1
|
Kohli S, Kohli D, Gupta R. Stent strategies: Endothelial progenitor cell coated stents vs sirolimus eluting stents in a pairwise meta-analysis. Am J Surg 2025; 239:116055. [PMID: 39514989 DOI: 10.1016/j.amjsurg.2024.116055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/03/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) capturing stents were developed to enhance endothelial repair and reduce the risk of stent thrombosis, addressing limitations of Sirolimus-Eluting Stents (SES). This study aims to compare the safety and efficacy of EPC stents versus SES in patients undergoing percutaneous coronary intervention (PCI). METHODS We performed a meta-analysis following PRISMA guidelines in patients undergoing PCI treated with Sirolimus eluting stent (SES) vs the use of EPC stents and recognized 8 clinical trials with patients undergoing PCI and reporting outcomes such as Target Lesion Failure (TLF), stent thrombosis, and revascularisation. Relative risks were calculated using a random effects model and heterogeneity was assessed with I^2 statistics. RESULTS The EPC group showed higher incidence of TLF (RR = 1.28), MI(RR = 1.10), and cardiac death (RR = 1.19) compared to SES, though these differences were not statistically significant. Revascularisation rates were significantly higher in EPC group with TVR (RR = 1.60) and TLR(RR = 2.20) while stent thrombosis was lower (RR = 0.93). CONCLUSION The results of this EPC study reveals that while EPC stents show promise in revascularisation and lowering stent thrombosis, they are also associated with higher incidence of adverse events. The utility of EPC, especially vast reendothelialization, may have niche applications but their full potential can be realized with more rigorous trials as a clear advantage over SES remains lacking.
Collapse
Affiliation(s)
- Srishti Kohli
- Government Medical College, Amritsar, Majitha Road, Amritsar, Punjab, 143001, India.
| | - Daksh Kohli
- Government Medical College Patiala, Sangrur Road, opp. Rajindra Hospital, Patiala, Punjab, 147001, India.
| | - Raghav Gupta
- Government Medical College, Amritsar, Majitha Road, Amritsar, Punjab, 143001, India.
| |
Collapse
|
2
|
Shimizu Y, Kawashiri SY, Yamanashi H, Nakamichi S, Hayashida N, Nagata Y, Maeda T. Beneficial influence of low-density lipoprotein cholesterol on the endothelium in relation to endothelial repair. Environ Health Prev Med 2025; 30:24. [PMID: 40189259 PMCID: PMC11986262 DOI: 10.1265/ehpm.24-00332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/20/2025] [Indexed: 04/13/2025] Open
Abstract
BACKGROUND Low-density lipoprotein cholesterol (LDLc) is regarded as a risk factor for endothelial dysfunction. However, LDLc stimulates the proliferation of hematopoietic stem cells (CD34-positive cells), which contribute to endothelial repair. Therefore, LDLc may have a beneficial influence on the endothelium of individuals with lower endothelial repair activity. METHODS This cross-sectional study included 245 men aged 60-69 years. Endothelial repair activity was categorized by the circulating levels of CD34-positive cells based on median values. The status of endothelium was evaluated using the cardio-ankle vascular index (CAVI). RESULTS Among individuals with low levels of circulating CD34-positive cells, LDL-c levels were significantly inversely correlated with CAVI and positively correlated with circulating CD34-positive cells. No significant correlations were observed among the participants with high levels of circulating CD34-positive cells. Among low levels of CD34-positive cells, the adjusted standardized parameter (β) and p value were -0.24 (p = 0.021) for CAVI and 0.41 (p < 0.001) for CD34-positive cells, whereas among high levels of CD34-positive cells, the corresponding values were 0.03 (p = 0.738) and -0.09 (p = 0.355). CONCLUSION LDLc has a beneficial influence on endothelial health among individuals with low endothelial repair activity, possibly by stimulating the proliferation of hematopoietic stem cells.
Collapse
Affiliation(s)
- Yuji Shimizu
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Epidemiology Section, Division of Public Health, Osaka Institute of Public Health, Osaka, Japan
| | - Shin-Ya Kawashiri
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hirotomo Yamanashi
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Seiko Nakamichi
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Naomi Hayashida
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Division of Strategic Collaborative Research, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Yasuhiro Nagata
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takahiro Maeda
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Islands and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
3
|
Hershenson R, Nardi-Agmon I, Leshem-Lev D, Kornowski R, Eisen A. The effect of empagliflozin on circulating endothelial progenitor cells in patients with diabetes and stable coronary artery disease. Cardiovasc Diabetol 2024; 23:386. [PMID: 39468546 PMCID: PMC11520434 DOI: 10.1186/s12933-024-02466-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Diabetes mellitus (DM) is associated with premature atherosclerotic disease, coronary artery disease (CAD) and chronic heart failure (HF), leading to increased morbidity and mortality. Sodium-Glucose Co-transporter 2 Inhibitors (SGLT2i) exhibit cardioprotective benefits beyond glucose lowering, reducing the risk of major cardiovascular events (MACE) and HF hospitalizations in patients with DM and CAD. Endothelial progenitor cells (EPCs) are bone marrow-derived cells involved in vascular repair, mobilized in response to vascular injury. The number and function of circulating EPCs (cEPCs) are negatively affected by cardiovascular risk factors, including DM. This study aimed to examine the response of cEPCs to SGLT2i treatment in DM patients with stable CAD. METHODS A prospective single-center study included patients with DM and stable CAD who were started on an SGLT2i (empagliflozin). Peripheral blood samples were collected at baseline, 1 month, and 3 months to evaluate cEPC levels and function by flow cytometry, immunohistochemistry and MTT assays. RESULTS Eighteen patients were included in the study (median age 73, (IQR 69, 77) years, 67% male). After 1 month of treatment with empagliflozin, there was no significant change in cEPCs level or function. However, following 3 months of treatment, a significant increase was observed both in cell levels (CD34(+)/VEGFR-2(+): from 0.49% (IQR 0.32, 0.64) to 1.58% (IQR 0.93, 1.82), p = 0.0006; CD133(+)/VEGFR-2(+): from 0.38% (IQR 0.27, 0.6) to 0.82% (IQR 0.7, 1.95), p = 0.0001) and in cell function (from 0.25 CFUs (IQR 0, 0.5) at baseline, to 2 CFUs (IQR 1, 2) at 3 months, p = 0.0012). CONCLUSIONS Empagliflozin treatment in patients with DM and stable CAD increases cEPC levels and function, implying a cardioprotective mechanism. These findings highlight the potential of SGLT2i in treating cardiovascular diseases, warranting further research to explore these effects and their long-term implications.
Collapse
Affiliation(s)
- Roy Hershenson
- Department of Cardiology, Rabin Medical Center, 39 Jabotinsky St., 49100, Petah Tikva, Israel.
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Inbar Nardi-Agmon
- Department of Cardiology, Rabin Medical Center, 39 Jabotinsky St., 49100, Petah Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
| | - Dorit Leshem-Lev
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
| | - Ran Kornowski
- Department of Cardiology, Rabin Medical Center, 39 Jabotinsky St., 49100, Petah Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alon Eisen
- Department of Cardiology, Rabin Medical Center, 39 Jabotinsky St., 49100, Petah Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
4
|
Santillán-Cortez D, Vera-Gómez E, Hernández-Patricio A, Ruíz-Hernández AS, Gutiérrez-Buendía JA, De la Vega-Moreno K, Rizo-García YA, Loman-Zuñiga OA, Escotto-Sánchez I, Rodríguez-Trejo JM, Téllez-González MA, Toledo-Lozano CG, Ortega-Rosas T, García S, Mondragón-Terán P, Suárez-Cuenca JA. Endothelial Progenitor Cells May Be Related to Major Amputation after Angioplasty in Patients with Critical Limb Ischemia. Cells 2023; 12:cells12040584. [PMID: 36831250 PMCID: PMC9954311 DOI: 10.3390/cells12040584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/28/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Critical limb ischemia represents an advanced stage of peripheral arterial disease. Angioplasty improves blood flow to the limb; however, some patients progress irreversibly to lower limb amputation. Few studies have explored the predictive potential of biomarkers during postangioplasty outcomes. AIM To evaluate the behavior of endothelial progenitor cells in patients with critical limb ischemia, in relation to their postangioplasty outcome. METHODS Twenty patients with critical limb ischemia, candidates for angioplasty, were enrolled. Flow-mediated dilation, as well as endothelial progenitor cells (subpopulations CD45+/CD34+/CD133+/CD184+ and CD45+/CD/34+/KDR[VEGFR-2]+ estimated by flow cytometry) from blood flow close to vascular damage, were evaluated before and after angioplasty. Association with lower limb amputation during a 30-day follow-up was analyzed. RESULTS Endothelial progenitor cells were related with flow-mediated dilation. A higher number of baseline EPCs CD45+CD34+KDR+, as well as an impaired reactivity of endothelial progenitor cells CD45+CD34+CD133+CD184+ after angioplasty, were observed in cases further undergoing major limb amputation, with a significant discrimination ability and risk (0.75, specificity 0.83 and RR 4.5 p < 0.05). CONCLUSIONS Endothelial progenitor cells were related with endothelial dysfunction, whereas a higher baseline number of the subpopulation CD45+CD34+KDR+, as well as an impaired reactivity of subpopulation CD45+CD34+CD133+CD184+ after angioplasty, showed a predictive ability for major limb amputation in patients with critical limb ischemia.
Collapse
Affiliation(s)
- Daniel Santillán-Cortez
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
- Vascular Surgery and Angiology Department, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
- Regenerative Medicine and Tissue Engineering Laboratory, Coordination of Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Eduardo Vera-Gómez
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Alejandro Hernández-Patricio
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Atzín Suá Ruíz-Hernández
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Juan Ariel Gutiérrez-Buendía
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Karen De la Vega-Moreno
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Yasser Alberto Rizo-García
- Vascular Surgery and Angiology Department, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Oscar Antonio Loman-Zuñiga
- Vascular Surgery and Angiology Department, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Ignacio Escotto-Sánchez
- Vascular Surgery and Angiology Department, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Juan Miguel Rodríguez-Trejo
- Vascular Surgery and Angiology Department, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Mario Antonio Téllez-González
- Regenerative Medicine and Tissue Engineering Laboratory, Coordination of Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Christian Gabriel Toledo-Lozano
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Tania Ortega-Rosas
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Silvia García
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Paul Mondragón-Terán
- Regenerative Medicine and Tissue Engineering Laboratory, Coordination of Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
| | - Juan Antonio Suárez-Cuenca
- Experimental Metabolism and Clinical Research Laboratory, Clinical Research Department, Division of Biomedical Research, Centro Médico Nacional “20 de Noviembre”, Instituto de Seguridad y Servicios Sociales para los Trabajadores del Estado, Mexico City P.O. 03100, Mexico
- Correspondence: ; Tel.: +5255-52005003 (ext. 14661)
| |
Collapse
|
5
|
Pelliccia F, Zimarino M, De Luca G, Viceconte N, Tanzilli G, De Caterina R. Endothelial Progenitor Cells in Coronary Artery Disease: From Bench to Bedside. Stem Cells Transl Med 2022; 11:451-460. [PMID: 35365823 PMCID: PMC9154346 DOI: 10.1093/stcltm/szac010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 02/04/2022] [Indexed: 11/14/2022] Open
Abstract
Endothelial progenitor cells (EPCs) are a heterogeneous group of cells present in peripheral blood at various stages of endothelial differentiation. EPCs have been extensively investigated in patients with coronary artery disease (CAD), with controversial findings both on their role in atherosclerosis progression and in the process of neointimal growth after a percutaneous coronary intervention (PCI). Despite nearly 2 decades of experimental and clinical investigations, however, the significance of EPCs in clinical practice remains unclear and poorly understood. This review provides an update on the role of EPCs in the most common clinical scenarios that are experienced by cardiologists managing patients with CAD. We here summarize the main findings on the association of EPCs with cardiovascular risk factors, coronary atherosclerosis, and myocardial ischemia. We then discuss the potential effects of EPCs in post-PCI in-stent restenosis, as well as most recent findings with EPC-coated stents. Based on the mounting evidence of the relationship between levels of EPCs and several different adverse cardiovascular events, EPCs are emerging as novel predictive biomarkers of long-term outcomes in patients with CAD.
Collapse
Affiliation(s)
| | - Marco Zimarino
- Institute of Cardiology, “G. d’Annunzio” University, Chieti, Italy
- Cath Lab, SS. Annunziata Hospital, Chieti, Italy
| | - Giuseppe De Luca
- Division of Cardiology, Azienda Ospedaliero-Universitaria Maggiore della Carità, Università del Piemonte Orientale, Novara, Italy
| | - Nicola Viceconte
- Department of Cardiovascular Sciences, Sapienza University, Rome, Italy
| | - Gaetano Tanzilli
- Department of Cardiovascular Sciences, Sapienza University, Rome, Italy
| | | |
Collapse
|
6
|
The impact of different forms of exercise on endothelial progenitor cells in healthy populations. Eur J Appl Physiol 2022; 122:1589-1625. [PMID: 35305142 PMCID: PMC9197818 DOI: 10.1007/s00421-022-04921-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 02/18/2022] [Indexed: 11/03/2022]
Abstract
Circulating endothelial progenitor cells (EPCs) contribute to vascular healing and neovascularisation, while exercise is an effective means to mobilise EPCs into the circulation. OBJECTIVES to systematically examine the acute and chronic effects of different forms of exercise on circulating EPCs in healthy populations. METHODS Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines were followed. RESULTS thirty-one articles met the inclusion criteria including 747 participants aged 19 to 76 years. All included trials used flow cytometry for identification of circulating EPCs. Eight and five different EPC phenotypes were identified in the acute and chronic trials, respectively. In the acute trials, moderate intensity continuous (MICON), maximal, prolonged endurance, resistance and high intensity interval training (HIIT) exercise protocols were utilised. Prolonged endurance and resistance exercise had the most profound effect on circulating EPCs followed by maximal exercise. In the chronic trials, MICON exercise, HIIT, HIIT compared to MICON and MICON compared to exergame (exercise modality based on an interactive video game) were identified. MICON exercise had a positive effect on circulating EPCs in older sedentary individuals which was accompanied by improvements in endothelial function and arterial stiffness. Long-stage HIIT (4 min bouts) appears to be an effective means and superior than MICON exercise in mobilising circulating EPCs. In conclusion, both in acute and chronic trials the degree of exercise-induced EPC mobilisation depends upon the exercise regime applied. In future, more research is warranted to examine the dose-response relationship of different exercise forms on circulating EPCs using standardised methodology and EPC phenotype.
Collapse
|
7
|
Pelliccia F, Pasceri V, Zimarino M, De Luca G, De Caterina R, Mehran R, Dangas G. Endothelial progenitor cells in coronary atherosclerosis and percutaneous coronary intervention: A systematic review and meta-analysis. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2022; 42:94-99. [DOI: 10.1016/j.carrev.2022.02.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 12/19/2022]
|
8
|
Reno TA, Tarnus L, Tracy R, Landay AL, Sereti I, Apetrei C, Pandrea I. The Youngbloods. Get Together. Hypercoagulation, Complement, and NET Formation in HIV/SIV Pathogenesis. FRONTIERS IN VIROLOGY 2022. [DOI: 10.3389/fviro.2021.795373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chronic, systemic T-cell immune activation and inflammation (IA/INFL) have been reported to be associated with disease progression in persons with HIV (PWH) since the inception of the AIDS pandemic. IA/INFL persist in PWH on antiretroviral therapy (ART), despite complete viral suppression and increases their susceptibility to serious non-AIDS events (SNAEs). Increased IA/INFL also occur during pathogenic SIV infections of macaques, while natural hosts of SIVs that control chronic IA/INFL do not progress to AIDS, despite having persistent high viral replication and severe acute CD4+ T-cell loss. Moreover, natural hosts of SIVs do not present with SNAEs. Multiple mechanisms drive HIV-associated IA/INFL, including the virus itself, persistent gut dysfunction, coinfections (CMV, HCV, HBV), proinflammatory lipids, ART toxicity, comorbidities, and behavioral factors (diet, smoking, and alcohol). Other mechanisms could also significantly contribute to IA/INFL during HIV/SIV infection, notably, a hypercoagulable state, characterized by elevated coagulation biomarkers, including D-dimer and tissue factor, which can accurately identify patients at risk for thromboembolic events and death. Coagulation biomarkers strongly correlate with INFL and predict the risk of SNAE-induced end-organ damage. Meanwhile, the complement system is also involved in the pathogenesis of HIV comorbidities. Despite prolonged viral suppression, PWH on ART have high plasma levels of C3a. HIV/SIV infections also trigger neutrophil extracellular traps (NETs) formation that contribute to the elimination of viral particles and infected CD4+ T-cells. However, as SIV infection progresses, generation of NETs can become excessive, fueling IA/INFL, destruction of multiple immune cells subsets, and microthrombotic events, contributing to further tissue damages and SNAEs. Tackling residual IA/INFL has the potential to improve the clinical course of HIV infection. Therefore, therapeutics targeting new pathways that can fuel IA/INFL such as hypercoagulation, complement activation and excessive formation of NETs might be beneficial for PWH and should be considered and evaluated.
Collapse
|
9
|
Lee CH, Liu KS, Roth JG, Hung KC, Liu YW, Wang SH, Kuo CC, Liu SJ. Telmisartan Loaded Nanofibers Enhance Re-Endothelialization and Inhibit Neointimal Hyperplasia. Pharmaceutics 2021; 13:1756. [PMID: 34834171 PMCID: PMC8623288 DOI: 10.3390/pharmaceutics13111756] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/15/2021] [Accepted: 10/16/2021] [Indexed: 12/12/2022] Open
Abstract
Stent implantation impairs local endothelial function and may be associated with subsequent adverse cardiovascular events. Telmisartan, an angiotensin II receptor blocker that has unique peroxisome proliferator-activated-receptor-gamma-mediated effects on cardiovascular disease, has been shown to enhance endothelial function and limit neointimal hyperplasia. This study utilized hybrid biodegradable/stent nanofibers to facilitate sustained and local delivery of telmisartan to injured arterial vessels. Telmisartan and poly(d,l)-lactide-co-glycolide (PLGA) (75:25) were dissolved in hexafluoroisopropyl alcohol and electrospun into biodegradable nanofibrous tubes which were coated onto metal stents. By releasing 20% of the loaded telmisartan in 30 days, these hybrid biodegradable/stent telmisartan-loaded nanofibers increased the migration of endothelial progenitor cells in vitro, promoted endothelialization, and reduced intimal hyperplasia. As such, this work provides insights into the use of PLGA nanofibers for treating patients with an increased risk of stent restenosis.
Collapse
Affiliation(s)
- Chen-Hung Lee
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital-Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan; (K.-C.H.); (Y.-W.L.); (S.-H.W.)
| | - Kuo-Sheng Liu
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan;
| | - Julien George Roth
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Kuo-Chun Hung
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital-Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan; (K.-C.H.); (Y.-W.L.); (S.-H.W.)
| | - Yen-Wei Liu
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital-Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan; (K.-C.H.); (Y.-W.L.); (S.-H.W.)
| | - Shin-Huei Wang
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital-Linkou, Chang Gung University College of Medicine, Taoyuan 33305, Taiwan; (K.-C.H.); (Y.-W.L.); (S.-H.W.)
| | - Chi-Ching Kuo
- Research and Development Center of Smart Textile Technology, Institute of Organic and Polymeric Materials, National Taipei University of Technology, Taipei 10608, Taiwan
| | - Shih-Jung Liu
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan
| |
Collapse
|
10
|
Shimizu Y, Kawashiri SY, Nobusue K, Yamanashi H, Nagata Y, Maeda T. Associations between handgrip strength and hypertension in relation to circulating CD34-positive cell levels among Japanese older men: a cross-sectional study. Environ Health Prev Med 2021; 26:62. [PMID: 34088260 PMCID: PMC8178925 DOI: 10.1186/s12199-021-00982-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/14/2021] [Indexed: 01/09/2023] Open
Abstract
Background A positive association between handgrip strength and blood pressure has been reported. Since these factors are linked to the condition of the endothelium, the activity of endothelial repair might influence the association between handgrip strength and hypertension. Methods A cross-sectional study was conducted with 257 Japanese men aged 60–69 years who underwent an annual health checkup. As individuals with high level of circulating CD34-positive cells might show active endothelial repair, which plays an important role in vascular homeostasis, participants were stratified by circulating CD34-positive cell levels, using the median value of this population (0.96 cells/μL) as the cutoff. Results Independent of known cardiovascular risk factors, for participants with a high CD34-positive cell, handgrip strength is significantly positively associated with hypertension (odds ratio and 95% confidence interval of hypertension for 1 standard deviation increment of handgrip strength were 1.85 (1.19, 2.88) but not for participants with a low CD34-positive cell (0.91 (0.61, 1.37)). Conclusion The positive association between handgrip strength and hypertension is limited to high CD34-positive cells. This result may help clarify the role of vascular homeostasis in maintaining muscle strength.
Collapse
Affiliation(s)
- Yuji Shimizu
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Sakamoto 1-12-4, Nagasaki, 852-8523, Japan. .,Department of Cardiovascular Disease Prevention, Osaka Center for Cancer and Cardiovascular Diseases Prevention, Osaka, Japan.
| | - Shin-Ya Kawashiri
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Sakamoto 1-12-4, Nagasaki, 852-8523, Japan
| | - Kenichi Nobusue
- Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hirotomo Yamanashi
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yasuhiro Nagata
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Sakamoto 1-12-4, Nagasaki, 852-8523, Japan
| | - Takahiro Maeda
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki-shi, Sakamoto 1-12-4, Nagasaki, 852-8523, Japan.,Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
11
|
Birer M, Acartürk F. Telmisartan loaded polycaprolactone/gelatin-based electrospun vascular scaffolds. INT J POLYM MATER PO 2021. [DOI: 10.1080/00914037.2021.1915785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Mehmet Birer
- Department of Pharmaceutical Technology, Gazi University Faculty of Pharmacy, Ankara, Turkey
| | - Füsun Acartürk
- Department of Pharmaceutical Technology, Gazi University Faculty of Pharmacy, Ankara, Turkey
| |
Collapse
|
12
|
Kettelhut A, Bowman E, Funderburg NT. Immunomodulatory and Anti-Inflammatory Strategies to Reduce Comorbidity Risk in People with HIV. Curr HIV/AIDS Rep 2020; 17:394-404. [PMID: 32535769 DOI: 10.1007/s11904-020-00509-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW In this review, we will discuss treatment interventions targeting drivers of immune activation and chronic inflammation in PWH. RECENT FINDINGS Potential treatment strategies to prevent the progression of comorbidities in PWH have been identified. These studies include, among others, the use of statins to modulate lipid alterations and subsequent innate immune receptor activation, probiotics to restore healthy gut microbiota and reduce microbial translocation, hydroxychloroquine to reduce immune activation by altering Toll-like receptors function and expression, and canakinumab to block the action of a major pro-inflammatory cytokine IL-1β. Although many of the treatment strategies discussed here show promise, due to the complex nature of chronic inflammation and comorbidities in PWH, larger clinical studies are needed to understand and target the prominent drivers and inflammatory cascades underlying these end-organ diseases.
Collapse
Affiliation(s)
- Aaren Kettelhut
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University College of Medicine, Columbus, OH, USA
| | - Emily Bowman
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University College of Medicine, Columbus, OH, USA
| | - Nicholas T Funderburg
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
13
|
Fadini GP, Mehta A, Dhindsa DS, Bonora BM, Sreejit G, Nagareddy P, Quyyumi AA. Circulating stem cells and cardiovascular outcomes: from basic science to the clinic. Eur Heart J 2020; 41:4271-4282. [PMID: 31891403 PMCID: PMC7825095 DOI: 10.1093/eurheartj/ehz923] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 10/19/2019] [Accepted: 12/05/2019] [Indexed: 02/06/2023] Open
Abstract
The cardiovascular and haematopoietic systems have fundamental inter-relationships during development, as well as in health and disease of the adult organism. Although haematopoietic stem cells (HSCs) emerge from a specialized haemogenic endothelium in the embryo, persistence of haemangioblasts in adulthood is debated. Rather, the vast majority of circulating stem cells (CSCs) is composed of bone marrow-derived HSCs and the downstream haematopoietic stem/progenitors (HSPCs). A fraction of these cells, known as endothelial progenitor cells (EPCs), has endothelial specification and vascular tropism. In general, the levels of HSCs, HSPCs, and EPCs are considered indicative of the endogenous regenerative capacity of the organism as a whole and, particularly, of the cardiovascular system. In the last two decades, the research on CSCs has focused on their physiologic role in tissue/organ homoeostasis, their potential application in cell therapies, and their use as clinical biomarkers. In this review, we provide background information on the biology of CSCs and discuss in detail the clinical implications of changing CSC levels in patients with cardiovascular risk factors or established cardiovascular disease. Of particular interest is the mounting evidence available in the literature on the close relationships between reduced levels of CSCs and adverse cardiovascular outcomes in different cohorts of patients. We also discuss potential mechanisms that explain this association. Beyond CSCs' ability to participate in cardiovascular repair, levels of CSCs need to be interpreted in the context of the broader connections between haematopoiesis and cardiovascular function, including the role of clonal haematopoiesis and inflammatory myelopoiesis.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Anurag Mehta
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, USA
| | - Devinder Singh Dhindsa
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, USA
| | | | - Gopalkrishna Sreejit
- Division of Cardiac Surgery, Department of Surgery, Ohio State University, Columbus, OH 43210, USA
| | - Prabhakara Nagareddy
- Division of Cardiac Surgery, Department of Surgery, Ohio State University, Columbus, OH 43210, USA
| | - Arshed Ali Quyyumi
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, USA
| |
Collapse
|
14
|
Pelliccia F, Pasceri V, Moretti A, Tanzilli G, Speciale G, Gaudio C. Endothelial progenitor cells predict long-term outcome in patients with coronary artery disease: Ten-year follow-up of the PROCREATION extended study. Int J Cardiol 2020; 318:123-125. [DOI: 10.1016/j.ijcard.2020.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/13/2020] [Accepted: 06/01/2020] [Indexed: 01/26/2023]
|
15
|
Oikonomou E, Siasos G, Tsigkou V, Bletsa E, Panoilia ME, Oikonomou IN, Sinanidis I, Spinou M, Papastavrou A, Kokosias G, Zaromitidou M, Stampouloglou P, Spartalis M, Vavuranakis M, Stefanadis C, Papavassiliou AG, Tousoulis D. Coronary Artery Disease and Endothelial Dysfunction: Novel Diagnostic and Therapeutic Approaches. Curr Med Chem 2020; 27:1052-1080. [PMID: 31470773 DOI: 10.2174/0929867326666190830103219] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 04/02/2019] [Accepted: 04/30/2019] [Indexed: 12/30/2022]
Abstract
Coronary artery disease is the leading cause of morbidity and mortality worldwide. The most common pathophysiologic substrate is atherosclerosis which is an inflammatory procedure that starts at childhood and develops throughout life. Endothelial dysfunction is associated with the initiation and progression of atherosclerosis and is characterized by the impaired production of nitric oxide. In general, endothelial dysfunction is linked to poor cardiovascular prognosis and different methods, both invasive and non-invasive, have been developed for its evaluation. Ultrasound evaluation of flow mediated dilatation of the branchial artery is the most commonly used method to assessed endothelial function while intracoronary administration of vasoactive agents may be also be used to test directly endothelial properties of the coronary vasculature. Endothelial dysfunction has also been the subject of therapeutic interventions. This review article summarizes the knowledge about evaluation of endothelial function in acute coronary syndromes and stable coronary artery disease and demonstrates the current therapeutic approaches against endothelial dysfunction.
Collapse
Affiliation(s)
- Evangelos Oikonomou
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Gerasimos Siasos
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School and Harvard-MIT Biomedical Engineering Center, Massachusetts Institute of Technology, Boston, MA, United States
| | - Vasiliki Tsigkou
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Evanthia Bletsa
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Maria-Evi Panoilia
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Iris Niovi Oikonomou
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Ilias Sinanidis
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Marianna Spinou
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Angeliki Papastavrou
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Georgios Kokosias
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Marina Zaromitidou
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School and Harvard-MIT Biomedical Engineering Center, Massachusetts Institute of Technology, Boston, MA, United States
| | - Panagiota Stampouloglou
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Michail Spartalis
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Manolis Vavuranakis
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | | | - Athanasios G Papavassiliou
- Department of Biological Chemistry, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Dimitris Tousoulis
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
16
|
Tentolouris A, Eleftheriadou I, Tzeravini E, Tsilingiris D, Paschou SA, Siasos G, Tentolouris N. Endothelium as a Therapeutic Target in Diabetes Mellitus: From Basic Mechanisms to Clinical Practice. Curr Med Chem 2020; 27:1089-1131. [PMID: 30663560 DOI: 10.2174/0929867326666190119154152] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/28/2018] [Accepted: 01/09/2019] [Indexed: 12/12/2022]
Abstract
Endothelium plays an essential role in human homeostasis by regulating arterial blood pressure, distributing nutrients and hormones as well as providing a smooth surface that modulates coagulation, fibrinolysis and inflammation. Endothelial dysfunction is present in Diabetes Mellitus (DM) and contributes to the development and progression of macrovascular disease, while it is also associated with most of the microvascular complications such as diabetic retinopathy, nephropathy and neuropathy. Hyperglycemia, insulin resistance, hyperinsulinemia and dyslipidemia are the main factors involved in the pathogenesis of endothelial dysfunction. Regarding antidiabetic medication, metformin, gliclazide, pioglitazone, exenatide and dapagliflozin exert a beneficial effect on Endothelial Function (EF); glimepiride and glibenclamide, dipeptidyl peptidase-4 inhibitors and liraglutide have a neutral effect, while studies examining the effect of insulin analogues, empagliflozin and canagliflozin on EF are limited. In terms of lipid-lowering medication, statins improve EF in subjects with DM, while data from short-term trials suggest that fenofibrate improves EF; ezetimibe also improves EF but further studies are required in people with DM. The effect of acetylsalicylic acid on EF is dose-dependent and lower doses improve EF while higher ones do not. Clopidogrel improves EF, but more studies in subjects with DM are required. Furthermore, angiotensin- converting-enzyme inhibitors /angiotensin II receptor blockers improve EF. Phosphodiesterase type 5 inhibitors improve EF locally in the corpus cavernosum. Finally, cilostazol exerts favorable effect on EF, nevertheless, more data in people with DM are required.
Collapse
Affiliation(s)
- Anastasios Tentolouris
- Diabetes Center, 1st Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Ioanna Eleftheriadou
- Diabetes Center, 1st Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Evangelia Tzeravini
- Diabetes Center, 1st Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Dimitrios Tsilingiris
- Diabetes Center, 1st Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Stavroula A Paschou
- Diabetes Center, 1st Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Gerasimos Siasos
- First Department of Cardiology, Hippokration Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Nikolaos Tentolouris
- Diabetes Center, 1st Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| |
Collapse
|
17
|
Hu Z, Wang H, Fan G, Zhang H, Wang X, Mao J, Zhao Y, An Y, Huang Y, Li C, Chang L, Chu X, Li Y, Zhang Y, Qin G, Gao X, Zhang B. Danhong injection mobilizes endothelial progenitor cells to repair vascular endothelium injury via upregulating the expression of Akt, eNOS and MMP-9. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 61:152850. [PMID: 31035054 DOI: 10.1016/j.phymed.2019.152850] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 01/23/2019] [Accepted: 01/27/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUD Endothelial progenitor cells (EPCs) have been characterized as one of the key effectors of endothelial healing. The effect of Danhong injection (DHI), the most widely prescribed Chinese medicine for coronary heart disease (CHD), on EPCs mobilization remains unclear. PURPOSE We aimed to assess the effect of DHI on EPCs mobilization to repair percutaneous coronary intervention (PCI) induced vascular injury, and to investigate the characteristics and potential mechanism of DHI on EPCs mobilization. METHOD Forty-two patients with CHD underwent PCI and received stent implantation were enrolled in a Phase II clinical trials. All patients received routine western medical treatment after PCI, patients of DHI group received DHI in addition. The levels of CECs, cytokines (vWF, IL-6, CRP) and EPCs were analyzed at baseline, post-PCI and after treatment. To investigate the characteristics of DHI on EPCs mobilization, 12 healthy volunteers received intravenous infusion of DHI once and the other 12 received for 7 days. EPCs enumeration were done at a series of time points. At last we tested the effect of DHI and three chemical constituents of DHI (danshensu; lithospermic acid, LA; salvianolic acid D, SaD) on EPCs level and expression of Akt, eNOS and MMP-9 in bone marrow cells of myocardial infarction (MI) mice. RESULTS In the DHI group the angina symptoms were improved, the levels of cytokines and CECs were reduced; while EPCs population was increased after treatment. In the phase I clinical trials, EPCs counts reached a plateau phase in 9 h and maintained for more than 10 h after a single dose. After continuous administration, EPCs levels plateaued on the 3rd or 4th day, and maintain till 1 day after the withdrawal, then its levels gradually declined. DHI treatment induced a timely dependent mobilization of EPCs. DHI promoted EPCs mobilization via upregulating the expression of Akt, eNOS and MMP-9 in BM. LA and SaD have played a valuable role in EPCs mobilization. CONCLUSION These initial results demonstrated that DHI is effective in alleviating endothelial injury and promoting endothelial repair through enhancing EPCs mobilization and revealed the effect feature and possible mechanisms of DHI in mobilizing EPCs.
Collapse
Affiliation(s)
- Zhen Hu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 314 Anshan West Road, Tianjin 300193, China; Tianjin State Key Laboratory of Modern Chinese Medicine and key research department of prescription component compatibility, Tianjin University of Traditional Chinese Medicine, 312 Anshan West Road, Tianjin 300193, China; Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 312 Anshan West Road, Tianjin 300193, China
| | - Hong Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine and key research department of prescription component compatibility, Tianjin University of Traditional Chinese Medicine, 312 Anshan West Road, Tianjin 300193, China; Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 312 Anshan West Road, Tianjin 300193, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 314 Anshan West Road, Tianjin 300193, China; Tianjin State Key Laboratory of Modern Chinese Medicine and key research department of prescription component compatibility, Tianjin University of Traditional Chinese Medicine, 312 Anshan West Road, Tianjin 300193, China; Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 312 Anshan West Road, Tianjin 300193, China.
| | - Han Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine and key research department of prescription component compatibility, Tianjin University of Traditional Chinese Medicine, 312 Anshan West Road, Tianjin 300193, China; Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 312 Anshan West Road, Tianjin 300193, China
| | - Xiaoying Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine and key research department of prescription component compatibility, Tianjin University of Traditional Chinese Medicine, 312 Anshan West Road, Tianjin 300193, China; Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 312 Anshan West Road, Tianjin 300193, China
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 314 Anshan West Road, Tianjin 300193, China
| | - Yingqiang Zhao
- Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 816 Zhenli Road, Tianjin 300150, China
| | - Yi An
- The affiliated cardiovascular hospital of Qingdao university, 5 Zhiquan Road, Qingdao 266071, China
| | - Yuhong Huang
- Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 816 Zhenli Road, Tianjin 300150, China
| | - Chuan Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
| | - Lianying Chang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 314 Anshan West Road, Tianjin 300193, China
| | - Xianming Chu
- The affiliated cardiovascular hospital of Qingdao university, 5 Zhiquan Road, Qingdao 266071, China
| | - Yanfen Li
- Second Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 816 Zhenli Road, Tianjin 300150, China
| | - Yuan Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine and key research department of prescription component compatibility, Tianjin University of Traditional Chinese Medicine, 312 Anshan West Road, Tianjin 300193, China
| | - Gangjian Qin
- Tianjin State Key Laboratory of Modern Chinese Medicine and key research department of prescription component compatibility, Tianjin University of Traditional Chinese Medicine, 312 Anshan West Road, Tianjin 300193, China; Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Xiumei Gao
- Tianjin State Key Laboratory of Modern Chinese Medicine and key research department of prescription component compatibility, Tianjin University of Traditional Chinese Medicine, 312 Anshan West Road, Tianjin 300193, China; Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 312 Anshan West Road, Tianjin 300193, China.
| | - Boli Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine and key research department of prescription component compatibility, Tianjin University of Traditional Chinese Medicine, 312 Anshan West Road, Tianjin 300193, China; Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, 312 Anshan West Road, Tianjin 300193, China
| |
Collapse
|
18
|
Shimizu Y, Yamanashi H, Noguchi Y, Koyamatsu J, Kiyoura K, Kawashiri SY, Maeda T. Consumptive reduction following increased production of CD34-positive cells and carotid intima-media thickness in non-hypertensive elderly Japanese men. COGENT MEDICINE 2019. [DOI: 10.1080/2331205x.2019.1629169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Affiliation(s)
- Yuji Shimizu
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Cardiovascular Disease Prevention, Osaka Center for Cancer and Cardiovascular Disease Prevention, Osaka, Japan
| | - Hirotomo Yamanashi
- Department of General Medicine, Nagasaki University Hospital, Nagasaki, Japan
| | - Yuko Noguchi
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Jun Koyamatsu
- Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kairi Kiyoura
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shin-Ya Kawashiri
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takahiro Maeda
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of General Medicine, Nagasaki University Hospital, Nagasaki, Japan
- Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
19
|
Skrzypkowska MW, Gutknecht PG, Ryba-Stanisławowska ME, Słomiński B, Siebert J, Myśliwska JM. CD34+ and CD34+VEGFR2+ cells in poorly controlled hypertensive patients. J Hum Hypertens 2018; 33:863-872. [DOI: 10.1038/s41371-018-0145-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/07/2018] [Accepted: 11/29/2018] [Indexed: 11/09/2022]
|
20
|
Hou Y, Li C. Stem/Progenitor Cells and Their Therapeutic Application in Cardiovascular Disease. Front Cell Dev Biol 2018; 6:139. [PMID: 30406100 PMCID: PMC6200850 DOI: 10.3389/fcell.2018.00139] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 09/28/2018] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular disease is the leading cause of death in the world. The stem/progenitor cell-based therapy has emerged as a promising approach for the treatment of a variety of cardiovascular diseases including myocardial infarction, stroke, peripheral arterial disease, and diabetes. An increasing number of evidence has shown that stem/progenitor cell transplantation could replenish damaged cells, improve cardiac and vascular functions, and repair injured tissues in many pre-clinical studies and clinical trials. In this review, we have outlined the major types of stem/progenitor cells, and summarized the studies in applying these cells, especially endothelial stem/progenitor cells and their derivatives, in the treatment of cardiovascular disease. Here the strategies used to improve the stem/progenitor cell-based therapies in cardiovascular disease and the challenges with these therapies in clinical applications are also reviewed.
Collapse
Affiliation(s)
| | - Chunying Li
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
21
|
Shimizu Y, Sato S, Noguchi Y, Koyamatsu J, Yamanashi H, Higashi M, Nagayoshi M, Kawashiri SY, Nagata Y, Takamura N, Maeda T. Association between tongue pressure and subclinical carotid atherosclerosis in relation to platelet levels in hypertensive elderly men: a cross-sectional study. Environ Health Prev Med 2018; 23:31. [PMID: 30021529 PMCID: PMC6052576 DOI: 10.1186/s12199-018-0720-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 06/13/2018] [Indexed: 12/31/2022] Open
Abstract
Background Age-related low-grade inflammation causing endothelial disruption influences sarcopenia, hypertension, and atherosclerosis. We reported previously that maintenance of muscle strength in elderly hypertensive men with high platelet levels is positively associated with subclinical atherosclerosis but not in those with low platelet levels. Since reduced tongue pressure is related to sarcopenia, tongue pressure may be associated with subclinical carotid atherosclerosis in hypertensive elderly subjects, and platelet levels may function as an indicator of the association between tongue pressure and subclinical carotid atherosclerosis. Methods We conducted a cross-sectional study of 342 hypertensive elderly Japanese men aged 60–89 who participated in an annual health check-up in 2015 and 2016. Subclinical carotid atherosclerosis was defined as a common carotid intima-media thickness (CIMT) of 1.1 mm or more. Results In the overall study population, 171 subjects demonstrated low platelets (< 21.4 × 104/μL). Tongue pressure was significantly inversely associated with subclinical carotid atherosclerosis in these subjects, but not in subjects with high platelets. The known cardiovascular risk factor adjusted odds ratios (ORs) and 95% confidence intervals (CIs) of subclinical carotid atherosclerosis for a 1 standard deviation (SD) increment in tongue pressure (10.4 kPa) were 0.54 (0.35, 0.85) and 1.31 (0.87, 1.96), respectively. Conclusion Tongue pressure is inversely associated with subclinical carotid atherosclerosis in hypertensive elderly men with low platelet levels, but not in those with high levels. This finding may thus constitute an efficient tool for clarifying the background mechanism of age-related diseases such as sarcopenia, hypertension, and atherosclerosis.
Collapse
Affiliation(s)
- Yuji Shimizu
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan. .,Department of Cardiovascular Disease Prevention, Osaka Center for Cancer and Cardiovascular Disease Prevention, Osaka, Japan.
| | - Shimpei Sato
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yuko Noguchi
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Jun Koyamatsu
- Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hirotomo Yamanashi
- Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Miho Higashi
- Department of Global Health, Medicine and Welfare, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Mako Nagayoshi
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shin-Ya Kawashiri
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yasuhiro Nagata
- Center for Comprehensive Community Care Education, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Noboru Takamura
- Department of Global Health, Medicine and Welfare, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takahiro Maeda
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
22
|
Yang JX, Pan YY, Wang XX, Qiu YG, Mao W. Endothelial progenitor cells in age-related vascular remodeling. Cell Transplant 2018; 27:786-795. [PMID: 29882417 PMCID: PMC6047273 DOI: 10.1177/0963689718779345] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence has demonstrated that endothelial progenitor cells (EPCs) could facilitate the reendothelialization of injured arteries by replacing the dysfunctional endothelial cells, thereby suppressing the formation of neointima. Meanwhile, other findings suggest that EPCs may be involved in the pathogenesis of age-related vascular remodeling. This review is presented to summarize the characteristics of EPCs and age-related vascular remodeling. In addition, the role of EPCs in age-related vascular remodeling and possible solutions for improving the therapeutic effects of EPCs in the treatment of age-related diseases are discussed.
Collapse
Affiliation(s)
- Jin-Xiu Yang
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China.,2 Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Yan-Yun Pan
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
| | - Xing-Xiang Wang
- 2 Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Yuan-Gang Qiu
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
| | - Wei Mao
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
| |
Collapse
|
23
|
Jin Z, Tan Q, Sun B. Telmisartan ameliorates vascular endothelial dysfunction in coronary slow flow phenomenon (CSFP). Cell Biochem Funct 2018; 36:18-26. [PMID: 29314204 DOI: 10.1002/cbf.3313] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/22/2017] [Accepted: 11/29/2017] [Indexed: 11/07/2022]
Affiliation(s)
- Zhe Jin
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, Tsinghua University, Beijing, China
| | - Qindong Tan
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bei Sun
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
24
|
Haberzettl P, Conklin DJ, Abplanalp WT, Bhatnagar A, O'Toole TE. Inhalation of Fine Particulate Matter Impairs Endothelial Progenitor Cell Function Via Pulmonary Oxidative Stress. Arterioscler Thromb Vasc Biol 2017; 38:131-142. [PMID: 29191925 DOI: 10.1161/atvbaha.117.309971] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 11/20/2017] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Exposure to fine particulate matter (PM2.5) air pollution is associated with the depletion of circulating endothelial progenitor cells (EPCs), as well as vascular injury and dysfunction. Nevertheless, it remains unclear whether PM2.5 exposure leads to significant impairments in EPC function. Hence, we studied the effects of PM2.5 on EPC-mediated recovery of vascular perfusion after hindlimb ischemia and examined the mechanisms whereby PM2.5 exposure affects EPC abundance and function. APPROACH AND RESULTS In comparison with EPCs isolated from mice breathing filtered air, EPCs from mice exposed for 9 consecutive days (6 hours per day) to concentrated ambient PM2.5 (CAP) had defects in both proliferation and tube formation. However, CAP exposure of mice overexpressing extracellular superoxide dismutase (ecSOD-Tg) in the lungs did not affect EPC tube formation. Exposure to CAP also suppressed circulating EPC levels, VEGF (vascular endothelial growth factor)-stimulated aortic Akt phosphorylation, and plasma NO levels in wild-type but not in ecSOD-Tg mice. EPCs from CAP-exposed wild-type mice failed to augment basal recovery of hindlimb perfusion when injected into unexposed mice subjected to hindlimb ischemia; however, these deficits in recovery of hindlimb perfusion were absent when using EPCs derived from CAP-exposed ecSOD-Tg mice. The improved reparative function of EPCs from CAP-exposed ecSOD-Tg mice was also reflected by greater expression of Mmp-9 and Nos3 when compared with EPCs from CAP-exposed wild-type mice. CONCLUSIONS Exposure to PM2.5 impairs EPC abundance and function and prevents EPC-mediated vascular recovery after hindlimb ischemia. This defect is attributed, in part, to pulmonary oxidative stress and was associated with vascular VEGF resistance and a decrement in NO bioavailability.
Collapse
Affiliation(s)
- Petra Haberzettl
- From the Department of Medicine, Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, KY
| | - Daniel J Conklin
- From the Department of Medicine, Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, KY
| | - Wesley T Abplanalp
- From the Department of Medicine, Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, KY
| | - Aruni Bhatnagar
- From the Department of Medicine, Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, KY
| | - Timothy E O'Toole
- From the Department of Medicine, Diabetes and Obesity Center, Institute of Molecular Cardiology, University of Louisville, KY.
| |
Collapse
|
25
|
Park SJ, Son JW, Park SM, Choi HH, Hong KS. Relationship between inter-arm blood pressure difference and severity of coronary atherosclerosis. Atherosclerosis 2017. [DOI: 10.1016/j.atherosclerosis.2017.06.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
26
|
Patel P. Telmisartan: clinical evidence across the cardiovascular and renal disease continuum. DRUGS & THERAPY PERSPECTIVES 2016. [DOI: 10.1007/s40267-016-0366-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
27
|
Lake JE, Seang S, Kelesidis T, Currier JS, Yang OO. Telmisartan increases vascular reparative capacity in older HIV-infected adults: a pilot study. HIV CLINICAL TRIALS 2016; 17:225-232. [PMID: 27658740 DOI: 10.1080/15284336.2016.1234222] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) are bone marrow-derived cells that contribute to vascular repair. EPCs may be reduced in HIV-infected (HIV+) persons, contributing to cardiovascular disease (CVD). Telmisartan is an angiotensin receptor blocker that increases EPCs in HIV-uninfected adults. OBJECTIVE To assess telmisartan's effects on EPC number and immunophenotype in older HIV + adults at risk for CVD. METHODS HIV + persons ≥50 years old with HIV-1 RNA < 50 copies/mL on suppressive antiretroviral therapy and ≥1 CVD risk factor participated in a prospective, open-label, pilot study of oral telmisartan 80 mg daily for 12 weeks. Using CD34 and CD133 as markers of early maturity and KDR as a marker of endothelial lineage commitment, EPCs were quantified via flow cytometry and defined as viable CD3-/CD33-/CD19-/glycophorin- cells of four immunophenotypes: CD133+/KDR+, CD34+/KDR+, CD34+/CD133+, or CD34+/KDR+/CD133+. The primary endpoint was a 12-week change in EPC subsets (NCT01578772). RESULTS Seventeen participants (88% men, median age 60 years and peripheral CD4+ T lymphocyte count 625 cells/mm3) enrolled and completed the study. After 6 and 12 weeks of telmisartan, frequencies of all EPC immunophenotypes were higher than baseline (all p < 0.10 except week 12 CD133+/KDR+ EPC, p = 0.13). Participants with lower baseline EPC levels had the largest gains. Additionally, the percentage of CD34+ cells with endothelial commitment (KDR+) increased. CONCLUSIONS Our data suggest that telmisartan use is associated with an increase in circulating EPCs in older HIV + individuals with CVD risk factors. Further controlled studies are needed to assess whether EPC increases translate to a reduction in CVD risk in this population.
Collapse
Affiliation(s)
- Jordan E Lake
- a Division of Infectious Diseases, Department of Medicine , University of California , Los Angeles , CA , USA
| | - Sophie Seang
- a Division of Infectious Diseases, Department of Medicine , University of California , Los Angeles , CA , USA
| | - Theodoros Kelesidis
- a Division of Infectious Diseases, Department of Medicine , University of California , Los Angeles , CA , USA
| | - Judith S Currier
- a Division of Infectious Diseases, Department of Medicine , University of California , Los Angeles , CA , USA
| | - Otto O Yang
- a Division of Infectious Diseases, Department of Medicine , University of California , Los Angeles , CA , USA.,b Department of Microbiology, Immunology, and Molecular Genetics , University of California , Los Angeles , CA , USA.,c AIDS Healthcare Foundation , Los Angeles , CA , USA
| |
Collapse
|
28
|
Wang H, Yin YG, Huang H, Zhao XH, Yu J, Wang Q, Li W, Cai KY, Ding SF. Transplantation of EPCs overexpressing PDGFR-β promotes vascular repair in the early phase after vascular injury. BMC Cardiovasc Disord 2016; 16:179. [PMID: 27619504 PMCID: PMC5020463 DOI: 10.1186/s12872-016-0353-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 08/26/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) play important roles in the regeneration of the vascular endothelial cells (ECs). Platelet-derived growth factor receptor (PDGFR)-β is known to contribute to proliferation, migration, and angiogenesis of EPCs, this study aims to investigate effects of transplantation of EPCs overexpressing PDGFR-β on vascular regeneration. METHODS We transplanted genetically modified EPCs overexpressing PDGFR-β into a mouse model with carotid artery injury. After 3 days of EPCs transplantation, the enhanced green fluorescent protein (EGFP)-expressing cells were found at the injury site and the lining of the lumen by laser scanning confocal microscope (LSCM). At 4, 7, and 14 days of the carotid artery injury, reendothelialization was evaluated by Evans Blue staining. Neointima formation was evaluated at day 14 with hematoxylin and eosin (HE) staining by calculating the neointimal area, medial area, and neointimal/media (NI/M) ratio. Intimal cell apoptosis was evaluated using TUNEL assay. Then we tested whether PDGF-BB-induced VSMC migration and PDGF-BB's function in reducing VSMC apoptosis can be attenuated by EPCs overexpressing PDGFR-β in a transwell co-culture system. RESULTS Our results showed that EPCs overexpressing PDGFR-β accelerates reendothelialization and mitigates neointimal formation at 14 days after injury. Moreover, we found that there is great possibility that EPCs overexpressing PDGFR-β enhanc VSMC apoptosis and suppress VSMC migration by competitive consumption of PDGF-BB in the early phase after carotid artery injury in mice. CONCLUSIONS We report the first in vivo and in vitro evidence that transplantation of genetically modified EPC can have a combined effect of both amplifying the reendothelialization capacity of EPCs and inhibiting neointima formation so as to facilitate better inhibition of adverse remodeling after vascular injury.
Collapse
Affiliation(s)
- Hang Wang
- Cadre Ward Two, Wuhan General Hospital of Guangzhou Military Command, Wuhan, 430070, China
| | - Yang-Guang Yin
- Intensive Care Unit, The sixth people's hospital of Chongqing, Nan'an District, Chongqing, 400060, China
| | - Hao Huang
- Clinic center, Shenzhen Hornetcorn Biotechnology Company, Ltd, Shenzhen, 518400, China
| | - Xiao-Hui Zhao
- Institute of Cardiovascular Science, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Jie Yu
- Institute of Cardiovascular Science, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Qiang Wang
- Institute of Cardiovascular Science, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Wei Li
- Institute of Cardiovascular Science, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Ke-Yin Cai
- Cadre Ward Two, Wuhan General Hospital of Guangzhou Military Command, Wuhan, 430070, China
| | - Shi-Fang Ding
- Institute of Cardiovascular Science, Wuhan General Hospital of Guangzhou Military Command, Wuhan, 430070, China.
| |
Collapse
|
29
|
Guo Y, Ledesma RA, Peng R, Liu Q, Xu D. The Beneficial Effects of Cardiac Rehabilitation on the Function and Levels of Endothelial Progenitor Cells. Heart Lung Circ 2016; 26:10-17. [PMID: 27614559 DOI: 10.1016/j.hlc.2016.06.1210] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 04/08/2016] [Accepted: 06/06/2016] [Indexed: 12/19/2022]
Abstract
Cardiac rehabilitation (CR) is a comprehensive program, which mainly focusses on exercise training, disease evaluation, cardiovascular risk factors control, medication therapy, psychosocial intervention, and patient education. Although the beneficial properties of CR have been widely evidenced, its mechanism is still not completely clarified. To date, endothelial progenitor cells (EPCs) have been explored by emerging studies, and evidence has suggested that CR, especially exercise training, significantly increases the function and levels of EPCs, which is likely to elucidate the profiting mechanism of CR. Thus, this review summarises the potential relationship between CR and EPCs with an aim of providing novel directions for future CR research.
Collapse
Affiliation(s)
- Yuan Guo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Robert Andre Ledesma
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, USA
| | - Ran Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Qiong Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
30
|
Altabas V, Altabas K, Kirigin L. Endothelial progenitor cells (EPCs) in ageing and age-related diseases: How currently available treatment modalities affect EPC biology, atherosclerosis, and cardiovascular outcomes. Mech Ageing Dev 2016; 159:49-62. [PMID: 26919825 DOI: 10.1016/j.mad.2016.02.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/25/2016] [Accepted: 02/22/2016] [Indexed: 12/15/2022]
Abstract
Endothelial progenitor cells (EPCs) are mononuclear cells that circulate in the blood and are derived from different tissues, expressing cell surface markers that are similar to mature endothelial cells. The discovery of EPCs has lead to new insights in vascular repair and atherosclerosis and also a new theory for ageing. EPCs from the bone marrow and some other organs aid in vascular repair by migrating to distant vessels where they differentiate into mature endothelial cells and replace old and injured endothelial cells. The ability of EPCs to repair vascular damage depends on their number and functionality. Currently marketed drugs used in a variety of diseases can modulate these characteristics. In this review, the effect of currently available treatment options for cardiovascular and metabolic disorders on EPC biology will be discussed. The various EPC-based therapies that will be discussed include lipid-lowering agents, antihypertensive agents, antidiabetic drugs, phosphodiesteraze inhibitors, hormones, as well as EPC capturing stents.
Collapse
Affiliation(s)
- Velimir Altabas
- Department of Internal Medicine, University Clinical Hospital "Sestre milosrdnice", Zagreb, Croatia.
| | - Karmela Altabas
- Department of Internal Medicine, University Clinical Hospital "Sestre milosrdnice", Zagreb, Croatia.
| | - Lora Kirigin
- Department of Internal Medicine, University Clinical Hospital "Sestre milosrdnice", Zagreb, Croatia.
| |
Collapse
|
31
|
Ahmadian E, Jafari S, Yari Khosroushahi A. Role of angiotensin II in stem cell therapy of cardiac disease. J Renin Angiotensin Aldosterone Syst 2015; 16:702-11. [PMID: 26670032 DOI: 10.1177/1470320315621225] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 09/01/2015] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION The renin angiotensin system (RAS) is closely related to the cardiovascular system, body fluid regulation and homeostasis. MATERIALS AND METHODS Despite common therapeutic methods, stem cell/progenitor cell therapy is daily increasing as a term of regenerative medicine. RAS and its pharmacological inhibitors are not only involved in physiological and pathological aspects of the cardiovascular system, but also affect the different stages of stem cell proliferation, differentiation and function, via interfering cell signaling pathways. RESULTS This study reviews the new role of RAS, in particular Ang II distinct from other common roles, by considering its regulating impact on the different signaling pathways involved in the cardiac and endothelial tissue, as well as in stem cell transplantation. CONCLUSIONS This review focuses on the impact of stem cell therapy on the cardiovascular system, the role of RAS in stem cell differentiation, and the role of RAS inhibition in cardiac stem cell growth and development.
Collapse
Affiliation(s)
- Elham Ahmadian
- Biotechnology Research Center, Tabriz University of Medical Science, Tabriz, Iran Department of Pharmacology and Toxicology, Tabriz University of Medical Science, Tabriz, Iran Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
| | - Samira Jafari
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran Department of Pharmaceutical Nanotechnology, Tabriz University of Medical Science, Tabriz, Iran
| | - Ahmad Yari Khosroushahi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran Department of Pharmacognosy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
32
|
Chen JD, Liu M, Chen XH, Yang ZJ. Effect of Angiotensin Receptor Blockers on Flow-Mediated Vasodilation: A Meta-Analysis of Randomized Controlled Trials. Cardiology 2015; 131:69-79. [DOI: 10.1159/000375259] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 01/14/2015] [Indexed: 11/19/2022]
Abstract
Background: In a meta-analysis, we investigated the effects of angiotensin receptor blockers (ARBs) in comparison to placebo or other classes of antihypertensive drugs on endothelial function, which was measured by brachial flow-mediated vasodilation (FMD). Methods: We searched for randomized controlled trials that compared ARBs with placebo or other classes of antihypertensive drugs in improving FMD. A random-effect model was used to compute pooled estimates. Results: In 13 trials (n = 529), ARBs were more efficacious in improving brachial FMD than placebo [pooled weighted mean change difference (WMD) 1.34%, 95% CI, 0.93-1.75%, p < 0.001]. In 15 trials (n = 918), treatment with ARBs had a significant effect on brachial FMD when compared with other antihypertensive drugs (pooled WMD 0.59%, 95% CI, 0.20-0.98%, p = 0.003 with significant heterogeneity). ARBs were also more efficacious in improving brachial FMD than calcium channel blockers (CCBs; pooled WMD 1.61%, 95% CI, 0.72-2.49%, p < 0.001) but not the other classes of drugs (p ≥ 0.072). Conclusions: This meta-analysis shows that ARBs improve brachial FMD, a marker of endothelial function, and that they are superior to placebo and CCBs. There was no significant difference in the effect on brachial FMD between ARBs and the other antihypertensive drugs.
Collapse
|
33
|
Altabas V. Diabetes, Endothelial Dysfunction, and Vascular Repair: What Should a Diabetologist Keep His Eye on? Int J Endocrinol 2015; 2015:848272. [PMID: 26089898 PMCID: PMC4452196 DOI: 10.1155/2015/848272] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 01/13/2015] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular complications are the most common complications of diabetes mellitus. A prominent attribute of diabetic cardiovascular complications is accelerated atherosclerosis, considered as a still incurable disease, at least at more advanced stages. The discovery of endothelial progenitor cells (EPCs), able to replace old and injured mature endothelial cells and capable of differentiating into healthy and functional endothelial cells, has offered the prospect of merging the traditional theories on the pathogenesis of atherosclerosis with evolving concepts of vascular biology. The literature supports the notion that EPC alterations are involved in the pathogenesis of vascular diseases in diabetics, but at present many questions remain unanswered. In this review the aspects linking endothelial progenitor cells to the altered vascular biology in diabetes mellitus are discussed.
Collapse
Affiliation(s)
- V. Altabas
- Department for Endocrinology, Diabetes and Metabolic Diseases “Mladen Sekso”, Clinic for Internal Medicine, University Hospital Center “Sestre Milosrdnice”, 10000 Zagreb, Croatia
- *V. Altabas:
| |
Collapse
|
34
|
Lee PSS, Poh KK. Endothelial progenitor cells in cardiovascular diseases. World J Stem Cells 2014; 6:355-366. [PMID: 25126384 PMCID: PMC4131276 DOI: 10.4252/wjsc.v6.i3.355] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 04/03/2014] [Indexed: 02/06/2023] Open
Abstract
Endothelial dysfunction has been associated with the development of atherosclerosis and cardiovascular diseases. Adult endothelial progenitor cells (EPCs) are derived from hematopoietic stem cells and are capable of forming new blood vessels through a process of vasculogenesis. There are studies which report correlations between circulating EPCs and cardiovascular risk factors. There are also studies on how pharmacotherapies may influence levels of circulating EPCs. In this review, we discuss the potential role of endothelial progenitor cells as both diagnostic and prognostic biomarkers. In addition, we look at the interaction between cardiovascular pharmacotherapies and endothelial progenitor cells. We also discuss how EPCs can be used directly and indirectly as a therapeutic agent. Finally, we evaluate the challenges facing EPC research and how these may be overcome.
Collapse
|
35
|
A meta-analysis of randomized controlled trials of telmisartan for flow-mediated dilatation. Hypertens Res 2014; 37:845-51. [PMID: 24718299 DOI: 10.1038/hr.2014.81] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 03/03/2014] [Accepted: 03/04/2014] [Indexed: 01/06/2023]
Abstract
There have been a number of small-sized underpowered randomized controlled trials to assess effects of telmisartan on flow-mediated dilatation (FMD). To determine whether telmisartan increases FMD, we performed a meta-analysis of these trials. MEDLINE, EMBASE and the Cochrane Central Register of Controlled Trials were searched through December 2013. Eligible studies were prospective randomized controlled trials of telmisartan reporting FMD as an outcome. Search terms included: telmisartan; endothelial function/dysfunction; flow-mediated dilation/dilatation/vasodilation/vasodilatation; and randomized, randomly or randomization. Included studies were reviewed to determine the number of patients randomized, mean duration of treatment and percent changes of FMD. Of 25 potentially relevant articles screened initially, seven reports of randomized trials enrolling a total of 398 patients were identified and included. A pooled analysis of the seven trials demonstrated a statistically significant increase in FMD by 48.7%, with telmisartan relative to control in the random-effects model (mean difference, 48.72%; 95% confidence interval, 15.37-82.08%; P for effect=0.004; P for heterogeneity <0.00001). Exclusion of any single trial from the analysis did not substantively alter the overall result of our analysis. There was no evidence of significant publication bias. In conclusion, the present meta-analysis of seven randomized controlled trials enrolling a total of 398 patients confirmed the evidence of a significant increase in FMD with telmisartan, which suggests that telmisartan may improve endothelial dysfunction.
Collapse
|
36
|
Li S, Wu Y, Yu G, Xia Q, Xu Y. Angiotensin II receptor blockers improve peripheral endothelial function: a meta-analysis of randomized controlled trials. PLoS One 2014; 9:e90217. [PMID: 24595033 PMCID: PMC3940822 DOI: 10.1371/journal.pone.0090217] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 01/26/2014] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE(S) Several studies have assessed the effect of angiotensin II receptor blockers (ARBs) on peripheral endothelial dysfunction as measured by flow-mediated vasodilatation (FMD), a widely-used indicator for endothelial function. We conducted a meta-analysis to investigate the effect in comparison to placebo or no treatment and other antihypertensives. METHODS MEDLINE, Cochrane library and EMBASE were searched to September 2013 for randomized controlled trials (RCTs) that assessed the effect of ARBs versus placebo or no treatment and other antihypertensives (angiotensin-converting enzyme inhibitors (ACEIs), calcium channel blockers (CCBs), β-blockers, diuretics) by forearm FMD. Furthermore, we also use meta-regression to analyze the relationship between the endothelial function and the duration of ARBs treatments. RESULTS In 11 trials including 590 patients, ARBs (n = 315) significantly improved FMD (1.36%, 95% confidence internal [CI]:1.28 to 1.44) versus placebo or no treatment (n = 275). In 16 trials that included 1028 patients, ARBs (n = 486) had a significant effect (0.59%, 95% CI: 0.25 to 0.94) on FMD when compared with other antihypertensives (n = 542). In 8 trials, ARBs (n = 174) had no significant effect (-0.14%, 95% CI: -0.32 to 0.03) compared with ACEI (n = 173). Compared with others, the benefits of ARBs, respectively, were 1.67% (95% CI: 0.65 to 0.93) in 7 trials with CCBs, 0.79% (95% CI: 0.42 to 1.01) with β-blockers in 3 trials and 0.9% (95% CI: 0.77 to 1.03) with diuretics in 3 trials. Importantly, we found ARBs were less effective in a long time span (95% CI: -1.990 to -0.622) than the first 6 months (95% CI: -0.484 to 0.360). CONCLUSIONS This study shows that ARBs improve peripheral endothelial function and are superior to CCBs, β-blockers and diuretics. However, the effect couldn't be maintained for a long time. In addition, there was no significant difference between ARBs and ACEI.
Collapse
Affiliation(s)
- Shuang Li
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yan Wu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ge Yu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qing Xia
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
37
|
Artom N, Montecucco F, Mach F, Dallegri F, Pende A. Angiotensin II receptor antagonists in acute coronary syndromes. Eur J Clin Invest 2014; 44:219-230. [PMID: 24289238 DOI: 10.1111/eci.12198] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 11/01/2013] [Indexed: 12/25/2022]
Abstract
BACKGROUND It is well known that inappropriate or exaggerated activity of the renin-angiotensin system might contribute to the development of systemic hypertension with consequent organ injury and associated increased risk of acute cardiovascular (CV) diseases. This review will discuss evidence form basic research and clinical studies, investigating the efficacy of angiotensin II receptor blockers (ARBs) in the management of acute coronary syndromes (ACS). MATERIALS AND METHODS This narrative review is based on the material found on MEDLINE and PubMed up to June 2013. We looked for the terms 'angiotensin, AT1 receptor, ACE inhibitors' in combination with 'acute coronary syndromes, acute myocardial infarction, pathophysiology'. RESULTS Preclinical studies showed relevant protective effects of ARBs to reduce adverse cardiac remodelling in animal models of acute cardiac ischaemia. However, although recommended in Consensus guidelines as a good alternative to angiotensin-converting enzyme inhibitors (ACEIs), clinical studies did not confirm a superior efficacy of the ARBs as compared to ACEIs. As a matter of fact for some authors, these drugs might potentially have deleterious effects increasing the CV risk. CONCLUSIONS Emerging evidence from clinical trials suggests that the use of ARBs in ACS might be controversial, and caution should be used for their clinical use to replace ACEIs in ACS.
Collapse
Affiliation(s)
- Nathan Artom
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | | | | | | | | |
Collapse
|
38
|
Hu ZP, Fang XL, Qian HY, Fang N, Wang BN, Wang Y. Telmisartan prevents angiotensin II-induced endothelial dysfunction in rabbit aorta via activating HGF/Met system and PPARγ pathway. Fundam Clin Pharmacol 2013; 28:501-11. [PMID: 24188213 DOI: 10.1111/fcp.12057] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Revised: 10/11/2013] [Accepted: 10/31/2013] [Indexed: 01/06/2023]
Affiliation(s)
- Ze-Ping Hu
- Department of Cardiology; The First Affiliated Hospital; Anhui Medical University; 218 Jixi Road Hefei Anhui Province 230022 China
| | - Xiao-Ling Fang
- Operating Room; The First Affiliated Hospital; Anhui Medical University; 218 Jixi Road Hefei Anhui Province 230022 China
| | - Hai-Yan Qian
- Department of Cardiology; State Key Laboratory of Cardiovascular Disease; Fuwai Hospital; National Center for Cardiovascular Diseases; Chinese Academy of Medical Sciences and Peking Union Medical College; 167 North Lishi Road Xicheng District Beijing 100037 China
| | - Nan Fang
- Department of Cardiology; The First Affiliated Hospital; Anhui Medical University; 218 Jixi Road Hefei Anhui Province 230022 China
| | - Bang-Ning Wang
- Department of Cardiology; The First Affiliated Hospital; Anhui Medical University; 218 Jixi Road Hefei Anhui Province 230022 China
| | - Yuan Wang
- Laboratory of Molecular Biology and Department of Biochemistry; Anhui Medical University; 81 Meishan Road Hefei Anhui Province 230032 China
| |
Collapse
|
39
|
Desouza CV. Does drug therapy reverse endothelial progenitor cell dysfunction in diabetes? J Diabetes Complications 2013; 27:519-25. [PMID: 23809765 DOI: 10.1016/j.jdiacomp.2013.04.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 04/03/2013] [Accepted: 04/09/2013] [Indexed: 12/17/2022]
Abstract
Endothelial progenitor cells (EPCs) are vital for the maintenance and repair of the endothelium. Decreased EPC number and function have been associated with increased cardiovascular (CVD) risk. Patients with diabetes have decreased number of circulating EPCs and decreased EPC function. This may account for some of the increased CVD risk seen in patients with diabetes that is not explained by traditional risk factors such as glycemic control, dyslipidemia and hypertension. Recent studies seem to indicate that drugs commonly used in diabetes patients such as metformin, thiazolidinediones, GLP-1 agonists, DPP-4 inhibitors, insulin, statins and ACE inhibitors may increase EPC number and improve EPC function. The mechanisms by which these drugs modulate EPC function may involve reduction in inflammation, oxidative stress and insulin resistance as well as an increase in nitric oxide (NO) bioavailability. This review will discuss the evidence in the literature regarding the above mentioned topics.
Collapse
Affiliation(s)
- Cyrus V Desouza
- Omaha VA Medical Center, Omaha, NE, USA; University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
40
|
Dessì M, Madeddu C, Piras A, Cadeddu C, Antoni G, Mercuro G, Mantovani G. Long-term, up to 18 months, protective effects of the angiotensin II receptor blocker telmisartan on Epirubin-induced inflammation and oxidative stress assessed by serial strain rate. SPRINGERPLUS 2013; 2:198. [PMID: 23741643 PMCID: PMC3664751 DOI: 10.1186/2193-1801-2-198] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 04/24/2013] [Indexed: 01/16/2023]
Abstract
PURPOSE The primary objective of the present study was to show the long lasting cardioprotective activity, at different time-points, up to 18 month-follow-up, of telmisartan in preserving the systolic function (assessed as Strain Rate-SR) in cancer patients treated with EPI both in the adjuvant and metastatic setting; the secondary objective was to confirm the correlation of the cardioprotective activity of telmisartan with a reduction of inflammation and oxidative stress induced by EPI. METHODS Phase II single blind placebo-controlled randomized trial. Sample size 50 patients per arm: based on a pre-planned interim analysis for early stopping rules, the study was discontinued for ethical reasons at 49 patients. Cardiovascular disease-free patients with cancer at different sites eligible for EPI-based treatment randomized to: telmisartan n = 25 or placebo n = 24. Echocardiography Tissue Doppler imaging (TDI) strain and strain rate was performed, serum levels of proinflammatory cytokines (IL-6, TNF-α) and oxidative stress (reactive oxygen species, ROS) were assessed at baseline, every 100 mg/m(2) EPI dose and at 6-, 12- and 18-month follow-up (FU). RESULTS Significant SR peak reduction in both arms was observed at t2 (cumulative dose EPI 200 mg/m(2)) vs t0. Conversely, at t3, t4, 6-, 12- and 18-month FU SR increased towards normal range in the telmisartan arm, while in the placebo arm SR remained significantly lower. Differences between SR changes in the placebo and telmisartan arm were significant from t3 up to 18 month-FU. IL-6 and ROS increased significantly in the placebo arm at t2 but did not change in the telmisartan arm. A significant (p < 0.05) correlation between changes of SR vs IL-6 and ROS was observed. CONCLUSIONS Our results suggest that the protective effect of telmisartan is long lasting, probably by ensuring a permanent (at least up to 18-month FU) defense against chronic or late-onset types of anthracycline-induced cardiotoxicity.
Collapse
Affiliation(s)
- Mariele Dessì
- Department of Medical Sciences: Medical Oncology, University of Cagliari, Cagliari, Italy
| | | | | | | | | | | | | |
Collapse
|
41
|
Patarroyo Aponte MM, Francis GS. Effect of Angiotensin-converting enzyme inhibitors and Angiotensin receptor antagonists in atherosclerosis prevention. Curr Cardiol Rep 2012; 14:433-42. [PMID: 22562592 DOI: 10.1007/s11886-012-0275-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Atherosclerosis is a highly complex biological process that has become the scourge of modern civilization. Endothelial dysfunction is the first step in the development of atherosclerosis. The renin-angiotensin-aldosterone system (RAAS) plays an important role in the development of endothelial dysfunction and atherosclerosis. Several studies have shown that in vitro blockade of the RAAS is associated with improvement in markers of endothelial dysfunction and inflammation. Many clinical trials have demonstrated a clear benefit of angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) manifested by a reduction of cardiovascular events. These findings suggest that ACEIs and ARBs can play an important role in prevention of atherosclerosis and in the delay of its progression. In this review we focus on the importance of RAAS blockade to prevent or delay progression of atherosclerosis and its impact on reduction of cardiovascular events.
Collapse
Affiliation(s)
- Maria M Patarroyo Aponte
- Division of Cardiovascular Medicine, Lillehei Heart Institute, University of Minnesota Medical Center, 420 Delaware Street SE, Minneapolis, MN 55455, USA.
| | | |
Collapse
|
42
|
Jung C, Florvaag A, Oberle V, Fritzenwanger M, Kretschmar D, Kuethe F, Betge S, Goebel B, Franz M, Barz D, Ferrari M, Figulla HR. Positive effect of eplerenone treatment on endothelial progenitor cells in patients with chronic heart failure. J Renin Angiotensin Aldosterone Syst 2012; 13:401-6. [DOI: 10.1177/1470320312447650] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Background: Endothelial progenitor cells (EPCs) are known to play a significant role in reendothelialization and vascular repair. Recently, a mineralocorticoid receptor was demonstrated to be expressed by EPCs. The study aimed to evaluate a potential influence of eplerenone treatment on the total number of EPCs in patients with chronic heart failure. Methods: Eighty-seven male patients with chronic heart failure were included (age: 23–83 years; body mass index 29.1 ± 5.1 kg/m2; New York Heart failure classification (NYHA) I: 29 patients, NYHA II: 32 patients, NYHA III: 26 patients). Numbers of circulating EPCs were quantified immediately using flow cytometry. Twenty-eight patients received therapy with eplerenone. Patients were further characterized by echocardiography, spirometry and laboratory markers. Results: Patients with ongoing eplerenone administration showed higher levels of circulating cells expressing CD34+ ( p<0.05) and CD34+KDR+ ( p<0.05) and CD34+CD133+KDR+ cells ( p<0.05). The effects of eplerenone treatment could be shown to be independent of NYHA status, genesis of the underlying cardiovascular morbidity, left ventricular function and co-medication. Conclusion: Patients with chronic heart failure treated with eplerenone show higher numbers of EPCs. The clinical benefit for treatment with eplerenone has been demonstrated even for patients with mild heart failure and might be partially mediated by EPCs.
Collapse
Affiliation(s)
- Christian Jung
- First Department of Medicine, Friedrich-Schiller-University Jena, Germany
| | - Anna Florvaag
- First Department of Medicine, Friedrich-Schiller-University Jena, Germany
| | - Volker Oberle
- Division of Cardiology, Institute for Transfusion Medicine, Friedrich-Schiller-University Jena, Germany
| | | | - Daniel Kretschmar
- First Department of Medicine, Friedrich-Schiller-University Jena, Germany
| | - Friedhelm Kuethe
- First Department of Medicine, Friedrich-Schiller-University Jena, Germany
| | - Stefan Betge
- First Department of Medicine, Friedrich-Schiller-University Jena, Germany
| | - Bjoern Goebel
- First Department of Medicine, Friedrich-Schiller-University Jena, Germany
| | - Marcus Franz
- First Department of Medicine, Friedrich-Schiller-University Jena, Germany
| | - Dagmar Barz
- Division of Cardiology, Institute for Transfusion Medicine, Friedrich-Schiller-University Jena, Germany
| | - Markus Ferrari
- First Department of Medicine, Friedrich-Schiller-University Jena, Germany
| | - Hans R Figulla
- First Department of Medicine, Friedrich-Schiller-University Jena, Germany
| |
Collapse
|
43
|
Abstract
Modulation of the RAS (renin–angiotensin system), in particular of the function of the hormones AngII (angiotensin II) and Ang-(1–7) [angiotensin-(1–7)], is an important target for pharmacotherapy in the cardiovascular system. In the classical view, such modulation affects cardiovascular cells to decrease hypertrophy, fibrosis and endothelial dysfunction, and improves diuresis. In this view, excessive stimulation of AT1 receptors (AngII type 1 receptors) fulfils a detrimental role, as it promotes cardiovascular pathogenesis, and this is opposed by stimulation of the AT2 receptor (angiotensin II type 2 receptor) and the Ang-(1–7) receptor encoded by the Mas proto-oncogene. In recent years, this view has been broadened with the observation that the RAS regulates bone marrow stromal cells and stem cells, thus involving haematopoiesis and tissue regeneration by progenitor cells. This change of paradigm has enlarged the field of perspectives for therapeutic application of existing as well as newly developed medicines that alter angiotensin signalling, which now stretches beyond cardiovascular therapy. In the present article, we review the role of AngII and Ang-(1–7) and their respective receptors in haematopoietic and mesenchymal stem cells, and discuss possible pharmacotherapeutical implications.
Collapse
|
44
|
Roks AJM, Rodgers K, Walther T. Effects of the renin angiotensin system on vasculogenesis-related progenitor cells. Curr Opin Pharmacol 2011; 11:162-74. [PMID: 21296616 DOI: 10.1016/j.coph.2011.01.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Revised: 01/12/2011] [Accepted: 01/13/2011] [Indexed: 11/27/2022]
Abstract
The current concept is that there are both cells that integrate into the vasculature, true endothelial progenitor cells (EPC), and cells with hematopoietic markers that support neovascularisation. As identification of the EPC is controversial and studies refer cells that might fall into either pools, we will use the term, vasculogenesis-related progenitor cells (VRPC), for this review. VRPC are considered to be an important target for the treatment of cardiovascular diseases (CVD). Angiotensin II is known to be an important player in neovascularisation and the modulation of renin angiotensin system (RAS) is one of the major pharmacotherapeutic strategies for the treatment of CVD. We will review the effects of different components of the RAS on such VRPC under physiological conditions and in CVD. The reviewed research strongly supports a critical role of the RAS in vasculogenesis and vascular regeneration. Therefore, pharmacological intervention on the components of the RAS does not only target directly end-organ remodelling and blood pressure but also influence tissue healing and/or regeneration by influencing specific progenitor cells. Thus, the interrogation of RAS effects on VRPC will be important in the optimisation of RAS intervention or regenerative therapy.
Collapse
Affiliation(s)
- Anton J M Roks
- Department of Internal Medicine, Section of Pharmacology, Vascular and Metabolic Disease, Erasmus Medical Centre, Rotterdam, The Netherlands
| | | | | |
Collapse
|
45
|
Marketou ME, Kontaraki JE, Tsakountakis NA, Zacharis EA, Kochiadakis GE, Arfanakis DA, Parthenakis F, Chlouverakis G, Vardas PE. Differential effect of telmisartan and amlodipine on monocyte chemoattractant protein-1 and peroxisome proliferator-activated receptor-gamma gene expression in peripheral monocytes in patients with essential hypertension. Am J Cardiol 2011; 107:59-63. [PMID: 21146687 DOI: 10.1016/j.amjcard.2010.08.048] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Revised: 08/24/2010] [Accepted: 08/24/2010] [Indexed: 10/18/2022]
Abstract
Monocyte chemoattractant protein-1 (MCP-1) and peroxisome proliferator-activated receptor-γ (PPAR-γ) play a significant role in monocyte activation, vascular inflammation, and atherogenesis. Angiotensin receptor blockers and calcium channel blockers are antihypertensive drugs with established efficacy and a favorable safety profile. We investigated the effect of telmisartan--an angiotensin receptor blocker with PPAR-γ agonist activity--and amlodipine on the activation state of peripheral blood monocytes with respect to MCP-1 and PPAR-γ gene expression in hypertensives. We recruited 31 previously untreated patients with essential hypertension who were randomly assigned to receive treatment with telmisartan (n = 16) or amlodipine (n = 15). Blood samples were taken before and 3 months after therapy initiation. Mononuclear cells were isolated and mRNAs of MCP-1 and PPAR-γ were estimated by real-time quantitative reverse transcription-polymerase chain reaction each time. The 2 treatments decreased all blood pressure components significantly (p <0.001). In contrast, in the amlodipine group, MCP-1 gene expression was significantly downregulated after treatment with telmisartan (from 21.4 ± 20.5 to 8.1 ± 6.5, p = 0.009), whereas the amlodipine group did not show any significant change (12.5 ± 8.5 vs 17.6 ± 16.4, p = NS). In addition, PPAR-γ mRNA levels showed a significant increase in telmisartan-treated patients (from 20 ± 18.5 to 42.6 ± 36, p = 0.006) and no significant alterations in the amlodipine group (from 29.6 ± 42.5 to 24.2 ± 27.7, p = NS). In conclusion, treatment with telmisartan results in a significant attenuation of MCP-1 gene expression and an increase of PPAR-γ gene expression in peripheral monocytes in patients with essential hypertension. Our findings may provide new insights into the cardiovascular protection of telmisartan in hypertensives.
Collapse
|
46
|
Everaert BR, Van Craenenbroeck EM, Hoymans VY, Haine SE, Van Nassauw L, Conraads VM, Timmermans JP, Vrints CJ. Current perspective of pathophysiological and interventional effects on endothelial progenitor cell biology: focus on PI3K/AKT/eNOS pathway. Int J Cardiol 2010; 144:350-66. [PMID: 20444511 DOI: 10.1016/j.ijcard.2010.04.018] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2010] [Accepted: 04/04/2010] [Indexed: 12/24/2022]
Abstract
For more than a decade, endothelial progenitor cells (EPCs) have been implicated in cardiovascular homeostasis. EPCs are believed to reside within the bone marrow in close contact with surrounding stromal cells, and, under stimulation of pro-inflammatory cytokines, EPCs are mobilized out of the bone marrow. Hereafter circulating EPCs home to peripheral tissues, undergoing further proliferation and differentiation. Under certain pathophysiologic conditions this process seems to be blunted, resulting in a reduced capacity of EPCs to engage in vasculogenesis at sites of endothelial injury or tissue ischemia. In this review, we focus on the effects of traditional cardiovascular risk factors on EPC biology and we explore whether pharmacological, dietary and lifestyle interventions can favorably restore EPC mobilization, differentiation, homing and angiogenic properties. Because the PI3K/Akt/eNOS pathway plays a pivotal role in the process of EPC mobilization, migration and homing, we specifically emphasize the involvement of PI3K, Akt and eNOS in EPC biology under these different (patho)physiologic conditions. (Pre)clinically used drugs or lifestyle interventions that have been shown to ameliorate EPC biology are reviewed. These treatment strategies remain attractive targets to restore the regenerative capacity of EPCs in cardiovascular diseases.
Collapse
Affiliation(s)
- Bert R Everaert
- Laboratory of Cell Biology and Histology, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | | | |
Collapse
|