1
|
Huang S, Xu M, Li M, Cheng J, Wu Y. The Expression of Circ-Astn1 Inhibits High Glucose Induced Endothelial Progenitor Cell Dysfunction by Activating Autophagy. Endocr Res 2024; 49:213-222. [PMID: 38867680 DOI: 10.1080/07435800.2024.2365887] [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: 02/10/2024] [Revised: 05/06/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Diabetes mellitus (DM) and complications such as chronic kidney disease and cardiovascular symptoms pose a substantial public health burden. Increasing studies have shown that circular RNAs (circRNAs) regulate many gene expressions that are essential in diverse pathological and biological procedures. However, the roles of particular circRNAs in DM are unclear. METHODS In the current investigation, endothelial progenitor cells (EPCs) were used to search for abnormal expression of circRNAs by using high-throughput sequencing under high glucose (HG) conditions. The regulatory mechanisms and targets were then studied through bioinformatics analysis, luciferase reporter analysis, angiogenic differentiation experiments, flow cytometry detection of apoptosis and RT-qPCR analysis. RESULTS The circ-Astn1 expression in EPCs decreased after HG treatment. Overexpression or circ-Astn1 suppressed HG induced endothelial cell damage. MicroRNA (miR)-138-5p and SIRT5 were found to be the downstream targets of circ-Astn1 through luciferase reporter analysis. SIRT5 downregulation or miR-138-5p overexpression reversed circ-Astn1's protective effect against HG induced endothelial cell dysfunction, including apoptosis and abnormal vascular differentiation. Furthermore, circ-Astn1 overexpression promoted autophagy activation by increasing SIRT5 expression under HG conditions. Our findings suggest that circ-Astn1 mediated promotion of SIRT5 facilitates autophagy by sponging miR-138-5p. CONLUSION Together, our findings show that the overexpression of circ-Astn1 suppresses HG induced endothelial cell damage by targeting miR-138-5p/SIRT5 axis.
Collapse
Affiliation(s)
- Shiying Huang
- Department of Interventional & Vascular Surgery, Tenth People's Hospital of Tongji University, Shanghai, China
- Department of Pharmacy, Shanghai Children's Medical Center, Shanghai Jiao Tong University, Shanghai, China
| | - Minjie Xu
- Department of Interventional & Vascular Surgery, Tenth People's Hospital of Tongji University, Shanghai, China
- Institute of Interventional & Vascular Surgery, Tongji University, Shanghai, China
| | - Maoquan Li
- Department of Interventional & Vascular Surgery, Tenth People's Hospital of Tongji University, Shanghai, China
- Institute of Interventional & Vascular Surgery, Tongji University, Shanghai, China
| | - Jie Cheng
- Department of Interventional & Vascular Surgery, Tenth People's Hospital of Tongji University, Shanghai, China
- Institute of Interventional & Vascular Surgery, Tongji University, Shanghai, China
| | - Yongfa Wu
- Department of Interventional & Vascular Surgery, Tenth People's Hospital of Tongji University, Shanghai, China
- Institute of Interventional & Vascular Surgery, Tongji University, Shanghai, China
| |
Collapse
|
2
|
Lee M, Tariq AR, Kim M. Gemigliptin, a potent selective dipeptidyl peptidase 4 inhibitor, protects endothelial progenitor cells by oxidative stress via caspase-3 dependent pathway. Biochem Biophys Rep 2024; 38:101673. [PMID: 38444735 PMCID: PMC10914559 DOI: 10.1016/j.bbrep.2024.101673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 03/07/2024] Open
Abstract
Endothelial progenitor cells (EPCs) are exclusive players in vasculogenesis and endothelial regeneration. EPCs are of two types and their differentiation is mediated by different growth factors. A decrease in EPC number and function causes cardiovascular abnormalities and reduced angiogenesis. Various studies has documented a role of EPCs in diabetes. EPCs treatment with different drugs improve insulin secretion but causes other abnormalities. In vivo and in vitro studies have reported anti glycation effect of gemigliptin but no data is available on in vitro effect of gemigliptin on EPC number and functional credibility. The current study was aimed to find an in vitro effect of gemigliptin on EPC number and function along with an effective treatment dose of gemigliptin. EPCs were isolated, cultured and phenotypically characterized using Dil- AcLDL and ulex-lectin fluorescence staining. EPCs were then treated with different doses of Zemiglo and their viability analyzed with viability assay using water-soluble tetrazolium salt (WST-1), by Annexin V and Propidium Iodide (PI) staining, senescence-associated beta-galactosidase (SA-β-gal) staining, western blot and Flow cytometric analysis of apoptotic signals. The results demonstrated that the isolated EPCs has typical endothelial phenotypes. And these EPCs were of two types based on morphology i.e., early and late EPCs. Gemigliptin dose dependently improved the EPCs morphology and increased EPCs viability, the most effective dose being the 20 μM. Gemigliptin at 10 μM, 20 μM and 50 μM significantly increased the BCL-2 levels and at 20 μM significantly decreased the Caspase-3 levels in EPCs. In conclusion, gemigliptin dose dependently effects the EPCs viability and morphology through Caspase-3 signaling. Our results are the first report of gemigliptin effect on EPC viability and morphology.
Collapse
Affiliation(s)
- Mijung Lee
- Neurology, Center for Medical Innovation, Seoul National University Hospital, Seoul, South Korea
| | - Amna Rashid Tariq
- Neurology, Center for Medical Innovation, Seoul National University Hospital, Seoul, South Korea
| | - Manho Kim
- Neurology, Seoul National University Hospital, Neuroscience and Dementia Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
3
|
Xiujin Z, Lili G, Jing F, Wenhai Y, Sikai L, Wan-Yin S. HOXD9 regulated mitophagy to promote endothelial progenitor cells angiogenesis and deep vein thrombosis recanalization and resolution. Mol Med 2024; 30:84. [PMID: 38867168 PMCID: PMC11167931 DOI: 10.1186/s10020-024-00852-5] [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: 12/26/2023] [Accepted: 05/31/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Deep vein thrombosis (DVT) is a common vascular surgical disease caused by the coagulation of blood in the deep veins, and predominantly occur in the lower limbs. Endothelial progenitor cells (EPCs) are multi-functional stem cells, which are precursors of vascular endothelial cells. EPCs have gradually evolved into a promising treatment strategy for promoting deep vein thrombus dissolution and recanalization through the stimulation of various physical and chemical factors. METHODS In this study, we utilized a mouse DVT model and performed several experiments including qRT-PCR, Western blot, tube formation, wound healing, Transwell assay, immunofluorescence, flow cytometry analysis, and immunoprecipitation to investigate the role of HOXD9 in the function of EPCs cells. The therapeutic effect of EPCs overexpressing HOXD9 on the DVT model and its mechanism were also explored. RESULTS Overexpression of HOXD9 significantly enhanced the angiogenesis and migration abilities of EPCs, while inhibiting cell apoptosis. Additionally, results indicated that HOXD9 specifically targeted the HRD1 promoter region and regulated the downstream PINK1-mediated mitophagy. Interestingly, intravenous injection of EPCs overexpressing HOXD9 into mice promoted thrombus dissolution and recanalization, significantly decreasing venous thrombosis. CONCLUSIONS The findings of this study reveal that HOXD9 plays a pivotal role in stimulating vascular formation in endothelial progenitor cells, indicating its potential as a therapeutic target for DVT management.
Collapse
Affiliation(s)
- Zhang Xiujin
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Guo Lili
- Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Fan Jing
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Ye Wenhai
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Liu Sikai
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Shi Wan-Yin
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
4
|
Altabas V, Marinković Radošević J, Špoljarec L, Uremović S, Bulum T. The Impact of Modern Anti-Diabetic Treatment on Endothelial Progenitor Cells. Biomedicines 2023; 11:3051. [PMID: 38002051 PMCID: PMC10669792 DOI: 10.3390/biomedicines11113051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Diabetes is one of the leading chronic diseases globally with a significant impact on mortality. This condition is associated with chronic microvascular and macrovascular complications caused by vascular damage. Recently, endothelial progenitor cells (EPCs) raised interest due to their regenerative properties. EPCs are mononuclear cells that are derived from different tissues. Circulating EPCs contribute to regenerating the vessel's intima and restoring vascular function. The ability of EPCs to repair vascular damage depends on their number and functionality. Diabetic patients have a decreased circulating EPC count and impaired EPC function. This may at least partially explain the increased risk of diabetic complications, including the increased cardiovascular risk in these patients. Recent studies have confirmed that many currently available drugs with proven cardiovascular benefits have beneficial effects on EPC count and function. Among these drugs are also medications used to treat different types of diabetes. This manuscript aims to critically review currently available evidence about the ways anti-diabetic treatment affects EPC biology and to provide a broader context considering cardiovascular complications. The therapies that will be discussed include lifestyle adjustments, metformin, sulphonylureas, gut glucosidase inhibitors, thiazolidinediones, dipeptidyl peptidase 4 inhibitors, glucagon-like peptide 1 receptor analogs, sodium-glucose transporter 2 inhibitors, and insulin.
Collapse
Affiliation(s)
- Velimir Altabas
- Department of Endocrinology, Diabetes and Metabolic Diseases, Sestre Milosrdnice University Clinical Hospital, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Jelena Marinković Radošević
- Department of Endocrinology, Diabetes and Metabolic Diseases, Sestre Milosrdnice University Clinical Hospital, 10000 Zagreb, Croatia
| | - Lucija Špoljarec
- Department of Endocrinology, Diabetes and Metabolic Diseases, Sestre Milosrdnice University Clinical Hospital, 10000 Zagreb, Croatia
| | | | - Tomislav Bulum
- Department of Endocrinology, Diabetes and Metabolic Diseases, Sestre Milosrdnice University Clinical Hospital, 10000 Zagreb, Croatia
- Vuk Vrhovac University Clinic for Diabetes, Endocrinology and Metabolic Diseases, Merkur University Hospital, 10000 Zagreb, Croatia
| |
Collapse
|
5
|
Akashi N, Umemoto T, Yamada H, Fujiwara T, Yamamoto K, Taniguchi Y, Sakakura K, Wada H, Momomura SI, Fujita H. Teneligliptin, a DPP-4 Inhibitor, Improves Vascular Endothelial Function via Divergent Actions Including Changes in Circulating Endothelial Progenitor Cells. Diabetes Metab Syndr Obes 2023; 16:1043-1054. [PMID: 37077576 PMCID: PMC10108873 DOI: 10.2147/dmso.s403125] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/03/2023] [Indexed: 04/20/2023] Open
Abstract
PURPOSE Dipeptidyl peptidase-4 (DPP-4) inhibitors increase endothelial progenitor cells (EPCs) in peripheral blood circulation. However, the underlying mechanisms and effects on vascular endothelial function remain unclear. We evaluated whether the DPP-4 inhibitor teneligliptin increases circulating EPCs by inhibiting stromal-derived factor-1α (SDF-1α) and improves flow-mediated vascular dilatation (FMD) in type 2 diabetes mellitus patients with acute coronary syndrome (ACS) or its risk factors. PATIENTS AND METHODS This single-center, open-label, prospective, randomized controlled trial evaluated 17 patients (hemoglobin A1c ≤7.5% and peak creatinine phosphokinase <2000 IU/mL) with ACS or a history of ACS or multiple cardiovascular risk factors. Metabolic variables of glucose and lipids, circulating EPCs, plasma DPP-4 activity, and SDF-1α levels, and FMD were evaluated at baseline and 28 ± 4 weeks after enrollment. Patients were randomly assigned to either the teneligliptin (n = 8) or control (n = 9) groups. RESULTS The DPP-4 activity (∆-509.5 ± 105.7 vs ∆32.8 ± 53.4 μU/mL) and SDF-1α levels (∆-695.6 ± 443.2 vs ∆11.1 ± 193.7 pg/mL) were significantly decreased after 28 weeks in the teneligliptin group than those in the control group. The number of EPCs showed an increasing trend in the teneligliptin treated group; albeit this did not reach statistical significance. Glucose and lipid levels were not significantly different between the groups before and after 28 weeks. However, FMD was significantly improved in the teneligliptin group when compared to the control group (∆3.8% ± 2.1% vs ∆-0.3% ± 2.9%, P=0.006). CONCLUSION Teneligliptin improved FMD through a mechanism other than increasing the number of circulating EPCs.
Collapse
Affiliation(s)
- Naoyuki Akashi
- Division of Cardiovascular Medicine, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Tomio Umemoto
- Division of Cardiovascular Medicine, Jichi Medical University Saitama Medical Center, Saitama, Japan
- Correspondence: Tomio Umemoto, Division of Cardiovascular Medicine, Jichi Medical University Saitama Medical Center, 1-847 Amanuma-cho, Omiya-ku, Saitama, 330-8503, Japan, Tel +81-48-647-2111, Fax +81-48-648-5188, Email
| | - Hodaka Yamada
- Division of Endocrinology and Metabolism, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Takayuki Fujiwara
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Kei Yamamoto
- Division of Cardiovascular Medicine, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Yousuke Taniguchi
- Division of Cardiovascular Medicine, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Kenichi Sakakura
- Division of Cardiovascular Medicine, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Hiroshi Wada
- Division of Cardiovascular Medicine, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Shin-ichi Momomura
- Division of Cardiovascular Medicine, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Hideo Fujita
- Division of Cardiovascular Medicine, Jichi Medical University Saitama Medical Center, Saitama, Japan
| |
Collapse
|
6
|
Changes in Arterial Stiffness in Response to Various Types of Exercise Modalities: A Narrative Review on Physiological and Endothelial Senescence Perspectives. Cells 2022; 11:cells11223544. [PMID: 36428973 PMCID: PMC9688701 DOI: 10.3390/cells11223544] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/12/2022] Open
Abstract
Arterial stiffness is a reliable independent predictor of cardiovascular events. Exercise training might enhance arterial compliance through improved metabolic health status. Different modes of exercise may have different effects on arterial stiffness. However, the interactions among different modes of exercise on endothelial senescence, the development of arterial vascular stiffness, and the associated molecular mechanisms are not completely understood. In this narrative review, we evaluate the current evidence focusing on the effects of various exercise modes on arterial stiffness and vascular health, and the known underlying physiological mechanisms are discussed as well. Here, we discuss the most recent evidence of aerobic exercise, high-intensity interval training (HIIT), and resistance exercise (RE) on arterial stiffness and endothelial senescence in physiological and cellular studies. Indeed, aerobic, HIIT, and progression RE-induced arterial compliance may reduce arterial stiffness by effectively promoting nitric oxide (NO) bioavailability and reducing endothelial senescence. However, the transient increase in inflammation and sympathetic activation may contribute to the temporary elevation in arterial stiffness following whole-body high-intensity acute resistance exercise.
Collapse
|
7
|
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
|
8
|
Endothelial progenitor cells predict vascular damage progression in naive hypertensive patients according to sex. Hypertens Res 2021; 44:1451-1461. [PMID: 34471254 DOI: 10.1038/s41440-021-00716-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/16/2021] [Accepted: 07/11/2021] [Indexed: 02/07/2023]
Abstract
Low levels of endothelial progenitor cells (EPCs) are associated with cardiovascular (CV) morbidity and mortality. Early indicators of vascular damage represent independent predictors of CV prognosis. The aim of this study was to evaluate the possible association of EPCs and circulating cytokine levels with vascular damage markers in naive hypertensive patients according to sex and to evaluate the role of EPCs in vascular damage progression. We enrolled 60 subjects; circulating EPCs were determined by cytometric analysis, and serum cytokines were determined by chemiluminescence microarray technology. Endothelial function was estimated with the measurement of the reactive hyperemia index (RHI), arterial stiffness (AS) was evaluated with the measurement of carotid-femoral pulse wave velocity (PWV) and carotid intima-media thickness (IMT) was determined by a high-resolution ultrasound B-mode system. Patients were evaluated at baseline and after an average follow-up of 3.0 ± 0.6 years. RHI was correlated with EPCs and inversely related to HOMA, TNF-α, IL-6, hs-CRP, and IL-1β. PWV was positively correlated with HOMA, TNF-α, IL-6, IL-1β, and hs-CRP, and it was inversely related to EPCs. An inverse relationship was observed between c-IMT and EPCs and e-GFR. EPCs were the major predictor of the RHI and PWV. After adjustment for vascular index basal values and the other covariates, EPCs explained 17.0%, 27.7%, and 10.6% of the variability in ΔRHI, ΔPWV, and Δc-IMT at follow-up, respectively. Our study results support the hypothesis of an etiological link between circulating EPCs and morphofunctional vascular parameters in hypertensive subjects. Of interest, circulating EPCs, after adjusting for possible confounding factors, may indicate vascular damage progression.
Collapse
|
9
|
Wu W, Zhang J, Shao L, Huang H, Meng Q, Shen Z, Teng X. Evaluation of Circulating Endothelial Progenitor Cells in Abdominal Aortic Aneurysms after Endovascular Aneurysm Repair. Int J Stem Cells 2021; 15:136-143. [PMID: 34711694 PMCID: PMC9148833 DOI: 10.15283/ijsc21027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 06/11/2021] [Accepted: 08/02/2021] [Indexed: 11/10/2022] Open
Abstract
Background and Objectives Circulating endothelial progenitor cells (EPCs) participate in vascular repair and predict cardiovascular outcomes. The aim of this study was to investigate the correlation between EPCs and abdominal aortic aneurysms (AAAs). Methods and Results Patients (age 67±9.41 years) suffering from AAAs (aortic diameters 58.09±11.24 mm) were prospectively enrolled in this study. All patients received endovascular aneurysm repair (EVAR). Blood samples were taken preoperatively and 14 days after surgery from patients with aortic aneurysms. Samples were also obtained from age-matched control subjects. Circulating EPCs were defined as those cells that were double positive for CD34 and CD309. Rat models of AAA formation were generated by the peri-adventitial elastase application of either saline solution (control; n=10), or porcine pancreatic elastase (PPE; n=14). The aortas were analyzed using an ultrasonic video system and immunohistochemistry. The levels of CD34+/CD309+ cells in the peripheral blood mononuclear cell populations were measured by flow cytometry. The baseline numbers of circulating EPCs (CD34+/CD309+) in the peripheral blood were significantly smaller in AAA patients compared with control subjects. The number of EPCs doubled by the 14th day after EVAR. A total of 78.57% of rats in the PPE group (11/14) formed AAAs (dilation ratio >150%). The numbers of EPCs from defined AAA rats were significantly decreased compared with the control group. Conclusions EPC levels may be useful for monitoring abdominal aorta aneurysms and rise after EVAR in patients with aortic aneurysms, and might contribute to the rapid endothelialization of vessels.
Collapse
Affiliation(s)
- Weihua Wu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China.,Center of Clinical Laboratory, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jinlong Zhang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Lianbo Shao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Haoyue Huang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Qingyou Meng
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Xiaomei Teng
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| |
Collapse
|
10
|
Poniedziałek-Czajkowska E, Mierzyński R, Dłuski D, Leszczyńska-Gorzelak B. Prevention of Hypertensive Disorders of Pregnancy-Is There a Place for Metformin? J Clin Med 2021; 10:jcm10132805. [PMID: 34202343 PMCID: PMC8268471 DOI: 10.3390/jcm10132805] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/20/2021] [Accepted: 06/23/2021] [Indexed: 12/13/2022] Open
Abstract
The possibility of prophylaxis of hypertensive disorders of pregnancy (HDPs) such as preeclampsia (PE) and pregnancy-induced hypertension is of interest due to the unpredictable course of these diseases and the risks they carry for both mother and fetus. It has been proven that their development is associated with the presence of the placenta, and the processes that initiate it begin at the time of the abnormal invasion of the trophoblast in early pregnancy. The ideal HDP prophylaxis should alleviate the influence of risk factors and, at the same time, promote physiological trophoblast invasion and maintain the physiologic endothelium function without any harm to both mother and fetus. So far, aspirin is the only effective and recommended pharmacological agent for the prevention of HDPs in high-risk groups. Metformin is a hypoglycemic drug with a proven protective effect on the cardiovascular system. Respecting the anti-inflammatory properties of metformin and its favorable impact on the endothelium, it seems to be an interesting option for HDP prophylaxis. The results of previous studies on such use of metformin are ambiguous, although they indicate that in a certain group of pregnant women, it might be effective in preventing hypertensive complications. The aim of this study is to present the possibility of metformin in the prevention of hypertensive disorders of pregnancy with respect to its impact on the pathogenic elements of development
Collapse
|
11
|
Endothelial Progenitor Cells Dysfunctions and Cardiometabolic Disorders: From Mechanisms to Therapeutic Approaches. Int J Mol Sci 2021; 22:ijms22136667. [PMID: 34206404 PMCID: PMC8267891 DOI: 10.3390/ijms22136667] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/10/2021] [Accepted: 06/17/2021] [Indexed: 12/12/2022] Open
Abstract
Metabolic syndrome (MetS) is a cluster of several disorders, such as hypertension, central obesity, dyslipidemia, hyperglycemia, insulin resistance and non-alcoholic fatty liver disease. Despite health policies based on the promotion of physical exercise, the reduction of calorie intake and the consumption of healthy food, there is still a global rise in the incidence and prevalence of MetS in the world. This phenomenon can partly be explained by the fact that adverse events in the perinatal period can increase the susceptibility to develop cardiometabolic diseases in adulthood. Individuals born after intrauterine growth restriction (IUGR) are particularly at risk of developing cardiovascular diseases (CVD) and metabolic disorders later in life. It has been shown that alterations in the structural and functional integrity of the endothelium can lead to the development of cardiometabolic diseases. The endothelial progenitor cells (EPCs) are circulating components of the endothelium playing a major role in vascular homeostasis. An association has been found between the maintenance of endothelial structure and function by EPCs and their ability to differentiate and repair damaged endothelial tissue. In this narrative review, we explore the alterations of EPCs observed in individuals with cardiometabolic disorders, describe some mechanisms related to such dysfunction and propose some therapeutical approaches to reverse the EPCs dysfunction.
Collapse
|
12
|
Singh P, O'Toole TE, Conklin DJ, Hill BG, Haberzettl P. Endothelial progenitor cells as critical mediators of environmental air pollution-induced cardiovascular toxicity. Am J Physiol Heart Circ Physiol 2021; 320:H1440-H1455. [PMID: 33606580 PMCID: PMC8260385 DOI: 10.1152/ajpheart.00804.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/26/2021] [Accepted: 02/14/2021] [Indexed: 01/15/2023]
Abstract
Environmental air pollution exposure is a leading cause of death worldwide, and with increasing industrialization and urbanization, its disease burden is expected to rise even further. The majority of air pollution exposure-associated deaths are linked to cardiovascular disease (CVD). Although ample research demonstrates a strong correlation between air pollution exposure and CVD risk, the mechanisms by which inhalation of polluted air affects cardiovascular health are not completely understood. Inhalation of environmental air pollution has been associated with endothelial dysfunction, which suggests that air pollution exposure impacts CVD health by inducing endothelial injury. Interestingly, recent studies demonstrate that air pollution exposure affects the number and function of endothelial progenitor cells (EPCs), subpopulations of bone marrow-derived proangiogenic cells that have been shown to play an essential role in maintaining cardiovascular health. In line with their beneficial function, chronically low levels of circulating EPCs and EPC dysfunction (e.g., in diabetic patients) have been associated with vascular dysfunction, poor cardiovascular health, and increases in the severity of cardiovascular outcomes. In contrast, treatments that improve EPC number and function (e.g., exercise) have been found to attenuate cardiovascular dysfunction. Considering the critical, nonredundant role of EPCs in maintaining vascular health, air pollution exposure-induced impairments in EPC number and function could lead to endothelial dysfunction, consequently increasing the risk for CVD. This review article covers novel aspects and new mechanistic insights of the adverse effects of air pollution exposure on cardiovascular health associated with changes in EPC number and function.
Collapse
Affiliation(s)
- Parul Singh
- Division of Environmental Medicine, Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Timothy E O'Toole
- Division of Environmental Medicine, Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Daniel J Conklin
- Division of Environmental Medicine, Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Bradford G Hill
- Division of Environmental Medicine, Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Petra Haberzettl
- Division of Environmental Medicine, Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
13
|
Cao F, Wu K, Zhu YZ, Bao ZW. Roles and Mechanisms of Dipeptidyl Peptidase 4 Inhibitors in Vascular Aging. Front Endocrinol (Lausanne) 2021; 12:731273. [PMID: 34489872 PMCID: PMC8416540 DOI: 10.3389/fendo.2021.731273] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 07/21/2021] [Indexed: 12/22/2022] Open
Abstract
Vascular aging is characterized by alterations in the constitutive properties and biological functions of the blood vessel wall. Endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) are indispensability elements in the inner layer and the medial layer of the blood vessel wall, respectively. Dipeptidyl peptidase-4 (DPP4) inhibitors, as a hypoglycemic agent, play a protective role in reversing vascular aging regardless of their effects in meliorating glycemic control in humans and animal models of type 2 diabetes mellitus (T2DM) through complex cellular mechanisms, including improving EC dysfunction, promoting EC proliferation and migration, alleviating EC senescence, obstructing EC apoptosis, suppressing the proliferation and migration of VSMCs, increasing circulating endothelial progenitor cell (EPC) levels, and preventing the infiltration of mononuclear macrophages. All of these showed that DPP4 inhibitors may exert a positive effect against vascular aging, thereby preventing vascular aging-related diseases. In the current review, we will summarize the cellular mechanism of DPP4 inhibitors regulating vascular aging; moreover, we also intend to compile the roles and the promising therapeutic application of DPP4 inhibitors in vascular aging-related diseases.
Collapse
Affiliation(s)
- Fen Cao
- Department of Cardiology, Huaihua First People’s Hospital, Huaihua, China
| | - Kun Wu
- Department of Neurology, Huaihua First People’s Hospital, Huaihua, China
| | - Yong-Zhi Zhu
- Department of Cardiology, Huaihua First People’s Hospital, Huaihua, China
| | - Zhong-Wu Bao
- Department of Cardiology, Huaihua First People’s Hospital, Huaihua, China
- *Correspondence: Zhong-Wu Bao,
| |
Collapse
|
14
|
Zhang Y, Liu X, Yang L, Zou L. Current Researches, Rationale, Plausibility, and Evidence Gaps on Metformin for the Management of Hypertensive Disorders of Pregnancy. Front Pharmacol 2020; 11:596145. [PMID: 33381040 PMCID: PMC7768035 DOI: 10.3389/fphar.2020.596145] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/22/2020] [Indexed: 12/16/2022] Open
Abstract
Hypertensive disorders of pregnancy (HDP) are a group of morbid pregnancy complications, with preeclampsia (PE) being the most common subclassification among them. PE affects 2%–8% of pregnancies globally and threatens maternal and fetal health seriously. However, the only effective treatment of PE to date is the timely termination of pregnancy, albeit with increased perinatal risks. Hence, more emerging therapies for PE management are in urgent need. Originally introduced as the first-line therapy for type 2 diabetes mellitus, metformin (MET) has now been found in clinical trials to significantly reduce the incidence of gestational hypertension and PE in pregnant women with PE-related risks, including but not limited to pregestational diabetes mellitus, gestational diabetes mellitus, polycystic ovary syndrome, or obesity. Additionally, existing clinical data have preliminarily ensured the safety of taking MET during human pregnancies. Relevant lab studies have indicated that the underlying mechanism includes angiogenesis promotion, endothelial protection, anti-inflammatory effects, and particularly protective effects on trophoblast cells against the risk factors, which are beneficial to placental development. Together with its global availability, easy administration, and low cost, MET is expected to be a promising option for the prevention and treatment of PE. Nevertheless, there are still some limitations in current studies, and the design of the relevant research scheme is supposed to be further improved in the future. Herein, we summarize the relevant clinical and experimental researches to discuss the rationale, safety, and feasibility of MET for the management of HDP. At the end of the article, gaps in current researches are proposed. Concretely, experimental MET concentration and PE models should be chosen cautiously. Besides, the clinical trial protocol should be further optimized to evaluate the reduction in the prevalence of PE as a primary endpoint. All of those evidence gaps may be of guiding significance to improve the design of relevant experiments and clinical trials in the future.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxia Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liu Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Zou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Vinci MC, Gambini E, Bassetti B, Genovese S, Pompilio G. When Good Guys Turn Bad: Bone Marrow's and Hematopoietic Stem Cells' Role in the Pathobiology of Diabetic Complications. Int J Mol Sci 2020; 21:ijms21113864. [PMID: 32485847 PMCID: PMC7312629 DOI: 10.3390/ijms21113864] [Citation(s) in RCA: 15] [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: 04/12/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 12/15/2022] Open
Abstract
Diabetes strongly contributes to the development of cardiovascular disease, the leading cause of mortality and morbidity in these patients. It is widely accepted that hyperglycemia impairs hematopoietic stem/progenitor cell (HSPC) mobilization from the bone marrow (BM) by inducing stem cell niche dysfunction. Moreover, a recent study demonstrated that type 2 diabetic patients are characterized by significant depletion of circulating provascular progenitor cells and increased frequency of inflammatory cells. This unbalance, potentially responsible for the reduction of intrinsic vascular homeostatic capacity and for the establishment of a low-grade inflammatory status, suggests that bone BM-derived HSPCs are not only victims but also active perpetrators in diabetic complications. In this review, we will discuss the most recent literature on the molecular mechanisms underpinning hyperglycemia-mediated BM dysfunction and differentiation abnormality of HSPCs. Moreover, a section will be dedicated to the new glucose-lowering therapies that by specifically targeting the culprits may prevent or treat diabetic complications.
Collapse
Affiliation(s)
- Maria Cristina Vinci
- Unit of Vascular Biology and Regenerative Medicine, IRCCS Centro Cardiologico Monzino, I-20138- Milan, Italy; (E.G.); (B.B.); (G.P.)
- Correspondence: ; Tel.: +39-02-5800-2028
| | - Elisa Gambini
- Unit of Vascular Biology and Regenerative Medicine, IRCCS Centro Cardiologico Monzino, I-20138- Milan, Italy; (E.G.); (B.B.); (G.P.)
| | - Beatrice Bassetti
- Unit of Vascular Biology and Regenerative Medicine, IRCCS Centro Cardiologico Monzino, I-20138- Milan, Italy; (E.G.); (B.B.); (G.P.)
| | - Stefano Genovese
- Unit of Diabetes, Endocrine and Metabolic Diseases, IRCCS Centro Cardiologico Monzino, I-20138- Milan, Italy;
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, IRCCS Centro Cardiologico Monzino, I-20138- Milan, Italy; (E.G.); (B.B.); (G.P.)
| |
Collapse
|
16
|
Hu C, Sun Y, Yang X. Pioglitazone up-regulates MALAT1 and promotes the proliferation of endothelial progenitor cells through increasing c-Myc expression in type 2 diabetes mellitus. ACTA ACUST UNITED AC 2020. [DOI: 10.31491/apt.2020.03.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
17
|
Liu H, Guo L, Xing J, Li P, Sang H, Hu X, Du Y, Zhao L, Song R, Gu H. The protective role of DPP4 inhibitors in atherosclerosis. Eur J Pharmacol 2020; 875:173037. [PMID: 32097656 DOI: 10.1016/j.ejphar.2020.173037] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 01/30/2020] [Accepted: 02/21/2020] [Indexed: 12/16/2022]
Abstract
Diabetes is a chronic non-communicable disease whose incidence continues to grow rapidly, and it is one of the most serious and critical public health problems. Diabetes complications, especially atherosclerosis-related chronic vascular complications, are a serious threat to human life and health. Growing evidence suggests that dipeptidyl peptidase 4 (DPP4) inhibitors, beyond their role in improving glycemic control, are helpful in ameliorating endothelial dysfunction in humans and animal models of T2DM. In fact, DPP4 inhibitors have been shown by successive studies to play a protective effect against vascular complications. On one hand, in addition to their hypoglycemic effects, DPP4 inhibitors participate in the control of atherosclerotic risk factors by regulating blood lipids and lowering blood pressure. On the other hand, DPP4 inhibitors exert anti-atherosclerotic effects directly through multiple mechanisms, including improving endothelial cell dysfunction, increasing circulating endothelial progenitor cell (EPCs) levels, regulating mononuclear macrophages and smooth muscle cells, inhibiting inflammation and oxidative stress and improving plaque instability. Herein, we review the beneficial roles of DPP4 inhibitors in atherosclerosis as detailed.
Collapse
Affiliation(s)
- Hengdao Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Lingli Guo
- Department of General Medicine, The Third People's Provincial Hospital of Henan Province, Zhengzhou, 450000, Henan, China
| | - Junhui Xing
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Peicheng Li
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University. Xinxiang, Henan, 453100, China
| | - Haiqiang Sang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xiaofang Hu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders (Xiangya), Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yunpeng Du
- Department of Cardiology, Huixian People's Hospital, Xinxiang, Henan, 453600, China
| | - Liangping Zhao
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University. Xinxiang, Henan, 453100, China
| | - Ruipeng Song
- Department of Endocrinology, The Third People's Provincial Hospital of Henan Province, Zhengzhou, 450000, Henan, China.
| | - Heping Gu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| |
Collapse
|
18
|
Shen Z, Chen Q, Ying H, Ma Z, Bi X, Li X, Wang M, Jin C, Lai D, Zhao Y, Fu G. Identification of differentially expressed genes in the endothelial precursor cells of patients with type 2 diabetes mellitus by bioinformatics analysis. Exp Ther Med 2019; 19:499-510. [PMID: 31897097 PMCID: PMC6923743 DOI: 10.3892/etm.2019.8239] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 10/18/2019] [Indexed: 12/21/2022] Open
Abstract
Type 2 diabetes mellitus (DM) is a metabolic disease with worldwide prevalence that is associated with a decrease in the number and function of endothelial progenitor cells (EPCs). The aim of the present study was to explore the potential hub genes of EPCs in patients with type 2 DM. Differentially expressed genes (DEGs) were screened from a public microarray dataset (accession no. GSE43950). Pathway and functional enrichment analyses were performed using the Database for Annotation, Visualization and Integrated Discovery. The protein-protein interaction (PPI) network was visualized. The most significantly clustered modules and hub genes were identified using Cytoscape. Furthermore, hub genes were validated by quantitative PCR analysis of EPCs isolated from diabetic and normal subjects. Subsequently, weighted gene co-expression network analysis (WGCNA) was performed to identify the modules incorporating the genes exhibiting the most significant variance. A total of 970 DEGs were obtained and they were mainly accumulated in inflammation-associated pathways. A total of 9 hub genes were extracted from the PPI network and the highest differential expression was determined for the interleukin 8 (IL8) and CXC chemokine ligand 1 (CXCL1) genes. In the WGCNA performed to determine the modules associated with type 2 DM, one module incorporated IL8 and CXCL1. Finally, pathway enrichment of 10% genes in the pink module ordered by intramodular connectivity (IC) was associated with the IL17 and the chemokine signaling pathways. The present results revealed that the expression of IL8 and CXCL1 may serve important roles in the pathophysiology of EPCs during type 2 DM and inflammatory response may be critical for the reduced number and hypofunction of EPCs isolated from patients with diabetes.
Collapse
Affiliation(s)
- Zhida Shen
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | - Qi Chen
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | - Hangying Ying
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | - Zetao Ma
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | - Xukun Bi
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | - Xiaoting Li
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | - Meihui Wang
- Biomedical Research Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | - Chongying Jin
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | - Dongwu Lai
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | - Yanbo Zhao
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| |
Collapse
|
19
|
Loader J, Khouri C, Taylor F, Stewart S, Lorenzen C, Cracowski JL, Walther G, Roustit M. The continuums of impairment in vascular reactivity across the spectrum of cardiometabolic health: A systematic review and network meta-analysis. Obes Rev 2019; 20:906-920. [PMID: 30887713 DOI: 10.1111/obr.12831] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/03/2019] [Accepted: 01/03/2019] [Indexed: 12/12/2022]
Abstract
This study aimed to assess, for the first time, the change in vascular reactivity across the full spectrum of cardiometabolic health. Systematic searches were conducted in MEDLINE and EMBASE databases from their inception to March 13, 2017, including studies that assessed basal vascular reactivity in two or more of the following health groups (aged ≥18 years old): healthy, overweight, obesity, impaired glucose tolerance, metabolic syndrome, or type 2 diabetes with or without complications. Direct and indirect comparisons of vascular reactivity were combined using a network meta-analysis. Comparing data from 193 articles (7226 healthy subjects and 19344 patients), the network meta-analyses revealed a progressive impairment in vascular reactivity (flow-mediated dilation data) from the clinical onset of an overweight status (-0.41%, 95% CI, -0.98 to 0.15) through to the development of vascular complications in those with type 2 diabetes (-4.26%, 95% CI, -4.97 to -3.54). Meta-regressions revealed that for every 1 mmol/l increase in fasting blood glucose concentration, flow-mediated dilation decreased by 0.52%. Acknowledging that the time course of disease may vary between patients, this study demonstrates multiple continuums of vascular dysfunction where the severity of impairment in vascular reactivity progressively increases throughout the pathogenesis of obesity and/or insulin resistance, providing information that is important to enhancing the timing and effectiveness of strategies that aim to improve cardiovascular outcomes.
Collapse
Affiliation(s)
- Jordan Loader
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Australia
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia
- LAPEC EA4278, Avignon Université, Avignon, France
| | - Charles Khouri
- Inserm U1042, Université Grenoble Alpes, Grenoble, France
- Clinical Pharmacology, Grenoble Alpes University Hospital, Grenoble, France
| | - Frances Taylor
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia
| | - Simon Stewart
- Hatter Institute for Reducing Cardiovascular Disease in Africa, The University of Cape Town, Cape Town, South Africa
| | - Christian Lorenzen
- School of Exercise Science, Australian Catholic University, Melbourne, Australia
| | - Jean-Luc Cracowski
- Inserm U1042, Université Grenoble Alpes, Grenoble, France
- Clinical Pharmacology, Grenoble Alpes University Hospital, Grenoble, France
| | - Guillaume Walther
- LAPEC EA4278, Avignon Université, Avignon, France
- School of Exercise Science, Australian Catholic University, Melbourne, Australia
| | - Matthieu Roustit
- Inserm U1042, Université Grenoble Alpes, Grenoble, France
- Clinical Pharmacology, Grenoble Alpes University Hospital, Grenoble, France
| |
Collapse
|
20
|
Endothelial Progenitor Cells Biology in Diabetes Mellitus and Peripheral Arterial Disease and their Therapeutic Potential. Stem Cell Rev Rep 2018; 15:157-165. [DOI: 10.1007/s12015-018-9863-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
21
|
Mehra P, Guo Y, Nong Y, Lorkiewicz P, Nasr M, Li Q, Muthusamy S, Bradley JA, Bhatnagar A, Wysoczynski M, Bolli R, Hill BG. Cardiac mesenchymal cells from diabetic mice are ineffective for cell therapy-mediated myocardial repair. Basic Res Cardiol 2018; 113:46. [PMID: 30353243 PMCID: PMC6314032 DOI: 10.1007/s00395-018-0703-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/04/2018] [Indexed: 01/17/2023]
Abstract
Although cell therapy improves cardiac function after myocardial infarction, highly variable results and limited understanding of the underlying mechanisms preclude its clinical translation. Because many heart failure patients are diabetic, we examined how diabetic conditions affect the characteristics of cardiac mesenchymal cells (CMC) and their ability to promote myocardial repair in mice. To examine how diabetes affects CMC function, we isolated CMCs from non-diabetic C57BL/6J (CMCWT) or diabetic B6.BKS(D)-Leprdb/J (CMCdb/db) mice. When CMCs were grown in 17.5 mM glucose, CMCdb/db cells showed > twofold higher glycolytic activity and a threefold higher expression of Pfkfb3 compared with CMCWT cells; however, culture of CMCdb/db cells in 5.5 mM glucose led to metabolic remodeling characterized by normalization of metabolism, a higher NAD+/NADH ratio, and a sixfold upregulation of Sirt1. These changes were associated with altered extracellular vesicle miRNA content as well as proliferation and cytotoxicity parameters comparable to CMCWT cells. To test whether this metabolic improvement of CMCdb/db cells renders them suitable for cell therapy, we cultured CMCWT or CMCdb/db cells in 5.5 mM glucose and then injected them into infarcted hearts of non-diabetic mice (CMCWT, n = 17; CMCdb/db, n = 13; Veh, n = 14). Hemodynamic measurements performed 35 days after transplantation showed that, despite normalization of their properties in vitro, and unlike CMCWT cells, CMCdb/db cells did not improve load-dependent and -independent parameters of left ventricular function. These results suggest that diabetes adversely affects the reparative capacity of CMCs and that modulating CMC characteristics via culture in lower glucose does not render them efficacious for cell therapy.
Collapse
Affiliation(s)
- Parul Mehra
- Division of Cardiovascular Medicine, Department of Medicine, Institute of Molecular Cardiology, Envirome Institute, Diabetes and Obesity Center, University of Louisville School of Medicine, 580 S. Preston St., Rm 321E, Louisville, KY, 40202, USA
| | - Yiru Guo
- Division of Cardiovascular Medicine, Department of Medicine, Institute of Molecular Cardiology, Envirome Institute, Diabetes and Obesity Center, University of Louisville School of Medicine, 580 S. Preston St., Rm 321E, Louisville, KY, 40202, USA
| | - Yibing Nong
- Division of Cardiovascular Medicine, Department of Medicine, Institute of Molecular Cardiology, Envirome Institute, Diabetes and Obesity Center, University of Louisville School of Medicine, 580 S. Preston St., Rm 321E, Louisville, KY, 40202, USA
| | - Pawel Lorkiewicz
- Division of Cardiovascular Medicine, Department of Medicine, Institute of Molecular Cardiology, Envirome Institute, Diabetes and Obesity Center, University of Louisville School of Medicine, 580 S. Preston St., Rm 321E, Louisville, KY, 40202, USA
| | - Marjan Nasr
- Division of Cardiovascular Medicine, Department of Medicine, Institute of Molecular Cardiology, Envirome Institute, Diabetes and Obesity Center, University of Louisville School of Medicine, 580 S. Preston St., Rm 321E, Louisville, KY, 40202, USA
| | - Qianhong Li
- Division of Cardiovascular Medicine, Department of Medicine, Institute of Molecular Cardiology, Envirome Institute, Diabetes and Obesity Center, University of Louisville School of Medicine, 580 S. Preston St., Rm 321E, Louisville, KY, 40202, USA
| | - Senthilkumar Muthusamy
- Division of Cardiovascular Medicine, Department of Medicine, Institute of Molecular Cardiology, Envirome Institute, Diabetes and Obesity Center, University of Louisville School of Medicine, 580 S. Preston St., Rm 321E, Louisville, KY, 40202, USA
| | - James A Bradley
- Division of Cardiovascular Medicine, Department of Medicine, Institute of Molecular Cardiology, Envirome Institute, Diabetes and Obesity Center, University of Louisville School of Medicine, 580 S. Preston St., Rm 321E, Louisville, KY, 40202, USA
| | - Aruni Bhatnagar
- Division of Cardiovascular Medicine, Department of Medicine, Institute of Molecular Cardiology, Envirome Institute, Diabetes and Obesity Center, University of Louisville School of Medicine, 580 S. Preston St., Rm 321E, Louisville, KY, 40202, USA
| | - Marcin Wysoczynski
- Division of Cardiovascular Medicine, Department of Medicine, Institute of Molecular Cardiology, Envirome Institute, Diabetes and Obesity Center, University of Louisville School of Medicine, 580 S. Preston St., Rm 321E, Louisville, KY, 40202, USA
| | - Roberto Bolli
- Division of Cardiovascular Medicine, Department of Medicine, Institute of Molecular Cardiology, Envirome Institute, Diabetes and Obesity Center, University of Louisville School of Medicine, 580 S. Preston St., Rm 321E, Louisville, KY, 40202, USA
| | - Bradford G Hill
- Division of Cardiovascular Medicine, Department of Medicine, Institute of Molecular Cardiology, Envirome Institute, Diabetes and Obesity Center, University of Louisville School of Medicine, 580 S. Preston St., Rm 321E, Louisville, KY, 40202, USA.
| |
Collapse
|
22
|
Abstract
The dysfunction of endothelial progenitor cells (EPCs) was found to be associated with vascular complications in diabetes mellitus (DM) patients. Previous studies found that regular exercise could improve the function of EPCs in DM patients, but the underling mechanism was unclear. Irisin, a newly identified myokine, was induced by exercise and has been demonstrated to mediate some of the positive effects of exercise. In this study, we hypothesize that irisin may have direct effects on EPC function in DM mice. These data showed for the first time that irisin increased the number of EPCs in peripheral blood of DM mice and improved the function of EPCs derived from DM mice bone marrow. The mechanism for the effect of irisin is related to the PI3K/Akt/eNOS pathway. Furthermore, irisin was demonstrated to improve endothelial repair in DM mice that received EPC transplants after carotid artery injury. The results of this study indicate a novel effect of irisin in regulating the number and function of EPCs via the PI3K/Akt/eNOS pathway, suggesting a potential for the administration of exogenous irisin as a succedaneum to improve EPC function in diabetic patients who fail to achieve such improvements through regular exercise.
Collapse
|
23
|
Mitchell A, Fujisawa T, Mills NL, Brittan M, Newby DE, Cruden NLM. Endothelial Progenitor Cell Biology and Vascular Recovery Following Transradial Cardiac Catheterization. J Am Heart Assoc 2017; 6:e006610. [PMID: 29080864 PMCID: PMC5721759 DOI: 10.1161/jaha.117.006610] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 07/28/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Transradial catheterization is associated with radial artery injury and vasomotor dysfunction and represents an accessible model of acute vascular injury in humans. We characterized vascular injury and functional recovery to understand the role of circulating endothelial progenitor cells in vascular repair. METHODS AND RESULTS In 50 patients (aged 64±10 years, 70% male) undergoing transradial cardiac catheterization, radial artery injury was assessed by optical coherence tomography and examination of explanted vascular sheaths. Flow- and nitrate-mediated dilatation of the radial artery was assessed in both arms at baseline, at 24 hours, and at 1, 4, and 12 weeks. Circulating endothelial progenitor cell populations were quantified using flow cytometry. Late endothelial outgrowth colonies were isolated and examined in vitro. Optical coherence tomography identified macroscopic injury in 12 of 50 patients (24%), but endothelial cells (1.9±1.2×104 cells) were isolated from all arterial sheaths examined. Compared with the noncatheterized radial artery, flow-mediated vasodilatation was impaired in the catheterized artery at 24 hours (9.9±4.6% versus 4.1±3.1%, P<0.0001) and recovered by 12 weeks (8.1±4.9% versus 10.1±4.9%, P=0.09). Although the number of CD133+ cells increased 24 hours after catheterization (P=0.02), the numbers of CD34+ cells and endothelial outgrowth colonies were unchanged. Migration of endothelial cells derived from endothelial outgrowth colonies correlated with arterial function before catheterization but was not related to recovery of function following injury. CONCLUSIONS Transradial cardiac catheterization causes endothelial denudation, vascular injury, and vasomotor dysfunction that recover over 12 weeks. Recovery of vascular function does not appear to be dependent on the mobilization or function of endothelial progenitor cells. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrials.gov. Unique identifier: NCT02147119.
Collapse
Affiliation(s)
- Andrew Mitchell
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, United Kingdom
- Edinburgh Heart Centre, NHS Lothian, Edinburgh, United Kingdom
- BHF Centre for Vascular Regeneration, Scottish Centre for Regenerative Medicine, University of Edinburgh, United Kingdom
| | - Takeshi Fujisawa
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, United Kingdom
- BHF Centre for Vascular Regeneration, Scottish Centre for Regenerative Medicine, University of Edinburgh, United Kingdom
| | - Nicholas L Mills
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, United Kingdom
- Edinburgh Heart Centre, NHS Lothian, Edinburgh, United Kingdom
- BHF Centre for Vascular Regeneration, Scottish Centre for Regenerative Medicine, University of Edinburgh, United Kingdom
| | - Mairi Brittan
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, United Kingdom
- BHF Centre for Vascular Regeneration, Scottish Centre for Regenerative Medicine, University of Edinburgh, United Kingdom
| | - David E Newby
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, United Kingdom
- Edinburgh Heart Centre, NHS Lothian, Edinburgh, United Kingdom
- BHF Centre for Vascular Regeneration, Scottish Centre for Regenerative Medicine, University of Edinburgh, United Kingdom
| | - Nicholas L M Cruden
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, United Kingdom
- Edinburgh Heart Centre, NHS Lothian, Edinburgh, United Kingdom
- BHF Centre for Vascular Regeneration, Scottish Centre for Regenerative Medicine, University of Edinburgh, United Kingdom
| |
Collapse
|
24
|
Han X, Tao Y, Deng Y, Yu J, Sun Y, Jiang G. Metformin accelerates wound healing in type 2 diabetic db/db mice. Mol Med Rep 2017; 16:8691-8698. [PMID: 28990070 PMCID: PMC5779947 DOI: 10.3892/mmr.2017.7707] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 08/10/2017] [Indexed: 12/31/2022] Open
Abstract
Wound healing impairment is increasingly recognized to be a consequence of hyperglycemia-induced dysfunction of endothelial precursor cells (EPCs) in type 2 diabetes mellitus (T2DM). Metformin exhibits potential for the improvement of endothelial function and the wound healing process. However, the underlying mechanisms for the observed beneficial effects of metformin application remain to be completely understood. The present study assessed whether metformin, a widely used therapeutic drug for T2DM, may accelerate wound closure in T2DM db/db mice. Genetically hyperglycemic db/db mice were used as the T2DM model. Metformin (250 mg/kg/day; intragastric) was administered for two weeks prior to EPC collection and wound model creation in db/db mice. Wound healing was evaluated by alterations in the wound area and the number of platelet endothelial cell adhesion molecule-positive cells. The function of the isolated bone marrow-derived EPCs (BM-EPCs) was assessed by a tube formation assay. The number of circulating EPCs, and the levels of intracellular nitric oxide (NO) and superoxide (O2−) were detected by flow cytometry. Thrombospondin-1 (TSP-1) expression was determined by western blot analysis. It was observed that treatment with metformin accelerated wound healing, improved angiogenesis and increased the circulating EPC number in db/db mice. In vitro, treatment with metformin reversed the impaired BM-EPC function reflected by tube formation, and significantly increased NO production while decreasing O2− levels in BM-EPCs from db/db mice. In addition, TSP-1 expression was markedly attenuated by treatment with metformin in cultured BM-EPCs. Metformin contributed to wound healing and improved angiogenesis in T2DM mice, which was, in part, associated with stimulation of NO, and inhibition of O2− and TSP-1 in EPCs from db/db mice.
Collapse
Affiliation(s)
- Xue Han
- Department of Pharmacy, Xiaoshan Hospital, Hangzhou, Zhejiang 311202, P.R. China
| | - Yulong Tao
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Yaping Deng
- Department of Pharmacy, Xiaoshan Hospital, Hangzhou, Zhejiang 311202, P.R. China
| | - Jiawen Yu
- Department of Pharmacy, Xiaoshan Hospital, Hangzhou, Zhejiang 311202, P.R. China
| | - Yuannan Sun
- Department of Pharmacy, Xiaoshan Hospital, Hangzhou, Zhejiang 311202, P.R. China
| | - Guojun Jiang
- Department of Pharmacy, Xiaoshan Hospital, Hangzhou, Zhejiang 311202, P.R. China
| |
Collapse
|
25
|
Aikawa T, Matsubara H, Ugaji S, Shirakawa J, Nagai R, Munesue S, Harashima A, Yamamoto Y, Tsuchiya H. Contribution of methylglyoxal to delayed healing of bone injury in diabetes. Mol Med Rep 2017; 16:403-409. [DOI: 10.3892/mmr.2017.6589] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 03/06/2017] [Indexed: 11/06/2022] Open
|
26
|
Lee PSS, Ye L, Khoo EYH, Yeo TC, Tan HC, Richards AM, Poh KK. Impairment in the number and function of CD34+/KDR+ circulating cells in diabetes and obesity with functional improvement after thymosin β4 treatment. Cardiovasc Endocrinol 2016. [DOI: 10.1097/xce.0000000000000076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
27
|
Endothelial Progenitor Cells in Diabetic Microvascular Complications: Friends or Foes? Stem Cells Int 2016; 2016:1803989. [PMID: 27313624 PMCID: PMC4903148 DOI: 10.1155/2016/1803989] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/05/2016] [Accepted: 04/18/2016] [Indexed: 12/24/2022] Open
Abstract
Despite being featured as metabolic disorder, diabetic patients are largely affected by hyperglycemia-induced vascular abnormality. Accumulated evidence has confirmed the beneficial effect of endothelial progenitor cells (EPCs) in coronary heart disease. However, antivascular endothelial growth factor (anti-VEGF) treatment is the main therapy for diabetic retinopathy and nephropathy, indicating the uncertain role of EPCs in the pathogenesis of diabetic microvascular disease. In this review, we first illustrate how hyperglycemia induces metabolic and epigenetic changes in EPCs, which exerts deleterious impact on their number and function. We then discuss how abnormal angiogenesis develops in eyes and kidneys under diabetes condition, focusing on “VEGF uncoupling with nitric oxide” and “competitive angiopoietin 1/angiopoietin 2” mechanisms that are shared in both organs. Next, we dissect the nature of EPCs in diabetic microvascular complications. After we overview the current EPCs-related strategies, we point out new EPCs-associated options for future exploration. Ultimately, we hope that this review would uncover the mysterious nature of EPCs in diabetic microvascular disease for therapeutics.
Collapse
|
28
|
Haberzettl P, McCracken JP, Bhatnagar A, Conklin DJ. Insulin sensitizers prevent fine particulate matter-induced vascular insulin resistance and changes in endothelial progenitor cell homeostasis. Am J Physiol Heart Circ Physiol 2016; 310:H1423-38. [PMID: 27016579 DOI: 10.1152/ajpheart.00369.2015] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 03/23/2016] [Indexed: 12/15/2022]
Abstract
Exposure to fine particular matter (PM2.5) increases the risk of developing cardiovascular disease and Type 2 diabetes. Because blood vessels are sensitive targets of air pollutant exposure, we examined the effects of concentrated ambient PM2.5 (CAP) on vascular insulin sensitivity and circulating levels of endothelial progenitor cells (EPCs), which reflect cardiovascular health. We found that CAP exposure for 9 days decreased insulin-stimulated Akt phosphorylation in the aorta of mice maintained on control diet. This change was accompanied by the induction of IL-1β and increases in the abundance of cleaved IL-18 and p10 subunit of Casp-1, consistent with the activation of the inflammasome pathway. CAP exposure also suppressed circulating levels of EPCs (Flk-1(+)/Sca-1(+) cells), while enhancing the bone marrow abundance of these cells. Although similar changes in vascular insulin signaling and EPC levels were observed in mice fed high-fat diet, CAP exposure did not exacerbate diet-induced changes in vascular insulin resistance or EPC homeostasis. Treatment with an insulin sensitizer, metformin or rosiglitazone, prevented CAP-induced vascular insulin resistance and NF-κB and inflammasome activation and restored peripheral blood and bone marrow EPC levels. These findings suggest that PM2.5 exposure induces diet-independent vascular insulin resistance and inflammation and prevents EPC mobilization, and that this EPC mobilization defect could be mediated by vascular insulin resistance. Impaired vascular insulin sensitivity may be an important mechanism underlying PM2.5-induced vascular injury, and pharmacological sensitization to insulin action could potentially prevent deficits in vascular repair and mitigate vascular inflammation due to exposure to elevated levels of ambient air pollution.
Collapse
Affiliation(s)
- Petra Haberzettl
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - James P McCracken
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Aruni Bhatnagar
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Daniel J Conklin
- Diabetes and Obesity Center, Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
29
|
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
|
30
|
Thom SR, Hampton M, Troiano MA, Mirza Z, Malay DS, Shannon S, Jennato NB, Donohue CM, Hoffstad O, Woltereck D, Yang M, Yu K, Bhopale VM, Kovtun S, Margolis DJ. Measurements of CD34+/CD45-dim Stem Cells Predict Healing of Diabetic Neuropathic Wounds. Diabetes 2016; 65:486-97. [PMID: 26487786 PMCID: PMC4747459 DOI: 10.2337/db15-0517] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 10/14/2015] [Indexed: 12/12/2022]
Abstract
Management of neuropathic foot ulcers in patients with diabetes (DFUs) has changed little over the past decade, and there is currently no objective method to gauge probability of successful healing. We hypothesized that studies of stem/progenitor cells (SPCs) in the early weeks of standard wound management could predict who will heal within 16 weeks. Blood and debrided wound margins were collected for 8 weeks from 100 patients undergoing weekly evaluations and treatment. SPC number and intracellular content of hypoxia-inducible factors (HIFs) were evaluated by flow cytometry and immunohistochemistry. More SPCs entered the bloodstream in the first 2 weeks of care in patients who healed (n = 37) than in those who did not (n = 63). Logistic regression demonstrated that the number of blood-borne SPCs and the cellular content of HIFs at study entry and the first-week follow-up visit predicted healing. Strong correlations were found among week-to-week assessments of blood-borne SPC HIF factors. We conclude that assays of SPCs during the first weeks of care in patients with DFUs can provide insight into how well wounds will respond and may aid with decisions on the use of adjunctive measures.
Collapse
Affiliation(s)
- Stephen R Thom
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Michelle Hampton
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Michael A Troiano
- Podiatric Surgery and Medicine, Penn Presbyterian Medical Center, Philadelphia, PA
| | - Ziad Mirza
- Department of Medicine, Greater Baltimore Medical Center, Baltimore, MD
| | - D Scot Malay
- Podiatric Surgery and Medicine, Penn Presbyterian Medical Center, Philadelphia, PA
| | - Steven Shannon
- Podiatric Surgery and Medicine, Penn Presbyterian Medical Center, Philadelphia, PA
| | - Nathan B Jennato
- Podiatric Surgery and Medicine, Penn Presbyterian Medical Center, Philadelphia, PA
| | | | - Ole Hoffstad
- Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Diana Woltereck
- Department of Medicine, Greater Baltimore Medical Center, Baltimore, MD
| | - Ming Yang
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Kevin Yu
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Veena M Bhopale
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Svitlana Kovtun
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - David J Margolis
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
31
|
Endothelial Progenitor Cells for Diagnosis and Prognosis in Cardiovascular Disease. Stem Cells Int 2015; 2016:8043792. [PMID: 26839569 PMCID: PMC4709789 DOI: 10.1155/2016/8043792] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/16/2015] [Accepted: 09/20/2015] [Indexed: 12/12/2022] Open
Abstract
Objective. To identify, evaluate, and synthesize evidence on the predictive power of circulating endothelial progenitor cells (EPCs) in cardiovascular disease, through a systematic review of quantitative studies. Data Sources. MEDLINE was searched using keywords related to "endothelial progenitor cells" and "endothelium" and, for the different categories, respectively, "smoking"; "blood pressure"; "diabetes mellitus" or "insulin resistance"; "dyslipidemia"; "aging" or "elderly"; "angina pectoris" or "myocardial infarction"; "stroke" or "cerebrovascular disease"; "homocysteine"; "C-reactive protein"; "vitamin D". Study Selection. Database hits were evaluated against explicit inclusion criteria. From 927 database hits, 43 quantitative studies were included. Data Syntheses. EPC count has been suggested for cardiovascular risk estimation in the clinical practice, since it is currently accepted that EPCs can work as proangiogenic support cells, maintaining their importance as regenerative/reparative potential, and also as prognostic markers. Conclusions. EPCs showed an important role in identifying cardiovascular risk conditions, and to suggest their evaluation as predictor of outcomes appears to be reasonable in different defined clinical settings. Due to their capability of proliferation, circulation, and the development of functional progeny, great interest has been directed to therapeutic use of progenitor cells in atherosclerotic diseases. This trial is registered with registration number: Prospero CRD42015023717.
Collapse
|
32
|
Bitterli L, Afan S, Bühler S, DiSanto S, Zwahlen M, Schmidlin K, Yang Z, Baumgartner I, Diehm N, Kalka C. Endothelial progenitor cells as a biological marker of peripheral artery disease. Vasc Med 2015; 21:3-11. [PMID: 26511986 DOI: 10.1177/1358863x15611225] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The role of endothelial progenitor cells (EPCs) in peripheral artery disease (PAD) remains unclear. We hypothesized that EPC mobilization and function play a central role in the development of endothelial dysfunction and directly influence the degree of atherosclerotic burden in peripheral artery vessels. The number of circulating EPCs, defined as CD34(+)/KDR(+) cells, were assessed by flow cytometry in 91 subjects classified according to a predefined sample size of 31 non-diabetic PAD patients, 30 diabetic PAD patients, and 30 healthy volunteers. Both PAD groups had undergone endovascular treatment in the past. As a functional parameter, EPC colony-forming units were determined ex vivo. Apart from a broad laboratory analysis, a series of clinical measures using the ankle-brachial index (ABI), flow-mediated dilatation (FMD) and carotid intima-media thickness (cIMT) were investigated. A significant reduction of EPC counts and proliferation indices in both PAD groups compared to healthy subjects were observed. Low EPC number and pathological findings in the clinical assessment were strongly correlated to the group allocation. Multivariate statistical analysis revealed these findings to be independent predictors of disease appearance. Linear regression analysis showed the ABI to be a predictor of circulating EPC number (p=0.02). Moreover, the functionality of EPCs was correlated by linear regression (p=0.017) to cIMT. The influence of diabetes mellitus on EPCs in our study has to be considered marginal in already disease-affected patients. This study demonstrated that EPCs could predict the prevalence and severity of symptomatic PAD, with ABI as the determinant of the state of EPC populations in disease-affected groups.
Collapse
Affiliation(s)
- Lukas Bitterli
- Division of Clinical and Interventional Angiology, Swiss Cardiovascular Center, Inselspital, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Samuel Afan
- Division of Cardiovascular Medicine, Marienhospital Brühl, Brühl, Germany
| | - Stephan Bühler
- Division of Clinical and Interventional Angiology, Swiss Cardiovascular Center, Inselspital, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Stefano DiSanto
- Division of Clinical and Interventional Angiology, Swiss Cardiovascular Center, Inselspital, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Marcel Zwahlen
- Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
| | - Kurt Schmidlin
- Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
| | - Zijang Yang
- Division of Clinical and Interventional Angiology, Swiss Cardiovascular Center, Inselspital, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Iris Baumgartner
- Division of Clinical and Interventional Angiology, Swiss Cardiovascular Center, Inselspital, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Nicolas Diehm
- Division of Clinical and Interventional Angiology, Swiss Cardiovascular Center, Inselspital, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Christoph Kalka
- Division of Cardiovascular Medicine, Marienhospital Brühl, Brühl, Germany
| |
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: 55] [Impact Index Per Article: 5.5] [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
|
Zeng YC, Mu GP, Huang SF, Zeng XH, Cheng H, Li ZX. Effects of lycopene on number and function of human peripheral blood endothelial progenitor cells cultivated with high glucose. Nutr Res Pract 2014; 8:368-76. [PMID: 25110555 PMCID: PMC4122707 DOI: 10.4162/nrp.2014.8.4.368] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 02/16/2014] [Accepted: 02/25/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND/OBJECTIVES The objectives of this study were to investigate the effects of lycopene on the migration, adhesion, tube formation capacity, and p38 mitogen-activated protein kinase (p38 MAPK) activity of endothelial progenitor cells (EPCs) cultivated with high glucose (HG) and as well as explore the mechanism behind the protective effects of lycopene on peripheral blood EPCs. MATERIALS/METHODS Mononuclear cells were isolated from human peripheral blood by Ficoll density gradient centrifugation. EPCs were identified after induction of cellular differentiation. Third generation EPCs were incubated with HG (33 mmol/L) or 10, 30, and 50 µg/mL of lycopene plus HG. MTT assay and flow cytometry were performed to assess proliferation and apoptosis of EPCs. EPC migration was assessed by MTT assay with a modified boyden chamber. Adhesion assay was performed by replating EPCs on fibronectin-coated dishes, after which adherent cells were counted. In vitro vasculogenesis activity was assayed by Madrigal network formation assay. Western blotting was performed to analyze protein expression of both phosphorylated and non-phosphorylated p38 MAPK. RESULTS The proliferation, migration, adhesion, and in vitro vasculogenesis capacity of EPCs treated with 10, 30, and 50 µg/mL of lycopene plus HG were all significantly higher comapred to the HG group (P < 0.05). Rates of apoptosis were also significantly lower than that of the HG group. Moreover, lycopene blocked phosphorylation of p38 MAPK in EPCs (P < 0.05). To confirm the causal relationship between MAPK inhibition and the protective effects of lycopene against HG-induced cellular injury, we treated cells with SB203580, a phosphorylation inhibitor. The inhibitor significantly inhibited HG-induced EPC injury. CONCLUSIONS Lycopene promotes proliferation, migration, adhesion, and in vitro vasculogenesis capacity as well as reduces apoptosis of EPCs. Further, the underlying molecular mechanism of the protective effects of lycopene against HG-induced EPC injury may involve the p38 MAPK signal transduction pathway. Specifically, lycopene was shown to inhibit HG-induced EPC injury by inhibiting p38 MAPKs.
Collapse
Affiliation(s)
- Yao-Chi Zeng
- Department of Clinical Nutrition, Shenzhen Traditional Chinese Medicine Hospital, China
| | - Gui-Ping Mu
- Department of Central Laboratory, Shenzhen Traditional Chinese Medicine Hospital, China
| | - Shu-Fen Huang
- Department of Health Education, Shenzhen Traditional Chinese Medicine Hospital, China
| | - Xue-Hui Zeng
- Department of Clinical Laboratory, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Hong Cheng
- Department of Cardiovascular Medicine, Shenzhen Traditional Chinese Medicine Hospital, China
| | - Zhong-Xin Li
- Department of Clinical Laboratory, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| |
Collapse
|
35
|
Nakamura K, Oe H, Kihara H, Shimada K, Fukuda S, Watanabe K, Takagi T, Yunoki K, Miyoshi T, Hirata K, Yoshikawa J, Ito H. DPP-4 inhibitor and alpha-glucosidase inhibitor equally improve endothelial function in patients with type 2 diabetes: EDGE study. Cardiovasc Diabetol 2014; 13:110. [PMID: 25074318 PMCID: PMC4149239 DOI: 10.1186/s12933-014-0110-2] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 06/29/2014] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Alpha glucosidase inhibitor (GI) attenuates postprandial hyperglycemia (PPH) and reduces the risk of cardiovascular events in patients with impaired glucose tolerance or type 2 diabetes. Dipeptidyl peptidase 4 (DPP-4) inhibitors also attenuate PPH. PPH is one of the factors leading to endothelial dysfunction which is an early event in the pathogenesis of atherosclerosis. Furthermore, DPP-4 inhibitors protect endothelial function through a GLP-1-dependent mechanism. However, the impact of these two types of drugs on endothelial dysfunction in patients with type 2 diabetes has not been fully elucidated. We compared the effects of sitagliptin, a DPP-4 inhibitor, and voglibose, an alpha GI, on endothelial function in patients with diabetes. METHODS We conducted a randomized prospective multicenter study in 66 patients with type 2 diabetes who did not achieve the treatment goal with sulfonylurea, metformin or pioglitazone treatment; 31 patients received sitagliptin treatment and 35 patients, voglibose treatment. The flow-mediated dilatation (FMD) of the brachial artery was measured in the fasting state at baseline and after 12 weeks of treatment. The primary endpoint was a change in FMD (ΔFMD) from the baseline to the end of follow-up. The effects of sitagliptin and voglibose on FMD were assessed by ANCOVA after adjustment for the baseline FMD, age, sex, current smoking, diabetes duration and body mass index. Secondary efficacy measures included changes in HbA1c, GIP, GLP-1, C-peptide, CD34, lipid profile, oxidative stress markers, inflammatory markers and eGFR and any adverse events. RESULTS ΔFMD was significantly improved after 12 weeks of treatment in both groups, and there was no significant difference in ΔFMD between the two groups. There were no significant differences in changes in HbA1c, GIP, GLP-1, C-peptide, lipid profile, oxidative stress marker, inflammatory marker and eGFR between the two groups. Compared with voglibose, sitagliptin significantly increased the circulating CD34, a marker of endothelial progenitor cells. Adverse events were observed in 5 patients in only the voglibose group (diarrhea 1, nausea 1, edema 2 and abdominal fullness 1). CONCLUSIONS Sitagliptin improved endothelial dysfunction just as well as voglibose in patients with type 2 diabetes. Sitagliptin had protective effects on endothelial function without adverse events. TRIAL REGISTRATION registered at http://www.umin.ac.jp/ctrj/ under UMIN000003951.
Collapse
Affiliation(s)
- Kazufumi Nakamura
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, 700-8558 Japan
| | - Hiroki Oe
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, 700-8558 Japan
| | - Hajime Kihara
- Department of Internal Medicine, Kihara Cardiovascular Clinic, Asahikawa, Japan
| | - Kenei Shimada
- Department of Internal Medicine and Cardiology, Osaka City University of Medicine, Osaka, Japan
| | - Shota Fukuda
- Department of Medicine, Osaka Ekisaikai Hospital, Osaka, Japan
| | - Kyoko Watanabe
- Department of Internal Medicine, Okayama Saiseikai General Hospital, Okayama, Japan
| | | | - Kei Yunoki
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, 700-8558 Japan
| | - Toru Miyoshi
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, 700-8558 Japan
| | - Kumiko Hirata
- Department of Cardiovascular Medicine, Wakayama Medical University, Wakayama, Japan
| | | | - Hiroshi Ito
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, 700-8558 Japan
| |
Collapse
|
36
|
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
|
37
|
Yiu KH, Tse HF. Specific role of impaired glucose metabolism and diabetes mellitus in endothelial progenitor cell characteristics and function. Arterioscler Thromb Vasc Biol 2014; 34:1136-43. [PMID: 24743430 DOI: 10.1161/atvbaha.114.302192] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The disease burden of diabetes mellitus (DM) and its associated cardiovascular complications represent a growing and major global health problem. Recent studies suggest that circulating exogenous endothelial progenitor cells (EPCs) play an important role in endothelial repair and neovascularization at sites of injury or ischemia. Both experimental and clinical studies have demonstrated that hyperglycemia related to DM can induce alterations to EPCs. The reduction and dysfunction of EPCs related to DM correlate with the occurrence and severity of microvascular and macrovascular complications, suggesting a close mechanistic link between EPC dysfunction and impaired vascular function/repair in DM. These alterations to EPCs, likely mediated by multiple pathophysiological mechanisms, including inflammation, oxidative stress, and alterations in Akt and the nitric oxide pathway, affect EPCs at multiple stages: differentiation and mobilization in the bone marrow, trafficking and survival in the circulation, and homing and neovascularization. Several different therapeutic approaches have consequently been proposed to reverse the reduction and dysfunction of EPCs in DM and may represent a novel therapeutic approach to prevent and treat DM-related cardiovascular complications.
Collapse
Affiliation(s)
- Kai-Hang Yiu
- From the Division of Cardiology, Department of Medicine, Queen Mary Hospital (K.-H.Y., H.-F.T.) and Shenzhen Institute of Research and Innovation (H.-F.T.), University of Hong Kong, Hong Kong, China; and Research Centre of Heart, Brain, Hormone, and Healthy Aging (K.-H.Y., H.-F.T.) and Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine (H.-F.T.), Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Hung-Fat Tse
- From the Division of Cardiology, Department of Medicine, Queen Mary Hospital (K.-H.Y., H.-F.T.) and Shenzhen Institute of Research and Innovation (H.-F.T.), University of Hong Kong, Hong Kong, China; and Research Centre of Heart, Brain, Hormone, and Healthy Aging (K.-H.Y., H.-F.T.) and Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine (H.-F.T.), Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China.
| |
Collapse
|
38
|
Mao L, Tamura Y, Kawao N, Okada K, Yano M, Okumoto K, Kaji H. Influence of diabetic state and vitamin D deficiency on bone repair in female mice. Bone 2014; 61:102-8. [PMID: 24378215 DOI: 10.1016/j.bone.2013.12.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 12/14/2013] [Accepted: 12/19/2013] [Indexed: 12/17/2022]
Abstract
Type 1 diabetes is associated with an increased fracture risk, an impaired fracture healing, and an increased vitamin D insufficiency. However, the role of vitamin D in diabetic bone repair process remains unclear. We therefore examined the effects of vitamin D deficiency on the impaired bone repair in streptozotocin (STZ)-induced diabetes using female mice. Diabetes was induced by STZ injection into female mice after feeding with normal or vitamin D-deficient diet for 6weeks from the age of 4weeks. A femoral bone defect was induced in mice 4 weeks after induction of diabetes. The repair of damaged site on the femur was significantly delayed at days 7 and 10 after bone defect by diabetic state in mice, as assessed by quantitative computed tomography, while vitamin D deficiency did not affect the bone repair both in mice with normal and diabetic state. The decreases in bone mineral density (BMD) at cortical and trabecular bone by diabetic state were significantly augmented by vitamin D deficiency in tibia at the undamaged side in mice. Diabetic state blunted the levels of osteogenic and chondrogenic genes enhanced by vitamin D deficiency. Moreover, vitamin D deficiency significantly aggravated the decreases in osteocalcin and IGF-1 mRNA by diabetic state. In conclusion, our study showed that vitamin D deficiency aggravates the decrease in BMD by diabetic state in female mice, although vitamin D deficiency did not affect bone repair delayed by diabetic state.
Collapse
Affiliation(s)
- Li Mao
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka-Sayama, 589-8511, Japan
| | - Yukinori Tamura
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka-Sayama, 589-8511, Japan
| | - Naoyuki Kawao
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka-Sayama, 589-8511, Japan
| | - Kiyotaka Okada
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka-Sayama, 589-8511, Japan
| | - Masato Yano
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka-Sayama, 589-8511, Japan
| | - Katsumi Okumoto
- Life Science Research Institute, Kinki University Faculty of Medicine, Osaka-Sayama, 589-8511, Japan
| | - Hiroshi Kaji
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka-Sayama, 589-8511, Japan.
| |
Collapse
|
39
|
Mao L, Kawao N, Tamura Y, Okumoto K, Okada K, Yano M, Matsuo O, Kaji H. Plasminogen activator inhibitor-1 is involved in impaired bone repair associated with diabetes in female mice. PLoS One 2014; 9:e92686. [PMID: 24651693 PMCID: PMC3961397 DOI: 10.1371/journal.pone.0092686] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 02/24/2014] [Indexed: 11/18/2022] Open
Abstract
Previous studies suggest that fracture healing is impaired in diabetes; however, the underlying mechanism remains unclear. Here, we investigated the roles of plasminogen activator inhibitor-1 (PAI-1) in the impaired bone repair process by using streptozotocin (STZ)-induced diabetic female wild-type (PAI-1+/+) and PAI-1-deficient (PAI-1−/−) mice. Bone repair and the number of alkaline phosphatase (ALP)-positive cells at the site of a femoral bone damage were comparable in PAI-1+/+ and PAI-1−/− mice without STZ treatment. Although the bone repair process was delayed by STZ treatment in PAI-1+/+ mice, this delayed bone repair was blunted in PAI-1−/− mice. The reduction in the number of ALP-positive cells at the site of bone damage induced by STZ treatment was attenuated in PAI-1−/− mice compared to PAI-1+/+ mice. On the other hand, PAI-1 deficiency increased the levels of ALP and type I collagen mRNA in female mice with or without STZ treatment, and the levels of Osterix and osteocalcin mRNA, suppressed by diabetic state in PAI-1+/+ mice, were partially protected in PAI-1−/− mice. PAI-1 deficiency did not affect formation of the cartilage matrix and the levels of types II and X collagen and aggrecan mRNA suppressed by STZ treatment, although PAI-1 deficiency increased the expression of chondrogenic markers in mice without STZ treatment. The present study indicates that PAI-1 is involved in the impaired bone repair process induced by the diabetic state in part through a decrease in the number of ALP-positive cells.
Collapse
Affiliation(s)
- Li Mao
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka, Japan
| | - Naoyuki Kawao
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka, Japan
| | - Yukinori Tamura
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka, Japan
| | - Katsumi Okumoto
- Life Science Research Institute, Kinki University, Osaka, Japan
| | - Kiyotaka Okada
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka, Japan
| | - Masato Yano
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka, Japan
| | - Osamu Matsuo
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka, Japan
| | - Hiroshi Kaji
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka, Japan
- * E-mail:
| |
Collapse
|
40
|
Liao YF, Feng Y, Chen LL, Zeng TS, Yu F, Hu LJ. Coronary heart disease risk equivalence in diabetes and arterial diseases characterized by endothelial function and endothelial progenitor cell. J Diabetes Complications 2014; 28:214-8. [PMID: 24332936 DOI: 10.1016/j.jdiacomp.2013.09.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 09/24/2013] [Accepted: 09/26/2013] [Indexed: 10/26/2022]
Abstract
AIMS Peripheral Arterial Disease (PAD), Carotid Artery Disease (CAD), and Type 2 Diabetes Mellitus (DM) were considered as "Coronary Heart Disease (CHD) risk equivalents". Vascular endothelial dysfunction was recognized as an early event in the development of atherosclerosis. Involved in neovasculogenesis and maintenance of vascular homeostasis, endothelial progenitor cell (EPC) has been considered as a biological marker of cardiovascular disease. The purpose of this study was to assess the CHD risk equivalents concept by investigating the endothelial function and circulating EPC number in patients with CHD, PAD, CAD and T2DM. METHODS There were four groups in the study: CHD (n = 19), AD [PAD and CAD (n = 17)], DM (n = 21) and healthy controls (HC, n = 20). PAD and CAD were assessed by ultrasonography. Coronal artery angiography was used to identify CHD. The diagnosis of T2DM was based on oral glucose tolerance test and medical history. Vascular endothelial function was assessed by flow-mediated brachial artery dilatation (FMD). Circulating EPC was quantified by flow cytometry. RESULTS The circulating EPC numbers in four groups were CHD, 973 ± 96; AD, 1048 ± 97; T2DM, 1210 ± 125; HC, 1649 ± 112 cells/ml. There were no significant differences in circulating EPC numbers between CHD and AD groups (P > 0.05). Compared with CHD or AD group, T2DM group was associated with a slight increase in circulating EPC numbers (P < 0.05). The results of FMD were almost similar to the circulating EPC numbers(CHD, 4.06 ± 0.54; AD, 3.90 ± 0.48; DM, 3.85 ± 0.57; HC, 5.52 ± 0.67%)except that there was no significant difference among the CHD, AD and T2DM groups (P > 0.05). Age, glycosylated hemoglobin, low density lipoprotein cholesterol, systolic blood pressure, body mass index (BMI) and medical history were the independent risk factors of circulating EPC number in all the patients (P < 0.05). Age, total cholesterol, BMI and medical history were the independent risk factors of FMD in all of the patients (P<0.05). CONCLUSIONS The results of this study supported the equivalents hypothesis and revealed that "CHD risk equivalents" were characterized by the consistent physiological changes of blood vessels in angiogenesis, repairing ability and endothelial function.
Collapse
Affiliation(s)
- Yun-fei Liao
- Department of Endocrinology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Feng
- Department of Orthopedics, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Lu-Lu Chen
- Department of Endocrinology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China.
| | - Tian-shu Zeng
- Department of Endocrinology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Yu
- Department of Endocrinology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Li-jun Hu
- Department of Ultrasound Diagnosis, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
41
|
Role of endothelial progenitor cells and inflammatory cytokines in healing of diabetic foot ulcers. PLoS One 2013; 8:e83314. [PMID: 24358275 PMCID: PMC3865213 DOI: 10.1371/journal.pone.0083314] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 11/01/2013] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND To evaluate changes in endothelial progenitor cells (EPCs) and cytokines in patients with diabetic foot ulceration (DFU) in association with wound healing. METHODS We studied healthy subjects, diabetic patients not at risk of DFU, at risk of DFU and with active DFU. We prospectively followed the DFU patients over a 12-week period. We also investigated similar changes in diabetic rabbit and mouse models of wound healing. RESULTS All EPC phenotypes except the kinase insert domain receptor (KDR)(+)CD133(+) were reduced in the at risk and the DFU groups compared to the controls. There were no major EPC differences between the control and not at risk group, and between the at risk and DFU groups. Serum stromal-cell derived factor-1 (SDF-1) and stem cell factor (SCF) were increased in DFU patients. DFU patients who healed their ulcers had lower CD34(+)KDR(+) count at visits 3 and 4, serum c-reactive protein (CRP) and granulocyte-macrophage colony-stimulating factor (GM-CSF) at visit 1, interleukin-1 (IL-1) at visits 1 and 4. EPCs tended to be higher in both diabetic animal models when compared to their non-diabetic counterparts both before and ten days after wounding. CONCLUSIONS Uncomplicated diabetes does not affect EPCs. EPCs are reduced in patients at risk or with DFU while complete wound healing is associated with CD34(+)KDR(+) reduction, suggesting possible increased homing. Low baseline CRP, IL-1α and GM-CSF serum levels were associated with complete wound healing and may potentially serve as prognostic markers of DFU healing. No animal model alone is representative of the human condition, indicating the need for multiple experimental models.
Collapse
|
42
|
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
|
43
|
Lev EI, Singer J, Leshem-Lev D, Rigler M, Dadush O, Vaduganathan M, Battler A, Kornowski R. Effect of intensive glycaemic control on endothelial progenitor cells in patients with long-standing uncontrolled type 2 diabetes. Eur J Prev Cardiol 2013; 21:1153-62. [DOI: 10.1177/2047487313488300] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Eli I Lev
- Tel-Aviv University, Tel-Aviv, Israel
- The Felsenstein Medical Research Institute, Petah-Tikva, Israel
| | | | | | | | - Oshrat Dadush
- The Felsenstein Medical Research Institute, Petah-Tikva, Israel
| | | | - Alexander Battler
- Tel-Aviv University, Tel-Aviv, Israel
- The Felsenstein Medical Research Institute, Petah-Tikva, Israel
| | - Ran Kornowski
- Tel-Aviv University, Tel-Aviv, Israel
- The Felsenstein Medical Research Institute, Petah-Tikva, Israel
| |
Collapse
|
44
|
Huang JV, Greyson CR, Schwartz GG. PPAR-γ as a therapeutic target in cardiovascular disease: evidence and uncertainty. J Lipid Res 2012; 53:1738-54. [PMID: 22685322 DOI: 10.1194/jlr.r024505] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPAR-γ) is a key regulator of fatty acid metabolism, promoting its storage in adipose tissue and reducing circulating concentrations of free fatty acids. Activation of PPAR-γ has favorable effects on measures of adipocyte function, insulin sensitivity, lipoprotein metabolism, and vascular structure and function. Despite these effects, clinical trials of thiazolidinedione PPAR-γ activators have not provided conclusive evidence that they reduce cardiovascular morbidity and mortality. The apparent disparity between effects on laboratory measurements and clinical outcomes may be related to limitations of clinical trials, adverse effects of PPAR-γ activation, or off-target effects of thiazolidinedione agents. This review addresses these issues from a clinician's perspective and highlights several ongoing clinical trials that may help to clarify the therapeutic role of PPAR-γ activators in cardiovascular disease.
Collapse
Affiliation(s)
- Janice V Huang
- Cardiology Section, Denver VA Medical Center, US Department of Veterans Affairs, Denver, CO, USA
| | | | | |
Collapse
|
45
|
Shin L, Peterson DA. Impaired therapeutic capacity of autologous stem cells in a model of type 2 diabetes. Stem Cells Transl Med 2012. [PMID: 23197759 DOI: 10.5966/sctm.2012-0031] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Endogenous stem cells in the bone marrow respond to environmental cues and contribute to tissue maintenance and repair. In type 2 diabetes, a multifaceted metabolic disease characterized by insulin resistance and hyperglycemia, major complications are seen in multiple organ systems. To evaluate the effects of this disease on the endogenous stem cell population, we used a type 2 diabetic mouse model (db/db), which recapitulates these diabetic phenotypes. Bone marrow-derived mesenchymal stem cells (MSCs) from db/db mice were characterized in vitro using flow cytometric cell population analysis, differentiation, gene expression, and proliferation assays. Diabetic MSCs were evaluated for their therapeutic potential in vivo using an excisional splint wound model in both nondiabetic wild-type and diabetic mice. Diabetic animals possessed fewer MSCs, which were proliferation and survival impaired in vitro. Examination of the recruitment response of stem and progenitor cells after wounding revealed that significantly fewer endogenous MSCs homed to the site of injury in diabetic subjects. Although direct engraftment of healthy MSCs accelerated wound closure in both healthy and diabetic subjects, diabetic MSC engraftment produced limited improvement in the diabetic subjects and could not produce the same therapeutic outcomes as in their nondiabetic counterparts in vivo. Our data reveal stem cell impairment as a major complication of type 2 diabetes in mice and suggest that the disease may stably alter endogenous MSCs. These results have implications for the efficiency of autologous therapies in diabetic patients and identify endogenous MSCs as a potential therapeutic target.
Collapse
Affiliation(s)
- Laura Shin
- Center for Stem Cell and Regenerative Medicine and Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, USA
| | | |
Collapse
|
46
|
Yue WS, Lau KK, Siu CW, Wang M, Yan GH, Yiu KH, Tse HF. Impact of glycemic control on circulating endothelial progenitor cells and arterial stiffness in patients with type 2 diabetes mellitus. Cardiovasc Diabetol 2011; 10:113. [PMID: 22185563 PMCID: PMC3258289 DOI: 10.1186/1475-2840-10-113] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2011] [Accepted: 12/20/2011] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Patients with type 2 diabetes mellitus (DM) have increased risk of endothelial dysfunction and arterial stiffness. Levels of circulating endothelial progenitor cells (EPCs) are also reduced in hyperglycemic states. However, the relationships between glycemic control, levels of EPCs and arterial stiffness are unknown. METHODS We measured circulating EPCs and brachial-ankle pulse wave velocity (baPWV) in 234 patients with type 2 DM and compared them with 121 age- and sex-matched controls. RESULTS Patients with DM had significantly lower circulating Log CD34/KDR+ and Log CD133/KDR+ EPC counts, and higher Log baPWV compared with controls (all P < 0.05). Among those 120/234 (51%) of DM patients with satisfactory glycemic control (defined by Hemoglobin A1c, HbA1c < 6.5%), they had significantly higher circulating Log CD34/KDR+ and Log CD133/KDR+ EPC counts, and lower Log baPWV compared with patients with poor glycemic control (all P < 0.05). The circulating levels of Log CD34/KDR+ EPC (r = -0.46, P < 0.001) and Log CD133/KDR+ EPC counts (r = -0.45, P < 0.001) were negatively correlated with Log baPWV. Whilst the level of HbA1c positively correlated with Log baPWV (r = 0.20, P < 0.05) and negatively correlated with circulating levels of Log CD34/KDR+ EPC (r = -0.40, P < 0.001) and Log CD133/KDR+ EPC (r = -0.41, P < 0.001). Multivariate analysis revealed that HbA1c, Log CD34/KDR+ and Log CD133/KDR+ EPC counts were independent predictors of Log baPWV (P < 0.05). CONCLUSIONS In patients with type 2 DM, the level of circulating EPCs and arterial stiffness were closely related to their glycemic control. Furthermore, DM patients with satisfactory glycemic control had higher levels of circulating EPCs and were associated with lower arterial stiffness.
Collapse
Affiliation(s)
- Wen-Sheng Yue
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong SAR, China
| | | | | | | | | | | | | |
Collapse
|
47
|
Zhang Z, Qun J, Cao C, Wang J, Li W, Wu Y, Du L, Zhao P, Gong K. Apolipoprotein A-I mimetic peptide D-4F promotes human endothelial progenitor cell proliferation, migration, adhesion though eNOS/NO pathway. Mol Biol Rep 2011; 39:4445-54. [PMID: 21947883 DOI: 10.1007/s11033-011-1233-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Accepted: 09/13/2011] [Indexed: 02/07/2023]
Abstract
Circulating endothelial progenitor cells (EPCs) have a critical role in endothelial maintenance and repair. Apolipoprotein A-I mimetic peptide D-4F has been shown to posses anti-atherogenic properties via sequestration of oxidized phospholipids, induction of remodeling of high density lipoprotein and promotion of cholesterol efflux from macrophage-derived foam cells. In this study, we test the effects of D-4F on EPC biology. EPCs were isolated from the peripheral venous blood of healthy male volunteers and characterized by 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine-labeled acetylated LDL uptake and ulex europaeus agglutinin binding and flow cytometry. Cell proliferation, migration, adhesion, nitric oxide production and endothelial nitric oxide synthase (eNOS) expression in the absence and presence of D-4F or simvastatin (as a positive control), were assayed. We demonstrated that D-4F significantly enhanced EPC proliferation, migration and adhesion in a dose-dependent manner compared with vehicle. However, all of the favorable effects of D-4F on EPCs were dramatically attenuated by preincubation with NOS inhibitor L-NAME. Further, D-4F also increased nitric oxide production in culture supernatant and the levels of eNOS expression and phosphorylation. The stimulatory effects of D-4F (10 μg/ml) on EPC biology were comparable to 0.5 μM simvastatin. These results suggest that eNOS/NO pathway mediates the functional modulation of EPC biology in response to D-4F treatment and support the notion that the beneficial role of D-4F on EPCs may be one of the important components of its anti-atherogenic potential.
Collapse
Affiliation(s)
- Zhengang Zhang
- Department of Cardiology, The Second Clinic Medical College, Yangzhou University, Yangzhou, 225001, China
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Chen LL, Yu F, Zeng TS, Liao YF, Li YM, Ding HC. Effects of gliclazide on endothelial function in patients with newly diagnosed type 2 diabetes. Eur J Pharmacol 2011; 659:296-301. [PMID: 21453695 DOI: 10.1016/j.ejphar.2011.02.044] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Revised: 01/25/2011] [Accepted: 02/17/2011] [Indexed: 02/08/2023]
Abstract
Endothelial dysfunction is thought to be a critical event in the pathogenesis of vasculopathy in type 2 diabetes and oxidant stress is a major etiological factor. Gliclazide, a second generation sulfonylurea, contains an azabicyclo-octyl ring, which has been described to have antioxidant properties. However, the effect of gliclazide on endothelial function is unknown. Therefore, in this study, we examined the effect of gliclazide on endothelial function in patients with newly diagnosed type 2 diabetes (diabetic group; n=33). A control group of non-diabetic subjects was also enrolled (n=25). All of the diabetic patients were treated with gliclazide for 12 weeks. Endothelial function was evaluated by flow-mediated vasodilation (FMD) before and after treatment. We also determined the number of circulating endothelial progenitor cells (EPCs), which were defined by CD45(low)/CD34(+)/VEGFR2(+) and quantified by flow cytometry, because these cells may offer a new biomarker for circulatory diseases. Oxidative stress was evaluated in terms of the serum levels of malondialdehyde, superoxide dismutase and nitric oxide. FMD, circulating EPC count and superoxide dismutase activity were significantly lower in the diabetic group than in the control group at baseline (P<0.05), and improved significantly following gliclazide treatment (P<0.05). Malondialdehyde and nitric oxide levels were higher in the diabetic group than in the control group at baseline (P<0.05), and decreased following gliclazide treatment. These results suggest that gliclazide could improve endothelial function in diabetes, which may be related to its antioxidant properties.
Collapse
Affiliation(s)
- Lu-Lu Chen
- Department of Endocrinology, Union Hospital, Huazhong University of Science and Technology, Wuhan, China.
| | | | | | | | | | | |
Collapse
|
49
|
Lenk K, Uhlemann M, Schuler G, Adams V. Role of endothelial progenitor cells in the beneficial effects of physical exercise on atherosclerosis and coronary artery disease. J Appl Physiol (1985) 2011; 111:321-8. [PMID: 21350026 DOI: 10.1152/japplphysiol.01464.2010] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In clinical trials as well as in several animal experiments it is evident that physical exercise is a powerful tool to positively influence the development and/or progression of atherosclerosis and coronary artery disease (CAD). The main target of physical exercise seems to be the maintenance of an intact endothelial cell layer. Since the discovery that endothelial progenitor cells (EPCs) are present in the circulation and the knowledge that exercise, either as a single exercise bout or an exercise training program, have the potency to mobilize EPCs from the bone marrow, the contribution of the EPCs for the preservation or repair of the endothelial cell layer is still under debate. Either the EPCs differentiate into mature endothelial cells, or they stimulate via a paracrine mechanism mature endothelial cells to proliferate. It is still unclear, if the exercise-induced mobilization of EPCs is casually related to the improvement of endothelial function. This review will discuss the role of endothelial progenitor cells in the beneficial effects of physical exercise on atherosclerosis and coronary artery disease.
Collapse
Affiliation(s)
- Karsten Lenk
- University Leipzig, Heart Center, Department of Internal Medicine/Cardiology, Leipzig, Germany
| | | | | | | |
Collapse
|