1
|
Filatov NS, Khismatullin RR, Bilyalov AI, Khabirova AI, Salyakhutdinova SM, Ursan RV, Kasimova RN, Peshkova AD, Gazizov II, Shagimardanova EI, Woroncow MV, Kiyasov AP, Litvinov RI, Gusev OA. Distinct Hemostasis and Blood Composition in Spiny Mouse Acomys cahirinus. Int J Mol Sci 2024; 25:12867. [PMID: 39684578 DOI: 10.3390/ijms252312867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
The spiny mouse (Acomys species) is capable of scarless wound regeneration through largely yet unknown mechanisms. To investigate whether this capacity is related to peculiarities of the hemostatic system, we studied the blood of Acomys cahirinus in comparison to Mus musculus (Balb/c) to reveal differences in blood composition and clotting in both males and females. In response to surgical manipulations, blood clots formed in wounds of Acomys comprised a stronger hemostatic seal with reduced surgical bleeding in comparison with Balb/c. Acomys demonstrated notably shorter tail bleeding times and elevated clottable fibrinogen levels. Histological analysis revealed that clots from Acomys blood had densely packed fibrin-rich clots with pronounced fibrin segregation from erythrocytes. Acomys exhibited superior plasma clot stiffness as revealed with thromboelastography. The latter two characteristics are likely due to hyperfibrinogenemia. Light transmission platelet aggregometry demonstrated that ADP-induced platelet aggregates in Acomys males are stable, unlike the aggregates formed in the plasma of Balb/c undergoing progressive disaggregation over time. There were no apparent distinctions in platelet contractility and baseline expression of phosphatidylserine. Hematological profiling revealed a reduced erythrocytes count but increased mean corpuscular volume and hemoglobin content in Acomys. These results demonstrate the distinctive hemostatic potential of Acomys cahirinus, which may contribute to their remarkable regenerative capacity.
Collapse
Affiliation(s)
- Nikita S Filatov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Rafael R Khismatullin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Airat I Bilyalov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Loginov Moscow Clinical Scientific Center, 111123 Moscow, Russia
| | - Alina I Khabirova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | | | - Roman V Ursan
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | | | - Alina D Peshkova
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Insaf I Gazizov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Elena I Shagimardanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Loginov Moscow Clinical Scientific Center, 111123 Moscow, Russia
| | - Mary V Woroncow
- Institute for Regenerative Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Andrey P Kiyasov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Rustem I Litvinov
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Oleg A Gusev
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- LIFT, Life Improvement by Future Technologies Institute, 121205 Moscow, Russia
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| |
Collapse
|
2
|
Everts PA, Lana JF, Alexander RW, Dallo I, Kon E, Ambach MA, van Zundert A, Podesta L. Profound Properties of Protein-Rich, Platelet-Rich Plasma Matrices as Novel, Multi-Purpose Biological Platforms in Tissue Repair, Regeneration, and Wound Healing. Int J Mol Sci 2024; 25:7914. [PMID: 39063156 PMCID: PMC11277244 DOI: 10.3390/ijms25147914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/07/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Autologous platelet-rich plasma (PRP) preparations are prepared at the point of care. Centrifugation cellular density separation sequesters a fresh unit of blood into three main fractions: a platelet-poor plasma (PPP) fraction, a stratum rich in platelets (platelet concentrate), and variable leukocyte bioformulation and erythrocyte fractions. The employment of autologous platelet concentrates facilitates the biological potential to accelerate and support numerous cellular activities that can lead to tissue repair, tissue regeneration, wound healing, and, ultimately, functional and structural repair. Normally, after PRP preparation, the PPP fraction is discarded. One of the less well-known but equally important features of PPP is that particular growth factors (GFs) are not abundantly present in PRP, as they reside outside of the platelet alpha granules. Precisely, insulin-like growth factor-1 (IGF-1) and hepatocyte growth factor (HGF) are mainly present in the PPP fraction. In addition to their roles as angiogenesis activators, these plasma-based GFs are also known to inhibit inflammation and fibrosis, and they promote keratinocyte migration and support tissue repair and wound healing. Additionally, PPP is known for the presence of exosomes and other macrovesicles, exerting cell-cell communication and cell signaling. Newly developed ultrafiltration technologies incorporate PPP processing methods by eliminating, in a fast and efficient manner, plasma water, cytokines, molecules, and plasma proteins with a molecular mass (weight) less than the pore size of the fibers. Consequently, a viable and viscous protein concentrate of functional total proteins, like fibrinogen, albumin, and alpha-2-macroglobulin is created. Consolidating a small volume of high platelet concentrate with a small volume of highly concentrated protein-rich PPP creates a protein-rich, platelet-rich plasma (PR-PRP) biological preparation. After the activation of proteins, mainly fibrinogen, the PR-PRP matrix retains and facilitates interactions between invading resident cells, like macrophages, fibroblast, and mesenchymal stem cells (MSCs), as well as the embedded concentrated PRP cells and molecules. The administered PR-PRP biologic will ultimately undergo fibrinolysis, leading to a sustained release of concentrated cells and molecules that have been retained in the PR-PRP matrix until the matrix is dissolved. We will discuss the unique biological and tissue reparative and regenerative properties of the PR-PRP matrix.
Collapse
Affiliation(s)
- Peter A. Everts
- Gulf Coast Biologics, A Non-Profit Organization, Fort Myers, FL 33916, USA
- OrthoRegen Group, Max-Planck University, Indaiatuba 13334-170, SP, Brazil;
| | - José Fábio Lana
- OrthoRegen Group, Max-Planck University, Indaiatuba 13334-170, SP, Brazil;
| | - Robert W. Alexander
- Regenevita Biocellular Aesthetic & Reconstructive Surgery, Cranio-Maxillofacial Surgery, Regenerative and Wound Healing, Hamilton, MT 59840, USA;
- Department of Surgery & Maxillofacial Surgery, School of Medicine & Dentistry, University of Washington, Seattle, WA 98195, USA
| | - Ignacio Dallo
- Unit of Biological Therapies and MSK Interventionism, Department of Orthopaedic Surgery and Sports Medicine, Sport Me Medical Center, 41013 Seville, Spain;
| | - Elizaveta Kon
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy;
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Mary A. Ambach
- BioEvolve, San Diego Orthobiologics and Sports Center, San Diego, CA 92024, USA
| | - André van Zundert
- Department of Anaesthesia and Perioperative Medicine, Royal Brisbane and Women’s Hospital, Brisbane and The University of Queensland, Brisbane 4072, Australia;
| | - Luga Podesta
- Bluetail Medical Group & Podesta Orthopedic Sports Medicine, Naples, FL 34109, USA;
- Physical Medicine & Rehabilitation Orlando College of Osteopathic Medicine, Orlando, FL 32806, USA
| |
Collapse
|
3
|
VerHulst EM, Galarza RMR, Herring IP, Ramos RV, Kemper AR. Comparison of conjunctival pedicle flap to corneal fixation strength achieved by Tisseel® fibrin glue, ethyl cyanoacrylate adhesive, ReSure® hydrogel sealant, and conventional suturing with 8-0 VICRYL® ophthalmic suture. Vet Ophthalmol 2024. [PMID: 38529836 DOI: 10.1111/vop.13197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 12/04/2023] [Accepted: 02/11/2024] [Indexed: 03/27/2024]
Abstract
OBJECTIVE To determine and compare the fixation strength of conjunctival pedicle flaps to cornea achieved via conventional ophthalmic suture and three different adhesive compounds. ANIMALS STUDIED Ex vivo porcine globes. PROCEDURES Following a 6 mm wide 500-micron-restricted depth lamellar keratectomy, conjunctival pedicle flaps were secured to the keratectomy site with either 8-0 VICRYL® suture or one of three adhesive products, including Tisseel® bioadhesive, ReSure® synthetic adhesive, or ethyl cyanoacrylate adhesive (n = 10 per surgical group). Adhesive application protocol varied by product based upon adhesive biocompatibility. Corneoconjunctival tissues were then harvested, clamped in a tensile testing device, and loaded at a rate of 1 mm/s under video surveillance until the point of failure. Peak load was determined for each test and used to compare fixation strength between samples. RESULTS Forty conjunctival flaps were performed, with 6 omitted from evaluation due to dehiscence prior to tensile testing. Of the 34 flaps analyzed, 10 were secured with suture, 10 with cyanoacrylate, 8 with ReSure®, and 6 with Tisseel®. Flaps secured with suture withstood significantly higher applied tensile force compared with cyanoacrylate (p = .02474), ReSure® (p = .00000), and Tisseel® (p = .00002). Flaps secured with cyanoacrylate withstood significantly greater force than those secured with ReSure® and Tisseel® (p = .01194 and 0.01798, respectively). There was no significant difference in fixation strength between ReSure® and Tisseel® glue (p = .95675). CONCLUSIONS Conjunctival pedicle flap fixation using 8-0 VICRYL® suture fixation was able to withstand significantly greater maximum tensile force compared to ReSure®, Tisseel®, or cyanoacrylate adhesives. Fixation strength achieved with cyanoacrylate adhesive was significantly greater than that achieved with ReSure® or Tisseel®.
Collapse
Affiliation(s)
- Elodie M VerHulst
- Department of Small Animal Clinical Sciences, Virginia Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Roxanne M Rodriguez Galarza
- Department of Small Animal Clinical Sciences, Virginia Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Ian P Herring
- Department of Small Animal Clinical Sciences, Virginia Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Renata Velloso Ramos
- Department of Small Animal Clinical Sciences, Virginia Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Andrew R Kemper
- Department of Biomedical Engineering and Mechanics, College of Engineering, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
4
|
Fang W, Zhai J, Qian ZB, Li HD, Wang MD, Shen LJ. Effect of a novel tyrosine kinase inhibitor nintedanib on bFGF and VEGF concentrations in a rabbit retinal vein occlusion model. Int J Ophthalmol 2023; 16:1450-1455. [PMID: 37724275 PMCID: PMC10475619 DOI: 10.18240/ijo.2023.09.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 06/21/2023] [Indexed: 09/20/2023] Open
Abstract
AIM To evaluate whether a novel tyrosine kinase inhibitor nintedanib could inhibit basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) simultaneously for retinal vascular disease in vivo. METHODS After a laser induced rabbit retinal vein occlusion (RVO) model was made, 0.5 mg of nintedanib was injected intravitreally in the left eye on the third day while the right eye was as a control. Intracameral samples were taken on the day before laser treatment and days 1, 3, 7, 14, 21, and 28 after treatment. Enzyme-linked immunosorbent assay (ELISA) was used to test the bFGF and VEGF-A concentrations in the aqueous humor. RESULTS Both bFGF and VEGF-A rose significantly on the third day after laser treatment in both eyes. In the control eye the bFGF concentration peaked on the 14th day while the VEGF-A concentration dropped rapidly soon after the third day. After nintadanib injection in the study eye, both bFGF and VEGF-A showed a significant reduction on the 4th day (7th day after laser treatment) when compared to the control eye, and kept on low level in the following several weeks. CONCLUSION Intravitreal injection of nintedanib can inhibit the expression of bFGF and VEGF in the process of RVO model to a certain extent, which is expected to become a new method for the treatment of retinal vascular diseases or fibrotic diseases.
Collapse
Affiliation(s)
- Wei Fang
- The Eye Hospital of Wenzhou Medical University, School of Opthalmology & Optometry, WMU, Zhejiang Eye Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Jing Zhai
- The Eye Hospital of Wenzhou Medical University, School of Opthalmology & Optometry, WMU, Zhejiang Eye Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Zhen-Bin Qian
- The Eye Hospital of Wenzhou Medical University, School of Opthalmology & Optometry, WMU, Zhejiang Eye Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Hai-Dong Li
- The Eye Hospital of Wenzhou Medical University, School of Opthalmology & Optometry, WMU, Zhejiang Eye Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Meng-Di Wang
- The Eye Hospital of Wenzhou Medical University, School of Opthalmology & Optometry, WMU, Zhejiang Eye Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Li-Jun Shen
- The Eye Hospital of Wenzhou Medical University, School of Opthalmology & Optometry, WMU, Zhejiang Eye Hospital, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
5
|
Podieh F, Wensveen R, Overboom M, Abbas L, Majolée J, Hordijk P. Differential role for rapid proteostasis in Rho GTPase-mediated control of quiescent endothelial integrity. J Biol Chem 2023; 299:104593. [PMID: 36894017 PMCID: PMC10124901 DOI: 10.1016/j.jbc.2023.104593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/09/2023] Open
Abstract
Endothelial monolayer permeability is regulated by actin dynamics and vesicular traffic. Recently, ubiquitination was also implicated in the integrity of quiescent endothelium, as it differentially controls the localization and stability of adhesion- and signaling proteins. However, the more general effect of fast protein turnover on endothelial integrity is not clear. Here, we found that inhibition of E1 ubiquitin ligases induces a rapid, reversible loss of integrity in quiescent, primary human endothelial monolayers, accompanied by increased F-actin stress fibers and the formation of intercellular gaps. Concomitantly, total protein and activity of the actin-regulating GTPase RhoB, but not its close homologue RhoA, increase ∼10-fold in 5-8 h. We determined that, the depletion of RhoB, but not of RhoA, the inhibition of actin contractility and the inhibition of protein synthesis all significantly rescue the loss of cell-cell contact induced by E1 ligase inhibition. Collectively, our data suggest that in quiescent human endothelial cells, the continuous and fast turnover of short-lived proteins that negatively regulate cell-cell contact, is essential to preserve monolayer integrity.
Collapse
Affiliation(s)
- Fabienne Podieh
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Roos Wensveen
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - MaxC Overboom
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Lotte Abbas
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Jisca Majolée
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands; Developmental Biology and Stem Cell Research, Hubrecht Institute, 3584 CT, Utrecht, The Netherlands
| | - PeterL Hordijk
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands.
| |
Collapse
|
6
|
Palermo A, Giannotti L, Di Chiara Stanca B, Ferrante F, Gnoni A, Nitti P, Calabriso N, Demitri C, Damiano F, Batani T, Lungherini M, Carluccio MA, Rapone B, Qorri E, Scarano A, Siculella L, Stanca E, Rochira A. Use of CGF in Oral and Implant Surgery: From Laboratory Evidence to Clinical Evaluation. Int J Mol Sci 2022; 23:15164. [PMID: 36499489 PMCID: PMC9736623 DOI: 10.3390/ijms232315164] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/03/2022] Open
Abstract
Edentulism is the condition of having lost natural teeth, and has serious social, psychological, and emotional consequences. The need for implant services in edentulous patients has dramatically increased during the last decades. In this study, the effects of concentrated growth factor (CGF), an autologous blood-derived biomaterial, in improving the process of osseointegration of dental implants have been evaluated. Here, permeation of dental implants with CGF has been obtained by using a Round up device. These CGF-coated dental implants retained a complex internal structure capable of releasing growth factors (VEGF, TGF-β1, and BMP-2) and matrix metalloproteinases (MMP-2 and MMP-9) over time. The CGF-permeated implants induced the osteogenic differentiation of human bone marrow stem cells (hBMSC) as confirmed by matrix mineralization and the expression of osteogenic differentiation markers. Moreover, CGF provided dental implants with a biocompatible and biologically active surface that significantly improved adhesion of endothelial cells on CGF-coated implants compared to control implants (without CGF). Finally, data obtained from surgical interventions with CGF-permeated dental implants presented better results in terms of optimal osseointegration and reduced post-surgical complications. These data, taken together, highlight new and interesting perspectives in the use of CGF in the dental implantology field to improve osseointegration and promote the healing process.
Collapse
Affiliation(s)
- Andrea Palermo
- College of Medicine and Dentistry Birmingham, University of Birmingham, Birmingham B4 6BN, UK
| | - Laura Giannotti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy
| | - Benedetta Di Chiara Stanca
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy
| | | | - Antonio Gnoni
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Paola Nitti
- Department of Engineering for Innovation, Campus Ecotekne, University of Salento, Via per Monteroni, 73100 Lecce, Italy
| | - Nadia Calabriso
- National Research Council (CNR) Institute of Clinical Physiology (IFC), 73100 Lecce, Italy
| | - Christian Demitri
- Department of Engineering for Innovation, Campus Ecotekne, University of Salento, Via per Monteroni, 73100 Lecce, Italy
| | - Fabrizio Damiano
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy
| | | | | | | | - Biagio Rapone
- Interdisciplinary Department of Medicine, Aldo Moro University of Bari, 70121 Bari, Italy
| | - Erda Qorri
- Faculty of Medical Science, Albanian University, Bulevardi Zogu I, 1001 Tirana, Albania
| | - Antonio Scarano
- Department of Oral Science, Nano and Biotechnology and CeSi-Met, University of Chieti-Pescara, 66100 Chieti, Italy
| | - Luisa Siculella
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy
| | - Eleonora Stanca
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy
| | - Alessio Rochira
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy
| |
Collapse
|
7
|
Gomez-Sosa JF, Cardier JE, Caviedes-Bucheli J. The hypoxia-dependent angiogenic process in dental pulp. J Oral Biosci 2022; 64:381-391. [PMID: 35998752 DOI: 10.1016/j.job.2022.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 08/09/2022] [Accepted: 08/15/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND In this review, we analyzed the existing literature to elucidate how the hypoxia-dependent angiogenic processes work in dental pulp. Angiogenesis is an essential biological process in the maturation and homeostasis of teeth. It involves multiple sequential steps such as endothelial cell proliferation and migration, cell-to-cell contact, and tube formation. HIGHLIGHT Clinical implications of understanding the process of angiogenesis include how the mineralization processes of dental pulp occur and how dental pulp maintains its homeostasis, preventing irreversible inflammation or necrosis. CONCLUSION The angiogenesis process in dental pulp regulates adequate concentrations of oxygen required for mineralization in root development and defense mechanisms against chronic stimuli.
Collapse
Affiliation(s)
- Jose Francisco Gomez-Sosa
- Unidad de Terapia Celular - Centro de Medicina Regenerativa, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas-Venezuela.
| | - Jose E Cardier
- Unidad de Terapia Celular - Centro de Medicina Regenerativa, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas-Venezuela
| | | |
Collapse
|
8
|
Cutini PH, Campelo AE, Massheimer VL. Vascular response to stress: Protective action of the bisphosphonate alendronate. Vasc Med 2022; 27:425-432. [PMID: 35879908 DOI: 10.1177/1358863x221112168] [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: 11/16/2022]
Abstract
BACKGROUND Since several additional actions of bone bisphosphonates have been proposed, we studied the effect of the bisphosphonate alendronate (ALN) on the vascular response to environmental stress. METHODS Primary cultures of endothelial cells (EC) and vascular smooth muscle cells (VSMC) exposed to strained conditions were employed for experimental evaluation. After ALN treatment, cell migration, proliferation, and angiogenesis assays were performed. The participation of signal transduction pathways in the biochemical action of ALN was also assessed. RESULTS In VSMC cultures, ALN counteracted the stimulation of cellular migration elicited by the proinflammatory agent lipopolysaccharide (LPS) or by high levels of calcium and phosphorus (osteogenic medium). Indeed, ALN reduced the increase of VSMC proliferation evoked by the stressors. When LPS and osteogenic medium were added simultaneously, the enhancement of cell proliferation dropped to control values in the presence of ALN. The mechanism of action of ALN involved the participation of nitric oxide synthase, mitogen-activated protein kinase (MAPK), and protein kinase C (PKC) signaling pathways. The study revealed that ALN exhibits a proangiogenic action. On EC, ALN enhanced vascular endothelial growth factor (VEGF) synthesis, and induced capillary-like tube formation in a VEGF-dependent manner. The presence of vascular stress conditions (LPS or osteogenic medium) did not modify the proangiogenic action elicited by ALN. CONCLUSION The findings presented suggest an extra-bone biological action of ALN, which could contribute to the maintenance of vascular homeostasis avoiding cellular damage elicited by environmental stress.
Collapse
Affiliation(s)
- Pablo H Cutini
- Departamento de Biología, Bioquímica y Farmacia, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Sur (UNS), Bahía Blanca, Buenos Aires, Argentina
| | - Adrián E Campelo
- Departamento de Biología, Bioquímica y Farmacia, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Sur (UNS), Bahía Blanca, Buenos Aires, Argentina
| | - Virginia L Massheimer
- Departamento de Biología, Bioquímica y Farmacia, Instituto de Ciencias Biológicas y Biomédicas del Sur (INBIOSUR), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional del Sur (UNS), Bahía Blanca, Buenos Aires, Argentina
| |
Collapse
|
9
|
Human Milk Oligosaccharides in Cord Blood Are Altered in Gestational Diabetes and Stimulate Feto-Placental Angiogenesis In Vitro. Nutrients 2021; 13:nu13124257. [PMID: 34959807 PMCID: PMC8705424 DOI: 10.3390/nu13124257] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 12/11/2022] Open
Abstract
(1) Background: Human milk oligosaccharides (HMOs) are present in maternal serum during pregnancy and their composition is altered in gestational diabetes (GDM). HMOs are also in fetal cord blood and in contact with the feto-placental endothelium, potentially affecting its functions, such as angiogenesis. We hypothesized that cord blood HMOs are changed in GDM and contribute to increased feto-placental angiogenesis, hallmark of GDM. (2) Methods: Using HPLC, we quantified HMOs in cord blood of women with normal glucose tolerance (NGT, n = 25) or GDM (n = 26). We investigated in vitro angiogenesis using primary feto-placental endothelial cells (fpECs) from term placentas after healthy pregnancy (n = 10), in presence or absence of HMOs (100 µg/mL) isolated from human milk, 3′-sialyllactose (3′SL, 30 µg/mL) and lactose (glycan control) and determined network formation (Matrigel assay), proliferation (MTT assays), actin organization (F-actin staining), tube formation (fibrin tube formation assay) and sprouting (spheroid sprouting assay). (3) Results: 3′SL was higher in GDM cord blood. HMOs increased network formation, HMOs and 3’SL increased proliferation and F-actin staining. In fibrin assays, HMOs and 3’SL increased total tube length by 24% and 25% (p < 0.05), in spheroid assays, by 32% (p < 0.05) and 21% (p = 0.056), respectively. Lactose had no effect. (4) Conclusions: Our study suggests a novel role of HMOs in feto-placental angiogenesis and indicates a contribution of HMO composition to altered feto-placental vascularization in GDM.
Collapse
|
10
|
Atallah R, Gindlhuber J, Platzer W, Bärnthaler T, Tatzl E, Toller W, Strutz J, Rittchen S, Luschnig P, Birner-Gruenberger R, Wadsack C, Heinemann A. SUCNR1 Is Expressed in Human Placenta and Mediates Angiogenesis: Significance in Gestational Diabetes. Int J Mol Sci 2021; 22:12048. [PMID: 34769478 PMCID: PMC8585094 DOI: 10.3390/ijms222112048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 12/19/2022] Open
Abstract
Placental hypervascularization has been reported in pregnancy-related pathologies such as gestational diabetes mellitus (GDM). Nevertheless, the underlying causes behind this abnormality are not well understood. In this study, we addressed the expression of SUCNR1 (cognate succinate receptor) in human placental endothelial cells and hypothesized that the succinate-SUCNR1 axis might play a role in the placental hypervascularization reported in GDM. We measured significantly higher succinate levels in placental tissue lysates from women with GDM relative to matched controls. In parallel, SUCNR1 protein expression was upregulated in GDM tissue lysates as well as in isolated diabetic fetoplacental arterial endothelial cells (FpECAds). A positive correlation of SUCNR1 and vascular endothelial growth factor (VEGF) protein levels in tissue lysates indicated a potential link between the succinate-SUCNR1 axis and placental angiogenesis. In our in vitro experiments, succinate prompted hallmarks of angiogenesis in human umbilical vein endothelial cells (HUVECs) such as proliferation, migration and spheroid sprouting. These results were further validated in fetoplacental arterial endothelial cells (FpECAs), where succinate induced endothelial tube formation. VEGF gene expression was increased in response to succinate in both HUVECs and FpECAs. Yet, knockdown of SUCNR1 in HUVECs led to suppression of VEGF gene expression and abrogated the migratory ability and wound healing in response to succinate. In conclusion, our data underline SUCNR1 as a promising metabolic target in human placenta and as a potential driver of enhanced placental angiogenesis in GDM.
Collapse
MESH Headings
- Adult
- Case-Control Studies
- Cells, Cultured
- Diabetes, Gestational/genetics
- Diabetes, Gestational/metabolism
- Diabetes, Gestational/physiopathology
- Endothelium, Vascular/metabolism
- Female
- Human Umbilical Vein Endothelial Cells
- Humans
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Neovascularization, Physiologic/genetics
- Placenta/blood supply
- Placenta/metabolism
- Pregnancy
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/physiology
Collapse
Affiliation(s)
- Reham Atallah
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (R.A.); (W.P.); (T.B.); (S.R.); (P.L.)
- National Research Centre, Cairo 12622, Egypt
| | - Juergen Gindlhuber
- Diagnostic and Research Institute of Pathology, Medical University of Graz, 8010 Graz, Austria; (J.G.); (R.B.-G.)
- Institute of Chemical Technologies and Analytics, Technische Universität Wien, 1060 Vienna, Austria
| | - Wolfgang Platzer
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (R.A.); (W.P.); (T.B.); (S.R.); (P.L.)
| | - Thomas Bärnthaler
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (R.A.); (W.P.); (T.B.); (S.R.); (P.L.)
| | - Eva Tatzl
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, 8036 Graz, Austria; (E.T.); (W.T.)
| | - Wolfgang Toller
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, 8036 Graz, Austria; (E.T.); (W.T.)
| | - Jasmin Strutz
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (J.S.); (C.W.)
- Institute of Biomedical Science, Carinthia University of Applied Sciences, 9020 Klagenfurt, Austria
| | - Sonja Rittchen
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (R.A.); (W.P.); (T.B.); (S.R.); (P.L.)
| | - Petra Luschnig
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (R.A.); (W.P.); (T.B.); (S.R.); (P.L.)
| | - Ruth Birner-Gruenberger
- Diagnostic and Research Institute of Pathology, Medical University of Graz, 8010 Graz, Austria; (J.G.); (R.B.-G.)
- Institute of Chemical Technologies and Analytics, Technische Universität Wien, 1060 Vienna, Austria
| | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (J.S.); (C.W.)
| | - Akos Heinemann
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (R.A.); (W.P.); (T.B.); (S.R.); (P.L.)
| |
Collapse
|
11
|
Fazil S, Shah H, Noreen M, Yar M, Farooq Khan A, Zaman Safi S, Yousef Alomar S, Fahad Alkhuriji A, Mualla Alharbi H, Sohail Afzal M. Evaluation of molecular mechanisms of heparin-induced angiogenesis, in human umbilical vein endothelial cells. JOURNAL OF KING SAUD UNIVERSITY - SCIENCE 2021; 33:101534. [DOI: 10.1016/j.jksus.2021.101534] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/07/2024]
|
12
|
Yao B, Wang F, Zhao X, Wang B, Yue X, Ding Y, Liu G. Effect of a Topical Nonsteroidal Anti-Inflammatory Drug (0.1% Pranoprofen) on VEGF and COX-2 Expression in Primary Pterygium. Front Pharmacol 2021; 12:709251. [PMID: 34305615 PMCID: PMC8298757 DOI: 10.3389/fphar.2021.709251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 06/30/2021] [Indexed: 11/17/2022] Open
Abstract
Purpose: To evaluate the effect of a topical nonsteroidal anti-inflammatory drug (0.1% pranoprofen) on the expression of VEGF and Cox-2 in primary pterygium. Methods: This was a prospective, randomized study. Between January 2019 and April 2020, 120 patients diagnosed with primary pterygium were enrolled and randomly divided into three groups before operation: 1) 40 patients in group 1 received topical pranoprofen 0.1% four times daily for 4 weeks, 2) 40 patients in group 2 received topical fluorometholone 0.1% four times daily for 4 weeks, and 3) patients in group 3 did not receive treatment. For each group, the age, sex, eye type, best-corrected visual acuity (BCVA), intraocular pressure (IOP), duration of onset, combined systemic diseases, and the results regarding vascular endothelial growth factor (VEGF) and cyclo-oxygen-ase-2 (COX-2) in postoperative pterygial tissues were evaluated in detail. Results: There were no significant differences regarding age, sex, eye type, combined systemic diseases, duration of onset, IOP, and BCVA within the three groups (p > 0.05). The reduction of VEGF and CoX-2 expression of pterygial vascular endothelial cells in group 1 were statistically significant compared to group 2 and group 3 (p < 0.05). There were significant correlations between COX-2 and VEGF expression of pterygial tissues within the three groups (p < 0.05). Conclusion: The present findings suggested that the topical pranoprofen 0.1% could reduce the expression of VEGF and COX-2 in primary pterygium. We confirmed that treatment with pranoprofen offers advantages in early intervention and has therapeutic potential in reducing the postoperative recurrence of primary pterygium patients. Clinical Trial registration: The study was registered with the Chinese Clinical Trial Registry. (http://www.chictr.org.cn/index.aspx, Registration Number: ChiCTR2100047726).
Collapse
Affiliation(s)
- Bangtao Yao
- Department of Ophthalmology, Nanjing Lishui District People's Hospital, Lishui Branch of Southeast University Affiliated Zhongda Hospital, Nanjing, China
| | - Fei Wang
- Department of Ophthalmology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaogui Zhao
- Department of Ophthalmology, Nanjing Lishui District People's Hospital, Lishui Branch of Southeast University Affiliated Zhongda Hospital, Nanjing, China
| | - Bei Wang
- Department of Ophthalmology, Nanjing Lishui District People's Hospital, Lishui Branch of Southeast University Affiliated Zhongda Hospital, Nanjing, China
| | - Xiaoli Yue
- Department of Pathology, Nanjing Lishui District People's Hospital, Lishui Branch of Southeast University Affiliated Zhongda Hospital, Nanjing, China
| | - Yuhua Ding
- Department of Ophthalmology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Gang Liu
- Department of Ophthalmology, Nanjing Lishui District People's Hospital, Lishui Branch of Southeast University Affiliated Zhongda Hospital, Nanjing, China
| |
Collapse
|
13
|
Ding X, Zhao H, Li Y, Lee AL, Li Z, Fu M, Li C, Yang YY, Yuan P. Synthetic peptide hydrogels as 3D scaffolds for tissue engineering. Adv Drug Deliv Rev 2020; 160:78-104. [PMID: 33091503 DOI: 10.1016/j.addr.2020.10.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 09/25/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022]
Abstract
The regeneration of tissues and organs poses an immense challenge due to the extreme complexity in the research work involved. Despite the tissue engineering approach being considered as a promising strategy for more than two decades, a key issue impeding its progress is the lack of ideal scaffold materials. Nature-inspired synthetic peptide hydrogels are inherently biocompatible, and its high resemblance to extracellular matrix makes peptide hydrogels suitable 3D scaffold materials. This review covers the important aspects of peptide hydrogels as 3D scaffolds, including mechanical properties, biodegradability and bioactivity, and the current approaches in creating matrices with optimized features. Many of these scaffolds contain peptide sequences that are widely reported for tissue repair and regeneration and these peptide sequences will also be discussed. Furthermore, 3D biofabrication strategies of synthetic peptide hydrogels and the recent advances of peptide hydrogels in tissue engineering will also be described to reflect the current trend in the field. In the final section, we will present the future outlook in the design and development of peptide-based hydrogels for translational tissue engineering applications.
Collapse
Affiliation(s)
- Xin Ding
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| | - Huimin Zhao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yuzhen Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Ashlynn Lingzhi Lee
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Zongshao Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Mengjing Fu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Chengnan Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yi Yan Yang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore.
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
14
|
Chen M, Bao L, Zhao M, Cao J, Zheng H. Progress in Research on the Role of FGF in the Formation and Treatment of Corneal Neovascularization. Front Pharmacol 2020; 11:111. [PMID: 32158390 PMCID: PMC7052042 DOI: 10.3389/fphar.2020.00111] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/28/2020] [Indexed: 12/23/2022] Open
Abstract
Corneal neovascularization (CNV) is a sight-threatening disease usually associated with inflammatory, infectious, degenerative, and traumatic disorders of the ocular surface. Fibroblast growth factor (FGF) family members play an important role in angiogenesis to induce corneal neovascularization, which significantly affects the differentiation, proliferation, metastasis, and chemotaxis of vascular endothelial cells. Both acidic fibroblast growth factor (aFGF) and basic fibroblast growth factor (bFGF) demonstrate positive staining in capillaries and induce corneal stromal cells. The anabolism of endothelial cells is induced by bFGF in corneal neovascularization. FGFs exert their effects via specific binding to cell surface-expressed specific receptors. We believe that both anti-FGF antibodies and anti-FGF receptor antibodies represent new directions for the treatment of CNV. Similar to anti-vascular endothelial growth factor antibodies, subconjunctival injection and eye drops can be considered effective forms of drug delivery.
Collapse
Affiliation(s)
- Mengji Chen
- Department of Ophthalmology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Licheng Bao
- Department of Ophthalmology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mengying Zhao
- Department of Ophthalmology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiarong Cao
- Department of Ophthalmology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haihua Zheng
- Department of Ophthalmology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
15
|
Beamish JA, Juliar BA, Cleveland DS, Busch ME, Nimmagadda L, Putnam AJ. Deciphering the relative roles of matrix metalloproteinase- and plasmin-mediated matrix degradation during capillary morphogenesis using engineered hydrogels. J Biomed Mater Res B Appl Biomater 2019; 107:2507-2516. [PMID: 30784190 PMCID: PMC6699943 DOI: 10.1002/jbm.b.34341] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/09/2019] [Accepted: 01/26/2019] [Indexed: 12/20/2022]
Abstract
Extracellular matrix (ECM) remodeling is essential for the process of capillary morphogenesis. Here we employed synthetic poly(ethylene glycol) (PEG) hydrogels engineered with proteolytic specificity to either matrix metalloproteinases (MMPs), plasmin, or both to investigate the relative contributions of MMP- and plasmin-mediated ECM remodeling to vessel formation in a 3D-model of capillary self-assembly analogous to vasculogenesis. We first demonstrated a role for both MMP- and plasmin-mediated mechanisms of ECM remodeling in an endothelial-fibroblast co-culture model of vasculogenesis in fibrin hydrogels using inhibitors of MMPs and plasmin. When this co-culture model was employed in engineered PEG hydrogels with selective protease sensitivity, we observed robust capillary morphogenesis only in MMP-sensitive matrices. Fibroblast spreading in plasmin-selective hydrogels confirmed this difference was due to protease preference by endothelial cells, not due to limitations of the matrix itself. In hydrogels engineered with crosslinks that were dually susceptible to MMPs and plasmin, capillary morphogenesis was unchanged. These findings highlight the critical importance of MMP-mediated degradation during vasculogenesis and provide strong evidence to justify the preferential selection of MMP-degradable peptide crosslinkers in synthetic hydrogels used to study vascular morphogenesis and promote vascularization. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B:2507-2516, 2019.
Collapse
Affiliation(s)
- Jeffrey A. Beamish
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Benjamin A. Juliar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - David S. Cleveland
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Megan E. Busch
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Likitha Nimmagadda
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Andrew J. Putnam
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
16
|
Outgrowth, proliferation, viability, angiogenesis and phenotype of primary human endothelial cells in different purchasable endothelial culture media: feed wisely. Histochem Cell Biol 2019; 152:377-390. [PMID: 31541300 PMCID: PMC6842357 DOI: 10.1007/s00418-019-01815-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2019] [Indexed: 12/15/2022]
Abstract
Function and dysfunction of endothelial cells are regulated by a multitude of factors. Endothelial cell research often requires in vitro cell culture experiments. Hence, various culture media specifically designed to promote endothelial cell growth are available. These strikingly differ in their composition: complex media contain endothelial cell growth supplement (ECGS), an extract produced of bovine brain with undefined amounts of biologically active compounds, whilst defined media contain selected growth factors in defined concentrations. We here compared the effect of seven purchasable endothelial cell culture media on colony outgrowth, proliferation, viability, in vitro angiogenesis and phenotype of mature primary human endothelial cells using feto-placental endothelial cells isolated from chorionic arteries (fpEC). The effect of media on colony outgrowth was additionally tested on umbilical cord blood-derived endothelial progenitor cells (ECFCs). Outgrowth, purity, proliferation and viability differed between media. Outgrowth of fpEC and ECFCs was best in a defined medium containing EGF, FGF2 and VEGF. By contrast, established fpEC isolations proliferated best in complex media containing ECGS, heparin and ascorbic acid. Also viability of cells was higher in complex media. In vitro angiogenesis was most intense in a defined medium containing the highest number of individual growth factors. FACS analysis of surface markers for endothelial cell subtypes revealed that endothelial phenotype of fpEC was unaffected by media composition. Our data demonstrate the fundamental effect of endothelial cell culture media on primary cell isolation success and behaviour. Whether the composition of supplements is suitable also for individual experiments needs to be tested specifically.
Collapse
|
17
|
Monsuur HN, Weijers EM, Gibbs S, van den Broek LJ. Skin substitutes are more potent than dermal or epidermal substitutes in stimulating endothelial cell sprouting. BMC Biomed Eng 2019; 1:18. [PMID: 32903380 PMCID: PMC7422578 DOI: 10.1186/s42490-019-0018-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 06/11/2019] [Indexed: 12/01/2022] Open
Abstract
Background Therapy resistant ulcers are wounds that remain open for a long time period and often arise from chronic venous disease, prolonged pressure or diabetes. For healing of chronic wounds, revitalization of the inert wound bed, which is achieved by angiogenic sprouting of new blood vessels is of great importance. An alternative treatment option to conventional therapies is the use of skin substitutes: dermal (DS), epidermal (ES) or bi-layered skin substitutes (SS). The aim of this study was to determine the mode of action of an autologous SS, ES and DS with regards to endothelial cell proliferation, migration and angiogenic sprouting into a fibrin hydrogel. Results SS consists of a fully differentiated epidermis expanding over the acellular donor dermis (AD) which has become repopulated with fibroblasts. DS is the same construct as SS but without the epidermis and ES is the same construct as SS but without the fibroblasts. As a control, AD was used throughout. It was found that the bi-layered SS was the most potent substitute in inducing migration and sprouting of endothelial cells. The cross talk between dermis and epidermis resulted in the strongest induction of sprouting via VEGF and uPAR. ES stimulated sprouting more than DS again via VEGF and uPAR. The slight induction of sprouting mediated by DS was not mediated by VEGF, but was in part stimulated through uPAR. Conclusion This in vitro study supports our clinical observations that a bi-layered SS is a strong stimulator of angiogenesis and therefore has the potential to revitalize an inert wound bed.
Collapse
Affiliation(s)
- Hanneke N Monsuur
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ester M Weijers
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdan and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Lenie J van den Broek
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Tasev D, Dekker-Vroling L, van Wijhe M, Broxterman HJ, Koolwijk P, van Hinsbergh VWM. Hypoxia Impairs Initial Outgrowth of Endothelial Colony Forming Cells and Reduces Their Proliferative and Sprouting Potential. Front Med (Lausanne) 2018; 5:356. [PMID: 30619865 PMCID: PMC6306419 DOI: 10.3389/fmed.2018.00356] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 12/06/2018] [Indexed: 01/09/2023] Open
Abstract
Vascular homeostasis and regeneration in ischemic tissue relies on intrinsic competence of the tissue to rapidly recruit endothelial cells for vascularization. The mononuclear cell (MNC) fraction of blood contains circulating progenitors committed to endothelial lineage. These progenitors give rise to endothelial colony-forming cells (ECFCs) that actively participate in neovascularization of ischemic tissue. To evaluate if the initial clonal outgrowth of ECFCs from cord (CB) and peripheral blood (PB) was stimulated by hypoxic conditions, MNCs obtained from CB and PB were subjected to 20 and 1% O2 cell culture conditions. Clonal outgrowth was followed during a 30 day incubation period. Hypoxia impaired the initial outgrowth of ECFC colonies from CB and also reduced their number that were developing from PB MNCs. Three days of oxygenation (20% O2) prior to hypoxia could overcome the initial CB-ECFC outgrowth. Once proliferating and subcultured the CB-ECFCs growth was only modestly affected by hypoxia; proliferation of PB-ECFCs was reduced to a similar extent (18-30% reduction). Early passages of subcultured CB- and PB-ECFCs contained only viable cells and few if any senescent cells. Tube formation by subcultured PB-ECFCs was also markedly inhibited by continuous exposure to 1% O2. Gene expression profiles point to regulation of the cell cycle and metabolism as major altered gene clusters. Finally we discuss our counterintuitive observations in the context of the important role that hypoxia has in promoting neovascularization.
Collapse
Affiliation(s)
- Dimitar Tasev
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Laura Dekker-Vroling
- Department of Medical Oncology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Michiel van Wijhe
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Henk J Broxterman
- Department of Medical Oncology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Pieter Koolwijk
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Victor W M van Hinsbergh
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| |
Collapse
|
19
|
Boas SEM, Carvalho J, van den Broek M, Weijers EM, Goumans MJ, Koolwijk P, Merks RMH. A local uPAR-plasmin-TGFβ1 positive feedback loop in a qualitative computational model of angiogenic sprouting explains the in vitro effect of fibrinogen variants. PLoS Comput Biol 2018; 14:e1006239. [PMID: 29979675 PMCID: PMC6072121 DOI: 10.1371/journal.pcbi.1006239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 08/02/2018] [Accepted: 05/28/2018] [Indexed: 11/19/2022] Open
Abstract
In experimental assays of angiogenesis in three-dimensional fibrin matrices, a temporary scaffold formed during wound healing, the type and composition of fibrin impacts the level of sprouting. More sprouts form on high molecular weight (HMW) than on low molecular weight (LMW) fibrin. It is unclear what mechanisms regulate the number and the positions of the vascular-like structures in cell cultures. To address this question, we propose a mechanistic simulation model of endothelial cell migration and fibrin proteolysis by the plasmin system. The model is a hybrid, cell-based and continuum, computational model based on the cellular Potts model and sets of partial-differential equations. Based on the model results, we propose that a positive feedback mechanism between uPAR, plasmin and transforming growth factor β1 (TGFβ1) selects cells in the monolayer for matrix invasion. Invading cells releases TGFβ1 from the extracellular matrix through plasmin-mediated fibrin degradation. The activated TGFβ1 further stimulates fibrin degradation and keeps proteolysis active as the sprout invades the fibrin matrix. The binding capacity for TGFβ1 of LMW is reduced relative to that of HMW. This leads to reduced activation of proteolysis and, consequently, reduced cell ingrowth in LMW fibrin compared to HMW fibrin. Thus our model predicts that endothelial cells in LMW fibrin matrices compared to HMW matrices show reduced sprouting due to a lower bio-availability of TGFβ1. Therapies for a range of medical conditions, including cancer, wound healing and diabetic retinopathy can benefit from a better control over the growth of blood vessels. The chemical properties of fibrin, the material that forms scabs in wounds and can also occur in large concentrations in tumors, can regulate the degree of blood vessel growth (angiogenesis). Angiogenesis can be mimicked in cell cultures. These allow us to modulate the chemical properties of fibrin and study the effect on angiogenesis. Fibrin occurs in high molecular weight (HMW) and in low molecular weight (LMW) forms. Interestingly, there is more ingrowth of angiogenic-like structures into HMW than in LMW fibrin, but the mechanisms are poorly understood. To get more insight into these, we constructed a computational model. Using the model, we propose and analyse a hypothetical mechanism for sprouting that could explain the differences in endothelial cell sprouting in LMW and HMW fibrin matrices. Our model suggests that cells digest fibrin, thus creating space for ingrowth. At the same time, digestion frees growth factors bound to fibrin, that activates further secretion of digestive enzymes by the cells. We propose that the resulting positive feedback loop spontaneously selects cells in the endothelial monolayer for ingrowth and helps the blood vessel sprout move deeper into the fibrin. This could be a complementary mechanism to lateral-inhibition by Delta-Notch for the selection of leader cells, also called ‘tip cells’. Our model predicts that endothelial cells in LMW fibrin compared to HMW fibrin show reduced sprouting due to a lower bio-availability of TGFβ1.
Collapse
Affiliation(s)
- Sonja E. M. Boas
- Centrum Wiskunde & Informatica (CWI), Amsterdam, The Netherlands
- Mathematical Institute, Leiden University, Leiden, The Netherlands
| | - Joao Carvalho
- Centrum Wiskunde & Informatica (CWI), Amsterdam, The Netherlands
- CFisUC, Department of Physics, University of Coimbra, Coimbra, Portugal
| | - Marloes van den Broek
- Amsterdam Cardiovascular Sciences, VU University medical Center, Dept. of Physiology, Amsterdam, The Netherlands
| | - Ester M. Weijers
- Amsterdam Cardiovascular Sciences, VU University medical Center, Dept. of Physiology, Amsterdam, The Netherlands
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Pieter Koolwijk
- Amsterdam Cardiovascular Sciences, VU University medical Center, Dept. of Physiology, Amsterdam, The Netherlands
| | - Roeland M. H. Merks
- Centrum Wiskunde & Informatica (CWI), Amsterdam, The Netherlands
- Mathematical Institute, Leiden University, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
20
|
Jung B, Hong S, Kim SC, Hwang C. In Vivo Observation of Endothelial Cell-Assisted Vascularization in Pancreatic Cancer Xenograft Engineering. Tissue Eng Regen Med 2018; 15:275-285. [PMID: 30603553 PMCID: PMC6171679 DOI: 10.1007/s13770-018-0113-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/10/2017] [Accepted: 01/02/2018] [Indexed: 02/07/2023] Open
Abstract
In this study, for better understanding of patient-derived xenograft (PDX) generation, angiogenic characteristics during PDX cancerous tissue generation was investigated with different initial cell seeding conditions in the hydrogel. We monitored the angiogenic changes during the formation of in vivo cancer cell line xenografts induced by endothelial cells. Our in vivo cancer tissue formation system was designed with the assistance of tissue engineering technology to mimic patient-derived xenograft formation. Endothelial cells and MIA PaCa-2 pancreatic carcinoma cells were encapsulated in fibrin gel at different mixing configurations and subcutaneously implanted into nude mice. To investigate the effect of the initial cancerous cell distribution in the fibrin gel, MIA PaCa-2 cells were encapsulated as a homogeneous cell distribution or as a cell aggregate, with endothelial cells homogeneously distributed in the fibrin gel. Histological observation of the explanted tissues after different implantation periods revealed three different stages: isolated vascular tubes, leaky blood vessels, and mature cancerous tissue formation. The in vivo engineered cancerous tissues had leaky blood vessels with low expression of the vascular tight junction marker CD31. Under our experimental conditions, complex cancer-like tissue formation was most successful when tumorous cells and endothelial cells were homogeneously mixed in the fibrin gel. The present study implies that tumorous xenograft tissue formation can be achieved with a low number of initial cells and that effective vascularization conditions can be attained with a limited volume of patient-derived cancer tissue. Endothelial cell-assisted vascularization can be a potent choice for the effective development of vascularized cancerous tissues for studying patient-derived xenografts, cancer angiogenesis, cancer metastasis, and anticancer drugs.
Collapse
Affiliation(s)
- Boyoung Jung
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 South Korea
- University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 South Korea
| | - Soyoung Hong
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 South Korea
| | - Song Cheol Kim
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 South Korea
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine and Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 South Korea
| | - Changmo Hwang
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 South Korea
- University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 South Korea
| |
Collapse
|
21
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
22
|
The minor histocompatibility antigen 1 (HMHA1)/ArhGAP45 is a RacGAP and a novel regulator of endothelial integrity. Vascul Pharmacol 2017; 101:38-47. [PMID: 29174013 DOI: 10.1016/j.vph.2017.11.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/14/2017] [Accepted: 11/18/2017] [Indexed: 12/25/2022]
Abstract
Endothelial cells line the vasculature and act as gatekeepers that control the passage of plasma, macromolecules and cells from the circulation to the interstitial space. Dysfunction of the endothelial barrier can lead to uncontrolled leak or edema. Vascular leakage is a hallmark of a range of diseases and despite its large impact no specialized therapies are available to prevent or reduce it. RhoGTPases are known key regulators of cellular behavior that are directly involved in the regulation of the endothelial barrier. We recently performed a comprehensive analysis of the effect of all RhoGTPases and their regulators on basal endothelial integrity. In addition to novel positive regulators of endothelial barrier function, we also identified novel negative regulators, of which the ArhGAP45 (also known as HMHA1) was the most significant. We now demonstrate that ArhGAP45 acts as a Rac-GAP (GTPase-Activating Protein) in endothelial cells, which explains its negative effect on endothelial barrier function. Silencing ArhGAP45 not only promotes basal endothelial barrier function, but also increases cellular surface area and induces sprout formation in a 3D-fibrin matrix. Our data further shows that loss of ArhGAP45 promotes migration and shear stress adaptation. In conclusion, we identify ArhGAP45 (HMHA1) as a novel regulator, which contributes to the fine-tuning of the regulation of basal endothelial integrity.
Collapse
|
23
|
Liu D, Peng C, Jiang Z, Tao L. Relationship between expression of cyclooxygenase 2 and neovascularization in human pterygia. Oncotarget 2017; 8:105630-105636. [PMID: 29285278 PMCID: PMC5739665 DOI: 10.18632/oncotarget.22351] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/25/2017] [Indexed: 11/25/2022] Open
Abstract
In present study, we are to test the relationship between cyclooxygenase 2 (COX-2) and angiogenesis in pterygium tissues from a group of Chinese patients. Here forty-five primary pterygium tissues and twenty-three normal bulbar conjunctival tissues were obtained during ophthalmologic surgeries. The primary pterygium samples were treated for the immunohistochemical evaluation of COX-2, CD31 and vascular endothelial growth factor (VEGF) antibodies for different tissues. In order to evaluate the relationship between COX-2 and neovascularization, a statistical analysis was performed using the SPSS 13.0 statistical software package. As results, in our study, 36 (80%) of the primary pterygia samples were found to be positive for COX-2 staining, which was not found in the normal conjunctivas. The density of the microvessels (MVD) was significantly increased in the COX-2 positive patients when compared to the COX-2 negative ones (19.06 ± 1.84 vs.10.44 ± 2.98, P=1.36×10-5) in the pterygia cases. In the group that was positive for COX-2, there were 39 (86.7%) samples with VEGF expression. Furthermore, the staining of both COX-2 and VEGF was localized to the lower and middle layers of the epithelium and the endothelial cells of the microvessels. When analyzed the relation between them, the expression of COX-2 showed a significant correlation with the MVD (P = 4.02×10-4) and VEGF (p = 2.72×10-4). In conclusion, the present study showed that COX-2 may play an important role in stimulating the angiogenesis of pterygium in concert with VEGF.
Collapse
Affiliation(s)
- Dongwei Liu
- Department of Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, PR China
| | - Chang Peng
- Department of Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, PR China
| | - Zhengxuan Jiang
- Department of Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, PR China
| | - Liming Tao
- Department of Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, PR China
| |
Collapse
|
24
|
Stepanova V, Dergilev KV, Holman KR, Parfyonova YV, Tsokolaeva ZI, Teter M, Atochina-Vasserman EN, Volgina A, Zaitsev SV, Lewis SP, Zabozlaev FG, Obraztsova K, Krymskaya VP, Cines DB. Urokinase-type plasminogen activator (uPA) is critical for progression of tuberous sclerosis complex 2 (TSC2)-deficient tumors. J Biol Chem 2017; 292:20528-20543. [PMID: 28972182 DOI: 10.1074/jbc.m117.799593] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/20/2017] [Indexed: 12/20/2022] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a fatal lung disease associated with germline or somatic inactivating mutations in tuberous sclerosis complex genes (TSC1 or TSC2). LAM is characterized by neoplastic growth of smooth muscle-α-actin-positive cells that destroy lung parenchyma and by the formation of benign renal neoplasms called angiolipomas. The mammalian target of rapamycin complex 1 (mTORC1) inhibitor rapamycin slows progression of these diseases but is not curative and associated with notable toxicity at clinically effective doses, highlighting the need for better understanding LAM's molecular etiology. We report here that LAM lesions and angiomyolipomas overexpress urokinase-type plasminogen activator (uPA). Tsc1-/- and Tsc2-/- mouse embryonic fibroblasts expressed higher uPA levels than their WT counterparts, resulting from the TSC inactivation. Inhibition of uPA expression in Tsc2-null cells reduced the growth and invasiveness and increased susceptibility to apoptosis. However, rapamycin further increased uPA expression in TSC2-null tumor cells and immortalized TSC2-null angiomyolipoma cells, but not in cells with intact TSC. Induction of glucocorticoid receptor signaling or forkhead box (FOXO) 1/3 inhibition abolished the rapamycin-induced uPA expression in TSC-compromised cells. Moreover, rapamycin-enhanced migration of TSC2-null cells was inhibited by the uPA inhibitor UK122, dexamethasone, and a FOXO inhibitor. uPA-knock-out mice developed fewer and smaller TSC2-null lung tumors, and introduction of uPA shRNA in tumor cells or amiloride-induced uPA inhibition reduced tumorigenesis in vivo These findings suggest that interference with the uPA-dependent pathway, when used along with rapamycin, might attenuate LAM progression and potentially other TSC-related disorders.
Collapse
Affiliation(s)
| | - Konstantin V Dergilev
- the Angiogenesis Laboratory, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Moscow 121552, Russia
| | - Kelci R Holman
- the College of Arts and Sciences, Drexel University, Philadelphia, Pennsylvania 19104, and
| | - Yelena V Parfyonova
- the Angiogenesis Laboratory, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Moscow 121552, Russia
| | - Zoya I Tsokolaeva
- the Angiogenesis Laboratory, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Moscow 121552, Russia
| | - Mimi Teter
- the College of Arts and Sciences, Drexel University, Philadelphia, Pennsylvania 19104, and
| | - Elena N Atochina-Vasserman
- Penn Center for Pulmonary Biology, Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | - Alla Volgina
- Penn Center for Pulmonary Biology, Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | | | - Shane P Lewis
- the College of Arts and Sciences, Drexel University, Philadelphia, Pennsylvania 19104, and
| | - Fedor G Zabozlaev
- the Department of Pathology, Federal Research Clinical Center Federal Medical and Biological Agency of Russia, Moscow 115682, Russia
| | - Kseniya Obraztsova
- Penn Center for Pulmonary Biology, Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | - Vera P Krymskaya
- Penn Center for Pulmonary Biology, Pulmonary, Allergy, and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | - Douglas B Cines
- From the Department of Pathology and Laboratory Medicine and
| |
Collapse
|
25
|
Pirro M, Bianconi V, Paciullo F, Mannarino MR, Bagaglia F, Sahebkar A. Lipoprotein(a) and inflammation: A dangerous duet leading to endothelial loss of integrity. Pharmacol Res 2017; 119:178-187. [DOI: 10.1016/j.phrs.2017.02.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 01/08/2017] [Accepted: 02/02/2017] [Indexed: 12/15/2022]
|
26
|
Ma R, Chen J, Liang Y, Lin S, Zhu L, Liang X, Cai X. Sorafenib: A potential therapeutic drug for hepatic fibrosis and its outcomes. Biomed Pharmacother 2017; 88:459-468. [PMID: 28122312 DOI: 10.1016/j.biopha.2017.01.107] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/17/2017] [Accepted: 01/17/2017] [Indexed: 12/16/2022] Open
|
27
|
Correlation of Plasma Vascular Endothelial Growth Factor and Endostatin Levels with Symptomatic Intra- and Extracranial Atherosclerotic Stenosis in a Chinese Han Population. J Stroke Cerebrovasc Dis 2017; 26:1061-1070. [PMID: 28189572 DOI: 10.1016/j.jstrokecerebrovasdis.2016.12.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 12/16/2016] [Accepted: 12/24/2016] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Symptomatic intracranial atherosclerotic stenosis (ICAS) and extracranial atherosclerotic stenosis (ECAS) are different in many aspects. Here, we explored the association between the location or severity of atherosclerotic stenosis and pro- or antiangiogenic factors, specifically vascular endothelial growth factor (VEGF) and endostatin (ES). METHODS We evaluated 198 consecutive patients with acute ischemia stroke: 132 with large-artery atherosclerosis (LAA) and 66 with small-artery occlusion (small-vessel occlusion). The LAA group was subclassified into 102 patients with ICAS and 30 with ECAS. Independent associations of VEGF, ES levels, and VEGF/ES ratio with the location of cerebral stenosis and the severity or short-term prognosis (14th day modified Rankin Scale) of ICAS were evaluated. RESULTS Plasma concentrations of VEGF and ES were lower (P < .05) in ICAS (38.07, 32.76-46.28 pg/mL and 58.95, 55.04-59.77 ng/mL) than those in ECAS (45.00, 34.30-83.34 pg/mL and 140.74, 85.63-231.21 ng/mL). Logistic regression analysis showed that VEGF concentrations and dyslipidemia were independently associated with ICAS, with odds ratios of .987 [95% CI = (.976, .998)] and .265 [95% CI = (.103, .792)], respectively. Moreover, plasmatic VEGF levels increased gradually along with the severity of ICAS (P = .003), and lower levels of ES (P = .040) or a higher VEGF/ES ratio (P = .048) were related to unfavorable short-term prognosis of ICAS. CONCLUSION Lower VEGF levels are associated with the presence of symptomatic ICAS, but not with ECAS. Furthermore, the severity of ICAS is positively correlated with the levels of VEGF, and lower ES levels or a predominance of VEGF over ES are predictors of poor short-term prognosis of ICAS.
Collapse
|
28
|
Wojtukiewicz MZ, Hempel D, Sierko E, Tucker SC, Honn KV. Thrombin-unique coagulation system protein with multifaceted impacts on cancer and metastasis. Cancer Metastasis Rev 2017; 35:213-33. [PMID: 27189210 DOI: 10.1007/s10555-016-9626-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The association between blood coagulation and cancer development is well recognized. Thrombin, the pleiotropic enzyme best known for its contribution to fibrin formation and platelet aggregation during vascular hemostasis, may also trigger cellular events through protease-activated receptors, PAR-1 and PAR-4, leading to cancer progression. Our pioneering findings provided evidence that thrombin contributes to cancer metastasis by increasing adhesive potential of malignant cells. However, there is evidence that thrombin regulates every step of cancer dissemination: (1) cancer cell invasion, detachment from primary tumor, migration; (2) entering the blood vessel; (3) surviving in vasculature; (4) extravasation; (5) implantation in host organs. Recent studies have provided new molecular data about thrombin generation in cancer patients and the mechanisms by which thrombin contributes to transendothelial migration, platelet/tumor cell interactions, angiogenesis, and other processes. Though a great deal is known regarding the role of thrombin in cancer dissemination, there are new data for multiple thrombin-mediated events that justify devoting focus to this topic with a comprehensive approach.
Collapse
Affiliation(s)
- Marek Z Wojtukiewicz
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa St., 15-025, Bialystok, Poland. .,Department of Clinical Oncology, Comprehensive Cancer Center in Bialystok, Bialystok, Poland.
| | - Dominika Hempel
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa St., 15-025, Bialystok, Poland.,Department of Radiotherapy, Comprehensive Cancer Center in Bialystok, Bialystok, Poland
| | - Ewa Sierko
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa St., 15-025, Bialystok, Poland.,Department of Radiotherapy, Comprehensive Cancer Center in Bialystok, Bialystok, Poland
| | - Stephanie C Tucker
- Bioactive Lipids Research Program, Department of Pathology-School of Medicine, Wayne State University, Detroit, MI, USA
| | - Kenneth V Honn
- Bioactive Lipids Research Program, Department of Pathology-School of Medicine, Wayne State University, Detroit, MI, USA.,Department of Chemistry, Wayne State University, Detroit, MI, USA.,Department of Oncology, Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
29
|
Desmedt S, Desmedt V, Delanghe JR, Speeckaert R, Speeckaert MM. The intriguing role of soluble urokinase receptor in inflammatory diseases. Crit Rev Clin Lab Sci 2017; 54:117-133. [DOI: 10.1080/10408363.2016.1269310] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
| | | | - J. R. Delanghe
- Department of Clinical Chemistry, Ghent University Hospital, Gent, Belgium
| | - R. Speeckaert
- Department of Clinical Chemistry, Ghent University Hospital, Gent, Belgium
| | | |
Collapse
|
30
|
Extensive Characterization and Comparison of Endothelial Cells Derived from Dermis and Adipose Tissue: Potential Use in Tissue Engineering. PLoS One 2016; 11:e0167056. [PMID: 27902740 PMCID: PMC5130240 DOI: 10.1371/journal.pone.0167056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 11/08/2016] [Indexed: 02/04/2023] Open
Abstract
Tissue-engineered constructs need to become quickly vascularized in order to ensure graft take. One way of achieving this is to incorporate endothelial cells (EC) into the construct. The adipose tissue stromal vascular fraction (adipose-SVF) might provide an alternative source for endothelial cells as adipose tissue can easily be obtained by liposuction. Since adipose-EC are now gaining more interest in tissue engineering, we aimed to extensively characterize endothelial cells from adipose tissue (adipose-EC) and compare them with endothelial cells from dermis (dermal-EC). The amount of endothelial cells before purification varied between 4–16% of the total stromal population. After MACS selection for CD31 positive cells, a >99% pure population of endothelial cells was obtained within two weeks of culture. Adipose- and dermal-EC expressed the typical endothelial markers PECAM-1, ICAM-1, Endoglin, VE-cadherin and VEGFR2 to a similar extent, with 80–99% of the cell population staining positive. With the exception of CXCR4, which was expressed on 29% of endothelial cells, all other chemokine receptors (CXCR1, 2, 3, and CCR2) were expressed on less than 5% of the endothelial cell populations. Adipose-EC proliferated similar to dermal-EC, but responded less to the mitogens bFGF and VEGF. A similar migration rate was found for both adipose-EC and dermal-EC in response to bFGF. Sprouting of adipose-EC and dermal-EC was induced by bFGF and VEGF in a 3D fibrin matrix. After stimulation of adipose-EC and dermal-EC with TNF-α an increased secretion was seen for PDGF-BB, but not uPA, PAI-1 or Angiopoietin-2. Furthermore, secretion of cytokines and chemokines (IL-6, CCL2, CCL5, CCL20, CXCL1, CXCL8 and CXCL10) was also upregulated by both adipose- and dermal-EC. The similar characteristics of adipose-EC compared to their dermal-derived counterpart make them particularly interesting for skin tissue engineering. In conclusion, we show here that adipose tissue provides for an excellent source of endothelial cells for tissue engineering purposes, since they are readily available, and easily isolated and amplified.
Collapse
|
31
|
Identification of HIF-2α-regulated genes that play a role in human microvascular endothelial sprouting during prolonged hypoxia in vitro. Angiogenesis 2016; 20:39-54. [PMID: 27699500 PMCID: PMC5306362 DOI: 10.1007/s10456-016-9527-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/28/2016] [Indexed: 12/24/2022]
Abstract
During prolonged hypoxic conditions, endothelial cells change their gene expression to adjust to the low oxygen environment. This process is mainly regulated by the hypoxia-inducible factors, HIF-1α and HIF-2α. Although endothelial cells do not form sprouts during prolonged hypoxic culturing, silencing of HIF-2α partially restores sprout formation. The present study identifies novel HIF-2α-target genes that may regulate endothelial sprouting during prolonged hypoxia. The gene expression profile of primary human microvascular endothelial cells (hMVECs) that were cultured at 20 % oxygen was compared to hMVECs that were cultured at 1 % oxygen for 14 days by using genome-wide RNA-sequencing. The differentially regulated genes in hypoxia were compared to the genes that were differentially regulated upon silencing of HIF-2α in hypoxia. Surprisingly, KEGG pathway analysis showed that metabolic pathways were enriched within genes upregulated in response to hypoxia and enriched within genes downregulated upon HIF-2α silencing. Moreover, 51 HIF-2α-regulated genes were screened for their role in endothelial sprouting in hypoxia, of which four genes ARRDC3, MME, PPARG and RALGPS2 directly influenced endothelial sprouting during prolonged hypoxic culturing. The manipulation of specific downstream targets of HIF-2α provides a new, but to be further evaluated, perspective for restoring reduced neovascularization in several pathological conditions, such as diabetic ulcers or other chronic wounds, for improvement of vascularization of implanted tissue-engineered scaffolds.
Collapse
|
32
|
HIF-2α Expression Regulates Sprout Formation into 3D Fibrin Matrices in Prolonged Hypoxia in Human Microvascular Endothelial Cells. PLoS One 2016; 11:e0160700. [PMID: 27490118 PMCID: PMC4973926 DOI: 10.1371/journal.pone.0160700] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 07/22/2016] [Indexed: 11/20/2022] Open
Abstract
Background During short-term hypoxia, Hypoxia Inducible Factors (particular their subunits HIF-1α and HIF-2α) regulate the expression of many genes including the potent angiogenesis stimulator VEGF. However, in some pathological conditions chronic hypoxia occurs and is accompanied by reduced angiogenesis. Objectives We investigated the effect of prolonged hypoxia on the proliferation and sprouting ability of human microvascular endothelial cells and the involvement of the HIFs and Dll4/Notch signaling. Methods and Results Human microvascular endothelial cells (hMVECs), cultured at 20% oxygen for 14 days and seeded on top of 3D fibrin matrices, formed sprouts when stimulated with VEGF-A/TNFα. In contrast, hMVECs precultured at 1% oxygen for 14 days were viable and proliferative, but did not form sprouts into fibrin upon VEGF-A/TNFα stimulation at 1% oxygen. Silencing of HIF-2α with si-RNA partially restored the inhibition of endothelial sprouting, whereas HIF-1α or HIF-3α by si-RNA had no effect. No involvement of Dll4/Notch pathway in the inhibitory effect on endothelial sprouting by prolonged hypoxia was found. In addition, hypoxia decreased the production of urokinase-type plasminogen activator (uPA), needed for migration and invasion, without a significant effect on its inhibitor PAI-1. This was independent of HIF-2α, as si-HIF-2α did not counteract uPA reduction. Conclusion Prolonged culturing of hMVECs at 1% oxygen inhibited endothelial sprouting into fibrin. Two independent mechanisms contribute. Silencing of HIF-2α with si-RNA partially restored the inhibition of endothelial sprouting pointing to a HIF-2α-dependent mechanism. In addition, reduction of uPA contributed to reduced endothelial tube formation in a fibrin matrix during prolonged hypoxia.
Collapse
|
33
|
Belair DG, Schwartz MP, Knudsen T, Murphy WL. Human iPSC-derived endothelial cell sprouting assay in synthetic hydrogel arrays. Acta Biomater 2016; 39:12-24. [PMID: 27181878 PMCID: PMC5228278 DOI: 10.1016/j.actbio.2016.05.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/10/2016] [Accepted: 05/11/2016] [Indexed: 01/09/2023]
Abstract
UNLABELLED Activation of vascular endothelial cells (ECs) by growth factors initiates a cascade of events during angiogenesis in vivo consisting of EC tip cell selection, sprout formation, EC stalk cell proliferation, and ultimately vascular stabilization by support cells. Although EC functional assays can recapitulate one or more aspects of angiogenesis in vitro, they are often limited by undefined substrates and lack of dependence on key angiogenic signaling axes. Here, we designed and characterized a chemically-defined model of endothelial sprouting behavior in vitro using human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs). We rapidly encapsulated iPSC-ECs at high density in poly(ethylene glycol) (PEG) hydrogel spheres using thiol-ene chemistry and subsequently encapsulated cell-dense hydrogel spheres in a cell-free hydrogel layer. The hydrogel sprouting array supported pro-angiogenic phenotype of iPSC-ECs and supported growth factor-dependent proliferation and sprouting behavior. iPSC-ECs in the sprouting model responded appropriately to several reference pharmacological angiogenesis inhibitors of vascular endothelial growth factor, NF-κB, matrix metalloproteinase-2/9, protein kinase activity, and β-tubulin, which confirms their functional role in endothelial sprouting. A blinded screen of 38 putative vascular disrupting compounds from the US Environmental Protection Agency's ToxCast library identified six compounds that inhibited iPSC-EC sprouting and five compounds that were overtly cytotoxic to iPSC-ECs at a single concentration. The chemically-defined iPSC-EC sprouting model (iSM) is thus amenable to enhanced-throughput screening of small molecular libraries for effects on angiogenic sprouting and iPSC-EC toxicity assessment. STATEMENT OF SIGNIFICANCE Angiogenesis assays that are commonly used for drug screening and toxicity assessment applications typically utilize natural substrates like Matrigel(TM) that are difficult to spatially pattern, costly, ill-defined, and may exhibit lot-to-lot variability. Herein, we describe a novel angiogenic sprouting assay using chemically-defined, bioinert poly(ethylene glycol) hydrogels functionalized with biomimetic peptides to promote cell attachment and degradation in a reproducible format that may mitigate the need for natural substrates. The quantitative assay of angiogenic sprouting here enables precise control over the initial conditions and can be formulated into arrays for screening. The sprouting assay here was dependent on key angiogenic signaling axes in a screen of angiogenesis inhibitors and a blinded screen of putative vascular disrupting compounds from the US-EPA.
Collapse
Affiliation(s)
- David G Belair
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael P Schwartz
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Thomas Knudsen
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA; Material Science Program, University of Wisconsin-Madison, Madison, WI, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
34
|
Govindaraj S, Gordon R, Genden EM. Effect of Fibrin Matrix and Vascular Endothelial Growth Factor on Reepithelialization of Orthotopic Murine Tracheal Transplants. Ann Otol Rhinol Laryngol 2016; 113:797-804. [PMID: 15535142 DOI: 10.1177/000348940411301005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To determine whether the administration of vascular endothelial growth factor (VEGF) alone and in combination with fibrin matrix accelerates murine tracheal allograft reepithelialization, we randomly assigned 40 age-matched mice to 5 experimental groups. BALB/c tracheal grafts were transplanted orthotopically into allogeneic C57BL/6 recipients. The recipients were immunosuppressed with cyclosporine (25 mg/kg per day) and treated with a single topical dose of fibrin matrix, a single topical dose of VEGF, or a single topical dose of a combination of VEGF and fibrin matrix. Thirty-five and 50 days after transplantation, a mixed lymphocyte reaction was performed to assess adequate immunosuppression and the grafts were assessed for rejection, rate and quality of allograft reepithelialization, and cartilage viability. The administration of a combination of fibrin matrix and VEGF to tracheal allografts demonstrated an increased rate of reepithelialization and increased density (37% ± 2.9%) of morphologically normal ciliated pseudostratified epithelium when compared with an immunosuppressed control group (29.3% ± 9.1%) 35 days after transplantation. The treated allografts demonstrated no significant change in cartilage viability or rejection. We conclude that the administration of fibrin matrix and VEGF to immunosuppressed tracheal allografts improves the rate and density of tracheal allograft reepithelialization. Carrier-bound growth factors may represent a novel approach to accelerating tracheal allograft reepithelialization and decreasing the need for prolonged immunosuppression following tracheal transplantation.
Collapse
Affiliation(s)
- Satish Govindaraj
- Department of Otolaryngology-Head and Neck Surgery, The Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | |
Collapse
|
35
|
Uzunalli G, Mammadov R, Yesildal F, Alhan D, Ozturk S, Ozgurtas T, Guler MO, Tekinay AB. Angiogenic Heparin-Mimetic Peptide Nanofiber Gel Improves Regenerative Healing of Acute Wounds. ACS Biomater Sci Eng 2016; 3:1296-1303. [DOI: 10.1021/acsbiomaterials.6b00165] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Gozde Uzunalli
- Institute
of Materials Science and Nanotechnology, National Nanotechnology Research
Center (UNAM), Bilkent University, Ankara, Turkey 06800
| | - Rashad Mammadov
- Institute
of Materials Science and Nanotechnology, National Nanotechnology Research
Center (UNAM), Bilkent University, Ankara, Turkey 06800
| | - Fatih Yesildal
- Department
of Medical Biochemistry, Diyarbakir Military Hospital, Diyarbakir, Turkey
| | - Dogan Alhan
- Gulhane Military Medical Academy, Ankara, Turkey
| | | | | | - Mustafa O. Guler
- Institute
of Materials Science and Nanotechnology, National Nanotechnology Research
Center (UNAM), Bilkent University, Ankara, Turkey 06800
| | - Ayse B. Tekinay
- Institute
of Materials Science and Nanotechnology, National Nanotechnology Research
Center (UNAM), Bilkent University, Ankara, Turkey 06800
| |
Collapse
|
36
|
Chen YW, Yin S, Lai YJJ, Johnson MD, Lin CY. Plasminogen-Dependent Matriptase Activation Accelerates Plasmin Generation by Differentiating Primary Human Keratinocytes. J Invest Dermatol 2016; 136:1210-1218. [DOI: 10.1016/j.jid.2016.01.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 01/19/2016] [Accepted: 01/25/2016] [Indexed: 11/25/2022]
|
37
|
Stepanova V, Jayaraman PS, Zaitsev SV, Lebedeva T, Bdeir K, Kershaw R, Holman KR, Parfyonova YV, Semina EV, Beloglazova IB, Tkachuk VA, Cines DB. Urokinase-type Plasminogen Activator (uPA) Promotes Angiogenesis by Attenuating Proline-rich Homeodomain Protein (PRH) Transcription Factor Activity and De-repressing Vascular Endothelial Growth Factor (VEGF) Receptor Expression. J Biol Chem 2016; 291:15029-45. [PMID: 27151212 DOI: 10.1074/jbc.m115.678490] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Indexed: 01/09/2023] Open
Abstract
Urokinase-type plasminogen activator (uPA) regulates angiogenesis and vascular permeability through proteolytic degradation of extracellular matrix and intracellular signaling initiated upon its binding to uPAR/CD87 and other cell surface receptors. Here, we describe an additional mechanism by which uPA regulates angiogenesis. Ex vivo VEGF-induced vascular sprouting from Matrigel-embedded aortic rings isolated from uPA knock-out (uPA(-/-)) mice was impaired compared with vessels emanating from wild-type mice. Endothelial cells isolated from uPA(-/-) mice show less proliferation and migration in response to VEGF than their wild type counterparts or uPA(-/-) endothelial cells in which expression of wild type uPA had been restored. We reported previously that uPA is transported from cell surface receptors to nuclei through a mechanism that requires its kringle domain. Intranuclear uPA modulates gene transcription by binding to a subset of transcription factors. Here we report that wild type single-chain uPA, but not uPA variants incapable of nuclear transport, increases the expression of cell surface VEGF receptor 1 (VEGFR1) and VEGF receptor 2 (VEGFR2) by translocating to the nuclei of ECs. Intranuclear single-chain uPA binds directly to and interferes with the function of the transcription factor hematopoietically expressed homeodomain protein or proline-rich homeodomain protein (HHEX/PRH), which thereby lose their physiologic capacity to repress the activity of vehgr1 and vegfr2 gene promoters. These studies identify uPA-dependent de-repression of vegfr1 and vegfr2 gene transcription through binding to HHEX/PRH as a novel mechanism by which uPA mediates the pro-angiogenic effects of VEGF and identifies a potential new target for control of pathologic angiogenesis.
Collapse
Affiliation(s)
| | - Padma-Sheela Jayaraman
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B152TT, United Kingdom
| | - Sergei V Zaitsev
- Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | | | - Khalil Bdeir
- From the Departments of Pathology and Laboratory Medicine and
| | - Rachael Kershaw
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B152TT, United Kingdom
| | - Kelci R Holman
- College of Arts and Sciences, Drexel University, Philadelphia, Pennsylvania 19104
| | - Yelena V Parfyonova
- Russian Cardiology Research Center, Moscow 121552, Russia, School (Faculty) of Fundamental Medicine, Lomonosov Moscow State University, Moscow, 117192, Russia, and
| | - Ekaterina V Semina
- Russian Cardiology Research Center, Moscow 121552, Russia, School (Faculty) of Fundamental Medicine, Lomonosov Moscow State University, Moscow, 117192, Russia, and
| | | | - Vsevolod A Tkachuk
- Russian Cardiology Research Center, Moscow 121552, Russia, School (Faculty) of Fundamental Medicine, Lomonosov Moscow State University, Moscow, 117192, Russia, and
| | - Douglas B Cines
- From the Departments of Pathology and Laboratory Medicine and
| |
Collapse
|
38
|
Methods to study differences in cell mobility during skin wound healing in vitro. J Biomech 2016; 49:1381-1387. [DOI: 10.1016/j.jbiomech.2016.01.040] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 01/19/2016] [Accepted: 01/28/2016] [Indexed: 01/19/2023]
|
39
|
Tasev D, Konijnenberg LSF, Amado-Azevedo J, van Wijhe MH, Koolwijk P, van Hinsbergh VWM. CD34 expression modulates tube-forming capacity and barrier properties of peripheral blood-derived endothelial colony-forming cells (ECFCs). Angiogenesis 2016; 19:325-38. [PMID: 27043316 PMCID: PMC4930476 DOI: 10.1007/s10456-016-9506-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 03/19/2016] [Indexed: 11/30/2022]
Abstract
Endothelial colony-forming cells (ECFC) are grown from circulating CD34+ progenitors present in adult peripheral blood, but during in vitro expansion part of the cells lose CD34. To evaluate whether the regulation of CD34 characterizes the angiogenic phenotypical features of PB-ECFCs, we investigated the properties of CD34+ and CD34− ECFCs with respect to their ability to form capillary-like tubes in 3D fibrin matrices, tip-cell gene expression, and barrier integrity. Selection of CD34+ and CD34− ECFCs from subcultured ECFCs was accomplished by magnetic sorting (FACS: CD34+: 95 % pos; CD34−: 99 % neg). Both fractions proliferated at same rate, while CD34+ ECFCs exhibited higher tube-forming capacity and tip-cell gene expression than CD34− cells. However, during cell culture CD34− cells re-expressed CD34. Cell-seeding density, cell–cell contact formation, and serum supplements modulated CD34 expression. CD34 expression in ECFCs was strongly suppressed by newborn calf serum. Stimulation with FGF-2, VEGF, or HGF prepared in medium supplemented with 3 % albumin did not change CD34 mRNA or surface expression. Silencing of CD34 with siRNA resulted in strengthening of cell–cell contacts and increased barrier function of ECFC monolayers as measured by ECIS. Furthermore, CD34 siRNA reduced tube formation by ECFC, but did not affect tip-cell gene expression. These findings demonstrate that CD34+ and CD34− cells are different phenotypes of similar cells and that CD34 (1) can be regulated in ECFC; (2) is positively involved in capillary-like sprout formation; (3) is associated but not causally related to tip-cell gene expression; and (4) can affect endothelial barrier function.
Collapse
Affiliation(s)
- Dimitar Tasev
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center Amsterdam, De Boelelaan 1118, 1081 HV, Amsterdam, The Netherlands.,A-Skin Nederland BV, De Boelelaan 1117, 1007 MB, Amsterdam, The Netherlands
| | - Lara S F Konijnenberg
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center Amsterdam, De Boelelaan 1118, 1081 HV, Amsterdam, The Netherlands
| | - Joana Amado-Azevedo
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center Amsterdam, De Boelelaan 1118, 1081 HV, Amsterdam, The Netherlands
| | - Michiel H van Wijhe
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center Amsterdam, De Boelelaan 1118, 1081 HV, Amsterdam, The Netherlands
| | - Pieter Koolwijk
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center Amsterdam, De Boelelaan 1118, 1081 HV, Amsterdam, The Netherlands
| | - Victor W M van Hinsbergh
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center Amsterdam, De Boelelaan 1118, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
40
|
Bookholt FD, Monsuur HN, Gibbs S, Vermolen FJ. Mathematical modelling of angiogenesis using continuous cell-based models. Biomech Model Mechanobiol 2016; 15:1577-1600. [PMID: 27037954 PMCID: PMC5106520 DOI: 10.1007/s10237-016-0784-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 03/15/2016] [Indexed: 11/25/2022]
Abstract
In this work, we develop a mathematical formalism based on a 3D in vitro model that is used to simulate the early stages of angiogenesis. The model treats cells as individual entities that are migrating as a result of chemotaxis and durotaxis. The phenotypes used here are endothelial cells that can be distinguished into stalk and tip (leading) cells. The model takes into account the dynamic interaction and interchange between both phenotypes. Next to the cells, the model takes into account several proteins such as vascular endothelial growth factor, delta-like ligand 4, urokinase plasminogen activator and matrix metalloproteinase, which are computed through the solution of a system of reaction–diffusion equations. The method used in the present study is classified into the hybrid approaches. The present study, implemented in three spatial dimensions, demonstrates the feasibility of the approach that is qualitatively confirmed by experimental results.
Collapse
Affiliation(s)
- F D Bookholt
- Delft Institute of Applied Mathematics, Delft University of Technology, Delft, The Netherlands
| | - H N Monsuur
- Department of Dermatology (VUmc), VU University Medical Center, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - S Gibbs
- Department of Dermatology (VUmc), VU University Medical Center, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - F J Vermolen
- Delft Institute of Applied Mathematics, Delft University of Technology, Delft, The Netherlands.
| |
Collapse
|
41
|
Huber B, Czaja AM, Kluger PJ. Influence of epidermal growth factor (EGF) and hydrocortisone on the co-culture of mature adipocytes and endothelial cells for vascularized adipose tissue engineering. Cell Biol Int 2016; 40:569-78. [DOI: 10.1002/cbin.10595] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 02/14/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Birgit Huber
- Institute for Interfacial Process Engineering and Plasma Technology; University of Stuttgart; Nobelstraße 12 Stuttgart 70569 Germany
| | - Alina Maria Czaja
- Esslingen University of Applied Sciences; Kanalstraße 33 Esslingen 73728 Germany
| | - Petra Juliane Kluger
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB; Nobelstraße 12 Stuttgart 70569 Germany
- Reutlingen University; Alteburgstr. 150 Reutlingen 72762 Germany
| |
Collapse
|
42
|
Soares TS, Oliveira F, Torquato RJ, Sasaki SD, Araujo MS, Paschoalin T, Tanaka AS. BmTI-A, a Kunitz type inhibitor from Rhipicephalus microplus able to interfere in vessel formation. Vet Parasitol 2016; 219:44-52. [DOI: 10.1016/j.vetpar.2016.01.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 12/23/2015] [Accepted: 01/27/2016] [Indexed: 11/30/2022]
|
43
|
Yıldırım C, Favre J, Weijers EM, Fontijn RD, van Wijhe MH, van Vliet SJ, Boon RA, Koolwijk P, van der Pouw Kraan TCTM, Horrevoets AJG. IFN-β affects the angiogenic potential of circulating angiogenic cells by activating calpain 1. Am J Physiol Heart Circ Physiol 2015; 309:H1667-78. [DOI: 10.1152/ajpheart.00810.2014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 09/01/2015] [Indexed: 01/15/2023]
Abstract
Circulating angiogenic cells (CACs) are monocyte-derived cells with endothelial characteristics, which contribute to both angiogenesis and arteriogenesis in a paracrine way. Interferon-β (IFN-β) is known to inhibit these divergent processes in animals and patients. We hypothesized that IFN-β might act by affecting the differentiation and function of CACs. CACs were cultured from peripheral blood mononuclear cells and phenotypically characterized by surface expression of monocytic and endothelial markers. IFN-β significantly reduced the number of CACs by 18–64%. Apoptosis was not induced by IFN-β, neither in mononuclear cells during differentiation, nor after maturation to CACs. Rather, IFN-β impaired adhesion to, and spreading on, fibronectin, which was dependent on α5β1 (VLA-5)-integrin. IFN-β affected the function of VLA-5 in mature CACs, leading to rounding and detachment of cells, by induction of calpain 1 activity. Cell rounding and detachment was completely reversed by inhibition of calpain 1 activity in mature CACs. During in vitro capillary formation, CAC addition and calpain 1 inhibition enhanced sprouting of endothelial cells to a comparable extent, but were not sufficient to rescue tube formation in the presence of IFN-β. We show that the IFN-β-induced reduction of the numbers of in vitro differentiated CACs is based on activation of calpain 1, resulting in an attenuated adhesion to extracellular matrix proteins via VLA-5. In vivo, this could lead to inhibition of vessel formation due to reduction of the locally recruited CAC numbers and their paracrine angiogenic factors.
Collapse
Affiliation(s)
- Cansu Yıldırım
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Julie Favre
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Ester M. Weijers
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands; and
| | - Ruud D. Fontijn
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Michiel H. van Wijhe
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands; and
| | - Sandra J. van Vliet
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Reinier A. Boon
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Pieter Koolwijk
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands; and
| | | | - Anton J. G. Horrevoets
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
44
|
Understanding the effects of mature adipocytes and endothelial cells on fatty acid metabolism and vascular tone in physiological fatty tissue for vascularized adipose tissue engineering. Cell Tissue Res 2015; 362:269-79. [PMID: 26340984 DOI: 10.1007/s00441-015-2274-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 07/31/2015] [Indexed: 01/27/2023]
Abstract
Engineering of large vascularized adipose tissue constructs is still a challenge for the treatment of extensive high-graded burns or the replacement of tissue after tumor removal. Communication between mature adipocytes and endothelial cells is important for homeostasis and the maintenance of adipose tissue mass but, to date, is mainly neglected in tissue engineering strategies. Thus, new co-culture strategies are needed to integrate adipocytes and endothelial cells successfully into a functional construct. This review focuses on the cross-talk of mature adipocytes and endothelial cells and considers their influence on fatty acid metabolism and vascular tone. In addition, the properties and challenges with regard to these two cell types for vascularized tissue engineering are highlighted.
Collapse
|
45
|
Tasev D, van Wijhe MH, Weijers EM, van Hinsbergh VWM, Koolwijk P. Long-Term Expansion in Platelet Lysate Increases Growth of Peripheral Blood-Derived Endothelial-Colony Forming Cells and Their Growth Factor-Induced Sprouting Capacity. PLoS One 2015; 10:e0129935. [PMID: 26076450 PMCID: PMC4468160 DOI: 10.1371/journal.pone.0129935] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 05/14/2015] [Indexed: 12/27/2022] Open
Abstract
Introduction Efficient implementation of peripheral blood-derived endothelial-colony cells (PB-ECFCs) as a therapeutical tool requires isolation and generation of a sufficient number of cells in ex vivo conditions devoid of animal-derived products. At present, little is known how the isolation and expansion procedure in xenogeneic-free conditions affects the therapeutical capacity of PB-ECFCs. Results The findings presented in this study indicate that human platelet lysate (PL) as a serum substitute yields twice more colonies per mL blood compared to the conventional isolation with fetal bovine serum (FBS). Isolated ECFCs displayed a higher proliferative ability in PL supplemented medium than cells in FBS medium during 30 days expansion. The cells at 18 cumulative population doubling levels (CPDL) retained their proliferative capacity, showed higher sprouting ability in fibrin matrices upon stimulation with FGF-2 and VEGF-A than the cells at 6 CPDL, and displayed low β-galactosidase activity. The increased sprouting of PB-ECFCs at 18 CPDL was accompanied by an intrinsic activation of the uPA/uPAR fibrinolytic system. Induced deficiency of uPA (urokinase-type plasminogen activator) or uPAR (uPA receptor) by siRNA technology completely abolished the angiogenic ability of PB-ECFCs in fibrin matrices. During the serial expansion, the gene induction of the markers associated with inflammatory activation such as VCAM-1 and ICAM-1 did not occur or only to limited extent. While further propagation up to 31 CPDL proceeded at a comparable rate, a marked upregulation of inflammatory markers occurred in all donors accompanied by a further increase of uPA/uPAR gene induction. The observed induction of inflammatory genes at later stages of long-term propagation of PB-ECFCs underpins the necessity to determine the right time-point for harvesting of sufficient number of cells with preserved therapeutical potential. Conclusion The presented isolation method and subsequent cell expansion in platelet lysate supplemented culture medium permits suitable large-scale propagation of PB-ECFC. For optimal use of PB-ECFCs in clinical settings, our data suggest that 15–20 CPDL is the most adequate maturation stage.
Collapse
Affiliation(s)
- Dimitar Tasev
- Dept. of Physiology, ICaR-VU, VU University Medical Center, Amsterdam, The Netherlands
| | - Michiel H. van Wijhe
- Dept. of Physiology, ICaR-VU, VU University Medical Center, Amsterdam, The Netherlands
| | - Ester M. Weijers
- Dept. of Physiology, ICaR-VU, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Pieter Koolwijk
- Dept. of Physiology, ICaR-VU, VU University Medical Center, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
46
|
Enocsson H, Sjöwall C, Wetterö J. Soluble urokinase plasminogen activator receptor--a valuable biomarker in systemic lupus erythematosus? Clin Chim Acta 2015; 444:234-41. [PMID: 25704300 DOI: 10.1016/j.cca.2015.02.031] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 02/13/2015] [Accepted: 02/14/2015] [Indexed: 12/29/2022]
Abstract
Systemic lupus erythematosus (SLE) is a potentially severe autoimmune condition with an unpredictable disease course, often with fluctuations in disease activity over time. Long term inflammation and drug-related side-effects may subsequently lead to permanent organ damage, a consequence which is intimately connected to decreased quality of life and mortality. New lupus biomarkers that convey information regarding inflammation and/or organ damage are thus warranted. Today, there is no clinical biomarker that indicates the risk of damage accrual. Herein we highlight the urokinase plasminogen activator receptor (uPAR) and especially its soluble form (suPAR) that besides having biological functions in e.g. proteolysis, cell migration and tissue homeostasis, recently has emerged as a promising biomarker of inflammation and prognosis of several disorders. A strong association between suPAR and organ damage in SLE was recently demonstrated, and preliminary data (presented in this review) suggests the possibility of a predictive value of suPAR blood levels. The involvement of suPAR in the pathogenesis of SLE remains obscure, but its effects in leukocyte recruitment, phagocytic uptake of dying cells (efferocytosis) and complement regulation suggests that the central parts of the SLE pathogenesis could be regulated by suPAR, and vice versa.
Collapse
Affiliation(s)
- Helena Enocsson
- Rheumatology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
| | - Christopher Sjöwall
- Rheumatology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
| | - Jonas Wetterö
- Rheumatology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
| |
Collapse
|
47
|
Noort AR, van Zoest KPM, Weijers EM, Koolwijk P, Maracle CX, Novack DV, Siemerink MJ, Schlingemann RO, Tak PP, Tas SW. NF-κB-inducing kinase is a key regulator of inflammation-induced and tumour-associated angiogenesis. J Pathol 2014; 234:375-85. [PMID: 25043127 PMCID: PMC4194146 DOI: 10.1002/path.4403] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 06/23/2014] [Accepted: 07/02/2014] [Indexed: 01/08/2023]
Abstract
Angiogenesis is essential during development and in pathological conditions such as chronic inflammation and cancer progression. Inhibition of angiogenesis by targeting vascular endothelial growth factor (VEGF) blocks disease progression, but most patients eventually develop resistance which may result from compensatory signalling pathways. In endothelial cells (ECs), expression of the pro-angiogenic chemokine CXCL12 is regulated by non-canonical nuclear factor (NF)-κB signalling. Here, we report that NF-κB-inducing kinase (NIK) and subsequent non-canonical NF-κB signalling regulate both inflammation-induced and tumour-associated angiogenesis. NIK is highly expressed in endothelial cells (ECs) in tumour tissues and inflamed rheumatoid arthritis synovial tissue. Furthermore, non-canonical NF-κB signalling in human microvascular ECs significantly enhanced vascular tube formation, which was completely blocked by siRNA targeting NIK. Interestingly, Nik(-/-) mice exhibited normal angiogenesis during development and unaltered TNFα- or VEGF-induced angiogenic responses, whereas angiogenesis induced by non-canonical NF-κB stimuli was significantly reduced. In addition, angiogenesis in experimental arthritis and a murine tumour model was severely impaired in these mice. These studies provide evidence for a role of non-canonical NF-κB signalling in pathological angiogenesis, and identify NIK as a potential therapeutic target in chronic inflammatory diseases and tumour neoangiogenesis.
Collapse
Affiliation(s)
- Ae R Noort
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of AmsterdamThe Netherlands
- Department of Experimental Immunology, Academic Medical Center/University of AmsterdamThe Netherlands
| | - Katinka PM van Zoest
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of AmsterdamThe Netherlands
- Department of Experimental Immunology, Academic Medical Center/University of AmsterdamThe Netherlands
| | - Ester M Weijers
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU Medical CenterAmsterdam, The Netherlands
| | - Pieter Koolwijk
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU Medical CenterAmsterdam, The Netherlands
| | - Chrissta X Maracle
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of AmsterdamThe Netherlands
- Department of Experimental Immunology, Academic Medical Center/University of AmsterdamThe Netherlands
| | - Deborah V Novack
- Division of Bone and Mineral Diseases, Departments of Medicine and Pathology, Washington University School of MedicineSt Louis, Missouri, USA
| | - Martin J Siemerink
- Ocular Angiogenesis Group, Department of Ophthalmology and Department of Cell Biology and Histology, Academic Medical Center/University of AmsterdamThe Netherlands
| | - Reinier O Schlingemann
- Ocular Angiogenesis Group, Department of Ophthalmology and Department of Cell Biology and Histology, Academic Medical Center/University of AmsterdamThe Netherlands
| | - Paul P Tak
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of AmsterdamThe Netherlands
- Department of Medicine, University of CambridgeCambridge, UK
| | - Sander W Tas
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of AmsterdamThe Netherlands
- Department of Experimental Immunology, Academic Medical Center/University of AmsterdamThe Netherlands
| |
Collapse
|
48
|
Fontijn RD, Favre J, Naaijkens BA, Meinster E, Paauw NJ, Ragghoe SL, Nauta TD, van den Broek MA, Weijers EM, Niessen HW, Koolwijk P, Horrevoets AJ. Adipose tissue-derived stromal cells acquire endothelial-like features upon reprogramming with SOX18. Stem Cell Res 2014; 13:367-78. [PMID: 25290189 DOI: 10.1016/j.scr.2014.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 07/29/2014] [Accepted: 09/17/2014] [Indexed: 01/08/2023] Open
Abstract
Adipose tissue-derived stromal cells (ASC) form a rich source of autologous cells for use in regenerative medicine. In vitro induction of an endothelial phenotype may improve performance of ASCs in cardiovascular repair. Here, we report on an in vitro strategy using direct reprogramming of ASCs by means of ectopic expression of the endothelial-specific transcription factor SRY (sex determining region Y)-box18 (SOX18). SOX18 induces ASCs to express a set of genes involved in vascular patterning: MMP7, KDR, EFNB2, SEMA3G and CXCR4. Accordingly, SOX18 transduced ASCs reorganize under conditions of shear stress, display VEGF-induced chemotaxis and form tubular structures in 3D matrices in an MMP7-dependent manner. These in vitro findings provide insight into molecular and cellular processes downstream of SOX18 and show that reprogramming using SOX18 is sufficient to induce several endothelial-like features in ASCs.
Collapse
Affiliation(s)
- R D Fontijn
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands.
| | - J Favre
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands.
| | - B A Naaijkens
- Department of Pathology and Cardiac Surgery, VU University Medical Center, de Boelelaan 117, 1081 HV Amsterdam, The Netherlands.
| | - E Meinster
- Department of Pathology and Cardiac Surgery, VU University Medical Center, de Boelelaan 117, 1081 HV Amsterdam, The Netherlands.
| | - N J Paauw
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands.
| | - S L Ragghoe
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - T D Nauta
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands.
| | - M A van den Broek
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands.
| | - E M Weijers
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands.
| | - H W Niessen
- Department of Pathology and Cardiac Surgery, VU University Medical Center, de Boelelaan 117, 1081 HV Amsterdam, The Netherlands.
| | - P Koolwijk
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands.
| | - A J Horrevoets
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands.
| |
Collapse
|
49
|
Cheung EYL, Weijers EM, Tuk B, Scheffer R, Leebeek FW, van Neck JW, Koolwijk P, de Maat MPM. Specific effects of fibrinogen and the γA and γ'-chain fibrinogen variants on angiogenesis and wound healing. Tissue Eng Part A 2014; 21:106-14. [PMID: 24974891 DOI: 10.1089/ten.tea.2014.0020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In a newly formed wound, the natural fibrin network provides the first temporary matrix for tissue repair. Topical application of fibrin to a new wound may improve wound healing. A matrix of the common natural γ' fibrin variant may further improve wound healing because it is expected to have a different architecture and this will influence angiogenesis, because it possesses increased thrombin and factor XIII binding and decreased platelet binding, when compared with the common γA fibrin matrix. Our objective was to determine the effect of fibrinogen and its γA and γ' variants on angiogenesis and wound healing. We used in vitro angiogenesis models and an in vivo rat full-thickness excisional wound healing model. When comparing γA and γ' fibrin in vitro, more tube-like structures were formed on day 7 in γA fibrin than in γ' fibrin (13.83±6.12 AU vs. 6.1±1.46 AU). Wounds treated with fibrin demonstrated improved healing in vivo with more perfusion (47%±3% vs. 26%±4%, p<0.01 in placebo) and higher CD34 density score (2.0±0.4 vs. 2.8±0.1, p<0.01) on day 21 with fibrin matrices when compared with placebo-treated wounds. Increased perfusion was observed in γA fibrin-treated wounds on day 21 (53%±10% vs. 41%±7% for γ' fibrin). The other parameters showed slightly improved (not significant) wound healing with γA fibrin compared with γ' fibrin matrices. In conclusion, the use of fibrin and fibrin variant matrices offers an interesting methodology to stimulate the wound healing process.
Collapse
Affiliation(s)
- Elim Y L Cheung
- 1 Department of Hematology, Erasmus University Medical Center , Rotterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
50
|
van Oers RFM, Rens EG, LaValley DJ, Reinhart-King CA, Merks RMH. Mechanical cell-matrix feedback explains pairwise and collective endothelial cell behavior in vitro. PLoS Comput Biol 2014; 10:e1003774. [PMID: 25121971 PMCID: PMC4133044 DOI: 10.1371/journal.pcbi.1003774] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 06/20/2014] [Indexed: 12/14/2022] Open
Abstract
In vitro cultures of endothelial cells are a widely used model system of the collective behavior of endothelial cells during vasculogenesis and angiogenesis. When seeded in an extracellular matrix, endothelial cells can form blood vessel-like structures, including vascular networks and sprouts. Endothelial morphogenesis depends on a large number of chemical and mechanical factors, including the compliancy of the extracellular matrix, the available growth factors, the adhesion of cells to the extracellular matrix, cell-cell signaling, etc. Although various computational models have been proposed to explain the role of each of these biochemical and biomechanical effects, the understanding of the mechanisms underlying in vitro angiogenesis is still incomplete. Most explanations focus on predicting the whole vascular network or sprout from the underlying cell behavior, and do not check if the same model also correctly captures the intermediate scale: the pairwise cell-cell interactions or single cell responses to ECM mechanics. Here we show, using a hybrid cellular Potts and finite element computational model, that a single set of biologically plausible rules describing (a) the contractile forces that endothelial cells exert on the ECM, (b) the resulting strains in the extracellular matrix, and (c) the cellular response to the strains, suffices for reproducing the behavior of individual endothelial cells and the interactions of endothelial cell pairs in compliant matrices. With the same set of rules, the model also reproduces network formation from scattered cells, and sprouting from endothelial spheroids. Combining the present mechanical model with aspects of previously proposed mechanical and chemical models may lead to a more complete understanding of in vitro angiogenesis.
Collapse
Affiliation(s)
- René F. M. van Oers
- Life Sciences group, Centrum Wiskunde & Informatica, Amsterdam, The Netherlands
- Netherlands Consortium for System Biology - Netherlands Institute for Systems Biology, Amsterdam, The Netherlands
| | - Elisabeth G. Rens
- Life Sciences group, Centrum Wiskunde & Informatica, Amsterdam, The Netherlands
- Netherlands Consortium for System Biology - Netherlands Institute for Systems Biology, Amsterdam, The Netherlands
| | - Danielle J. LaValley
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
| | - Cynthia A. Reinhart-King
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, United States of America
| | - Roeland M. H. Merks
- Life Sciences group, Centrum Wiskunde & Informatica, Amsterdam, The Netherlands
- Netherlands Consortium for System Biology - Netherlands Institute for Systems Biology, Amsterdam, The Netherlands
- Mathematical Institute, Leiden University, Leiden, The Netherlands
| |
Collapse
|