1
|
Renzelmann J, Heene S, Jonczyk R, Krüger J, Alnajjar S, Blume C. Sustainability of shear stress conditioning in endothelial colony-forming cells compared to human aortic endothelial cells to underline suitability for tissue-engineered vascular grafts. Microvasc Res 2025; 157:104746. [PMID: 39278537 DOI: 10.1016/j.mvr.2024.104746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/05/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
The endothelialization of cardiovascular implants is supposed to improve the long-term patency of these implants. In addition, in previous studies, it has been shown, that the conditioning of endothelial cells by dynamic cultivation leads to the expression of an anti-thrombogenic phenotype. For the creation of a tissue-engineered vascular graft (TEVG), these two strategies were combined to achieve optimal hemocompatibility. In a clinical setup, this would require the transfer of the already endothelialized construct from the conditioning bioreactor to the patient. Therefore, the reversibility of the dynamic conditioning of the endothelial cells with arterial-like high shear stress (20 dyn/cm2) was investigated to define the timeframe (tested in a range of up to 24 h) for the perseverance of dynamically induced phenotypical changes. Two types of endothelial cells were compared: endothelial colony-forming cells (ECFCs) and human aortic endothelial cells (HAECs). The results showed that ECFCs respond far more sensitively and rapidly to flow than HAECs. The resulting cell alignment and increased protein expression of KLF-2, Notch-4, Thrombomodulin, Tie2 and eNOS monomer was paralleled by increased eNOS and unaltered KLF-2 mRNA levels even under stopped-flow conditions. VCAM-1 mRNA and protein expression was downregulated under flow and did not recover under stopped flow. From these time kinetic results, we concluded, that the maximum time gap between the TEVG cultivated with autologous ECFCs in future reactor cultivations and the transfer to the potential TEVG recipient should be limited to ∼6 h.
Collapse
Affiliation(s)
- Jannis Renzelmann
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany.
| | - Sebastian Heene
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany.
| | - Rebecca Jonczyk
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany.
| | - Jana Krüger
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany.
| | - Suhayla Alnajjar
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany.
| | - Cornelia Blume
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 5, 30167 Hannover, Germany.
| |
Collapse
|
2
|
Fischer KS, Henn D, Zhao ET, Sivaraj D, Litmanovich B, Hahn WW, Hostler AC, Mojadidi SM, Gonzalez J, Knochel AB, Mora Pinos MG, Holley J, Kussie H, Granoski M, Yasmeh JP, Kneser U, Chen K, Gurtner GC. Elevated Shear Stress Modulates Heterogenous Cellular Subpopulations to Induce Vascular Remodeling. Tissue Eng Part A 2024; 30:752-765. [PMID: 38753711 DOI: 10.1089/ten.tea.2023.0362] [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] [Indexed: 05/18/2024] Open
Abstract
Rationale: Elevated shear stress (ESS) induces vascular remodeling in veins exposed to arterial blood flow, which can lead to arteriovenous (AV) fistula failure. The molecular mechanisms driving remodeling have not been comprehensively examined with a single-cell resolution before. Objective: Using an in vivo animal mode, single-cell RNA sequencing, and histopathology, we precisely manipulate blood flow to comprehensively characterize all cell subpopulations important during vascular remodeling. Methods: AV loops were created in saphenous vessels of rats using a contralateral saphenous vein interposition graft to promote ESS. Saphenous veins with no elevated shear stress (NSS) were anastomosed as controls. Findings: ESS promoted transcriptional homogeneity, and NSS promoted considerable heterogeneity. Specifically, ESS endothelial cells (ECs) showed a more homogeneous transcriptional response promoting angiogenesis and upregulating endothelial-to-mesenchymal transition inhibiting genes (Klf2). NSS ECs upregulated antiproliferation genes such as Cav1, Cst3, and Btg1. In macrophages, ESS promoted a large homogeneous subpopulation, creating a mechanically activated, proinflammatory and thus proangiogenic myeloid phenotype, whereas NSS myeloid cells expressed the anti-inflammatory and antiangiogenetic marker Mrc1. Conclusion: ESS activates unified gene expression profiles to induce adaption of the vessel wall to hemodynamic alterations. Targeted depletion of the identified cellular subpopulations may lead to novel therapies to prevent excessive venous remodeling, intimal hyperplasia, and AV fistula failure.
Collapse
Affiliation(s)
- Katharina S Fischer
- Department of Surgery, University of Arizona, Tucson, Arizona, USA
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
- Department of Plastic and Reconstructive Surgery and Hand surgery, BG Trauma Clinic Ludwigshafen, University of Heidelberg, Heidelberg, Germany
| | - Dominic Henn
- Department of Surgery, University of Arizona, Tucson, Arizona, USA
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
- Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Eric T Zhao
- Department of Chemical Engineering, Stanford University, Stanford, California, USA
| | - Dharshan Sivaraj
- Department of Surgery, University of Arizona, Tucson, Arizona, USA
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
| | - Ben Litmanovich
- Department of Surgery, University of Arizona, Tucson, Arizona, USA
| | - William W Hahn
- Department of Surgery, University of Arizona, Tucson, Arizona, USA
| | - Andrew C Hostler
- Department of Surgery, University of Arizona, Tucson, Arizona, USA
| | | | - Javier Gonzalez
- Department of Surgery, University of Arizona, Tucson, Arizona, USA
| | - Amelia B Knochel
- Department of Surgery, University of Arizona, Tucson, Arizona, USA
| | | | - Jared Holley
- Department of Surgery, University of Arizona, Tucson, Arizona, USA
| | - Hudson Kussie
- Department of Surgery, University of Arizona, Tucson, Arizona, USA
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
| | - Maia Granoski
- Department of Surgery, University of Arizona, Tucson, Arizona, USA
| | | | - Ulrich Kneser
- Department of Plastic and Reconstructive Surgery and Hand surgery, BG Trauma Clinic Ludwigshafen, University of Heidelberg, Heidelberg, Germany
| | - Kellen Chen
- Department of Surgery, University of Arizona, Tucson, Arizona, USA
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
| | - Geoffrey C Gurtner
- Department of Surgery, University of Arizona, Tucson, Arizona, USA
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
| |
Collapse
|
3
|
Wolfe JT, Chen V, Chen Y, Tefft BJ. Identification of a subpopulation of highly adherent endothelial cells for seeding synthetic vascular grafts. J Thorac Cardiovasc Surg 2024:S0022-5223(24)00550-6. [PMID: 38972570 PMCID: PMC11700231 DOI: 10.1016/j.jtcvs.2024.06.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/07/2024] [Accepted: 06/27/2024] [Indexed: 07/09/2024]
Abstract
OBJECTIVE There is an unmet clinical need for alternatives to autologous vessel grafts. Small-diameter (<6 mm) synthetic vascular grafts are not suitable because of unacceptable patency rates. This mainly occurs due to the lack of an endothelial cell (EC) monolayer to prevent platelet activation, thrombosis, and intimal hyperplasia. There are no reliable methods to endothelialize small-diameter grafts because most seeded ECs are lost due to exposure to fluid shear stress after implantation. The goal of this work is to determine if EC loss is a random process or if it is possible to predict which cells are more likely to remain adherent. METHODS In initial studies, we sorted ECs using fluid shear stress and identified a subpopulation of ECs that are more likely to resist detachment. We use RNA sequencing to examine gene expression of adherent ECs compared with the whole population. Using fluorescence activated cell sorting, we sorted ECs based on the expression level of a candidate marker and studied their retention in small-diameter vascular grafts in vitro. RESULTS Transcriptomic analysis revealed that fibronectin leucine rich transmembrane protein 2 (FLRT2), encoding protein FLRT2, is downregulated in the ECs that are more likely to resist detachment. When seeded onto vascular grafts and exposed to shear stress, ECs expressing low levels of FLRT2 exhibit 59.2% ± 7.4% retention compared with 24.5% ± 6.1% retention for the remainder of the EC population. CONCLUSIONS For the first time, we show EC detachment is not an entirely random process. This provides validation for the concept that we can seed small-diameter vascular grafts only with highly adherent ECs to maintain a stable endothelium and improve graft patency rates.
Collapse
Affiliation(s)
- Jayne T Wolfe
- Joint Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, Wis
| | - Vaya Chen
- Versiti Blood Research Institute, Milwaukee, Wis
| | - Yiliang Chen
- Versiti Blood Research Institute, Milwaukee, Wis; Department of Medicine, Medical College of Wisconsin, Milwaukee, Wis; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wis
| | - Brandon J Tefft
- Joint Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, Wis; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wis.
| |
Collapse
|
4
|
Laan SNJ, de Boer S, Dirven RJ, van Moort I, Kuipers TB, Mei H, Bierings R, Eikenboom J. Transcriptional and functional profiling identifies inflammation and endothelial-to-mesenchymal transition as potential drivers for phenotypic heterogeneity within a cohort of endothelial colony forming cells. J Thromb Haemost 2024; 22:2027-2038. [PMID: 38574861 DOI: 10.1016/j.jtha.2024.03.018] [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: 10/19/2023] [Revised: 03/14/2024] [Accepted: 03/24/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Endothelial colony-forming cells (ECFCs) derived from patients can be used to investigate pathogenic mechanisms of vascular diseases like von Willebrand disease. Considerable phenotypic heterogeneity has been observed between ECFC clones derived from healthy donors. This heterogeneity needs to be well understood in order to use ECFCs as endothelial models for disease. OBJECTIVES Therefore, we aimed to determine phenotypic and gene expression differences between control ECFCs. METHODS A total of 34 ECFC clones derived from 16 healthy controls were analyzed. The transcriptome of a selection of ECFC clones (n = 15) was analyzed by bulk RNA sequencing and gene set enrichment analysis. Gene expression was measured in all ECFC clones by quantitative polymerase chain reaction. Phenotypic profiling was performed and migration speed of the ECFCs was measured using confocal microscopy, followed by automated quantification of cell morphometrics and migration speed. RESULTS Through hierarchical clustering of RNA expression profiles, we could distinguish 2 major clusters within the ECFC cohort. Major differences were associated with proliferation and migration in cluster 1 and inflammation and endothelial-to-mesenchymal transition in cluster 2. Phenotypic profiling showed significantly more and smaller ECFCs in cluster 1, which contained more and longer Weibel-Palade bodies. Migration speed in cluster 1 was also significantly higher. CONCLUSION We observed a range of different RNA expression patterns between ECFC clones, mostly associated with inflammation and clear differences in Weibel-Palade body count and structure. We developed a quantitative polymerase chain reaction panel that can be used for the characterization of ECFC clones, which is essential for the correct analysis of pathogenic mechanisms in vascular disorders.
Collapse
Affiliation(s)
- Sebastiaan N J Laan
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, the Netherlands; Department of Hematology, Erasmus University Medical Centre, Rotterdam, the Netherlands. https://twitter.com/laan_bas
| | - Suzan de Boer
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, the Netherlands
| | - Richard J Dirven
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, the Netherlands
| | - Iris van Moort
- Department of Hematology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Thomas B Kuipers
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, the Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, the Netherlands
| | - Ruben Bierings
- Department of Hematology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Jeroen Eikenboom
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, the Netherlands.
| |
Collapse
|
5
|
Weekes A, Wasielewska JM, Pinto N, Jenkins J, Patel J, Li Z, Klein TJ, Meinert C. Harnessing the Regenerative Potential of Fetal Mesenchymal Stem Cells and Endothelial Colony-Forming Cells in the Biofabrication of Tissue-Engineered Vascular Grafts (TEVGs). J Tissue Eng Regen Med 2024; 2024:8707377. [PMID: 40225752 PMCID: PMC11919237 DOI: 10.1155/2024/8707377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/08/2024] [Indexed: 04/15/2025]
Abstract
Tissue engineering is a promising approach for the production of small-diameter vascular grafts; however, there are limited data directly comparing the suitability of applicable cell types for vessel biofabrication. Here, we investigated the potential of adult smooth muscle cells (SMCs), placental mesenchymal stem cells (MSCs), placental endothelial colony-forming cells (ECFCs), and a combination of MSCs and ECFCs on highly porous biocompatible poly(ɛ-caprolactone) (PCL) scaffolds produced via melt electrowriting (MEW) for the biofabrication of tissue-engineered vascular grafts (TEVGs). Cellular attachment, proliferation, and deposition of essential extracellular matrix (ECM) components were analysed in vitro over four weeks. TEVGs cultured with MSCs accumulated the highest levels of collagenous components within a dense ECM, while SMCs and the coculture were more sparsely populated, ascertained via histological and immunofluorescence imaging, and biochemical assessment. Scanning electron microscopy (SEM) enabled visualisation of morphological differences in cell attachment and growth, with MSCs and SMCs infiltrating and covering scaffolds completely within the 28-day culture period. Coverage and matrix deposition by ECFCs was limited. However, ECFCs lined the ECM formed by MSCs in coculture, visualised via immunostaining. Thus, of cells investigated, placental MSCs were identified as the preferred cell source for the fabrication of tissue-engineered constructs, exhibiting extensive population of porous polymer scaffolds and production of ECM components; with the inclusion of ECFCs for luminal endothelialisation, an encouraging outcome warranting further consideration in future studies. In combination, these findings represent a substantial step toward the development of the next generation of small-diameter vascular grafts in the management of cardiovascular disease.
Collapse
Affiliation(s)
- Angus Weekes
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Herston Biofabrication Institute, Metro North Hospital and Health Services, Herston, QLD, Australia
| | - Joanna M. Wasielewska
- Herston Biofabrication Institute, Metro North Hospital and Health Services, Herston, QLD, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Nigel Pinto
- Herston Biofabrication Institute, Metro North Hospital and Health Services, Herston, QLD, Australia
- Department of Vascular Surgery, The Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Jason Jenkins
- Herston Biofabrication Institute, Metro North Hospital and Health Services, Herston, QLD, Australia
- Department of Vascular Surgery, The Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Jatin Patel
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Woolloongabba, QLD, Australia
| | - Zhiyong Li
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Travis J. Klein
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Christoph Meinert
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, Australia
- Herston Biofabrication Institute, Metro North Hospital and Health Services, Herston, QLD, Australia
| |
Collapse
|
6
|
Liu Y, Lyons CJ, Ayu C, O'Brien T. Recent advances in endothelial colony-forming cells: from the transcriptomic perspective. J Transl Med 2024; 22:313. [PMID: 38532420 PMCID: PMC10967123 DOI: 10.1186/s12967-024-05108-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/18/2024] [Indexed: 03/28/2024] Open
Abstract
Endothelial colony-forming cells (ECFCs) are progenitors of endothelial cells with significant proliferative and angiogenic ability. ECFCs are a promising treatment option for various diseases, such as ischemic heart disease and peripheral artery disease. However, some barriers hinder the clinical application of ECFC therapeutics. One of the current obstacles is that ECFCs are dysfunctional due to the underlying disease states. ECFCs exhibit dysfunctional phenotypes in pathologic states, which include but are not limited to the following: premature neonates and pregnancy-related diseases, diabetes mellitus, cancers, haematological system diseases, hypoxia, pulmonary arterial hypertension, coronary artery diseases, and other vascular diseases. Besides, ECFCs are heterogeneous among donors, tissue sources, and within cell subpopulations. Therefore, it is important to elucidate the underlying mechanisms of ECFC dysfunction and characterize their heterogeneity to enable clinical application. In this review, we summarize the current and potential application of transcriptomic analysis in the field of ECFC biology. Transcriptomic analysis is a powerful tool for exploring the key molecules and pathways involved in health and disease and can be used to characterize ECFC heterogeneity.
Collapse
Affiliation(s)
- Yaqiong Liu
- Regenerative Medicine Institute (REMEDI), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Caomhán J Lyons
- Regenerative Medicine Institute (REMEDI), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Christine Ayu
- Regenerative Medicine Institute (REMEDI), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI), Biomedical Sciences Building, University of Galway, Galway, Ireland.
| |
Collapse
|
7
|
Lutter G, Pommert NS, Zhang X, Seiler J, Saeid Nia M, Meier D, Sellers SL, Gorb SN, Hansen JH, Seoudy H, Müller OJ, Saad M, Haneya A, Frank D, Puehler T, Sathananthan J. Producing and Testing Prototype Tissue-Engineered 3D Tri-Leaflet Valved Stents on Biodegradable Poly-ε-Caprolactone Scaffolds. Int J Mol Sci 2023; 24:17357. [PMID: 38139185 PMCID: PMC10744316 DOI: 10.3390/ijms242417357] [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: 07/21/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Transcatheter pulmonary valve replacement is a minimally-invasive alternative treatment for right ventricular outflow tract dysfunction and has been rapidly evolving over the past years. Heart valve prostheses currently available still have major limitations. Therefore, one of the significant challenges for the future is the roll out of transcatheter tissue engineered pulmonary valve replacement to more patients. In the present study, biodegradable poly-ε-caprolactone (PCL) nanofiber scaffolds in the form of a 3D leaflet matrix were successfully seeded with human endothelial colony-forming cells (ECFCs), human induced pluripotent stem cell-derived MSCs (hMSCs), and porcine MSCs (pMSCs) for three weeks for the generation of 3D tissue-engineered tri-leaflet valved stent grafts. The cell adhesion, proliferation, and distribution of these 3D heart leaflets was analyzed using fluorescence microscopy and scanning electron microscopy (SEM). All cell lineages were able to increase the overgrown leaflet area within the three-week timeframe. While hMSCs showed a consistent growth rate over the course of three weeks, ECFSs showed almost no increase between days 7 and 14 until a growth spurt appeared between days 14 and 21. More than 90% of heart valve leaflets were covered with cells after the full three-week culturing cycle in nearly all leaflet areas, regardless of which cell type was used. This study shows that seeded biodegradable PCL nanofiber scaffolds incorporated in nitinol or biodegradable stents will offer a new therapeutic option in the future.
Collapse
Affiliation(s)
- Georg Lutter
- Department of Cardiac Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (N.S.P.); (X.Z.); (M.S.N.); (A.H.); (T.P.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
| | - Nina Sophie Pommert
- Department of Cardiac Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (N.S.P.); (X.Z.); (M.S.N.); (A.H.); (T.P.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
| | - Xiling Zhang
- Department of Cardiac Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (N.S.P.); (X.Z.); (M.S.N.); (A.H.); (T.P.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
| | - Jette Seiler
- Department of Cardiac Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (N.S.P.); (X.Z.); (M.S.N.); (A.H.); (T.P.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
| | - Monireh Saeid Nia
- Department of Cardiac Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (N.S.P.); (X.Z.); (M.S.N.); (A.H.); (T.P.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
| | - David Meier
- Department of Cardiology, Lausanne University Hospital and University of Lausanne, 1015 Lausanne, Switzerland;
| | - Stephanie L. Sellers
- Centre for Cardiovascular Innovation, St Paul’s and Vancouver General Hospital, Vancouver, BC V6Z 1Y6, Canada; (S.L.S.); (J.S.)
- Cardiovascular Translational Laboratory, Providence Research & Centre for Heart Lung Innovation, Vancouver, BC V6Z 1Y6, Canada
- Centre for Heart Valve Innovation, St. Paul’s Hospital, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Stanislav N. Gorb
- Department of Functional Morphology and Biomechanics, Zoological Institute, Christian-Albrecht University of Kiel, 24105 Kiel, Germany
| | - Jan-Hinnerk Hansen
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
- Department of Congenital Heart Disease and Pediatric Cardiology, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Hatim Seoudy
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany
| | - Oliver J. Müller
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany
| | - Mohammed Saad
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany
| | - Assad Haneya
- Department of Cardiac Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (N.S.P.); (X.Z.); (M.S.N.); (A.H.); (T.P.)
| | - Derk Frank
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany
| | - Thomas Puehler
- Department of Cardiac Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (N.S.P.); (X.Z.); (M.S.N.); (A.H.); (T.P.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 69120 Hamburg, Germany; (J.-H.H.); (H.S.); (O.J.M.); (M.S.); (D.F.)
| | - Janarthanan Sathananthan
- Centre for Cardiovascular Innovation, St Paul’s and Vancouver General Hospital, Vancouver, BC V6Z 1Y6, Canada; (S.L.S.); (J.S.)
- Cardiovascular Translational Laboratory, Providence Research & Centre for Heart Lung Innovation, Vancouver, BC V6Z 1Y6, Canada
- Centre for Heart Valve Innovation, St. Paul’s Hospital, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
8
|
Hall E, Alderfer L, Neu E, Saha S, Johandes E, Haas DM, Haneline LS, Hanjaya-Putra D. The Effects of Preeclamptic Milieu on Cord Blood Derived Endothelial Colony-Forming Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.03.569585. [PMID: 38105991 PMCID: PMC10723349 DOI: 10.1101/2023.12.03.569585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Preeclampsia is one of the leading causes of infant and maternal mortality worldwide. Many infants born from preeclamptic pregnancies are born prematurely with higher risk of developing cardiovascular later in their life. A key mechanism by which these complications occur is through stress-induced dysfunction of endothelial progenitor cells (EPCs), including endothelial colony-forming cells (ECFCs). To gain insight into this, cord blood derived ECFCs isolated from preeclamptic pregnancies (PRECs) were analyzed and compared to their healthy counterparts. While PRECs preserve key endothelial markers, they upregulate several markers associated with oxidative stress and inflammatory response. Compared to ECFCs, PRECs also exhibit lower migratory behaviors and impaired angiogenic potential. Interestingly, treatment of neuropilin-1 can improve tube formation in vitro. Collectively, this study reports that preeclamptic milieu influence phenotypes and functionality of PRECs, which can be rejuvenated using exogenous molecules. Promising results from this study warrant future investigations on the prospect of the rejuvenated PRECs to improve lung function of infants born from preeclamptic pregnancies.
Collapse
Affiliation(s)
- Eva Hall
- Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame
| | - Laura Alderfer
- Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame
| | - Erin Neu
- Department of Obstetrics & Gynecology, Indiana University School of Medicine, Indianapolis, IN
| | - Sanjoy Saha
- Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame
| | - Ellie Johandes
- Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame
| | - David M. Haas
- Department of Obstetrics & Gynecology, Indiana University School of Medicine, Indianapolis, IN
| | - Laura S. Haneline
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Riley Hospital for Children at Indiana University Health, Indianapolis, IN
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN
| | - Donny Hanjaya-Putra
- Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame
- Department of Obstetrics & Gynecology, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
9
|
Liang J, Zhao J, Chen Y, Li B, Li Y, Lu F, Dong Z. New Insights and Advanced Strategies for In Vitro Construction of Vascularized Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:692-709. [PMID: 37409413 DOI: 10.1089/ten.teb.2023.0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
Inadequate vascularization is a significant barrier to clinical application of large-volume tissue engineered grafts. In contrast to in vivo vascularization, in vitro prevascularization shortens the time required for host vessels to grow into the graft core and minimizes necrosis in the core region of the graft. However, the challenge of prevascularization is to construct hierarchical perfusable vascular networks, increase graft volume, and form a vascular tip that can anastomose with host vessels. Understanding advances in in vitro prevascularization techniques and new insights into angiogenesis could overcome these obstacles. In the present review, we discuss new perspectives on angiogenesis, the differences between in vivo and in vitro tissue vascularization, the four elements of prevascularized constructs, recent advances in perfusion-based in vitro prevascularized tissue fabrication, and prospects for large-volume prevascularized tissue engineering.
Collapse
Affiliation(s)
- Jiancong Liang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jing Zhao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yunzi Chen
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Bin Li
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Ye Li
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Ziqing Dong
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
10
|
Frolov A, Lobov A, Kabilov M, Zainullina B, Tupikin A, Shishkova D, Markova V, Sinitskaya A, Grigoriev E, Markova Y, Kutikhin A. Multi-Omics Profiling of Human Endothelial Cells from the Coronary Artery and Internal Thoracic Artery Reveals Molecular but Not Functional Heterogeneity. Int J Mol Sci 2023; 24:15032. [PMID: 37834480 PMCID: PMC10573276 DOI: 10.3390/ijms241915032] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/02/2023] [Accepted: 10/08/2023] [Indexed: 10/15/2023] Open
Abstract
Major adverse cardiovascular events occurring upon coronary artery bypass graft surgery are typically accompanied by endothelial dysfunction. Total arterial revascularisation, which employs both left and right internal thoracic arteries instead of the saphenous vein to create a bypass, is associated with better mid- and long-term outcomes. We suggested that molecular profiles of human coronary artery endothelial cells (HCAECs) and human internal mammary artery endothelial cells (HITAECs) are coherent in terms of transcriptomic and proteomic signatures, which were then investigated by RNA sequencing and ultra-high performance liquid chromatography-mass spectrometry, respectively. Both HCAECs and HITAECs overexpressed molecules responsible for the synthesis of extracellular matrix (ECM) components, basement membrane assembly, cell-ECM adhesion, organisation of intercellular junctions, and secretion of extracellular vesicles. HCAECs were characterised by higher enrichment with molecular signatures of basement membrane construction, collagen biosynthesis and folding, and formation of intercellular junctions, whilst HITAECs were notable for augmented pro-inflammatory signaling, intensive synthesis of proteins and nitrogen compounds, and enhanced ribosome biogenesis. Despite HCAECs and HITAECs showing a certain degree of molecular heterogeneity, no specific markers at the protein level have been identified. Coherence of differentially expressed molecular categories in HCAECs and HITAECs suggests synergistic interactions between these ECs in a bypass surgery scenario.
Collapse
Affiliation(s)
- Alexey Frolov
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia; (A.F.); (D.S.); (V.M.); (A.S.); (E.G.); (Y.M.)
| | - Arseniy Lobov
- Laboratory for Regenerative Biomedicine, Research Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretskiy Prospekt, St. Petersburg 194064, Russia;
| | - Marsel Kabilov
- SB RAS Genomics Core Facility, Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, 8 Prospekt Akademika Lavrentieva, Novosibirsk 630090, Russia; (M.K.); (A.T.)
| | - Bozhana Zainullina
- Centre for Molecular and Cell Technologies, Research Park, Saint Petersburg State University, 7/9 Universitetskaya Embankment, St. Petersburg 199034, Russia;
| | - Alexey Tupikin
- SB RAS Genomics Core Facility, Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, 8 Prospekt Akademika Lavrentieva, Novosibirsk 630090, Russia; (M.K.); (A.T.)
| | - Daria Shishkova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia; (A.F.); (D.S.); (V.M.); (A.S.); (E.G.); (Y.M.)
| | - Victoria Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia; (A.F.); (D.S.); (V.M.); (A.S.); (E.G.); (Y.M.)
| | - Anna Sinitskaya
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia; (A.F.); (D.S.); (V.M.); (A.S.); (E.G.); (Y.M.)
| | - Evgeny Grigoriev
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia; (A.F.); (D.S.); (V.M.); (A.S.); (E.G.); (Y.M.)
| | - Yulia Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia; (A.F.); (D.S.); (V.M.); (A.S.); (E.G.); (Y.M.)
| | - Anton Kutikhin
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia; (A.F.); (D.S.); (V.M.); (A.S.); (E.G.); (Y.M.)
| |
Collapse
|
11
|
Schwarz N, Yadegari H. Potentials of Endothelial Colony-Forming Cells: Applications in Hemostasis and Thrombosis Disorders, from Unveiling Disease Pathophysiology to Cell Therapy. Hamostaseologie 2023; 43:325-337. [PMID: 37857295 DOI: 10.1055/a-2101-5936] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
Endothelial colony-forming cells (ECFCs) are endothelial progenitor cells circulating in a limited number in peripheral blood. They can give rise to mature endothelial cells (ECs) and, with intrinsically high proliferative potency, contribute to forming new blood vessels and restoring the damaged endothelium in vivo. ECFCs can be isolated from peripheral blood or umbilical cord and cultured to generate large amounts of autologous ECs in vitro. Upon differentiation in culture, ECFCs are excellent surrogates for mature ECs showing the same phenotypic, genotypic, and functional features. In the last two decades, the ECFCs from various vascular disease patients have been widely used to study the diseases' pathophysiology ex vivo and develop cell-based therapeutic approaches, including vascular regenerative therapy, tissue engineering, and gene therapy. In the current review, we will provide an updated overview of past studies, which have used ECFCs to elucidate the molecular mechanisms underlying the pathogenesis of hemostatic disorders in basic research. Additionally, we summarize preceding studies demonstrating the utility of ECFCs as cellular tools for diagnostic or therapeutic clinical applications in thrombosis and hemostasis.
Collapse
Affiliation(s)
- Nadine Schwarz
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Hamideh Yadegari
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
12
|
Tang Y, Yin L, Gao S, Long X, Du Z, Zhou Y, Zhao S, Cao Y, Pan S. A small-diameter vascular graft immobilized peptides for capturing endothelial colony-forming cells. Front Bioeng Biotechnol 2023; 11:1154986. [PMID: 37101749 PMCID: PMC10123284 DOI: 10.3389/fbioe.2023.1154986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/23/2023] [Indexed: 04/28/2023] Open
Abstract
Combining synthetic polymers and biomacromolecules prevents the occurrence of thrombogenicity and intimal hyperplasia in small-diameter vascular grafts (SDVGs). In the present study, an electrospinning poly (L)-lactic acid (PLLA) bilayered scaffold is developed to prevent thrombosis after implantation by promoting the capture and differentiation of endothelial colony-forming cells (ECFCs). The scaffold consists of an outer PLLA scaffold and an inner porous PLLA biomimetic membrane combined with heparin (Hep), peptide Gly-Gly-Gly-Arg-Glu-Asp-Val (GGG-REDV), and vascular endothelial growth factor (VEGF). Attenuated total reflection Fourier transform infrared (ATR-FTIR) spectroscopy, X-ray photoelectron spectroscopy (XPS), and contact angle goniometry were performed to determine successful synthesis. The tensile strength of the outer layer was obtained using the recorded stress/strain curves, and hemocompatibility was evaluated using the blood clotting test. The proliferation, function, and differentiation properties of ECFCs were measured on various surfaces. Scanning electronic microscopy (SEM) was used to observe the morphology of ECFCs on the surface. The outer layer of scaffolds exhibited a similar strain and stress performance as the human saphenous vein via the tensile experiment. The contact angle decreased continuously until it reached 56° after REDV/VEGF modification, and SEM images of platelet adhesion showed a better hemocompatibility surface after modification. The ECFCs were captured using the REDV + VEGF + surface successfully under flow conditions. The expression of mature ECs was constantly increased with the culture of ECFCs on REDV + VEGF + surfaces. SEM images showed that the ECFCs captured by the REDV + VEGF + surface formed capillary-like structures after 4 weeks of culture. The SDVGs modified by REDV combined with VEGF promoted ECFC capture and rapid differentiation into ECs, forming capillary-like structures in vitro. The bilayered SDVGs could be used as vascular devices that achieved a high patency rate and rapid re-endothelialization.
Collapse
Affiliation(s)
- Yaqi Tang
- Heart Center, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, China
| | - Lu Yin
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou, China
| | - Shuai Gao
- Heart Center, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, China
| | - Xiaojing Long
- State Key Laboratory of Bio-fibers and Eco-textiles, Qingdao University, Qingdao, China
| | - Zhanhui Du
- Heart Center, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, China
| | - Yingchao Zhou
- Heart Center, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, China
| | - Shuiyan Zhao
- Heart Center, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, China
| | - Yue Cao
- Heart Center, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, China
| | - Silin Pan
- Heart Center, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
13
|
Guo Z, Wan X, Luo Y, Liang F, Jiang S, Yuan X, Mo Z. The vicious circle of UHRF1 down-regulation and KEAP1/NRF2/HO-1 pathway impairment promotes oxidative stress-induced endothelial cell apoptosis in diabetes. Diabet Med 2023; 40:e15026. [PMID: 36510823 DOI: 10.1111/dme.15026] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 12/05/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Oxidative stress is recognized as a key factor in the induction of endothelial dysfunction in diabetes. However, the specific mechanisms have not been fully elucidated. We herein hypothesized that ubiquitin-like containing PHD and RING finger domains 1 (UHRF1) might have a role in oxidative stress-induced endothelial cell (EC) apoptosis in diabetes. METHODS Western blot, qPCR, wound healing assay, apoptosis assay, reactive oxygen species (ROS) detection, dual-luciferase reporter assay, methylation-specific PCR, bisulfite sequencing PCR and chromatin immunoprecipitation assay were performed. RESULTS UHRF1 expression levels were significantly decreased in endothelial colony-forming cells derived from peripheral blood of participants with type 2 diabetes compared with individuals without diabetes. ECs treated with high glucose, palmitate or hydrogen peroxide in vitro also exhibited decreased UHRF1 protein levels. Silencing of UHRF1 led to decreased migration ability and increased apoptosis and ROS production in ECs, which might be related to impaired Kelch-like ECH-associated protein 1 (KEAP1)/nuclear factor erythroid 2-related factor 2 (NRF2)/haeme oxygenase-1 pathway. Mechanistically, UHRF1 is closely implicated in epigenetic regulation of chromatin modification status at KEAP1 genomic locus via histone acetylation. NRF2 down-regulation in turn inhibits UHRF1 protein level, which might be due to increased ROS generation. CONCLUSION Diabetes-induced oxidative stress can mediate down-regulation of UHRF1, which enhances ROS production by regulating KEAP1/p-NRF2 pathway through histone acetylation and might also form a self-perpetuating feedback loop with KEAP1/p-NRF2 to further promote oxidative stress-induced apoptosis of ECs in diabetes.
Collapse
Affiliation(s)
- Zi Guo
- Department of Endocrinology, The Third Xiangya Hospital and Diabetic Foot Research Centre of Central South University, Changsha, China
| | - Xinxing Wan
- Department of Endocrinology, The Third Xiangya Hospital and Diabetic Foot Research Centre of Central South University, Changsha, China
| | - Yufang Luo
- Department of Endocrinology, The Third Xiangya Hospital and Diabetic Foot Research Centre of Central South University, Changsha, China
| | - Fang Liang
- Department of Endocrinology, The Third Xiangya Hospital and Diabetic Foot Research Centre of Central South University, Changsha, China
| | - Siwei Jiang
- Department of Endocrinology, The Third Xiangya Hospital and Diabetic Foot Research Centre of Central South University, Changsha, China
| | - Xiuhong Yuan
- Department of Clinical Psychology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhaohui Mo
- Department of Endocrinology, The Third Xiangya Hospital and Diabetic Foot Research Centre of Central South University, Changsha, China
| |
Collapse
|
14
|
Wu Z, Thierry K, Bachy S, Zhang X, Gamradt P, Hernandez-Vargas H, Mikaelian I, Tonon L, Pommier R, Zhao Y, Bertolino P, Hennino A. Pericyte stem cells induce Ly6G + cell accumulation and immunotherapy resistance in pancreatic cancer. EMBO Rep 2023; 24:e56524. [PMID: 36802267 PMCID: PMC10074138 DOI: 10.15252/embr.202256524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/23/2023] [Accepted: 02/01/2023] [Indexed: 02/23/2023] Open
Abstract
We report the identification of a cell population that shares pericyte, stromal and stemness features, does not harbor the KrasG12D mutation and drives tumoral growth in vitro and in vivo. We term these cells pericyte stem cells (PeSCs) and define them as CD45- EPCAM- CD29+ CD106+ CD24+ CD44+ cells. We perform studies with p48-Cre;KrasG12D (KC), pdx1-Cre;KrasG12D ;Ink4a/Arffl/fl (KIC) and pdx1-Cre;KrasG12D ;p53R172H (KPC) and tumor tissues from PDAC and chronic pancreatitis patients. We also perform single-cell RNAseq analysis and reveal a unique signature of PeSC. Under steady-state conditions, PeSCs are barely detectable in the pancreas but present in the neoplastic microenvironment both in humans and mice. The coinjection of PeSCs and tumor epithelial cells leads to increased tumor growth, differentiation of Ly6G+ myeloid-derived suppressor cells, and a decreased amount of F4/80+ macrophages and CD11c+ dendritic cells. This population induces resistance to anti-PD-1 immunotherapy when coinjected with epithelial tumor cells. Our data reveal the existence of a cell population that instructs immunosuppressive myeloid cell responses to bypass PD-1 targeting and thus suggest potential new approaches for overcoming resistance to immunotherapy in clinical settings.
Collapse
Affiliation(s)
- Zhichong Wu
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France.,Université Lyon 1, Lyon, France.,Centre Léon Bérard, Lyon, France.,Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kevin Thierry
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France.,Université Lyon 1, Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Sophie Bachy
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France.,Université Lyon 1, Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Xinyi Zhang
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France.,Université Lyon 1, Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Pia Gamradt
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France.,Université Lyon 1, Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Hector Hernandez-Vargas
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France.,Université Lyon 1, Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Ivan Mikaelian
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France.,Université Lyon 1, Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Laurie Tonon
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France.,Université Lyon 1, Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Roxanne Pommier
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France.,Université Lyon 1, Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Yajie Zhao
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France.,Université Lyon 1, Lyon, France.,Centre Léon Bérard, Lyon, France.,Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Philippe Bertolino
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France.,Université Lyon 1, Lyon, France.,Centre Léon Bérard, Lyon, France
| | - Ana Hennino
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France.,Université Lyon 1, Lyon, France.,Centre Léon Bérard, Lyon, France
| |
Collapse
|
15
|
Markova V, Bogdanov L, Velikanova E, Kanonykina A, Frolov A, Shishkova D, Lazebnaya A, Kutikhin A. Endothelial Cell Markers Are Inferior to Vascular Smooth Muscle Cells Markers in Staining Vasa Vasorum and Are Non-Specific for Distinct Endothelial Cell Lineages in Clinical Samples. Int J Mol Sci 2023; 24:ijms24031959. [PMID: 36768296 PMCID: PMC9916324 DOI: 10.3390/ijms24031959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/20/2023] Open
Abstract
Current techniques for the detection of vasa vasorum (VV) in vascular pathology include staining for endothelial cell (EC) markers such as CD31 or VE-cadherin. However, this approach does not permit an objective assessment of vascular geometry upon vasospasm and the clinical relevance of endothelial specification markers found in developmental biology studies remains unclear. Here, we performed a combined immunostaining of rat abdominal aorta (rAA) and human saphenous vein (hSV) for various EC or vascular smooth muscle cell (VSMC) markers and found that the latter (e.g., alpha smooth muscle actin (α-SMA) or smooth muscle myosin heavy chain (SM-MHC)) ensure a several-fold higher signal-to-noise ratio irrespective of the primary antibody origin, fluorophore, or VV type (arterioles, venules, or capillaries). Further, α-SMA or SM-MHC staining allowed unbiased evaluation of the VV area under vasospasm. Screening of the molecular markers of endothelial heterogeneity (mechanosensitive transcription factors KLF2 and KLF4, arterial transcription factors HES1, HEY1, and ERG, venous transcription factor NR2F2, and venous/lymphatic markers PROX1, LYVE1, VEGFR3, and NRP2) have not revealed specific markers of any lineage in hSV (although KLF2 and PROX1 were restricted to venous endothelium in rAA), suggesting the need in high-throughput searches for the clinically relevant signatures of arterial, venous, lymphatic, or capillary differentiation.
Collapse
|
16
|
Antonova L, Kutikhin A, Sevostianova V, Lobov A, Repkin E, Krivkina E, Velikanova E, Mironov A, Mukhamadiyarov R, Senokosova E, Khanova M, Shishkova D, Markova V, Barbarash L. Controlled and Synchronised Vascular Regeneration upon the Implantation of Iloprost- and Cationic Amphiphilic Drugs-Conjugated Tissue-Engineered Vascular Grafts into the Ovine Carotid Artery: A Proteomics-Empowered Study. Polymers (Basel) 2022; 14:polym14235149. [PMID: 36501545 PMCID: PMC9736446 DOI: 10.3390/polym14235149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/17/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
Implementation of small-diameter tissue-engineered vascular grafts (TEVGs) into clinical practice is still delayed due to the frequent complications, including thrombosis, aneurysms, neointimal hyperplasia, calcification, atherosclerosis, and infection. Here, we conjugated a vasodilator/platelet inhibitor, iloprost, and an antimicrobial cationic amphiphilic drug, 1,5-bis-(4-tetradecyl-1,4-diazoniabicyclo [2.2.2]octan-1-yl) pentane tetrabromide, to the luminal surface of electrospun poly(ε-caprolactone) (PCL) TEVGs for preventing thrombosis and infection, additionally enveloped such TEVGs into the PCL sheath to preclude aneurysms, and implanted PCLIlo/CAD TEVGs into the ovine carotid artery (n = 12) for 6 months. The primary patency was 50% (6/12 animals). TEVGs were completely replaced with the vascular tissue, free from aneurysms, calcification, atherosclerosis and infection, completely endothelialised, and had clearly distinguishable medial and adventitial layers. Comparative proteomic profiling of TEVGs and contralateral carotid arteries found that TEVGs lacked contractile vascular smooth muscle cell markers, basement membrane components, and proteins mediating antioxidant defense, concurrently showing the protein signatures of upregulated protein synthesis, folding and assembly, enhanced energy metabolism, and macrophage-driven inflammation. Collectively, these results suggested a synchronised replacement of PCL with a newly formed vascular tissue but insufficient compliance of PCLIlo/CAD TEVGs, demanding their testing in the muscular artery position or stimulation of vascular smooth muscle cell specification after the implantation.
Collapse
Affiliation(s)
- Larisa Antonova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Anton Kutikhin
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
- Correspondence: ; Tel.: +7-9609077067
| | - Viktoriia Sevostianova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Arseniy Lobov
- Department of Regenerative Biomedicine, Research Institute of Cytology, 4 Tikhoretskiy Prospekt, Saint Petersburg 194064, Russia
| | - Egor Repkin
- Centre for Molecular and Cell Technologies, Saint Petersburg State University, Universitetskaya Embankment, 7/9, Saint Petersburg 199034, Russia
| | - Evgenia Krivkina
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Elena Velikanova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Andrey Mironov
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Rinat Mukhamadiyarov
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Evgenia Senokosova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Mariam Khanova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Daria Shishkova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Victoria Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Leonid Barbarash
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| |
Collapse
|
17
|
Sinitsky M, Sinitskaya A, Shishkova D, Ponasenko A. Genotoxic stress leads to the proinflammatory response of endothelial cells: an in vitro study. BIOMEDITSINSKAYA KHIMIYA 2022; 68:361-366. [DOI: 10.18097/pbmc20226805361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
It was shown, that genotoxic stress can trigger endothelial disfunction and atherosclerosis, but the molecular genetic mechanisms of this process are poorly investigated. At the same time, inflammation also plays the important role in atherogenesis. This study aimed access of inflammatory marker expression in the endothelial cells exposed to alkylating mutagen mitomycin C (MMC). Primary human coronary (HCAEC) and internal thoracic artery endothelial cells (HITAEC) exposed to 500 ng/ml MMC (experimental group) and 0.9% NaCl (control) were used in this research. A gene expression profile was evaluated by quantitative reverse transcription PCR after 6 h exposure of endothelial cells to MMC (or 0.9% NaCl) followed by subsequent 24 h incubation in the mutagen-free cell growth media. The cytokine profile of endotheliocytes was studied by dot blotting. We found that MIF, IL-8, MCP-1, IP-10 and PDGFB were upregulated both in HCAEC and HITAEC, while MIP-1β release remained unchanged. TIMP-2 was upregulated in HCAEC but not in HITAEC. sTNF RI was expressed only in HCAEC. According to gene expression analysis, HCAEC exposed to MMC are characterized by the increased mRNA level of IL-8, MCP-1 and IP-10; decreased expression of TIMP-2 and no differences in the expression of MIF, MIP-1β and PDGFB compared to the control. In HITAEC, increased mRNA level of IL-8 and IP-10; decreased expression of MIF and TIMP-2, no differences in the expression of MCP-1, MIP-1β and PDGFB was shown. TNF-RI expression was not detected in both cell lines. Thus, genotoxic stress in endothelial cells induced by MMC leads to differential inflammatory response that can trigger endothelial dysfunction.
Collapse
Affiliation(s)
- M.Y. Sinitsky
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - A.V. Sinitskaya
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - D.K. Shishkova
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - A.V. Ponasenko
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| |
Collapse
|
18
|
Kraus X, van de Flierdt E, Renzelmann J, Thoms S, Witt M, Scheper T, Blume C. Peripheral blood derived endothelial colony forming cells as suitable cell source for pre-endothelialization of arterial vascular grafts under dynamic flow conditions. Microvasc Res 2022; 143:104402. [PMID: 35753506 DOI: 10.1016/j.mvr.2022.104402] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/11/2022] [Accepted: 06/14/2022] [Indexed: 11/26/2022]
Abstract
In regenerative medicine, autologous peripheral blood derived endothelial colony forming cells (PB-derived ECFC) represent a promising source of endothelial cells (EC) for pre-endothelialization of arterial tissue engineered vascular grafts (TEVG) since they are readily attainable, can easily be isolated and possess a high proliferation potential. The aim of this study was to compare the phenotype of PB-derived ECFC with arterial and venous model cells such as human aortic endothelial cells (HAEC) and human umbilical vein endothelial cells (HUVEC) under dynamic cell culture conditions to find a suitable cell source of EC for pre-endothelialization. In this study PB-derived ECFC were cultivated over 24 h under a high pulsatile shear stress (20 dyn/cm2, 1 Hz) and subsequently analyzed. ECFC oriented and elongated in the direction of flow and expressed similar anti-thrombotic and endothelial differentiation markers compared to HAEC. There were significant differences observable in gene expression levels of CD31, CD34 and NOTCH4 between ECFC and HUVEC. These results therefore suggest an arterial phenotype for PB-derived ECFC both under static and flow conditions, and this was supported by NOTCH4 protein expression profiles. ECFC also significantly up-regulated gene expression levels of anti-thrombotic genes such as krueppel-like factor 2, endothelial nitric oxide synthase 3 and thrombomodulin under shear stress cultivation as compared to static conditions. Dynamically cultured PB-derived ECFC therefore may be a promising cell source for pre-endothelialization of arterial TEVGs.
Collapse
Affiliation(s)
- Xenia Kraus
- Leibniz University Hannover, Institute of Technical Chemistry, Callinstr. 5, D-30167 Hannover, Germany; Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany.
| | - Edda van de Flierdt
- Leibniz University Hannover, Institute of Technical Chemistry, Callinstr. 5, D-30167 Hannover, Germany; Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Jannis Renzelmann
- Leibniz University Hannover, Institute of Technical Chemistry, Callinstr. 5, D-30167 Hannover, Germany; Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Stefanie Thoms
- Leibniz University Hannover, Institute of Technical Chemistry, Callinstr. 5, D-30167 Hannover, Germany; Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Martin Witt
- Leibniz University Hannover, Institute of Technical Chemistry, Callinstr. 5, D-30167 Hannover, Germany; Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Thomas Scheper
- Leibniz University Hannover, Institute of Technical Chemistry, Callinstr. 5, D-30167 Hannover, Germany; Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany
| | - Cornelia Blume
- Leibniz University Hannover, Institute of Technical Chemistry, Callinstr. 5, D-30167 Hannover, Germany; Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany
| |
Collapse
|
19
|
Kutikhin AG, Shishkova DK, Velikanova EA, Sinitsky MY, Sinitskaya AV, Markova VE. Endothelial Dysfunction in the Context of Blood–Brain Barrier Modeling. J EVOL BIOCHEM PHYS+ 2022; 58:781-806. [PMID: 35789679 PMCID: PMC9243926 DOI: 10.1134/s0022093022030139] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 01/04/2023]
Abstract
Here, we discuss pathophysiological approaches to the defining
of endothelial dysfunction criteria (i.e., endothelial activation,
impaired endothelial mechanotransduction, endothelial-to-mesenchymal
transition, reduced nitric oxide release, compromised endothelial
integrity, and loss of anti-thrombogenic properties) in different
in vitro and in vivo models. The canonical definition of endothelial
dysfunction includes insufficient production of vasodilators, pro-thrombotic
and pro-inflammatory activation of endothelial cells, and pathologically
increased endothelial permeability. Among the clinical consequences
of endothelial dysfunction are arterial hypertension, macro- and
microangiopathy, and microalbuminuria. We propose to extend the definition
of endothelial dysfunction by adding altered endothelial mechanotransduction
and endothelial-to-mesenchymal transition to its criteria. Albeit
interleukin-6, interleukin-8, and MCP-1/CCL2 dictate the pathogenic
paracrine effects of dysfunctional endothelial cells and are therefore
reliable endothelial dysfunction biomarkers in vitro, they are non-specific
for endothelial cells and cannot be used for the diagnostics of
endothelial dysfunction in vivo. Conceptual improvements in the
existing methods to model endothelial dysfunction, specifically,
in relation to the blood–brain barrier, include endothelial cell
culturing under pulsatile flow, collagen IV coating of flow chambers,
and endothelial lysate collection from the blood vessels of laboratory
animals in situ for the subsequent gene and protein expression profiling.
Combined with the simulation of paracrine effects by using conditioned
medium from dysfunctional endothelial cells, these flow-sensitive
models have a high physiological relevance, bringing the experimental
conditions to the physiological scenario.
Collapse
Affiliation(s)
- A. G. Kutikhin
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - D. K. Shishkova
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - E. A. Velikanova
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - M. Yu. Sinitsky
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - A. V. Sinitskaya
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - V. E. Markova
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| |
Collapse
|
20
|
Matveeva VG, Senokosova EA, Sevostianova VV, Khanova MY, Glushkova TV, Akentieva TN, Antonova LV, Barbarash LS. Advantages of Fibrin Polymerization Method without the Use of Exogenous Thrombin for Vascular Tissue Engineering Applications. Biomedicines 2022; 10:biomedicines10040789. [PMID: 35453539 PMCID: PMC9026760 DOI: 10.3390/biomedicines10040789] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/16/2022] [Accepted: 03/25/2022] [Indexed: 01/01/2023] Open
Abstract
Fibrin is widely used in vascular tissue engineering. Typically, fibrin polymerization is initiated by adding exogenous thrombin. In this study, we proposed a protocol for the preparation of completely autologous fibrin without the use of endogenous thrombin and compared the properties of the prepared fibrin matrix with that obtained by the traditional method. Fibrinogen was obtained by ethanol precipitation followed by fibrin polymerization by adding either exogenous thrombin and calcium chloride (ExThr), or only calcium chloride (EnThr). We examined the structure, mechanical properties, thrombogenicity, degradation rate and cytocompatibility of fibrin matrices. Factor XIII (FXIII) quantitative assay was performed by ELISA, and FXIII activity was assessed by SDS-PAGE detection of γ-γ cross-links. The results show that network structure of EnThr fibrin was characterized by thinner fibers. The EnThr fibrin matrices had higher strength, stiffness and resistance to proteolytic degradation compared to ExThr fibrin. EnThr fibrin matrices exhibited less thrombogenicity in vitro than ExThr, and retained high cytocompatibility. Thus, the proposed approach has several advantages over the traditional method, namely the fabrication of a completely autologous coating material that has better mechanical properties, higher resistance to proteolysis and lower thrombogenicity.
Collapse
|
21
|
Failure Analysis of TEVG’s II: Late Failure and Entering the Regeneration Pathway. Cells 2022; 11:cells11060939. [PMID: 35326390 PMCID: PMC8946846 DOI: 10.3390/cells11060939] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/03/2022] [Accepted: 03/01/2022] [Indexed: 12/21/2022] Open
Abstract
Tissue-engineered vascular grafts (TEVGs) are a promising alternative to treat vascular disease under complex hemodynamic conditions. However, despite efforts from the tissue engineering and regenerative medicine fields, the interactions between the material and the biological and hemodynamic environment are still to be understood, and optimization of the rational design of vascular grafts is an open challenge. This is of special importance as TEVGs not only have to overcome the surgical requirements upon implantation, they also need to withhold the inflammatory response and sustain remodeling of the tissue. This work aims to analyze and evaluate the bio-molecular interactions and hemodynamic phenomena between blood components, cells and materials that have been reported to be related to the failure of the TEVGs during the regeneration process once the initial stages of preimplantation have been resolved, in order to tailor and refine the needed criteria for the optimal design of TEVGs.
Collapse
|
22
|
Regulation of endothelial progenitor cell functions during hyperglycemia: new therapeutic targets in diabetic wound healing. J Mol Med (Berl) 2022; 100:485-498. [PMID: 34997250 DOI: 10.1007/s00109-021-02172-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/16/2021] [Accepted: 12/02/2021] [Indexed: 11/09/2022]
Abstract
Diabetes is primarily characterized by hyperglycemia, and its high incidence is often very costly to patients, their families, and national economies. Unsurprisingly, the number and function of endothelial progenitor cells (EPCs) decrease in patients resulting in diabetic wound non-healing. As precursors of endothelial cells (ECs), these cells were discovered in 1997 and found to play an essential role in wound healing. Their function, number, and role in wound healing has been widely investigated. Hitherto, a lot of complex molecular mechanisms have been discovered. In this review, we summarize the mechanisms of how hyperglycemia affects the function and number of EPCs and how the affected cells impact wound healing. We aim to provide a complete summary of the relationship between diabetic hyperglycosemia, EPCs, and wound healing, as well as a better comprehensive platform for subsequent related research.
Collapse
|
23
|
Lutter G, Puehler T, Cyganek L, Seiler J, Rogler A, Herberth T, Knueppel P, Gorb SN, Sathananthan J, Sellers S, Müller OJ, Frank D, Haben I. Biodegradable Poly-ε-Caprolactone Scaffolds with ECFCs and iMSCs for Tissue-Engineered Heart Valves. Int J Mol Sci 2022; 23:527. [PMID: 35008953 PMCID: PMC8745109 DOI: 10.3390/ijms23010527] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 12/16/2022] Open
Abstract
Clinically used heart valve prostheses, despite their progress, are still associated with limitations. Biodegradable poly-ε-caprolactone (PCL) nanofiber scaffolds, as a matrix, were seeded with human endothelial colony-forming cells (ECFCs) and human induced-pluripotent stem cells-derived MSCs (iMSCs) for the generation of tissue-engineered heart valves. Cell adhesion, proliferation, and distribution, as well as the effects of coating PCL nanofibers, were analyzed by fluorescence microscopy and SEM. Mechanical properties of seeded PCL scaffolds were investigated under uniaxial loading. iPSCs were used to differentiate into iMSCs via mesoderm. The obtained iMSCs exhibited a comparable phenotype and surface marker expression to adult human MSCs and were capable of multilineage differentiation. EFCFs and MSCs showed good adhesion and distribution on PCL fibers, forming a closed cell cover. Coating of the fibers resulted in an increased cell number only at an early time point; from day 7 of colonization, there was no difference between cell numbers on coated and uncoated PCL fibers. The mechanical properties of PCL scaffolds under uniaxial loading were compared with native porcine pulmonary valve leaflets. The Young's modulus and mean elongation at Fmax of unseeded PCL scaffolds were comparable to those of native leaflets (p = ns.). Colonization of PCL scaffolds with human ECFCs or iMSCs did not alter these properties (p = ns.). However, the native heart valves exhibited a maximum tensile stress at a force of 1.2 ± 0.5 N, whereas it was lower in the unseeded PCL scaffolds (0.6 ± 0.0 N, p < 0.05). A closed cell layer on PCL tissues did not change the values of Fmax (ECFCs: 0.6 ± 0.1 N; iMSCs: 0.7 ± 0.1 N). Here, a successful two-phase protocol, based on the timed use of differentiation factors for efficient differentiation of human iPSCs into iMSCs, was developed. Furthermore, we demonstrated the successful colonization of a biodegradable PCL nanofiber matrix with human ECFCs and iMSCs suitable for the generation of tissue-engineered heart valves. A closed cell cover was already evident after 14 days for ECFCs and 21 days for MSCs. The PCL tissue did not show major mechanical differences compared to native heart valves, which was not altered by short-term surface colonization with human cells in the absence of an extracellular matrix.
Collapse
Affiliation(s)
- Georg Lutter
- Department of Cardiovascular Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (T.P.); (J.S.); (A.R.); (T.H.); (P.K.); (I.H.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany; (O.J.M.); (D.F.)
| | - Thomas Puehler
- Department of Cardiovascular Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (T.P.); (J.S.); (A.R.); (T.H.); (P.K.); (I.H.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany; (O.J.M.); (D.F.)
| | - Lukas Cyganek
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany;
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Jette Seiler
- Department of Cardiovascular Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (T.P.); (J.S.); (A.R.); (T.H.); (P.K.); (I.H.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany; (O.J.M.); (D.F.)
| | - Anita Rogler
- Department of Cardiovascular Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (T.P.); (J.S.); (A.R.); (T.H.); (P.K.); (I.H.)
| | - Tanja Herberth
- Department of Cardiovascular Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (T.P.); (J.S.); (A.R.); (T.H.); (P.K.); (I.H.)
| | - Philipp Knueppel
- Department of Cardiovascular Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (T.P.); (J.S.); (A.R.); (T.H.); (P.K.); (I.H.)
| | - Stanislav N. Gorb
- Department of Functional Morphology and Biomechanics, Zoological Institute, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany;
| | - Janarthanan Sathananthan
- Department of Centre for Heart Valve Innovation, St Paul’s Hospital, University of British Columbia, Vancouver, BC V6T 174, Canada; (J.S.); (S.S.)
| | - Stephanie Sellers
- Department of Centre for Heart Valve Innovation, St Paul’s Hospital, University of British Columbia, Vancouver, BC V6T 174, Canada; (J.S.); (S.S.)
| | - Oliver J. Müller
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany; (O.J.M.); (D.F.)
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany
| | - Derk Frank
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany; (O.J.M.); (D.F.)
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany
| | - Irma Haben
- Department of Cardiovascular Surgery, University Hospital Schleswig-Holstein (UKSH), 24105 Kiel, Germany; (T.P.); (J.S.); (A.R.); (T.H.); (P.K.); (I.H.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany; (O.J.M.); (D.F.)
| |
Collapse
|
24
|
Chambers SE, Pathak V, Pedrini E, Soret L, Gendron N, Guerin CL, Stitt AW, Smadja DM, Medina RJ. Current concepts on endothelial stem cells definition, location, and markers. Stem Cells Transl Med 2021; 10 Suppl 2:S54-S61. [PMID: 34724714 PMCID: PMC8560200 DOI: 10.1002/sctm.21-0022] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/12/2021] [Accepted: 03/25/2021] [Indexed: 12/20/2022] Open
Abstract
Ischemic vascular disease is a major cause of mortality and morbidity worldwide, and regeneration of blood vessels in perfusion-deficient tissues is a worthwhile therapeutic goal. The idea of delivering endothelial stem/progenitor cells to repair damaged vasculature, reperfuse hypoxic tissue, prevent cell death, and consequently diminish tissue inflammation and fibrosis has a strong scientific basis and clinical value. Various labs have proposed endothelial stem/progenitor cell candidates. This has created confusion, as there are profound differences between these cell definitions based on isolation methodology, characterization, and reparative biology. Here, a stricter definition based on stem cell biology principles is proposed. Although preclinical studies have often been promising, results from clinical trials have been highly contradictory and served to highlight multiple challenges associated with disappointing therapeutic benefit. This article reviews recent accomplishments in the field and discusses current difficulties when developing endothelial stem cell therapies. Emerging evidence that disputes the classic view of the bone marrow as the source for these cells and supports the vascular wall as the niche for these tissue-resident endothelial stem cells is considered. In addition, novel markers to identify endothelial stem cells, including CD157, EPCR, and CD31low VEGFR2low IL33+ Sox9+ , are described.
Collapse
Affiliation(s)
- Sarah E.J. Chambers
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| | - Varun Pathak
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| | - Edoardo Pedrini
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| | - Lou Soret
- Université de ParisInnovative Therapies in Haemostasis, INSERMParisFrance
- Hematology department and Biosurgical research lab (Carpentier Foundation)Assistance Publique Hôpitaux de Paris.Centre‐Université de Paris (APHP‐CUP)ParisFrance
| | - Nicolas Gendron
- Université de ParisInnovative Therapies in Haemostasis, INSERMParisFrance
- Hematology department and Biosurgical research lab (Carpentier Foundation)Assistance Publique Hôpitaux de Paris.Centre‐Université de Paris (APHP‐CUP)ParisFrance
| | - Coralie L. Guerin
- Université de ParisInnovative Therapies in Haemostasis, INSERMParisFrance
- Cytometry Platform, Institut CurieParisFrance
| | - Alan W. Stitt
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| | - David M. Smadja
- Université de ParisInnovative Therapies in Haemostasis, INSERMParisFrance
- Hematology department and Biosurgical research lab (Carpentier Foundation)Assistance Publique Hôpitaux de Paris.Centre‐Université de Paris (APHP‐CUP)ParisFrance
| | - Reinhold J. Medina
- Wellcome‐Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queen's University BelfastBelfastUK
| |
Collapse
|
25
|
Ferreira-Facio CDS, Botafogo V, Ferrão PM, Canellas MC, Milito CB, Romano S, Lopes DV, Teixeira LC, Oliveira E, Bruno-Riscarolli E, Mello FV, Siqueira PFR, Moura P, Macedo FN, Forny DN, Simião L, Pureza AL, Land MGP, Pedreira CE, van Dongen JJM, Orfao A, da Costa ES. Flow Cytometry Immunophenotyping for Diagnostic Orientation and Classification of Pediatric Cancer Based on the EuroFlow Solid Tumor Orientation Tube (STOT). Cancers (Basel) 2021; 13:cancers13194945. [PMID: 34638431 PMCID: PMC8508207 DOI: 10.3390/cancers13194945] [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: 08/12/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 11/15/2022] Open
Abstract
Simple Summary Pediatric solid tumors are a heterogenous group of diseases that comprise ≈ 40% of all pediatric cancers, early diagnosis being key for improved survival. Here we designed, tested, and validated a single eight-color tube for the diagnostic screening of pediatric cancer—solid tumor orientation tube (STOT)—based on multiparameter flow cytometry vs. conventional diagnostic procedures. Prospective clinical validation of STOT in 149 samples (63 tumor mass, 38 bone marrow, 30 lymph node, and 18 body fluid samples) screened for pediatric cancer, apart from 26 blood specimens that were excluded from analysis, showed concordant results with the final WHO/ICCC-3 diagnosis in 138/149 cases (92.6%). This included correct diagnostic orientation by STOT in 43/44 (98%) malignant and 4/4 (100%) benign non-hematopoietic tumors, together with 28/38 (74%) leukemia/lymphoma cases. The only recurrently missed diagnosis was Hodgkin lymphoma (0/8), which would require additional markers. These results support the use of STOT as a complementary tool for fast and accurate diagnostic screening, orientation, and classification of pediatric cancer in suspicious patients. Abstract Early diagnosis of pediatric cancer is key for adequate patient management and improved outcome. Although multiparameter flow cytometry (MFC) has proven of great utility in the diagnosis and classification of hematologic malignancies, its application to non-hematopoietic pediatric tumors remains limited. Here we designed and prospectively validated a new single eight-color antibody combination—solid tumor orientation tube, STOT—for diagnostic screening of pediatric cancer by MFC. A total of 476 samples (139 tumor mass, 138 bone marrow, 86 lymph node, 58 peripheral blood, and 55 other body fluid samples) from 296 patients with diagnostic suspicion of pediatric cancer were analyzed by MFC vs. conventional diagnostic procedures. STOT was designed after several design–test–evaluate–redesign cycles based on a large panel of monoclonal antibody combinations tested on 301 samples. In its final version, STOT consists of a single 8-color/12-marker antibody combination (CD99-CD8/numyogenin/CD4-EpCAM/CD56/GD2/smCD3-CD19/cyCD3-CD271/CD45). Prospective validation of STOT in 149 samples showed concordant results with the patient WHO/ICCC-3 diagnosis in 138/149 cases (92.6%). These included: 63/63 (100%) reactive/disease-free samples, 43/44 (98%) malignant and 4/4 (100%) benign non-hematopoietic tumors together with 28/38 (74%) leukemia/lymphoma cases; the only exception was Hodgkin lymphoma that required additional markers to be stained. In addition, STOT allowed accurate discrimination among the four most common subtypes of malignant CD45− CD56++ non-hematopoietic solid tumors: 13/13 (GD2++ numyogenin− CD271−/+ nuMyoD1− CD99− EpCAM−) neuroblastoma samples, 5/5 (GD2− numyogenin++ CD271++ nuMyoD1++ CD99−/+ EpCAM−) rhabdomyosarcomas, 2/2 (GD2−/+ numyogenin− CD271+ nuMyoD1− CD99+ EpCAM−) Ewing sarcoma family of tumors, and 7/7 (GD2− numyogenin− CD271+ nuMyoD1− CD99− EpCAM+) Wilms tumors. In summary, here we designed and validated a new standardized antibody combination and MFC assay for diagnostic screening of pediatric solid tumors that might contribute to fast and accurate diagnostic orientation and classification of pediatric cancer in routine clinical practice.
Collapse
Affiliation(s)
- Cristiane de Sá Ferreira-Facio
- Internal Medicine Postgraduate Program, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil; (C.d.S.F.-F.); (V.B.); (L.C.T.); (E.O.); (E.B.-R.); (P.F.R.S.); (M.G.P.L.)
- Cytometry Service, Institute of Paediatrics and Puericultura Martagão Gesteira (IPPMG), Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-612, Brazil; (P.M.F.); (M.C.C.); (D.V.L.); (F.V.M.); (L.S.); (A.L.P.)
| | - Vitor Botafogo
- Internal Medicine Postgraduate Program, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil; (C.d.S.F.-F.); (V.B.); (L.C.T.); (E.O.); (E.B.-R.); (P.F.R.S.); (M.G.P.L.)
- Cytometry Service, Institute of Paediatrics and Puericultura Martagão Gesteira (IPPMG), Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-612, Brazil; (P.M.F.); (M.C.C.); (D.V.L.); (F.V.M.); (L.S.); (A.L.P.)
| | - Patrícia Mello Ferrão
- Cytometry Service, Institute of Paediatrics and Puericultura Martagão Gesteira (IPPMG), Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-612, Brazil; (P.M.F.); (M.C.C.); (D.V.L.); (F.V.M.); (L.S.); (A.L.P.)
| | - Maria Clara Canellas
- Cytometry Service, Institute of Paediatrics and Puericultura Martagão Gesteira (IPPMG), Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-612, Brazil; (P.M.F.); (M.C.C.); (D.V.L.); (F.V.M.); (L.S.); (A.L.P.)
| | - Cristiane B. Milito
- Department of Pathology, Faculty of Medicine, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil;
| | - Sérgio Romano
- Laboratory of Anatomical Pathology and Cytopathology, Instituto Nacional de Câncer (INCa), Rio de Janeiro 20220-400, Brazil;
| | - Daiana V. Lopes
- Cytometry Service, Institute of Paediatrics and Puericultura Martagão Gesteira (IPPMG), Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-612, Brazil; (P.M.F.); (M.C.C.); (D.V.L.); (F.V.M.); (L.S.); (A.L.P.)
| | - Lisandra C. Teixeira
- Internal Medicine Postgraduate Program, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil; (C.d.S.F.-F.); (V.B.); (L.C.T.); (E.O.); (E.B.-R.); (P.F.R.S.); (M.G.P.L.)
- Cytometry Service, Institute of Paediatrics and Puericultura Martagão Gesteira (IPPMG), Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-612, Brazil; (P.M.F.); (M.C.C.); (D.V.L.); (F.V.M.); (L.S.); (A.L.P.)
| | - Elen Oliveira
- Internal Medicine Postgraduate Program, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil; (C.d.S.F.-F.); (V.B.); (L.C.T.); (E.O.); (E.B.-R.); (P.F.R.S.); (M.G.P.L.)
- Cytometry Service, Institute of Paediatrics and Puericultura Martagão Gesteira (IPPMG), Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-612, Brazil; (P.M.F.); (M.C.C.); (D.V.L.); (F.V.M.); (L.S.); (A.L.P.)
| | - Enrico Bruno-Riscarolli
- Internal Medicine Postgraduate Program, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil; (C.d.S.F.-F.); (V.B.); (L.C.T.); (E.O.); (E.B.-R.); (P.F.R.S.); (M.G.P.L.)
- Cytometry Service, Institute of Paediatrics and Puericultura Martagão Gesteira (IPPMG), Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-612, Brazil; (P.M.F.); (M.C.C.); (D.V.L.); (F.V.M.); (L.S.); (A.L.P.)
| | - Fabiana V. Mello
- Cytometry Service, Institute of Paediatrics and Puericultura Martagão Gesteira (IPPMG), Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-612, Brazil; (P.M.F.); (M.C.C.); (D.V.L.); (F.V.M.); (L.S.); (A.L.P.)
| | - Patrícia F. R. Siqueira
- Internal Medicine Postgraduate Program, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil; (C.d.S.F.-F.); (V.B.); (L.C.T.); (E.O.); (E.B.-R.); (P.F.R.S.); (M.G.P.L.)
- Cytometry Service, Institute of Paediatrics and Puericultura Martagão Gesteira (IPPMG), Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-612, Brazil; (P.M.F.); (M.C.C.); (D.V.L.); (F.V.M.); (L.S.); (A.L.P.)
| | - Patrícia Moura
- I’Dor Institute, Hospital Estadual da Criança, Rio de Janeiro 21330-400, Brazil; (P.M.); (F.N.M.)
| | - Francisco Nicanor Macedo
- I’Dor Institute, Hospital Estadual da Criança, Rio de Janeiro 21330-400, Brazil; (P.M.); (F.N.M.)
| | - Danielle N. Forny
- Department of Pediatric Surgery, Institute of Paediatrics and Puericultura Martagão Gesteira (IPPMG), Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-612, Brazil;
| | - Luíza Simião
- Cytometry Service, Institute of Paediatrics and Puericultura Martagão Gesteira (IPPMG), Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-612, Brazil; (P.M.F.); (M.C.C.); (D.V.L.); (F.V.M.); (L.S.); (A.L.P.)
| | - Ana Luíza Pureza
- Cytometry Service, Institute of Paediatrics and Puericultura Martagão Gesteira (IPPMG), Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-612, Brazil; (P.M.F.); (M.C.C.); (D.V.L.); (F.V.M.); (L.S.); (A.L.P.)
| | - Marcelo Gerardin Poirot Land
- Internal Medicine Postgraduate Program, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil; (C.d.S.F.-F.); (V.B.); (L.C.T.); (E.O.); (E.B.-R.); (P.F.R.S.); (M.G.P.L.)
| | - Carlos Eduardo Pedreira
- Systems and Computing Engineering Department (COPPE-PESC), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-972, Brazil;
| | - Jacques J. M. van Dongen
- Department of Immunohematology and Blood Transfusion (IHB), Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands;
| | - Alberto Orfao
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and IBMCC (CSIC-University of Salamanca), Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca (USAL), Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Correspondence: (A.O.); (E.S.d.C.); Tel.: +34-9232-9481 (A.O.); +55-21-3938-4725 (E.S.d.C.)
| | - Elaine Sobral da Costa
- Internal Medicine Postgraduate Program, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-617, Brazil; (C.d.S.F.-F.); (V.B.); (L.C.T.); (E.O.); (E.B.-R.); (P.F.R.S.); (M.G.P.L.)
- Cytometry Service, Institute of Paediatrics and Puericultura Martagão Gesteira (IPPMG), Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-612, Brazil; (P.M.F.); (M.C.C.); (D.V.L.); (F.V.M.); (L.S.); (A.L.P.)
- Correspondence: (A.O.); (E.S.d.C.); Tel.: +34-9232-9481 (A.O.); +55-21-3938-4725 (E.S.d.C.)
| |
Collapse
|
26
|
Patient Endothelial Colony-Forming Cells to Model Coronary Artery Disease Susceptibility and Unravel the Role of Dysregulated Mitochondrial Redox Signalling. Antioxidants (Basel) 2021; 10:antiox10101547. [PMID: 34679682 PMCID: PMC8532880 DOI: 10.3390/antiox10101547] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 09/25/2021] [Accepted: 09/27/2021] [Indexed: 01/02/2023] Open
Abstract
Mechanisms involved in the individual susceptibility to atherosclerotic coronary artery disease (CAD) beyond traditional risk factors are poorly understood. Here, we describe the utility of cultured patient-derived endothelial colony-forming cells (ECFCs) in examining novel mechanisms of CAD susceptibility, particularly the role of dysregulated redox signalling. ECFCs were selectively cultured from peripheral blood mononuclear cells from 828 patients from the BioHEART-CT cohort, each with corresponding demographic, clinical and CT coronary angiographic imaging data. Spontaneous growth occurred in 178 (21.5%) patients and was more common in patients with hypertension (OR 1.45 (95% CI 1.03-2.02), p = 0.031), and less likely in patients with obesity (OR 0.62 [95% CI 0.40-0.95], p = 0.027) or obstructive CAD (stenosis > 50%) (OR 0.60 [95% CI 0.38-0.95], p = 0.027). ECFCs from patients with CAD had higher mitochondrial production of superoxide (O2--MitoSOX assay). The latter was strongly correlated with the severity of CAD as measured by either coronary artery calcium score (R2 = 0.46; p = 0.0051) or Gensini Score (R2 = 0.67; p = 0.0002). Patient-derived ECFCs were successfully cultured in 3D culture pulsatile mini-vessels. Patient-derived ECFCs can provide a novel resource for discovering mechanisms of CAD disease susceptibility, particularly in relation to mitochondrial redox signalling.
Collapse
|
27
|
Ewald ML, Chen YH, Lee AP, Hughes CCW. The vascular niche in next generation microphysiological systems. LAB ON A CHIP 2021; 21:3244-3262. [PMID: 34396383 PMCID: PMC8635227 DOI: 10.1039/d1lc00530h] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
In recent years, microphysiological system (MPS, also known as, organ-on-a-chip or tissue chip) platforms have emerged with great promise to improve the predictive capacity of preclinical modeling thereby reducing the high attrition rates when drugs move into trials. While their designs can vary quite significantly, in general MPS are bioengineered in vitro microenvironments that recapitulate key functional units of human organs, and that have broad applications in human physiology, pathophysiology, and clinical pharmacology. A critical next step in the evolution of MPS devices is the widespread incorporation of functional vasculature within tissues. The vasculature itself is a major organ that carries nutrients, immune cells, signaling molecules and therapeutics to all other organs. It also plays critical roles in inducing and maintaining tissue identity through expression of angiocrine factors, and in providing tissue-specific milieus (i.e., the vascular niche) that can support the survival and function of stem cells. Thus, organs are patterned, maintained and supported by the vasculature, which in turn receives signals that drive tissue specific gene expression. In this review, we will discuss published vascularized MPS platforms and present considerations for next-generation devices looking to incorporate this critical constituent. Finally, we will highlight the organ-patterning processes governed by the vasculature, and how the incorporation of a vascular niche within MPS platforms will establish a unique opportunity to study stem cell development.
Collapse
Affiliation(s)
- Makena L Ewald
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697, USA.
| | | | | | | |
Collapse
|
28
|
Dawson A, Wang Y, Li Y, LeMaire SA, Shen YH. New Technologies With Increased Precision Improve Understanding of Endothelial Cell Heterogeneity in Cardiovascular Health and Disease. Front Cell Dev Biol 2021; 9:679995. [PMID: 34513826 PMCID: PMC8430032 DOI: 10.3389/fcell.2021.679995] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/17/2021] [Indexed: 01/08/2023] Open
Abstract
Endothelial cells (ECs) are vital for blood vessel integrity and have roles in maintaining normal vascular function, healing after injury, and vascular dysfunction. Extensive phenotypic heterogeneity has been observed among ECs of different types of blood vessels in the normal and diseased vascular wall. Although ECs with different phenotypes can share common functions, each has unique features that may dictate a fine-tuned role in vascular health and disease. Recent studies performed with single-cell technology have generated powerful information that has significantly improved our understanding of EC biology. Here, we summarize a variety of EC types, states, and phenotypes recently identified by using new, increasingly precise techniques in transcriptome analysis.
Collapse
Affiliation(s)
- Ashley Dawson
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Yidan Wang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Yanming Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Scott A. LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX, United States
| | - Ying H. Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX, United States
| |
Collapse
|
29
|
Tan CMJ, Lewandowski AJ, Williamson W, Huckstep OJ, Yu GZ, Fischer R, Simon JN, Alsharqi M, Mohamed A, Leeson P, Bertagnolli M. Proteomic Signature of Dysfunctional Circulating Endothelial Colony-Forming Cells of Young Adults. J Am Heart Assoc 2021; 10:e021119. [PMID: 34275329 PMCID: PMC8475699 DOI: 10.1161/jaha.121.021119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022]
Abstract
Background A subpopulation of endothelial progenitor cells called endothelial colony-forming cells (ECFCs) may offer a platform for cellular assessment in clinical studies because of their remarkable angiogenic and expansion potentials in vitro. Despite endothelial cell function being influenced by cardiovascular risk factors, no studies have yet provided a comprehensive proteomic profile to distinguish functional (ie, more angiogenic and expansive cells) versus dysfunctional circulating ECFCs of young adults. The aim of this study was to provide a detailed proteomic comparison between functional and dysfunctional ECFCs. Methods and Results Peripheral blood ECFCs were isolated from 11 subjects (45% men, aged 27±5 years) using Ficoll density gradient centrifugation. ECFCs expressed endothelial and progenitor surface markers and displayed cobblestone-patterned morphology with clonal and angiogenic capacities in vitro. ECFCs were deemed dysfunctional if <1 closed tube formed during the in vitro tube formation assay and proliferation rate was <20%. Hierarchical functional clustering revealed distinct ECFC proteomic signatures between functional and dysfunctional ECFCs with changes in cellular mechanisms involved in exocytosis, vesicle transport, extracellular matrix organization, cell metabolism, and apoptosis. Targeted antiangiogenic proteins in dysfunctional ECFCs included SPARC (secreted protein acidic and rich in cysteine), CD36 (cluster of differentiation 36), LUM (lumican), and PTX3 (pentraxin-related protein PYX3). Conclusions Circulating ECFCs with impaired angiogenesis and expansion capacities have a distinct proteomic profile and significant phenotype changes compared with highly angiogenic endothelial cells. Impaired angiogenesis in dysfunctional ECFCs may underlie the link between endothelial dysfunction and cardiovascular disease risks in young adults.
Collapse
Affiliation(s)
- Cheryl M. J. Tan
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Adam J. Lewandowski
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Wilby Williamson
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Odaro J. Huckstep
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
- Department of BiologyUnited States Air Force AcademyColorado SpringsCOUSA
| | - Grace Z. Yu
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
- Wellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Roman Fischer
- Target Discovery Institute (TDI) Mass Spectrometry Laboratory, Target Discovery Institute, Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Jillian N. Simon
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUnited Kingdom
| | - Maryam Alsharqi
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
- Department of Cardiac TechnologyImam Abdulrahman Bin Faisal UniversityDammamSaudi Arabia
| | - Afifah Mohamed
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
- Department of Diagnostic Imaging & Applied Health Sciences, Faculty of Health SciencesUniversiti Kebangsaan MalaysiaKuala LumpurMalaysia
| | - Paul Leeson
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Mariane Bertagnolli
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
- Montreal Hospital Sacré‐Cœur Research CentreCentre Intégré Universitaire de Santé et de Services Sociaux du Nord‐de‐l'Île‐de‐MontréalMontréalQCCanada
- School of Physical and Occupational Therapy, Faculty of MedicineMcGill UniversityMontréalQCCanada
| |
Collapse
|
30
|
Vašíček J, Baláži A, Bauer M, Svoradová A, Tirpáková M, Tomka M, Chrenek P. Molecular Profiling and Gene Banking of Rabbit EPCs Derived from Two Biological Sources. Genes (Basel) 2021; 12:genes12030366. [PMID: 33806502 PMCID: PMC7998175 DOI: 10.3390/genes12030366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/27/2021] [Accepted: 03/03/2021] [Indexed: 12/20/2022] Open
Abstract
Endothelial progenitor cells (EPCs) have been broadly studied for several years due to their outstanding regenerative potential. Moreover, these cells might be a valuable source of genetic information for the preservation of endangered animal species. However, a controversy regarding their characterization still exists. The aim of this study was to isolate and compare the rabbit peripheral blood- and bone marrow-derived EPCs with human umbilical vein endothelial cells (HUVECs) in terms of their phenotype and morphology that could be affected by the passage number or cryopreservation as well as to assess their possible neuro-differentiation potential. Briefly, cells were isolated and cultured under standard endothelial conditions until passage 3. The morphological changes during the culture were monitored and each passage was analyzed for the typical phenotype using flow cytometry, quantitative real–time polymerase chain reaction (qPCR) and novel digital droplet PCR (ddPCR), and compared to HUVECs. The neurogenic differentiation was induced using a commercial kit. Rabbit cells were also cryopreserved for at least 3 months and then analyzed after thawing. According to the obtained results, both rabbit EPCs exhibit a spindle-shaped morphology and high proliferation rate. The both cell lines possess same stable phenotype: CD14−CD29+CD31−CD34−CD44+CD45−CD49f+CD73+CD90+CD105+CD133−CD146−CD166+VE-cadherin+VEGFR-2+SSEA-4+MSCA-1−vWF+eNOS+AcLDL+ALDH+vimentin+desmin+α-SMA+, slightly different from HUVECs. Moreover, both induced rabbit EPCs exhibit neuron-like morphological changes and expression of neuronal markers ENO2 and MAP2. In addition, cryopreserved rabbit cells maintained high viability (>85%) and endothelial phenotype after thawing. In conclusion, our findings suggest that cells expanded from the rabbit peripheral blood and bone marrow are of the endothelial origin with a stable marker expression and interesting proliferation and differentiation capacity.
Collapse
Affiliation(s)
- Jaromír Vašíček
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
- Department of Biochemistry and Biotechnology, Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia;
- Correspondence: (J.V.); (P.C.); Tel.: +421-37-654-6600 (J.V.); +421-37-641-4274 (P.C.)
| | - Andrej Baláži
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
| | - Miroslav Bauer
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
- Department of Botany and Genetics, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, Nábrežie mládeže 91, 949 74 Nitra, Slovakia
| | - Andrea Svoradová
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
| | - Mária Tirpáková
- Department of Biochemistry and Biotechnology, Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia;
- AgroBioTech Research Center, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia
| | - Marián Tomka
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
| | - Peter Chrenek
- NPPC, Research Institute for Animal Production Nitra, Institute of Farm Animal Genetics and Reproduction, Hlohovecká 2, 951 41 Lužianky, Slovakia; (A.B.); (M.B.); (A.S.); (M.T.)
- Department of Biochemistry and Biotechnology, Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia;
- Correspondence: (J.V.); (P.C.); Tel.: +421-37-654-6600 (J.V.); +421-37-641-4274 (P.C.)
| |
Collapse
|
31
|
Co-Culture of Primary Human Coronary Artery and Internal Thoracic Artery Endothelial Cells Results in Mutually Beneficial Paracrine Interactions. Int J Mol Sci 2020; 21:ijms21218032. [PMID: 33126651 PMCID: PMC7663246 DOI: 10.3390/ijms21218032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/21/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022] Open
Abstract
Although saphenous veins (SVs) are commonly used as conduits for coronary artery bypass grafting (CABG), internal thoracic artery (ITA) grafts have significantly higher long-term patency. As SVs and ITA endothelial cells (ECs) have a considerable level of heterogeneity, we suggested that synergistic paracrine interactions between CA and ITA ECs (HCAECs and HITAECs, respectively) may explain the increased resistance of ITA grafts and adjacent CAs to atherosclerosis and restenosis. In this study, we measured the gene and protein expression of the molecules responsible for endothelial homeostasis, pro-inflammatory response, and endothelial-to-mesenchymal transition in HCAECs co-cultured with either HITAECs or SV ECs (HSaVECs) for an ascending duration. Upon the co-culture, HCAECs and HITAECs showed augmented expression of endothelial nitric oxide synthase (eNOS) and reduced expression of endothelial-to-mesenchymal transition transcription factors Snail and Slug when compared to the HCAEC–HSaVEC model. HCAECs co-cultured with HITAECs demonstrated an upregulation of HES1, a master regulator of arterial specification, of which the expression was also exclusively induced in HSaVECs co-cultured with HCAECs, suggestive of their arterialisation. In addition, co-culture of HCAECs and HITAECs promoted the release of pro-angiogenic molecules. To conclude, co-culture of HCAECs and HITAECs results in reciprocal and beneficial paracrine interactions that might contribute to the better performance of ITA grafts upon CABG.
Collapse
|
32
|
Therapeutic Potential of Endothelial Colony-Forming Cells in Ischemic Disease: Strategies to Improve their Regenerative Efficacy. Int J Mol Sci 2020; 21:ijms21197406. [PMID: 33036489 PMCID: PMC7582994 DOI: 10.3390/ijms21197406] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/02/2020] [Accepted: 10/02/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease (CVD) comprises a range of major clinical cardiac and circulatory diseases, which produce immense health and economic burdens worldwide. Currently, vascular regenerative surgery represents the most employed therapeutic option to treat ischemic disorders, even though not all the patients are amenable to surgical revascularization. Therefore, more efficient therapeutic approaches are urgently required to promote neovascularization. Therapeutic angiogenesis represents an emerging strategy that aims at reconstructing the damaged vascular network by stimulating local angiogenesis and/or promoting de novo blood vessel formation according to a process known as vasculogenesis. In turn, circulating endothelial colony-forming cells (ECFCs) represent truly endothelial precursors, which display high clonogenic potential and have the documented ability to originate de novo blood vessels in vivo. Therefore, ECFCs are regarded as the most promising cellular candidate to promote therapeutic angiogenesis in patients suffering from CVD. The current briefly summarizes the available information about the origin and characterization of ECFCs and then widely illustrates the preclinical studies that assessed their regenerative efficacy in a variety of ischemic disorders, including acute myocardial infarction, peripheral artery disease, ischemic brain disease, and retinopathy. Then, we describe the most common pharmacological, genetic, and epigenetic strategies employed to enhance the vasoreparative potential of autologous ECFCs by manipulating crucial pro-angiogenic signaling pathways, e.g., extracellular-signal regulated kinase/Akt, phosphoinositide 3-kinase, and Ca2+ signaling. We conclude by discussing the possibility of targeting circulating ECFCs to rescue their dysfunctional phenotype and promote neovascularization in the presence of CVD.
Collapse
|
33
|
Martínez-González J, García de Frutos P. Cells in Cardiovascular Disease: Using Diversity to Confront Adversity. Cells 2020; 9:cells9102192. [PMID: 33003290 PMCID: PMC7600927 DOI: 10.3390/cells9102192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 09/16/2020] [Indexed: 11/16/2022] Open
Abstract
The present Special Issue on "Cells in Cardiovascular Disease" wants to offer a general overview of current cardiovascular research and illustrate how advances in the molecular characterization at the cellular level are providing unique insights into pathologies of the circulatory system [...].
Collapse
Affiliation(s)
- José Martínez-González
- Institute for Biomedical Research of Barcelona, IIBB-CSIC, 08036 Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institut d’Investigacions Biomèdiques Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- Correspondence: (J.M.-G.); (P.G.d.F.)
| | - Pablo García de Frutos
- Institute for Biomedical Research of Barcelona, IIBB-CSIC, 08036 Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Correspondence: (J.M.-G.); (P.G.d.F.)
| |
Collapse
|