1
|
Zhang Y, Guan Z, Gong H, Ni Z, Xiao Q, Guo X, Xu Q. The Role of Progenitor Cells in the Pathogenesis of Arteriosclerosis. CARDIOLOGY DISCOVERY 2024; 4:231-244. [DOI: 10.1097/cd9.0000000000000130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The increasing incidence of arteriosclerosis has become a significant global health burden. Arteriosclerosis is characterized by the thickening and hardening of arterial walls, which can lead to the narrowing or complete blockage of blood vessels. However, the pathogenesis of the disease remains incompletely understood. Recent research has shown that stem and progenitor cells found in the bone marrow and local vessel walls play a role in the development of arteriosclerosis by differentiating into various types of vascular cells, including endothelial cells, smooth muscle cells, fibroblasts, and inflammatory cells. This review aims to provide a comprehensive understanding of the role of stem and progenitor cells in the pathogenesis of arteriosclerosis, shedding light on the underlying mechanisms and potential therapeutic approaches for this disease.
Collapse
Affiliation(s)
- Yuesheng Zhang
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Ziyin Guan
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Hui Gong
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Zhichao Ni
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Xiaogang Guo
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Qingbo Xu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
2
|
Li Z, Zhang Y, Ma M, Wang W, Hui H, Tian J, Chen Y. Targeted mitigation of neointimal hyperplasia via magnetic field-directed localization of superparamagnetic iron oxide nanoparticle-labeled endothelial progenitor cells following carotid balloon catheter injury in rats. Biomed Pharmacother 2024; 177:117022. [PMID: 38917756 DOI: 10.1016/j.biopha.2024.117022] [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: 04/03/2024] [Revised: 06/10/2024] [Accepted: 06/19/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND The transplantation of endothelial progenitor cells (EPCs) has been shown to reduce neointimal hyperplasia following arterial injury. However, the efficacy of this approach is hampered by limited homing of EPCs to the injury site. Additionally, the in vivo recruitment and metabolic activity of transplanted EPCs have not been continuously monitored. METHODS EPCs were labeled with indocyanine green (ICG)-conjugated superparamagnetic iron oxide nanoparticles (SPIONs) and subjected to external magnetic field targeting to enhance their delivery to a carotid balloon injury (BI) model in Sprague-Dawley rats. Magnetic particle imaging (MPI)/ fluorescence imaging (FLI) multimodal in vivo imaging, 3D MPI/CT imaging and MPI/FLI ex vivo imaging was performed after injury. Carotid arteries were collected and analyzed for pathology and immunofluorescence staining. The paracrine effects were analyzed by enzyme-linked immunosorbent assay. RESULTS The application of a magnetic field significantly enhanced the localization and retention of SPIONs@PEG-ICG-EPCs at the site of arterial injury, as evidenced by both in vivo continuous monitoring and ex vivo by observation. This targeted delivery approach effectively inhibited neointimal hyperplasia and increased the presence of CD31-positive cells at the injury site. Moreover, serum levels of SDF-1α, VEGF, IGF-1, and TGF-β1 were significantly elevated, indicating enhanced paracrine activity. CONCLUSIONS Our findings demonstrate that external magnetic field-directed delivery of SPIONs@PEG-ICG-EPCs to areas of arterial injury can significantly enhance their therapeutic efficacy. This enhancement is likely mediated through increased paracrine signaling. These results underscore the potential of magnetically guided SPIONs@PEG-ICG-EPCs delivery as a promising strategy for treating arterial injuries.
Collapse
Affiliation(s)
- Zhongxuan Li
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China; Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing 100853, China
| | - Yingqian Zhang
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Mingrui Ma
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing 100853, China
| | - Wei Wang
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Hui Hui
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing 100190, China; Beijing Key Laboratory of Molecular Imaging, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100080, China; National Key Laboratory of Kidney Diseases, Beijing 100853, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing 100190, China; Beijing Key Laboratory of Molecular Imaging, Beijing 100190, China; School of Engineering Medicine & School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China; Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beijing 100191, China; National Key Laboratory of Kidney Diseases, Beijing 100853, China.
| | - Yundai Chen
- Senior Department of Cardiology, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China.
| |
Collapse
|
3
|
Kologrivova IV, Suslova TE, Koshelskaya OA, Kravchenko ES, Kharitonova OA, Romanova EA, Vyrostkova AI, Boshchenko AA. Intermediate Monocytes and Circulating Endothelial Cells: Interplay with Severity of Atherosclerosis in Patients with Coronary Artery Disease and Type 2 Diabetes Mellitus. Biomedicines 2023; 11:2911. [PMID: 38001912 PMCID: PMC10669450 DOI: 10.3390/biomedicines11112911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
The aim was to investigate the association of monocyte heterogeneity and presence of circulating endothelial cells with the severity of coronary atherosclerosis in patients with coronary artery disease (CAD) and type 2 diabetes mellitus (T2DM). We recruited 62 patients with CAD, including 22 patients with DM2. The severity of atherosclerosis was evaluated using Gensini Score. Numbers of classical (CD14++CD16-), intermediate (CD14++CD16+), and non-classical (CD14+CD16++) monocyte subsets; circulating endothelial progenitor cells; and the presence of circulating endothelial cells were evaluated. Counts and frequencies of intermediate monocytes, but not glycaemia parameters, were associated with the severity of atherosclerosis in diabetic CAD patients (rs = 0.689; p = 0.001 and rs = 0.632; p = 0.002, respectively). Frequency of Tie2+ cells was lower in classical than in non-classical monocytes in CAD patients (p = 0.007), while in patients with association of CAD and T2DM, differences between Tie2+ monocytes subsets disappeared (p = 0.080). Circulating endothelial cells were determined in 100% of CAD+T2DM patients, and counts of CD14++CD16+ monocytes and concentration of TGF-β predicted the presence of circulating endothelial cells (sensitivity 92.3%; specificity 90.9%; AUC = 0.930). Thus, intermediate monocytes represent one of the key determinants of the appearance of circulating endothelial cells in all the patients with CAD, but are associated with the severity of atherosclerosis only in patients with association of CAD and T2DM.
Collapse
Affiliation(s)
- Irina V. Kologrivova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (T.E.S.); (O.A.K.); (E.S.K.); (O.A.K.); (A.A.B.)
| | - Tatiana E. Suslova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (T.E.S.); (O.A.K.); (E.S.K.); (O.A.K.); (A.A.B.)
| | - Olga A. Koshelskaya
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (T.E.S.); (O.A.K.); (E.S.K.); (O.A.K.); (A.A.B.)
| | - Elena S. Kravchenko
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (T.E.S.); (O.A.K.); (E.S.K.); (O.A.K.); (A.A.B.)
| | - Olga A. Kharitonova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (T.E.S.); (O.A.K.); (E.S.K.); (O.A.K.); (A.A.B.)
| | - Ekaterina A. Romanova
- Department of Biomedicine, Siberian State Medical University, 2 Moskovskii trakt, Tomsk 634050, Russia; (E.A.R.); (A.I.V.)
| | - Alexandra I. Vyrostkova
- Department of Biomedicine, Siberian State Medical University, 2 Moskovskii trakt, Tomsk 634050, Russia; (E.A.R.); (A.I.V.)
| | - Alla A. Boshchenko
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (T.E.S.); (O.A.K.); (E.S.K.); (O.A.K.); (A.A.B.)
| |
Collapse
|
4
|
Li T, Wang B, Ding H, Chen S, Cheng W, Li Y, Wu X, Wang L, Jiang Y, Lu Z, Teng Y, Su S, Han X, Zhao M. Effect of Extracellular Vesicles From Multiple Cells on Vascular Smooth Muscle Cells in Atherosclerosis. Front Pharmacol 2022; 13:857331. [PMID: 35620296 PMCID: PMC9127356 DOI: 10.3389/fphar.2022.857331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/05/2022] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis (AS)-related diseases are still the main cause of death in clinical patients. The phenotype switching, proliferation, migration, and secretion of vascular smooth muscle cells (VSMCs) have a pivotal role in atherosclerosis. Although numerous research studies have elucidated the role of VSMCs in AS, their potential functional regulations continue to be explored. The formation of AS involves various cells, such as endothelial cells, smooth muscle cells, and macrophages. Therefore, intercellular communication of blood vessels cannot be ignored due to closely connected endothelia, media, and adventitia. Extracellular vesicles (EVs), as the vectors of cell-to-cell communication, can deliver proteins and nucleic acids of parent cells to the recipient cells. EVs have emerged as being central in intercellular communication and play a vital role in the pathophysiologic mechanisms of AS. This review summarizes the effects of extracellular vesicles (EVs) derived from multiple cells (endothelial cells, macrophages, mesenchymal stem cells, etc.) on VSMCs in AS. The key findings of this review are as follows: 1) endothelial cell–derived EVs (EEVs) have anti- or pro-atherogenic effects on VSMCs; 2) macrophage-derived EVs (MEVs) aggravate the proliferation and migration of VSMCs; 3) mesenchymal stem cells can inhibit VSMCs; and 4) the proliferation and migration of VSMCs can be inhibited by the treatment of EVs with atherosclerosis-protective factors and promoted by noxious stimulants. These results suggested that EVs have the same functional properties as treated parent cells, which might provide vital guidance for treating AS.
Collapse
Affiliation(s)
- Tong Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Baofu Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Hao Ding
- Department of Oncology, Shanxi Traditional Chinese Medical Hospital, Taiyuan, China
| | - Shiqi Chen
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Weiting Cheng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoxiao Wu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yangyang Jiang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ziwen Lu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Teng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Sha Su
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaowan Han
- Department of Cardiac Rehabilitation, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mingjing Zhao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
5
|
Phillippi JA. On vasa vasorum: A history of advances in understanding the vessels of vessels. SCIENCE ADVANCES 2022; 8:eabl6364. [PMID: 35442731 PMCID: PMC9020663 DOI: 10.1126/sciadv.abl6364] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/01/2022] [Indexed: 05/09/2023]
Abstract
The vasa vasorum are a vital microvascular network supporting the outer wall of larger blood vessels. Although these dynamic microvessels have been studied for centuries, the importance and impact of their functions in vascular health and disease are not yet fully realized. There is now rich knowledge regarding what local progenitor cell populations comprise and cohabitate with the vasa vasorum and how they might contribute to physiological and pathological changes in the network or its expansion via angiogenesis or vasculogenesis. Evidence of whether vasa vasorum remodeling incites or governs disease progression or is a consequence of cardiovascular pathologies remains limited. Recent advances in vasa vasorum imaging for understanding cardiovascular disease severity and pathophysiology open the door for theranostic opportunities. Approaches that strive to control angiogenesis and vasculogenesis potentiate mitigation of vasa vasorum-mediated contributions to cardiovascular diseases and emerging diseases involving the microcirculation.
Collapse
Affiliation(s)
- Julie A. Phillippi
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
6
|
Wang W, Zhang Y, Hui H, Tong W, Wei Z, Li Z, Zhang S, Yang X, Tian J, Chen Y. The effect of endothelial progenitor cell transplantation on neointimal hyperplasia and reendothelialisation after balloon catheter injury in rat carotid arteries. Stem Cell Res Ther 2021; 12:99. [PMID: 33536065 PMCID: PMC7860581 DOI: 10.1186/s13287-021-02135-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/01/2021] [Indexed: 12/20/2022] Open
Abstract
Background Reendothelialisation is the natural pathway that inhibits neointimal hyperplasia and in-stent restenosis. Circulating endothelial progenitor cells (EPCs) derived from bone marrow (BM) might contribute to endothelial repair. However, the temporal and spatial distributions of reendothelialisation and neointimal hyperplasia after EPC transplantation in injured arteries are currently unclear. Methods A carotid balloon injury (BI) model was established in Sprague-Dawley rats, and PKH26-labelled BM-derived EPCs were transplanted after BI. The carotid arteries were harvested on the first, fourth, seventh, and 14th day post-injury and analysed via light-sheet fluorescence microscopy and pathological staining (n = 3). EPC and human umbilical vein endothelial cell culture supernatants were collected, and blood samples were collected before and after transplantation. The paracrine effects of VEGF, IGF-1, and TGF-β1 in cell culture supernatants and serum were analysed by enzyme-linked immunosorbent assay (n = 4). Results Transplanted EPCs labelled with PKH26 were attached to the injured luminal surface the first day after BI. In the sham operation group, the transplanted EPCs did not adhere to the luminal surface. From the fourth day after BI, the mean fluorescence intensity of PKH26 decreased significantly. However, reendothelialisation and inhibition of neointimal hyperplasia were significantly promoted by transplanted EPCs. The degree of reendothelialisation of the EPC7d and EPC14d groups was higher than that of the BI7d and BI14d groups, and the difference in neointimal hyperplasia was observed between the EPC14d and BI14d groups. The number of endothelial cells on the luminal surface of the EPC14d group was higher than that of the BI14d group. The number of infiltrated macrophages in the injured artery decreased in the EPC transplanted groups. Conclusions Transplanted EPCs had chemotactic enrichment and attached to the injured arterial luminal surface. Although decreasing significantly after the fourth day at the site of injury after transplantation, transplanted EPCs could still promote reendothelialisation and inhibit neointimal hyperplasia. The underlying mechanism is through paracrine cytokines and not differentiation into mature endothelial cells. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02135-w.
Collapse
Affiliation(s)
- Wei Wang
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, China.,Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China.,CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Yingqian Zhang
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Hui Hui
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wei Tong
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, China.,Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China.,CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Zechen Wei
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhongxuan Li
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Suhui Zhang
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, 100853, China.,Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China.,CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xin Yang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China. .,University of Chinese Academy of Sciences, Beijing, China. .,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, 100083, China.
| | - Yundai Chen
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
7
|
Pashova A, Work LM, Nicklin SA. The role of extracellular vesicles in neointima formation post vascular injury. Cell Signal 2020; 76:109783. [PMID: 32956789 DOI: 10.1016/j.cellsig.2020.109783] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/15/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022]
Abstract
Pathological neointimal growth can develop in patients as a result of vascular injury following percutaneous coronary intervention and coronary artery bypass grafting using autologous saphenous vein, leading to arterial or vein graft occlusion. Neointima formation driven by intimal hyperplasia occurs as a result of a complex interplay between molecular and cellular processes involving different cell types including endothelial cells, vascular smooth muscle cells and various inflammatory cells. Therefore, understanding the intercellular communication mechanisms underlying this process remains of fundamental importance in order to develop therapeutic strategies to preserve endothelial integrity and vascular health post coronary interventions. Extracellular vesicles (EVs), including microvesicles and exosomes, are membrane-bound particles secreted by cells which mediate intercellular signalling in physiological and pathophysiological states, however their role in neointima formation is not fully understood. The purification and characterization techniques currently used in the field are associated with many limitations which significantly hinder the ability to comprehensively study the role of specific EV types and make direct functional comparisons between EV subpopulations. In this review, the current knowledge focusing on EV signalling in neointima formation post vascular injury is discussed.
Collapse
Affiliation(s)
- A Pashova
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow, UK
| | - L M Work
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow, UK
| | - S A Nicklin
- Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
8
|
Rasheed A, Shawky SA, Tsai R, Jung RG, Simard T, Saikali MF, Hibbert B, Rayner KJ, Cummins CL. The secretome of liver X receptor agonist-treated early outgrowth cells decreases atherosclerosis in Ldlr-/- mice. Stem Cells Transl Med 2020; 10:479-491. [PMID: 33231376 PMCID: PMC7900590 DOI: 10.1002/sctm.19-0390] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 09/11/2020] [Accepted: 09/26/2020] [Indexed: 12/14/2022] Open
Abstract
Endothelial progenitor cells (EPCs) promote the maintenance of the endothelium by secreting vasoreparative factors. A population of EPCs known as early outgrowth cells (EOCs) is being investigated as novel cell‐based therapies for the treatment of cardiovascular disease. We previously demonstrated that the absence of liver X receptors (LXRs) is detrimental to the formation and function of EOCs under hypercholesterolemic conditions. Here, we investigate whether LXR activation in EOCs is beneficial for the treatment of atherosclerosis. EOCs were differentiated from the bone marrow of wild‐type (WT) and LXR‐knockout (Lxrαβ−/−) mice in the presence of vehicle or LXR agonist (GW3965). WT EOCs treated with GW3965 throughout differentiation showed reduced mRNA expression of endothelial lineage markers (Cd144, Vegfr2) compared with WT vehicle and Lxrαβ−/− EOCs. GW3965‐treated EOCs produced secreted factors that reduced monocyte adhesion to activated endothelial cells in culture. When injected into atherosclerosis‐prone Ldlr−/− mice, GW3965‐treated EOCs, or their corresponding conditioned media (CM) were both able to reduce aortic sinus plaque burden compared with controls. Furthermore, when human EOCs (obtained from patients with established CAD) were treated with GW3965 and the CM applied to endothelial cells, monocyte adhesion was decreased, indicating that our results in mice could be translated to patients. Ex vivo LXR agonist treatment of EOCs therefore produces a secretome that decreases early atherosclerosis in Ldlr−/− mice, and additionally, CM from human EOCs significantly inhibits monocyte to endothelial adhesion. Thus, active factor(s) within the GW3965‐treated EOC secretome may have the potential to be useful for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Adil Rasheed
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Sarah A Shawky
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Ricky Tsai
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Richard G Jung
- Capital Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Trevor Simard
- Capital Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario, Canada.,Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Michael F Saikali
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin Hibbert
- Capital Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario, Canada.,Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Katey J Rayner
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Banting and Best Diabetes Centre, Toronto, Ontario, Canada.,The Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Lin Y, Zhu W, Chen X. The involving progress of MSCs based therapy in atherosclerosis. Stem Cell Res Ther 2020; 11:216. [PMID: 32503682 PMCID: PMC7275513 DOI: 10.1186/s13287-020-01728-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/25/2020] [Accepted: 05/13/2020] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a chronic progressive vascular inflammation characterized by lipid deposition and plaque formation, for which vascular cell dysfunction and impaired immune responses are involved. Up to now, lipid-lowering drugs remain the main therapy for treating atherosclerosis; however, the surgical or interventional therapy is often applied, and yet, morbidity and mortality of such cardiovascular disease remain high worldwide. Over the past decades, an anti-inflammatory approach has become an important therapeutic target for dealing with atherosclerosis, as altered immune responses have been regarded as an essential player in the pathological process of vascular abnormality induced by hyperlipidemia. Interestingly, mesenchymal stem cells, one type of stem cells with the capabilities of self-renewal and multi-potential, have demonstrated their unique immunomodulatory function in the various pathological process, especially in atherosclerosis. While some controversies remain regarding their therapeutic efficacy and working mechanisms, our present review aims to summarize the current research progress on stem cell-based therapy, focusing on its immunomodulatory effects on the pathogenesis of atherosclerosis and how endothelial cells, smooth muscle cells, and other immune cells are regulated by MSC-based therapy.
Collapse
Affiliation(s)
- Ying Lin
- School of Medicine, Ningbo University, Ningbo, Zhejiang, China.,Department of Cardiology, Ningbo First hospital, Ningbo, Zhejiang, China.,Department of Cardiology and Key Lab of Cardiovascular Disease, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Zhu
- Department of Cardiology and Key Lab of Cardiovascular Disease, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.
| | - Xiaomin Chen
- School of Medicine, Ningbo University, Ningbo, Zhejiang, China. .,Department of Cardiology, Ningbo First hospital, Ningbo, Zhejiang, China.
| |
Collapse
|
10
|
Steffen E, Mayer von Wittgenstein WBE, Hennig M, Niepmann ST, Zietzer A, Werner N, Rassaf T, Nickenig G, Wassmann S, Zimmer S, Steinmetz M. Murine sca1/flk1-positive cells are not endothelial progenitor cells, but B2 lymphocytes. Basic Res Cardiol 2020; 115:18. [PMID: 31980946 PMCID: PMC6981106 DOI: 10.1007/s00395-020-0774-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/02/2020] [Indexed: 12/12/2022]
Abstract
Circulating sca1+/flk1+ cells are hypothesized to be endothelial progenitor cells (EPCs) in mice that contribute to atheroprotection by replacing dysfunctional endothelial cells. Decreased numbers of circulating sca1+/flk1+ cells correlate with increased atherosclerotic lesions and impaired reendothelialization upon electric injury of the common carotid artery. However, legitimate doubts remain about the identity of the putative EPCs and their contribution to endothelial restoration. Hence, our study aimed to establish a phenotype for sca1+/flk1+ cells to gain a better understanding of their role in atherosclerotic disease. In wild-type mice, sca1+/flk1+ cells were mobilized into the peripheral circulation by granulocyte-colony stimulating factor (G-CSF) treatment and this movement correlated with improved endothelial regeneration upon carotid artery injury. Multicolor flow cytometry analysis revealed that sca1+/flk1+ cells predominantly co-expressed surface markers of conventional B cells (B2 cells). In RAG2-deficient mice and upon B2 cell depletion, sca1+/flk1+ cells were fully depleted. In the absence of monocytes, sca1+/flk1+ cell levels were unchanged. A PCR array focused on cell surface markers and next-generation sequencing (NGS) of purified sca1+/flk1+ cells confirmed their phenotype to be predominantly that of B cells. Finally, the depletion of B2 cells, including sca1+/flk1+ cells, in G-CSF-treated wild-type mice partly abolished the endothelial regenerating effect of G-CSF, indicating an atheroprotective role for sca1+/flk1+ B2 cells. In summary, we characterized sca1+/flk1+ cells as a subset of predominantly B2 cells, which are apparently involved in endothelial regeneration.
Collapse
Affiliation(s)
- Eva Steffen
- Herzzentrum Bonn, Medizinische Klinik und Poliklinik II, Universitätsklinikum Bonn, Venusberg Campus 1, 53127, Bonn, Germany.
| | | | - Marie Hennig
- Herzzentrum Bonn, Medizinische Klinik und Poliklinik II, Universitätsklinikum Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Sven Thomas Niepmann
- Herzzentrum Bonn, Medizinische Klinik und Poliklinik II, Universitätsklinikum Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Andreas Zietzer
- Herzzentrum Bonn, Medizinische Klinik und Poliklinik II, Universitätsklinikum Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Nikos Werner
- Krankenhaus der Barmherzigen Brüder, Innere Medizin III, Trier, Germany
| | - Tienush Rassaf
- Westdeutsches Herz- und Gefäßzentrum, Klinik für Kardiologie und Angiologie, Universitätsklinikum Essen, Essen, Germany
| | - Georg Nickenig
- Herzzentrum Bonn, Medizinische Klinik und Poliklinik II, Universitätsklinikum Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Sven Wassmann
- Cardiology Pasing, Munich, Germany.,University of the Saarland, Homburg, Saar, Germany
| | - Sebastian Zimmer
- Herzzentrum Bonn, Medizinische Klinik und Poliklinik II, Universitätsklinikum Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Martin Steinmetz
- Westdeutsches Herz- und Gefäßzentrum, Klinik für Kardiologie und Angiologie, Universitätsklinikum Essen, Essen, Germany
| |
Collapse
|
11
|
Yi M, Wu Y, Long J, Liu F, Liu Z, Zhang YH, Sun XP, Fan ZX, Gao J, Si J, Zuo XB, Zhang LM, Shi N, Miao ZP, Bai ZR, Liu BY, Liu HR, Li J. Exosomes secreted from osteocalcin-overexpressing endothelial progenitor cells promote endothelial cell angiogenesis. Am J Physiol Cell Physiol 2019; 317:C932-C941. [PMID: 31411920 DOI: 10.1152/ajpcell.00534.2018] [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: 01/13/2023]
Abstract
Exosome secretion is an important paracrine way of endothelial progenitor cells (EPCs) to modulate resident endothelial cells. The osteocalcin (OCN)-expressing EPCs have been found to be increased in cardiovascular disease patients and are considered to be involved in the process of coronary atherosclerosis. Since OCN has been proven to prevent endothelial dysfunction, this study aimed to evaluate the effect of exosomes derived from OCN-overexpressed EPCs on endothelial cells. Exosomes derived from EPCs (Exos) and OCN-overexpressed EPCs (OCN-Exos) were isolated and incubated with rat aorta endothelial cells (RAOECs) with or without the inhibition of OCN receptor G protein-coupled receptor family C group 6 member A (GPRC6A). The effects of exosomes on the proliferation activity of endothelial cells were evaluated by CCK-8 assay, and the migration of endothelial cells was detected by wound healing assay. A tube formation assay was used to test the influence of exosomes on the angiogenesis performance of endothelial cells. Here, we presented that OCN was packed into Exos and was able to be transferred to the RAOECs via exosome incorporation, which was increased in OCN-Exos groups. Compared with Exos, OCN-Exos had better efficiency in promoting RAOEC proliferation and migration and tube formation. The promoting effects were impeded after the inhibition of GPRC6A expression in RAOECs. These data suggest that exosomes from OCN-overexpressed EPCs have a beneficial regulating effect on endothelial cells, which involved enhanced OCN-GPRC6A signaling.
Collapse
Affiliation(s)
- Ming Yi
- Division of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China.,Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Beijing, People's Republic of China
| | - Ye Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China.,Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Beijing, People's Republic of China
| | - Jun Long
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Fei Liu
- Division of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Zhi Liu
- Division of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Ying-Hua Zhang
- Division of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xi-Peng Sun
- Division of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Zhen-Xing Fan
- Division of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jing Gao
- Division of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jin Si
- Division of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xue-Bing Zuo
- Division of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Lei-Min Zhang
- Division of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Ning Shi
- Division of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Zu-Pei Miao
- Division of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Zhao-Run Bai
- Division of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Bin-Yu Liu
- Division of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Hui-Rong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China.,Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Beijing, People's Republic of China
| | - Jing Li
- Division of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China.,Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Beijing, People's Republic of China
| |
Collapse
|
12
|
Lu W, Li X. PDGFs and their receptors in vascular stem/progenitor cells: Functions and therapeutic potential in retinal vasculopathy. Mol Aspects Med 2018; 62:22-32. [DOI: 10.1016/j.mam.2017.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/04/2017] [Indexed: 02/07/2023]
|
13
|
Exosomes of Endothelial Progenitor Cells Inhibit Neointima Formation After Carotid Artery Injury. J Surg Res 2018; 232:398-407. [PMID: 30463748 DOI: 10.1016/j.jss.2018.06.066] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 06/04/2018] [Accepted: 06/20/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Exosomes released from endothelial progenitor cells (EPCs) play a protective role in various disease models. Both endothelial cell (EC) damage and smooth muscle cell (SMC) proliferation are involved in the pathological process of restenosis after angioplasty and stenting. Few studies have focused on the therapeutic role of exosomes in EC damage and SMC proliferation. In this study, we sought to investigate the effect of exosomes released by human fetal aorta-derived EPCs on the rat carotid artery balloon injury model in vivo. We also sought to determine the effect of exosomes on both ECs and SMCs in vitro. METHODS Exosomes (Exo group) or saline (Con group) were injected in rat carotid balloon injury model animals. The rats were sacrificed after 2, 4, 14, and 28 d, and injured carotid specimens were collected for Evans blue staining, hematoxylin-eosin staining, and immunohistochemistry. RESULTS When the Con group and the Exo group were compared, the reendothelialized areas were not significantly different after 2 or 4 d, as shown by Evans blue staining. The hematoxylin-eosin results showed that the intimal to medial area ratio was slightly but not significantly higher in the Exo group after 2 and 4 d. The immunohistochemistry results showed that the proliferation of SMCs was slightly higher in the Exo group after 2 and 4 d, but the difference was not significant. The reendothelialization area of the Con group was significantly smaller than that of the Exo group at day 14. Both the intimal to medial area ratio and SMC proliferation in the Exo group were significantly smaller than those of the Con group at 14 or 28 d. In the in vitro study, exosome treatment significantly enhanced the proliferation and migration of both ECs and SMCs. CONCLUSIONS Exosomes derived from EPCs could inhibit neointimal hyperplasia after carotid artery injury in rats. The protective effect of exosomes may manifest through the promotion of EC repair rather than direct suppression of proliferation and migration of smooth muscles cells.
Collapse
|
14
|
Rasheed A, Tsai R, Cummins CL. Loss of the Liver X Receptors Disrupts the Balance of Hematopoietic Populations, With Detrimental Effects on Endothelial Progenitor Cells. J Am Heart Assoc 2018; 7:JAHA.117.007787. [PMID: 29739800 PMCID: PMC6015321 DOI: 10.1161/jaha.117.007787] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background The liver X receptors (LXRs; α/β) are nuclear receptors known to regulate cholesterol homeostasis and the production of select hematopoietic populations. The objective of this study was to determine the importance of LXRs and a high‐fat high‐cholesterol diet on global hematopoiesis, with special emphasis on endothelial progenitor cells (EPCs), a vasoreparative cell type that is derived from bone marrow hematopoietic stem cells. Methods and Results Wild‐type and LXR double‐knockout (Lxrαβ−/−) mice were fed a Western diet (WD) to increase plasma cholesterol levels. In WD‐fed Lxrαβ−/− mice, flow cytometry and complete blood cell counts revealed that hematopoietic stem cells, a myeloid progenitor, and mature circulating myeloid cells were increased; EPC numbers were significantly decreased. Hematopoietic stem cells from WD‐fed Lxrαβ−/− mice showed increased cholesterol content, along with increased myeloid colony formation compared with chow‐fed mice. In contrast, EPCs from WD‐fed Lxrαβ−/− mice also demonstrated increased cellular cholesterol content that was associated with greater expression of the endothelial lineage markers Cd144 and Vegfr2, suggesting accelerated differentiation of the EPCs. Treatment of human umbilical vein endothelial cells with conditioned medium collected from these EPCs increased THP‐1 monocyte adhesion. Increased monocyte adhesion to conditioned medium–treated endothelial cells was recapitulated with conditioned medium from Lxrαβ−/−EPCs treated with cholesterol ex vivo, suggesting cholesterol is the main component of the WD inducing EPC dysfunction. Conclusions LXRs are crucial for maintaining the balance of hematopoietic cells in a hypercholesterolemic environment and for mitigating the negative effects of cholesterol on EPC differentiation/secretome changes that promote monocyte‐endothelial adhesion.
Collapse
Affiliation(s)
- Adil Rasheed
- Department of Pharmaceutical Sciences, University of Toronto, Ontario, Canada
| | - Ricky Tsai
- Department of Pharmaceutical Sciences, University of Toronto, Ontario, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, University of Toronto, Ontario, Canada .,Banting and Best Diabetes Centre, Toronto, Ontario, Canada.,The Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario, Canada
| |
Collapse
|
15
|
Yoder MC. Endothelial stem and progenitor cells (stem cells): (2017 Grover Conference Series). Pulm Circ 2018; 8:2045893217743950. [PMID: 29099663 PMCID: PMC5731724 DOI: 10.1177/2045893217743950] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 10/31/2017] [Indexed: 12/11/2022] Open
Abstract
The capacity of existing blood vessels to give rise to new blood vessels via endothelial cell sprouting is called angiogenesis and is a well-studied biologic process. In contrast, little is known about the mechanisms for endothelial cell replacement or regeneration within established blood vessels. Since clear definitions exist for identifying cells with stem and progenitor cell properties in many tissues and organs of the body, several groups have begun to accumulate evidence that endothelial stem and progenitor cells exist within the endothelial intima of existing blood vessels. This paper will review stem and progenitor cell definitions and highlight several recent papers purporting to have identified resident vascular endothelial stem and progenitor cells.
Collapse
Affiliation(s)
- Mervin C. Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
16
|
Wang D, Li LK, Dai T, Wang A, Li S. Adult Stem Cells in Vascular Remodeling. Am J Cancer Res 2018; 8:815-829. [PMID: 29344309 PMCID: PMC5771096 DOI: 10.7150/thno.19577] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 10/01/2017] [Indexed: 01/03/2023] Open
Abstract
Understanding the contribution of vascular cells to blood vessel remodeling is critical for the development of new therapeutic approaches to cure cardiovascular diseases (CVDs) and regenerate blood vessels. Recent findings suggest that neointimal formation and atherosclerotic lesions involve not only inflammatory cells, endothelial cells, and smooth muscle cells, but also several types of stem cells or progenitors in arterial walls and the circulation. Some of these stem cells also participate in the remodeling of vascular grafts, microvessel regeneration, and formation of fibrotic tissue around biomaterial implants. Here we review the recent findings on how adult stem cells participate in CVD development and regeneration as well as the current state of clinical trials in the field, which may lead to new approaches for cardiovascular therapies and tissue engineering.
Collapse
|
17
|
Bentzon JF, Daemen M, Falk E, Garcia-Garcia HM, Herrmann J, Hoefer I, Jukema JW, Krams R, Kwak BR, Marx N, Naruszewicz M, Newby A, Pasterkamp G, Serruys PWJC, Waltenberger J, Weber C, Tokgözoglu L, Ylä-Herttuala S. Stabilisation of atherosclerotic plaques. Thromb Haemost 2017; 106:1-19. [DOI: 10.1160/th10-12-0784] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Accepted: 04/29/2011] [Indexed: 01/04/2023]
Abstract
SummaryPlaque rupture and subsequent thrombotic occlusion of the coronary artery account for as many as three quarters of myocardial infarctions. The concept of plaque stabilisation emerged about 20 years ago to explain the discrepancy between the reduction of cardiovascular events in patients receiving lipid lowering therapy and the small decrease seen in angiographic evaluation of atherosclerosis. Since then, the concept of a vulnerable plaque has received a lot of attention in basic and clinical research leading to a better understanding of the pathophysiology of the vulnerable plaque and acute coronary syndromes. From pathological and clinical observations, plaques that have recently ruptured have thin fibrous caps, large lipid cores, exhibit outward remodelling and invasion by vasa vasorum. Ruptured plaques are also focally inflamed and this may be a common denominator of the other pathological features. Plaques with similar characteristics, but which have not yet ruptured, are believed to be vulnerable to rupture. Experimental studies strongly support the validity of anti-inflammatory approaches to promote plaque stability. Unfortunately, reliable non-invasive methods for imaging and detection of such plaques are not yet readily available. There is a strong biological basis and supportive clinical evidence that low-density lipoprotein lowering with statins is useful for the stabilisation of vulnerable plaques. There is also some clinical evidence for the usefulness of antiplatelet agents, beta blockers and renin-angiotensin-aldosterone system inhibitors for plaque stabilisation. Determining the causes of plaque rupture and designing diagnostics and interventions to prevent them are urgent priorities for current basic and clinical research in cardiovascular area.
Collapse
|
18
|
Daugherty A, Tall AR, Daemen MJ, Falk E, Fisher EA, García-Cardeña G, Lusis AJ, Owens AP, Rosenfeld ME, Virmani R. Recommendation on Design, Execution, and Reporting of Animal Atherosclerosis Studies: A Scientific Statement From the American Heart Association. Circ Res 2017; 121:e53-e79. [DOI: 10.1161/res.0000000000000169] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Animal studies are a foundation for defining mechanisms of atherosclerosis and potential targets of drugs to prevent lesion development or reverse the disease. In the current literature, it is common to see contradictions of outcomes in animal studies from different research groups, leading to the paucity of extrapolations of experimental findings into understanding the human disease. The purpose of this statement is to provide guidelines for development and execution of experimental design and interpretation in animal studies. Recommendations include the following: (1) animal model selection, with commentary on the fidelity of mimicking facets of the human disease; (2) experimental design and its impact on the interpretation of data; and (3) standard methods to enhance accuracy of measurements and characterization of atherosclerotic lesions.
Collapse
|
19
|
Abstract
The introduction of molecularly targeted therapies with tyrosine kinase inhibitors has revolutionized cancer therapy and has contributed to a steady decline in cancer-related mortality since the late 1990s. However, not only cardiac but also vascular toxicity has been reported for these agents, some as expected on-target effects (e.g., VEGF receptor inhibitors) and others as unanticipated events (e.g., BCR-Abl inhibitors). A sound understanding of these cardiovascular toxic effects is critical to advance mechanistic insight into vascular disease and clinical care. From a conceptual standpoint, there might be value in defining type I (permanent) and type II (transient) vascular toxicity. This review will focus on the tyrosine kinase inhibitors in current clinical use and their associated vascular side effects.
Collapse
|
20
|
Daugherty A, Tall AR, Daemen MJAP, Falk E, Fisher EA, García-Cardeña G, Lusis AJ, Owens AP, Rosenfeld ME, Virmani R. Recommendation on Design, Execution, and Reporting of Animal Atherosclerosis Studies: A Scientific Statement From the American Heart Association. Arterioscler Thromb Vasc Biol 2017; 37:e131-e157. [PMID: 28729366 DOI: 10.1161/atv.0000000000000062] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Animal studies are a foundation for defining mechanisms of atherosclerosis and potential targets of drugs to prevent lesion development or reverse the disease. In the current literature, it is common to see contradictions of outcomes in animal studies from different research groups, leading to the paucity of extrapolations of experimental findings into understanding the human disease. The purpose of this statement is to provide guidelines for development and execution of experimental design and interpretation in animal studies. Recommendations include the following: (1) animal model selection, with commentary on the fidelity of mimicking facets of the human disease; (2) experimental design and its impact on the interpretation of data; and (3) standard methods to enhance accuracy of measurements and characterization of atherosclerotic lesions.
Collapse
|
21
|
Green L, Ofstein RH, Rapp B, Saadatzadeh MR, Bhavsar JR, Fajardo A, Dalsing MC, Ingram DA, Murphy MP. Adult venous endothelium is a niche for highly proliferative and vasculogenic endothelial colony-forming cells. J Vasc Surg 2017; 66:1854-1863. [PMID: 28655551 DOI: 10.1016/j.jvs.2016.11.059] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 11/30/2016] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Postnatal resident endothelium of blood vessels has been proposed to represent terminally differentiated tissue that does not replicate. We previously isolated endothelial colony-forming cells (ECFCs) from human umbilical cord blood (CB) and term placenta by using colony-forming assays and immunocytochemistry. We showed that ECFCs are highly proliferative and form functioning vessels in vivo, the defining characteristics of a true endothelial progenitor cell. This exploratory investigation was conducted to determine whether the endothelium of healthy adult blood vessels contained resident ECFCs. METHODS The endothelium of great saphenous vein (GSV) obtained from vein stripping procedures was collected with mechanical scraping, and ECFCs were isolated according to established protocols. RESULTS GSV ECFCs incorporated acetylated low-density lipoprotein, formed tubules in Matrigel (BD Biosciences, San Jose, Calif) at 24 hours, and expressed endothelial antigens cluster of differentiation (CD) 144, CD31, CD105, and kinase insert domain receptor but not hematopoietic antigen CD45. Using cumulative population doublings and single-cell assays, we demonstrated that GSV ECFCs exhibited comparable proliferative capacities compared with CB ECFCs, including similar numbers of highly proliferative cells. When injected in collagen/fibronectin gels implanted in nonobese diabetic/severe combined immune deficiency mice, GSV ECFCs formed blood vessels with circulating murine red blood cells, demonstrating their vasculogenic potential. CONCLUSIONS The ECFCs of the GSV contain a hierarchy of progenitor cells with a comparable number of highly proliferative clones as ECFCs of CB. The results of this investigation demonstrate that the adult endothelium contains resident progenitor cells that may have a critical role in vascular homeostasis and repair and could potentially be used as a source of autologous cells for cell therapies focusing on vasculogenesis.
Collapse
Affiliation(s)
- Linden Green
- Health Center for Aortic Disease, Indiana University Health Center for Aortic Disease, Indianapolis, Ind; Department of Cellular and Integrative Physiology, Indiana University Health Center for Aortic Disease, Indianapolis, Ind.
| | - Richard H Ofstein
- Department of Surgery, Indiana University Health Center for Aortic Disease, Indianapolis, Ind
| | - Brian Rapp
- Department of Surgery, Indiana University Health Center for Aortic Disease, Indianapolis, Ind
| | - M Reza Saadatzadeh
- Department of Surgery, Indiana University Health Center for Aortic Disease, Indianapolis, Ind; Indiana Center for Vascular Biology and Medicine, Indiana University Health Center for Aortic Disease, Indianapolis, Ind
| | - Janak R Bhavsar
- Department of Surgery, Indiana University Health Center for Aortic Disease, Indianapolis, Ind; Indiana Center for Vascular Biology and Medicine, Indiana University Health Center for Aortic Disease, Indianapolis, Ind
| | - Andres Fajardo
- Department of Surgery, Indiana University Health Center for Aortic Disease, Indianapolis, Ind
| | - Michael C Dalsing
- Department of Surgery, Indiana University Health Center for Aortic Disease, Indianapolis, Ind
| | - David A Ingram
- Indiana Center for Vascular Biology and Medicine, Indiana University Health Center for Aortic Disease, Indianapolis, Ind; Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University Health Center for Aortic Disease, Indianapolis, Ind; Department of Biochemistry and Molecular Biology, Indiana University Health Center for Aortic Disease, Indianapolis, Ind
| | - Michael P Murphy
- Health Center for Aortic Disease, Indiana University Health Center for Aortic Disease, Indianapolis, Ind; Department of Cellular and Integrative Physiology, Indiana University Health Center for Aortic Disease, Indianapolis, Ind; Department of Surgery, Indiana University Health Center for Aortic Disease, Indianapolis, Ind; Indiana Center for Vascular Biology and Medicine, Indiana University Health Center for Aortic Disease, Indianapolis, Ind
| |
Collapse
|
22
|
Leukocyte Overexpression of Intracellular NAMPT Attenuates Atherosclerosis by Regulating PPARγ-Dependent Monocyte Differentiation and Function. Arterioscler Thromb Vasc Biol 2017; 37:1157-1167. [DOI: 10.1161/atvbaha.116.308187] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 03/28/2017] [Indexed: 11/16/2022]
Abstract
Objective—
Extracellular nicotinamide phosphoribosyltransferase (eNAMPT) mediates inflammatory and potentially proatherogenic effects, whereas the role of intracellular NAMPT (iNAMPT), the rate limiting enzyme in the salvage pathway of nicotinamide adenine dinucleotide (NAD)
+
generation, in atherogenesis is largely unknown. Here we investigated the effects of iNAMPT overexpression in leukocytes on inflammation and atherosclerosis.
Approach and Results—
Low-density lipoprotein receptor–deficient mice with hematopoietic overexpression of human iNAMPT (iNAMPT
hi
), on a western type diet, showed attenuated plaque burden with features of lesion stabilization. This anti-atherogenic effect was caused by improved resistance of macrophages to apoptosis by attenuated chemokine (C–C motif) receptor 2-dependent monocyte chemotaxis and by skewing macrophage polarization toward an anti-inflammatory M2 phenotype. The iNAMPT
hi
phenotype was almost fully reversed by treatment with the NAMPT inhibitor FK866, indicating that iNAMPT catalytic activity is instrumental in the atheroprotection. Importantly, iNAMPT overexpression did not induce any increase in eNAMPT, and eNAMPT had no effect on chemokine (C–C motif) receptor 2 expression and promoted an inflammatory M1 phenotype in macrophages. The iNAMPT-mediated effects at least partly involved sirtuin 1–dependent molecular crosstalk of NAMPT and peroxisome proliferator–activated receptor γ. Finally, iNAMPT and peroxisome proliferator–activated receptor γ showed a strong correlation in human atherosclerotic, but not healthy arteries, hinting to a relevance of iNAMPT/peroxisome proliferator–activated receptor γ pathway also in human carotid atherosclerosis.
Conclusions—
This study highlights the functional dichotomy of intracellular versus extracellular NAMPT, and unveils a critical role for the iNAMPT–peroxisome proliferator–activated receptor γ axis in atherosclerosis.
Collapse
|
23
|
Otto S, Nitsche K, Jung C, Kryvanos A, Zhylka A, Heitkamp K, Gutiérrez-Chico JL, Goebel B, Schulze PC, Figulla HR, Poerner TC. Endothelial progenitor cells and plaque burden in stented coronary artery segments: an optical coherence tomography study six months after elective PCI. BMC Cardiovasc Disord 2017; 17:103. [PMID: 28441929 PMCID: PMC5405468 DOI: 10.1186/s12872-017-0534-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 04/08/2017] [Indexed: 02/03/2023] Open
Abstract
Background Endothelial progenitor cells (EPC) are involved in neovascularization and endothelial integrity. They might be protective in atherosclerosis. Optical coherence tomography (OCT) is a precise intracoronary imaging modality that allows assessment of subintimal plaque development. We evaluated the influence of EPC on coronary plaque burden in stable disease and implemented a novel computational plaque analysis algorithm using OCT. Methods Forty-three patients (69.8% males, 69.6 ± 7.7 years) were investigated by OCT during re-angiography 6 months after elective stent implantation. Different subpopulations of EPCs were identified by flow cytometry according to their co-expression of antigens (CD34+, CD133+, kinase domain receptor, KDR+). An algorithm was applied to calculate the underlying total plaque burden of the stented segments from OCT images. Plaque morphology was assessed according to international consensus in OCT imaging. Results A cumulative sub-strut plaque volume of 10.87 ± 12.7 mm3 and a sub-stent plaque area of 16.23 ± 17.0 mm2 were found within the stented vessel segments with no significant differences between different stent types. All EPC subpopulations (mean of EPC levels: CD34+/CD133+: 2.66 ± 2.0%; CD34+/KDR+: 7.50 ± 5.0%; CD34+/CD133+/KDR+: 1.12 ± 1.0%) inversely correlated with the identified underlying total plaque volume and plaque area (p ≤ 0.012). Conclusions This novel analysis algorithm allows for the first time comprehensive quantification of coronary plaque burden by OCT and illustration as spread out vessel charts. Increased EPC levels are associated with less sub-stent coronary plaque burden which adds to previous findings of their protective role in atherosclerosis.
Collapse
Affiliation(s)
- Sylvia Otto
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, University Hospital Jena Friedrich-Schiller-University Jena, Am Klinikum 1, 07747, Jena, Germany.
| | - Kristina Nitsche
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, University Hospital Jena Friedrich-Schiller-University Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Christian Jung
- Department of Cardiology, Pulmonology, University, Duesseldorf, Medical Faculty and Vascular Diseases, Düsseldorf, Germany
| | | | - Andrey Zhylka
- Belarusian State University, Faculty of Applied Mathematics and Computer Science, Minsk, Belarus
| | - Kerstin Heitkamp
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, University Hospital Jena Friedrich-Schiller-University Jena, Am Klinikum 1, 07747, Jena, Germany
| | | | - Björn Goebel
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, University Hospital Jena Friedrich-Schiller-University Jena, Am Klinikum 1, 07747, Jena, Germany
| | - P Christian Schulze
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, University Hospital Jena Friedrich-Schiller-University Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Hans R Figulla
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, University Hospital Jena Friedrich-Schiller-University Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Tudor C Poerner
- Department of Internal Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, University Hospital Jena Friedrich-Schiller-University Jena, Am Klinikum 1, 07747, Jena, Germany
| |
Collapse
|
24
|
Mierke J, Christoph M, Pfluecke C, Jellinghaus S, Wunderlich C, Strasser RH, Ibrahim K, Poitz DM. Atheroprotective role of Caveolin-1 and eNOS in an innovative transplantation model is mainly mediated by local effects. Biochim Biophys Acta Mol Basis Dis 2017; 1863:529-536. [DOI: 10.1016/j.bbadis.2016.11.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 11/03/2016] [Accepted: 11/29/2016] [Indexed: 12/16/2022]
|
25
|
Santini MP, Forte E, Harvey RP, Kovacic JC. Developmental origin and lineage plasticity of endogenous cardiac stem cells. Development 2016; 143:1242-58. [PMID: 27095490 DOI: 10.1242/dev.111591] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Over the past two decades, several populations of cardiac stem cells have been described in the adult mammalian heart. For the most part, however, their lineage origins and in vivo functions remain largely unexplored. This Review summarizes what is known about different populations of embryonic and adult cardiac stem cells, including KIT(+), PDGFRα(+), ISL1(+)and SCA1(+)cells, side population cells, cardiospheres and epicardial cells. We discuss their developmental origins and defining characteristics, and consider their possible contribution to heart organogenesis and regeneration. We also summarize the origin and plasticity of cardiac fibroblasts and circulating endothelial progenitor cells, and consider what role these cells have in contributing to cardiac repair.
Collapse
Affiliation(s)
- Maria Paola Santini
- Cardiovascular Research Centre, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Elvira Forte
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst 2010, Australia St Vincent's Clinical School, University of New South Wales, Kensington 2052, Australia Stem Cells Australia, Melbourne Brain Centre, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Richard P Harvey
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst 2010, Australia St Vincent's Clinical School, University of New South Wales, Kensington 2052, Australia Stem Cells Australia, Melbourne Brain Centre, The University of Melbourne, Parkville, Victoria 3010, Australia School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington 2052, Australia
| | - Jason C Kovacic
- Cardiovascular Research Centre, Icahn School of Medicine at Mount Sinai, New York City, NY, USA Stem Cells Australia, Melbourne Brain Centre, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
26
|
Fadini GP, Ciciliot S, Albiero M. Concise Review: Perspectives and Clinical Implications of Bone Marrow and Circulating Stem Cell Defects in Diabetes. Stem Cells 2016; 35:106-116. [PMID: 27401837 DOI: 10.1002/stem.2445] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 05/27/2016] [Accepted: 05/28/2016] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus is a complex systemic disease characterized by severe morbidity and excess mortality. The burden of its multiorgan complications relies on an imbalance between hyperglycemic cell damage and defective endogenous reparative mechanisms. Inflammation and abnormalities in several hematopoietic components are typically found in diabetes. The discovery that diabetes reduces circulating stem/progenitor cells and impairs their function has opened an entire new field of study where diabetology comes into contact with hematology and regenerative medicine. It is being progressively recognized that such rare circulating cell populations mirror finely regulated processes involved in hematopoiesis, immunosurveillance, and peripheral tissue homeostasis. From a clinical perspective, pauperization of circulating stem cells predicts adverse outcomes and death. Furthermore, studies in murine models and humans have identified the bone marrow (BM) as a previously neglected site of diabetic end-organ damage, characterized by microangiopathy, neuropathy, fat deposition, and inflammation. As a result, diabetes impairs the mobilization of BM stem/progenitor cells, a defect known as mobilopathy or myelokathexis, with negative consequences for physiologic hematopoiesis, immune regulation, and tissue regeneration. A better understanding of the molecular and cellular processes that govern the BM stem cell niche, cell mobilization, and kinetics in peripheral tissues may uncover new therapeutic strategies for patients with diabetes. This concise review summarizes the current knowledge on the interplay between the BM, circulating stem cells, and diabetes, and sets the stages for future developments in the field. Stem Cells 2017;35:106-116.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, and Venetian Institute of Molecular Medicine, Padova, 35128, Italy
| | - Stefano Ciciliot
- Department of Medicine, University of Padova, and Venetian Institute of Molecular Medicine, Padova, 35128, Italy
| | - Mattia Albiero
- Department of Medicine, University of Padova, and Venetian Institute of Molecular Medicine, Padova, 35128, Italy
| |
Collapse
|
27
|
Cahill PA, Redmond EM. Vascular endothelium - Gatekeeper of vessel health. Atherosclerosis 2016; 248:97-109. [PMID: 26994427 PMCID: PMC6478391 DOI: 10.1016/j.atherosclerosis.2016.03.007] [Citation(s) in RCA: 354] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 03/03/2016] [Accepted: 03/04/2016] [Indexed: 02/08/2023]
Abstract
The vascular endothelium is an interface between the blood stream and the vessel wall. Changes in this single cell layer of the artery wall are believed of primary importance in the pathogenesis of vascular disease/atherosclerosis. The endothelium responds to humoral, neural and especially hemodynamic stimuli and regulates platelet function, inflammatory responses, vascular smooth muscle cell growth and migration, in addition to modulating vascular tone by synthesizing and releasing vasoactive substances. Compromised endothelial function contributes to the pathogenesis of cardiovascular disease; endothelial 'dysfunction' is associated with risk factors, correlates with disease progression, and predicts cardiovascular events. Therapies for atherosclerosis have been developed, therefore, that are directed towards improving endothelial function.
Collapse
Affiliation(s)
- Paul A Cahill
- Vascular Biology and Therapeutics Laboratory, Dublin City University, Dublin, Ireland
| | - Eileen M Redmond
- Department of Surgery, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
28
|
Uryga AK, Bennett MR. Ageing induced vascular smooth muscle cell senescence in atherosclerosis. J Physiol 2015; 594:2115-24. [PMID: 26174609 DOI: 10.1113/jp270923] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 07/08/2015] [Indexed: 12/16/2022] Open
Abstract
Atherosclerosis is a disease of ageing in that its incidence and prevalence increase with age. However, atherosclerosis is also associated with biological ageing, manifest by a number of typical hallmarks of ageing in the atherosclerotic plaque. Thus, accelerated biological ageing may be superimposed on the effects of chronological ageing in atherosclerosis. Tissue ageing is seen in all cells that comprise the plaque, but particularly in vascular smooth muscle cells (VSMCs). Hallmarks of ageing include evidence of cell senescence, DNA damage (including telomere attrition), mitochondrial dysfunction, a pro-inflammatory secretory phenotype, defects in proteostasis, epigenetic changes, deregulated nutrient sensing, and exhaustion of progenitor cells. In this model, initial damage to DNA (genomic, telomeric, mitochondrial and epigenetic changes) results in a number of cellular responses (cellular senescence, deregulated nutrient sensing and defects in proteostasis). Ultimately, ongoing damage and attempts at repair by continued proliferation overwhelm reparative capacity, causing loss of specialised cell functions, cell death and inflammation. This review summarises the evidence for accelerated biological ageing in atherosclerosis, the functional consequences of cell ageing on cells comprising the plaque, and the causal role that VSMC senescence plays in atherogenesis.
Collapse
Affiliation(s)
- Anna K Uryga
- Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, Box 110, Cambridge, CB2 0QQ, UK
| | - Martin R Bennett
- Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, Box 110, Cambridge, CB2 0QQ, UK
| |
Collapse
|
29
|
Ikutomi M, Sahara M, Nakajima T, Minami Y, Morita T, Hirata Y, Komuro I, Nakamura F, Sata M. Diverse contribution of bone marrow-derived late-outgrowth endothelial progenitor cells to vascular repair under pulmonary arterial hypertension and arterial neointimal formation. J Mol Cell Cardiol 2015; 86:121-35. [DOI: 10.1016/j.yjmcc.2015.07.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 07/10/2015] [Accepted: 07/24/2015] [Indexed: 01/29/2023]
|
30
|
Lupieri A, Smirnova N, Malet N, Gayral S, Laffargue M. PI3K signaling in arterial diseases: Non redundant functions of the PI3K isoforms. Adv Biol Regul 2015; 59:4-18. [PMID: 26238239 DOI: 10.1016/j.jbior.2015.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 06/15/2015] [Accepted: 06/15/2015] [Indexed: 06/04/2023]
Abstract
Cardiovascular diseases are the most common cause of death around the world. This includes atherosclerosis and the adverse effects of its treatment, such as restenosis and thrombotic complications. The development of these arterial pathologies requires a series of highly-intertwined interactions between immune and arterial cells, leading to specific inflammatory and fibroproliferative cellular responses. In the last few years, the study of phosphoinositide 3-kinase (PI3K) functions has become an attractive area of investigation in the field of arterial diseases, especially since inhibitors of specific PI3K isoforms have been developed. The PI3K family includes 8 members divided into classes I, II or III depending on their substrate specificity. Although some of the different isoforms are responsible for the production of the same 3-phosphoinositides, they each have specific, non-redundant functions as a result of differences in expression levels in different cell types, activation mechanisms and specific subcellular locations. This review will focus on the functions of the different PI3K isoforms that are suspected as having protective or deleterious effects in both the various immune cells and types of cell found in the arterial wall. It will also discuss our current understanding in the context of which PI3K isoform(s) should be targeted for future therapeutic interventions to prevent or treat arterial diseases.
Collapse
Affiliation(s)
- Adrien Lupieri
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Natalia Smirnova
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Nicole Malet
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Stéphanie Gayral
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Muriel Laffargue
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France.
| |
Collapse
|
31
|
Madonna R, De Caterina R. Circulating endothelial progenitor cells: Do they live up to their name? Vascul Pharmacol 2015; 67-69:2-5. [DOI: 10.1016/j.vph.2015.02.018] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 02/11/2015] [Accepted: 02/14/2015] [Indexed: 10/23/2022]
|
32
|
Steinmetz M, Lucanus E, Zimmer S, Nickenig G, Werner N. Mobilization of sca1/flk-1 positive endothelial progenitor cells declines in apolipoprotein E-deficient mice with a high-fat diet. J Cardiol 2015; 66:532-8. [PMID: 25818640 DOI: 10.1016/j.jjcc.2015.02.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 01/13/2015] [Accepted: 02/05/2015] [Indexed: 01/14/2023]
Abstract
BACKGROUND Atherosclerosis features a deterioration of the endothelial layer in all stages. Restoration of the endothelium is associated with circulating stem cell antigen 1 (sca1) and vascular endothelial growth factor receptor type 2 (flk-1) positive endothelial progenitor cells (EPCs). We investigated whether EPC production and/or a mobilization from bone marrow are reduced in severe atherosclerosis. METHODS AND RESULTS EPCs in peripheral blood were diminished in ApoE-/- mice with high-fat diet (HFD) whereas bone marrow levels of these cells were not significantly altered compared to controls. In situ perfusion of the hind limbs demonstrated that EPC mobilization was reduced compared to ApoE-/- mice with normal chow, although increased plasma stromal cell-derived factor (SDF) 1α and responsivity suggested a mobilizing stimulus. The proliferation of sca1/flk-1 positive cells showed no functional impairment. EPCs could not only be significantly mobilized from the bone marrow through the application of granulocyte colony stimulating factor (GCSF), but also led by trend to a depletion of the bone marrow pool. GCSF levels in plasma were equal in ApoE-/- mice with normal chow or HFD, which excluded a decline in GCSF production. CONCLUSION The capability of the bone marrow pool to adapt the proliferation and mobilization of sca1/flk-1 positive EPCs seems overstrained in ApoE-/- mice with a HFD.
Collapse
Affiliation(s)
- Martin Steinmetz
- Medizinische Klinik und Poliklinik II, Kardiologie/Angiologie/Pulmologie/Internistische Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany; Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Paris, France.
| | - Eva Lucanus
- Medizinische Klinik und Poliklinik II, Kardiologie/Angiologie/Pulmologie/Internistische Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Sebastian Zimmer
- Medizinische Klinik und Poliklinik II, Kardiologie/Angiologie/Pulmologie/Internistische Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Georg Nickenig
- Medizinische Klinik und Poliklinik II, Kardiologie/Angiologie/Pulmologie/Internistische Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Nikos Werner
- Medizinische Klinik und Poliklinik II, Kardiologie/Angiologie/Pulmologie/Internistische Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany
| |
Collapse
|
33
|
Zhang Z, Dong J, Lobe CG, Gong P, Liu J, Liao L. CCR5 facilitates endothelial progenitor cell recruitment and promotes the stabilization of atherosclerotic plaques in ApoE-/- mice. Stem Cell Res Ther 2015; 6:36. [PMID: 25889019 PMCID: PMC4404610 DOI: 10.1186/s13287-015-0026-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 02/27/2015] [Accepted: 02/27/2015] [Indexed: 12/04/2022] Open
Abstract
Introduction Unstable atherosclerotic plaques are prone to rupture, which leads to atherothrombosis. Endothelial progenitor cells (EPCs) are bone marrow-derived precursor cells that may repair vascular injury in atherosclerosis. Chemokine (C-C motif) receptor 5 (CCR5) promotes mobilization of EPCs. In this study, we investigated the therapeutic potential of CCR5-overexpressing EPCs on plaque stabilization in an apolipoprotein E (ApoE)−/− mouse model. Methods The expression of CCR5 and its cognate ligand chemokine (C-C motif) ligand 5 (CCL5) was examined in atherosclerotic aortas of humans and mice by immunohistochemistry. Splenectomized ApoE−/− C57BL/6 J mice fed a high-fat diet for 24 weeks were intravenously injected with EPCs transfected with CCR5 overexpression lentivirus. The recruitment of EPCs over the atherosclerotic plaques was evaluated by immunofluorescence. The content of lipid, smooth muscle cells, monocytes/macrophages, and endothelial cells in atherosclerotic plaques was assayed by specific immunostaining. The serum levels of atherosclerosis-related inflammatory factors in ApoE−/− mice were measured by mouse atherosclerosis antibody array I. Results CCR5 and CCL5 are highly expressed in atherosclerotic plaques in both humans and mice. The ApoE−/− mice with CCR5-overexpressing EPC treatment demonstrated a more stable plaque formation with enhanced recruitment of EPC, reduced lipid, and macrophage content in the atherosclerotic plaques. CCR5-overexpressing EPC treatment also increased the content of endothelial cells and nitric oxide production in the plaques. In addition, the serum levels of interleukin-3 (IL-3), IL-5, IL-6, IL-13, CD40, and tumor necrosis factor-alpha and the plaque contents of IL-6 and matrix metalloproteinase-9 were reduced in mice with CCR5-overexpressing EPC treatment. Conclusions These findings suggest that CCR5 is a novel therapeutic target in EPC treatment for stabilization of atherosclerotic plaques. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0026-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhongwen Zhang
- Department of Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, 16766 Jingshi Road, Jinan, Shandong, 250014, China.
| | - Jianjun Dong
- Department of Medicine, Qilu Hospital of Shandong University, Wenhua Road, Jinan, 250012, China.
| | - Corrinne G Lobe
- Miami Mice Research Corp., 101 College Street, Toronto, Ontario, M5G 1 L7, Canada.
| | - Peiyun Gong
- Department of Medicine, Qilu Hospital of Shandong University, Wenhua Road, Jinan, 250012, China.
| | - Ju Liu
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, 16766 Jingshi Road, Jinan, Shandong, 250014, China.
| | - Lin Liao
- Department of Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, 16766 Jingshi Road, Jinan, Shandong, 250014, China.
| |
Collapse
|
34
|
Minami Y, Nakajima T, Ikutomi M, Morita T, Komuro I, Sata M, Sahara M. Angiogenic potential of early and late outgrowth endothelial progenitor cells is dependent on the time of emergence. Int J Cardiol 2015; 186:305-14. [PMID: 25838182 DOI: 10.1016/j.ijcard.2015.03.166] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 02/18/2015] [Accepted: 03/16/2015] [Indexed: 12/25/2022]
Abstract
BACKGROUND Recent studies have suggested that late-outgrowth endothelial progenitor cells (EPCs) derived from human peripheral blood mononuclear cells (hPBMNCs) might have higher angiogenic potential than classically-defined early-outgrowth EPCs (EOCs). However, it still remains unclear which of "so-called" EPC subpopulations defined in a variety of ways has the highest angiogenic potential. METHODS AND RESULTS We classified hPBMNC-derived EPC subpopulations by the time of their emergence in culture. EOCs were defined as attached cells on culture days 3-7. Late-outgrowth EPCs, defined as the cell forming colonies with cobblestone appearance since day 10, were further classified as follows: "moderate"-outgrowth EPCs (MOCs) emerging on days 10-16, "late"-outgrowth EPCs (LOCs) on days 17-23, and "very late"-outgrowth EPCs (VOCs) on days 24-30. Flow cytometry analyses showed the clear differences of hematopoietic/endothelial markers between EOC (CD31(+)VE-cadherin(-)CD34(-)CD14(+)CD45(+)) and LOC (CD31(+)VE-cadherin(+)CD34(+)CD14(-)CD45(-)). We found that LOCs had the highest proliferation and tube formation capabilities in vitro along with the highest expression of angiogenic genes including KDR and eNOS. To investigate the in vivo therapeutic efficacies, each EPC subpopulation was intravenously transplanted into immunocompromised mice (total 4 × 10(5) cells) after unilateral hindlimb ischemia surgery. The LOC-treated mice exhibited significantly-enhanced blood flow recovery (flow ratios of ischemic/non-ischemic leg: 0.99±0.02 [LOC group] versus 0.67 ± 0.07 to 0.78 ± 0.09 [other groups]; P < 0.05) and augmented capillary collateral formation in ischemic leg, which were attributable to their direct engraftment into host angiogenic vessels (approximately 10%) and paracrine effects. CONCLUSION hPBMNC-derived late-outgrowth EPCs emerging on culture days 17-23 are superior to other EPC subpopulations with regard to therapeutic angiogenic potential.
Collapse
Affiliation(s)
- Yoshiyasu Minami
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; Department of Ischemic Circulatory Physiology, 22nd Century Medical and Research Center, University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Toshiaki Nakajima
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; Department of Ischemic Circulatory Physiology, 22nd Century Medical and Research Center, University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Masayasu Ikutomi
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; Department of Cardiovascular Medicine, Teikyo University Chiba Medical Center, 3426-3 Anegasaki, Ichihara 299-0111, Japan
| | - Toshihiro Morita
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; Department of Ischemic Circulatory Physiology, 22nd Century Medical and Research Center, University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Masataka Sata
- Department of Cardiovascular Medicine, Institute of Health Biosciences, The University of Tokushima Graduate School of Medicine, 2-10-1 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Makoto Sahara
- Department of Cardiovascular Medicine, University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; Department of Medicine-Cardiology/Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
35
|
Santulli G. microRNAs Distinctively Regulate Vascular Smooth Muscle and Endothelial Cells: Functional Implications in Angiogenesis, Atherosclerosis, and In-Stent Restenosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 887:53-77. [PMID: 26662986 PMCID: PMC4871245 DOI: 10.1007/978-3-319-22380-3_4] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Endothelial cells (EC) and vascular smooth muscle cells (VSMC) are the main cell types within the vasculature. We describe here how microRNAs (miRs)--noncoding RNAs that can regulate gene expression via translational repression and/or post-transcriptional degradation--distinctively modulate EC and VSMC function in physiology and disease. In particular, the specific roles of miR-126 and miR-143/145, master regulators of EC and VSMC function, respectively, are deeply explored. We also describe the mechanistic role of miRs in the regulation of the pathophysiology of key cardiovascular processes including angiogenesis, atherosclerosis, and in-stent restenosis post-angioplasty. Drawbacks of currently available therapeutic options are discussed, pointing at the challenges and potential clinical opportunities provided by miR-based treatments.
Collapse
MESH Headings
- Angioplasty
- Animals
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Endothelial Cells/cytology
- Endothelial Cells/metabolism
- Gene Expression Regulation
- Graft Occlusion, Vascular/genetics
- Graft Occlusion, Vascular/metabolism
- Humans
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Physiologic
- RNA, Untranslated/genetics
- RNA, Untranslated/metabolism
- Stents/adverse effects
- Vascular Remodeling
Collapse
|
36
|
Van Linthout S, Frias M, Singh N, De Geest B. Therapeutic potential of HDL in cardioprotection and tissue repair. Handb Exp Pharmacol 2015; 224:527-565. [PMID: 25523001 DOI: 10.1007/978-3-319-09665-0_17] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Epidemiological studies support a strong association between high-density lipoprotein (HDL) cholesterol levels and heart failure incidence. Experimental evidence from different angles supports the view that low HDL is unlikely an innocent bystander in the development of heart failure. HDL exerts direct cardioprotective effects, which are mediated via its interactions with the myocardium and more specifically with cardiomyocytes. HDL may improve cardiac function in several ways. Firstly, HDL may protect the heart against ischaemia/reperfusion injury resulting in a reduction of infarct size and thus in myocardial salvage. Secondly, HDL can improve cardiac function in the absence of ischaemic heart disease as illustrated by beneficial effects conferred by these lipoproteins in diabetic cardiomyopathy. Thirdly, HDL may improve cardiac function by reducing infarct expansion and by attenuating ventricular remodelling post-myocardial infarction. These different mechanisms are substantiated by in vitro, ex vivo, and in vivo intervention studies that applied treatment with native HDL, treatment with reconstituted HDL, or human apo A-I gene transfer. The effect of human apo A-I gene transfer on infarct expansion and ventricular remodelling post-myocardial infarction illustrates the beneficial effects of HDL on tissue repair. The role of HDL in tissue repair is further underpinned by the potent effects of these lipoproteins on endothelial progenitor cell number, function, and incorporation, which may in particular be relevant under conditions of high endothelial cell turnover. Furthermore, topical HDL therapy enhances cutaneous wound healing in different models. In conclusion, the development of HDL-targeted interventions in these strategically chosen therapeutic areas is supported by a strong clinical rationale and significant preclinical data.
Collapse
Affiliation(s)
- Sophie Van Linthout
- Charité-University-Medicine Berlin, Campus Virchow, Berlin-Brandenburg Center for Regenerative Therapy (BCRT), Berlin, Germany
| | | | | | | |
Collapse
|
37
|
Michelis KC, Boehm M, Kovacic JC. New vessel formation in the context of cardiomyocyte regeneration--the role and importance of an adequate perfusing vasculature. Stem Cell Res 2014; 13:666-82. [PMID: 24841067 PMCID: PMC4213356 DOI: 10.1016/j.scr.2014.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 03/16/2014] [Accepted: 04/18/2014] [Indexed: 02/08/2023] Open
Abstract
The history of revascularization for cardiac ischemia dates back to the early 1960's when the first coronary artery bypass graft procedures were performed in humans. With this 50 year history of providing a new vasculature to ischemic and hibernating myocardium, a profound depth of experience has been amassed in clinical cardiovascular medicine as to what does, and does not work in the context of cardiac revascularization, alleviating ischemia and adequacy of myocardial perfusion. These issues are of central relevance to contemporary cell-based cardiac regenerative approaches. While the cardiovascular cell therapy field is surging forward on many exciting fronts, several well accepted clinical axioms related to the cardiac arterial supply appear to be almost overlooked by some of our current basic conceptual and experimental cell therapy paradigms. We present here information drawn from five decades of the clinical revascularization experience, review relevant new data on vascular formation via cell therapy, and put forward the case that for optimal cell-based cardiac regeneration due attention must be paid to providing an adequate vascular supply.
Collapse
Affiliation(s)
- Katherine C Michelis
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manfred Boehm
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
38
|
In vivo bio-distribution and homing of endothelial outgrowth cells in a tumour model. Nucl Med Biol 2014; 41:848-55. [DOI: 10.1016/j.nucmedbio.2014.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 05/28/2014] [Accepted: 07/14/2014] [Indexed: 01/16/2023]
|
39
|
Yuldasheva NY, Rashid ST, Haywood NJ, Cordell P, Mughal R, Viswambharan H, Imrie H, Sukumar P, Cubbon RM, Aziz A, Gage M, Mbonye KA, Smith J, Galloway S, Skromna A, Scott DJA, Kearney MT, Wheatcroft SB. Haploinsufficiency of the Insulin-Like Growth Factor-1 Receptor Enhances Endothelial Repair and Favorably Modifies Angiogenic Progenitor Cell Phenotype. Arterioscler Thromb Vasc Biol 2014; 34:2051-8. [DOI: 10.1161/atvbaha.114.304121] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objectives—
Defective endothelial regeneration predisposes to adverse arterial remodeling and is thought to contribute to cardiovascular disease in type 2 diabetes mellitus. We recently demonstrated that the type 1 insulin-like growth factor receptor (IGF1R) is a negative regulator of insulin sensitivity and nitric oxide bioavailability. In this report, we examined partial deletion of the IGF1R as a potential strategy to enhance endothelial repair.
Approach and Results—
We assessed endothelial regeneration after wire injury in mice and abundance and function of angiogenic progenitor cells in mice with haploinsufficiency of the IGF1R (IGF1R
+/−
). Endothelial regeneration after arterial injury was accelerated in IGF1R
+/−
mice. Although the yield of angiogenic progenitor cells was lower in IGF1R
+/−
mice, these angiogenic progenitor cells displayed enhanced adhesion, increased secretion of insulin-like growth factor-1, and enhanced angiogenic capacity. To examine the relevance of IGF1R manipulation to cell-based therapy, we transfused IGF1R
+/−
bone marrow–derived CD117
+
cells into wild-type mice. IGF1R
+/−
cells accelerated endothelial regeneration after arterial injury compared with wild-type cells and did not alter atherosclerotic lesion formation.
Conclusions—
Haploinsufficiency of the IGF1R is associated with accelerated endothelial regeneration in vivo and enhanced tube forming and adhesive potential of angiogenic progenitor cells in vitro. Partial deletion of IGF1R in transfused bone marrow–derived CD117
+
cells enhanced their capacity to promote endothelial regeneration without altering atherosclerosis. Our data suggest that manipulation of the IGF1R could be exploited as novel therapeutic approach to enhance repair of the arterial wall after injury.
Collapse
Affiliation(s)
- Nadira Y. Yuldasheva
- From the Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - Sheikh Tawqeer Rashid
- From the Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - Natalie J. Haywood
- From the Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - Paul Cordell
- From the Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - Romana Mughal
- From the Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - Hema Viswambharan
- From the Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - Helen Imrie
- From the Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - Piruthivi Sukumar
- From the Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - Richard M. Cubbon
- From the Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - Amir Aziz
- From the Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - Matthew Gage
- From the Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - Kamatamu Amanda Mbonye
- From the Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - Jessica Smith
- From the Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - Stacey Galloway
- From the Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - Anna Skromna
- From the Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - D. Julian A. Scott
- From the Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - Mark T. Kearney
- From the Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| | - Stephen B. Wheatcroft
- From the Division of Cardiovascular and Diabetes Research, Leeds Multidisciplinary Cardiovascular Research Centre, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
40
|
Tian XL, Li Y. Endothelial cell senescence and age-related vascular diseases. J Genet Genomics 2014; 41:485-95. [PMID: 25269674 DOI: 10.1016/j.jgg.2014.08.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Revised: 07/31/2014] [Accepted: 08/06/2014] [Indexed: 10/24/2022]
Abstract
Advanced age is an independent risk factor for ageing-related complex diseases, such as coronary artery disease, stroke, and hypertension, which are common but life threatening and related to the ageing-associated vascular dysfunction. On the other hand, patients with progeria syndromes suffer from serious atherosclerosis, suggesting that the impaired vascular functions may be critical to organismal ageing, or vice versa. However, it remains largely unknown how vascular cells, particularly endothelial cell, become senescent and how the senescence impairs the vascular functions and contributes to the age-related vascular diseases over time. Here, we review the recent progress on the characteristics of vascular ageing and endothelial cell senescence in vitro and in vivo, evaluate how genetic and environmental factors as well as autophagy and stem cell influence endothelial cell senescence and how the senescence contributes to the age-related vascular phenotypes, such as atherosclerosis and increased vascular stiffness, and explore the possibility whether we can delay the age-related vascular diseases through the control of vascular ageing.
Collapse
Affiliation(s)
- Xiao-Li Tian
- Department of Human Population Genetics and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine (IMM), Peking University, Beijing 100871, China.
| | - Yang Li
- Department of Human Population Genetics and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine (IMM), Peking University, Beijing 100871, China
| |
Collapse
|
41
|
Hernandez SL, Gong JH, Chen L, Wu IH, Sun JK, Keenan HA, King GL. Characterization of circulating and endothelial progenitor cells in patients with extreme-duration type 1 diabetes. Diabetes Care 2014; 37:2193-201. [PMID: 24780357 PMCID: PMC4113171 DOI: 10.2337/dc13-2547] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE We characterized and correlated endothelial progenitor cells (EPCs) and circulating progenitor cells (CPCs) with lack of vascular complications in the Joslin Medalist Study in patients with type 1 diabetes for 50 years or longer. RESEARCH DESIGN AND METHODS EPC and CPC levels were ascertained by flow cytometry and compared among Medalists (n = 172) with or without diabetic retinopathy (DR; n = 84 of 162), neuropathy (n = 94 of 165), diabetic nephropathy (DN; n = 18 of 172), cardiovascular disease (CVD; n = 63 of 168), age-matched controls (n = 83), type 2 diabetic patients (n = 36), and younger type 1 diabetic patients (n = 31). Mitogens, inflammatory cytokines, and oxidative markers were measured in blood or urine. Migration of cultured peripheral blood mononuclear cells (PBMCs) from Medalists and age-matched controls were compared. RESULTS Medalists' EPC and CPC levels equaled those of their nondiabetic age-matched controls, were 10% higher than those in younger type 1 diabetic patients, and were 20% higher than those in age-matched type 2 diabetic patients. CPC levels were 15% higher in Medalists without CVD and nephropathy than in those affected, whereas EPC levels were significantly higher in those without peripheral vascular disease (PVD) than those with PVD. Stromal-derived factor 1 (SDF-1) levels were higher in Medalists with CVD, DN, and DR than in those not affected and their controls. IGF-I levels were lower in Medalists and correlated inversely with CPC levels. Additionally, cultured PBMCs from Medalists migrated more than those from nondiabetic controls. CONCLUSIONS Normal levels of EPC and CPC in the Medalists, unlike other groups with diabetes, especially those without CVD, support the idea that endogenous factors exist to neutralize the adverse effects of metabolic abnormalities of diabetes on vascular tissues.
Collapse
Affiliation(s)
- Sonia L Hernandez
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Jennifer H Gong
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MADepartment of Internal Medicine, Harvard Medical School, Boston, MA
| | - Liming Chen
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MATianjin Medical University, Tianjin, China
| | - I-Hsien Wu
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Jennifer K Sun
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MABeetham Eye Institute, Joslin Diabetes Center, Harvard Medical School, Boston, MADepartment of Ophthalmology, Harvard Medical School, Boston, MA
| | - Hillary A Keenan
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MADepartment of Internal Medicine, Harvard Medical School, Boston, MA
| | - George L King
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| |
Collapse
|
42
|
Zhang N, Xie X, Chen H, Chen H, Yu H, Wang JA. Stem cell-based therapies for atherosclerosis: perspectives and ongoing controversies. Stem Cells Dev 2014; 23:1731-40. [PMID: 24702267 DOI: 10.1089/scd.2014.0078] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Atherosclerosis is a major contributor to life-threatening cardiovascular events, the leading cause of death worldwide. Since the mechanisms of atherosclerosis have not been fully understood, currently, there are no effective approaches to regressing atherosclerosis. Therefore, there is a dire need to explore the mechanisms and potential therapeutic strategies to prevent or reverse the progression of atherosclerosis. In recent years, stem cell-based therapies have held promises to various diseases, including atherosclerosis. Unfortunately, the efficacy of stem cell-based therapies for atherosclerosis as reported in the literature has been inconsistent or even conflicting. In this review, we summarize the current literature of stem cell-based therapies for atherosclerosis and discuss possible mechanisms and future directions of these potential therapies.
Collapse
Affiliation(s)
- Na Zhang
- 1 Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine , Hangzhou, China
| | | | | | | | | | | |
Collapse
|
43
|
Noels H, Zhou B, Tilstam PV, Theelen W, Li X, Pawig L, Schmitz C, Akhtar S, Simsekyilmaz S, Shagdarsuren E, Schober A, Adams RH, Bernhagen J, Liehn EA, Döring Y, Weber C. Deficiency of endothelial CXCR4 reduces reendothelialization and enhances neointimal hyperplasia after vascular injury in atherosclerosis-prone mice. Arterioscler Thromb Vasc Biol 2014; 34:1209-20. [PMID: 24723559 DOI: 10.1161/atvbaha.113.302878] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The Cxcl12/Cxcr4 chemokine ligand/receptor axis mediates the mobilization of smooth muscle cell progenitors, driving injury-induced neointimal hyperplasia. This study aimed to investigate the role of endothelial Cxcr4 in neointima formation. APPROACH AND RESULTS β-Galactosidase staining using bone marrow x kinase (Bmx)-CreER(T2) reporter mice and double immunofluorescence revealed an efficient and endothelial-specific deletion of Cxcr4 in Bmx-CreER(T2+) compared with Bmx-CreER(T2-) Cxcr4-floxed apolipoprotein E-deficient (Apoe(-/-)) mice (referred to as Cxcr4(EC-KO)ApoE(-/-) and Cxcr4(EC-WT) ApoE(-/-), respectively). Endothelial Cxcr4 deficiency significantly increased wire injury-induced neointima formation in carotid arteries from Cxcr4(EC-KO)ApoE(-/-) mice. The lesions displayed a higher number of macrophages, whereas the smooth muscle cell and collagen content were reduced. This was associated with a significant reduction in reendothelialization and endothelial cell proliferation in injured Cxcr4(EC-KO)ApoE(-/-) carotids compared with Cxcr4(EC-WT)ApoE(-/-) controls. Furthermore, stimulation of human aortic endothelial cells with chemokine (C-X-C motif) ligand 12 (CXCL12) significantly enhanced their wound-healing capacity in an in vitro scratch assay, an effect that could be reversed with the CXCR4 antagonist AMD3100. Also, flow cytometric analysis showed a reduced mobilization of Sca1(+)Flk1(+)Cd31(+) and of Lin(-)Sca1(+) progenitors in Cxcr4(EC-KO) ApoE(-/-) mice after vascular injury, although Cxcr4 surface expression was unaltered. No differences could be detected in plasma concentrations of Cxcl12, vascular endothelial growth factor, sphingosine 1-phosphate, or Flt3 (fms-related tyrosine kinase 3) ligand, all cytokines with an established role in progenitor cell mobilization. Nonetheless, double immunofluorescence revealed a significant reduction in local endothelial Cxcl12 staining in injured carotids from Cxcr4(EC-KO)ApoE(-/-) mice. CONCLUSIONS Endothelial Cxcr4 is crucial for efficient reendothelialization after vascular injury through endothelial wound healing and proliferation, and through the mobilization of Sca1(+)Flk1(+)Cd31(+) cells, often referred to as circulating endothelial progenitor cells.
Collapse
Affiliation(s)
- Heidi Noels
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.).
| | - Baixue Zhou
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Pathricia V Tilstam
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Wendy Theelen
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Xiaofeng Li
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Lukas Pawig
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Corinna Schmitz
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Shamima Akhtar
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Sakine Simsekyilmaz
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Erdenechimeg Shagdarsuren
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Andreas Schober
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Ralf H Adams
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Jürgen Bernhagen
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Elisa A Liehn
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Yvonne Döring
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Christian Weber
- From the Institute for Molecular Cardiovascular Research (H.N., B.Z., P.V.T., W.T., X.L., L.P., S.A., S.S., E.S., E.A.L.) and Institute of Biochemistry and Molecular Cell Biology (C.S., J.B.), University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (A.S., Y.D., C.W.) and August-Lenz-Stiftung, Institute for Cardiovascular Research (J.B.), Ludwig-Maximilians-University Munich, Munich, Germany; Max Planck Institute for Molecular Biomedicine, University of Münster, Münster, Germany (R.H.A.); Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands (C.W.); and German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung), partner site Munich Heart Alliance, Munich, Germany (C.W.).
| |
Collapse
|
44
|
Pelosi E, Castelli G, Testa U. Endothelial progenitors. Blood Cells Mol Dis 2014; 52:186-94. [DOI: 10.1016/j.bcmd.2013.11.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 11/13/2013] [Accepted: 11/13/2013] [Indexed: 12/31/2022]
|
45
|
Tahir H, Bona-Casas C, Narracott AJ, Iqbal J, Gunn J, Lawford P, Hoekstra AG. Endothelial repair process and its relevance to longitudinal neointimal tissue patterns: comparing histology with in silico modelling. J R Soc Interface 2014; 11:20140022. [PMID: 24621816 DOI: 10.1098/rsif.2014.0022] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Re-establishing a functional endothelium following endovascular treatment is an important factor in arresting neointimal proliferation. In this study, both histology (in vivo) and computational simulations (in silico) are used to evaluate neointimal growth patterns within coronary arteries along the axial direction of the stent. Comparison of the growth configurations in vivo and in silico was undertaken to identify candidate mechanisms for endothelial repair. Stent, lumen and neointimal areas were measured from histological sections obtained from eight right coronary stented porcine arteries. Two re-endothelialization scenarios (endothelial cell (EC) random seeding and EC growth from proximal and distal ends) were implemented in silico to evaluate their influence on the morphology of the simulated lesions. Subject to the assumptions made in the current simulations, comparison between in vivo and in silico results suggests that endothelial growth does not occur from the proximal and distal ends alone, but is more consistent with the assumption of a random seeding process. This may occur either from the patches of endothelium which survive following stent implantation or from attachment of circulating endothelial progenitor cells.
Collapse
Affiliation(s)
- Hannan Tahir
- Computational Science, Informatics Institute, University of Amsterdam, , Science Park 904, Amsterdam 1098 XH, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
46
|
Fadini GP. A reappraisal of the role of circulating (progenitor) cells in the pathobiology of diabetic complications. Diabetologia 2014; 57:4-15. [PMID: 24173366 DOI: 10.1007/s00125-013-3087-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 10/01/2013] [Indexed: 01/10/2023]
Abstract
Traditionally, the development of diabetic complications has been attributed to the biochemical pathways driving hyperglycaemic cell damage, while reparatory mechanisms have been long overlooked. A more comprehensive view of the balance between damage and repair suggests that an impaired regenerative capacity of bone marrow (BM)-derived cells strongly contributes to defective re-endothelisation and neoangiogenesis in diabetes. Although recent technological advances have redefined the biology and function of endothelial progenitor cells (EPCs), interest in BM-derived vasculotropic cells in the setting of diabetes and its complications remains high. Several circulating cell types of haematopoietic and non-haematopoietic origin are affected by diabetes and are potentially involved in the pathobiology of chronic complications. In addition to classical EPCs, these include circulating (pro-)angiogenic cells, polarised monocytes/macrophages (M1 and M2), myeloid calcifying cells and smooth muscle progenitor cells, having disparate roles in vascular biology. In parallel with the study of elusive progenitor cell phenotypes, it has been recognised that diabetes induces a profound remodelling of the BM stem cell niche. The alteration of circulating (progenitor) cells in the BM is now believed to be the link among distant end-organ complications. The field is rapidly evolving and interest is shifting from specific cell populations to the complex network of interactions that orchestrate trafficking of circulating vasculotropic cells.
Collapse
Affiliation(s)
- G P Fadini
- Department of Medicine, University Hospital of Padova, University of Padova, Via Giustiniani, 2, 35100, Padova, Italy,
| |
Collapse
|
47
|
Endothelial colony-forming cells for preparing prevascular three-dimensional cell-dense tissues using cell-sheet engineering. J Tissue Eng Regen Med 2013; 10:739-47. [DOI: 10.1002/term.1858] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 09/03/2013] [Accepted: 11/10/2013] [Indexed: 12/27/2022]
|
48
|
Kimura W, Sadek HA. The cardiac hypoxic niche: emerging role of hypoxic microenvironment in cardiac progenitors. Cardiovasc Diagn Ther 2013; 2:278-89. [PMID: 24282728 DOI: 10.3978/j.issn.2223-3652.2012.12.02] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Accepted: 12/10/2012] [Indexed: 12/11/2022]
Abstract
Resident stem cells persist throughout the entire lifetime of an organism where they replenishing damaged cells. Numerous types of resident stem cells are housed in a low-oxygen tension (hypoxic) microenvironment, or niches, which seem to be critical for survival and maintenance of stem cells. Recently our group has identified the adult mammalian epicardium and subepicardium as a hypoxic niche for cardiac progenitor cells. Similar to hematopoietic stem cells (LT-HSCs), progenitor cells in the hypoxic epicardial niche utilize cytoplasmic glycolysis instead of mitochondrial oxidative phosphorylation, where hypoxia inducible factor 1α (Hif-1α) maintains them in glycolytic undifferentiated state. In this review we summarize the relationship between hypoxic signaling and stem cell function, and discuss potential roles of several cardiac stem/progenitor cells in cardiac homeostasis and regeneration.
Collapse
Affiliation(s)
- Wataru Kimura
- Department of Internal Medicine, Division of Cardiology, UT Southwestern Medical Center, Dallas, TX, USA
| | | |
Collapse
|
49
|
Tousoulis D, Briasoulis A, Vogiatzi G, Valatsou A, Kourkouti P, Pantopoulou A, Papageorgiou N, Perrea D, Stefanadis C. Effects of direct infusion of bone marrow-derived progenitor cells and indirect mobilization of hematopoietic progenitor cells on atherosclerotic plaque and inflammatory process in atherosclerosis. Int J Cardiol 2013; 168:4769-4774. [PMID: 23958421 DOI: 10.1016/j.ijcard.2013.07.229] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 07/15/2013] [Accepted: 07/25/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND We sought to investigate the effects of lin-/sca+ cells, endothelial progenitor cells (EPCs) and granulocyte colony-stimulating factor (G-CSF) administration on atherosclerotic plaque progression. METHODS Apolipoprotein E-deficient (apoE(-/-)) mice were splenectomized and treated with high-cholesterol diet for 6 weeks in order to induce atherosclerotic plaque development. Bone marrow-derived Lin-/sca-1+ cells were isolated and further cultured to early growth endothelial progenitor cells (EPCs). Mice were divided in four groups (n=10/group) and received two intravenous injections of 5×10(5) cells (lin-/sca-1+ or EPCs), or granulocyte colony-stimulating factor (G-CSF 100 μg/kg/day) for 7 days or normal saline. The same interventions were administered to animals, which had undergone unilateral hind-limb ischemia. Effects on inflammatory parameters, lesion severity, and atherosclerotic plaque area size were assessed. RESULTS The administration of both G-CSF and progenitor cells significantly decreased the levels of IL-6, 6 weeks after the initiation of treatment. Atherosclerotic lesion area was reduced by G-CSF (atherosclerotic plaque area percentage 22.94%±3.68, p=0.001), by lin-/sca-1+ (23.27%±5.98, p=0.002) and cultured EPCs (23.16±4.86%, p=0.002) compared to control (32.75%±7.05). In the atherosclerotic mice that underwent limb ischemia, the atherosclerotic plaque area, was not significantly different between the treatment groups cultured EPCs-treated mice and the control group (p=NS, for all). CONCLUSIONS Direct infusion of progenitor cells and indirect mobilization of hematopoietic progenitor cells decreased plaque progression and levels of inflammatory molecules in a murine model of atherosclerosis. Treatment with G-CSF, lin-/sca-1+, or EPCs may exert beneficial effects on vascular inflammation and atherosclerotic plaque progression. However, the effects are diminished in an ischemic setting.
Collapse
|
50
|
Jansen F, Yang X, Hoelscher M, Cattelan A, Schmitz T, Proebsting S, Wenzel D, Vosen S, Franklin BS, Fleischmann BK, Nickenig G, Werner N. Endothelial microparticle-mediated transfer of MicroRNA-126 promotes vascular endothelial cell repair via SPRED1 and is abrogated in glucose-damaged endothelial microparticles. Circulation 2013; 128:2026-38. [PMID: 24014835 DOI: 10.1161/circulationaha.113.001720] [Citation(s) in RCA: 364] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Repair of the endothelium after vascular injury is crucial for preserving endothelial integrity and preventing the development of vascular disease. The underlying mechanisms of endothelial cell repair are largely unknown. We sought to investigate whether endothelial microparticles (EMPs), released from apoptotic endothelial cells (ECs), influence EC repair. METHODS AND RESULTS Systemic treatment of mice with EMPs after electric denudation of the endothelium accelerated reendothelialization in vivo. In vitro experiments revealed that EMP uptake in ECs promotes EC migration and proliferation, both critical steps in endothelial repair. To dissect the underlying mechanisms, Taqman microRNA array was performed, and microRNA (miR)-126 was identified as the predominantly expressed miR in EMPs. The following experiments demonstrated that miR-126 was transported into recipient human coronary artery endothelial cells by EMPs and functionally regulated the target protein sprouty-related, EVH1 domain-containing protein 1 (SPRED1). Knockdown of miR-126 in EMPs abrogated EMP-mediated effects on human coronary artery endothelial cell migration and proliferation in vitro and reendothelialization in vivo. Interestingly, after simulating diabetic conditions, EMPs derived from glucose-treated ECs contained significantly lower amounts of miR-126 and showed reduced endothelial repair capacity in vitro and in vivo. Finally, expression analysis of miR-126 in circulating microparticles from 176 patients with stable coronary artery disease with and without diabetes mellitus revealed a significantly reduced miR-126 expression in circulating microparticles from diabetic patients. CONCLUSIONS Endothelial microparticles promote vascular endothelial repair by delivering functional miR-126 into recipient cells. In pathological hyperglycemic conditions, EMP-mediated miR-126-induced EC repair is altered.
Collapse
Affiliation(s)
- Felix Jansen
- Department of Internal Medicine II, University Hospital Bonn, Rheinische Friedrich-Wilhelms University, Bonn, Germany (F.J., M.H., A.C., T.S., S.P., G.N., N.W.); Feinberg Cardiovascular Research Institute, Northwestern University School of Medicine, Chicago, IL (X.Y.); Institute of Physiology, University Hospital Bonn, Rheinische Friedrich-Wilhelms University, Bonn, Germany (D.W., S.V., B.K.F.); and Institute of Innate Immunity, Rheinische Friedrich-Wilhelms University, Bonn, Germany (B.S.F.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|