Zhongnan Hospital of Wuhan University, Department of Orthopedics, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Kangquan Shou
- Zhongnan Hospital of Wuhan University, Department of Orthopedics, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Chao Jian
- Zhongnan Hospital of Wuhan University, Department of Orthopedics, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Pengcheng Li
- Zhongnan Hospital of Wuhan University, Department of Orthopedics, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yahui Niu
- Zhongnan Hospital of Wuhan University, Department of Orthopedics, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Baiwen Qi
- Zhongnan Hospital of Wuhan University, Department of Orthopedics, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Aixi Yu
- Zhongnan Hospital of Wuhan University, Department of Orthopedics, Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
215
|
You WK, Stallcup WB. Localization of VEGF to Vascular ECM Is an Important Aspect of Tumor Angiogenesis. Cancers (Basel) 2017; 9:cancers9080097. [PMID: 28788063 PMCID: PMC5575600 DOI: 10.3390/cancers9080097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 07/22/2017] [Accepted: 07/26/2017] [Indexed: 12/02/2022] Open
Abstract
Our research has identified several examples in which reduced VEGF-A binding to deficient vascular extracellular matrix leads to deficits in tumor vascularization and tumor growth: (1) germline ablation of collagen VI in the stroma of intracranial B16F10 melanomas; (2) knockdown of the Tks5 scaffolding protein in MDA-MB-231 mammary tumor cells; (3) germline ablation of NG2 proteoglycan in the stroma of MMTV-PyMT mammary tumors; and (4) myeloid-specific ablation of NG2 in the stroma of intracranial B16F10 melanomas. Tumor hypoxia is increased in each of the four types of experimental mice, accompanied by increases in total VEGF-A. However, while VEGF-A is highly associated with tumor blood vessels in control mice, it is much more diffusely distributed in tumors in all four sets of experimental mice, likely due to reduced extent of the vascular extracellular matrix. In parallel to lost VEGF-A localization, tumor vessels in each case have smaller diameters and are leakier than tumor vessels in control mice. Tumor growth is decreased as a result of this poor vascular function. The fact that the observed vascular changes occur in the absence of alterations in vascular density suggests that examination of vessel structure and function is more useful than vascular density for understanding the importance of angiogenesis in tumor progression.
Collapse
Affiliation(s)
| | - William B Stallcup
- Tumor Microenvironment and Cancer Immunology Program, Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
216
|
Li X, Chen M, Lei X, Huang M, Ye W, Zhang R, Zhang D. Luteolin inhibits angiogenesis by blocking Gas6/Axl signaling pathway. Int J Oncol 2017. [DOI: 10.3892/ijo.2017.4041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
217
|
Lauridsen HM, Pellowe AS, Ramanathan A, Liu R, Miller-Jensen K, McNiff JM, Pober JS, Gonzalez AL. Tumor Necrosis Factor-α and IL-17A Activation Induces Pericyte-Mediated Basement Membrane Remodeling in Human Neutrophilic Dermatoses. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1893-1906. [PMID: 28609645 DOI: 10.1016/j.ajpath.2017.04.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/03/2017] [Accepted: 04/24/2017] [Indexed: 12/11/2022]
Abstract
Sweet syndrome (SS) is a prototypical neutrophilic dermatosis, a class of inflammatory diseases marked by elevated levels of tumor necrosis factor (TNF)-α and IL-17A, pathologic neutrophil recruitment, and microvascular remodeling. Histologic analyses of four matrix proteins-collagen I and IV, laminin, and fibronectin-in skin biopsies of patients with SS reveal that the basement membrane of dermal postcapillary venules undergoes changes in structure and composition. Increased neutrophil recruitment in vivo was associated with increases in collagen IV, decreases in laminin, and varied changes in fibronectin. In vitro studies using TNF-α and IL-17A were conducted to dissect basement membrane remodeling. Prolonged dual activation of cultured human pericytes with TNF-α and IL-17A augmented collagen IV production, similar to in vivo remodeling. Co-activation of pericytes with TNF-α and IL-17A also elevated fibronectin levels with little direct effect on laminin. However, the expression of fibronectin- and laminin-specific matrix metalloproteinases (MMPs), particularly MMP-3, was significantly up-regulated. Interactions between pericytes and neutrophils in culture yielded even higher levels of active MMPs, facilitating fibronectin and laminin degradation, and likely contributing to the varied levels of detectable fibronectin and the decreases in laminin observed in vivo. These data indicate that pericyte-neutrophil interactions play a role in mediating microvascular changes in SS and suggest that targeting MMP-3 may be effective in protecting vascular wall integrity.
Collapse
Affiliation(s)
- Holly M Lauridsen
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Amanda S Pellowe
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Anand Ramanathan
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Rebecca Liu
- Department of Immunobiology, Yale University, New Haven, Connecticut
| | | | - Jennifer M McNiff
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut
| | - Jordan S Pober
- Department of Immunobiology, Yale University, New Haven, Connecticut; Department of Dermatology, Yale School of Medicine, New Haven, Connecticut
| | - Anjelica L Gonzalez
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut.
| |
Collapse
|
218
|
The Importance of Pericytes in Healing: Wounds and other Pathologies. Int J Mol Sci 2017; 18:ijms18061129. [PMID: 28538706 PMCID: PMC5485953 DOI: 10.3390/ijms18061129] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/12/2017] [Accepted: 05/15/2017] [Indexed: 12/20/2022] Open
Abstract
Much of current research investigates the beneficial properties of mesenchymal stem cells (MSCs) as a treatment for wounds and other forms of injury. In this review, we bring attention to and discuss the role of the pericyte, a cell type which shares much of the differentiation potential and regenerative properties of the MSC as well as specific roles in the regulation of angiogenesis, inflammation and fibrosis. Pericytes have been identified as dysfunctional or depleted in many disease states, and observing the outcomes of pericyte perturbation in models of disease and wound healing informs our understanding of overall pericyte function and identifies these cells as an important target in the development of therapies to encourage healing.
Collapse
|
219
|
Du P, Suhaeri M, Ha SS, Oh SJ, Kim SH, Park K. Human lung fibroblast-derived matrix facilitates vascular morphogenesis in 3D environment and enhances skin wound healing. Acta Biomater 2017; 54:333-344. [PMID: 28351680 DOI: 10.1016/j.actbio.2017.03.035] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 03/23/2017] [Accepted: 03/24/2017] [Indexed: 10/19/2022]
Abstract
Extracellular matrix (ECM) is crucial to many aspects of vascular morphogenesis and maintenance of vasculature function. Currently the recapitulation of angiogenic ECM microenvironment is still challenging, due mainly to its diverse components and complex organization. Here we investigate the angiogenic potential of human lung fibroblast-derived matrix (hFDM) in creating a three-dimensional (3D) vascular construct. hFDM was obtained via decellularization of in vitro cultured human lung fibroblasts and analyzed via immunofluorescence staining and ELISA, which detect multiple ECM macromolecules and angiogenic growth factors (GFs). Human umbilical vein endothelial cells (HUVECs) morphology was more elongated and better proliferative on hFDM than on gelatin-coated substrate. To prepare 3D construct, hFDM is collected, quantitatively analyzed, and incorporated in collagen hydrogel (Col) with HUVECs. Capillary-like structure (CLS) formation at 7day was significantly better with the groups containing higher doses of hFDM compared to the Col group (control). Moreover, the group (Col/hFDM/GFs) with both hFDM and angiogenic GFs (VEGF, bFGF, SDF-1) showed the synergistic activity on CLS formation and found much larger capillary lumen diameters with time. Further analysis of hFDM via angiogenesis antibody array kit reveals abundant biochemical cues, such as angiogenesis-related cytokines, GFs, and proteolytic enzymes. Significantly up-regulated expression of VE-cadherin and ECM-specific integrin subunits was also noticed in Col/hFDM/GFs. In addition, transplantation of Col/hFMD/GFs with HUVECs in skin wound model presents more effective re-epithelialization, many regenerated hair follicles, better transplanted cells viability, and advanced neovascularization. We believe that current system is a very promising platform for 3D vasculature construction in vitro and for cell delivery toward therapeutic applications in vivo. STATEMENT OF SIGNIFICANCE Functional 3D vasculature construction in vitro is still challenging due to the difficulty of recapitulating the complex angiogenic extracellular matrix (ECM) environment. Herein, we present a simple and practical method to create an angiogenic 3D environment via incorporation of human lung fibroblast-derived matrix (hFDM) into collagen hydrogel. We found that hFDM offers a significantly improved angiogenic microenvironment for HUVECs on 2D substrates and in 3D construct. A synergistic effect of hFDM and angiogenic growth factors has been well confirmed in 3D condition. The prevascularized 3D collagen constructs also facilitate skin wound healing. We believe that current system should be a convenient and powerful platform in engineering 3D vasculature in vitro, and in delivering cells for therapeutic purposes in vivo.
Collapse
|
220
|
Reynolds LE, D'Amico G, Lechertier T, Papachristodoulou A, Muñoz-Félix JM, De Arcangelis A, Baker M, Serrels B, Hodivala-Dilke KM. Dual role of pericyte α6β1-integrin in tumour blood vessels. J Cell Sci 2017; 130:1583-1595. [PMID: 28289267 PMCID: PMC5450232 DOI: 10.1242/jcs.197848] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 03/08/2017] [Indexed: 12/18/2022] Open
Abstract
The α6β1-integrin is a major laminin receptor, and formation of a laminin-rich basement membrane is a key feature in tumour blood vessel stabilisation and pericyte recruitment, processes that are important in the growth and maturation of tumour blood vessels. However, the role of pericyte α6β1-integrin in angiogenesis is largely unknown. We developed mice where the α6-integrin subunit is deleted in pericytes and examined tumour angiogenesis and growth. These mice had: (1) reduced pericyte coverage of tumour blood vessels; (2) reduced tumour blood vessel stability; (3) increased blood vessel diameter; (4) enhanced blood vessel leakiness, and (5) abnormal blood vessel basement membrane architecture. Surprisingly, tumour growth, blood vessel density and metastasis were not altered. Analysis of retinas revealed that deletion of pericyte α6-integrin did not affect physiological angiogenesis. At the molecular level, we provide evidence that pericyte α6-integrin controls PDGFRβ expression and AKT-mTOR signalling. Taken together, we show that pericyte α6β1-integrin regulates tumour blood vessels by both controlling PDGFRβ and basement membrane architecture. These data establish a novel dual role for pericyte α6-integrin as modulating the blood vessel phenotype during pathological angiogenesis.
Collapse
Affiliation(s)
- Louise E Reynolds
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Biology, Barts Cancer Institute - A CRUK Centre of Excellence, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Gabriela D'Amico
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Biology, Barts Cancer Institute - A CRUK Centre of Excellence, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Tanguy Lechertier
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Biology, Barts Cancer Institute - A CRUK Centre of Excellence, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Alexandros Papachristodoulou
- Laboratory for Molecular Neuro-Oncology, Dept. of Neurology, University Hospital Zurich, Frauenklinikstrasse 26, Zurich CH-8091, Switzerland
| | - José M Muñoz-Félix
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Biology, Barts Cancer Institute - A CRUK Centre of Excellence, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Adèle De Arcangelis
- IGBMC, UMR 7104, INSERM U964, Université de Strasbourg, BP. 10142, 1, Rue Laurent Fries, Illkirch Cedex 67404, France
| | - Marianne Baker
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Biology, Barts Cancer Institute - A CRUK Centre of Excellence, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Bryan Serrels
- Cancer Research UK Edinburgh Centre, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK
| | - Kairbaan M Hodivala-Dilke
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Biology, Barts Cancer Institute - A CRUK Centre of Excellence, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
221
|
Chrifi I, Louzao-Martinez L, Brandt M, van Dijk CGM, Burgisser P, Zhu C, Kros JM, Duncker DJ, Cheng C. CMTM3 (CKLF-Like Marvel Transmembrane Domain 3) Mediates Angiogenesis by Regulating Cell Surface Availability of VE-Cadherin in Endothelial Adherens Junctions. Arterioscler Thromb Vasc Biol 2017; 37:1098-1114. [PMID: 28428220 DOI: 10.1161/atvbaha.116.308792] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 04/10/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Decrease in VE-cadherin adherens junctions reduces vascular stability, whereas disruption of adherens junctions is a requirement for neovessel sprouting during angiogenesis. Endocytosis plays a key role in regulating junctional strength by altering bioavailability of cell surface proteins, including VE-cadherin. Identification of new mediators of endothelial endocytosis could enhance our understanding of angiogenesis. Here, we assessed the function of CMTM3 (CKLF-like MARVEL transmembrane domain 3), which we have previously identified as highly expressed in Flk1+ endothelial progenitor cells during embryonic development. APPROACH AND RESULTS Using a 3-dimensional coculture of human umbilical vein endothelial cells-GFP (green fluorescent protein) and pericytes-RFP (red fluorescent protein), we demonstrated that siRNA-mediated CMTM3 silencing in human umbilical vein endothelial cells impairs angiogenesis. In vivo CMTM3 inhibition by morpholino injection in developing zebrafish larvae confirmed that CMTM3 expression is required for vascular sprouting. CMTM3 knockdown in human umbilical vein endothelial cells does not affect proliferation or migration. Intracellular staining demonstrated that CMTM3 colocalizes with early endosome markers EEA1 (early endosome marker 1) and Clathrin+ vesicles and with cytosolic VE-cadherin in human umbilical vein endothelial cells. Adenovirus-mediated CMTM3 overexpression enhances endothelial endocytosis, shown by an increase in Clathrin+, EEA1+, Rab11+, Rab5+, and Rab7+ vesicles. CMTM3 overexpression enhances, whereas CMTM3 knockdown decreases internalization of cell surface VE-cadherin in vitro. CMTM3 promotes loss of endothelial barrier function in thrombin-induced responses, shown by transendothelial electric resistance measurements in vitro. CONCLUSIONS In this study, we have identified a new regulatory function for CMTM3 in angiogenesis. CMTM3 is involved in VE-cadherin turnover and is a regulator of the cell surface pool of VE-cadherin. Therefore, CMTM3 mediates cell-cell adhesion at adherens junctions and contributes to the control of vascular sprouting.
Collapse
Affiliation(s)
- Ihsan Chrifi
- From the Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (I.C., M.B., P.B., D.J.D., C.C.); Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands (L.L.-M., C.G.M.v.D., C.C.); Netherlands Heart Institute, Utrecht (L.L.-M.); and Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands (C.Z., J.M.K.)
| | - Laura Louzao-Martinez
- From the Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (I.C., M.B., P.B., D.J.D., C.C.); Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands (L.L.-M., C.G.M.v.D., C.C.); Netherlands Heart Institute, Utrecht (L.L.-M.); and Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands (C.Z., J.M.K.)
| | - Maarten Brandt
- From the Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (I.C., M.B., P.B., D.J.D., C.C.); Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands (L.L.-M., C.G.M.v.D., C.C.); Netherlands Heart Institute, Utrecht (L.L.-M.); and Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands (C.Z., J.M.K.)
| | - Christian G M van Dijk
- From the Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (I.C., M.B., P.B., D.J.D., C.C.); Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands (L.L.-M., C.G.M.v.D., C.C.); Netherlands Heart Institute, Utrecht (L.L.-M.); and Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands (C.Z., J.M.K.)
| | - Petra Burgisser
- From the Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (I.C., M.B., P.B., D.J.D., C.C.); Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands (L.L.-M., C.G.M.v.D., C.C.); Netherlands Heart Institute, Utrecht (L.L.-M.); and Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands (C.Z., J.M.K.)
| | - Changbin Zhu
- From the Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (I.C., M.B., P.B., D.J.D., C.C.); Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands (L.L.-M., C.G.M.v.D., C.C.); Netherlands Heart Institute, Utrecht (L.L.-M.); and Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands (C.Z., J.M.K.)
| | - Johan M Kros
- From the Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (I.C., M.B., P.B., D.J.D., C.C.); Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands (L.L.-M., C.G.M.v.D., C.C.); Netherlands Heart Institute, Utrecht (L.L.-M.); and Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands (C.Z., J.M.K.)
| | - Dirk J Duncker
- From the Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (I.C., M.B., P.B., D.J.D., C.C.); Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands (L.L.-M., C.G.M.v.D., C.C.); Netherlands Heart Institute, Utrecht (L.L.-M.); and Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands (C.Z., J.M.K.)
| | - Caroline Cheng
- From the Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands (I.C., M.B., P.B., D.J.D., C.C.); Division of Internal Medicine and Dermatology, Department of Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands (L.L.-M., C.G.M.v.D., C.C.); Netherlands Heart Institute, Utrecht (L.L.-M.); and Department of Pathology, Erasmus University Medical Center, Rotterdam, the Netherlands (C.Z., J.M.K.).
| |
Collapse
|
222
|
Ramakrishnan VM, Boyd NL. The Adipose Stromal Vascular Fraction as a Complex Cellular Source for Tissue Engineering Applications. TISSUE ENGINEERING PART B-REVIEWS 2017; 24:289-299. [PMID: 28316259 DOI: 10.1089/ten.teb.2017.0061] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
A major challenge in tissue engineering is the generation of sufficient volumes of viable tissue for organ transplant. The development of a stable, mature vasculature is required to sustain the metabolic and functional activities of engineered tissues. Adipose stromal vascular fraction (SVF) cells are an easily accessible, heterogeneous cell system comprised of endothelial cells, macrophages, pericytes, and various stem cell populations. Collectively, SVF has been shown to spontaneously form vessel-like networks in vitro and robust, patent, and functional vasculatures in vivo. Capitalizing on this ability, we and others have demonstrated adipose SVF's utility in generating and augmenting engineered liver, cardiac, and vascular tissues, to name a few. This review highlights the scientific origins of SVF, the use of SVF as a clinically relevant vascular source, various SVF constituents and their roles, and practical considerations associated with isolating SVF for various tissue engineering applications.
Collapse
Affiliation(s)
- Venkat M Ramakrishnan
- Cardiovascular Innovation Institute, Department of Physiology, University of Louisville School of Medicine , Louisville, Kentucky
| | - Nolan L Boyd
- Cardiovascular Innovation Institute, Department of Physiology, University of Louisville School of Medicine , Louisville, Kentucky
| |
Collapse
|
223
|
Wang S, Zeng H, Chen ST, Zhou L, Xie XJ, He X, Tao YK, Tuo QH, Deng C, Liao DF, Chen JX. Ablation of endothelial prolyl hydroxylase domain protein-2 promotes renal vascular remodelling and fibrosis in mice. J Cell Mol Med 2017; 21:1967-1978. [PMID: 28266128 PMCID: PMC5571552 DOI: 10.1111/jcmm.13117] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/02/2017] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence demonstrates that hypoxia-inducible factor (HIF-α) hydroxylase system has a critical role in vascular remodelling. Using an endothelial-specific prolyl hydroxylase domain protein-2 (PHD2) knockout (PHD2EC KO) mouse model, this study investigates the regulatory role of endothelial HIF-α hydroxylase system in the development of renal fibrosis. Knockout of PHD2 in EC up-regulated the expression of HIF-1α and HIF-2α, resulting in a significant decline of renal function as evidenced by elevated levels of serum creatinine. Deletion of PHD2 increased the expression of Notch3 and transforming growth factor (TGF-β1) in EC, thus further causing glomerular arteriolar remodelling with an increased pericyte and pericyte coverage. This was accompanied by a significant elevation of renal resistive index (RI). Moreover, knockout of PHD2 in EC up-regulated the expression of fibroblast-specific protein-1 (FSP-1) and increased interstitial fibrosis in the kidney. These alterations were strongly associated with up-regulation of Notch3 and TGF-β1. We concluded that the expression of PHD2 in endothelial cells plays a critical role in renal fibrosis and vascular remodelling in adult mice. Furthermore, these changes were strongly associated with up-regulation of Notch3/TGF-β1 signalling and excessive pericyte coverage.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Heng Zeng
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Sean T Chen
- Duke University School of Medicine, Durham, NC, USA
| | - Liying Zhou
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Xue-Jiao Xie
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA.,Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xiaochen He
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yong-Kang Tao
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Qin-Hui Tuo
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Changqin Deng
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Duan-Fang Liao
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jian-Xiong Chen
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
224
|
Takeshita Y, Omoto M, Fujikawa S, Kanda T. Immunohistochemical analysis of laminin components in the blood-nerve barrier and blood-brain barrier. ACTA ACUST UNITED AC 2017. [DOI: 10.1111/cen3.12359] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Yukio Takeshita
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Yamaguchi Japan
| | - Masatoshi Omoto
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Yamaguchi Japan
| | - Susumu Fujikawa
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Yamaguchi Japan
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Yamaguchi Japan
| |
Collapse
|
225
|
Laminin differentially regulates the stemness of type I and type II pericytes. Stem Cell Res Ther 2017; 8:28. [PMID: 28173861 PMCID: PMC5297126 DOI: 10.1186/s13287-017-0479-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 01/03/2017] [Accepted: 01/10/2017] [Indexed: 01/22/2023] Open
Abstract
Background Laminin, a major basement membrane component that has direct contact with pericytes under physiological conditions, actively regulates the proliferation and differentiation/fate determination of pericytes. Recently, two types of pericytes (type I and type II) with different molecular markers and functions have been identified in skeletal muscles. Whether laminin differentially regulates the proliferation and differentiation of these two subpopulations remains unclear. Methods Wild-type and pericytic laminin-deficient mice under Nestin-GFP background were used to determine if laminin differentially regulates the proliferation and differentiation of type I and type II pericytes. Specifically, type I and type II pericytes were isolated from these mice, and their proliferation and differentiation were examined in vitro. Moreover, in vivo studies were also performed. Results We demonstrate that, although laminin inhibits the proliferation of both type I and type II pericytes in vitro, loss of laminin predominantly induces proliferation of type II pericytes in vivo. In addition, laminin negatively regulates the adipogenic differentiation of type I pericytes and positively regulates the myogenic differentiation of type II pericytes in vitro. Conclusions Laminin differentially regulates the proliferation and differentiation of type I and type II pericytes. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0479-4) contains supplementary material, which is available to authorized users.
Collapse
|
226
|
Boutin ME, Kramer LL, Livi LL, Brown T, Moore C, Hoffman-Kim D. A three-dimensional neural spheroid model for capillary-like network formation. J Neurosci Methods 2017; 299:55-63. [PMID: 28143748 DOI: 10.1016/j.jneumeth.2017.01.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 01/19/2017] [Accepted: 01/24/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND In vitro three-dimensional neural spheroid models have an in vivo-like cell density, and have the potential to reduce animal usage and increase experimental throughput. The aim of this study was to establish a spheroid model to study the formation of capillary-like networks in a three-dimensional environment that incorporates both neuronal and glial cell types, and does not require exogenous vasculogenic growth factors. NEW METHOD We created self-assembled, scaffold-free cellular spheroids using primary-derived postnatal rodent cortex as a cell source. The interactions between relevant neural cell types, basement membrane proteins, and endothelial cells were characterized by immunohistochemistry. Transmission electron microscopy was used to determine if endothelial network structures had lumens. RESULTS Endothelial cells within cortical spheroids assembled into capillary-like networks with lumens. Networks were surrounded by basement membrane proteins, including laminin, fibronectin and collagen IV, as well as key neurovascular cell types. COMPARISON WITH EXISTING METHOD(S) Existing in vitro models of the cortical neurovascular environment study monolayers of endothelial cells, either on transwell inserts or coating cellular spheroids. These models are not well suited to study vasculogenesis, a process hallmarked by endothelial cell cord formation and subsequent lumenization. CONCLUSIONS The neural spheroid is a new model to study the formation of endothelial cell capillary-like structures in vitro within a high cell density three-dimensional environment that contains both neuronal and glial populations. This model can be applied to investigate vascular assembly in healthy or disease states, such as stroke, traumatic brain injury, or neurodegenerative disorders.
Collapse
Affiliation(s)
- Molly E Boutin
- Center for Biomedical Engineering, 175 Meeting Street, Brown University, Providence, RI, 02912, United States; Department of Molecular Pharmacology, Physiology, and Biotechnology, 175 Meeting Street, Brown University, Providence, RI, 02912, United States.
| | - Liana L Kramer
- Department of Molecular Pharmacology, Physiology, and Biotechnology, 175 Meeting Street, Brown University, Providence, RI, 02912, United States.
| | - Liane L Livi
- Center for Biomedical Engineering, 175 Meeting Street, Brown University, Providence, RI, 02912, United States; Department of Molecular Pharmacology, Physiology, and Biotechnology, 175 Meeting Street, Brown University, Providence, RI, 02912, United States.
| | - Tyler Brown
- Department of Neuroscience, 175 Meeting Street, Brown University, Providence, RI, 02912, United States; Brown Institute for Brain Science, 175 Meeting Street, Brown University, Providence, RI, 02912, United States.
| | - Christopher Moore
- Department of Neuroscience, 175 Meeting Street, Brown University, Providence, RI, 02912, United States; Brown Institute for Brain Science, 175 Meeting Street, Brown University, Providence, RI, 02912, United States.
| | - Diane Hoffman-Kim
- Center for Biomedical Engineering, 175 Meeting Street, Brown University, Providence, RI, 02912, United States; Department of Molecular Pharmacology, Physiology, and Biotechnology, 175 Meeting Street, Brown University, Providence, RI, 02912, United States; Brown Institute for Brain Science, 175 Meeting Street, Brown University, Providence, RI, 02912, United States.
| |
Collapse
|
227
|
Abdeen AA, Lee J, Li Y, Kilian KA. Cytoskeletal Priming of Mesenchymal Stem Cells to a Medicinal Phenotype. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2017. [DOI: 10.1007/s40883-016-0021-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
228
|
Calderon GA, Thai P, Hsu CW, Grigoryan B, Gibson SM, Dickinson ME, Miller JS. Tubulogenesis of co-cultured human iPS-derived endothelial cells and human mesenchymal stem cells in fibrin and gelatin methacrylate gels. Biomater Sci 2017; 5:1652-1660. [DOI: 10.1039/c7bm00223h] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Here, we investigate the tubulogenic potential of commercially-sourced iPS-ECs with and without supporting commercially-sourced hMSCs within 3D natural fibrin or semi-synthetic gelatin methacrylate (GelMA) hydrogels.
Collapse
Affiliation(s)
| | - P. Thai
- Department of Bioengineering
- Rice University
- Houston
- USA
| | - C. W. Hsu
- Department of Molecular Physiology and Biophysics
- Baylor College of Medicine
- Houston
- USA
| | - B. Grigoryan
- Department of Bioengineering
- Rice University
- Houston
- USA
| | - S. M. Gibson
- Department of Bioengineering
- Rice University
- Houston
- USA
- Department of Molecular Physiology and Biophysics
| | - M. E. Dickinson
- Department of Molecular Physiology and Biophysics
- Baylor College of Medicine
- Houston
- USA
| | - J. S. Miller
- Department of Bioengineering
- Rice University
- Houston
- USA
| |
Collapse
|
229
|
Fabian KL, Storkus WJ. Immunotherapeutic Targeting of Tumor-Associated Blood Vessels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1036:191-211. [PMID: 29275473 DOI: 10.1007/978-3-319-67577-0_13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Pathological angiogenesis occurs during tumor progression and leads in the formation of an abnormal vasculature in the tumor microenvironment (TME). The tumor vasculature is disorganized, tortuous and leaky, resulting in high interstitial pressure and hypoxia in the TME, all of which are events that support tumor growth and survival. Given the sustaining role of the tumor vasculature, it has become an increasingly attractive target for the development of anti-cancer therapies. Antibodies, tyrosine kinase inhibitors and cancer vaccines that target pro-angiogenic factors, angiogenesis-associated receptors or tumor blood vessel-associated antigens continue to be developed and tested for therapeutic efficacy. Preferred anti-angiogenic protocols include those that "normalize" the tumor-associated vasculature which reduce hypoxia and improve tumor blood perfusion, resulting in tumor cell apoptosis, decreased immunosuppression, and enhanced effector immune cell infiltration/tumoricidal action within the TME.
Collapse
Affiliation(s)
- Kellsye L Fabian
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Dermatology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
230
|
Wu Q, Jing Y, Yuan X, Li B, Wang B, Liu M, Li H, Xiu R. The distinct abilities of tube-formation and migration between brain and spinal cord microvascular pericytes in rats. Clin Hemorheol Microcirc 2016; 60:231-40. [PMID: 24946754 DOI: 10.3233/ch-141856] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Pericytes are contractile cells that wrap around the endothelial cells of capillaries throughout the body. They play an important role in regulating the blood brain barrier (BBB) and blood spinal cord barrier (BSCB). The differences between brain and spinal cord microvascular endothelial cells have been investigated. However, no report has elucidated the similarities and differences between brain microvascular pericytes (BMPs) and spinal cord microvascular pericytes (SCMPs) in vitro. The similarities were found between the two types of pericytes not only in the proliferation ability but also in the expression of toll like receptor 4. On the other hand, BMPs showed more than 2 fold in tubular length formation compared with SCMPs. The number of migratory SCMPs was larger than that of migratory BMPs. The expressions of connexin 43 and vascular endothelial growth factor (VEGF) in BMPs were increased compared with those in SCMPs, while SCMPs expressed more desmin and N-cadherin than BMPs. The abilities of tube-formation and migration between BMPs and SCMPs were markedly different, which might be mediated by VEGF, connexin 43, N-cadherin and desmin. These distinguishing features may reflect the more widespread differences between the BBB and BSCB which directly impact pathophysiological processes in various major diseases.
Collapse
|
231
|
Núñez-Gómez E, Pericacho M, Ollauri-Ibáñez C, Bernabéu C, López-Novoa JM. The role of endoglin in post-ischemic revascularization. Angiogenesis 2016; 20:1-24. [PMID: 27943030 DOI: 10.1007/s10456-016-9535-4] [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: 05/16/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022]
Abstract
Following arterial occlusion, blood vessels respond by forming a new network of functional capillaries (angiogenesis), by reorganizing preexisting capillaries through the recruitment of smooth muscle cells to generate new arteries (arteriogenesis) and by growing and remodeling preexisting collateral arterioles into physiologically relevant arteries (collateral development). All these processes result in the recovery of organ perfusion. The importance of endoglin in post-occlusion reperfusion is sustained by several observations: (1) endoglin expression is increased in vessels showing active angiogenesis/remodeling; (2) genetic endoglin haploinsufficiency in humans causes deficient angiogenesis; and (3) the reduction of endoglin expression by gene disruption or the administration of endoglin-neutralizing antibodies reduces angiogenesis and revascularization. However, the precise role of endoglin in the several processes associated with revascularization has not been completely elucidated and, in some cases, the function ascribed to endoglin by different authors is controversial. The purpose of this review is to organize in a critical way the information available for the role of endoglin in several phenomena (angiogenesis, arteriogenesis and collateral development) associated with post-ischemic revascularization.
Collapse
Affiliation(s)
- Elena Núñez-Gómez
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Miguel Pericacho
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Claudia Ollauri-Ibáñez
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Carmelo Bernabéu
- Centro de Investigaciones Biológicas, Spanish National Research Council (CIB, CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - José M López-Novoa
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain. .,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain.
| |
Collapse
|
232
|
Stratman AN, Pezoa SA, Farrelly OM, Castranova D, Dye LE, Butler MG, Sidik H, Talbot WS, Weinstein BM. Interactions between mural cells and endothelial cells stabilize the developing zebrafish dorsal aorta. Development 2016; 144:115-127. [PMID: 27913637 DOI: 10.1242/dev.143131] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/17/2016] [Indexed: 12/13/2022]
Abstract
Mural cells (vascular smooth muscle cells and pericytes) play an essential role in the development of the vasculature, promoting vascular quiescence and long-term vessel stabilization through their interactions with endothelial cells. However, the mechanistic details of how mural cells stabilize vessels are not fully understood. We have examined the emergence and functional role of mural cells investing the dorsal aorta during early development using the zebrafish. Consistent with previous literature, our data suggest that cells ensheathing the dorsal aorta emerge from a sub-population of cells in the adjacent sclerotome. Inhibition of mural cell recruitment to the dorsal aorta through disruption of pdgfr signaling leads to a reduced vascular basement membrane, which in turn results in enhanced dorsal aorta vessel elasticity and failure to restrict aortic diameter. Our results provide direct in vivo evidence for a functional role for mural cells in patterning and stabilization of the early vasculature through production and maintenance of the vascular basement membrane to prevent abnormal aortic expansion and elasticity.
Collapse
Affiliation(s)
- Amber N Stratman
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sofia A Pezoa
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Olivia M Farrelly
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel Castranova
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Louis E Dye
- Microscopy & Imaging Core, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew G Butler
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Harwin Sidik
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - William S Talbot
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Brant M Weinstein
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
233
|
Abstract
Pericytes are a heterogeneous population of cells located in the blood vessel wall. They were first identified in the 19th century by Rouget, however their biological role and potential for drug targeting have taken time to be recognised. Isolation of pericytes from several different tissues has allowed a better phenotypic and functional characterization. These findings revealed a tissue-specific, multi-functional group of cells with multilineage potential. Given this emerging evidence, pericytes have acquired specific roles in pathobiological events in vascular diseases. In this review article, we will provide a compelling overview of the main diseases in which pericytes are involved, from well-established mechanisms to the latest findings. Pericyte involvement in diabetes and cancer will be discussed extensively. In the last part of the article we will review therapeutic approaches for these diseases in light of the recently acquired knowledge. To unravel pericyte-related vascular pathobiological events is pivotal not only for more tailored treatments of disease but also to establish pericytes as a therapeutic tool.
Collapse
|
234
|
Reuten R, Patel TR, McDougall M, Rama N, Nikodemus D, Gibert B, Delcros JG, Prein C, Meier M, Metzger S, Zhou Z, Kaltenberg J, McKee KK, Bald T, Tüting T, Zigrino P, Djonov V, Bloch W, Clausen-Schaumann H, Poschl E, Yurchenco PD, Ehrbar M, Mehlen P, Stetefeld J, Koch M. Structural decoding of netrin-4 reveals a regulatory function towards mature basement membranes. Nat Commun 2016; 7:13515. [PMID: 27901020 PMCID: PMC5141367 DOI: 10.1038/ncomms13515] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 10/09/2016] [Indexed: 02/07/2023] Open
Abstract
Netrins, a family of laminin-related molecules, have been proposed to act as guidance cues either during nervous system development or the establishment of the vascular system. This was clearly demonstrated for netrin-1 via its interaction with the receptors DCC and UNC5s. However, mainly based on shared homologies with netrin-1, netrin-4 was also proposed to play a role in neuronal outgrowth and developmental/pathological angiogenesis via interactions with netrin-1 receptors. Here, we present the high-resolution structure of netrin-4, which shows unique features in comparison with netrin-1, and show that it does not bind directly to any of the known netrin-1 receptors. We show that netrin-4 disrupts laminin networks and basement membranes (BMs) through high-affinity binding to the laminin γ1 chain. We hypothesize that this laminin-related function is essential for the previously described effects on axon growth promotion and angiogenesis. Our study unveils netrin-4 as a non-enzymatic extracellular matrix protein actively disrupting pre-existing BMs. Netrins are secreted guidance factors that promote axon outgrowth and orientation during nervous system development. Here the authors present structural and biological evidence that netrin-4 is not a guidance cue per se, but rather functions to modulate laminin-laminin interactions.
Collapse
Affiliation(s)
- Raphael Reuten
- Institute for Dental Research and Oral Musculoskeletal Biology, Medical Faculty, University of Cologne, Joseph-Stelzmann-Strasse 52, Cologne 50931, Germany.,Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Joseph-Stelzmann-Strasse 52, Cologne 50931, Germany
| | - Trushar R Patel
- Alberta RNA Research and Training Institute, Department of Chemistry and Biochemistry, University of Lethbridge, 4401 University Drive, Lethbridge, Alberta T1K 3M4, Canada.,School of Biosciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Matthew McDougall
- Department of Chemistry, University of Manitoba, Winnipeg R3T 2N2, Canada
| | - Nicolas Rama
- Apoptosis, Cancer and Development Laboratory, Equipe labellisée 'La Ligue', LabEx DEVweCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, Lyon 69008, France
| | - Denise Nikodemus
- Institute for Dental Research and Oral Musculoskeletal Biology, Medical Faculty, University of Cologne, Joseph-Stelzmann-Strasse 52, Cologne 50931, Germany.,Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Joseph-Stelzmann-Strasse 52, Cologne 50931, Germany
| | - Benjamin Gibert
- Apoptosis, Cancer and Development Laboratory, Equipe labellisée 'La Ligue', LabEx DEVweCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, Lyon 69008, France
| | - Jean-Guy Delcros
- Apoptosis, Cancer and Development Laboratory, Equipe labellisée 'La Ligue', LabEx DEVweCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, Lyon 69008, France
| | - Carina Prein
- Center for Applied Tissue Engineering and Regenerative Medicine-CANTER, Munich University of Applied Sciences, Lothstrasse 34, Munich D-80335, Germany.,Laboratory of Experimental Surgery and Regenerative Medicine - ExperiMed, Department of Surgery, Clinical Center University of Munich, Nussbaumstrasse 20, Munich D-80336, Germany.,Center for Nanoscience-CeNS, Geschwister-Scholl-Platz 1, Munich D-80539, Germany
| | - Markus Meier
- Department of Chemistry, University of Manitoba, Winnipeg R3T 2N2, Canada
| | - Stéphanie Metzger
- Laboratory for Cell and Tissue Engineering, Department of Obstetrics, University Hospital Zurich, Schmelzbergstr. 12, Zurich 8091, Switzerland
| | - Zhigang Zhou
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK.,Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Jennifer Kaltenberg
- Institute for Dental Research and Oral Musculoskeletal Biology, Medical Faculty, University of Cologne, Joseph-Stelzmann-Strasse 52, Cologne 50931, Germany.,Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Joseph-Stelzmann-Strasse 52, Cologne 50931, Germany
| | - Karen K McKee
- Department of Pathology, Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA
| | - Tobias Bald
- Department of Dermatology, University Hospital Magdeburg, Magdeburg 39120, Germany.,Laboratory of Experimental Dermatology, Department of Dermatology and Allergy, University of Bonn, Bonn 53105, Germany
| | - Thomas Tüting
- Department of Dermatology, University Hospital Magdeburg, Magdeburg 39120, Germany.,Laboratory of Experimental Dermatology, Department of Dermatology and Allergy, University of Bonn, Bonn 53105, Germany
| | - Paola Zigrino
- Department of Dermatology and Venerology, University of Cologne, Cologne 50931, Germany
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, Bern 3000, Switzerland
| | - Wilhelm Bloch
- Institute of Cardiovascular Research and Sport Medicine, Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Cologne 50933, Germany
| | - Hauke Clausen-Schaumann
- Center for Applied Tissue Engineering and Regenerative Medicine-CANTER, Munich University of Applied Sciences, Lothstrasse 34, Munich D-80335, Germany.,Center for Nanoscience-CeNS, Geschwister-Scholl-Platz 1, Munich D-80539, Germany
| | - Ernst Poschl
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Peter D Yurchenco
- Department of Pathology, Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA
| | - Martin Ehrbar
- Laboratory for Cell and Tissue Engineering, Department of Obstetrics, University Hospital Zurich, Schmelzbergstr. 12, Zurich 8091, Switzerland
| | - Patrick Mehlen
- Apoptosis, Cancer and Development Laboratory, Equipe labellisée 'La Ligue', LabEx DEVweCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, Lyon 69008, France
| | - Jörg Stetefeld
- Department of Chemistry, University of Manitoba, Winnipeg R3T 2N2, Canada
| | - Manuel Koch
- Institute for Dental Research and Oral Musculoskeletal Biology, Medical Faculty, University of Cologne, Joseph-Stelzmann-Strasse 52, Cologne 50931, Germany.,Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Joseph-Stelzmann-Strasse 52, Cologne 50931, Germany
| |
Collapse
|
235
|
Abstract
The concept of pericyte has been changing over years. This cell type was believed to possess only a function of trophic support to endothelial cells and to maintain vasculature stabilization. In the last years, the discovery of multipotent ability of perivascular populations led to the concept of vessel/wall niche. Likewise, several perivascular populations have been identified in animal and human bone marrow. In this review, we provide an overview on bone marrow perivascular population, their cross-talk with other niche components, relationship with bone marrow stromal stem cells, and similarities and differences with the perivascular population of the vessel/wall niche. Finally, we focus on the regenerative potential of these cells and the forthcoming challenges related to their use as cell therapy products.
Collapse
Affiliation(s)
- Giuseppe Mangialardi
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, UK
| | - Andrea Cordaro
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, UK
| | - Paolo Madeddu
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, UK
| |
Collapse
|
236
|
Pinto MP, Arce M, Yameen B, Vilos C. Targeted brain delivery nanoparticles for malignant gliomas. Nanomedicine (Lond) 2016; 12:59-72. [PMID: 27876436 DOI: 10.2217/nnm-2016-0307] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Brain tumors display the highest mortality rates of all childhood cancers, and over the last decade its prevalence has steadily increased in elderly. To date, effective treatments for brain tumors and particularly for malignant gliomas remain a challenge mainly due to the low permeability and high selectivity of the blood-brain barrier (BBB) to conventional anticancer drugs. In recent years, the elucidation of the cellular mechanisms involved in the transport of substances into the brain has boosted the development of therapeutic-targeted nanoparticles (NPs) with the ability to cross the BBB. Here, we present a comprehensive overview of the available therapeutic strategies developed against malignant gliomas based on 'actively targeted' NPs, the challenges of crossing the BBB and blood-brain tumor barrier as well as its mechanisms and a critical assessment of clinical studies that have used targeted NPs for the treatment of malignant gliomas. Finally, we discuss the potential of actively targeted NP-based strategies in clinical settings, its possible side effects and future directions for therapeutic applications. First draft submitted: 4 October 2016; Accepted for publication: 14 October 2016; Published online: 23 November 2016.
Collapse
Affiliation(s)
- Mauricio P Pinto
- Laboratory of Immunology of Reproduction, Faculty of Chemistry & Biology, Universidad de Santiago de Chile, 9170022 Santiago, Chile
| | - Maximiliano Arce
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Catolica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Faculty of Biological Sciences, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Basit Yameen
- Laboratory of Nanomedicine & Biomaterials, Department of Anesthesiology, Harvard Medical School, Brigham & Women's Hospital, Boston, MA 02115, USA.,Department of Chemistry, SBA School of Science & Engineering, Lahore University of Management Sciences (LUMS), Lahore 54792, Pakistan
| | - Cristian Vilos
- Laboratory of Nanomedicine & Targeted Delivery, Center for Integrative Medicine & Innovative Science, Faculty of Medicine, Universidad Andres Bello, Santiago, 8370071 Santiago, Chile.,Center for Bioinformatics & Integrative Biology, Faculty of Biological Sciences, Universidad Andres Bello, Santiago, 8370071 Santiago, Chile.,Center for the Development of Nanoscience & Nanotechnology, CEDENNA, 9170124 Santiago, Chile
| |
Collapse
|
237
|
Gautam J, Zhang X, Yao Y. The role of pericytic laminin in blood brain barrier integrity maintenance. Sci Rep 2016; 6:36450. [PMID: 27808256 PMCID: PMC5093438 DOI: 10.1038/srep36450] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 10/14/2016] [Indexed: 11/09/2022] Open
Abstract
Laminin, a major component of the basement membrane, plays an important role in blood brain barrier regulation. At the neurovascular unit, brain endothelial cells, astrocytes, and pericytes synthesize and deposit different laminin isoforms into the basement membrane. It has been shown that laminin α4 (endothelial laminin) regulates vascular integrity at embryonic/neonatal stage, while astrocytic laminin maintains vascular integrity in adulthood. Here, we investigate the function of pericyte-derived laminin in vascular integrity. Using a conditional knockout mouse line, we report that loss of pericytic laminin leads to hydrocephalus and BBB breakdown in a small percentage (10.7%) of the mutants. Interestingly, BBB disruption always goes hand-in-hand with hydrocephalus in these mutants, and neither symptom is observed in the rest 89.3% of the mutants. Further mechanistic studies show that reduced tight junction proteins, diminished AQP4 expression, and decreased pericyte coverage are responsible for the BBB disruption. Together, these data suggest that pericyte-derived laminin is involved in the maintenance of BBB integrity and regulation of ventricular size/development.
Collapse
Affiliation(s)
- Jyoti Gautam
- College of Pharmacy, University of Minnesota, 1110 Kirby Drive, Duluth, MN, 55812, USA
| | - Xuanming Zhang
- College of Pharmacy, University of Minnesota, 1110 Kirby Drive, Duluth, MN, 55812, USA
| | - Yao Yao
- College of Pharmacy, University of Minnesota, 1110 Kirby Drive, Duluth, MN, 55812, USA
| |
Collapse
|
238
|
Warmke N, Griffin KJ, Cubbon RM. Pericytes in diabetes-associated vascular disease. J Diabetes Complications 2016; 30:1643-1650. [PMID: 27592245 DOI: 10.1016/j.jdiacomp.2016.08.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/01/2016] [Accepted: 08/08/2016] [Indexed: 12/21/2022]
Abstract
Pericytes are mural cells that support and stabilise the microvasculature, and are present in all vascular beds. Pericyte-endothelial cell crosstalk is essential in both remodelling and quiescent vasculature, and this complex interaction is often disrupted in disease states. Pericyte loss is believed to be an early hallmark of diabetes-associated microvascular disease, including retinopathy and nephropathy. Here we review the current literature defining pericyte biology in the context of diabetes-associated vascular disease, with a particular focus on whether pericytes contribute actively to disease progression. We also speculate regarding the role of pericytes in the recovery from macrovascular complications, such as critical limb ischaemia. It becomes clear that dysfunctional pericytes are likely to actively induce disease progression by causing vasoconstriction and basement membrane thickening, resulting in tissue ischaemia. Moreover, their altered interactions with endothelial cells are likely to cause abnormal and inadequate neovascularisation in diverse vascular beds. Further research is needed to identify mechanisms by which pericyte function is altered by diabetes, with a view to developing therapeutic approaches that normalise vascular function and remodelling.
Collapse
Affiliation(s)
- Nele Warmke
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Kathryn J Griffin
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom
| | - Richard M Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT laboratories, The University of Leeds, Clarendon Way, Leeds, LS2 9JT, United Kingdom.
| |
Collapse
|
239
|
Mazurek R, Dave JM, Chandran RR, Misra A, Sheikh AQ, Greif DM. Vascular Cells in Blood Vessel Wall Development and Disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 78:323-350. [PMID: 28212800 DOI: 10.1016/bs.apha.2016.08.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The vessel wall is composed of distinct cellular layers, yet communication among individual cells within and between layers results in a dynamic and versatile structure. The morphogenesis of the normal vascular wall involves a highly regulated process of cell proliferation, migration, and differentiation. The use of modern developmental biological and genetic approaches has markedly enriched our understanding of the molecular and cellular mechanisms underlying these developmental events. Additionally, the application of similar approaches to study diverse vascular diseases has resulted in paradigm-shifting insights into pathogenesis. Further investigations into the biology of vascular cells in development and disease promise to have major ramifications on therapeutic strategies to combat pathologies of the vasculature.
Collapse
Affiliation(s)
- R Mazurek
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - J M Dave
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - R R Chandran
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - A Misra
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - A Q Sheikh
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - D M Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States.
| |
Collapse
|
240
|
de Vries MR, Quax PHA. Plaque angiogenesis and its relation to inflammation and atherosclerotic plaque destabilization. Curr Opin Lipidol 2016; 27:499-506. [PMID: 27472406 DOI: 10.1097/mol.0000000000000339] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW The review discusses the recent literature on plaque angiogenesis and its relation to inflammation and plaque destabilization. Furthermore, it discusses how plaque angiogenesis can be used to monitor atherosclerosis and serve as a therapeutic target. RECENT FINDINGS Histopathologic studies have shown a clear relationship between plaque angiogenesis, intraplaque hemorrhage (IPH), plaque vulnerability, and cardiovascular events. Hypoxia is a main driver of plaque angiogenesis and the mechanism behind angiogenesis is only partly known. IPH, as the result of immature neovessels, is associated with increased influx of inflammatory cells in the plaques. Experimental models displaying certain features of human atherosclerosis such as plaque angiogenesis or IPH are developed and can contribute to unraveling the mechanism behind plaque vulnerability. New imaging techniques are established, with which plaque angiogenesis and vulnerability can be detected. Furthermore, antiangiogenic therapies in atherosclerosis gain much attention. SUMMARY Plaque angiogenesis, IPH, and inflammation contribute to plaque vulnerability. Histopathologic and imaging studies together with specific experimental studies have provided insights in plaque angiogenesis and plaque vulnerability. However, more extensive knowledge on the underlying mechanism is required for establishing new therapies for patients at risk.
Collapse
Affiliation(s)
- Margreet R de Vries
- Department of Surgery, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
241
|
Vascular Transdifferentiation in the CNS: A Focus on Neural and Glioblastoma Stem-Like Cells. Stem Cells Int 2016; 2016:2759403. [PMID: 27738435 PMCID: PMC5055959 DOI: 10.1155/2016/2759403] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 09/05/2016] [Indexed: 01/12/2023] Open
Abstract
Glioblastomas are devastating and extensively vascularized brain tumors from which glioblastoma stem-like cells (GSCs) have been isolated by many groups. These cells have a high tumorigenic potential and the capacity to generate heterogeneous phenotypes. There is growing evidence to support the possibility that these cells are derived from the accumulation of mutations in adult neural stem cells (NSCs) as well as in oligodendrocyte progenitors. It was recently reported that GSCs could transdifferentiate into endothelial-like and pericyte-like cells both in vitro and in vivo, notably under the influence of Notch and TGFβ signaling pathways. Vascular cells derived from GBM cells were also observed directly in patient samples. These results could lead to new directions for designing original therapeutic approaches against GBM neovascularization but this specific reprogramming requires further molecular investigations. Transdifferentiation of nontumoral neural stem cells into vascular cells has also been described and conversely vascular cells may generate neural stem cells. In this review, we present and discuss these recent data. As some of them appear controversial, further validation will be needed using new technical approaches such as high throughput profiling and functional analyses to avoid experimental pitfalls and misinterpretations.
Collapse
|
242
|
Interaction between integrin α5 and PDE4D regulates endothelial inflammatory signalling. Nat Cell Biol 2016; 18:1043-53. [PMID: 27595237 PMCID: PMC5301150 DOI: 10.1038/ncb3405] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 08/03/2016] [Indexed: 12/16/2022]
Abstract
Atherosclerosis is primarily a disease of lipid metabolism and inflammation; however, it is also closely associated with endothelial extracellular matrix (ECM) remodelling, with fibronectin accumulating in the laminin-collagen basement membrane. To investigate how fibronectin modulates inflammation in arteries, we replaced the cytoplasmic tail of the fibronectin receptor integrin α5 with that of the collagen/laminin receptor integrin α2. This chimaera suppressed inflammatory signalling in endothelial cells on fibronectin and in knock-in mice. Fibronectin promoted inflammation by suppressing anti-inflammatory cAMP. cAMP was activated through endothelial prostacyclin secretion; however, this was ECM-independent. Instead, cells on fibronectin suppressed cAMP via enhanced phosphodiesterase (PDE) activity, through direct binding of integrin α5 to phosphodiesterase-4D5 (PDE4D5), which induced PP2A-dependent dephosphorylation of PDE4D5 on the inhibitory site Ser651. In vivo knockdown of PDE4D5 inhibited inflammation at athero-prone sites. These data elucidate a molecular mechanism linking ECM remodelling and inflammation, thereby identifying a new class of therapeutic targets.
Collapse
|
243
|
Motegi SI, Sekiguchi A, Fujiwara C, Toki S, Ishikawa O. Possible association of elevated serum collagen type IV level with skin sclerosis in systemic sclerosis. J Dermatol 2016; 44:167-172. [DOI: 10.1111/1346-8138.13564] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 07/25/2016] [Indexed: 01/09/2023]
Affiliation(s)
- Sei-ichiro Motegi
- Department of Dermatology; Gunma University Graduate School of Medicine; Maebashi Japan
| | - Akiko Sekiguchi
- Department of Dermatology; Gunma University Graduate School of Medicine; Maebashi Japan
| | - Chisako Fujiwara
- Department of Dermatology; Gunma University Graduate School of Medicine; Maebashi Japan
| | - Sayaka Toki
- Department of Dermatology; Gunma University Graduate School of Medicine; Maebashi Japan
| | - Osamu Ishikawa
- Department of Dermatology; Gunma University Graduate School of Medicine; Maebashi Japan
| |
Collapse
|
244
|
Bersini S, Yazdi IK, Talò G, Shin SR, Moretti M, Khademhosseini A. Cell-microenvironment interactions and architectures in microvascular systems. Biotechnol Adv 2016; 34:1113-1130. [PMID: 27417066 DOI: 10.1016/j.biotechadv.2016.07.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 07/02/2016] [Accepted: 07/09/2016] [Indexed: 02/06/2023]
Abstract
In the past decade, significant advances have been made in the design and optimization of novel biomaterials and microfabrication techniques to generate vascularized tissues. Novel microfluidic systems have facilitated the development and optimization of in vitro models for exploring the complex pathophysiological phenomena that occur inside a microvascular environment. To date, most of these models have focused on engineering of increasingly complex systems, rather than analyzing the molecular and cellular mechanisms that drive microvascular network morphogenesis and remodeling. In fact, mutual interactions among endothelial cells (ECs), supporting mural cells and organ-specific cells, as well as between ECs and the extracellular matrix, are key driving forces for vascularization. This review focuses on the integration of materials science, microengineering and vascular biology for the development of in vitro microvascular systems. Various approaches currently being applied to study cell-cell/cell-matrix interactions, as well as biochemical/biophysical cues promoting vascularization and their impact on microvascular network formation, will be identified and discussed. Finally, this review will explore in vitro applications of microvascular systems, in vivo integration of transplanted vascularized tissues, and the important challenges for vascularization and controlling the microcirculatory system within the engineered tissues, especially for microfabrication approaches. It is likely that existing models and more complex models will further our understanding of the key elements of vascular network growth, stabilization and remodeling to translate basic research principles into functional, vascularized tissue constructs for regenerative medicine applications, drug screening and disease models.
Collapse
Affiliation(s)
- Simone Bersini
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| | - Iman K Yazdi
- Biomaterials Innovation Research Center, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02139, USA
| | - Giuseppe Talò
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| | - Su Ryon Shin
- Biomaterials Innovation Research Center, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02139, USA
| | - Matteo Moretti
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy; Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale, Lugano, Switzerland; Swiss Institute for Regenerative Medicine, Lugano, Switzerland; Cardiocentro Ticino, Lugano, Switzerland.
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02139, USA; Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia; College of Animal Bioscience and Technology, Department of Bioindustrial Technologies, Konkuk University, Hwayang-dong, Kwangjin-gu, Seoul 143-701, Republic of Korea.
| |
Collapse
|
245
|
A novel lineage restricted, pericyte-like cell line isolated from human embryonic stem cells. Sci Rep 2016; 6:24403. [PMID: 27109637 PMCID: PMC4842973 DOI: 10.1038/srep24403] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/29/2016] [Indexed: 12/22/2022] Open
Abstract
Pericytes (PCs) are endothelium-associated cells that play an important role in normal vascular function and maintenance. We developed a method comparable to GMP quality protocols for deriving self-renewing perivascular progenitors from the human embryonic stem cell (hESC), line ESI-017. We identified a highly scalable, perivascular progenitor cell line that we termed PC-A, which expressed surface markers common to mesenchymal stromal cells. PC-A cells were not osteogenic or adipogenic under standard differentiation conditions and showed minimal angiogenic support function in vitro. PC-A cells were capable of further differentiation to perivascular progenitors with limited differentiation capacity, having osteogenic potential (PC-O) or angiogenic support function (PC-M), while lacking adipogenic potential. Importantly, PC-M cells expressed surface markers associated with pericytes. Moreover, PC-M cells had pericyte-like functionality being capable of co-localizing with human umbilical vein endothelial cells (HUVECs) and enhancing tube stability up to 6 days in vitro. We have thus identified a self-renewing perivascular progenitor cell line that lacks osteogenic, adipogenic and angiogenic potential but is capable of differentiation toward progenitor cell lines with either osteogenic potential or pericyte-like angiogenic function. The hESC-derived perivascular progenitors described here have potential applications in vascular research, drug development and cell therapy.
Collapse
|
246
|
Lei X, Chen M, Nie Q, Hu J, Zhuo Z, Yiu A, Chen H, Xu N, Huang M, Ye K, Bai L, Ye W, Zhang D. In vitro and in vivo antiangiogenic activity of desacetylvinblastine monohydrazide through inhibition of VEGFR2 and Axl pathways. Am J Cancer Res 2016; 6:843-58. [PMID: 27186435 PMCID: PMC4859888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 03/06/2016] [Indexed: 02/14/2023] Open
Abstract
Tumor angiogenic process is regulated by multiple proangiogenic pathways, such as vascular endothelial growth factor receptor 2 (VEGFR2) and Axl receptor tyrosine kinase (Axl). Axl is one of many important factors involved in anti-VEGF resistance. Inhibition of VEGF/VEGFR2 signaling pathway alone fails to block tumor neovascularization. Therefore, discovery of novel agents targeting multiple angiogenesis pathways is in demand. Desacetylvinblastine monohydrazide (DAVLBH), a derivative of vinblastine (VLB), has been reported exhibit an anticancer activity via its cytotoxic effect. However, little attention has been paid to the antiangiogenic properties of DAVLBH. Here, we firstly reported that DAVLBH exerted a more potent antiangiogenic effect than VLB in vitro and in vivo, which was associated with inactivation of VEGF/VEGFR2 and Gas6/Axl signaling pathways. We found that DAVLBH inhibited VEGF- and Gas6-induced HUVECs proliferation, migration, tube formation and vessel sprouts formation in vitro and ex vivo. It significantly inhibited in vivo tumor angiogenesis and tumor growth in HeLa xenografts. It also inhibited Gas6-induced pericytes recruitment to endothelial tubes accompanied with a decrease in expression and activation of Axl. Besides, it could block the compensatory up-regulating expression and activation of Axl in response to bevacizumab treatment in HUVECs. Taken together, our results suggest that DAVLBH potently inhibits angiogenesis-mediated tumor growth through blockage of the activation of VEGF/VEGFR2 and Gas6/Axl pathways and it might serve as a promising antiangiogenic agent for the cancer therapy.
Collapse
Affiliation(s)
- Xueping Lei
- College of Pharmacy, Jinan UniversityGuangzhou 510632, China
| | - Minfeng Chen
- College of Pharmacy, Jinan UniversityGuangzhou 510632, China
| | - Qiulin Nie
- College of Pharmacy, Jinan UniversityGuangzhou 510632, China
| | - Jianyang Hu
- College of Pharmacy, Jinan UniversityGuangzhou 510632, China
| | - Zhenjian Zhuo
- College of Pharmacy, Jinan UniversityGuangzhou 510632, China
| | - Anita Yiu
- School of Life Sciences, The Chinese University of Hong KongHong Kong, China
| | - Heru Chen
- College of Pharmacy, Jinan UniversityGuangzhou 510632, China
| | - Nanhui Xu
- College of Pharmacy, Jinan UniversityGuangzhou 510632, China
| | - Maohua Huang
- College of Pharmacy, Jinan UniversityGuangzhou 510632, China
| | - Kaihe Ye
- College of Pharmacy, Jinan UniversityGuangzhou 510632, China
| | - Liangliang Bai
- College of Pharmacy, Jinan UniversityGuangzhou 510632, China
| | - Wencai Ye
- College of Pharmacy, Jinan UniversityGuangzhou 510632, China
| | - Dongmei Zhang
- College of Pharmacy, Jinan UniversityGuangzhou 510632, China
| |
Collapse
|
247
|
Tattersall IW, Du J, Cong Z, Cho BS, Klein AM, Dieck CL, Chaudhri RA, Cuervo H, Herts JH, Kitajewski J. In vitro modeling of endothelial interaction with macrophages and pericytes demonstrates Notch signaling function in the vascular microenvironment. Angiogenesis 2016; 19:201-15. [PMID: 26965898 DOI: 10.1007/s10456-016-9501-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 02/17/2016] [Indexed: 01/10/2023]
Abstract
Angiogenesis is regulated by complex interactions between endothelial cells and support cells of the vascular microenvironment, such as tissue myeloid cells and vascular mural cells. Multicellular interactions during angiogenesis are difficult to study in animals and challenging in a reductive setting. We incorporated stromal cells into an established bead-based capillary sprouting assay to develop assays that faithfully reproduce major steps of vessel sprouting and maturation. We observed that macrophages enhance angiogenesis, increasing the number and length of endothelial sprouts, a property we have dubbed "angiotrophism." We found that polarizing macrophages toward a pro-inflammatory profile further increased their angiotrophic stimulation of vessel sprouting, and this increase was dependent on macrophage Notch signaling. To study endothelial/pericyte interactions, we added vascular pericytes directly to the bead-bound endothelial monolayer. These pericytes formed close associations with the endothelial sprouts, causing increased sprout number and vessel caliber. We found that Jagged1 expression and Notch signaling are essential for the growth of both endothelial cells and pericytes and may function in their interaction. We observed that combining endothelial cells with both macrophages and pericytes in the same sprouting assay has multiplicative effects on sprouting. These results significantly improve bead-capillary sprouting assays and provide an enhanced method for modeling interactions between the endothelium and the vascular microenvironment. Achieving this in a reductive in vitro setting represents a significant step toward a better understanding of the cellular elements that contribute to the formation of mature vasculature.
Collapse
Affiliation(s)
| | - Jing Du
- Obstetrics/Gynecology, Columbia University, New York, NY, USA
- Pathology and Cell Biology, Columbia University, New York, NY, USA
| | | | - Bennet S Cho
- Obstetrics/Gynecology, Columbia University, New York, NY, USA
| | - Alyssa M Klein
- Obstetrics/Gynecology, Columbia University, New York, NY, USA
| | - Chelsea L Dieck
- Obstetrics/Gynecology, Columbia University, New York, NY, USA
| | | | - Henar Cuervo
- Obstetrics/Gynecology, Columbia University, New York, NY, USA
| | - James H Herts
- Obstetrics/Gynecology, Columbia University, New York, NY, USA
| | - Jan Kitajewski
- Obstetrics/Gynecology, Columbia University, New York, NY, USA.
- Pathology and Cell Biology, Columbia University, New York, NY, USA.
- Physiology and Biophysics, University of Illinois Chicago, College of Medicine, Chicago, IL, USA.
- Columbia University Medical Center, 1130 St. Nicholas Avenue, ICRC 926, New York, NY, 10032, USA.
| |
Collapse
|
248
|
Bodnar RJ, Satish L, Yates CC, Wells A. Pericytes: A newly recognized player in wound healing. Wound Repair Regen 2016; 24:204-14. [PMID: 26969517 DOI: 10.1111/wrr.12415] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 01/28/2016] [Indexed: 12/26/2022]
Abstract
Pericytes have generally been considered in the context of stabilizing vessels, ensuring the blood barriers, and regulating the flow through capillaries. However, new reports suggest that pericytes may function at critical times to either drive healing with minimal scarring or, perversely, contribute to fibrosis and ongoing scar formation. Beneficially, pericytes probably drive much of the vascular involution that occurs during the transition from the regenerative to the resolution phases of healing. Pathologically, pericytes can assume a fibrotic phenotype and promote scarring. This perspective will discuss pericyte involvement in wound repair and the relationship pericytes form with the parenchymal cells of the skin. We will further evaluate the role pericytes may have in disease progression in relation to chronic wounds and fibrosis.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.,McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania.,Veterans Affairs Medical Center, Pittsburgh, Pennsylvania
| | - Latha Satish
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania.,Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Cecelia C Yates
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.,McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania.,Veterans Affairs Medical Center, Pittsburgh, Pennsylvania.,Department of Health Promotions and Development, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.,McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania.,Veterans Affairs Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
249
|
Jha AK, Tharp KM, Browne S, Ye J, Stahl A, Yeghiazarians Y, Healy KE. Matrix metalloproteinase-13 mediated degradation of hyaluronic acid-based matrices orchestrates stem cell engraftment through vascular integration. Biomaterials 2016; 89:136-47. [PMID: 26967648 DOI: 10.1016/j.biomaterials.2016.02.023] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 02/17/2016] [Indexed: 12/15/2022]
Abstract
A critical design parameter for the function of synthetic extracellular matrices is to synchronize the gradual cell-mediated degradation of the matrix with the endogenous secretion of natural extracellular matrix (ECM) (e.g., creeping substitution). In hyaluronic acid (HyA)-based hydrogel matrices, we have investigated the effects of peptide crosslinkers with different matrix metalloproteinases (MMP) sensitivities on network degradation and neovascularization in vivo. The HyA hydrogel matrices consisted of cell adhesive peptides, heparin for both the presentation of exogenous and sequestration of endogenously synthesized growth factors, and MMP cleavable peptide linkages (i.e., QPQGLAK, GPLGMHGK, and GPLGLSLGK). Sca1(+)/CD45(-)/CD34(+)/CD44(+) cardiac progenitor cells (CPCs) cultured in the matrices with the slowly degradable QPQGLAK hydrogels supported the highest production of MMP-2, MMP-9, MMP-13, VEGF165, and a range of angiogenesis related proteins. Hydrogels with QPQGLAK crosslinks supported prolonged retention of these proteins via heparin within the matrix, stimulating rapid vascular development, and anastomosis with the host vasculature when implanted in the murine hindlimb.
Collapse
Affiliation(s)
- Amit K Jha
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA; Department of Material Science and Engineering, University of California, Berkeley, CA 94720, USA
| | - Kevin M Tharp
- Department of Nutritional Science and Toxicology, University of California, Berkeley, CA 94720, USA
| | - Shane Browne
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA; Department of Material Science and Engineering, University of California, Berkeley, CA 94720, USA; Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Ireland
| | - Jianqin Ye
- Department of Medicine, University of California, San Francisco, CA 94143, USA
| | - Andreas Stahl
- Department of Nutritional Science and Toxicology, University of California, Berkeley, CA 94720, USA
| | - Yerem Yeghiazarians
- Department of Medicine, University of California, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA; Cardiovascular Research Institute, University of California, San Francisco, CA 94143, USA
| | - Kevin E Healy
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA; Department of Material Science and Engineering, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
250
|
Role of the tumor stroma in resistance to anti-angiogenic therapy. Drug Resist Updat 2016; 25:26-37. [DOI: 10.1016/j.drup.2016.02.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 02/09/2016] [Accepted: 02/17/2016] [Indexed: 12/13/2022]
|