1
|
Hellenbrand J, Bloomer RJ, Van der Merwe M. The Effect of Short-Term NAD3® Supplementation on Circulating Adult Stem Cells in Healthy Individuals Aged 40-70 Years. Cureus 2024; 16:e55661. [PMID: 38590496 PMCID: PMC11000032 DOI: 10.7759/cureus.55661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2024] [Indexed: 04/10/2024] Open
Abstract
Objective This study aimed to assess the impact of acute and short-term supplementation with NAD3®, a theacrine-containing supplement, on circulating adult stem cell numbers in a healthy male and female population aged 40-70 years. Methods This was a double-blind, placebo-controlled crossover study with 12 participants randomized to receive either NAD3® or a placebo for seven days. Blood samples were collected after an overnight fast, before and after the seven-day supplementation period, and one and two hours after the final supplement dose. Using flow cytometry, circulating stem cells, including lymphocytoid CD34+ stem cells (CD45dimCD34+), stem cells associated with vascular maintenance and repair (CD45dimCD34+CD309+), CD34+ stem cells linked to a progenitor phenotype (CD45dimCD34+CD309neg), circulating endothelial stem cells (CD45negCD31+CD309+), and mesenchymal stem cells (CD45negCD90+) were quantified. Results Acute NAD3® supplementation did not result in the mobilization of stem cells from the bone marrow. However, seven days of daily NAD3® supplementation resulted in selective changes in circulating stem cell numbers. A significant time*treatment interaction was observed for CD45dimCD34+ cells (p=0.04) and CD45dimCD34+CD309neg cells (p=0.04), indicating a decrease in cell numbers with supplementation. There was also a trend toward an increase in circulating endothelial cells (p=0.08) with seven days of NAD3®supplementation. Conclusion Short-term NAD3® supplementation demonstrated an effect on the quantity of bone marrow-derived stem cells in circulation. The study suggests that this theacrine-containing supplement may play a role in modulating adult stem cell populations, emphasizing the potential impact of NAD3® on regenerative processes. Further research with extended supplementation periods and larger sample sizes is warranted to elucidate the functional consequences of these changes and explore the therapeutic implications for age-related declines in stem cell function.
Collapse
|
2
|
Dhindsa DS, Desai SR, Jin Q, Sandesara PB, Mehta A, Liu C, Tahhan AS, Nayak A, Ejaz K, Hooda A, Moazzami K, Islam SJ, Rogers SC, Almuwaqqat Z, Mokhtari A, Hesaroieh I, Ko YA, Sperling LS, Waller EK, Quyyumi AA. Circulating progenitor cells and outcomes in patients with coronary artery disease. Int J Cardiol 2023; 373:7-16. [PMID: 36460208 PMCID: PMC9840693 DOI: 10.1016/j.ijcard.2022.11.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 11/10/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Low quantities of circulating progenitor cells (CPCs), specifically CD34+ populations, reflect impairment of intrinsic regenerative capacity. This study investigates the relationship between subsets of CPCs and adverse outcomes. METHODS 1366 individuals undergoing angiography for evaluation of coronary artery disease (CAD) were enrolled into the Emory Cardiovascular Biobank. Flow cytometry identified CPCs as CD45med blood mononuclear cells expressing the CD34 epitope, with further enumeration of hematopoietic CPCs as CD133+/CXCR4+ cells and endothelial CPCs as vascular endothelial growth factor receptor-2 (VEGFR2+) cells. Adjusted Cox or Fine and Gray's sub-distribution hazard regression models analyzed the relationship between CPCs and 1) all-cause death and 2) a composite of cardiovascular death and non-fatal myocardial infarction (MI). RESULTS Over a median 3.1-year follow-up period (IQR 1.3-4.9), there were 221 (16.6%) all-cause deaths and 172 (12.9%) cardiovascular deaths/MIs. Hematopoietic CPCs were highly correlated, and the CD34+/CXCR4+ subset was the best independent predictor. Lower counts (≤median) of CD34+/CXCR4+ and CD34+/VEGFR2+ cells independently predicted all-cause mortality (HR 1.46 [95% CI 1.06-2.01], p = 0.02 and 1.59 [95% CI 1.15-2.18], p = 0.004) and cardiovascular death/MI (HR 1.50 [95% CI 1.04-2.17], p = 0.03 and 1.47 [95% CI 1.01-2.03], p = 0.04). A combination of low CD34+/CXCR4+ and CD34+/VEGFR2+ CPCs predicted all-cause death (HR 2.1, 95% CI 1.4-3.0; p = 0.0002) and cardiovascular death/MI (HR 2.0, 95% CI 1.3-3.2; p = 0.002) compared to those with both lineages above the cut-offs. CONCLUSIONS Lower levels of hematopoietic and endothelial CPCs indicate diminished endogenous regenerative capacity and independently correlate with greater mortality and cardiovascular risk in patients with CAD.
Collapse
Affiliation(s)
- Devinder S Dhindsa
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Shivang R Desai
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Qingchun Jin
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Pratik B Sandesara
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Anurag Mehta
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Chang Liu
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Ayman S Tahhan
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Aditi Nayak
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Kiran Ejaz
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Ananya Hooda
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Kasra Moazzami
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Shabatun J Islam
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Steven C Rogers
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Zakaria Almuwaqqat
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Ali Mokhtari
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Iraj Hesaroieh
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Yi-An Ko
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Laurence S Sperling
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Edmund K Waller
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Arshed A Quyyumi
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.
| |
Collapse
|
3
|
Moazzami K, Mehta A, Young A, Dhindsa DS, Martin G, Mokhtari A, Hesaroieh IG, Shah A, Bremner JD, Vaccarino V, Waller EK, Quyyumi AA. The association between baseline circulating progenitor cells and vascular function: The role of aging and risk factors. Vasc Med 2022; 27:532-541. [PMID: 36062298 PMCID: PMC10150400 DOI: 10.1177/1358863x221116411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND To investigate the cross-sectional and longitudinal relationships between vascular function and circulating progenitor cell (CPC) counts with respect to aging and exposure to risk factors. METHODS In 797 adult participants, CPCs were enumerated by flow cytometry as CD45med mononuclear cells expressing CD34 epitope and its subsets co-expressing CD133, and chemokine C-X-C motif receptor 4 (CXCR4+). Arterial stiffness was evaluated by tonometry-derived pulse wave velocity (PWV) and microvascular function was assessed as digital reactive hyperemia index (RHI). RESULTS In cross-sectional analyses, for every doubling in CD34+ cell counts, PWV was 15% higher and RHI was 9% lower, after adjusting for baseline characteristics and risk factors (p for all < 0.01). There were significant CPC-by-age-by-risk factor interactions (p <0.05) for both vascular measures. Among younger subjects (< 48 years), CPC counts were higher in those with risk factors and vascular function was better in those with higher compared to those with lower CPC counts (p for all < 0.0l). In contrast, in older participants, CPCs were not higher in those with risk factors, and vascular function was worse compared to the younger age group. A lower CPC count at baseline was an independent predictor of worsening vascular function during 2-year follow-up. CONCLUSION A higher CPC count in the presence of risk factors is associated with better vascular function among younger individuals. There is no increase in CPC count with risk factors in older individuals who have worse vascular function. Moreover, a higher CPC count is associated with less vascular dysfunction with aging.
Collapse
Affiliation(s)
- Kasra Moazzami
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Anurag Mehta
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - An Young
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Devinder Singh Dhindsa
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Greg Martin
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Ali Mokhtari
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Iraj Ghaini Hesaroieh
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Amit Shah
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, GA, USA
| | - J Douglas Bremner
- Atlanta VA Medical Center, Decatur, GA, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Viola Vaccarino
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Edmund K Waller
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Arshed A Quyyumi
- Department of Medicine, Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
4
|
Cardioprotective Mechanisms of Interrupted Anesthetic Preconditioning with Sevoflurane in the Setting of Ischemia/Reperfusion Injury in Rats. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12031476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Anesthetic preconditioning (AP) is known to mimic ischemic preconditioning. The purpose of this study was to investigate the effects of an interrupted sevoflurane administration protocol on myocardial ischemia/reperfusion (I/R) injury. Methods: Male Wistar rats (n = 60) were ventilated for 30 min with room air (control group, CG) or with a mixture of air and sevoflurane (1 minimum alveolar concentration—MAC) in 5-min cycles, alternating with 5-min wash-out periods (preconditioned groups). Cytokines implicated in the AP response were measured. An (I/R) lesion was produced immediately after the sham intervention (CG) and preconditioning protocol (early AP group, EAPG) or 24 h after the intervention (late AP group, LAPG). The area of fibrosis, the degree of apoptosis and the number of c-kit+ cells was estimated for each group. Results: Cytokine levels were increased post AP. The area of fibrosis decreased in both EAPG and LAPG compared to the CG (p < 0.0001). When compared to the CG, the degree of apoptosis was reduced in both LAPG (p = 0.006) and EAPG (p = 0.007) and the number of c-kit+ cells was the greatest for the LAPG (p < 0.0001). Conclusions: Sevoflurane preconditioning, using an interrupted anesthesia protocol, is efficient in myocardial protection and could be beneficial to reduce perioperative or periprocedural ischemia in patients with increased cardiovascular risk.
Collapse
|
5
|
Oprescu N, Micheu MM, Scafa-Udriste A, Popa-Fotea NM, Dorobantu M. Inflammatory markers in acute myocardial infarction and the correlation with the severity of coronary heart disease. Ann Med 2021; 53:1041-1047. [PMID: 34180324 PMCID: PMC8245096 DOI: 10.1080/07853890.2021.1916070] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 04/03/2021] [Accepted: 04/07/2021] [Indexed: 11/15/2022] Open
Abstract
INTRODUCTION The inflammatory hypothesis of atherosclerosis is appealing in acute coronary syndromes, but the dynamics and precise role are not established. OBJECTIVES The study investigates the levels of C reactive protein (CRP), interleukin 1β (IL-1β) and stromal-derived factor 1α (SDF-1α) at the time of acute myocardial infarction (AMI) and at 1 and 6 months afterwards, compared with a control group. RESULTS In the acute phase of AMI, CRP and SDF-1α were significantly higher, while IL-1β showed lower levels compared with controls. CRP positively correlated with coronary stenosis severity (rho = 0.3, p=.05) and negatively related with left ventricle ejection fraction (LVEF) at 1 month (rho= -0.43, p=.05). IL-1β weakly correlated with the severity of coronary lesions (rho =0.29, p=.02) and strongly with LVEF (rho= -0.8, p=.05). SDF-1α, slightly correlated with LVEF at 1 month (rho = 0.22, p=.01) and with the severity of coronary atherosclerosis (rho= -0.41, p=.003). CONCLUSIONS CRP, IL-1β and SDF-1α have important dynamic in the first 6 months after AMI and CRP and SDF-1α levels correlated with the severity of coronary lesions and LVEF at 1 month after the acute ischaemic event.
Collapse
Affiliation(s)
- Nicoleta Oprescu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Romania
| | | | - Alexandru Scafa-Udriste
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Romania
- Cardio-thoracic Department, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| | - Nicoleta-Monica Popa-Fotea
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Romania
- Cardio-thoracic Department, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| | - Maria Dorobantu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Romania
- Cardio-thoracic Department, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| |
Collapse
|
6
|
Chu CS, Law SH, Lenzen D, Tan YH, Weng SF, Ito E, Wu JC, Chen CH, Chan HC, Ke LY. Clinical Significance of Electronegative Low-Density Lipoprotein Cholesterol in Atherothrombosis. Biomedicines 2020; 8:biomedicines8080254. [PMID: 32751498 PMCID: PMC7460408 DOI: 10.3390/biomedicines8080254] [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: 06/30/2020] [Revised: 07/25/2020] [Accepted: 07/28/2020] [Indexed: 02/06/2023] Open
Abstract
Despite the numerous risk factors for atherosclerotic cardiovascular diseases (ASCVD), cumulative evidence shows that electronegative low-density lipoprotein (L5 LDL) cholesterol is a promising biomarker. Its toxicity may contribute to atherothrombotic events. Notably, plasma L5 LDL levels positively correlate with the increasing severity of cardiovascular diseases. In contrast, traditional markers such as LDL-cholesterol and triglyceride are the therapeutic goals in secondary prevention for ASCVD, but that is controversial in primary prevention for patients with low risk. In this review, we point out the clinical significance and pathophysiological mechanisms of L5 LDL, and the clinical applications of L5 LDL levels in ASCVD can be confidently addressed. Based on the previously defined cut-off value by receiver operating characteristic curve, the acceptable physiological range of L5 concentration is proposed to be below 1.7 mg/dL. When L5 LDL level surpass this threshold, clinically relevant ASCVD might be present, and further exams such as carotid intima-media thickness, pulse wave velocity, exercise stress test, or multidetector computed tomography are required. Notably, the ultimate goal of L5 LDL concentration is lower than 1.7 mg/dL. Instead, with L5 LDL greater than 1.7 mg/dL, lipid-lowering treatment may be required, including statin, ezetimibe or PCSK9 inhibitor, regardless of the low-density lipoprotein cholesterol (LDL-C) level. Since L5 LDL could be a promising biomarker, we propose that a high throughput, clinically feasible methodology is urgently required not only for conducting a prospective, large population study but for developing therapeutics strategies to decrease L5 LDL in the blood.
Collapse
Affiliation(s)
- Chih-Sheng Chu
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan;
- Division of Cardiology, Department of International Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Shi Hui Law
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
| | - David Lenzen
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
| | - Yong-Hong Tan
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
| | - Shih-Feng Weng
- Department of Healthcare Administration and Medical Informatics, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan;
| | - Etsuro Ito
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
- Department of Biology, Waseda University, Tokyo 162-8480, Japan
- Waseda Research Institute for Science and Engineering, Waseda University, Tokyo 162-8480, Japan
| | - Jung-Chou Wu
- Division of Cardiology, Department of Internal Medicine, Pingtung Christian Hospital, Pingtung 90059, Taiwan;
| | - Chu-Huang Chen
- Vascular and Medicinal Research, Texas Heart Institute, Houston, TX 77030, USA;
| | - Hua-Chen Chan
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807377, Taiwan;
- Correspondence: (H.-C.C.); (L.-Y.K.); Tel.: +886-73121101 (ext. 2296); Fax: +886-73111996 (L.-Y.K.)
| | - Liang-Yin Ke
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan; (S.H.L.); (D.L.); (Y.-H.T.); (E.I.)
- Graduate Institute of Medicine, College of Medicine, & Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Correspondence: (H.-C.C.); (L.-Y.K.); Tel.: +886-73121101 (ext. 2296); Fax: +886-73111996 (L.-Y.K.)
| |
Collapse
|
7
|
Akyol O, Chowdhury I, Akyol HR, Tessier K, Vural H, Akyol S. Why are cardiovascular diseases more common among patients with severe mental illness? The potential involvement of electronegative low-density lipoprotein (LDL) L5. Med Hypotheses 2020; 142:109821. [PMID: 32417641 DOI: 10.1016/j.mehy.2020.109821] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/22/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023]
Abstract
Despite tremendous efforts of experimental and clinical studies and knowledge, the pathophysiology of severe mental illness (SMI), including bipolar disorder (BD), unipolar depression (mood disorders, MD), and schizophrenia (SCZ), remains poorly understood. Besides their chronic course and high prevalence in society, mental and somatic comorbidities are really serious problems; patients with these disorders have increased risk of cardiovascular (CV) diseases (CVD) including coronary artery diseases (CAD, i.e. myocardial infarction and angina), stroke, sudden cardiac death, hypertension, cardiomyopathy, arrhythmia, and thromboembolic disease. Although it is determined that triglycerides, cholesterol, glucose, and low-density lipoprotein (LDL) levels are increased in MD and SCZ, the underlying reason remains unknown. Considering this, we propose that electronegative LDL (L5) is probably the main crucial element to understanding CVD induced by SMI and to discovering novel remedial approaches for these diseases. When it is hypothesized that L5 is greatly presupposed in CV system abnormalities, it follows that the anti-L5 therapies and even antioxidant treatment options may open new therapeutic opportunities to prevent CVD diseases secondary to SMI. In this review article, we tried to bring a very original subject to the attention of readers who are interested in lipoprotein metabolism in terms of experimental, clinical, and cell culture studies that corroborate the involvement of L5 in physiopathology of CVD secondary to SMI and also the new therapeutic approaches for these disorders.
Collapse
Affiliation(s)
- Omer Akyol
- Michigan Math & Science Academy, Department of Science, Warren, MI, USA.
| | - Imtihan Chowdhury
- Michigan Math & Science Academy, High School, 11th grade, Warren, MI, USA
| | - Hafsa Rana Akyol
- Illinois Institute of Technology, Biology, Sophomore, Chicago, IL, USA
| | - Kylie Tessier
- Michigan Math & Science Academy, High School, 11th grade, Warren, MI, USA
| | - Huseyin Vural
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, Aachen, Germany
| | - Sumeyya Akyol
- Beaumont Health, Beaumont Research Institute, Royal Oak, MI, USA
| |
Collapse
|
8
|
Tao Z, Tan S, Chen W, Chen X. Stem Cell Homing: a Potential Therapeutic Strategy Unproven for Treatment of Myocardial Injury. J Cardiovasc Transl Res 2018; 11:403-411. [PMID: 30324254 DOI: 10.1007/s12265-018-9823-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/26/2018] [Indexed: 02/06/2023]
Abstract
Despite advances in the prevention and therapeutic modalities of ischemic heart disease, morbidity and mortality post-infarction heart failure remain big challenges in modern society. Stem cell therapy is emerging as a promising therapeutic strategy. Stem cell homing, the ability of stem cells to find their destination, is receiving more attention. Identification of specific cues and understanding the signaling pathways that direct stem cells to targeted destination will improve stem cell homing efficiency. This review discusses the cellular and molecular mechanism of stem cell homing at length in the light of literature and analyzes the problem and considerations of this approach as a treatment strategy for the treatment of ischemic heart disease clinically.
Collapse
Affiliation(s)
- Zhonghao Tao
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Shihua Tan
- National Heart Centre Singapore, 5 Hospital Drive, Singapore, 169609, Singapore
| | - Wen Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China
| | - Xin Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, People's Republic of China.
| |
Collapse
|
9
|
Samman Tahhan A, Hammadah M, Raad M, Almuwaqqat Z, Alkhoder A, Sandesara PB, Mohamed-Kelli H, Hayek SS, Kim JH, O'Neal WT, Topel ML, Grant AJ, Sabbak N, Heinl RE, Gafeer MM, Obideen M, Kaseer B, Abdelhadi N, Ko YA, Liu C, Hesaroieh I, Mahar EA, Vaccarino V, Waller EK, Quyyumi AA. Progenitor Cells and Clinical Outcomes in Patients With Acute Coronary Syndromes. Circ Res 2018; 122:1565-1575. [PMID: 29514830 PMCID: PMC5970041 DOI: 10.1161/circresaha.118.312821] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 02/27/2018] [Accepted: 03/02/2018] [Indexed: 11/16/2022]
Abstract
RATIONALE Circulating progenitor cells (CPCs) mobilize in response to ischemic injury, but their predictive value remains unknown in acute coronary syndrome (ACS). OBJECTIVE We aimed to investigate the number of CPCs in ACS compared with those with stable coronary artery disease (CAD), relationship between bone marrow PCs and CPCs, and whether CPC counts predict mortality in patients with ACS. METHODS AND RESULTS In 2028 patients, 346 had unstable angina, 183 had an acute myocardial infarction (AMI), and the remaining 1499 patients had stable CAD. Patients with ACS were followed for the primary end point of all-cause death. CPCs were enumerated by flow cytometry as mononuclear cells expressing a combination of CD34+, CD133+, vascular endothelial growth factor receptor 2+, or chemokine (C-X-C motif) receptor 4+. CPC counts were higher in subjects with AMI compared those with stable CAD even after adjustment for age, sex, race, body mass index, renal function, hypertension, diabetes mellitus, hyperlipidemia, and smoking; CD34+, CD34+/CD133+, CD34+/CXCR4+, and CD34+/VEGFR2+ CPC counts were 19%, 25%, 28%, and 142% higher in those with AMI, respectively, compared with stable CAD. There were strong correlations between the concentrations of CPCs and the PC counts in bone marrow aspirates in 20 patients with AMI. During a 2 (interquartile range, 1.31-2.86)-year follow-up period of 529 patients with ACS, 12.4% died. In Cox regression models adjusted for age, sex, body mass index, heart failure history, estimated glomerular filtration rate, and AMI, subjects with low CD34+ cell counts had a 2.46-fold (95% confidence interval, 1.18-5.13) increase in all-cause mortality, P=0.01. CD34+/CD133+ and CD34+/CXCR4+, but not CD34+/VEGFR2+ PC counts, had similar associations with mortality. Results were validated in a separate cohort of 238 patients with ACS. CONCLUSIONS CPC levels are significantly higher in patients after an AMI compared with those with stable CAD and reflect bone marrow PC content. Among patients with ACS, a lower number of hematopoietic-enriched CPCs are associated with a higher mortality.
Collapse
Affiliation(s)
- Ayman Samman Tahhan
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Muhammad Hammadah
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Mohamad Raad
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Zakaria Almuwaqqat
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Ayman Alkhoder
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Pratik B Sandesara
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Heval Mohamed-Kelli
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Salim S Hayek
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Jeong Hwan Kim
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Wesley T O'Neal
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Matthew L Topel
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Aubrey J Grant
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Nabil Sabbak
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Robert E Heinl
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Mohamad Mazen Gafeer
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Malik Obideen
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Belal Kaseer
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Nasser Abdelhadi
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Yi-An Ko
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Department of Biostatistics and Bioinformatics (Y.-A.K., C.L., E.A.M.)
| | - Chang Liu
- Department of Biostatistics and Bioinformatics (Y.-A.K., C.L., E.A.M.)
| | - Iraj Hesaroieh
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Ernestine A Mahar
- Department of Biostatistics and Bioinformatics (Y.-A.K., C.L., E.A.M.)
| | - Viola Vaccarino
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| | - Edmund K Waller
- Department of Hematology and Oncology, Winship Cancer Institute (E.K.K.), Emory University, Atlanta, GA
| | - Arshed A Quyyumi
- From the Emory Clinical Cardiovascular Research Institute Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA (A.S.T., M.H., M.R., Z.A., A.A., P.B.S., H.M.-K., S.S.H., J.H.K., W.T.O., M.L.T., A.J.G., N.S., R.E.H., M.M.G., M.O., B.K., N.A., Y.-A.K., I.H., V.V., A.A.Q.)
| |
Collapse
|
10
|
van der Valk FM, Kuijk C, Verweij SL, Stiekema LCA, Kaiser Y, Zeerleder S, Nahrendorf M, Voermans C, Stroes ESG. Increased haematopoietic activity in patients with atherosclerosis. Eur Heart J 2018; 38:425-432. [PMID: 27357356 DOI: 10.1093/eurheartj/ehw246] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 05/25/2016] [Indexed: 12/31/2022] Open
Abstract
Aims Experimental work posits that acute ischaemic events trigger haematopoietic activity, driving monocytosis, and atherogenesis. Considering the chronic low-grade inflammatory state in atherosclerosis, we hypothesized that haematopoietic hyperactivity is a persistent feature in cardiovascular disease (CVD). Therefore, we aimed to assess the activity of haematopoietic organs and haematopoietic stem and progenitor cells (HSPCs) in humans. Methods and results First, we performed 18F-fluorodeoxyglucose positron emission tomographic (18F-FDG PET) imaging in 26 patients with stable atherosclerotic CVD (ischaemic event >12 months ago), and 25 matched controls. In splenic tissue, 18F-FDG uptake was 2.68 ± 0.65 in CVD patients vs. 1.75 ± 0.54 in controls (1.6-fold higher; P< 0.001), and in bone marrow 3.20 ± 0.76 vs. 2.72 ± 0.46 (1.2-fold higher; P = 0.003), closely related to LDL cholesterol levels (LDLc, r = 0.72). Subsequently, we determined progenitor potential of HSPCs harvested from 18 patients with known atherosclerotic CVD and 30 matched controls; both groups were selected from a cohort of cancer patients undergoing autologous stem cell transplantation. In CVD patients, the normalized progenitor potential, expressed as the number of colony-forming units-granulocyte/monocyte (CFU-GM) colonies/CD34+ cell, was 1.6-fold higher compared with matched controls (P < 0.001). Finally, we assessed the effects of native and oxidized lipoproteins on HSPCs harvested from healthy donors in vitro. Haematopoietic stem and progenitor cells displayed a 1.5-fold increased CFU-GM capacity in co-culture with oxidized LDL in vitro (P = 0.002), which was inhibited by blocking oxidized phospholipids via E06 (P = 0.001). Conclusion Collectively, these findings strengthen the case for a chronically affected haematopoietic system, potentially driving the low-grade inflammatory state in patients with atherosclerosis.
Collapse
Affiliation(s)
- Fleur M van der Valk
- Department of Vascular Medicine, AMC, Room F4-146, PO Box 22660, 1100 DD, Amsterdam, The Netherlands
| | - Carlijn Kuijk
- Department of Hematopoiesis, Sanquin Research, and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Simone L Verweij
- Department of Vascular Medicine, AMC, Room F4-146, PO Box 22660, 1100 DD, Amsterdam, The Netherlands
| | - Lotte C A Stiekema
- Department of Vascular Medicine, AMC, Room F4-146, PO Box 22660, 1100 DD, Amsterdam, The Netherlands
| | - Y Kaiser
- Department of Vascular Medicine, AMC, Room F4-146, PO Box 22660, 1100 DD, Amsterdam, The Netherlands
| | - Sacha Zeerleder
- Department of Hematology, AMC, Amsterdam, The Netherlands.,Department of Immunopathology, Sanquin Research, Amsterdam, The Netherlands
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | - Carlijn Voermans
- Department of Hematopoiesis, Sanquin Research, and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Erik S G Stroes
- Department of Vascular Medicine, AMC, Room F4-146, PO Box 22660, 1100 DD, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Yang TC, Chang PY, Kuo TL, Lu SC. Electronegative L5-LDL induces the production of G-CSF and GM-CSF in human macrophages through LOX-1 involving NF-κB and ERK2 activation. Atherosclerosis 2017; 267:1-9. [PMID: 29078142 DOI: 10.1016/j.atherosclerosis.2017.10.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/30/2017] [Accepted: 10/12/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS Circulating levels of granulocyte colony-stimulating factor (G-CSF) and granulocyte macrophage colony-stimulating factor (GM-CSF) are associated with the severity of acute myocardial infarction (AMI). However, what causes increases in G-CSF and GM-CSF is unclear. In this study, we investigated whether L5-low-density lipoprotein (LDL), a mildly oxidized LDL from AMI, can induce G-CSF and GM-CSF production in human macrophages. METHODS L1-LDL and L5-LDL were isolated through anion-exchange chromatography from AMI plasma. Human macrophages derived from THP-1 and peripheral blood mononuclear cells were treated with L1-LDL, L5-LDL, or copper-oxidized LDL (Cu-oxLDL) and G-CSF and GM-CSF protein levels in the medium were determined. In addition, the effects of L5-LDL on G-CSF and GM-CSF production were tested in lectin-type oxidized LDL receptor-1 (LOX-1), CD36, extracellular signal-regulated kinase (ERK) 1, and ERK2 knockdown THP-1 macrophages. RESULTS L5-LDL but not L1-LDL or Cu-oxLDL significantly induced production of G-CSF and GM-CSF in macrophages. In vitro oxidation of L1-LDL and L5-LDL altered their ability to induce G-CSF and GM-CSF, suggesting that the degree of oxidation is critical for the effects. Knockdown and antibody neutralization experiments suggested that the effects were caused by LOX-1. In addition, nuclear factor (NF)-κB and ERK1/2 inhibition resulted in marked reductions of L5-LDL-induced G-CSF and GM-CSF production. Moreover, knockdown of ERK2, but not ERK1, hindered L5-LDL-induced G-CSF and GM-CSF production. CONCLUSIONS The results indicate that L5-LDL, a naturally occurring mild oxidized LDL, induced G-CSF and GM-CSF production in human macrophages through LOX-1, ERK2, and NF-κB dependent pathways.
Collapse
Affiliation(s)
- Tzu-Ching Yang
- Department of Biochemistry and Molecular Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Po-Yuan Chang
- Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Tzu-Ling Kuo
- Department of Biochemistry and Molecular Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Shao-Chun Lu
- Department of Biochemistry and Molecular Biology, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
12
|
D'Amario D, Leone AM, Borovac JA, Cannata F, Siracusano A, Niccoli G, Crea F. Granulocyte colony-stimulating factor for the treatment of cardiovascular diseases: An update with a critical appraisal. Pharmacol Res 2017; 127:67-76. [PMID: 28602846 DOI: 10.1016/j.phrs.2017.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/30/2017] [Accepted: 06/06/2017] [Indexed: 01/22/2023]
Abstract
Heart failure and acute myocardial infarction are conditions that are associated with high morbidity and mortality. Significant dysfunction of the heart muscle can occur as the consequence of end-stage chronic cardiovascular diseases or acute ischemic events that are marked by large infarction area and significant tissue necrosis. Despite the remarkable improvement of conventional treatments, a substantial proportion of patients still develops severe heart failure that can only be resolved by heart transplantation or mechanical device implantation. Therefore, novel approaches based on stem-cell therapy can directly modify the disease process and alter its prognosis. The ability of the stem-cells to modify and repair the injured myocardium is a challenging but intriguing concept that can potentially replace expensive and invasive methods of treatment that are associated with increased risks and significant financial costs. In that sense, granulocyte colony-stimulating factor (G-CSF) seems as an attractive treatment approach. Based on the series of pre-clinical experiments and a limited amount of clinical data, it was demonstrated that G-CSF agents possess the ability to mobilize stem-cells from bone marrow and induce their differentiation into cardiomyocytes or endothelial cells when brought into contact with injured regions of the myocardium. However, clinical benefits of G-CSF use in damaged myocardium remain unclear and are the topic of expert discussion. The main goal of this review is to present relevant and up-to-date evidence on G-CSF therapy use in pre-clinical models and in humans and to provide a rationale for its potential clinical applications in the future.
Collapse
Affiliation(s)
- Domenico D'Amario
- Institute of Cardiology, Catholic University of the Sacred Heart, Largo Agostino Gemelli, 8, Rome, 00168, Italy
| | - Antonio Maria Leone
- Institute of Cardiology, Catholic University of the Sacred Heart, Largo Agostino Gemelli, 8, Rome, 00168, Italy
| | - Josip Anđelo Borovac
- Department of Pathophysiology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia
| | - Francesco Cannata
- Institute of Cardiology, Catholic University of the Sacred Heart, Largo Agostino Gemelli, 8, Rome, 00168, Italy
| | - Andrea Siracusano
- Institute of Cardiology, Catholic University of the Sacred Heart, Largo Agostino Gemelli, 8, Rome, 00168, Italy
| | - Giampaolo Niccoli
- Institute of Cardiology, Catholic University of the Sacred Heart, Largo Agostino Gemelli, 8, Rome, 00168, Italy
| | - Filippo Crea
- Institute of Cardiology, Catholic University of the Sacred Heart, Largo Agostino Gemelli, 8, Rome, 00168, Italy.
| |
Collapse
|
13
|
Bromage DI, Taferner S, Pillai M, Yellon DM, Davidson SM. A novel recombinant antibody specific to full-length stromal derived factor-1 for potential application in biomarker studies. PLoS One 2017; 12:e0174447. [PMID: 28379992 PMCID: PMC5381782 DOI: 10.1371/journal.pone.0174447] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/09/2017] [Indexed: 11/21/2022] Open
Abstract
Background Stromal derived factor-1α (SDF-1α/CXCL12) is a chemokine that is up-regulated in diseases characterised by tissue hypoxia, including myocardial infarction, ischaemic cardiomyopathy and remote ischaemic conditioning (RIC), a technique of cyclical, non-injurious ischaemia applied remote from the heart that protects the heat from lethal ischaemia-reperfusion injury. Accordingly, there is considerable interest in SDF-1α as a potential biomarker of such conditions. However, SDF-1α is rapidly degraded and inactivated by dipeptidyl peptidase 4 and other peptidases, and the kinetics of intact SDF-1α remain unknown. Methods & results To facilitate investigation of full-length SDF-1α we established an ELISA using a novel recombinant human antibody we developed called HCI.SDF1. HCI.SDF1 is specific to the N-terminal sequence of all isoforms of SDF-1 and has a comparable KD to commercially available antibodies. Together with a detection antibody specific to the α-isoform, HCI.SDF1 was used to specifically quantify full-length SDF-1α in blood for the first time. Using RIC applied to the hind limb of Sprague-Dawley rats or the arms of healthy human volunteers, we demonstrate an increase in SDF-1α using a commercially available antibody, as previously reported, but an unexpected decrease in full-length SDF-1α after RIC in both species. Conclusions We report for the first time the development of a novel recombinant antibody specific to full-length SDF-1. Applied to RIC, we demonstrate a significant decrease in SDF-1α that is at odds with the literature and suggests a need to investigate the kinetics of full-length SDF-1α in conditions characterised by tissue hypoxia.
Collapse
Affiliation(s)
- Daniel I. Bromage
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Stasa Taferner
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Mahesh Pillai
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Derek M. Yellon
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Sean M. Davidson
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
14
|
Mekonnen G, Hayek SS, Mehta PK, Li Q, Mahar E, Mou L, Kenkre TS, Petersen JW, Azarbal B, Samuels B, Anderson RD, Sedlak T, Zaya M, Agarwal M, Haftbaradaran A, Minissian M, Handberg E, Pepine CJ, Cogle CR, Bairey Merz CN, Waller EK, Quyyumi AA. Circulating progenitor cells and coronary microvascular dysfunction: Results from the NHLBI-sponsored Women's Ischemia Syndrome Evaluation - Coronary Vascular Dysfunction Study (WISE-CVD). Atherosclerosis 2016; 253:111-117. [PMID: 27596135 PMCID: PMC9197325 DOI: 10.1016/j.atherosclerosis.2016.08.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 08/14/2016] [Accepted: 08/23/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS Ischemia stimulates a reparative response resulting in mobilization of circulating progenitor cells (CPCs). We hypothesized that women with chronic myocardial ischemia from coronary microvascular disease (CMD) will mobilize CPCs. METHODS In 123 women with ischemic symptoms and signs but no obstructive coronary artery disease (CAD) enrolled in the Women's Ischemia Syndrome Evaluation - Coronary Vascular Dysfunction Study (WISE-CVD), we measured coronary flow reserve (CFR) in response to intracoronary adenosine. Peripheral blood CPCs were measured using flow cytometry for expression of CD34, CD133, CXCR4, and VEGFR2. RESULTS Subjects were 53 ± 11 years, BMI 30 ± 8; 44% hypertensive, 11% diabetic, 23% hyperlipidemic and 7% smokers. Lower CFR correlated inversely with higher levels of hematopoietic-enriched CD34+ (r = -0.23, p = 0.011), CD34+/CD133+ (r = -0.24, p = 0.008), and CD34+/CXCR4+ (r = -0.19, p = 0.036) cells. In multivariable regression analyses, after adjusting for traditional cardiovascular risk factors, lower CFR remained significantly associated with elevated levels of CD34+ (β -0.18, p = 0.042), CD34+/CD133+ (β -0.24, p = 0.036), and CD34+/CXCR4+ (β -0.22, p = 0.050) cells. We found no association between CFR and CD34+/VEGFR2+ cells. CONCLUSIONS In women with non-obstructive CAD, impaired CFR is associated with higher levels of CPCs, suggesting that chronic myocardial ischemia from CMD stimulates CPC mobilization. The functional significance of elevated CPCs in these subjects requires further investigation as a potential biomarker and treatment target.
Collapse
Affiliation(s)
| | - Salim S Hayek
- Emory University School of Medicine, Atlanta, GA, USA
| | - Puja K Mehta
- Barbra Streisand Women's Heart Center, Cedars-Sinai Heart Institute, Los Angeles, CA, USA
| | - Qunna Li
- Emory University School of Medicine, Atlanta, GA, USA
| | | | - Liping Mou
- Emory University School of Medicine, Atlanta, GA, USA
| | | | | | - Babak Azarbal
- Barbra Streisand Women's Heart Center, Cedars-Sinai Heart Institute, Los Angeles, CA, USA
| | - Bruce Samuels
- Barbra Streisand Women's Heart Center, Cedars-Sinai Heart Institute, Los Angeles, CA, USA
| | | | | | - Melody Zaya
- Barbra Streisand Women's Heart Center, Cedars-Sinai Heart Institute, Los Angeles, CA, USA
| | - Megha Agarwal
- Barbra Streisand Women's Heart Center, Cedars-Sinai Heart Institute, Los Angeles, CA, USA
| | - Afsaneh Haftbaradaran
- Barbra Streisand Women's Heart Center, Cedars-Sinai Heart Institute, Los Angeles, CA, USA
| | - Margo Minissian
- Barbra Streisand Women's Heart Center, Cedars-Sinai Heart Institute, Los Angeles, CA, USA
| | | | | | | | - C Noel Bairey Merz
- Barbra Streisand Women's Heart Center, Cedars-Sinai Heart Institute, Los Angeles, CA, USA
| | | | | |
Collapse
|
15
|
Al Mheid I, Hayek SS, Ko YA, Akbik F, Li Q, Ghasemzadeh N, Martin GS, Long Q, Hammadah M, Maziar Zafari A, Vaccarino V, Waller EK, Quyyumi AA. Age and Human Regenerative Capacity Impact of Cardiovascular Risk Factors. Circ Res 2016; 119:801-9. [PMID: 27436845 DOI: 10.1161/circresaha.116.308461] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/19/2016] [Indexed: 01/11/2023]
Abstract
RATIONALE We investigated aging of human endogenous reparative capacity and aimed to clarify whether it is affected by presence of cardiovascular disease or its risk factors (RFs). OBJECTIVE Circulating progenitor cell (PC) levels reflect endogenous regenerative potential. The effect on PC of healthy aging compared with aging with RFs or cardiovascular disease (CVD) is unknown. We examined whether exposure to RF and CVD leads to an accelerated decline in circulating PC with increasing age. METHODS AND RESULTS In 2792 adult subjects, 498 were free of RFs (smoking, diabetes mellitus, hypertension, or hyperlipidemia), 1036 subjects had 1 to 2 RF, and 1253 had ≥3 RFs or CVD. PC were enumerated by flow cytometry as CD45(med+) mononuclear cells expressing CD34 and subsets coexpressing CD133, CXCR4, and vascular endothelial growth factor receptor-2 epitopes. Younger age, male sex, and larger body size correlated with higher PC counts (P<0.01). After multivariable adjustment, both age and RF categories were independently associated with PC counts (P<0.05), with lower PC counts in older subjects and those with higher RF burden or CVD. PC counts remained unchanged with increasing age in healthy individuals. There were significant interactions between age and RF categories (P≤0.005), such that for younger subjects (<40 years), RFs were associated with increased PC counts, whereas for older subjects (>60 years), RFs and CVD were associated with lower PC counts. CONCLUSIONS Circulating PC levels do not decline with healthy aging; RF exposure at a younger age stimulates PC mobilization, whereas continued exposure is associated with lower PC levels in later life. Over the lifespan, exposure to RFs and CVD is associated with an initial stimulation and subsequent decline in circulating PC levels, which reflect endogenous regenerative capacity.
Collapse
Affiliation(s)
- Ibhar Al Mheid
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Salim S Hayek
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Yi-An Ko
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Faysal Akbik
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Qunna Li
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Nima Ghasemzadeh
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Greg S Martin
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Qi Long
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Muhammad Hammadah
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - A Maziar Zafari
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Viola Vaccarino
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Edmund K Waller
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA
| | - Arshed A Quyyumi
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory-Georgia Tech, Predictive Health Institute, Atlanta, GA.
| |
Collapse
|
16
|
Kim BS, Jacobs D, Emontzpohl C, Goetzenich A, Soppert J, Jarchow M, Schindler L, Averdunk L, Kraemer S, Marx G, Bernhagen J, Pallua N, Schlemmer HP, Simons D, Stoppe C. Myocardial Ischemia Induces SDF-1α Release in Cardiac Surgery Patients. J Cardiovasc Transl Res 2016; 9:230-238. [PMID: 27055858 DOI: 10.1007/s12265-016-9689-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 03/22/2016] [Indexed: 01/07/2023]
Abstract
In the present observational study, we measured serum levels of the chemokine stromal cell-derived factor-1α (SDF-1α) in 100 patients undergoing cardiac surgery with cardiopulmonary bypass at seven distinct time points including preoperative values, myocardial ischemia, reperfusion, and the postoperative course. Myocardial ischemia triggered a marked increase of SDF-1α serum levels whereas cardiac reperfusion had no significant influence. Perioperative SDF-1α serum levels were influenced by patients' characteristics (e.g., age, gender, aspirin intake). In an explorative analysis, we observed an inverse association between SDF-1α serum levels and the incidence of organ dysfunction. In conclusion, time of myocardial ischemia was identified as the key stimulus for a significant upregulation of SDF-1α, indicating its role as a marker of myocardial injury. The inverse association between SDF-1α levels and organ dysfunction association encourages further studies to evaluate its organoprotective properties in cardiac surgery patients.
Collapse
Affiliation(s)
- Bong-Sung Kim
- Department of Plastic Surgery, Hand Surgery, Burn Center, RWTH Aachen University, Aachen, Germany
| | - Denise Jacobs
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Christoph Emontzpohl
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Andreas Goetzenich
- Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany
| | - Josefin Soppert
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Mareike Jarchow
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Lisa Schindler
- Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany.,Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany
| | - Luisa Averdunk
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Sandra Kraemer
- Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany
| | - Gernot Marx
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Jürgen Bernhagen
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Norbert Pallua
- Department of Plastic Surgery, Hand Surgery, Burn Center, RWTH Aachen University, Aachen, Germany
| | | | - David Simons
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Christian Stoppe
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany.
| |
Collapse
|
17
|
Leone AM, D'Amario D, Teofili L, Basile E, Cannata F, Graziani F, Marzilli M, Russo AM, Tarantini G, Ceconi C, Leone G, Trani C, Rebuzzi AG, Crea F. The combined effect of subcutaneous granulocyte- colony stimulating factor and myocardial contrast echocardiography with intravenous infusion of sulfur hexafluoride on post-infarction left ventricular function, the RIGENERA 2.0 trial: study protocol for a randomized controlled trial. Trials 2016; 17:97. [PMID: 26891753 PMCID: PMC4759781 DOI: 10.1186/s13063-016-1172-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 01/12/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Several clinical trials and recent meta-analyses have demonstrated that administration of recombinant human granulocyte-colony stimulating factor (G-CSF) is safe and, only in patients with large acute myocardial infarction (AMI), is associated with an improvement in left ventricular ejection fraction. Moreover, the mobilization and engraftment of the bone marrow-derived cells may differ significantly among patients, interfering with the restoration of left ventricular function after treatment. Therefore, the clinical potential application of the G-CSF has not yet been fully elucidated. METHODS/DESIGN The RIGENERA 2.0 trial is a multicenter, phase II, placebo-controlled, randomized, open-label, with blinded evaluation of endpoints (PROBE) trial in which 120 patients with an acute ST-elevation myocardial infarction (STEMI) undergoing successful revascularization but with residual myocardial dysfunction will be enrolled. In cases where there is a left ventricular ejection fraction (LVEF) ≤ 45% the patient will be electronically randomized (1:1 ratio) to receive either subcutaneous recombinant human G-CSF (group 1) or placebo (group 2) both added on top of optimal standard of care. Both groups will undergo myocardial contrast echocardiography with intravenous infusion of sulfur hexafluoride (MCE) whilst undergoing the echocardiogram. The primary efficacy endpoint is the evaluation of the LVEF at 6 months after AMI assessed by cardiac magnetic resonance. Secondary efficacy endpoints are the evaluation of LVEF at 6 months after AMI assessed by echocardiography, left ventricular end-diastolic volume (LVEDV) and left ventricular end-systolic volume (LVESV) assessed by cardiac magnetic resonance and echocardiography at 6 months, together with the incidence of major adverse clinical events (MACE) defined as death, myocardial infarction, sustained cardiac arrhythmias, cardiogenic shock, stroke and re-hospitalization due to heart failure at 1 year. DISCUSSION The RIGENERA 2.0 trial will test whether G-CSF administration and MCE, through the enhancement of the bone marrow-derived cells homing in the myocardium, determines an improvement in regional and global contractile function, myocardial perfusion and infarct extension in patients with large AMI. The results of the present study are expected to envision routine clinical use of this safe, affordable and reproducible approach in patients with successful revascularization after AMI. TRIAL REGISTRATION ClinicalTrials.gov: NCT02502747 (29 June 2015); EudraCT: 2015-002189-21 (10 July 2015).
Collapse
Affiliation(s)
- Antonio Maria Leone
- Dipartimento di Scienze Cardiovascolari, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli, 8, Rome, 00168, Italy.
| | - Domenico D'Amario
- Dipartimento di Scienze Cardiovascolari, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli, 8, Rome, 00168, Italy.
| | - Luciana Teofili
- Istituto di Ematologia, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli, 8, Rome, 00168, Italy.
| | - Eloisa Basile
- Dipartimento di Scienze Cardiovascolari, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli, 8, Rome, 00168, Italy.
| | - Francesco Cannata
- Dipartimento di Scienze Cardiovascolari, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli, 8, Rome, 00168, Italy
| | - Francesca Graziani
- Dipartimento di Scienze Cardiovascolari, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli, 8, Rome, 00168, Italy.
| | - Mario Marzilli
- Dipartimento Cardio-Toraco-Vascolare, Università di Pisa, Via Paradisa, 2, Pisa, 56124, Italy.
| | | | - Giuseppe Tarantini
- Dipartimento di Scienze Cardiologiche Toraciche e Vascolari, Università di Padova, Via Giustiniani, 2, Padova, 35128, Italy.
| | - Claudio Ceconi
- Dipartimento di Medicina Clinica e Sperimentale, Università di Ferrara, Corso Giovecca, 203, Ferrara, 44100, Italy.
| | - Giuseppe Leone
- Istituto di Ematologia, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli, 8, Rome, 00168, Italy.
| | - Carlo Trani
- Dipartimento di Scienze Cardiovascolari, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli, 8, Rome, 00168, Italy.
| | - Antonio Giuseppe Rebuzzi
- Dipartimento di Scienze Cardiovascolari, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli, 8, Rome, 00168, Italy.
| | - Filippo Crea
- Dipartimento di Scienze Cardiovascolari, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli, 8, Rome, 00168, Italy.
| |
Collapse
|
18
|
Gallina C, Capelôa T, Saviozzi S, Accomasso L, Catalano F, Tullio F, Martra G, Penna C, Pagliaro P, Turinetto V, Giachino C. Human mesenchymal stem cells labelled with dye-loaded amorphous silica nanoparticles: long-term biosafety, stemness preservation and traceability in the beating heart. J Nanobiotechnology 2015; 13:77. [PMID: 26510588 PMCID: PMC4625930 DOI: 10.1186/s12951-015-0141-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 10/22/2015] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Treatment of myocardial infarction with mesenchymal stem cells (MSCs) has proven beneficial effects in both animal and clinical studies. Engineered silica nanoparticles (SiO2-NPs) have been extensively used as contrast agents in regenerative medicine, due to their resistance to degradation and ease of functionalization. However, there are still controversies on their effective biosafety on cellular systems. In this perspective, the aims of the present study are: 1) to deeply investigate the impact of amorphous 50 nm SiO2-NPs on viability and function of human bone marrow-derived MSCs (hMSCs); 2) to optimize a protocol of harmless hMSCs labelling and test its feasibility in a beating heart model. RESULTS Optimal cell labelling is obtained after 16 h exposure of hMSCs to fluorescent 50 nm SiO2-NPs (50 µg mL(-1)); interestingly, lysosomal activation consequent to NPs storage is not associated to oxidative stress. During prolonged culture hMSCs do not undergo cyto- or genotoxicity, preserve their proliferative potential and their stemness/differentiation properties. Finally, the bright fluorescence emitted by internalized SiO2-NPs allows both clear visualization of hMSCs in normal and infarcted rat hearts and ultrastructural analysis of cell engraftment inside myocardial tissue. CONCLUSIONS Overall, 50 nm SiO2-NPs display elevated compatibility with hMSCs in terms of lack of cyto- and genotoxicity and maintenance of important features of these cells. The demonstrated biosafety, combined with proper cell labelling and visualization in histological sections, make these SiO2-NPs optimal candidates for the purpose of stem cell tracking inside heart tissue.
Collapse
Affiliation(s)
- Clara Gallina
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy.
| | - Tânia Capelôa
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy.
| | - Silvia Saviozzi
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy.
| | - Lisa Accomasso
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy.
| | - Federico Catalano
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy. .,Department of Chemistry, Interdepartmental Centre "Nanostructured Interfaces and Surfaces", University of Turin, 7, Via P. Giuria, CAP 10125, Turin, Italy.
| | - Francesca Tullio
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy.
| | - Gianmario Martra
- Department of Chemistry, Interdepartmental Centre "Nanostructured Interfaces and Surfaces", University of Turin, 7, Via P. Giuria, CAP 10125, Turin, Italy.
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy.
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy.
| | - Valentina Turinetto
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy.
| | - Claudia Giachino
- Department of Clinical and Biological Sciences, University of Turin, 10, Regione Gonzole, CAP 10043, Orbassano, TO, Italy.
| |
Collapse
|
19
|
Achilli F, Malafronte C, Cesana F, Maggiolini S, Mauro C, De Ferrari GM, Lenatti L, Tespili M, Pasqualini P, Gentile F, Capogrossi MC, Maggioni A, Maseri A, Pontone G, Colombo GI, Pompilio G. Granulocyte-colony stimulating factor for large anterior ST-elevation myocardial infarction: rationale and design of the prospective randomized phase III STEM-AMI OUTCOME trial. Am Heart J 2015; 170:652-658.e7. [PMID: 26386788 DOI: 10.1016/j.ahj.2015.07.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 07/02/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Granulocyte-colony stimulating factor (G-CSF) has been clinically tested in ST-elevation myocardial infarction (STEMI) with mixed results. Our 3-year follow-up data from STEM-AMI trial documented a sustained benefit of G-CSF on adverse ventricular remodeling after large anterior STEMI, when administered early and at a high-dose in patients with left ventricular (LV) dysfunction. The Aim of the present trial is to establish whether G-CSF improves hard clinical long-term outcomes. METHODS The STEM-AMI OUTCOME is a prospective, multicenter, randomized, open-label, phase III trial. It will include 1,530 patients with anterior STEMI undergoing primary percutaneous coronary intervention 2 to 24 hours after symptoms onset and with LV ejection fraction ≤45% after successful reperfusion. Patients will be randomized 1:1 to G-CSF and/or standard treatment. The primary end point is a reduced occurrence of all-cause death, recurrence of myocardial infarction, or hospitalization due to heart failure in G-CSF-treated patients. Left ventricular remodeling will be assessed via cardiac ultrasound and a substudy with cardiac magnetic resonance will be carried out in 120 subjects. Accrual and follow-up periods will last 3 and 2 years, respectively. CONCLUSIONS The STEM-AMI OUTCOME study is designed to be a rigorous controlled phase III trial with adequate statistical power to conclusively assess efficacy of G-CSF treatment in STEMI.
Collapse
Affiliation(s)
- Felice Achilli
- Cardiology Unit and Intensive and Coronary Care Unit, San Gerardo Hospital, Monza, Italy.
| | - Cristina Malafronte
- Cardiology Unit and Intensive and Coronary Care Unit, San Gerardo Hospital, Monza, Italy
| | - Francesca Cesana
- Cardiology Unit and Intensive and Coronary Care Unit, San Gerardo Hospital, Monza, Italy
| | | | - Ciro Mauro
- Department of Cardiology, Cardarelli Hospital, Napoli, Italy
| | - Gaetano M De Ferrari
- Cardiology Unit and Intensive and Coronary Care Unit, Fondazione Policlinico San Matteo IRCCS, Pavia, Italy
| | - Laura Lenatti
- Department of Cardiology, Alessandro Manzoni Hospital, Lecco, Italy
| | | | | | - Francesco Gentile
- Department of Cardiology, Bassini Hospital, Cinisello Balsamo, Italy
| | - Maurizio C Capogrossi
- Laboratory of Vascular Pathology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| | - Aldo Maggioni
- Research Center of the Italian Association of Hospital Cardiologists (ANMCO), Florence, Italy
| | | | - Gianluca Pontone
- Department of Cardiovascular Imaging, Cardiac Magnetic Resonance Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Gualtiero I Colombo
- Immunology and Functional Genomics Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Giulio Pompilio
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, and Cardiovascular Section, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
20
|
Immunologic Network and Response to Intramyocardial CD34+ Stem Cell Therapy in Patients With Dilated Cardiomyopathy. J Card Fail 2015; 21:572-82. [PMID: 25863169 DOI: 10.1016/j.cardfail.2015.03.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 01/28/2015] [Accepted: 03/30/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Although stem cell therapy (SCT) is emerging as a potential treatment for patients with dilated cardiomyopathy (DCM), clinical response remains variable. Our objective was to determine whether baseline differences in circulating immunologic and nonimmunologic biomarkers may help to identify patients more likely to respond to intramyocardial injection of CD34(+)-based SCT. METHODS AND RESULTS We enrolled from January 3, 2011 to March 5, 2012 37 patients with longstanding DCM (left ventricular ejection fraction [LVEF] <40%, New York Heart Association functional class III) who underwent peripheral CD34(+) stem cell mobilization with granulocyte colony-stimulating factor (G-CSF) and collection by means of apheresis. CD34(+) cells were labeled with (99m)Tc-hexamethylpropyleneamine oxime to allow assessment of stem cell retention at 18 hours. Response to SCT was predefined as an increase in LVEF of ≥5% at 3 months. The majority (84%) of patients were male with an overall mean LVEF of 27 ± 7% and a median N-terminal pro-B-type natriuretic peptide (NT-proBNP) level of 2,774 pg/mL. Nineteen patients (51%) were responders to SCT. There was no significant difference between responders and nonresponders regarding to age, sex, baseline LVEF, NT-proBNP levels, or 6-minute walking distance. With the use of a partial least squares (PLS) predictive model, we identified 9 baseline factors that were associated with both stem cell response and stem cell retention (mechanistic validation). Among the baseline factors positively associated with both clinical response and stem cell retention were G-CSF, SDF-1, LIF, MCP-1, and MCP-3. Among baseline factors negatively associated with both clinical response and retention were IL-12p70, FASL, ICAM-1, and GGT. A decrease in G-CSF at 3-month follow-up was also observed in responders compared with nonresponders (P = .02). CONCLUSIONS If further validated, baseline immunologic and nonimmunologic biomarkers may help to identify patients with DCM who are more likely to respond to CD34(+)-based SCT.
Collapse
|
21
|
Michelucci A, Cesari F, Ricciardi G, Attanà P, Pieragnoli P, Ristalli F, Padeletti L, Gori AM, Gensini GF, Abbate R. Left ventricular mass and progenitor cells in chronic heart failure patients. Intern Emerg Med 2015; 10:329-35. [PMID: 25387824 DOI: 10.1007/s11739-014-1149-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 10/27/2014] [Indexed: 01/19/2023]
Abstract
The aim of the study was to evaluate the association between circulating (CPCs) and endothelial (EPCs) progenitor cells and left ventricular (LV) remodeling in chronic heart failure (HF). 85 HF patients, ranging 29-89 years, 83.5% males, 45.9% ischemic, NYHA functional class II-IV, with a LV ejection fraction ≤40% were studied. LV ejection fraction, LV end-diastolic and end-systolic (LVESV) volumes, LV mass and tricuspid annular plane systolic excursion (TAPSE) were evaluated, and, when indicated, indexed for body surface area (BSA). CPCs and EPCs number was assessed using flow cytometry. CPCs were defined as CD34+, CD133+ and CD34+/CD133+. EPCs, identified through their expression of KDR, were defined as CD34+/KDR+, CD133+/KDR+ and CD34+/CD133+/KDR+. All EPCs were negatively related to LVESV/BSA (r = -0.24, p = 0.02 for all EPC's populations), and to LVmass/BSA (CD34+KDR+; r = -0.30, p = 0.005; CD133+KDR+; r = -0.31, p = 0.004; CD34+CD133+KDR+; r = -0.29, p = 0.007). No differences in EPCs levels in relation to cardiovascular risk factors, medications, etiology, age or gender were observed. CPCs number was higher in women, and lower in ischemic patients. In logistic regression analyses, the low EPCs' number was associated with an increased likelihood of abnormal LVmass/BSA. CPCs proved to be higher and EPCs lower in patients with severely abnormal LVmass/BSA (gr/m(2), ≥122 in women and ≥149 in men). Our results suggest a correlation between LV remodeling and progenitor cells. This is noteworthy considering that it has been suggested that bone marrow-derived EPCs participate in cardiac regeneration and function recovery in the setting of progressive HF.
Collapse
Affiliation(s)
- Antonio Michelucci
- Section of Arrhythmology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134, Florence, Italy,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Improvement of endurance of DMD animal model using natural polyphenols. BIOMED RESEARCH INTERNATIONAL 2015; 2015:680615. [PMID: 25861640 PMCID: PMC4377377 DOI: 10.1155/2015/680615] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/13/2014] [Accepted: 08/27/2014] [Indexed: 12/11/2022]
Abstract
Duchenne muscular dystrophy (DMD), the most common form of muscular dystrophy, is characterized by muscular wasting caused by dystrophin deficiency that ultimately ends in force reduction and premature death. In addition to primary genetic defect, several mechanisms contribute to DMD pathogenesis. Recently, antioxidant supplementation was shown to be effective in the treatment of multiple diseases including muscular dystrophy. Different mechanisms were hypothesized such as reduced hydroxyl radicals, nuclear factor-κB deactivation, and NO protection from inactivation. Following these promising evidences, we investigated the effect of the administration of a mix of dietary natural polyphenols (ProAbe) on dystrophic mdx mice in terms of muscular architecture and functionality. We observed a reduction of muscle fibrosis deposition and myofiber necrosis together with an amelioration of vascularization. More importantly, the recovery of the morphological features of dystrophic muscle leads to an improvement of the endurance of treated dystrophic mice. Our data confirmed that ProAbe-based diet may represent a strategy to coadjuvate the treatment of DMD.
Collapse
|
23
|
Seropian IM, Sonnino C, Van Tassell BW, Biasucci LM, Abbate A. Inflammatory markers in ST-elevation acute myocardial infarction. EUROPEAN HEART JOURNAL-ACUTE CARDIOVASCULAR CARE 2015; 5:382-95. [PMID: 25681486 DOI: 10.1177/2048872615568965] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 01/02/2015] [Indexed: 01/05/2023]
Abstract
After acute myocardial infarction, ventricular remodeling is characterized by changes at the molecular, structural, geometrical and functional level that determine progression to heart failure. Inflammation plays a key role in wound healing and scar formation, affecting ventricular remodeling. Several, rather different, components of the inflammatory response were studied as biomarkers in ST-elevation acute myocardial infarction. Widely available and inexpensive tests, such as leukocyte count at admission, as well as more sophisticated immunoassays provide powerful predictors of adverse outcome in patients with ST-elevation acute myocardial infarction. We review the value of inflammatory markers in ST-elevation acute myocardial infarction and their association with ventricular remodeling, heart failure and sudden death. In conclusion, the use of these biomarkers may identify subjects at greater risk of adverse events and perhaps provide an insight into the mechanisms of disease progression.
Collapse
Affiliation(s)
- Ignacio M Seropian
- Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Argentina
| | - Chiara Sonnino
- VCU Pauley Heart Center, Virginia Commonwealth University, USA Victoria Johnson Research Laboratory, Virginia Commonwealth University, USA Department of Cardiovascular Medicine, Catholic University, Italy
| | - Benjamin W Van Tassell
- VCU Pauley Heart Center, Virginia Commonwealth University, USA Victoria Johnson Research Laboratory, Virginia Commonwealth University, USA School of Pharmacy, Virginia Commonwealth University, USA
| | - Luigi M Biasucci
- Department of Cardiovascular Medicine, Catholic University, Italy
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, USA Victoria Johnson Research Laboratory, Virginia Commonwealth University, USA
| |
Collapse
|
24
|
Chen Y, Lu B, Wang J, Chen S, Lin Z, Ma X, Liu Y, Zhao B, Chen Y. Circulating CD133+ CD34+ progenitor cells and plasma stromal-derived factor-1alpha: predictive role in ischemic stroke patients. J Stroke Cerebrovasc Dis 2014; 24:319-26. [PMID: 25444027 DOI: 10.1016/j.jstrokecerebrovasdis.2014.08.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/20/2014] [Accepted: 08/25/2014] [Indexed: 11/28/2022] Open
Abstract
Circulating progenitor cells and stromal-derived factor-1alpha (SDF-1α) have been suggested to participate in tissue repair after ischemic injury. However, the predictive role of circulating CD133+ CD34+ progenitors and plasma SDF-1α in ischemic stroke (IS) patients remains unknown. In this study, we recruited 95 acute IS patients, 40 at-risk subjects, and 30 normal subjects. The National Institutes of Health Stroke Scale (NIHSS), infarct volume, and carotid intima-media thickness (IMT) were determined at day 1 and the modified Rankin scale (mRS) of functional outcome was assessed at day 21. The levels of circulating CD133+ CD34+ cells and plasma SDF-1α were determined by flow cytometry and enzyme-linked immunosorbent assay, respectively. Our data showed that: (1) the levels of CD133+ CD34+ cells were lower in at-risk subjects and IS patients at admission (day 1) when compared with normal controls; (2) the day 1 level of CD133+ CD34+ cells varied in IS subgroups and inversely correlated with NIHSS and carotid IMT and the level of SDF-1α inversely correlated with NIHSS and infarct volume; (3) the increment rates of circulating CD133+ CD34+ cells and plasma SDF-1α within the first week were correlated; and (4) patients with a higher level of CD133+ CD34+ cells at day 7 had a low mRS. The increased rate of CD133+ CD34+ cells in the first week was inversely associated with mRS. In conclusion, our findings demonstrate that the circulating CD133+ CD34+ progenitor cells and plasma SDF-1α can be used as predictive parameters for IS severity and outcome.
Collapse
Affiliation(s)
- Yusen Chen
- Clinical Research Center and Department of Neurology, The Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, China
| | - Bing Lu
- Clinical Research Center and Department of Neurology, The Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, China
| | - Jinju Wang
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio
| | - Shuzhen Chen
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio
| | - Zhijun Lin
- Clinical Research Center and Department of Neurology, The Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, China
| | - Xiaotang Ma
- Clinical Research Center and Department of Neurology, The Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, China
| | - Yajing Liu
- Clinical Research Center and Department of Neurology, The Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, China
| | - Bin Zhao
- Clinical Research Center and Department of Neurology, The Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, China.
| | - Yanfang Chen
- Clinical Research Center and Department of Neurology, The Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, China; Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio.
| |
Collapse
|
25
|
Piatkowski A, Grieb G, Simons D, Bernhagen J, van der Hulst RR. Endothelial progenitor cells--potential new avenues to improve neoangiogenesis and reendothelialization. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 306:43-81. [PMID: 24016523 DOI: 10.1016/b978-0-12-407694-5.00002-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The term endothelial progenitor cell (EPC) was established more than 10 years ago and is used to refer to a group of circulating cells that display endothelial lineage qualities and are able to home to areas of ischemia or vascular injury and to facilitate the repair of damaged blood vessels or develop new vessels as needed. This chapter reviews the current lineage relationships among all the cells called EPC and will clear the terminology used in EPC research. Furthermore, an overview of the clinical and in vitro research, as well as cytokine and drug interactions and potential EPC applications, is given.
Collapse
Affiliation(s)
- Andrzej Piatkowski
- Department of Plastic Surgery, academisch ziekenhuis Maastricht, MUMC+, Maastricht, The Netherlands.
| | | | | | | | | |
Collapse
|
26
|
Elevated plasma stromal-cell-derived factor-1 protein levels correlate with severity in patients with community-acquired pneumonia. DISEASE MARKERS 2014; 2014:829706. [PMID: 25371597 PMCID: PMC4211303 DOI: 10.1155/2014/829706] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 09/02/2014] [Accepted: 09/18/2014] [Indexed: 11/17/2022]
Abstract
BACKGROUND The aim of this study was to investigate differential changes in plasma levels of stromal-cell-derived factor-1 (SDF-1) before and after antibiotic treatment in patients with community-acquired pneumonia (CAP) and observe the association between the severity of CAP and the plasma SDF-1 level. METHODS We gathered blood specimens from 61 adult CAP patients before and after antibiotic treatment and from 60 healthy controls to measure the plasma concentrations of SDF-1 by using an enzyme-linked immunosorbent assay. RESULTS The plasma SDF-1 concentration was elevated significantly in patients with CAP before receiving treatment compared with the controls and decreased significantly after the patients received treatment. Leukocyte (WBC) and neutrophil counts and C-reactive protein (CRP) levels decreased significantly after antibiotic treatment. Moreover, differences in the plasma concentration of SDF-1 were significantly correlated with PSI, CURB-65, and APACHE II scores (r = 0.389, P = 0.002, and n = 61; r = 0.449, P < 0.001, and n = 61; and r = 0.363, P = 0.004, and n = 61, resp.). CONCLUSIONS An elevated plasma SDF-1 concentration can be used as a biological marker for the early diagnosis of CAP and for the early detection of its severity.
Collapse
|
27
|
Bromage DI, Davidson SM, Yellon DM. Stromal derived factor 1α: a chemokine that delivers a two-pronged defence of the myocardium. Pharmacol Ther 2014; 143:305-15. [PMID: 24704323 PMCID: PMC4127789 DOI: 10.1016/j.pharmthera.2014.03.009] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 03/20/2014] [Indexed: 01/03/2023]
Abstract
Alleviating myocardial injury associated with ST elevation myocardial infarction is central to improving the global burden of coronary heart disease. The chemokine stromal cell-derived factor 1α (SDF-1α) has dual potential benefit in this regard. Firstly, SDF-1α is up-regulated in experimental and clinical studies of acute myocardial infarction (AMI) and regulates stem cell migration to sites of injury. SDF-1α delivery to the myocardium after AMI is associated with improved stem cell homing, angiogenesis, and left ventricular function in animal models, and improvements in heart failure and quality of life in humans. Secondly, SDF-1α may have a role in remote ischaemic conditioning (RIC), the phenomenon whereby non-lethal ischaemia–reperfusion applied to an organ or tissue remote from the heart protects the myocardium from lethal ischaemia–reperfusion injury (IRI). SDF-1α is increased in the serum of rats subjected to RIC and protects against myocardial IRI in ex vivo studies. Despite these potential pleiotropic effects, a limitation of SDF-1α is its short plasma half-life due to cleavage by dipeptidyl peptidase-4 (DPP-4). However, DPP-4 inhibitors increase the half-life of SDF-1α by preventing its degradation and are also protective against lethal IRI. In summary, SDF-1 potentially delivers a ‘two-pronged’ defence of the myocardium: acutely protecting it from IRI while simultaneously stimulating repair by recruiting stem cells to the site of injury. In this article we examine the evidence for acute and chronic cardioprotective roles of SDF-1α and discuss potential therapeutic manipulations of this mechanism with DPP-4 inhibitors to protect against lethal tissue injury in the clinical setting.
Collapse
Affiliation(s)
- Daniel I Bromage
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London WC1E 6HX, United Kingdom
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London WC1E 6HX, United Kingdom
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London WC1E 6HX, United Kingdom
| |
Collapse
|
28
|
Pfister O, Della Verde G, Liao R, Kuster GM. Regenerative therapy for cardiovascular disease. Transl Res 2014; 163:307-20. [PMID: 24378637 DOI: 10.1016/j.trsl.2013.12.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 11/04/2013] [Accepted: 12/05/2013] [Indexed: 11/25/2022]
Abstract
Recent insights into myocardial biology uncovered a hereto unknown regenerative capacity of the adult heart. The discovery of dividing cardiomyocytes and the identification and characterization of cardiac stem and progenitor cells with myogenic and angiogenic potential have generated new hopes that cardiac regeneration and repair might become a therapeutic option. During the past decade, multiple candidate cells have been proposed for cardiac regeneration, and their mechanisms of action in the myocardium have been explored. Initial clinical trials have focused on the use of bone marrow-derived cells to promote myocardial regeneration in ischemic heart disease and have yielded very mixed results, with no clear signs of clinically meaningful functional improvement. Although the efficiency of bona fide cardiomyocyte generation is generally low, stem cells delivered into the myocardium act mainly via paracrine mechanisms. More recent studies taking advantage of cardiac committed cells (eg, resident cardiac progenitor cells or primed cardiogenic mesenchymal stem cells) showed promising results in first clinical pilot trials. Also, transplantation of cardiomyogenic cells generated by induced pluripotent stem cells and genetic reprogramming of dividing nonmyocytes into cardiomyocytes may constitute attractive new regenerative approaches in cardiovascular medicine in the future. We discuss advantages and limitations of specific cell types proposed for cell-based therapy in cardiology and give an overview of the first clinical trials using this novel therapeutic approach in patients with cardiovascular disease.
Collapse
Affiliation(s)
- Otmar Pfister
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland; Division of Cardiology, University Hospital Basel, Basel, Switzerland.
| | - Giacomo Della Verde
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Ronglih Liao
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Gabriela M Kuster
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland; Division of Cardiology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
29
|
Eleuteri E, Di Stefano A, Vallese D, Gnemmi I, Pitruzzella A, Tarro Genta F, Delle Donne L, Cappello F, Ricciardolo FLM, Giannuzzi P. Fibrosis markers and CRIM1 increase in chronic heart failure of increasing severity. Biomarkers 2014; 19:214-21. [PMID: 24617547 DOI: 10.3109/1354750x.2014.896946] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Fibrosis suppressors/activators in chronic heart failure (CHF) is a topic of investigation. AIM To quantify serum levels of fibrosis regulators in CHF. METHODS ELISA tests were used to quantify fibrosis regulators, procollagen type-(PIP)I, (PIP)III, collagen-I, III, BMP1,2,3,7, SDF1α, CXCR4, fibulin 1,2,3, BMPER, CRIM1 and BAMBI in 66 CHF (NYHA class I, n = 9; II, n = 34; III n = 23), and in 14 controls. RESULTS In CHF, TGFβR2, PIPIII, SDF1α and CRIM1 were increased. PIPIII correlated with CRIM1. CONCLUSIONS The BMPs inhibitor CRIM1 is increased and correlates with higher levels of serum PIPIII showing an imbalance in favor of pro-fibrotic mechanisms in CHF.
Collapse
Affiliation(s)
- Ermanno Eleuteri
- Divisione di Cardiologia Riabilitativa e Laboratorio di Citoimmunopatologia Apparato Cardio-Respiratorio, Fondazione Salvatore Maugeri , IRCCS, Veruno, NO , Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Pfister O, Lorenz V, Oikonomopoulos A, Xu L, Häuselmann SP, Mbah C, Kaufmann BA, Liao R, Wodnar-Filipowicz A, Kuster GM. FLT3 Activation Improves Post-Myocardial Infarction Remodeling Involving a Cytoprotective Effect on Cardiomyocytes. J Am Coll Cardiol 2014; 63:1011-9. [DOI: 10.1016/j.jacc.2013.08.1647] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Revised: 08/01/2013] [Accepted: 08/26/2013] [Indexed: 12/16/2022]
|
31
|
Heo SC, Kwon YW, Jang IH, Jeong GO, Yoon JW, Kim CD, Kwon SM, Bae YS, Kim JH. WKYMVm-Induced Activation of Formyl Peptide Receptor 2 Stimulates Ischemic Neovasculogenesis by Promoting Homing of Endothelial Colony-Forming Cells. Stem Cells 2014; 32:779-90. [DOI: 10.1002/stem.1578] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Revised: 09/18/2013] [Accepted: 09/25/2013] [Indexed: 12/24/2022]
Affiliation(s)
- Soon Chul Heo
- Medical Research Center for Ischemic Tissue Regeneration; Yangsan Gyeongsangnam-do Republic of Korea
- Department of Physiology; School of Medicine; Pusan National University; Yangsan Gyeongsangnam-do Republic of Korea
| | - Yang Woo Kwon
- Medical Research Center for Ischemic Tissue Regeneration; Yangsan Gyeongsangnam-do Republic of Korea
- Department of Physiology; School of Medicine; Pusan National University; Yangsan Gyeongsangnam-do Republic of Korea
| | - Il Ho Jang
- Medical Research Center for Ischemic Tissue Regeneration; Yangsan Gyeongsangnam-do Republic of Korea
- Department of Physiology; School of Medicine; Pusan National University; Yangsan Gyeongsangnam-do Republic of Korea
| | - Geun Ok Jeong
- Medical Research Center for Ischemic Tissue Regeneration; Yangsan Gyeongsangnam-do Republic of Korea
- Department of Physiology; School of Medicine; Pusan National University; Yangsan Gyeongsangnam-do Republic of Korea
| | - Jung Won Yoon
- Medical Research Center for Ischemic Tissue Regeneration; Yangsan Gyeongsangnam-do Republic of Korea
- Department of Physiology; School of Medicine; Pusan National University; Yangsan Gyeongsangnam-do Republic of Korea
| | - Chi Dae Kim
- Medical Research Center for Ischemic Tissue Regeneration; Yangsan Gyeongsangnam-do Republic of Korea
- Department of Pharmacology; School of Medicine; Pusan National University; Yangsan Gyeongsangnam-do Republic of Korea
| | - Sang Mo Kwon
- Department of Physiology; School of Medicine; Pusan National University; Yangsan Gyeongsangnam-do Republic of Korea
| | - Yoe-Sik Bae
- Department of Biological Sciences; Sungkyunkwan University; Suwon Republic of Korea
| | - Jae Ho Kim
- Medical Research Center for Ischemic Tissue Regeneration; Yangsan Gyeongsangnam-do Republic of Korea
- Department of Physiology; School of Medicine; Pusan National University; Yangsan Gyeongsangnam-do Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital; Yangsan Gyeongsangnam-do Republic of Korea
| |
Collapse
|
32
|
Rubach M, Adelmann R, Haustein M, Drey F, Pfannkuche K, Xiao B, Koester A, Udink ten Cate FEA, Choi YH, Neef K, Fatima A, Hannes T, Pillekamp F, Hescheler J, Šarić T, Brockmeier K, Khalil M. Mesenchymal stem cells and their conditioned medium improve integration of purified induced pluripotent stem cell-derived cardiomyocyte clusters into myocardial tissue. Stem Cells Dev 2014; 23:643-53. [PMID: 24219308 DOI: 10.1089/scd.2013.0272] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Induced pluripotent stem cell-derived cardiomyocytes (iPS-CMs) might become therapeutically relevant to regenerate myocardial damage. Purified iPS-CMs exhibit poor functional integration into myocardial tissue. The aim of this study was to investigate whether murine mesenchymal stem cells (MSCs) or their conditioned medium (MScond) improves the integration of murine iPS-CMs into myocardial tissue. Vital or nonvital embryonic murine ventricular tissue slices were cocultured with purified clusters of iPS-CMs in combination with murine embryonic fibroblasts (MEFs), MSCs, or MScond. Morphological integration was assessed by visual scoring and functional integration by isometric force and field potential measurements. We observed a moderate morphological integration of iPS-CM clusters into vital, but a poor integration into nonvital, slices. MEFs and MSCs but not MScond improved morphological integration of CMs into nonvital slices and enabled purified iPS-CMs to confer force. Coculture of vital slices with iPS-CMs and MEFs or MSCs resulted in an improved electrical integration. A comparable improvement of electrical coupling was achieved with the cell-free MScond, indicating that soluble factors secreted by MSCs were involved in electrical coupling. We conclude that cells such as MSCs support the engraftment and adhesion of CMs, and confer force to noncontractile tissue. Furthermore, soluble factors secreted by MSCs mediate electrical coupling of purified iPS-CM clusters to myocardial tissue. These data suggest that MSCs may increase the functional engraftment and therapeutic efficacy of transplanted iPS-CMs into infarcted myocardium.
Collapse
Affiliation(s)
- Martin Rubach
- 1 Department of Pediatric Cardiology, University of Cologne , Cologne, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Yu CW, Choi SC, Hong SJ, Choi JH, Park CY, Kim JH, Park JH, Ahn CM, Lim DS. Cardiovascular event rates in patients with ST-elevation myocardial infarction were lower with early increases in mobilization of Oct4highNanoghigh stem cells into the peripheral circulation during a 4-year follow-up. Int J Cardiol 2013; 168:2533-9. [DOI: 10.1016/j.ijcard.2013.03.060] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 03/17/2013] [Indexed: 10/26/2022]
|
34
|
Fadini GP, Avogaro A. Diabetes impairs mobilization of stem cells for the treatment of cardiovascular disease. Int J Cardiol 2013. [DOI: 10.1016/j.ijcard.2012.10.089] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
35
|
Early Fibroblast Progenitor Cell Migration to the AngII-Exposed Myocardium Is Not CXCL12 or CCL2 Dependent as Previously Thought. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:459-69. [DOI: 10.1016/j.ajpath.2013.04.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 02/14/2013] [Accepted: 04/09/2013] [Indexed: 12/19/2022]
|
36
|
Intramyocardial transplantation of human adipose-derived stromal cell and endothelial progenitor cell mixture was not superior to individual cell type transplantation in improving left ventricular function in rats with myocardial infarction. Int J Cardiol 2013; 164:205-11. [DOI: 10.1016/j.ijcard.2011.06.128] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2011] [Revised: 05/07/2011] [Accepted: 06/27/2011] [Indexed: 11/20/2022]
|
37
|
Westerweel PE, Teraa M, Rafii S, Jaspers JE, White IA, Hooper AT, Doevendans PA, Verhaar MC. Impaired endothelial progenitor cell mobilization and dysfunctional bone marrow stroma in diabetes mellitus. PLoS One 2013; 8:e60357. [PMID: 23555959 PMCID: PMC3610687 DOI: 10.1371/journal.pone.0060357] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 02/26/2013] [Indexed: 01/04/2023] Open
Abstract
Background Circulating Endothelial Progenitor Cell (EPC) levels are reduced in diabetes mellitus. This may be a consequence of impaired mobilization of EPC from the bone marrow. We hypothesized that under diabetic conditions, mobilization of EPC from the bone marrow to the circulation is impaired –at least partly– due to dysfunction of the bone marrow stromal compartment. Methods Diabetes was induced in mice by streptozotocin injection. Circulating Sca-1+Flk-1+ EPC were characterized and quantified by flow cytometry at baseline and after mobilization with G-CSF/SCF injections. In vivo hemangiogenic recovery was tested by 5-FU challenge. Interaction within the bone marrow environment between CD34+ hematopoietic progenitor cells (HPC) and supporting stroma was assessed by co-cultures. To study progenitor cell–endothelial cell interaction under normoglycemic and hyperglycemic conditions, a co-culture model using E4Orf1-transfected human endothelial cells was employed. Results In diabetic mice, bone marrow EPC levels were unaffected. However, circulating EPC levels in blood were lower at baseline and mobilization was attenuated. Diabetic mice failed to recover and repopulate from 5-FU injection. In vitro, primary cultured bone marrow stroma from diabetic mice was impaired in its capacity to support human CFU-forming HPC. Finally, hyperglycemia hampered the HPC supportive function of endothelial cells in vitro. Conclusion EPC mobilization is impaired under experimental diabetic conditions and our data suggest that diabetes induces alterations in the progenitor cell supportive capacity of the bone marrow stroma, which could be partially responsible for the attenuated EPC mobilization and reduced EPC levels observed in diabetic patients.
Collapse
Affiliation(s)
- Peter E. Westerweel
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
- Howard Hughes Medical Institute, The Ansary Stem Cell Center for Regenerative Medicine, Weill Cornell Medical College, New York, New York, United States of America
- Department of Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Martin Teraa
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Vascular Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Shahin Rafii
- Howard Hughes Medical Institute, The Ansary Stem Cell Center for Regenerative Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Janneke E. Jaspers
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ian A. White
- Howard Hughes Medical Institute, The Ansary Stem Cell Center for Regenerative Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Andrea T. Hooper
- Howard Hughes Medical Institute, The Ansary Stem Cell Center for Regenerative Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Pieter A. Doevendans
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marianne C. Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
38
|
Domínguez-Franco A, González FJ, Rodríguez-Losada N, Marchal JA, Cabrera-Bueno F, Carrillo E, Gómez-Doblas JJ, Perán M, Alonso-Briales JH, Jiménez-Navarro MF, Aránega A, De Teresa Galván E. [Factors influencing mobilisation of endothelial progenitor cells and angiogenic cytokines after an extensive acute myocardial infarction]. Med Clin (Barc) 2012; 138:415-421. [PMID: 22197368 DOI: 10.1016/j.medcli.2011.05.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Revised: 05/26/2011] [Accepted: 05/31/2011] [Indexed: 10/14/2022]
Abstract
BACKGROUND AND OBJECTIVES Following an acute myocardial infarction (AMI), bone-marrow derived endothelial progenitor cells (EPC) are mobilised into the peripheral blood. Our aim was to examine the factors influencing this spontaneous cell mobilisation. PATIENTS AND METHODS In this study we analysed 47 patients with extensive AMI (left ventricular ejection fraction [LVEF] <50% by echocardiography during the first week post-AMI); we studied the peripheral blood EPC populations expressing CD133(+), CD34(+), KDR(+), CXCR4(+), as well as the cytokines VEGF (vascular endothelial growth factor), SDF-1 (stromal cell-derived factor 1) and TSP-1 (thrombospondin 1), measured on day 5±2.5 after AMI. RESULTS The extension of AMI (CPK peak) correlated with the number of CD133(+) mobilised cells: (r=0.40; P=.011). Patients who did not receive perfusion during the acute phase (34%) had more CD34(+)CXCR4(+) cells with a median (interquartile ranges) of 2,401 (498-7,004) vs. 999 (100-1,600), P=.048, and strong correlations between VEGF and CD133(+)CD34(+)KDR(+) (r=.84; P<.01) and SDF-1 and CD34(+)CXCR4(+) (r=.67; P<.01), and between these 2 cytokines (r=.57; P=.01). In the reperfused patients, the correlation between VEGF and CD133(+)CD34(+)KDR(+) was lower (r=.38; P=.03) and the correlation between SDF-1 and CD34(+)CXCR4(+) and VEGF disappeared. Multivariate analysis showed that a VEGF >7pg/mL (P<.01) predicted the mobilisation of CD133(+)CD34(+)KDR(+), whereas hypertension showed a trend (P=.055). Diabetes (P=.045) predicted the number of CD34(+)CXCR4(+), with reperfusion treatment showing a trend in this subpopulation (P=.054). CONCLUSIONS Mobilisation of progenitor cells after AMI is influenced by factors such as diabetes and the cytokine VEGF. Hypertension and reperfusion therapy during the acute phase also tend to influence the cell response.
Collapse
Affiliation(s)
- Antonio Domínguez-Franco
- Área del Corazón, Hospital Clínico Universitario Virgen de la Victoria, Málaga, Investigadores RECAVA, Málaga, España
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Carvalheiro T, Velada I, Valado A, Mendes F, Martinho A, António N, Gonçalves L, Providência L, Pais ML, Paiva A. Phenotypic and functional alterations on inflammatory peripheral blood cells after acute myocardial infarction. J Cardiovasc Transl Res 2012; 5:309-20. [PMID: 22528677 DOI: 10.1007/s12265-012-9365-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 04/04/2012] [Indexed: 01/25/2023]
Abstract
The frequency and function of T cells, monocytes, and dendritic cell subsets were investigated in 12 patients after acute myocardial infarction (AMI)-(T0), 1 month after the episode (T1), and in 12 healthy individuals (HG). The cell characterization and the functional studies were performed by flow cytometry and by RT-PCR, after cell sorting. The most important findings at T0 moment, when compared with T1 and HG, were: a decrease in the frequency of IL-2-producing T cells; a lower frequency of TNF-α- and IL-6-producing monocytes, myeloid dendritic cells, and CD14(-/low)CD16(+)DCs; and a lower TNF-α mRNA expression, after sorting these cells. Moreover, the regulatory function of Treg cells, at T0 moment, was upregulated, based on the FoxP3, CTLA-4, and TGF-β mRNA expression increase. The majority of these phenotypic and functional alterations disappeared at T1. Our data demonstrate that AMI induces a significant change in the immune system homeostasis.
Collapse
Affiliation(s)
- Tiago Carvalheiro
- Histocompatibility Centre of Coimbra, Edifício São Jerónimo, 4° Piso, Praceta Mota Pinto, 3001-301 Coimbra, Portugal
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Sanz-Ruiz R, Fernández-Avilés F. [Stimulating factors for cardiac repair: when the ischemic myocardium asks for help]. Med Clin (Barc) 2012; 138:435-7. [PMID: 22197361 DOI: 10.1016/j.medcli.2011.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 10/08/2011] [Accepted: 10/11/2011] [Indexed: 11/16/2022]
|
41
|
Plasma levels of stromal cell-derived factor-1 in patients with coronary artery disease: Effect of clinical presentation and cardiovascular risk factors. Atherosclerosis 2011; 219:913-6. [DOI: 10.1016/j.atherosclerosis.2011.09.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2011] [Revised: 09/13/2011] [Accepted: 09/13/2011] [Indexed: 11/20/2022]
|
42
|
Brunner S, Weinberger T, Huber BC, Segeth A, Zaruba MM, Theiss HD, Assmann G, Herbach N, Wanke R, Mueller-Hoecker J, Franz WM. The cardioprotective effects of parathyroid hormone are independent of endogenous granulocyte-colony stimulating factor release. Cardiovasc Res 2011; 93:330-9. [PMID: 22080594 DOI: 10.1093/cvr/cvr303] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AIMS Parathyroid hormone (PTH) administration after myocardial infarction (MI) is known to attenuate ischaemic cardiomyopathy. This effect mainly resulted from an increase in mobilization and homing of CD34+/CD45+ cells into the ischaemic myocardium. PTH-related stem cell mobilization was shown to be related to endogenous granulocyte-colony stimulating factor (G-CSF) release. The aim of our study is to determine the role of G-CSF on the cardioprotective effects of PTH. METHODS AND RESULTS G-CSF +/+ (C57BL/6) and G-CSF -/- mice were treated with PTH for 6 days after inducing a MI. The myocardial homing factor stromal cell-derived factor-1 (SDF-1) was analysed on day 2 with enzyme-linked immunosorbent assay. Stem cell populations in peripheral blood and hearts were examined by FACS on days 6 and 2, respectively. Cardiac function and immunohistochemistry were investigated on day 6 and day 30. PTH treatment resulted in a significant increase in CD45+/CD34+ cells in peripheral blood in G-CSF +/+ but not in G-CSF -/- mice. However, a significant increase in SDF-1 and enhanced migration of CD45+/CD34+ cells into the ischaemic myocardium was revealed after PTH administration in both G-CSF +/+ and G-CSF -/- mice. Enhanced stem cell homing was associated with improved cardiac function and post-MI survival after PTH treatment. Furthermore, infarct size, wall thickness, and neovascularization showed a significant improvement in both groups 30 days after MI. CONCLUSION The cardioprotective effects of PTH were shown to be independent of endogenous G-CSF release and therefore from stem cell mobilization. This puts more emphasis on the role of stem cell homing into ischaemic myocardium.
Collapse
Affiliation(s)
- Stefan Brunner
- Department of Internal Medicine I, Ludwig-Maximilians-University, Campus Grosshadern, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Baranski GM, Offin MD, Sifri ZC, Elhassan IO, Hannoush EJ, Alzate WD, Rameshwar P, Livingston DH, Mohr AM. β-blockade protection of bone marrow following trauma: the role of G-CSF. J Surg Res 2011; 170:325-31. [PMID: 21571320 PMCID: PMC3158299 DOI: 10.1016/j.jss.2011.03.059] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2011] [Revised: 02/24/2011] [Accepted: 03/18/2011] [Indexed: 01/03/2023]
Abstract
BACKGROUND Following severe trauma, there is a profound elevation of catecholamine that is associated with a persistent anemic state. We have previously shown that β-blockade (βB) prevents erythroid growth suppression and decreases hematopoietic progenitor cell (HPC) mobilization following injury. Under normal conditions, granulocyte colony stimulating factor (G-CSF) triggers the activation of matrix metalloprotease-9 (MMP-9), leading to the egress of progenitor cells from the bone marrow (BM). When sustained, this depletion of BM cellularity may contribute to BM failure. This study seeks to determine if G-CSF plays a role in the βB protection of BM following trauma. METHODS Male Sprague-Dawley rats were subjected to either unilateral lung contusion (LC) ± βB, hemorrhagic shock (HS) ± βB, or both LC/HS ± βB. Propranolol (βB) was given immediately following resuscitation. Animals were sacrificed at 3 and 24 h and HPC mobilization was assessed by evaluating BM cellularity and flow cytometric analysis of peripheral blood for HPCs. The concentration of G-CSF and MMP-9 was measured in plasma by ELISA. RESULTS BM cellularity is decreased at 3 h following LC, HS, and LC/HS. HS and LC/HS resulted in significant HPC mobilization in the peripheral blood. The addition of βB restored BM cellularity and reduced HPC mobilization. Three h following HS and LC/HS, plasma G-CSF levels more than double, however LC alone showed no change in G-CSF. βB significantly decreased G-CSF in both HS and LC/HS. Similarly, MMP-9 is elevated following LC/HS, and βB prevents this elevation (390 ± 100 pg/mL versus 275 ± 80 pg/mL). CONCLUSION βB protection of the BM following shock and injury may be due to reduced HPC mobilization and maintenance of BM cellularity. Following shock, there is an increase in plasma G-CSF and MMP-9, which is abrogated by βB and suggests a possible mechanism how βB decreases HPC mobilization thus preserving BM cellularity. In contrast, βB protection of BM following LC is not mediated by G-CSF. Therefore, the mechanism of progenitor cell mobilization from the BM is dependent on the type of injury.
Collapse
Affiliation(s)
- Gregg M. Baranski
- Department of Surgery, Division of Trauma, UMDNJ-New Jersey Medical School, Newark, NJ
| | - Michael D. Offin
- Department of Surgery, Division of Trauma, UMDNJ-New Jersey Medical School, Newark, NJ
| | - Ziad C. Sifri
- Department of Surgery, Division of Trauma, UMDNJ-New Jersey Medical School, Newark, NJ
| | - Ihab O. Elhassan
- Department of Surgery, Division of Trauma, UMDNJ-New Jersey Medical School, Newark, NJ
| | - Edward J. Hannoush
- Department of Surgery, Division of Trauma, UMDNJ-New Jersey Medical School, Newark, NJ
| | - Walter D. Alzate
- Department of Surgery, Division of Trauma, UMDNJ-New Jersey Medical School, Newark, NJ
| | - Pranela Rameshwar
- Department of Medicine-Hematology, UMDNJ-New Jersey Medical School, Newark, NJ
| | - David H. Livingston
- Department of Surgery, Division of Trauma, UMDNJ-New Jersey Medical School, Newark, NJ
| | - Alicia M. Mohr
- Department of Surgery, Division of Trauma, UMDNJ-New Jersey Medical School, Newark, NJ
| |
Collapse
|
44
|
Cangiano E, Cavazza C, Campo G, Valgimigli M, Francolini G, Malagutti P, Pratola C, Ferrari R. Different clinical models of CD34 + cells mobilization in patients with cardiovascular disease. J Thromb Thrombolysis 2011; 32:1-8. [PMID: 21197559 DOI: 10.1007/s11239-010-0543-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
To test the role of necrosis, ischemia or both in bone marrow cells (BMC) mobilization in patients with cardiovascular disease. We studied three groups of patients: group 1, Iatrogenic Necrosis, with pure necrosis (28 patients undergoing transcatheter radiofrequency ablation); group 2, Ischemic Necrosis (30 patients with myocardial infarction); group 3, Pure Ischemia (24 patients with unstable angina). As control groups, we studied 27 patients with stable coronary artery disease (CAD), and 20 patients without CAD undergoing angiography for valvular diseases or cardiomiopathy. CD34 + cells and cytokines were evaluated at: T(0) (baseline), 48 h and 5, 7, 10, 14 days thereafter. We observed a significant increase of CD34 + cells at T(3) and T(4) only in Iatrogenic Necrosis and Ischemic Necrosis group. The peak of mobilization was observed ten days after the necrotic event (2.8 ± 1.4 vs. 5.9 ± 1.9 in the group 1, P = 0.03; and 3 ± 1.5 vs. 5.6 ± 2 in the group 2, P = 0.04; respectively). We found a good correlation between CD34 + and vascular endothelial growth factor (VEGF) and stromal derived factor (SDF-1α) peak values (r = 0.77 and r = 0.63, respectively). At multivariable analysis, myocardial necrosis (OR 3.5, 95%CI 2.2-4.2, P < 0.01), VEGF (OR 2, 95%CI 1.1-3, P = 0.01 as above versus below median value), and SDF-1α (OR 1.6, 95%CI 1.1-2.5, P = 0.02 as above versus below median value) emerged as independent predictors of C34 + cells increase. Myocardial necrosis with simultaneous elevation of VEGF and SDF-1α causes a significant CD34 + cells mobilization in patients with cardiovascular disease.
Collapse
Affiliation(s)
- Elisa Cangiano
- U.O. Cardiologia, Cardiovascular Institute, Azienda Ospedaliero-Universitaria S.Anna, Ferrara, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Boyle AJ, Yeghiazarians Y, Shih H, Hwang J, Ye J, Sievers R, Zheng D, Palasubramaniam J, Palasubramaniam D, Karschimkus C, Whitbourn R, Jenkins A, Wilson AM. Myocardial production and release of MCP-1 and SDF-1 following myocardial infarction: differences between mice and man. J Transl Med 2011; 9:150. [PMID: 21910857 PMCID: PMC3180393 DOI: 10.1186/1479-5876-9-150] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 09/12/2011] [Indexed: 12/27/2022] Open
Abstract
Background Stem cell homing to the heart is mediated by the release of chemo-attractant cytokines. Stromal derived factor -1 alpha (SDF-1a) and monocyte chemotactic factor 1(MCP-1) are detectable in peripheral blood after myocardial infarction (MI). It remains unknown if they are produced by, and released from, the heart in order to attract stem cells to repair the damaged myocardium. Methods Murine hearts were studied for expression of MCP-1 and SDF-1a at day 3 and day 28 following myocardial infarction to determine whether production is increased following MI. In addition, we studied the coronary artery and coronary sinus (venous) blood from patients with normal coronary arteries, stable coronary artery disease (CAD), unstable angina and MI to determine whether these cytokines are released from the heart into the systemic circulation following MI. Results Both MCP-1 and SDF-1a are constitutively produced and released by the heart. MCP-1 mRNA is upregulated following murine experimental MI, but SDF-1a is suppressed. There is less release of SDF-1a into the systemic circulation in patients with all stages of CAD including MI, mimicking the animal model. However MCP-1 release from the human heart following MI is also suppressed, which is the exact opposite of the animal model. Conclusions SDF-1a and MCP-1 release from the human heart are suppressed following MI. In the case of SDF-1a, the animal model appropriately reflects the human situation. However, for MCP-1 the animal model is the exact opposite of the human condition. Human observational studies like this one are paramount in guiding translation from experimental studies to clinical trials.
Collapse
Affiliation(s)
- Andrew J Boyle
- Department of Medicine, Division of Cardiology, University of California San Francisco, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Sanganalmath SK, Abdel-Latif A, Bolli R, Xuan YT, Dawn B. Hematopoietic cytokines for cardiac repair: mobilization of bone marrow cells and beyond. Basic Res Cardiol 2011; 106:709-33. [PMID: 21541807 PMCID: PMC4281455 DOI: 10.1007/s00395-011-0183-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 04/11/2011] [Accepted: 04/15/2011] [Indexed: 12/20/2022]
Abstract
Hematopoietic cytokines, traditionally known to influence cellular proliferation, differentiation, maturation, and lineage commitment in the bone marrow, include granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor, stem cell factor, Flt-3 ligand, and erythropoietin among others. Emerging evidence suggests that these cytokines also exert multifarious biological effects on diverse nonhematopoietic organs and tissues. Although the precise mechanisms remain unclear, numerous studies in animal models of myocardial infarction (MI) and heart failure indicate that hematopoietic cytokines confer potent cardiovascular benefits, possibly through mobilization and subsequent homing of bone marrow-derived cells into the infarcted heart with consequent induction of myocardial repair involving multifarious mechanisms. In addition, these cytokines are also known to exert direct cytoprotective effects. However, results from small-scale clinical trials of G-CSF therapy as a single agent after acute MI have been discordant and largely disappointing. It is likely that cardiac repair following cytokine therapy depends on a number of known and unknown variables, and further experimental and clinical studies are certainly warranted to accurately determine the true therapeutic potential of such therapy. In this review, we discuss the biological features of several key hematopoietic cytokines and present the basic and clinical evidence pertaining to cardiac repair with hematopoietic cytokine therapy.
Collapse
Affiliation(s)
- Santosh K. Sanganalmath
- Division of Cardiovascular Diseases, Cardiovascular Research Institute, University of Kansas Medical Center, 3901 Rainbow Blvd, Rm. 1001 Eaton, MS 3006, Kansas City, KS 66160, USA
| | - Ahmed Abdel-Latif
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY 40292, USA
| | - Yu-Ting Xuan
- Division of Cardiovascular Diseases, Cardiovascular Research Institute, University of Kansas Medical Center, 3901 Rainbow Blvd, Rm. 1001 Eaton, MS 3006, Kansas City, KS 66160, USA
| | - Buddhadeb Dawn
- Division of Cardiovascular Diseases, Cardiovascular Research Institute, University of Kansas Medical Center, 3901 Rainbow Blvd, Rm. 1001 Eaton, MS 3006, Kansas City, KS 66160, USA
| |
Collapse
|
47
|
|
48
|
Cangiano E, Marchesini J, Campo G, Francolini G, Fortini C, Carrà G, Miccoli M, Ceconi C, Tavazzi L, Ferrari R. ACE Inhibition Modulates Endothelial Apoptosis and Renewal via Endothelial Progenitor Cells in Patients with Acute Coronary Syndromes. Am J Cardiovasc Drugs 2011; 11:189-98. [DOI: 10.2165/11589400-000000000-00000] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
49
|
Shah VK, Shalia KK. Stem Cell Therapy in Acute Myocardial Infarction: A Pot of Gold or Pandora's Box. Stem Cells Int 2011; 2011:536758. [PMID: 21804827 PMCID: PMC3142872 DOI: 10.4061/2011/536758] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 12/18/2010] [Accepted: 12/29/2010] [Indexed: 01/09/2023] Open
Abstract
Stem cell therapy for conditions characterized by myocyte loss in myocardial infarction and heart failure is intuitively appealing. Stem cells from various sources, including heart itself in preclinical and animal studies, have shown the potential to improve the function of ventricular muscle after ischaemic injury. The clinical experience from worldwide studies have indicated the safety profile but with modest benefits. The predominant mechanisms of transplanted cells for improving cardiac function have pointed towards paracrine effects rather than transdifferentiation into cardiomyocytes. Thus, further investigations should be encouraged towards bench side and bedside to resolve various issues for ensuring the correct type and dosing of cells, time, and method of delivery and identify correct mechanism of functional improvement. An interdisciplinary effort at the scientific, clinical, and the government front will bring successful realization of this therapy for healing the heart and may convert what seems now a Pandora's Box into a Pot of Gold.
Collapse
Affiliation(s)
- V K Shah
- Interventional Cardiologist, Sir H.N. Hospital and Research Centre, Raja Rammohan Roy Road, Mumbai 400 004, India
| | | |
Collapse
|
50
|
Bogoslovsky T, Spatz M, Chaudhry A, Maric D, Luby M, Frank J, Warach S, NINDS Natural History of Stroke Investigators. Stromal-derived factor-1[alpha] correlates with circulating endothelial progenitor cells and with acute lesion volume in stroke patients. Stroke 2011; 42:618-25. [PMID: 21257825 PMCID: PMC4751042 DOI: 10.1161/strokeaha.110.596007] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 09/02/2010] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND PURPOSE Endothelial progenitor cells (EPC) are important participants of neovascularization and are mobilized through signaling with stromal-derived factor (SDF-1α), vascular endothelial growth factor (VEGF), granulocyte colony-stimulating factor, and stem cell factor. The association between EPC levels and these growth factors (GF) in acute stroke has not been previously established. We aimed to determine the association between EPC and these GF, and to elucidate a relationship between these GF and stroke severity in acute stroke patients. METHODS Seventeen patients were selected from 175 patients with imaging-confirmed acute ischemic stroke. EPC were quantified using CD34, CD133, and VEGF-R2 markers. Plasma VEGF, SDF-1α, granulocyte colony-stimulating factor, and stem cell factor were determined by enzyme-linked immunosorbent assay on days 1 and 3, and brain MRI was performed at baseline and on days 1 and 5 after the stroke onset. RESULTS Levels of SDF-1α strongly (r=0.6) correlated with the numbers of EPC subsets CD133(+)VEFG-R2(+) (P<0.004), CD34(+)VEGF-R2(+) (P<0.01), and CD34(+)CD133(+)VEGF-R2(+) (P<0.01) on day 1. Stem cell factor strongly (r=0.5) correlated with CD133(+)VEGF-R2(+) (P<0.05). SDF-1α moderately inversely (r=-0.49) correlated with baseline diffusion-weighted imaging lesion volumes (P<0.04). Median levels of SDF-1α (1561 pg/mL) increased (P<0.04) on day 3 compared to day 1 (1379 pg/mL). Similarly, VEGF at day 3 (95 pg/mL) increased (P<0.03) compared to day 1 (64 pg/mL). CONCLUSIONS These results indicate that SDF-1α and stem cell factor correlate with an increase in EPC early in ischemic stroke patients.
Collapse
Affiliation(s)
- Tanya Bogoslovsky
- Center for Neuroscience & Regenerative Medicine, Uniformed Services University of the Health Sciences, National Institute of Nursing Research, National Institutes of Health, 12725 Twinbrook Parkway, Rockville, MD 20852, USA.
| | | | | | | | | | | | | | | |
Collapse
|