1
|
Hershenson R, Nardi-Agmon I, Leshem-Lev D, Kornowski R, Eisen A. The effect of empagliflozin on circulating endothelial progenitor cells in patients with diabetes and stable coronary artery disease. Cardiovasc Diabetol 2024; 23:386. [PMID: 39468546 PMCID: PMC11520434 DOI: 10.1186/s12933-024-02466-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Diabetes mellitus (DM) is associated with premature atherosclerotic disease, coronary artery disease (CAD) and chronic heart failure (HF), leading to increased morbidity and mortality. Sodium-Glucose Co-transporter 2 Inhibitors (SGLT2i) exhibit cardioprotective benefits beyond glucose lowering, reducing the risk of major cardiovascular events (MACE) and HF hospitalizations in patients with DM and CAD. Endothelial progenitor cells (EPCs) are bone marrow-derived cells involved in vascular repair, mobilized in response to vascular injury. The number and function of circulating EPCs (cEPCs) are negatively affected by cardiovascular risk factors, including DM. This study aimed to examine the response of cEPCs to SGLT2i treatment in DM patients with stable CAD. METHODS A prospective single-center study included patients with DM and stable CAD who were started on an SGLT2i (empagliflozin). Peripheral blood samples were collected at baseline, 1 month, and 3 months to evaluate cEPC levels and function by flow cytometry, immunohistochemistry and MTT assays. RESULTS Eighteen patients were included in the study (median age 73, (IQR 69, 77) years, 67% male). After 1 month of treatment with empagliflozin, there was no significant change in cEPCs level or function. However, following 3 months of treatment, a significant increase was observed both in cell levels (CD34(+)/VEGFR-2(+): from 0.49% (IQR 0.32, 0.64) to 1.58% (IQR 0.93, 1.82), p = 0.0006; CD133(+)/VEGFR-2(+): from 0.38% (IQR 0.27, 0.6) to 0.82% (IQR 0.7, 1.95), p = 0.0001) and in cell function (from 0.25 CFUs (IQR 0, 0.5) at baseline, to 2 CFUs (IQR 1, 2) at 3 months, p = 0.0012). CONCLUSIONS Empagliflozin treatment in patients with DM and stable CAD increases cEPC levels and function, implying a cardioprotective mechanism. These findings highlight the potential of SGLT2i in treating cardiovascular diseases, warranting further research to explore these effects and their long-term implications.
Collapse
Affiliation(s)
- Roy Hershenson
- Department of Cardiology, Rabin Medical Center, 39 Jabotinsky St., 49100, Petah Tikva, Israel.
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Inbar Nardi-Agmon
- Department of Cardiology, Rabin Medical Center, 39 Jabotinsky St., 49100, Petah Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
| | - Dorit Leshem-Lev
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
| | - Ran Kornowski
- Department of Cardiology, Rabin Medical Center, 39 Jabotinsky St., 49100, Petah Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alon Eisen
- Department of Cardiology, Rabin Medical Center, 39 Jabotinsky St., 49100, Petah Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
2
|
Hussein KH, Ahmadzada B, Correa JC, Sultan A, Wilken S, Amiot B, Nyberg SL. Liver tissue engineering using decellularized scaffolds: Current progress, challenges, and opportunities. Bioact Mater 2024; 40:280-305. [PMID: 38973992 PMCID: PMC11226731 DOI: 10.1016/j.bioactmat.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 07/09/2024] Open
Abstract
Liver transplantation represents the only definitive treatment for patients with end-stage liver disease. However, the shortage of liver donors provokes a dramatic gap between available grafts and patients on the waiting list. Whole liver bioengineering, an emerging field of tissue engineering, holds great potential to overcome this gap. This approach involves two main steps; the first is liver decellularization and the second is recellularization. Liver decellularization aims to remove cellular and nuclear materials from the organ, leaving behind extracellular matrices containing different structural proteins and growth factors while retaining both the vascular and biliary networks. Recellularization involves repopulating the decellularized liver with appropriate cells, theoretically from the recipient patient, to reconstruct the parenchyma, vascular tree, and biliary network. The aim of this review is to identify the major advances in decellularization and recellularization strategies and investigate obstacles for the clinical application of bioengineered liver, including immunogenicity of the designed liver extracellular matrices, the need for standardization of scaffold fabrication techniques, selection of suitable cell sources for parenchymal repopulation, vascular, and biliary tree reconstruction. In vivo transplantation models are also summarized for evaluating the functionality of bioengineered livers. Finally, the regulatory measures and future directions for confirming the safety and efficacy of bioengineered liver are also discussed. Addressing these challenges in whole liver bioengineering may offer new solutions to meet the demand for liver transplantation and improve patient outcomes.
Collapse
Affiliation(s)
- Kamal H. Hussein
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
- Department of Surgery, Anesthesiology, and Radiology, College of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Boyukkhanim Ahmadzada
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Julio Cisneros Correa
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Ahmer Sultan
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Silvana Wilken
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Bruce Amiot
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Scott L. Nyberg
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
3
|
Das A, Smith RJ, Andreadis ST. Harnessing the potential of monocytes/macrophages to regenerate tissue-engineered vascular grafts. Cardiovasc Res 2024; 120:839-854. [PMID: 38742656 PMCID: PMC11218695 DOI: 10.1093/cvr/cvae106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/19/2024] [Accepted: 04/02/2024] [Indexed: 05/16/2024] Open
Abstract
Cell-free tissue-engineered vascular grafts provide a promising alternative to treat cardiovascular disease, but timely endothelialization is essential for ensuring patency and proper functioning post-implantation. Recent studies from our lab showed that blood cells like monocytes (MCs) and macrophages (Mϕ) may contribute directly to cellularization and regeneration of bioengineered arteries in small and large animal models. While MCs and Mϕ are leucocytes that are part of the innate immune response, they share common developmental origins with endothelial cells (ECs) and are known to play crucial roles during vessel formation (angiogenesis) and vessel repair after inflammation/injury. They are highly plastic cells that polarize into pro-inflammatory and anti-inflammatory phenotypes upon exposure to cytokines and differentiate into other cell types, including EC-like cells, in the presence of appropriate chemical and mechanical stimuli. This review focuses on the developmental origins of MCs and ECs; the role of MCs and Mϕ in vessel repair/regeneration during inflammation/injury; and the role of chemical signalling and mechanical forces in Mϕ inflammation that mediates vascular graft regeneration. We postulate that comprehensive understanding of these mechanisms will better inform the development of strategies to coax MCs/Mϕ into endothelializing the lumen and regenerate the smooth muscle layers of cell-free bioengineered arteries and veins that are designed to treat cardiovascular diseases and perhaps the native vasculature as well.
Collapse
Affiliation(s)
- Arundhati Das
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, 908 Furnas Hall, Buffalo, NY 14260-4200, USA
| | - Randall J Smith
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, 332 Bonner Hall, Buffalo, NY 14260-1920, USA
| | - Stelios T Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, 908 Furnas Hall, Buffalo, NY 14260-4200, USA
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, 332 Bonner Hall, Buffalo, NY 14260-1920, USA
- Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, The State University of New York, 701 Ellicott St, Buffalo, NY 14203, USA
- Cell, Gene and Tissue Engineering (CGTE) Center, University at Buffalo, The State University of New York, 813 Furnas Hall, Buffalo, NY 14260-4200, USA
| |
Collapse
|
4
|
Tkacz M, Zgutka K, Tomasiak P, Tarnowski M. Responses of Endothelial Progenitor Cells to Chronic and Acute Physical Activity in Healthy Individuals. Int J Mol Sci 2024; 25:6085. [PMID: 38892272 PMCID: PMC11173310 DOI: 10.3390/ijms25116085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Endothelial progenitor cells (EPCs) are circulating cells of various origins that possess the capacity for renewing and regenerating the endothelial lining of blood vessels. During physical activity, in response to factors such as hypoxia, changes in osmotic pressure, and mechanical forces, endothelial cells undergo intense physiological stress that results in endothelial damage. Circulating EPCs participate in blood vessel repair and vascular healing mainly through paracrine signalling. Furthermore, physical activity may play an important role in mobilising this important cell population. In this narrative review, we summarise the current knowledge on the biology of EPCs, including their characteristics, assessment, and mobilisation in response to both chronic and acute physical activity in healthy individuals.
Collapse
Affiliation(s)
- Marta Tkacz
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, Zolnierska 48, 70-210 Szczecin, Poland
| | - Katarzyna Zgutka
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, Zolnierska 48, 70-210 Szczecin, Poland
| | - Patrycja Tomasiak
- Institute of Physical Culture Sciences, University of Szczecin, 70-453 Szczecin, Poland
| | - Maciej Tarnowski
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, Zolnierska 48, 70-210 Szczecin, Poland
- Institute of Physical Culture Sciences, University of Szczecin, 70-453 Szczecin, Poland
| |
Collapse
|
5
|
Santillán-Cortez D, Vera-Gómez E, Hernández-Patricio A, Ruíz-Hernández AS, Gutiérrez-Buendía JA, De la Vega-Moreno K, Rizo-García YA, Loman-Zuñiga OA, Escotto-Sánchez I, Rodríguez-Trejo JM, Téllez-González MA, Toledo-Lozano CG, Ortega-Rosas T, García S, Mondragón-Terán P, Suárez-Cuenca JA. Endothelial Progenitor Cells May Be Related to Major Amputation after Angioplasty in Patients with Critical Limb Ischemia. Cells 2023; 12:cells12040584. [PMID: 36831250 PMCID: PMC9954311 DOI: 10.3390/cells12040584] [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: 08/30/2022] [Revised: 01/28/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Critical limb ischemia represents an advanced stage of peripheral arterial disease. Angioplasty improves blood flow to the limb; however, some patients progress irreversibly to lower limb amputation. Few studies have explored the predictive potential of biomarkers during postangioplasty outcomes. AIM To evaluate the behavior of endothelial progenitor cells in patients with critical limb ischemia, in relation to their postangioplasty outcome. METHODS Twenty patients with critical limb ischemia, candidates for angioplasty, were enrolled. Flow-mediated dilation, as well as endothelial progenitor cells (subpopulations CD45+/CD34+/CD133+/CD184+ and CD45+/CD/34+/KDR[VEGFR-2]+ estimated by flow cytometry) from blood flow close to vascular damage, were evaluated before and after angioplasty. Association with lower limb amputation during a 30-day follow-up was analyzed. RESULTS Endothelial progenitor cells were related with flow-mediated dilation. A higher number of baseline EPCs CD45+CD34+KDR+, as well as an impaired reactivity of endothelial progenitor cells CD45+CD34+CD133+CD184+ after angioplasty, were observed in cases further undergoing major limb amputation, with a significant discrimination ability and risk (0.75, specificity 0.83 and RR 4.5 p < 0.05). CONCLUSIONS Endothelial progenitor cells were related with endothelial dysfunction, whereas a higher baseline number of the subpopulation CD45+CD34+KDR+, as well as an impaired reactivity of subpopulation CD45+CD34+CD133+CD184+ after angioplasty, showed a predictive ability for major limb amputation in patients with critical limb ischemia.
Collapse
Affiliation(s)
- Daniel Santillán-Cortez
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
- Vascular Surgery and Angiology Department, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
- Regenerative Medicine and Tissue Engineering Laboratory, Coordination of Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Eduardo Vera-Gómez
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Alejandro Hernández-Patricio
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Atzín Suá Ruíz-Hernández
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Juan Ariel Gutiérrez-Buendía
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Karen De la Vega-Moreno
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Yasser Alberto Rizo-García
- Vascular Surgery and Angiology Department, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Oscar Antonio Loman-Zuñiga
- Vascular Surgery and Angiology Department, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Ignacio Escotto-Sánchez
- Vascular Surgery and Angiology Department, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Juan Miguel Rodríguez-Trejo
- Vascular Surgery and Angiology Department, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Mario Antonio Téllez-González
- Regenerative Medicine and Tissue Engineering Laboratory, Coordination of Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Christian Gabriel Toledo-Lozano
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Tania Ortega-Rosas
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Silvia García
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Paul Mondragón-Terán
- Regenerative Medicine and Tissue Engineering Laboratory, Coordination of Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Juan Antonio Suárez-Cuenca
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
- Correspondence: ; Tel.: +5255-52005003 (ext. 14661)
| |
Collapse
|
6
|
Kerch G. Severe COVID-19-A Review of Suggested Mechanisms Based on the Role of Extracellular Matrix Stiffness. Int J Mol Sci 2023; 24:1187. [PMID: 36674700 PMCID: PMC9861790 DOI: 10.3390/ijms24021187] [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: 11/14/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
The severity of COVID-19 commonly depends on age-related tissue stiffness. The aim was to review publications that explain the effect of microenvironmental extracellular matrix stiffness on cellular processes. Platelets and endothelial cells are mechanosensitive. Increased tissue stiffness can trigger cytokine storm with the upregulated expression of pro-inflammatory cytokines, such as tumor necrosis factor alpha and interleukin IL-6, and tissue integrity disruption, leading to enhanced virus entry and disease severity. Increased tissue stiffness in critically ill COVID-19 patients triggers platelet activation and initiates plague formation and thrombosis development. Cholesterol content in cell membrane increases with aging and further enhances tissue stiffness. Membrane cholesterol depletion decreases virus entry to host cells. Membrane cholesterol lowering drugs, such as statins or novel chitosan derivatives, have to be further developed for application in COVID-19 treatment. Statins are also known to decrease arterial stiffness mitigating cardiovascular diseases. Sulfated chitosan derivatives can be further developed for potential use in future as anticoagulants in prevention of severe COVID-19. Anti-TNF-α therapies as well as destiffening therapies have been suggested to combat severe COVID-19. The inhibition of the nuclear factor kappa-light-chain-enhancer of activated B cells pathway must be considered as a therapeutic target in the treatment of severe COVID-19 patients. The activation of mechanosensitive platelets by higher matrix stiffness increases their adhesion and the risk of thrombus formation, thus enhancing the severity of COVID-19.
Collapse
Affiliation(s)
- Garry Kerch
- Faculty of Materials Science and Applied Chemistry, Riga Technical University, 1048 Riga, Latvia
| |
Collapse
|
7
|
Wiessman M, Kheifets M, Schamroth Pravda N, Leshem Lev D, Ziv E, Kornowski R, Spectre G, Perl L. Thrombogenicity and endothelial progenitor cells function during Acute myocardial infarction - comparison of Prasugrel versus Ticagrelor. J Thromb Thrombolysis 2023; 55:407-414. [PMID: 36598739 PMCID: PMC9811044 DOI: 10.1007/s11239-022-02759-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/18/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Thrombin generation (TG), platelet function and circulating endothelial progenitor cells (EPCs) have an important role in the pathophysiology of coronary artery disease (CAD). To date, the effect of novel P2Y12 inhibitors on these aspects has mostly been studied in the sub-acute phase following myocardial infarction. OBJECTIVES Comparing the effects of prasugrel and ticagrelor on TG and EPCs in the acute phase of ST-segment elevation myocardial infarction (STEMI). METHODS STEMI patients were randomized to either ticagrelor or prasugrel treatment. TG, platelet reactivity and EPCs were evaluated prior to P2Y12 inhibitor loading dose (T0), and one day following (T1). RESULTS Between December 2018 - July 2021, 83 consecutive STEMI patients were randomized to ticagrelor (N = 42) or prasugrel (N = 41) treatment. No differences were observed at T0 for all measurements. P2Y12 reactivity units (PRU) at T1 did not differ as well (prasugrel 13.2 [5.5-20.8] vs. ticagrelor 15.8 [4.0-26.3], p = 0.40). At T1, prasugrel was a significantly more potent TG inhibitor, with longer lag time to TG initiation (7.7 ± 7.5 vs. 3.9 ± 2.1 min, p < 0.01), longer time to peak (14.1 ± 12.6 vs. 8.3 ± 9.7 min, p = 0.03) and a lower endogenous thrombin potential (AUC 2186.1 ± 1123.1 vs. 3362.5 ± 2108.5 nM, p < 0.01). Furthermore, EPCs measured by percentage of cells expressing CD34 (2.6 ± 4.1 vs. 1.1 ± 1.1, p = 0.01) and CD133 (2.3 ± 1.8 vs. 1.4 ± 1.5, p = 0.01) and number of colony forming units (CFU, 2.1 ± 1.5 vs. 1.1 ± 1.0, p < 0.01) were significantly higher in the prasugrel group. CONCLUSION Among STEMI patients, prasugrel as compared to ticagrelor was associated with more potent TG inhibition and improved EPCs count and function.
Collapse
Affiliation(s)
- Maya Wiessman
- grid.413156.40000 0004 0575 344XDepartment of Cardiology, Rabin Medical Center, Beilinson Campus, 39 Ze’ev Jabotinsky St, 4941492 Petach Tikva, Israel ,grid.12136.370000 0004 1937 0546The Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mark Kheifets
- grid.413156.40000 0004 0575 344XDepartment of Cardiology, Rabin Medical Center, Beilinson Campus, 39 Ze’ev Jabotinsky St, 4941492 Petach Tikva, Israel ,grid.12136.370000 0004 1937 0546The Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nili Schamroth Pravda
- grid.413156.40000 0004 0575 344XDepartment of Cardiology, Rabin Medical Center, Beilinson Campus, 39 Ze’ev Jabotinsky St, 4941492 Petach Tikva, Israel ,grid.12136.370000 0004 1937 0546The Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dorit Leshem Lev
- grid.12136.370000 0004 1937 0546The Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel ,grid.413156.40000 0004 0575 344XFelsenstein Research Center, Rabin Medical Center, Petah-Tikva, Israel
| | - Eti Ziv
- grid.413156.40000 0004 0575 344XThrombosis and Haemostasis Unit, Institute of Hematology, Rabin Medical Center, Beilinson Campus, Petah-Tikva, Israel
| | - Ran Kornowski
- grid.413156.40000 0004 0575 344XDepartment of Cardiology, Rabin Medical Center, Beilinson Campus, 39 Ze’ev Jabotinsky St, 4941492 Petach Tikva, Israel ,grid.12136.370000 0004 1937 0546The Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel ,grid.413156.40000 0004 0575 344XFelsenstein Research Center, Rabin Medical Center, Petah-Tikva, Israel
| | - Galia Spectre
- grid.12136.370000 0004 1937 0546The Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel ,grid.413156.40000 0004 0575 344XThrombosis and Haemostasis Unit, Institute of Hematology, Rabin Medical Center, Beilinson Campus, Petah-Tikva, Israel
| | - Leor Perl
- grid.413156.40000 0004 0575 344XDepartment of Cardiology, Rabin Medical Center, Beilinson Campus, 39 Ze’ev Jabotinsky St, 4941492 Petach Tikva, Israel ,grid.12136.370000 0004 1937 0546The Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel ,grid.413156.40000 0004 0575 344XFelsenstein Research Center, Rabin Medical Center, Petah-Tikva, Israel
| |
Collapse
|
8
|
Moazzami K, Mehta A, Young A, Dhindsa DS, Martin G, Mokhtari A, Hesaroieh IG, Shah A, Bremner JD, Vaccarino V, Waller EK, Quyyumi AA. The association between baseline circulating progenitor cells and vascular function: The role of aging and risk factors. Vasc Med 2022; 27:532-541. [PMID: 36062298 PMCID: PMC10150400 DOI: 10.1177/1358863x221116411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND To investigate the cross-sectional and longitudinal relationships between vascular function and circulating progenitor cell (CPC) counts with respect to aging and exposure to risk factors. METHODS In 797 adult participants, CPCs were enumerated by flow cytometry as CD45med mononuclear cells expressing CD34 epitope and its subsets co-expressing CD133, and chemokine C-X-C motif receptor 4 (CXCR4+). Arterial stiffness was evaluated by tonometry-derived pulse wave velocity (PWV) and microvascular function was assessed as digital reactive hyperemia index (RHI). RESULTS In cross-sectional analyses, for every doubling in CD34+ cell counts, PWV was 15% higher and RHI was 9% lower, after adjusting for baseline characteristics and risk factors (p for all < 0.01). There were significant CPC-by-age-by-risk factor interactions (p <0.05) for both vascular measures. Among younger subjects (< 48 years), CPC counts were higher in those with risk factors and vascular function was better in those with higher compared to those with lower CPC counts (p for all < 0.0l). In contrast, in older participants, CPCs were not higher in those with risk factors, and vascular function was worse compared to the younger age group. A lower CPC count at baseline was an independent predictor of worsening vascular function during 2-year follow-up. CONCLUSION A higher CPC count in the presence of risk factors is associated with better vascular function among younger individuals. There is no increase in CPC count with risk factors in older individuals who have worse vascular function. Moreover, a higher CPC count is associated with less vascular dysfunction with aging.
Collapse
Affiliation(s)
- Kasra Moazzami
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Anurag Mehta
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - An Young
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Devinder Singh Dhindsa
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Greg Martin
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Ali Mokhtari
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Iraj Ghaini Hesaroieh
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Amit Shah
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, GA, USA
| | - J Douglas Bremner
- Atlanta VA Medical Center, Decatur, GA, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Viola Vaccarino
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Edmund K Waller
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Arshed A Quyyumi
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
9
|
Exarchos V, Zacharova E, Neuber S, Giampietro C, Motta SE, Hinkov H, Emmert MY, Nazari-Shafti TZ. The path to a hemocompatible cardiovascular implant: Advances and challenges of current endothelialization strategies. Front Cardiovasc Med 2022; 9:971028. [PMID: 36186971 PMCID: PMC9515323 DOI: 10.3389/fcvm.2022.971028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular (CV) implants are still associated with thrombogenicity due to insufficient hemocompatibility. Endothelialization of their luminal surface is a promising strategy to increase their hemocompatibility. In this review, we provide a collection of research studies and review articles aiming to summarize the recent efforts on surface modifications of CV implants, including stents, grafts, valves, and ventricular assist devises. We focus in particular on the implementation of micrometer or nanoscale surface modifications, physical characteristics of known biomaterials (such as wetness and stiffness), and surface morphological features (such as gratings, fibers, pores, and pits). We also review how biomechanical signals originating from the endothelial cell for surface interaction can be directed by topography engineering approaches toward the survival of the endothelium and its long-term adaptation. Finally, we summarize the regulatory and economic challenges that may prevent clinical implementation of endothelialized CV implants.
Collapse
Affiliation(s)
- Vasileios Exarchos
- Cardiosurgical Research Group, Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Translational Cardiovascular Regenerative Technologies Group, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
| | - Ema Zacharova
- Cardiosurgical Research Group, Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Translational Cardiovascular Regenerative Technologies Group, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
- Department of Life Sciences, IMC University of Applied Sciences Krems, Krems an der Donau, Austria
| | - Sebastian Neuber
- Cardiosurgical Research Group, Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Translational Cardiovascular Regenerative Technologies Group, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
| | - Costanza Giampietro
- Experimental Continuum Mechanics, Empa Swiss Federal Laboratories for Materials Science and Technology, Dübendorf, Switzerland
- Department of Mechanical and Process Engineering, Institute for Mechanical Systems, ETH Zürich, Zurich, Switzerland
| | - Sarah E. Motta
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Hristian Hinkov
- Cardiosurgical Research Group, Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Translational Cardiovascular Regenerative Technologies Group, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
| | - Maximilian Y. Emmert
- Cardiosurgical Research Group, Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Translational Cardiovascular Regenerative Technologies Group, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Clinic for Cardiovascular Surgery, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Timo Z. Nazari-Shafti
- Cardiosurgical Research Group, Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Translational Cardiovascular Regenerative Technologies Group, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité (Junior) (Digital) Clinician Scientist Program, Berlin, Germany
- *Correspondence: Timo Z. Nazari-Shafti,
| |
Collapse
|
10
|
Tan KX, Chang T, Lin XL. Secretomes as an emerging class of bioactive ingredients for enhanced cosmeceutical applications. Exp Dermatol 2022; 31:674-688. [PMID: 35338666 DOI: 10.1111/exd.14570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 02/23/2022] [Accepted: 03/22/2022] [Indexed: 11/30/2022]
Abstract
Skin aging is predominantly caused by either intrinsic or extrinsic factors, leading to undesirable skin features. Advancements in both molecular and cellular fields have created possibilities in developing novel stem cell-derived active ingredients for cosmeceutical applications and the beauty industry. Mesenchymal stromal cell (MSC)-derived secretomes or conditioned media hold great promise for advancing skin repair and regeneration due to the presence of varying cytokines. These cytokines signal our cells and trigger biological mechanisms associated with anti-inflammatory, antioxidant, anti-aging, proliferative, and immunomodulatory effects. In this review, we discuss the potential of MSC secretomes as novel biomaterials for skincare and rejuvenation by illustrating their mechanism of action related to wound healing, anti-aging, and whitening properties. The advantages and disadvantages of secretomes are compared to both plant-based and animal-derived extracts. In addition, this paper reviews the current safety standards, regulations, market products and research work related to the cosmeceutical applications of secretomes along with strategies to maintain and improve the therapeutic efficacy and production of secretomes. The future outlook of beauty industry is also presented. Lastly, we highlight significant challenges to be addressed for the clinical realization of MSC secretomes-based skin therapies as well as providing perspectives for the future direction of secretomes.
Collapse
Affiliation(s)
- Kei-Xian Tan
- Esco Aster, Block 67, Ayer Rajah Crescent, 139950, Singapore
| | - Trixie Chang
- Esco Aster, Block 67, Ayer Rajah Crescent, 139950, Singapore
| | - Xiang-Liang Lin
- Esco Aster, Block 67, Ayer Rajah Crescent, 139950, Singapore
| |
Collapse
|
11
|
Altabas V, Biloš LSK. The Role of Endothelial Progenitor Cells in Atherosclerosis and Impact of Anti-Lipemic Treatments on Endothelial Repair. Int J Mol Sci 2022; 23:ijms23052663. [PMID: 35269807 PMCID: PMC8910333 DOI: 10.3390/ijms23052663] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/16/2022] [Accepted: 02/26/2022] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular complications are associated with advanced atherosclerosis. Although atherosclerosis is still regarded as an incurable disease, at least in its more advanced stages, the discovery of endothelial progenitor cells (EPCs), with their ability to replace old and injured cells and differentiate into healthy and functional mature endothelial cells, has shifted our view of atherosclerosis as an incurable disease, and merged traditional theories of atherosclerosis pathogenesis with evolving concepts of vascular biology. EPC alterations are involved in the pathogenesis of vascular abnormalities in atherosclerosis, but many questions remain unanswered. Many currently available drugs that impact cardiovascular morbidity and mortality have shown a positive effect on EPC biology. This review examines the role of endothelial progenitor cells in atherosclerosis development, and the impact standard antilipemic drugs, including statins, fibrates, and ezetimibe, as well as more novel treatments such as proprotein convertase subtilisin/kexin type 9 (PCSK9) modulating agents and angiopoietin-like proteins (Angtpl3) inhibitors have on EPC biology.
Collapse
Affiliation(s)
- Velimir Altabas
- Department of Endocrinology, Diabetes and Metabolic Diseases, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia
- Correspondence: ; Tel.: +385-1-3787-692
| | | |
Collapse
|
12
|
Edlinger C, Paar V, Kheder SH, Krizanic F, Lalou E, Boxhammer E, Butter C, Dworok V, Bannehr M, Hoppe UC, Kopp K, Lichtenauer M. Endothelialization and Inflammatory Reactions After Intracardiac Device Implantation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1401:1-22. [DOI: 10.1007/5584_2022_712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
13
|
Huang C, Wen Z, Niu J, Lin S, Wang W. Steroid-Induced Osteonecrosis of the Femoral Head: Novel Insight Into the Roles of Bone Endothelial Cells in Pathogenesis and Treatment. Front Cell Dev Biol 2021; 9:777697. [PMID: 34917616 PMCID: PMC8670327 DOI: 10.3389/fcell.2021.777697] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/16/2021] [Indexed: 01/18/2023] Open
Abstract
Steroid-induced osteonecrosis of the femoral head (SONFH) is a disease characterized by the collapse of the femoral head. SONFH occurs due to the overuse of glucocorticoids (GCs) in patients with immune-related diseases. Among various pathogenesis proposed, the mechanism related to impaired blood vessels is gradually becoming the most convincing hypothesis. Bone endothelial cells including bone microvascular endothelial cells (BMECs) and endothelial progenitor cells (EPCs) play a crucial role in the maintenance of vascular homeostasis. Therefore, bone endothelial cells are key regulators in the occurrence and progression of SONFH. Impaired angiogenesis, abnormal apoptosis, thrombosis and fat embolism caused by the dysfunctions of bone endothelial cells are considered to be the pathogenesis of SONFH. In addition, even with high disability rates, SONFH lacks effective therapeutic approach. Icariin (ICA, a flavonoid extracted from Epimedii Herba), pravastatin, and VO-OHpic (a potent inhibitor of PTEN) are candidate reagents to prevent and treat SONFH through improving above pathological processes. However, these reagents are still in the preclinical stage and will not be widely used temporarily. In this case, bone tissue engineering represented by co-transplantation of bone endothelial cells and bone marrow mesenchymal stem cells (BMSCs) may be another feasible therapeutic strategy.
Collapse
Affiliation(s)
- Cheng Huang
- Department of Orthopedics, China-Japan Friendship Hospital, Beijing, China
| | - Zeqin Wen
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Junjie Niu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Subin Lin
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Weiguo Wang
- Department of Orthopedics, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
14
|
Evans WS, Sapp RM, Kim K, Heilman JM, Hagberg JM, Prior SJ. Re: Letter to the Editor on: "Effects of Exercise Training on the Paracrine Function of Circulating Angiogenic Cells.". Int J Sports Med 2021; 42:1139. [PMID: 34731896 DOI: 10.1055/a-1527-5024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- William S Evans
- Department of Kinesiology, University of Maryland School of Public Health, College Park, MD 20742, United States of America
| | - Ryan M Sapp
- Department of Kinesiology, University of Maryland School of Public Health, College Park, MD 20742, United States of America
| | - Katherine Kim
- Department of Kinesiology, University of Maryland School of Public Health, College Park, MD 20742, United States of America
| | - James M Heilman
- Department of Kinesiology, University of Maryland School of Public Health, College Park, MD 20742, United States of America
| | - James M Hagberg
- Department of Kinesiology, University of Maryland School of Public Health, College Park, MD 20742, United States of America
| | - Steven J Prior
- Department of Kinesiology, University of Maryland School of Public Health, College Park, MD 20742, United States of America.,Baltimore Veterans Affairs Geriatric Research, Education and Clinical Center, Baltimore, MD 21201, United States of America
| |
Collapse
|
15
|
Tokmakidis SP, Mitsiou G, Smilios I, Nanas S. Letter to the Editor on: "Effects of Exercise Training on the Paracrine Function of Circulating Angiogenic Cells.". Int J Sports Med 2021; 42:1137-1138. [PMID: 34731895 DOI: 10.1055/a-1527-5012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- S P Tokmakidis
- Clinical Ergophysiology and Exercise Physiology Laboratory, Department of Physical Education and Sport Science, Democritus University of Thrace, Panepistimioupoli, 69100 Komotini, Greece.,1st Critical Care Department, Evangelismos General Hospital, Department of Medicine, National and Kapodistrian University of Athens, 45-47 Ypsilantou Str., 106 75 Athens, Greece
| | - G Mitsiou
- Clinical Ergophysiology and Exercise Physiology Laboratory, Department of Physical Education and Sport Science, Democritus University of Thrace, Panepistimioupoli, 69100 Komotini, Greece.,1st Critical Care Department, Evangelismos General Hospital, Department of Medicine, National and Kapodistrian University of Athens, 45-47 Ypsilantou Str., 106 75 Athens, Greece
| | - I Smilios
- Clinical Ergophysiology and Exercise Physiology Laboratory, Department of Physical Education and Sport Science, Democritus University of Thrace, Panepistimioupoli, 69100 Komotini, Greece
| | - S Nanas
- 1st Critical Care Department, Evangelismos General Hospital, Department of Medicine, National and Kapodistrian University of Athens, 45-47 Ypsilantou Str., 106 75 Athens, Greece
| |
Collapse
|
16
|
Li C, Lin L, Zhang L, Xu R, Chen X, Ji J, Li Y. Long noncoding RNA p21 enhances autophagy to alleviate endothelial progenitor cells damage and promote endothelial repair in hypertension through SESN2/AMPK/TSC2 pathway. Pharmacol Res 2021; 173:105920. [PMID: 34601081 DOI: 10.1016/j.phrs.2021.105920] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 12/11/2022]
Abstract
Vascular damage of hypertension has been the focus of hypertension treatment, and endothelial progenitor cells (EPCs) play an important role in the repair of vascular endothelial damage. Functional damage and decreased number of EPCs are observed in the peripheral circulation of hypertensive patients, but its mechanism is not yet elucidated. Here, we show that the number of EPCs in hypertensive patients is significantly lower than that of normal population, and the cell function decreases with a higher proportion of EPCs at later stages. A decrease in autophagy is responsible for the senescence and damage of EPCs induced by AngII. Moreover, lncRNA-p21 plays a critical regulator role in EPCs' senescence and dysfunction. Furthermore, lncRNA-p21 activates SESN2/AMPK/TSC2 pathway by promoting the transcriptional activity of p53 and enhances autophagy to protect against AngII-induced EPC damage. The data provide evidence that a reversal of decreased autophagy serves as the protective mechanism of EPC injury in hypertensive patients, and lncRNA-p21 is a new therapeutic target for vascular endothelial repair in hypertension.
Collapse
Affiliation(s)
- Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lin Lin
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lei Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ran Xu
- Tianqiao District People's Hospital, Jinan 250031, China
| | - Xiaoqing Chen
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jingkang Ji
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yunlun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250000, China.
| |
Collapse
|
17
|
Razazian M, Khosravi M, Bahiraii S, Uzan G, Shamdani S, Naserian S. Differences and similarities between mesenchymal stem cell and endothelial progenitor cell immunoregulatory properties against T cells. World J Stem Cells 2021; 13:971-984. [PMID: 34567420 PMCID: PMC8422932 DOI: 10.4252/wjsc.v13.i8.971] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/28/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
Bone-marrow-derived mesenchymal stem cells and endothelial progenitor cells have some interesting biological properties that make them unique for cell therapy of degenerative and cardiovascular disorders. Although both cell populations have been already studied and used for their regenerative potentials, recently their special immunoregulatory features have brought much more attention. Mesenchymal stem cells and endothelial progenitor cells have both proangiogenic functions and have been shown to suppress the immune response, particularly T cell proliferation, activation, and cytokine production. This makes them suitable choices for allogeneic stem cell transplantation. Nevertheless, these two cells do not have equal immunoregulatory activities. Many elements including their extraction sources, age/passage, expression of different markers, secretion of bioactive mediators, and some others could change the efficiency of their immunosuppressive function. However, to our knowledge, no publication has yet compared mesenchymal stem cells and endothelial progenitor cells for their immunological interaction with T cells. This review aims to specifically compare the immunoregulatory effect of these two populations including their T cell suppression, deactivation, cytokine production, and regulatory T cells induction capacities. Moreover, it evaluates the implications of the tumor necrosis factor alpha-tumor necrosis factor receptor 2 axis as an emerging immune checkpoint signaling pathway controlling most of their immunological properties.
Collapse
Affiliation(s)
- Mehdi Razazian
- Institut national de la santé et de la recherche médicale (Inserm) Unité Mixte de Recherche-Inserm-Ministère de la Défense 1197, Hôpital Paul Brousse, Villejuif 94800, France
| | - Maryam Khosravi
- Microenvironment & Immunity Unit, Institut Pasteur, Paris 75724, France
- Institut national de la santé et de la recherche médicale (Inserm) Unit 1224, Paris 75724, France
| | - Sheyda Bahiraii
- Department of Pharmacognosy, University of Vienna, Vienna 1090, Austria
| | - Georges Uzan
- Institut national de la santé et de la recherche médicale (Inserm) Unité Mixte de Recherche-Inserm-Ministère de la Défense 1197, Hôpital Paul Brousse, Villejuif 94800, France
- Paris-Saclay University, Villejuif 94800, France
| | - Sara Shamdani
- Institut national de la santé et de la recherche médicale (Inserm) Unité Mixte de Recherche-Inserm-Ministère de la Défense 1197, Hôpital Paul Brousse, Villejuif 94800, France
- Paris-Saclay University, Villejuif 94800, France
- CellMedEx; Saint Maur Des Fossés 94100, France
| | - Sina Naserian
- Institut national de la santé et de la recherche médicale (Inserm) Unité Mixte de Recherche-Inserm-Ministère de la Défense 1197, Hôpital Paul Brousse, Villejuif 94800, France
- Paris-Saclay University, Villejuif 94800, France
- CellMedEx; Saint Maur Des Fossés 94100, France.
| |
Collapse
|
18
|
Heng JW, Yazid MD, Abdul Rahman MR, Sulaiman N. Coatings in Decellularized Vascular Scaffolds for the Establishment of a Functional Endothelium: A Scoping Review of Vascular Graft Refinement. Front Cardiovasc Med 2021; 8:677588. [PMID: 34395554 PMCID: PMC8358320 DOI: 10.3389/fcvm.2021.677588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
Developments in tissue engineering techniques have allowed for the creation of biocompatible, non-immunogenic alternative vascular grafts through the decellularization of existing tissues. With an ever-growing number of patients requiring life-saving vascular bypass grafting surgeries, the production of functional small diameter decellularized vascular scaffolds has never been more important. However, current implementations of small diameter decellularized vascular grafts face numerous clinical challenges attributed to premature graft failure as a consequence of common failure mechanisms such as acute thrombogenesis and intimal hyperplasia resulting from insufficient endothelial coverage on the graft lumen. This review summarizes some of the surface modifying coating agents currently used to improve the re-endothelialization efficiency and endothelial cell persistence in decellularized vascular scaffolds that could be applied in producing a better patency small diameter vascular graft. A comprehensive search yielding 192 publications was conducted in the PubMed, Scopus, Web of Science, and Ovid electronic databases. Careful screening and removal of unrelated publications and duplicate entries resulted in a total of 16 publications, which were discussed in this review. Selected publications demonstrate that the utilization of surface coating agents can induce endothelial cell adhesion, migration, and proliferation therefore leads to increased re-endothelialization efficiency. Unfortunately, the large variance in methodologies complicates comparison of coating effects between studies. Thus far, coating decellularized tissue gave encouraging results. These developments in re-endothelialization could be incorporated in the fabrication of functional, off-the-shelf alternative small diameter vascular scaffolds.
Collapse
Affiliation(s)
- Jun Wei Heng
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Mohd Ramzisham Abdul Rahman
- Department of Surgery, Hospital Canselor Tuanku Muhriz, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nadiah Sulaiman
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
19
|
Kumboyono K, Chomsy IN, Nurwidyaningtyas W, Cesa FY, Tjahjono CT, Wihastuti TA. Differences in senescence of late Endothelial Progenitor Cells in non-smokers and smokers. Tob Induc Dis 2021; 19:10. [PMID: 34131419 PMCID: PMC8171388 DOI: 10.18332/tid/135320] [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: 09/17/2020] [Revised: 01/30/2021] [Accepted: 03/31/2021] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Endothelial Progenitor Cells (EPCs) are part of hematopoietic stem cells that differentiate into endothelial cells during their blood vessels' maturation process. The role of EPCs is widely known to contribute to repair of the vascular wall when endothelial dysfunction occurs. However, various risk factors for cardiovascular disease (CVD) influence EPC performance, leading to endothelial dysfunction. One EPC dysfunction is decreased amount of EPC mobilization to the injured tissue. EPC dysfunction reduces the angiogenetic function of EPCs. The vital maturation process that the EPCs must pass is the late phase. The dysfunction of late EPCs is known as senescence. This study aimed to identify and compare senescence of late EPCs, through CD62E and CD41 markers, in non-smokers and smokers as a risk factor for CVD. METHODS EPC collection was from peripheral mononuclear cells (PBMCs) in non-smokers (n=30) and smokers (n=31). The EPCs were then marked by CD62E/CD41 and senescence β-galactosidase assay using FACS. Identification of senescence cells was based on fluorescence with DAPI. RESULTS Positive percentage of late EPCs in non-smokers was not significantly different from that in smokers (p=0.014). The number of senescent late EPCs in smokers was higher than in non-smokers (p<0.0001). CONCLUSIONS Endothelial progenitor cells that experienced senescence in the smokers showed EPC dysfunction, which resulted in decreased cell angiogenic function. Further research is needed to explain the mechanism of re-endothelialization failure in EPC dysfunction due to smoking.
Collapse
Affiliation(s)
- Kumboyono Kumboyono
- School of Nursing, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | | | | | | | - Cholid Tri Tjahjono
- Department of Cardiology, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Titin Andri Wihastuti
- Department of Basic Nursing Science, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| |
Collapse
|
20
|
Fang S, Ellman DG, Andersen DC. Review: Tissue Engineering of Small-Diameter Vascular Grafts and Their In Vivo Evaluation in Large Animals and Humans. Cells 2021; 10:713. [PMID: 33807009 PMCID: PMC8005053 DOI: 10.3390/cells10030713] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/10/2021] [Accepted: 03/15/2021] [Indexed: 12/15/2022] Open
Abstract
To date, a wide range of materials, from synthetic to natural or a mixture of these, has been explored, modified, and examined as small-diameter tissue-engineered vascular grafts (SD-TEVGs) for tissue regeneration either in vitro or in vivo. However, very limited success has been achieved due to mechanical failure, thrombogenicity or intimal hyperplasia, and improvements of the SD-TEVG design are thus required. Here, in vivo studies investigating novel and relative long (10 times of the inner diameter) SD-TEVGs in large animal models and humans are identified and discussed, with emphasis on graft outcome based on model- and graft-related conditions. Only a few types of synthetic polymer-based SD-TEVGs have been evaluated in large-animal models and reflect limited success. However, some polymers, such as polycaprolactone (PCL), show favorable biocompatibility and potential to be further modified and improved in the form of hybrid grafts. Natural polymer- and cell-secreted extracellular matrix (ECM)-based SD-TEVGs tested in large animals still fail due to a weak strength or thrombogenicity. Similarly, native ECM-based SD-TEVGs and in-vitro-developed hybrid SD-TEVGs that contain xenogeneic molecules or matrix seem related to a harmful graft outcome. In contrast, allogeneic native ECM-based SD-TEVGs, in-vitro-developed hybrid SD-TEVGs with allogeneic banked human cells or isolated autologous stem cells, and in-body tissue architecture (IBTA)-based SD-TEVGs seem to be promising for the future, since they are suitable in dimension, mechanical strength, biocompatibility, and availability.
Collapse
Affiliation(s)
- Shu Fang
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, J. B. Winsløwsvej 25, 5000 Odense C, Denmark; (D.G.E.); (D.C.A.)
- The Danish Regenerative Center, Odense University Hospital, J. B. Winsløwsvej 4, 5000 Odense C, Denmark
- Institute of Clinical Research, University of Southern Denmark, J. B. Winsløwsvej 19, 5000 Odense C, Denmark
| | - Ditte Gry Ellman
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, J. B. Winsløwsvej 25, 5000 Odense C, Denmark; (D.G.E.); (D.C.A.)
- Institute of Clinical Research, University of Southern Denmark, J. B. Winsløwsvej 19, 5000 Odense C, Denmark
| | - Ditte Caroline Andersen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, J. B. Winsløwsvej 25, 5000 Odense C, Denmark; (D.G.E.); (D.C.A.)
- The Danish Regenerative Center, Odense University Hospital, J. B. Winsløwsvej 4, 5000 Odense C, Denmark
- Institute of Clinical Research, University of Southern Denmark, J. B. Winsløwsvej 19, 5000 Odense C, Denmark
| |
Collapse
|
21
|
Itzhaki Ben Zadok O, Mager A, Leshem-Lev D, Lev E, Kornowski R, Eisen A. The Effect of Proprotein Convertase Subtilisin Kexin Type 9 Inhibitors on Circulating Endothelial Progenitor Cells in Patients with Cardiovascular Disease. Cardiovasc Drugs Ther 2021; 36:85-92. [PMID: 33394363 DOI: 10.1007/s10557-020-07119-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/25/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE Circulating endothelial progenitor cells (cEPCs) are vital to vascular repair by re-endothelialization. We aimed to explore the effect of proprotein convertase subtilisin kexin type 9 inhibitors (PCSK9i) on cEPCs hypothesizing a possible pleiotropic effect. METHODS Patients with cardiovascular disease (CVD) were sampled for cEPCs at baseline and following the initiation of PCSK9i. cEPCs were assessed using flow cytometry by the expression of CD34(+)/CD133(+) and vascular endothelial growth factor receptor (VEGFR)-2(+), and by the formation of colony-forming units (CFUs) and production of VEGF. RESULTS Our cohort included 26 patients (median age 68 (IQR 63, 73) years; 69% male). Following 3 months of treatment with PCSK9i and a decline in low-density lipoprotein cholesterol levels (153 (IQR 116, 176) to 56 (IQR 28, 72) mg/dl), p < 0.001), there was an increase in CD34(+)/CD133(+) and VEGFR-2(+) cell levels (0.98% (IQR 0.37, 1.55) to 1.43% (IQR 0.90, 4.51), p = 0.002 and 0.66% (IQR 0.22, 0.99) to 1.53% (IQR 0.73, 2.70), p = 0.05, respectively). Functionally, increase in EPCs-CFUs was microscopically evident following treatment with PCSK9i (1 CFUs (IQR 0.0, 1.0) to 2.5 (IQR 1.5, 3), p < 0.001) with a concomitant increase in EPC's viability as demonstrated by an MTT assay (0.15 (IQR 0.11, 0.19) to 0.21 (IQR 0.18, 0.23), p < 0.001). VEGF levels increased following PCSK9i treatment (57 (IQR 18, 24) to 105 (IQR 43, 245), p = 0.006). CONCLUSIONS Patients with CVD treated with PCSK9i demonstrate higher levels of active cEPCs, reflecting the promotion of endothelial repair. These findings may represent a novel mechanism of action of PCSK9i.
Collapse
Affiliation(s)
- Osnat Itzhaki Ben Zadok
- Department of Cardiology, Rabin Medical Center, 39 Jabotinsky St., 49100, Petah Tikva, Israel.
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Aviv Mager
- Department of Cardiology, Rabin Medical Center, 39 Jabotinsky St., 49100, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dorit Leshem-Lev
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
| | - Eli Lev
- Department of Cardiology, Assuta Ashdod Medical Center, Ashdod, Israel
- Faculty of Medicine, Ben-Gurion University, Beersheba, Israel
| | - Ran Kornowski
- Department of Cardiology, Rabin Medical Center, 39 Jabotinsky St., 49100, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alon Eisen
- Department of Cardiology, Rabin Medical Center, 39 Jabotinsky St., 49100, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
22
|
Ding X, Xiang W, He X. IFN-I Mediates Dysfunction of Endothelial Progenitor Cells in Atherosclerosis of Systemic Lupus Erythematosus. Front Immunol 2020; 11:581385. [PMID: 33262760 PMCID: PMC7686511 DOI: 10.3389/fimmu.2020.581385] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease including the cardiovascular system. Atherosclerosis is the most common cardiovascular complication of SLE and a significant risk factor for morbidity and mortality. Vascular damage/protection mechanism in SLE patients is out of balance, caused by the cascade reaction among oxidative stress, proinflammatory cytokines, Neutrophil Extracellular Traps, activation of B cells and autoantibodies and abnormal T cells. As a precursor cell repairing vascular endothelium, endothelial progenitor cells (EPCs) belong to the protective mechanism and show the reduced number and impaired function in SLE. However, the pathological mechanism of EPCs dysfunction in SLE remains ill-defined. This paper reviews the latest SLE epidemiology and pathogenesis, discusses the changes in the number and function of EPCs in SLE, expounds the role of EPCs in SLE atherosclerosis, and provides new guidance and theoretical basis for exploring novel targets for SLE treatment.
Collapse
Affiliation(s)
- Xuewei Ding
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wei Xiang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, NHC Key Laboratory of Control of Tropical diseases (Hainan Medical University), Haikou, China
| | - Xiaojie He
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
23
|
Endothelial progenitor cells as the target for cardiovascular disease prediction, personalized prevention, and treatments: progressing beyond the state-of-the-art. EPMA J 2020; 11:629-643. [PMID: 33240451 DOI: 10.1007/s13167-020-00223-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 09/11/2020] [Indexed: 02/06/2023]
Abstract
Stimulated by the leading mortalities of cardiovascular diseases (CVDs), various types of cardiovascular biomaterials have been widely investigated in the past few decades. Although great therapeutic effects can be achieved by bare metal stents (BMS) and drug-eluting stents (DES) within months or years, the long-term complications such as late thrombosis and restenosis have limited their further applications. It is well accepted that rapid endothelialization is a promising approach to eliminate these complications. Convincing evidence has shown that endothelial progenitor cells (EPCs) could be mobilized into the damaged vascular sites systemically and achieve endothelial repair in situ, which significantly contributes to the re-endothelialization process. Therefore, how to effectively capture EPCs via specific molecules immobilized on biomaterials is an important point to achieve rapid endothelialization. Further, in the context of predictive, preventive, personalized medicine (PPPM), the abnormal number alteration of EPCs in circulating blood and certain inflammation responses can also serve as important indicators for predicting and preventing early cardiovascular disease. In this contribution, we mainly focused on the following sections: the definition and classification of EPCs, the mechanisms of EPCs in treating CVDs, the potential diagnostic role of EPCs in predicting CVDs, as well as the main strategies for cardiovascular biomaterials to capture EPCs.
Collapse
|
24
|
Pei X, Kim H, Lee M, Wang N, Shin J, Lee S, Yoon M, Yang VC, He H. Local delivery of cardiac stem cells overexpressing HIF-1α promotes angiogenesis and muscular tissue repair in a hind limb ischemia model. J Control Release 2020; 322:610-621. [DOI: 10.1016/j.jconrel.2020.03.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/23/2020] [Accepted: 03/13/2020] [Indexed: 12/14/2022]
|
25
|
Summers T, Sookhoo B, Stumpo L, Parada S. Suprascapular Artery Aneurysm Secondary to Severe Shoulder Joint Osteoarthritis. CASE REPORTS IN ORTHOPEDIC RESEARCH 2020. [DOI: 10.1159/000507505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Aneurysms of the thyrocervical trunk and collateral branches are rarely encountered. Upon literature review, no documented cases of a suprascapular artery aneurysm resulting from osteoarthritis have been previously described. A 64-year-old female was found to have focal aneurysmal formation within the suprascapular artery. The extensive osteoarthritic changes to the glenoid, including medialization of her joint line, is hypothesized to have led to arterial injury and the observed aneurysm formation. Chronic mechanical stress on small vessels from abnormal bony contact in the setting of osteoarthritis can lead to aneurysmal formation. Arthritis as a cause of aneurysm formation in collateral vessels of the thyrocervical trunk has not been previously described.
Collapse
|
26
|
Balistreri CR, De Falco E, Bordin A, Maslova O, Koliada A, Vaiserman A. Stem cell therapy: old challenges and new solutions. Mol Biol Rep 2020; 47:3117-3131. [PMID: 32128709 DOI: 10.1007/s11033-020-05353-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 02/26/2020] [Indexed: 12/11/2022]
Abstract
Stem cell therapy (SCT), born as therapeutic revolution to replace pharmacological treatments, remains a hope and not yet an effective solution. Accordingly, stem cells cannot be conceivable as a "canonical" drug, because of their unique biological properties. A new reorientation in this field is emerging, based on a better understanding of stem cell biology and use of cutting-edge technologies and innovative disciplines. This will permit to solve the gaps, failures, and long-term needs, such as the retention, survival and integration of stem cells, by employing pharmacology, genetic manipulation, biological or material incorporation. Consequently, the clinical applicability of SCT for chronic human diseases will be extended, as well as its effectiveness and success, leading to long-awaited medical revolution. Here, some of these aspects are summarized, reviewing and discussing recent advances in this rapidly developing research field.
Collapse
Affiliation(s)
- Carmela Rita Balistreri
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo, Italy.
| | - Elena De Falco
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
- Mediterranea Cardiocentro, Napoli, Italy
| | - Antonella Bordin
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Olga Maslova
- National University of Life and Environmental Sciences of Ukraine, Kyiv, Ukraine
| | | | | |
Collapse
|
27
|
Frump AL, Lahm T. Tips for success in pulmonary hypertension treatment: progress in isolating endothelial cells from pulmonary artery catheters. Eur Respir J 2020; 55:55/3/2000122. [PMID: 32198271 DOI: 10.1183/13993003.00122-2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 01/21/2020] [Indexed: 11/05/2022]
Affiliation(s)
- Andrea L Frump
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Dept of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tim Lahm
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Dept of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Richard L. Roudebush VA Medical Center, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
28
|
Hussein KH, Park KM, Yu L, Song SH, Woo HM, Kwak HH. Vascular reconstruction: A major challenge in developing a functional whole solid organ graft from decellularized organs. Acta Biomater 2020; 103:68-80. [PMID: 31887454 DOI: 10.1016/j.actbio.2019.12.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 02/07/2023]
Abstract
Bioengineering a functional organ holds great potential to overcome the current gap between the organ need and shortage of available organs. Whole organ decellularization allows the removal of cells from large-scale organs, leaving behind extracellular matrices containing different growth factors, structural proteins, and a vascular network with a bare surface. Successful application of decellularized tissues as transplantable organs is hampered by the inability to completely reline the vasculature by endothelial cells (ECs), leading to blood coagulation, loss of vascular patency, and subsequent death of reseeded cells. Therefore, an intact, continuous layer of endothelium is essential to maintain proper functioning of the vascular system, which includes the transfer of nutrients to surrounding tissues and protecting other types of cells from shear stress. Here, we aimed to summarize the available cell sources that can be used for reendothelialization in addition to different trials performed by researchers to reconstruct vascularization of decellularized solid organs. Additionally, different techniques for enhancing reendothelialization and the methods used for evaluating reendothelialization efficiency along with the future prospective applications of this field are discussed. STATEMENT OF SIGNIFICANCE: Despite the great progress in whole organ decellularization, reconstruction of vasculature within the engineered constructs is still a major roadblock. Reconstructed endothelium acts as a multifunctional barrier of vessels, which can reduce thrombosis and help delivering of oxygen and nutrients throughout the whole organ. Successful reendothelialization can be achieved through reseeding of appropriate cell types on the naked vasculature with or without modification of its surface. Here, we present the current research milestones that so far established to reconstruct the vascular network in addition to the methods used for evaluating the efficiency of reendotheilization. Thus, this review is quite significant and will aid the researchers to know where we stand toward biofabricating a transplantable organ from decellularizd extracellular matrix.
Collapse
|
29
|
Fenofibrate Reverses Dysfunction of EPCs Caused by Chronic Heart Failure. J Cardiovasc Transl Res 2019; 13:158-170. [PMID: 31701352 DOI: 10.1007/s12265-019-09889-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/12/2019] [Indexed: 12/18/2022]
Abstract
The enhanced activity of endothelial progenitor cells (EPCs) by AMP-activated protein kinase (AMPK) agonists might explain the reversal of chronic heart failure (CHF)-mediated endothelial dysfunction. We studied baseline circulating EPC numbers in patients with heart failure and clarified the effect of fenofibrate on both circulating angiogenic cell (CAC) and late EPC activity. The numbers of circulating EPCs in CHF patients were quantified by flow cytometry. Blood-derived mononuclear cells were cultured, and CAC and late EPC functions, including fibronectin adhesion, tube formation, and migration, were evaluated. We focused on the effect of fenofibrate, an AMPK agonist, on EPC function and Akt/eNOS cascade activation in vitro. The number of circulating EPCs (CD34+/KDR+) was significantly lower in CHF patients (ischemic cardiomyopathy (ICMP): 0.07%, dilated cardiomyopathy (DCMP): 0.068%; p < 0.05) than in healthy subjects (0.102% of the gating region). In CACs, fibronectin adhesion function was reversed by fenofibrate treatment (p < 0.05). Similar results were also found for tube formation and migration in late EPCs, which were significantly improved by fenofibrate in an AMPK-dependent manner (p < 0.05), suggesting that fenofibrate reversed CACs and late EPC dysfunction in CHF patients. The present findings reveal the potential application of the AMPK agonist fenofibrate to reverse endothelial dysfunction in CHF patients.
Collapse
|
30
|
Vahdat S, Pahlavan S, Mahmoudi E, Barekat M, Ansari H, Bakhshandeh B, Aghdami N, Baharvand H. Expansion of Human Pluripotent Stem Cell-derived Early Cardiovascular Progenitor Cells by a Cocktail of Signaling Factors. Sci Rep 2019; 9:16006. [PMID: 31690816 PMCID: PMC6831601 DOI: 10.1038/s41598-019-52516-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 10/16/2019] [Indexed: 11/15/2022] Open
Abstract
Cardiovascular progenitor cells (CPCs) derived from human pluripotent stem cells (hPSCs) are proposed to be invaluable cell sources for experimental and clinical studies. This wide range of applications necessitates large-scale production of CPCs in an in vitro culture system, which enables both expansion and maintenance of these cells. In this study, we aimed to develop a defined and efficient culture medium that uses signaling factors for large-scale expansion of early CPCs, called cardiogenic mesodermal cells (CMCs), which were derived from hPSCs. Chemical screening resulted in a medium that contained a reproducible combination of three factors (A83-01, bFGF, and CHIR99021) that generated 1014 CMCs after 10 passages without the propensity for tumorigenicity. Expanded CMCs retained their gene expression pattern, chromosomal stability, and differentiation tendency through several passages and showed both the safety and possible cardio-protective potentials when transplanted into the infarcted rat myocardium. These CMCs were efficiently cryopreserved for an extended period of time. This culture medium could be used for both adherent and suspension culture conditions, for which the latter is required for large-scale CMC production. Taken together, hPSC-derived CMCs exhibited self-renewal capacity in our simple, reproducible, and defined medium. These cells might ultimately be potential, promising cell sources for cardiovascular studies.
Collapse
Affiliation(s)
- Sadaf Vahdat
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Elena Mahmoudi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maryam Barekat
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hassan Ansari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Behnaz Bakhshandeh
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Nasser Aghdami
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
31
|
Buffa S, Borzì D, Chiarelli R, Crapanzano F, Lena AM, Nania M, Candi E, Triolo F, Ruvolo G, Melino G, Balistreri CR. Biomarkers for vascular ageing in aorta tissues and blood samples. Exp Gerontol 2019; 128:110741. [PMID: 31648011 DOI: 10.1016/j.exger.2019.110741] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 07/26/2019] [Accepted: 09/18/2019] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Functional and quantitative alterations and senescence of circulating and expanded endothelial progenitor cells (EPC), as well as systemic and tissue modifications of angiogenetic and inflammatory molecules, were evaluated for predicting age-related vessel wall remodeling, correlating them to intima media thickness (IMT) in the common carotid artery (CCA), a biomarker of early cardiovascular disease and aortic root dilation. POPULATIONS AND METHODS A homogenous Caucasian population was included in the study, constituted by 160 healthy subjects (80 old subjects, mean age 72 ± 6.4, range 66-83 years; and 80 younger blood donors, mean age 26.2 ± 3.4, range 21-33 years), and 60 old subjects (mean age 73 ± 1.4 (range 66-83) years) with aortic root dilatation and hypertension, and 60 old people (70 ± 2.8 (age range 66-83)) with sporadic ascending aorta aneurysm (AAA). In addition, 20 control individuals (10 men and 10 women, mean age: 65 ± 8), were also included in the study for evaluating the gene expression's levels, in aorta tissues. Appropriate techniques, practises, protocols, gating strategies and statistical analyses were performed in our evaluations. RESULTS Interestingly, old people had a significantly reduced functionality and a high grade of senescence (high SA-β-Gal activity and high levels of TP53, p21 and p16 genes) of EPC expanded than younger subjects. The values of related parameters progressively augmented from the old subjects, in good healthy shape, to subjects with hypertension and aorta dilation, and AAA. Moreover, they significantly impacted the endothelium than the alterations in EPC number. No changes, but rather increased systemic levels of VEGF and SDF-1 were also assessed in old people vs. younger donors. Old people also showed significantly increased systemic levels of inflammatory cytokines, and a reciprocal significant reduction of systemic s-Notch 1 than younger subjects. These parameters, also including the number EPC alterations, resulted to be significantly sustained in old people bearers of an inflammatory combined genotype. Consistent with these data, a reduced expression of Notch-1 gene, accompanied by a sustained expression of inflammatory genes (i.e. TLR4, IL-1β, IL-6 and IL-17) were detected in aortic tissues from old control people and AAA cases. Finally, we detected the biological effects induced by all the detected alterations on vessel wall age-related remodeling, by evaluating the IMT in the population studied and correlating it to these alterations. The analysis demonstrated that the unique independent risk predictors for vascular ageing are age, the EPC reduced migratory activity and senescence, high grade of expression of genes inducing EPC senescence and chronic tissue and systemic inflammation. CONCLUSIONS Thus, we propose these parameters, of easy determination in biological samples (i.e. blood and tissue samples) from alive human population, as optimal biomarkers for vascular ageing.
Collapse
Affiliation(s)
- Silvio Buffa
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Davide Borzì
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Rita Chiarelli
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Floriana Crapanzano
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Anna Maria Lena
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Martina Nania
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome, Italy; Istituto Dermopatico dell'Immacolata, IRCCS, Rome, Italy
| | - Fabio Triolo
- Department of Internal Medicine and Cardiovascular Disease, Division of Cardiology and Cardiovascular Rehabilitation, University Hospital Paolo Giaccone, Palermo, Italy
| | - Giovanni Ruvolo
- Cardiac Surgery Unit, Department of Surgical Science, Tor Vergata University Hospital, Rome, Italy
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome, Italy; MRC-Toxicology Unit, University of Cambridge, UK
| | - Carmela Rita Balistreri
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo, Italy.
| |
Collapse
|
32
|
Suresh V, West JL. 3D Culture Facilitates VEGF-Stimulated Endothelial Differentiation of Adipose-Derived Stem Cells. Ann Biomed Eng 2019; 48:1034-1044. [PMID: 31165294 DOI: 10.1007/s10439-019-02297-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 05/25/2019] [Indexed: 01/15/2023]
Abstract
De novo vascularization of implantable tissue and whole organ constructs has been a significant challenge in the field of tissue engineering; the use of endothelial cell populations for this task is constrained by the cell population's limited regeneration capacity and potential for loss of function. Thus, there is a need for a stem-cell population that may be induced into an endothelial cell phenotype reliably. Adipose derived stem cells (ADSCs) are multipotent cells that can be readily isolated from donor fat and may have the potential to be readily induced into endothelial cells. The ability to stimulate endothelial differentiation of these cells has been limited in standard 2D culture. We hypothesized that 3D culture would yield better differentiation. To study the influence of cell density and culture conditions on the potential of ADSCs to differentiate into an endothelial-like state, we seeded these cells types within a 3D cell-adhesive, proteolytically degradable, peptide-modified poly(ethylene-glycol) (PEG) hydrogel. ADSCs were either cultured in basal media or pro-angiogenic media supplemented with 20 ng/mL of VEGF in 2D and then encapsulated at low or high densities within the PEG-based hydrogel. These encapsulated cells were maintained in either basal media or pro-angiogenic media. Cells were then isolated from the hydrogels and cultured in Matrigel to assess the potential for tubule formation. Our work shows that maintenance of ADSCs in a pro-angiogenic medium in 2D monoculture alone does not result in any CD31 expression. Furthermore, the level of CD31 expression was affected by the density of the cells encapsulated within the PEG-based hydrogel. Upon isolation of these cells, we found that these induced ADSCs were able to form tubules within Matrigel, indicative of endothelial function, while ADSCs cultured in basal medium could not. This finding points to the potential for this stem-cell population to serve as a safe and reliable source of endothelial cells for tissue engineering and regenerative medicine purposes.
Collapse
Affiliation(s)
- V Suresh
- Duke University School of Medicine, DUMC 3878, Durham, NC, 27710, USA
| | - J L West
- Department of Biomedical Engineering, Duke University, Box 90281, Durham, NC, 27708, USA.
| |
Collapse
|
33
|
Lamichane S, Baek SH, Kim YJ, Park JH, Dahal Lamichane B, Jang WB, Ji S, Lee NK, Dehua L, Kim DY, Kang S, Seong HJ, Yun J, Lee DH, Moon HR, Chung HY, Kwon SM. MHY2233 Attenuates Replicative Cellular Senescence in Human Endothelial Progenitor Cells via SIRT1 Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6492029. [PMID: 31223423 PMCID: PMC6556284 DOI: 10.1155/2019/6492029] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/09/2019] [Indexed: 12/13/2022]
Abstract
Cardiovascular diseases (CVDs) are a major cause of death worldwide. Due to the prevalence of many side effects and incomplete recovery from pharmacotherapies, stem cell therapy is being targeted for the treatment of CVDs. Among the different types of stem cells, endothelial progenitor cells (EPCs) have great potential. However, cellular replicative senescence decreases the proliferation, migration, and overall function of EPCs. Sirtuin 1 (SIRT1) has been mainly studied in the mammalian aging process. MHY2233 is a potent synthetic SIRT1 activator and a novel antiaging compound. We found that MHY2233 increased the expression of SIRT1, and its deacetylase activity thereby decreased expression of the cellular senescence biomarkers, p53, p16, and p21. In addition, MHY2233 decreased senescence-associated beta-galactosidase- (SA-β-gal-) positive cells and senescence-associated secretory phenotypes (SASPs), such as the secretion of interleukin- (IL-) 6, IL-8, IL-1α, and IL-1β. MHY2233 treatment protected senescent EPCs from oxidative stress by decreasing cellular reactive oxygen species (ROS) levels, thus enhancing cell survival and function. The angiogenesis, proliferation, and migration of senescent EPCs were enhanced by MHY2233 treatment. Thus, MHY2233 reduces replicative and oxidative stress-induced senescence in EPCs. Therefore, this novel antiaging compound MHY2233 might be considered a potent therapeutic agent for the treatment of age-associated CVDs.
Collapse
Affiliation(s)
- Shreekrishna Lamichane
- Convergence Stem Cell Research Center, Pusan National University, Yangsan, Republic of Korea
- Molecular Inflammation Research Center for Aging Intervention, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Sang Hong Baek
- Laboratory of Cardiovascular Disease, Division of Cardiology, School of Medicine, The Catholic University of Korea, Seoul 137-040, Republic of Korea
| | - Yeon-Ju Kim
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ji Hye Park
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Babita Dahal Lamichane
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Woong Bi Jang
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - SeungTaek Ji
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Na Kyung Lee
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Li Dehua
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Da Yeon Kim
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Songhwa Kang
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ha Jong Seong
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jisoo Yun
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Dong Hyung Lee
- Department of Obstetrics and Gynecology, Biomedical Research Institute, School of Medicine, Pusan National University, Busan 46241, Republic of Korea
| | - Hyung Ryong Moon
- Laboratory of Medicinal Chemistry, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Hae Young Chung
- Molecular Inflammation Research Center for Aging Intervention, College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Sang-Mo Kwon
- Convergence Stem Cell Research Center, Pusan National University, Yangsan, Republic of Korea
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| |
Collapse
|
34
|
Balistreri CR, Pisano C, Bertoldo F, Massoud R, Dolci S, Ruvolo G. Red Blood Cell Distribution Width, Vascular Aging Biomarkers, and Endothelial Progenitor Cells for Predicting Vascular Aging and Diagnosing/Prognosing Age-Related Degenerative Arterial Diseases. Rejuvenation Res 2019; 22:399-408. [PMID: 30572793 DOI: 10.1089/rej.2018.2144] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The emerging evidence emphasizes red blood cell distribution width (RDW) as optimal prognostic biomarker for cardiovascular diseases. However, several clinical biases impede its clinical application. Recent recommendations suggest combining RDW with other biomarkers. Accordingly, we propose evaluating the well-recognized biomarkers of vascular aging (i.e., the leukocyte telomere length and telomerase activity, and reduced levels of endothelial progenitor cells [EPCs]) with RDW, for predicting the risk for vascular aging and onset and prognosis of age-related degenerative arterial diseases, such as sporadic ascending aorta aneurysm (AAA), characterized to have an increased incidence in old people. Consequently, in this study (and for the first time), we simultaneously investigated the relationship between RDW values, systemic inflammatory molecules, mean values of leukocyte telomere length, telomerase activity and EPCs, and the risk for vascular aging and AAA onset and prognosis. To achieve this aim, we selected 80 old and 80 young healthy subjects and 80 AAA cases. Appropriate methodologies were used for assessing blood parameters, aorta alterations, genotyping, impairment of the leukocyte telomere length, and telomerase activity. The main findings obtained demonstrated that increased RDW values along with the augmented blood levels of high-sensitive C-reactive protein and the reduced mean values of both leukocyte telomere length, telomerase activity, and EPCs are independently associated with the high risk for both vascular aging and AAA onset and prognosis. They might be used as the best predictor biomarker profile for vascular aging, and for both diagnosis and outcome of sporadic AAA.
Collapse
Affiliation(s)
- Carmela Rita Balistreri
- Department of Pathobiology and Medical and Forensic Biotechnologies, University of Palermo, Palermo, Italy
| | - Calogera Pisano
- Department of Cardiac Surgery, University of Rome "Tor Vergata," Rome, Italy
| | - Fabio Bertoldo
- Department of Cardiac Surgery, University of Rome "Tor Vergata," Rome, Italy
| | - Renato Massoud
- Department of Clinical Biochemistry, Tor Vergata University Hospital, Rome, Italy
| | - Susanna Dolci
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Giovanni Ruvolo
- Department of Cardiac Surgery, University of Rome "Tor Vergata," Rome, Italy
| |
Collapse
|
35
|
Poz D, De Falco E, Pisano C, Madonna R, Ferdinandy P, Balistreri CR. Diagnostic and Prognostic Relevance of Red Blood Cell Distribution Width for Vascular Aging and Cardiovascular Diseases. Rejuvenation Res 2018; 22:146-162. [PMID: 30132390 DOI: 10.1089/rej.2018.2094] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Evidence suggests association of red blood cell distribution width (RDW) with cardiovascular diseases (CVDs). On the contrary, we underline that the sole RDW values cannot represent a valid CVD biomarker. High RDW values are expression of biological effects of a lot of both endogenous and exogenous factors (i.e., age, sex, genetic background, inflammation, hormones, drugs, diet, exercise, hematological analyzers, and ranges of values), modulating the biology and physiology of erythrocytes. Thus, the singular monitoring of RDW cannot be used to predict cardiovascular disorders. Accordingly, we have reviewed the evidence for potential relationship of RDW values with alterations in the cardiovascular system (i.e., regenerative capacity, endothelial turnover, and senescence of cardiovascular cells), associated with vascular aging and disease. In addition, we highlight the inevitable impact of biases in clinical application of RDW related to CVDs. Based on our thorough review of literature, we suggest a combined evaluation of RDW with other emerging biomarkers related to vascular aging and the diagnosis and prognosis of CVDs, including telomere length of leukocytes, circulating nucleated red blood cells (nRBCs) and endothelial progenitor cells (EPCs) in future large scale studies.
Collapse
Affiliation(s)
- Donatella Poz
- 1 Department of Laboratory Medicine, Institute of Clinical Pathology, Azienda Sanitaria Universitaria Integrata (ASUI) di Udine, Udine, Italy
| | - Elena De Falco
- 2 Department of Medical-Surgical Sciences and Biotechnologies, "Sapienza" University of Rome, Latina, Italy
| | - Calogera Pisano
- 3 Cardiac Surgery, Tor Vergata University, Cardiochirurgia Policlinico Tor Vergata, Rome, Italy
| | - Rosalinda Madonna
- 4 Heart Failure Research, Texas Heart Institute, St. Luke's Episcopal Hospital, Houston, Texas.,5 Department of Internal Medicine, Cardiology, The University of Texas Health Science Center at Houston, Houston, Texas.,6 Department of Neurosciences, Center of Aging Sciences and Translational Medicine, CESI-Met and Institute of Cardiology, Imaging and Clinical Sciences "G. D'Annunzio" University, Chieti, Italy
| | - Peter Ferdinandy
- 7 Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,8 Pharmahungary Group, Szeged, Hungary
| | - Carmela Rita Balistreri
- 9 Department of Pathobiology and Medical and Forensic Biotechnologies, University of Palermo, Palermo, Italy
| |
Collapse
|
36
|
Poletto V, Rosti V, Biggiogera M, Guerra G, Moccia F, Porta C. The role of endothelial colony forming cells in kidney cancer's pathogenesis, and in resistance to anti-VEGFR agents and mTOR inhibitors: A speculative review. Crit Rev Oncol Hematol 2018; 132:89-99. [PMID: 30447930 DOI: 10.1016/j.critrevonc.2018.09.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 08/07/2018] [Accepted: 09/08/2018] [Indexed: 12/22/2022] Open
Abstract
Renal cell carcinoma (RCC) is highly dependent on angiogenesis, due to the overactivation of the VHL/HIF/VEGF/VEGFRs axis; this justifies the marked sensitivity of this neoplasm to antiangiogenic agents which, however, ultimately fail to control tumor growth. RCC also frequently shows alterations in the mTOR signaling pathway, and mTOR inhibitors have shown a similar pattern of initial activity/late failure as pure antiangiogenic agents. Understanding mechanisms of resistance to these agents would be key to improve the outcome of our patients. Circulating endothelial cells are a family of mainly bone marrow-derived progenitors, which have been postulated to be responsible of the reactivation of angiogenesis in different tumors. In this review, we shall discuss the complex nature and function of these cells, the evidence pro and contra their contribution to tumor vascularization, especially as far as RCC is concerned, and their possible role in determining resistance to presently available treatments.
Collapse
Affiliation(s)
- Valentina Poletto
- Center for the Study of Myelofibrosis, Research and Experimental Biotechnology Laboratory Area, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Piazzale Golgi 19, 27100, Pavia, Italy.
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, Research and Experimental Biotechnology Laboratory Area, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Piazzale Golgi 19, 27100, Pavia, Italy.
| | - Marco Biggiogera
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Italy.
| | - Germano Guerra
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy.
| | - Francesco Moccia
- Laboratory of Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, via Forlanini 6, 27100, Pavia, Italy.
| | - Camillo Porta
- Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (I.R.C.C.S.) Policlinico San Matteo Foundation, Piazzale Golgi 19, 27100, Pavia, Italy; present address: Department of Internal Medicine, University of Pavia, and Division of Translational Oncology, IRCCS Istituti Clinici Scientifici Maugeri, via S. Maugeri 10, 27100 Pavia, Italy.
| |
Collapse
|
37
|
Deregulation of Notch1 pathway and circulating endothelial progenitor cell (EPC) number in patients with bicuspid aortic valve with and without ascending aorta aneurysm. Sci Rep 2018; 8:13834. [PMID: 30218064 PMCID: PMC6138685 DOI: 10.1038/s41598-018-32170-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/30/2018] [Indexed: 12/29/2022] Open
Abstract
Bicuspid aortic valve (BAV) is frequently associated with the development of ascending aortic aneurysm, even if the underlying mechanisms remain to be clarified. Here, we investigated if a deregulation of Notch1 signaling pathway and endothelial progenitor cells (EPCs) number is associated with BAV disease and an early ascending aortic aneurysm (AAA) onset. For this purpose, 70 subjects with BAV (M/F 50/20; mean age: 58.8 ± 14.8 years) and 70 subjects with tricuspid aortic valve (TAV) (M/F 35/35; mean age: 69.1 ± 12.8 years) and AAA complicated or not, were included. Interestingly, patients with AAA showed a significant increase in circulating Notch1 levels and EPC number than subjects without AAA. However, circulating Notch1 levels and EPC number were significantly lower in BAV subjects than TAV patients either in the presence or absence of AAA. Finally, Notch pathway was activated to a greater extent in aortic aneurysmatic portions with respect to healthy aortic fragments in both BAV and TAV patients. However, the expression of genes encoding components and ligands of Notch pathway in aortic tissues was significantly lower in BAV than TAV subjects. Our study demonstrates that BAV subjects are characterized by a significant decrease in both tissue and circulating levels of Notch pathway, and in blood EPC number than TAV patients, either in presence or absence of AAA disease.
Collapse
|
38
|
Sánchez PF, Brey EM, Briceño JC. Endothelialization mechanisms in vascular grafts. J Tissue Eng Regen Med 2018; 12:2164-2178. [PMID: 30079631 DOI: 10.1002/term.2747] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 05/18/2018] [Accepted: 07/09/2018] [Indexed: 12/12/2022]
Abstract
Despite the wide variety of tissue-engineered vascular grafts that are currently being developed, autologous vessels, such as the saphenous vein, are still the gold standard grafts for surgical treatment of vascular disease. Recently developed technologies have shown promising results in preclinical studies, but they still do not overcome the issues that native vessels present, and only a few have made the transition into clinical use. The endothelial lining is a key aspect for the success or failure of the grafts, especially on smaller diameter grafts (<5 mm). However, during the design and evaluation of the grafts, the mechanisms for the formation of this layer are not commonly examined. Therefore, a significant amount of established research might not be relevant to the clinical context, due to important differences that exist between the vascular regeneration mechanisms found in animal models and humans. This article reviews current knowledge about endothelialization mechanisms that have been so far identified: in vitro seeding, transanastomotic growth, transmural infiltration, and fallout endothelialization. Emphasis is placed on the models used for study of theses mechanisms and their effects on the development of tissue-engineering vascular conduits.
Collapse
Affiliation(s)
- Paolo F Sánchez
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá, Colombia
| | - Eric M Brey
- Biomedical Engineering, University of Texas at San Antonio, San Antonio, Texas.,Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois.,Research Service, South Texas Veterans Health Care System, San Antonio, Texas
| | - Juan Carlos Briceño
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá, Colombia.,Research Department, Fundación Cardioinfantil Instituto de Cardiología, Bogotá, Colombia
| |
Collapse
|
39
|
Yang JX, Pan YY, Wang XX, Qiu YG, Mao W. Endothelial progenitor cells in age-related vascular remodeling. Cell Transplant 2018; 27:786-795. [PMID: 29882417 PMCID: PMC6047273 DOI: 10.1177/0963689718779345] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence has demonstrated that endothelial progenitor cells (EPCs) could facilitate the reendothelialization of injured arteries by replacing the dysfunctional endothelial cells, thereby suppressing the formation of neointima. Meanwhile, other findings suggest that EPCs may be involved in the pathogenesis of age-related vascular remodeling. This review is presented to summarize the characteristics of EPCs and age-related vascular remodeling. In addition, the role of EPCs in age-related vascular remodeling and possible solutions for improving the therapeutic effects of EPCs in the treatment of age-related diseases are discussed.
Collapse
Affiliation(s)
- Jin-Xiu Yang
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China.,2 Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Yan-Yun Pan
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
| | - Xing-Xiang Wang
- 2 Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Yuan-Gang Qiu
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
| | - Wei Mao
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
| |
Collapse
|
40
|
Bauman E, Granja PL, Barrias CC. Fetal bovine serum-free culture of endothelial progenitor cells-progress and challenges. J Tissue Eng Regen Med 2018; 12:1567-1578. [PMID: 29701896 DOI: 10.1002/term.2678] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 03/22/2018] [Accepted: 04/16/2018] [Indexed: 12/19/2022]
Abstract
Two decades after the first report on endothelial progenitor cells (EPC), their key role in postnatal vasculogenesis and vascular repair is well established. The therapeutic potential of EPC and their growing use in clinical trials calls for the development of more robust, reproducible, and safer methods for the in vitro expansion and maintenance of these cells. Despite many limitations associated with its usage, fetal bovine serum (FBS) is still widely applied as a cell culture supplement. Although different approaches aiming at establishing FBS-free culture have been developed for many cell types, adequate solutions for endothelial cells, and for EPC in particular, are still scarce, possibly due to the multiple challenges that have to be faced when culturing these cells. In this review, we provide a brief overview on the therapeutic relevance of EPC and critically analyse the available literature on FBS-free endothelial cell culture methods, including xeno-free, serum-free, and chemically defined systems.
Collapse
Affiliation(s)
- E Bauman
- Instituto de Inovação e Investigação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Porto, Portugal
| | - P L Granja
- Instituto de Inovação e Investigação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Faculdade de Engenharia da Universidade do Porto (FEUP), Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - C C Barrias
- Instituto de Inovação e Investigação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
41
|
Shear stress: An essential driver of endothelial progenitor cells. J Mol Cell Cardiol 2018; 118:46-69. [PMID: 29549046 DOI: 10.1016/j.yjmcc.2018.03.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/08/2018] [Accepted: 03/09/2018] [Indexed: 02/06/2023]
Abstract
The blood flow through vessels produces a tangential, or shear, stress sensed by their innermost layer (i.e., endothelium) and representing a major hemodynamic force. In humans, endothelial repair and blood vessel formation are mainly performed by circulating endothelial progenitor cells (EPCs) characterized by a considerable expression of vascular endothelial growth factor receptor 2 (VEGFR2), CD34, and CD133, pronounced tube formation activity in vitro, and strong reendothelialization or neovascularization capacity in vivo. EPCs have been proposed as a promising agent to induce reendothelialization of injured arteries, neovascularization of ischemic tissues, and endothelialization or vascularization of bioartificial constructs. A number of preconditioning approaches have been suggested to improve the regenerative potential of EPCs, including the use of biophysical stimuli such as shear stress. However, in spite of well-defined influence of shear stress on mature endothelial cells (ECs), articles summarizing how it affects EPCs are lacking. Here we discuss the impact of shear stress on homing, paracrine effects, and differentiation of EPCs. Unidirectional laminar shear stress significantly promotes homing of circulating EPCs to endothelial injury sites, induces anti-thrombotic and anti-atherosclerotic phenotype of EPCs, increases their capability to form capillary-like tubes in vitro, and enhances differentiation of EPCs into mature ECs in a dose-dependent manner. These effects are mediated by VEGFR2, Tie2, Notch, and β1/3 integrin signaling and can be abrogated by means of complementary siRNA/shRNA or selective pharmacological inhibitors of the respective proteins. Although the testing of sheared EPCs for vascular tissue engineering or regenerative medicine applications is still an unaccomplished task, favorable effects of unidirectional laminar shear stress on EPCs suggest its usefulness for their preconditioning.
Collapse
|
42
|
Anti-Inflamm-Ageing and/or Anti-Age-Related Disease Emerging Treatments: A Historical Alchemy or Revolutionary Effective Procedures? Mediators Inflamm 2018; 2018:3705389. [PMID: 29576745 PMCID: PMC5822866 DOI: 10.1155/2018/3705389] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/13/2017] [Indexed: 01/16/2023] Open
Abstract
The “long-life elixir” has long represented for humans a dream, a vanity's sin for remaining young and to long survive. Today, because of ageing population phenomenon, the research of antiageing interventions appears to be more important than ever, for preserving health in old age and retarding/or delaying the onset of age-related diseases. A hope is given by experimental data, which evidence the possibility of retarding ageing in animal models. In addition, it has been also demonstrated in animal life-extending studies not only the possibility of increasing longevity but also the ability to retard the onset of age-related diseases. Interestingly, this recent evidence is leading to promise of obtaining the same effects in humans and resulting in benefits for their health in old ages. In order to achieve this goal, different approaches have been used ranging from pharmacological targeting of ageing, basic biological assays, and big data analysis to the recent use of young blood, stem cells, cellular, genetic, and epigenetic reprogramming, or other techniques of regenerative medicine. However, only a little fraction of these approaches has the features for being tested in clinical applications. Here, new emerging molecules, drugs, and procedures will be described, by evidencing potential benefits and limitations.
Collapse
|
43
|
Hinderer S, Sudrow K, Schneider M, Holeiter M, Layland SL, Seifert M, Schenke-Layland K. Surface functionalization of electrospun scaffolds using recombinant human decorin attracts circulating endothelial progenitor cells. Sci Rep 2018; 8:110. [PMID: 29311692 PMCID: PMC5758628 DOI: 10.1038/s41598-017-18382-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 12/11/2017] [Indexed: 01/25/2023] Open
Abstract
Decorin (DCN) is an important small leucine-rich proteoglycan present in the extracellular matrix (ECM) of many organs and tissues. Endothelial progenitor cells (EPCs) are able to interact with the surrounding ECM and bind to molecules such as DCN. Here, we recombinantly produced full-length human DCN under good laboratory practice (GLP) conditions, and after detailed immunological characterization, we investigated its potential to attract murine and human EPCs (mEPCs and hECFCs). Electrospun polymeric scaffolds were coated with DCN or stromal cell-derived factor-1 (SDF-1α) and were then dynamically cultured with both cell types. Cell viability was assessed via imaging flow cytometry. The number of captured cells was counted and compared with the non-coated controls. To characterize cell-scaffold interactions, immunofluorescence staining and scanning electron microscopy analyses were performed. We identified that DCN reduced T cell responses and attracted innate immune cells, which are responsible for ECM remodeling. A significantly higher number of EPCs attached on DCN- and SDF-1α-coated scaffolds, when compared with the uncoated controls. Interestingly, DCN showed a higher attractant effect on hECFCs than SDF-1α. Here, we successfully demonstrated DCN as promising EPC-attracting coating, which is particularily interesting when aiming to generate off-the-shelf biomaterials with the potential of in vivo cell seeding.
Collapse
Affiliation(s)
- Svenja Hinderer
- Department of Cell and Tissue Engineering, Fraunhofer-Institute for Interfacial Engineering and Biotechnology (IGB), 70569, Stuttgart, Germany
- Department of Women´s Health, Research Institute for Women's Health, Eberhard-Karls-University Tübingen, 72076, Tübingen, Germany
| | - Katrin Sudrow
- Institute of Medical Immunology and Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353, Berlin, Germany
| | - Maria Schneider
- Institute of Medical Immunology and Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353, Berlin, Germany
| | - Monika Holeiter
- Department of Women´s Health, Research Institute for Women's Health, Eberhard-Karls-University Tübingen, 72076, Tübingen, Germany
| | - Shannon Lee Layland
- Department of Cell and Tissue Engineering, Fraunhofer-Institute for Interfacial Engineering and Biotechnology (IGB), 70569, Stuttgart, Germany
- Department of Women´s Health, Research Institute for Women's Health, Eberhard-Karls-University Tübingen, 72076, Tübingen, Germany
| | - Martina Seifert
- Institute of Medical Immunology and Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353, Berlin, Germany
| | - Katja Schenke-Layland
- Department of Cell and Tissue Engineering, Fraunhofer-Institute for Interfacial Engineering and Biotechnology (IGB), 70569, Stuttgart, Germany.
- Department of Women´s Health, Research Institute for Women's Health, Eberhard-Karls-University Tübingen, 72076, Tübingen, Germany.
- Department of Medicine/Cardiology, Cardiovascular Research Laboratories, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
44
|
Angulski ABB, Capriglione LG, Batista M, Marcon BH, Senegaglia AC, Stimamiglio MA, Correa A. The Protein Content of Extracellular Vesicles Derived from Expanded Human Umbilical Cord Blood-Derived CD133 + and Human Bone Marrow-Derived Mesenchymal Stem Cells Partially Explains Why both Sources are Advantageous for Regenerative Medicine. Stem Cell Rev Rep 2017; 13:244-257. [PMID: 28054239 DOI: 10.1007/s12015-016-9715-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adult stem cells have beneficial effects when exposed to damaged tissue due, at least in part, to their paracrine activity, which includes soluble factors and extracellular vesicles (EVs). Given the multiplicity of signals carried by these vesicles through the horizontal transfer of functional molecules, human mesenchymal stem cell (hMSCs) and CD133+ cell-derived EVs have been tested in various disease models and shown to recover damaged tissues. In this study, we profiled the protein content of EVs derived from expanded human CD133+ cells and bone marrow-derived hMSCs with the intention of better understanding the functions performed by these vesicles/cells and delineating the most appropriate use of each EV in future therapeutic procedures. Using LC-MS/MS analysis, we identified 623 proteins for expanded CD133+-EVs and 797 proteins for hMSCs-EVs. Although the EVs from both origins were qualitatively similar, when protein abundance was considered, hMSCs-EVs and CD133+-EVs were different. Gene Ontology (GO) enrichment analysis in CD133+-EVs revealed proteins involved in a variety of angiogenesis-related functions as well proteins related to the cytoskeleton and highly implicated in cell motility and cellular activation. In contrast, when overrepresented proteins in hMSCs-EVs were analyzed, a GO cluster of immune response-related genes involved with immune response-regulating factors acting on phagocytosis and innate immunity was identified. Together our data demonstrate that from the point of view of protein content, expanded CD133+-EVs and hMSCs-EVs are in part similar but also sufficiently different to reflect the main beneficial paracrine effects widely reported in pre-clinical studies using expanded CD133+ cells and/or hBM-MSCs.
Collapse
Affiliation(s)
- Addeli B B Angulski
- Instituto Carlos Chagas, Fiocruz-Paraná, Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR, 81350-010, Brazil
| | - Luiz G Capriglione
- Núcleo de Tecnologia Celular, Pontifícia Universidade Católica do Paraná, Rua Imaculada Conceição, 1155, Curitiba, PR, 80215-901, Brazil
| | - Michel Batista
- Instituto Carlos Chagas, Fiocruz-Paraná, Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR, 81350-010, Brazil
| | - Bruna H Marcon
- Instituto Carlos Chagas, Fiocruz-Paraná, Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR, 81350-010, Brazil
| | - Alexandra C Senegaglia
- Núcleo de Tecnologia Celular, Pontifícia Universidade Católica do Paraná, Rua Imaculada Conceição, 1155, Curitiba, PR, 80215-901, Brazil
| | - Marco A Stimamiglio
- Instituto Carlos Chagas, Fiocruz-Paraná, Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR, 81350-010, Brazil.
| | - Alejandro Correa
- Instituto Carlos Chagas, Fiocruz-Paraná, Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR, 81350-010, Brazil.
| |
Collapse
|
45
|
Bianconi V, Sahebkar A, Kovanen P, Bagaglia F, Ricciuti B, Calabrò P, Patti G, Pirro M. Endothelial and cardiac progenitor cells for cardiovascular repair: A controversial paradigm in cell therapy. Pharmacol Ther 2017; 181:156-168. [PMID: 28827151 DOI: 10.1016/j.pharmthera.2017.08.004] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Stem cells have the potential to differentiate into cardiovascular cell lineages and to stimulate tissue regeneration in a paracrine/autocrine manner; thus, they have been extensively studied as candidate cell sources for cardiovascular regeneration. Several preclinical and clinical studies addressing the therapeutic potential of endothelial progenitor cells (EPCs) and cardiac progenitor cells (CPCs) in cardiovascular diseases have been performed. For instance, autologous EPC transplantation and EPC mobilization through pharmacological agents contributed to vascular repair and neovascularization in different animal models of limb ischemia and myocardial infarction. Also, CPC administration and in situ stimulation of resident CPCs have been shown to improve myocardial survival and function in experimental models of ischemic heart disease. However, clinical studies using EPC- and CPC-based therapeutic approaches have produced mixed results. In this regard, intracoronary, intra-myocardial or intramuscular injection of either bone marrow-derived or peripheral blood progenitor cells has improved pathological features of tissue ischemia in humans, despite modest or no clinical benefit has been observed in most cases. Also, the intriguing scientific background surrounding the potential clinical applications of EPC capture stenting is still waiting for a confirmatory proof. Moreover, clinical findings on the efficacy of CPC-based cell therapy in heart diseases are still very preliminary and based on small-size studies. Despite promising evidence, widespread clinical application of both EPCs and CPCs remains delayed due to several unresolved issues. The present review provides a summary of the different applications of EPCs and CPCs for cardiovascular cell therapy and underlies their advantages and limitations.
Collapse
Affiliation(s)
- Vanessa Bianconi
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Perugia, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Francesco Bagaglia
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Perugia, Italy
| | - Biagio Ricciuti
- Department of Medical Oncology, S. Maria della Misericordia Hospital, Perugia, Italy
| | - Paolo Calabrò
- Division of Cardiology, Second University of Naples, Department of Cardio-Thoracic and Respiratory Sciences, Italy
| | - Giuseppe Patti
- Unit of Cardiovascular Science, Campus Bio-Medico University of Rome, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Perugia, Italy.
| |
Collapse
|
46
|
Mistriotis P, Andreadis ST. Vascular aging: Molecular mechanisms and potential treatments for vascular rejuvenation. Ageing Res Rev 2017; 37:94-116. [PMID: 28579130 DOI: 10.1016/j.arr.2017.05.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 05/22/2017] [Accepted: 05/25/2017] [Indexed: 12/14/2022]
Abstract
Aging is the main risk factor contributing to vascular dysfunction and the progression of vascular diseases. In this review, we discuss the causes and mechanisms of vascular aging at the tissue and cellular level. We focus on Endothelial Cell (EC) and Smooth Muscle Cell (SMC) aging due to their critical role in mediating the defective vascular phenotype. We elaborate on two categories that contribute to cellular dysfunction: cell extrinsic and intrinsic factors. Extrinsic factors reflect systemic or environmental changes which alter EC and SMC homeostasis compromising vascular function. Intrinsic factors induce EC and SMC transformation resulting in cellular senescence. Replenishing or rejuvenating the aged/dysfunctional vascular cells is critical to the effective repair of the vasculature. As such, this review also elaborates on recent findings which indicate that stem cell and gene therapies may restore the impaired vascular cell function, reverse vascular aging, and prolong lifespan.
Collapse
Affiliation(s)
- Panagiotis Mistriotis
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
| | - Stelios T Andreadis
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA; Department of Biomedical Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA; Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14203, USA.
| |
Collapse
|
47
|
Abstract
The concept of pericyte has been changing over years. This cell type was believed to possess only a function of trophic support to endothelial cells and to maintain vasculature stabilization. In the last years, the discovery of multipotent ability of perivascular populations led to the concept of vessel/wall niche. Likewise, several perivascular populations have been identified in animal and human bone marrow. In this review, we provide an overview on bone marrow perivascular population, their cross-talk with other niche components, relationship with bone marrow stromal stem cells, and similarities and differences with the perivascular population of the vessel/wall niche. Finally, we focus on the regenerative potential of these cells and the forthcoming challenges related to their use as cell therapy products.
Collapse
Affiliation(s)
- Giuseppe Mangialardi
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, UK
| | - Andrea Cordaro
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, UK
| | - Paolo Madeddu
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, UK
| |
Collapse
|
48
|
Wils J, Favre J, Bellien J. Modulating putative endothelial progenitor cells for the treatment of endothelial dysfunction and cardiovascular complications in diabetes. Pharmacol Ther 2016; 170:98-115. [PMID: 27773788 DOI: 10.1016/j.pharmthera.2016.10.014] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Diabetes induces a decrease in the number and function of different pro-angiogenic cell types generically designated as putative endothelial progenitor cells (EPC), which encompasses cells from myeloid origin that act in a paracrine fashion to promote angiogenesis and putative "true" EPC that contribute to endothelial replacement. This not only compromises neovasculogenesis in ischemic tissues but also impairs, at an early stage, the reendotheliziation process at sites of injury, contributing to the development of endothelial dysfunction and cardiovascular complications. Hyperglycemia, insulin resistance and dyslipidemia promote putative EPC dysregulation by affecting the SDF-1/CXCR-4 and NO pathways and the p53/SIRT1/p66Shc axis that contribute to their mobilization, migration, homing and vasculogenic properties. To optimize the clinical management of patients with hypoglycemic agents, statins and renin-angiotensin system inhibitors, which display pleiotropic effects on putative EPC, is a first step to improve their number and angiogenic potential but specific strategies are needed. Among them, mobilizing therapies based on G-CSF, erythropoietin or CXCR-4 antagonism have been developed to increase putative EPC number to treat ischemic diseases with or without prior cell isolation and transplantation. Growth factors, genetic and pharmacological strategies are also evaluated to improve ex vivo cultured EPC function before transplantation. Moreover, pharmacological agents increasing in vivo the bioavailability of NO and other endothelial factors demonstrated beneficial effects on neovascularization in diabetic ischemic models but their effects on endothelial dysfunction remain poorly evaluated. More experiments are warranted to develop orally available drugs and specific agents targeting p66Shc to reverse putative EPC dysfunction in the expected goal of preventing endothelial dysfunction and diabetic cardiovascular complications.
Collapse
Affiliation(s)
- Julien Wils
- Department of Pharmacology, Rouen University Hospital, Rouen, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1096, Rouen, France; University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Julie Favre
- MITOVASC Institute, Angers, France; Centre National de la Recherche Scientifique (CNRS) UMR 6214, Angers, France; INSERM U1083, Angers, France; University of Angers, Angers, France
| | - Jérémy Bellien
- Department of Pharmacology, Rouen University Hospital, Rouen, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1096, Rouen, France; University of Rouen, Institute for Research and Innovation in Biomedicine, Rouen, France.
| |
Collapse
|
49
|
MicroRNA-195 regulates proliferation, migration, angiogenesis and autophagy of endothelial progenitor cells by targeting GABARAPL1. Biosci Rep 2016; 36:BSR20160139. [PMID: 27623937 PMCID: PMC5064457 DOI: 10.1042/bsr20160139] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 09/13/2016] [Indexed: 02/07/2023] Open
Abstract
Deep vein thrombosis (DVT) is a common type of venous thrombosis. Successful resolution of DVT-related thrombi is important in the treatment of DVT. Endothelial progenitor cells (EPCs) have emerged as a promising therapeutic choice for DVT-related thrombus resolution; however, the clinical application of EPCs faces many challenges. In the present study, the expression of miR-582, miR-195 and miR-532 under hypoxic or normoxic conditions was measured using quantitative real-time PCR analysis (qRT-PCR) and the results showed that the increased fold of miR-195 was highest in human EPCs (hEPCs) under hypoxic conditions. Then the role and regulating mechanism of miR-195 in improving the function of EPCs was investigated. To investigate the effect of miR-195 inhibition on the autophagy of hEPCs, the expression of the autophagy-related genes LC3B and beclin1 was examined using western blotting, and the formation of autophagosomes was observed using TEM. The results indicated that the inhibition of miR-195 expression could promote autophagy of hEPCs. In addition, we investigated the role of miR-195 on the proliferation, migration and angiogenesis of hEPCs under hypoxia. The results revealed that miR-195 inhibition promotes cell proliferation, migration and angiogenesis of hEPCs under hypoxia. Furthermore, GABA type A receptor associated protein like 1 (GABARAPL1) was identified as a directed target of miR-195 and GABARAPL1 silencing could decrease the effect of miR-195 knockdown on cell proliferation, migration, angiogenesis and autophagy of hEPCs under hypoxia. Together, these results indicate that miR-195 regulates cell proliferation, migration, angiogenesis and autophagy of hEPCs by targeting GABARAPL1.
Collapse
|
50
|
Madonna R, Novo G, Balistreri CR. Cellular and molecular basis of the imbalance between vascular damage and repair in ageing and age-related diseases: As biomarkers and targets for new treatments. Mech Ageing Dev 2016; 159:22-30. [DOI: 10.1016/j.mad.2016.03.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/07/2016] [Accepted: 03/12/2016] [Indexed: 12/24/2022]
|