1
|
Li N, Huang C, Zhang J, Zhang J, Huang J, Li S, Xia X, Wu Z, Chen C, Tang S, Xiao X, Gong H, Dai Y, Mao C, Wan M. Chemotactic NO/H 2S Nanomotors Realizing Cardiac Targeting of G-CSF against Myocardial Ischemia-Reperfusion Injury. ACS NANO 2023. [PMID: 37327056 DOI: 10.1021/acsnano.3c02781] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Recombinant granulocyte colony-stimulating factor (G-CSF), with a direct repair effect on injured cardiomyocytes against myocardial infarction ischemia-reperfusion-injury (IRI), displays a poor effect owing to the limited cardiac targeting efficacy. There are almost no reports of nanomaterials that deliver G-CSF to the IRI site. Herein, we propose a way to protect G-CSF by constructing one layer of nitric oxide (NO)/hydrogen sulfide (H2S) nanomotors on its outside. NO/H2S nanomotors with specific chemotactic ability to high expression of reactive oxygen species (ROS)/induced nitric oxide synthase (iNOS) at the IRI site can deliver G-CSF to the IRI site efficiently. Meanwhile, superoxide dismutase is covalently bound to the outermost part, reducing ROS at the IRI site through a cascade effect with NO/H2S nanomotors. The synergistic effect between NO and H2S on the effective regulation of the IRI microenvironment can not only avoid toxicity caused by excessive concentration of a single gas but also reduce inflammation level and relieve calcium overload, so as to promote G-CSF to play a cardioprotective role.
Collapse
Affiliation(s)
- Nan Li
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Chenxing Huang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200433, China
| | - Jie Zhang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Junyue Zhang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Jia Huang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200433, China
| | - Shangshang Li
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Xue Xia
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Ziyu Wu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Chenglong Chen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Shuwan Tang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Xiangyu Xiao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Hui Gong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200433, China
| | - Yuxiang Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200433, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| |
Collapse
|
2
|
Carbone RG, Monselise A, Bottino G, Negrini S, Puppo F. Stem cells therapy in acute myocardial infarction: a new era? Clin Exp Med 2021; 21:231-237. [PMID: 33484381 PMCID: PMC8053645 DOI: 10.1007/s10238-021-00682-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
Stem cells transplantation after acute myocardial infarction (AMI) has been claimed to restore cardiac function. However, this therapy is still restricted to experimental studies and clinical trials. Early un-blinded studies suggested a benefit from stem cell therapy following AMI. More recent blinded randomized trials have produced mixed results and, notably, the last largest pan-European clinical trial showed the inconclusive results. Furthermore, mechanisms of potential benefit remain uncertain. This review analytically evaluates 34 blinded and un-blinded clinical trials comprising 3142 patients and is aimed to: (1) identify the pros and cons of stem cell therapy up to a 6-month follow-up after AMI comparing benefit or no effectiveness reported in clinical trials; (2) provide useful information for planning future clinical programs of cardiac stem cell therapy.
Collapse
Affiliation(s)
- R G Carbone
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | | | - G Bottino
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - S Negrini
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - F Puppo
- Department of Internal Medicine, University of Genoa, Genoa, Italy.
| |
Collapse
|
3
|
The Effects of Granulocyte Colony-Stimulating Factor in Patients with a Large Anterior Wall Acute Myocardial Infarction to Prevent Left Ventricular Remodeling: A 10-Year Follow-Up of the RIGENERA Study. J Clin Med 2020; 9:jcm9041214. [PMID: 32340315 PMCID: PMC7230316 DOI: 10.3390/jcm9041214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023] Open
Abstract
Background: the RIGENERA trial assessed the efficacy of granulocyte-colony stimulating factor (G-CSF) in the improvement of clinical outcomes in patients with severe acute myocardial infarction. However, there is no evidence available regarding the long-term safety and efficacy of this treatment. Methods: in order to evaluate the long-term effects on the incidence of major adverse events, on the symptom burden, on the quality of life and the mean life expectancy and on the left ventricular (LV) function, we performed a clinical and echocardiographic evaluation together with an assessment using the Minnesota Living with Heart Failure Questionnaire (MLHFQ) and the Seattle Heart Failure Model (SHFM) at 10-years follow-up, in the patients cohorts enrolled in the RIGENERA trial. Results: thirty-two patients were eligible for the prospective clinical and echocardiography analyses. A significant reduction in adverse LV remodeling was observed in G-CSF group compared to controls, 9% vs. 48% (p = 0.030). The New York Heart Association (NYHA) functional class was lower in G-CSF group vs. controls (p = 0.040), with lower burden of symptoms and higher quality of life (p = 0.049). The mean life expectancy was significantly higher in G-CSF group compared to controls (15 ± 4 years vs. 12 ± 4 years, p = 0.046. No difference was found in the incidence of major adverse events. Conclusions: this longest available follow-up on G-CSF treatment in patients with severe acute myocardial infarction (AMI) showed that this treatment was safe and associated with a reduction of adverse LV remodeling and higher quality of life, in comparison with standard-of-care treatment.
Collapse
|
4
|
Nakao S, Tsukamoto T, Ueyama T, Kawamura T. STAT3 for Cardiac Regenerative Medicine: Involvement in Stem Cell Biology, Pathophysiology, and Bioengineering. Int J Mol Sci 2020; 21:ijms21061937. [PMID: 32178385 PMCID: PMC7139789 DOI: 10.3390/ijms21061937] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/07/2020] [Accepted: 03/10/2020] [Indexed: 12/28/2022] Open
Abstract
Heart disease is the most common cause of death in developed countries, but the medical treatments for heart failure remain limited. In this context, the development of cardiac regeneration therapy for severe heart failure is important. Owing to their unique characteristics, including multiple differentiation and infinitive self-renewal, pluripotent stem cells can be considered as a novel source for regenerative medicine. Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) signaling plays critical roles in the induction, maintenance, and differentiation of pluripotent stem cells. In the heart, JAK/STAT3 signaling has diverse cellular functions, including myocardial differentiation, cell cycle re-entry of matured myocyte after injury, and anti-apoptosis in pathological conditions. Therefore, regulating STAT3 activity has great potential as a strategy of cardiac regeneration therapy. In this review, we summarize the current understanding of STAT3, focusing on stem cell biology and pathophysiology, as they contribute to cardiac regeneration therapy. We also introduce a recently reported therapeutic strategy for myocardial regeneration that uses engineered artificial receptors that trigger endogenous STAT3 signal activation.
Collapse
Affiliation(s)
- Shu Nakao
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Japan; (S.N.); (T.T.); (T.U.)
- Ritsumeikan Global Innovation Research Institute, Ritsumeikan University, Kusatsu 525-8577, Japan
| | - Tasuku Tsukamoto
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Japan; (S.N.); (T.T.); (T.U.)
- Ritsumeikan Global Innovation Research Institute, Ritsumeikan University, Kusatsu 525-8577, Japan
| | - Tomoe Ueyama
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Japan; (S.N.); (T.T.); (T.U.)
- Ritsumeikan Global Innovation Research Institute, Ritsumeikan University, Kusatsu 525-8577, Japan
| | - Teruhisa Kawamura
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Japan; (S.N.); (T.T.); (T.U.)
- Ritsumeikan Global Innovation Research Institute, Ritsumeikan University, Kusatsu 525-8577, Japan
- Correspondence: ; Tel.: +81-75-599-4327
| |
Collapse
|
5
|
Singhal A, Subramanian M. Colony stimulating factors (CSFs): Complex roles in atherosclerosis. Cytokine 2019; 122:154190. [DOI: 10.1016/j.cyto.2017.10.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/10/2017] [Accepted: 10/11/2017] [Indexed: 12/11/2022]
|
6
|
Hu Z, Chen Z, Wang Y, Jiang J, Tse G, Xu W, Ge J, Sun B. Effects of granulocyte colony‑stimulating factor on rabbit carotid and porcine heart models of chronic obliterative arterial disease. Mol Med Rep 2019; 19:4569-4578. [PMID: 30942413 PMCID: PMC6522810 DOI: 10.3892/mmr.2019.10120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 03/01/2019] [Indexed: 11/16/2022] Open
Abstract
Previous studies suggest that granulocyte colony‑stimulating factor (G‑CSF) can promote bone marrow derived progenitor cells to mediate cardiovascular repair, potentially reversing mechanical dysfunction in chronic ischaemic heart disease and post myocardial infarction. Two models were used in the present study both using a surgical ameroid constrictor to induce arterial stenosis. The first model used the carotid artery of rabbits. They were divided into high fat diet (inducing atherosclerosis) or normal fat diet (control) groups. Each was subdivided into surgical exposure group without constrictor, ameroid constrictor receiving normal saline or receiving G‑CSF 15 µg/kg/day. Endothelial markers of endothelial nitric oxide synthase and endothelin 1 were increased by the use of ameroid constrictor in both atherosclerotic and non‑atherosclerotic mice, however were not further altered by G‑CSF. Scanning electron microscopy indicated that ameroid constrictor application altered endothelial morphology from an oval shape to a round shape and this was more prominent in the atherosclerotic compared with the non‑atherosclerotic group. G‑CSF injection increased the number of endothelial cells in all groups. The second model used the left coronary artery of pigs. They were equally divided into following groups, receiving normal saline (control), G‑CSF 2.5 µg/kg/day (low dose), 5 µg/kg/day (medium dose) and 10 µg/kg/day (high dose) for 5 days. G‑CSF at a low or high dose worsened intimal hyperplasia however at a medium dose improved it. In conclusion, G‑CSF had no effect in a rabbit carotid artery model of atherosclerosis. Its effects on the porcine heart were dose‑dependent; arterial disease worsened at a low or high dose, but improved at a medium dose.
Collapse
Affiliation(s)
- Zhaohui Hu
- Department of Cardiovascular Diseases, Tongji Hospital of Tongji University, Shanghai 200065, P.R. China
| | - Zhisong Chen
- Department of Cardiovascular Diseases, Tongji Hospital of Tongji University, Shanghai 200065, P.R. China
| | - Yiping Wang
- Department of Cardiovascular Diseases, Tongji Hospital of Tongji University, Shanghai 200065, P.R. China
| | - Jinfa Jiang
- Department of Cardiovascular Diseases, Tongji Hospital of Tongji University, Shanghai 200065, P.R. China
| | - Gary Tse
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong, SAR, P.R. China
- Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Wenjun Xu
- Department of Cardiovascular Diseases, Tongji Hospital of Tongji University, Shanghai 200065, P.R. China
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Bing Sun
- Department of Cardiovascular Diseases, Tongji Hospital of Tongji University, Shanghai 200065, P.R. China
| |
Collapse
|
7
|
Achilli F, Pontone G, Bassetti B, Squadroni L, Campodonico J, Corrada E, Facchini C, Mircoli L, Esposito G, Scarpa D, Pidello S, Righetti S, Di Gennaro F, Guglielmo M, Muscogiuri G, Baggiano A, Limido A, Lenatti L, Di Tano G, Malafronte C, Soffici F, Ceseri M, Maggiolini S, Colombo GI, Pompilio G. G-CSF for Extensive STEMI. Circ Res 2019; 125:295-306. [PMID: 31138020 DOI: 10.1161/circresaha.118.314617] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
RATIONALE In the exploratory Phase II STEM-AMI (Stem Cells Mobilization in Acute Myocardial Infarction) trial, we reported that early administration of G-CSF (granulocyte colony-stimulating factor), in patients with anterior ST-segment-elevation myocardial infarction and left ventricular (LV) dysfunction after successful percutaneous coronary intervention, had the potential to significantly attenuate LV adverse remodeling in the long-term. OBJECTIVE The STEM-AMI OUTCOME CMR (Stem Cells Mobilization in Acute Myocardial Infarction Outcome Cardiac Magnetic Resonance) Substudy was adequately powered to evaluate, in a population showing LV ejection fraction ≤45% after percutaneous coronary intervention for extensive ST-segment-elevation myocardial infarction, the effects of early administration of G-CSF in terms of LV remodeling and function, infarct size assessed by late gadolinium enhancement, and myocardial strain. METHODS AND RESULTS Within the Italian, multicenter, prospective, randomized, Phase III STEM-AMI OUTCOME trial, 161 ST-segment-elevation myocardial infarction patients were enrolled in the CMR Substudy and assigned to standard of care (SOC) plus G-CSF or SOC alone. In 119 patients (61 G-CSF and 58 SOC, respectively), CMR was available at baseline and 6-month follow-up. Paired imaging data were independently analyzed by 2 blinded experts in a core CMR lab. The 2 groups were similar for clinical characteristics, cardiovascular risk factors, and pharmacological treatment, except for a trend towards a larger infarct size and longer symptom-to-balloon time in G-CSF patients. ANCOVA showed that the improvement of LV ejection fraction from baseline to 6 months was 5.1% higher in G-CSF patients versus SOC (P=0.01); concurrently, there was a significant between-group difference of 6.7 mL/m2 in the change of indexed LV end-systolic volume in favor of G-CSF group (P=0.02). Indexed late gadolinium enhancement significantly decreased in G-CSF group only (P=0.04). Moreover, over time improvement of global longitudinal strain was 2.4% higher in G-CSF patients versus SOC (P=0.04). Global circumferential strain significantly improved in G-CSF group only (P=0.006). CONCLUSIONS Early administration of G-CSF exerted a beneficial effect on top of SOC in patients with LV dysfunction after extensive ST-segment-elevation myocardial infarction in terms of global systolic function, adverse remodeling, scar size, and myocardial strain. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrials.gov. Unique identifier: NCT01969890.
Collapse
Affiliation(s)
- Felice Achilli
- From the Departments of Cardiology (F.A., S.R., C.M., F.S.), ASST-Monza, San Gerardo Hospital, Monza, Italy
| | - Gianluca Pontone
- Cardiovascular Imaging (G. Pontone, M.G., G.M., A.B.), Centro Cardiologico Monzino IRCCS, Milano, Italy.,Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano, Italy (G.P.)
| | - Beatrice Bassetti
- Vascular Biology and Regenerative Medicine Unit (B.B., G. Pompilio), Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - Lidia Squadroni
- Department of Cardiology, San Carlo Borromeo Hospital, Milano, Italy (L.S.)
| | - Jeness Campodonico
- Intensive Cardiac Care Unit (J.C.), Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - Elena Corrada
- Cardiovascular Department, Humanitas Clinical and Research Center IRCCS, Rozzano, Italy (E.C.)
| | | | - Luca Mircoli
- Cardiology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy (L.M.)
| | - Giovanni Esposito
- Division of Cardiology, Department of Advanced Biomedical Sciences, University of Naples Federico II, Napoli, Italy (G.E.)
| | - Daniele Scarpa
- Cardiology, Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Italy (D.S.)
| | - Stefano Pidello
- Cardiology, Città della Salute e della Scienza University Hospital of Torino, Italy (S.P.)
| | - Stefano Righetti
- From the Departments of Cardiology (F.A., S.R., C.M., F.S.), ASST-Monza, San Gerardo Hospital, Monza, Italy
| | | | - Marco Guglielmo
- Cardiovascular Imaging (G. Pontone, M.G., G.M., A.B.), Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - Giuseppe Muscogiuri
- Cardiovascular Imaging (G. Pontone, M.G., G.M., A.B.), Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - Andrea Baggiano
- Cardiovascular Imaging (G. Pontone, M.G., G.M., A.B.), Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - Alberto Limido
- Coronary Intensive Care Unit, ASST-Settelaghi, Ospedale di Circolo-Fondazione Macchi, Varese, Italy (A.L.)
| | - Laura Lenatti
- Cardiology, Alessandro Manzoni Hospital, Lecco, Italy (L.L.)
| | | | - Cristina Malafronte
- From the Departments of Cardiology (F.A., S.R., C.M., F.S.), ASST-Monza, San Gerardo Hospital, Monza, Italy
| | - Federica Soffici
- From the Departments of Cardiology (F.A., S.R., C.M., F.S.), ASST-Monza, San Gerardo Hospital, Monza, Italy
| | - Martina Ceseri
- ANMCO Research Center, Heart Care Foundation, Firenze, Italy (M.C.)
| | | | - Gualtiero I Colombo
- Immunology and Functional Genomics Unit (G.I.C.), Centro Cardiologico Monzino IRCCS, Milano, Italy
| | - Giulio Pompilio
- Vascular Biology and Regenerative Medicine Unit (B.B., G. Pompilio), Centro Cardiologico Monzino IRCCS, Milano, Italy
| | | |
Collapse
|
8
|
French BA, Holmes JW. Implications of scar structure and mechanics for post-infarction cardiac repair and regeneration. Exp Cell Res 2019; 376:98-103. [PMID: 30610848 DOI: 10.1016/j.yexcr.2019.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/21/2018] [Accepted: 01/01/2019] [Indexed: 01/14/2023]
Abstract
Regenerating cardiac muscle lost during a heart attack is a topic of broad interest and enormous potential impact. One promising approach is to regenerate or re-engineer new myocardium in situ, at the site of damage, by injecting cells, growth factors, and other materials, or by reprogramming aspects of the normal wound healing process. A wide variety of strategies have been explored, from promoting angiogenesis to injection of a variety of different progenitor cell types, to re-engineering resident cells to produce key growth factors or even transdifferentiate into myocytes. Despite substantial progress and continued promise, clinical impact of this work has fallen short of expectations. One contributing factor may be that many efforts focus primarily on generating cardiomyocytes, with less attention to re-engineering the extracellular matrix (ECM). Yet the role of the ECM is particularly crucial to consider following myocardial infarction, which leads to rapid formation of a collagen-rich scar. This review combines a brief summary of current efforts to regenerate cardiomyocytes with what is currently known about the structure and mechanics of post-infarction scar, with the goal of identifying principles that can guide efforts to produce new myocytes embedded in an extracellular environment that facilitates their differentiation, maintenance, and function.
Collapse
Affiliation(s)
- Brent A French
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA; Department of Radiology, University of Virginia, Charlottesville, VA, USA; Department of Medicine, University of Virginia, Charlottesville, VA, USA; Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Jeffrey W Holmes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA; Department of Medicine, University of Virginia, Charlottesville, VA, USA; Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
9
|
Li X, Wan Q, Min J, Duan L, Liu J. Premobilization of CD133+ cells by granulocyte colony- stimulating factor attenuates ischemic acute kidney injury induced by cardiopulmonary bypass. Sci Rep 2019; 9:2470. [PMID: 30792422 PMCID: PMC6385363 DOI: 10.1038/s41598-019-38953-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 01/11/2019] [Indexed: 02/05/2023] Open
Abstract
Ischemic acute kidney injury (IAKI) is a common but severe complication after a cardiopulmonary bypass (CPB). Multiple studies have demonstrated that peripheral CD133+ or differentiated cells are able to home and repair the damaged tissues, but the number of available CD133+ cells is limited, and no efficient method published previously to mobilize them immediately. We analyzed the relationship between CD133+ cells and renal function in CPB patients, in addition, the efficacy of granulocyte colony-stimulating factor (G-CSF) pre-mobilized CD133+ cells in treating of mouse IAKI model have been investigated. In the clinical study, the prospective cohort study analyzed the correlation between BUN/Crea level and the peripheral CD133+ cell numbers. CPB was associated with postoperative renal dysfunction. The significant negative correlation was observed between patients' Crea and CD133+ cells (P < 0.05). The proposed mechanism studies were performed on the mouse IAKI model. The experimental mice were treated by G-CSF to mobilize CD133+ cells before implementing CPB. Data on cell count, inflammatory index, renal function/injury, and CD133+ cell mobilization were analyzed. The result demonstrated that pretreatment by G-CSF resulted in tremendous increase in the number of mouse peripheral blood and renal CD133+ cells, significantly reduces renal tissue inflammation and dramatically improves the renal function after CPB. In summary, we concluded that premobilization of CD133+ cells abated CPB induced IAKI, by promoting both repairing damaged epithelium and by its anti-inflammatory activity. Our findings stress the remarkable applications of CD133+ or differentiated cells-based therapies for potential preventing ischemic acute kidney injury.
Collapse
Affiliation(s)
- Xiaoqiang Li
- Department of Anesthesiology, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Qin Wan
- Department of Anesthesiology, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Jie Min
- Department of Integrated Traditional and Western Medicine, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Linjia Duan
- Department of Cardiology, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, 610041, Chengdu, China.
| |
Collapse
|
10
|
Yamada Y, Minatoguchi S, Endo N, Kanamori H, Kawasaki M, Nishigaki K, Mikami A, Minatoguchi S. Post-MI treatment with G-CSF and EPO-liposome with SLX repairs infarcted myocardium through EPCs mobilization and activation of prosurvival signals in rabbits. Pharmacol Res Perspect 2019; 7:e00451. [PMID: 30598826 PMCID: PMC6302719 DOI: 10.1002/prp2.451] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 11/19/2018] [Indexed: 01/02/2023] Open
Abstract
We investigated whether combination therapy of G-CSF and erythropoietin (EPO)-liposome with Siaryl Lewis X (SLX) is more cardioprotective than G-CSF or EPO-liposome with SLX alone. For the purpose of generating myocardial infarction (MI), rabbits underwent 30 minutes of coronary occlusion and 14 days of reperfusion. We administered saline (control group, i.v.,), G-CSF (G group, 10 μg/kg/day × 5 days, i.c., starting at 24 hours after reperfusion), EPO-liposome with SLX (LE group, i.v., 2500 IU/kg EPO containing liposome with SLX, immediately after reperfusion), and G-CSF + EPO-liposome with SLX (LE + G group) to the rabbits. The MI size was the smallest in the LE+G group (14.7 ± 0.8%), and smaller in the G group (22.4 ± 1.5%) and LE group (18.5 ± 1.1%) than in the control group (27.8 ± 1.5%). Compared with the control group, the cardiac function and remodeling of the G, LE, and LE + G groups were improved, and LE + G group tended to show the best improvement. The number of CD31-positive microvessels was the greatest in the LE + G group, greater in the G and LE groups than in the control group. Higher expressions of phosphorylated (p)-Akt and p-ERK were observed in the ischemic area of the LE and LE + G groups. The number of CD34+/CXCR4+ cells was significantly higher in the G and LE + G groups. The cardiac SDF-1 was more expressed in the G and LE + G groups. In conclusion, Post-MI combination therapy with G-CSF and EPO-liposome with SLX is more cardioprotective than G-CSF or EPO-liposome with SLX alone through EPCs mobilization, neovascularization, and activation of prosurvival signals.
Collapse
Affiliation(s)
- Yoshihisa Yamada
- Department of CardiologyGifu University Graduate School of MedicineGifuJapan
| | - Shingo Minatoguchi
- Department of CardiologyGifu University Graduate School of MedicineGifuJapan
| | - Noriko Endo
- Department of CardiologyGifu University Graduate School of MedicineGifuJapan
| | - Hiromitsu Kanamori
- Department of CardiologyGifu University Graduate School of MedicineGifuJapan
| | - Masanori Kawasaki
- Department of CardiologyGifu University Graduate School of MedicineGifuJapan
| | | | - Atsushi Mikami
- Department of CardiologyGifu University Graduate School of MedicineGifuJapan
| | - Shinya Minatoguchi
- Department of CardiologyGifu University Graduate School of MedicineGifuJapan
- CardiologyGifu Municipal HospitalGifuJapan
| |
Collapse
|
11
|
Study of tumor growth indicates the existence of an "immunological threshold" separating states of pro- and antitumoral peritumoral inflammation. PLoS One 2018; 13:e0202823. [PMID: 30388111 PMCID: PMC6214501 DOI: 10.1371/journal.pone.0202823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 08/09/2018] [Indexed: 11/28/2022] Open
Abstract
Background Peritumoral inflammation—a response mainly involving polimorphonuclear neutrophils—has traditionally been thought protumoral in its effects. In recent years, however, a number of studies have indicated that it may play an important antitumoral role. This discrepancy has been difficult to explain. Methods and findings This work describes a tool for simulating tumor growth that obeys the universal model of tumor growth dynamics, and shows through its use that low intensity peritumoral inflammation exerts a protumoral effect, while high intensity inflammation exerts a potent antitumoral effect. Indeed, the simulation results obtained indicate that a sufficiently strong antitumoral effect can reverse tumor growth, as has been suggested several times in the clinical literature. Conclusions The present result indicate that an ‘immunological threshold’ must exist, marking the boundary between states in which peritumoral inflammation is either harmful or beneficial. These findings lend support to the idea that stimulating intense peritumoral inflammation could be used as a treatment against solid tumors.
Collapse
|
12
|
Dubnika A, Manoukian MA, Mohammadi MR, Parekh MB, Gurjarpadhye AA, Inayathullah M, Dubniks V, Lakey JR, Rajadas J. Cytokines as therapeutic agents and targets in heart disease. Cytokine Growth Factor Rev 2018; 43:54-68. [DOI: 10.1016/j.cytogfr.2018.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/01/2018] [Accepted: 08/13/2018] [Indexed: 02/02/2023]
|
13
|
Banovic M, Pusnik-Vrckovnik M, Nakou E, Vardas P. Myocardial regeneration therapy in heart failure: Current status and future therapeutic implications in clinical practice. Int J Cardiol 2018; 260:124-130. [DOI: 10.1016/j.ijcard.2018.01.144] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 01/25/2018] [Accepted: 01/31/2018] [Indexed: 12/16/2022]
|
14
|
Acute Myocardial Infarction, Cardioprotection, and Muse Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1103:153-166. [PMID: 30484228 DOI: 10.1007/978-4-431-56847-6_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Acute myocardial infarction (AMI) is a common cause of morbidity and mortality worldwide. Severe MI leads to heart failure due to a marked loss of functional cardiomyocytes. First-line treatment for AMI is to reperfuse the occluded coronary artery by PCI as soon as possible. Besides PCI, there are several therapies to reduce the infarct size and improve the cardiac function and remodeling. These are drug therapies such as pharmacological pre- and postconditioning, cytokine therapies, and stem cell therapies. None of these therapies have been clinically developed as a standard treatment for AMI. Among many cell sources for stem cell therapies, the Muse cell is an endogenous non-tumorigenic pluripotent stem cell, which is able to differentiate into cells of all three germ layers from a single cell, suggesting that the Muse cell is a potential cell source for regenerative medicine. Endogenous Muse cell dynamics in the acute phase plays an important role in the prognosis of AMI patients; AMI patients with a higher number of Muse cells in the peripheral blood in the acute phase show more favorable improvement of the cardiac function and remodeling in the chronic phase, suggesting their innate reparative function for the heart. Intravenously administered exogenous Muse cells engrafted preferentially and efficiently to infarct border areas via the S1P-S1PR2 axis and differentiated spontaneously into working cardiomyocytes and vessels, showed paracrine effects, markedly reduced the myocardial infarct size, and delivered long-lasting improvement of the cardiac function and remodeling for 6 months. These findings suggest that Muse cells are reparative stem cells, and thus their clinical application is warranted.
Collapse
|
15
|
Fernández-Avilés F, Sanz-Ruiz R, Climent AM, Badimon L, Bolli R, Charron D, Fuster V, Janssens S, Kastrup J, Kim HS, Lüscher TF, Martin JF, Menasché P, Simari RD, Stone GW, Terzic A, Willerson JT, Wu JC. Global position paper on cardiovascular regenerative medicine. Eur Heart J 2017; 38:2532-2546. [PMID: 28575280 PMCID: PMC5837698 DOI: 10.1093/eurheartj/ehx248] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/13/2017] [Accepted: 04/20/2017] [Indexed: 12/11/2022] Open
Affiliation(s)
- Francisco Fernández-Avilés
- Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
- CIBERCV, ISCIII, Madrid, Spain
| | - Ricardo Sanz-Ruiz
- Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
- CIBERCV, ISCIII, Madrid, Spain
| | - Andreu M Climent
- Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
- CIBERCV, ISCIII, Madrid, Spain
| | - Lina Badimon
- CIBERCV, ISCIII, Madrid, Spain
- Cardiovascular Research Center (CSIC-ICCC), Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain
| | - Roberto Bolli
- Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville School of Medicine, Louisville, Kentucky
| | - Dominique Charron
- LabEx TRANSPLANTEX; HLA & Médecine "Jean Dausset" Laboratory Network, Hôpital Saint-Louis AP-HP, Université Paris Diderot, 75013, France
| | - Valentin Fuster
- CIBERCV, ISCIII, Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of medicine at Mount Sinai, New York, NY, USA
| | - Stefan Janssens
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Jens Kastrup
- Department of Cardiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Hyo-Soo Kim
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, Korea; Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Thomas F Lüscher
- Department of Cardiology, University Heart Center Zurich, Zurich, Switzerland; Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | | | - Philippe Menasché
- Department of Cardiovascular Surgery Hôpital Européen Georges Pompidou; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Robert D Simari
- School of Medicine, University of Kansas, 3901 Rainbow Boulevard, Kansas City, KS, USA
| | - Gregg W Stone
- Center for Clinical Trials, Cardiovascular Research Foundation, New York, New York; Center for Clinical Trials, NewYork-Presbyterian Hospital, Columbia University Medical Center, New York, NY, USA
| | - Andre Terzic
- Center for Regenerative Medicine, Department of Cardiovascular Diseases, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, NY, USA
| | - James T Willerson
- Department of Regenerative Medicine Research, Texas Heart Institute, Houston, TX, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine and Department of Radiology, Stanford University School of Medicine, CA, USA
| |
Collapse
|
16
|
Heo SC, Kwon YW, Jang IH, Jeong GO, Lee TW, Yoon JW, Shin HJ, Jeong HC, Ahn Y, Ko TH, Lee SC, Han J, Kim JH. Formyl Peptide Receptor 2 Is Involved in Cardiac Repair After Myocardial Infarction Through Mobilization of Circulating Angiogenic Cells. Stem Cells 2016; 35:654-665. [PMID: 27790799 DOI: 10.1002/stem.2535] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 10/02/2016] [Accepted: 10/10/2016] [Indexed: 12/11/2022]
Abstract
Increasing evidence suggests that circulating angiogenic cells (CACs) promote repair of ischemic tissues. Activation of formyl peptide receptor 2 (Fpr2) has been reported to stimulate repair of ischemic heart. This study was conducted to investigate the role of Fpr2 on CAC mobilization and cardiac protection in myocardial infarction (MI). WKYMVm, a strong agonist for Fpr2, was administered in a murine model of acute MI, and mobilization of CACs including endothelial progenitor cells (CD34+ Flk1+ or Sca1+ Flk1+ cells) in peripheral blood was monitored. CAC mobilization by daily injection of WKYMVm for the first 4 days after MI was as efficient as granulocyte colony-stimulating factor and provided myocardial protection from apoptosis with increased vascular density and preservation of cardiac function. Transplantation of bone marrow (BM) from green fluorescent protein mice showed that BM-derived cells homed to ischemic heart after WKYMVm treatment and contributed to tissue protection. Transplantation of BM from Fpr2 knockout mice showed that Fpr2 in BM cells is critical in mediation of WKYMVm-stimulated myocardial protection and neovascularization after MI. These results suggest that activation of Fpr2 in BM after WKYMVm treatment provides cardiac protection through mobilization of CACs after MI, which may lead to the development of a new clinical protocol for treating patients with ischemic heart conditions. Stem Cells 2017;35:654-665.
Collapse
Affiliation(s)
- Soon Chul Heo
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Yang Woo Kwon
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Il Ho Jang
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Geun Ok Jeong
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Tae Wook Lee
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Jung Won Yoon
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Ho Jin Shin
- Division of Hematology-Oncology, Department of Internal Medicine, School of Medicine, Medical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Hae Chang Jeong
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Youngkeun Ahn
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Tae Hee Ko
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea
| | - Sang Chul Lee
- Functional Genomics Research Center, KRIBB, Daejeon, Republic of Korea
| | - Jin Han
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea.,Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| |
Collapse
|
17
|
Abstract
Despite that advances in medical treatment and interventional procedures have reduced the mortality rate in patients with coronary artery disease, the number of patients with refractory myocardial ischemia and congestive heart failure is rapidly increasing. Experimental studies have demonstrated that bone marrow (BM) contains adult stem cells that can induce neovascularization and improve heart function in ischemic myocardium. Recent insights into the understanding of the mechanisms involved in proliferation, recruitment, mobilization, and incorporation of BM-derived stem cells into the myocardium and blood vessels have prompted development of cellular transplantation therapy for heart diseases refractory to conventional therapy. Initial preliminary clinical studies indicated potential clinical benefit of BM therapy in patients with acute myocardial infarction and chronic myocardial ischemia. Nevertheless, many obstacles remain, such as long-term safety and optimal timing and treatment strategies for BM cell therapy, and these issues need to be addressed in rationally designed, randomized clinical trials.
Collapse
Affiliation(s)
- Hung-Fat Tse
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China. hftse@ hkucc.hku.hk
| | | |
Collapse
|
18
|
Hare JM, Sanina C. Bone Marrow Mononuclear Cell Therapy and Granulocyte Colony-Stimulating Factor for Acute Myocardial Infarction: Is it Time to Reconsider? J Am Coll Cardiol 2015; 65:2383-7. [PMID: 26046731 DOI: 10.1016/j.jacc.2015.03.571] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 03/23/2015] [Indexed: 10/23/2022]
Affiliation(s)
- Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida; Division of Cardiology, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida; Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida.
| | - Cristina Sanina
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
19
|
Lluri G, Huang V, Touma M, Liu X, Harmon AW, Nakano A. Hematopoietic progenitors are required for proper development of coronary vasculature. J Mol Cell Cardiol 2015; 86:199-207. [PMID: 26241844 DOI: 10.1016/j.yjmcc.2015.07.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 06/29/2015] [Accepted: 07/24/2015] [Indexed: 10/23/2022]
Abstract
RATIONALE During embryogenesis, hematopoietic cells appear in the myocardium prior to the initiation of coronary formation. However, their role is unknown. OBJECTIVE Here we investigate whether pre-existing hematopoietic cells are required for the formation of coronary vasculature. METHODS AND RESULTS As a model of for hematopoietic cell deficient animals, we used Runx1 knockout embryos and Vav1-cre; R26-DTA embryos, latter of which genetically ablates 2/3 of CD45(+) hematopoietic cells. Both Runx1 knockout embryos and Vav1-cre; R26-DTA embryos revealed disorganized, hypoplastic microvasculature of coronary vessels on section and whole-mount stainings. Furthermore, coronary explant experiments showed that the mouse heart explants from Runx1 and Vav1-cre; R26-DTA embryos exhibited impaired coronary formation ex vivo. Interestingly, in both models it appears that epicardial to mesenchymal transition is adversely affected in the absence of hematopoietic progenitors. CONCLUSION Hematopoietic cells are not merely passively transported via coronary vessel, but substantially involved in the induction of the coronary growth. Our findings suggest a novel mechanism of coronary growth.
Collapse
Affiliation(s)
- Gentian Lluri
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, Section of Cardiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Vincent Huang
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marlin Touma
- Children's Discovery and Innovation Institute Department of Pediatrics, Department of Molecular Cell and Integrative Physiology, David Geffen School of Medicine, USA
| | - Xiaoqian Liu
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Andrew W Harmon
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Atsushi Nakano
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
20
|
Spinetti G, Mangialardi G, Specchia C, Madeddu P. Enhancing Stem Cell Mobility: New Hope for Treatment of Cardiovascular Complications in Patients With Diabetes? Diabetes 2015. [PMID: 26207034 DOI: 10.2337/db15-0433] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | | | - Claudia Specchia
- IRCCS MultiMedica, Milan, Italy University of Brescia, Brescia, Italy
| | - Paolo Madeddu
- Bristol Heart Institute, University of Bristol, Bristol, U.K.
| |
Collapse
|
21
|
Milasinovic D, Mohl W. Contemporary perspective on endogenous myocardial regeneration. World J Stem Cells 2015; 7:793-805. [PMID: 26131310 PMCID: PMC4478626 DOI: 10.4252/wjsc.v7.i5.793] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 03/01/2015] [Accepted: 04/20/2015] [Indexed: 02/06/2023] Open
Abstract
Considering the complex nature of the adult heart, it is no wonder that innate regenerative processes, while maintaining adequate cardiac function, fall short in myocardial jeopardy. In spite of these enchaining limitations, cardiac rejuvenation occurs as well as restricted regeneration. In this review, the background as well as potential mechanisms of endogenous myocardial regeneration are summarized. We present and analyze the available evidence in three subsequent steps. First, we examine the experimental research data that provide insights into the mechanisms and origins of the replicating cardiac myocytes, including cell populations referred to as cardiac progenitor cells (i.e., c-kit+ cells). Second, we describe the role of clinical settings such as acute or chronic myocardial ischemia, as initiators of pathways of endogenous myocardial regeneration. Third, the hitherto conducted clinical studies that examined different approaches of initiating endogenous myocardial regeneration in failing human hearts are analyzed. In conclusion, we present the evidence in support of the notion that regaining cardiac function beyond cellular replacement of dysfunctional myocardium via initiation of innate regenerative pathways could create a new perspective and a paradigm change in heart failure therapeutics. Reinitiating cardiac morphogenesis by reintroducing developmental pathways in the adult failing heart might provide a feasible way of tissue regeneration. Based on our hypothesis “embryonic recall”, we present first supporting evidence on regenerative impulses in the myocardium, as induced by developmental processes.
Collapse
|
22
|
San Roman JA, Sánchez PL, Villa A, Sanz-Ruiz R, Fernandez-Santos ME, Gimeno F, Ramos B, Arnold R, Serrador A, Gutiérrez H, Martin-Herrero F, Rollán MJ, Fernández-Vázquez F, López-Messa J, Ancillo P, Pérez-Ojeda G, Fernández-Avilés F. Comparison of Different Bone Marrow–Derived Stem Cell Approaches in Reperfused STEMI. J Am Coll Cardiol 2015; 65:2372-82. [DOI: 10.1016/j.jacc.2015.03.563] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 02/23/2015] [Accepted: 03/24/2015] [Indexed: 11/25/2022]
|
23
|
Jun Hong S, Rogers PI, Kihlken J, Warfel J, Bull C, Deuter-Reinhard M, Feng D, Xie J, Kyle A, Merfeld-Clauss S, Johnstone BH, Traktuev DO, Chen PS, Lindner JR, March KL. Intravenous xenogeneic transplantation of human adipose-derived stem cells improves left ventricular function and microvascular integrity in swine myocardial infarction model. Catheter Cardiovasc Interv 2015; 86:E38-48. [PMID: 24905889 DOI: 10.1002/ccd.25566] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 05/25/2014] [Indexed: 01/25/2023]
Abstract
OBJECTIVES The potential for beneficial effects of adipose-derived stem cells (ASCs) on myocardial perfusion and left ventricular dysfunction in myocardial ischemia (MI) has not been tested following intravenous delivery. METHODS Surviving pigs following induction of MI were randomly assigned to 1 of 3 different groups: the placebo group (n = 7), the single bolus group (SB) (n = 7, 15 × 10(7) ASCs), or the divided dose group (DD) (n = 7, 5 × 10(7) ASCs/day for three consecutive days). Myocardial perfusion defect area and coronary flow reserve (CFR) were compared during the 28-day follow-up. Also, serial changes in the absolute number of circulating CD4(+) T and CD8(+) T cells were measured. RESULTS The increases in ejection fraction were significantly greater in both the SB and the DD groups compared to the placebo group (5.4 ± 0.9%, 3.7 ± 0.7%, and -0.4 ± 0.6%, respectively), and the decrease in the perfusion defect area was significantly greater in the SB group than the placebo group (-36.3 ± 1.8 and -11.5 ± 2.8). CFR increased to a greater degree in the SB and the DD groups than in the placebo group (0.9 ± 0.2, 0.8 ± 0.1, and 0.2 ± 0.2, respectively). The circulating number of CD8(+) T cells was significantly greater in the SB and DD groups than the placebo group at day 7 (3,687 ± 317/µL, 3,454 ± 787/µL, and 1,928 ± 457/µL, respectively). The numbers of small vessels were significantly greater in the SB and the DD groups than the placebo group in the peri-infarct area. CONCLUSIONS Both intravenous SB and DD delivery of ASCs are effective modalities for the treatment of MI in swine. Intravenous delivery of ASCs, with its immunomodulatory and angiogenic effects, is an attractive noninvasive approach for myocardial rescue.
Collapse
Affiliation(s)
- Soon Jun Hong
- Krannert Institute of Cardiology, Indiana University, Indianapolis, Indiana.,Indiana Center for Vascular Biology and Medicine, Indianapolis, Indiana.,Indiana University School of Medicine, Indianapolis, Indiana.,R.L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana.,Korea University Anam Hospital, Seoul, Korea
| | - Pamela I Rogers
- Krannert Institute of Cardiology, Indiana University, Indianapolis, Indiana.,Indiana Center for Vascular Biology and Medicine, Indianapolis, Indiana.,Indiana University School of Medicine, Indianapolis, Indiana.,R.L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| | - John Kihlken
- Indiana University School of Medicine, Indianapolis, Indiana
| | - Jessica Warfel
- Indiana University School of Medicine, Indianapolis, Indiana
| | - Chris Bull
- Indiana University School of Medicine, Indianapolis, Indiana
| | - Maja Deuter-Reinhard
- Krannert Institute of Cardiology, Indiana University, Indianapolis, Indiana.,Indiana Center for Vascular Biology and Medicine, Indianapolis, Indiana.,Indiana University School of Medicine, Indianapolis, Indiana.,R.L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| | - Dongni Feng
- Krannert Institute of Cardiology, Indiana University, Indianapolis, Indiana.,Indiana Center for Vascular Biology and Medicine, Indianapolis, Indiana.,Indiana University School of Medicine, Indianapolis, Indiana.,R.L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| | - Jie Xie
- Krannert Institute of Cardiology, Indiana University, Indianapolis, Indiana.,Indiana Center for Vascular Biology and Medicine, Indianapolis, Indiana.,Indiana University School of Medicine, Indianapolis, Indiana.,R.L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| | - Aaron Kyle
- Indiana University School of Medicine, Indianapolis, Indiana
| | - Stephanie Merfeld-Clauss
- Krannert Institute of Cardiology, Indiana University, Indianapolis, Indiana.,Indiana Center for Vascular Biology and Medicine, Indianapolis, Indiana.,Indiana University School of Medicine, Indianapolis, Indiana.,R.L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| | - Brian H Johnstone
- Krannert Institute of Cardiology, Indiana University, Indianapolis, Indiana.,Indiana Center for Vascular Biology and Medicine, Indianapolis, Indiana.,Indiana University School of Medicine, Indianapolis, Indiana.,R.L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| | - Dmitry O Traktuev
- Krannert Institute of Cardiology, Indiana University, Indianapolis, Indiana.,Indiana Center for Vascular Biology and Medicine, Indianapolis, Indiana.,Indiana University School of Medicine, Indianapolis, Indiana.,R.L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| | - Peng-Sheng Chen
- Krannert Institute of Cardiology, Indiana University, Indianapolis, Indiana.,Indiana Center for Vascular Biology and Medicine, Indianapolis, Indiana.,Indiana University School of Medicine, Indianapolis, Indiana
| | | | - Keith L March
- Krannert Institute of Cardiology, Indiana University, Indianapolis, Indiana.,Indiana Center for Vascular Biology and Medicine, Indianapolis, Indiana.,Indiana University School of Medicine, Indianapolis, Indiana.,R.L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| |
Collapse
|
24
|
Jang IH, Heo SC, Kwon YW, Choi EJ, Kim JH. Role of formyl peptide receptor 2 in homing of endothelial progenitor cells and therapeutic angiogenesis. Adv Biol Regul 2014; 57:162-72. [PMID: 25304660 DOI: 10.1016/j.jbior.2014.09.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 09/03/2014] [Indexed: 12/30/2022]
Abstract
Endothelial progenitor cells (EPCs) hold a great promise as a therapeutic mediator in treatment of ischemic disease conditions. The discovery of EPCs in adult blood has been a cause of significant enthusiasm in the field of endothelial cell research and numerous clinical trials have been expedited. After more than a decade of research in basic science and clinical applications, limitations and new strategies of EPC therapeutics have emerged. With various phenotypes, vague definitions, and uncertain distinction from hematopoietic cells, understanding EPC biology remains challenging. However, EPCs, still hold great hope for treatment of critical ischemic injury as low concern regarding safety can accelerate the clinical applications from basic findings. This review provides an introduction to EPC as cellular therapeutics, which highlights a recent finding that EPC homing was promoted through FPR2 signaling.
Collapse
Affiliation(s)
- Il Ho Jang
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Republic of Korea
| | - Soon Chul Heo
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Republic of Korea
| | - Yang Woo Kwon
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Republic of Korea
| | - Eun Jung Choi
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Republic of Korea
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Republic of Korea; Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 626-770, Gyeongsangnam-do, Republic of Korea.
| |
Collapse
|
25
|
Ghodsizad A, Ruhparwar A, Bordel V, Mirsaidighazi E, Klein HM, Koerner MM, Karck M, El-Banayosy A. Clinical application of adult stem cells for therapy for cardiac disease. Cardiovasc Ther 2014; 31:323-34. [PMID: 23773460 DOI: 10.1111/1755-5922.12032] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
INTRODUCTION Cardiovascular disease is a major cause of death worldwide. Different medical and surgical therapeutic options are well established, but a significant number of patients are not amenable to standard therapeutic options. Cell-based therapies after clinical application have shown different results in recent years. Here, we are giving a comprehensive overview on major available clinical data regarding cell therapy. BACKGROUND Cell-based therapies and tissue engineering provide new promising platforms to develop upcoming therapeutic options. Initial clinical trials were able to generate promising results. A variety of different stem cell types have been used for the clinical application. Different adult cardiac stem cells and progenitor cells, including mesenchymal, CD34(+) and CD133(+) autologous human bone marrow-derived stem cells (BMCs), human myoblasts, and peripheral blood-derived stem and progenitor cells (PBSCs) have been used for the therapy for end-stage heart failure. Future experiments will show the importance of novel cell populations and clarify the mechanism causing cell therapy-mediated observed effects. CONCLUSION Several clinical trials have reported on sole therapy, as well as combined application of autologous adult stem cells with conventional revascularization. The reported promising findings encourage further research in the field of the translational research.
Collapse
Affiliation(s)
- Ali Ghodsizad
- Heart and Vascular Institute, Milton S. Hershey Medical Center, College of Medicine, The Pennsylvania State University, Hershey, PA, USA; Department of Cardiac Surgery, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Zhou Y, Singh AK, Hoyt RF, Wang S, Yu Z, Hunt T, Kindzelski B, Corcoran PC, Mohiuddin MM, Horvath KA. Regulatory T cells enhance mesenchymal stem cell survival and proliferation following autologous cotransplantation in ischemic myocardium. J Thorac Cardiovasc Surg 2014; 148:1131-7; discussiom 1117. [PMID: 25052825 DOI: 10.1016/j.jtcvs.2014.06.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 06/06/2014] [Accepted: 06/13/2014] [Indexed: 12/29/2022]
Abstract
OBJECTIVES We sought to investigate if autologous freshly isolated regulatory T cells (Tregs) provide a protective and supportive role when cotransplanted with mesenchymal stem cells (MSCs). METHODS In a porcine model of chronic ischemia, autologous MSCs were isolated and expanded ex vivo for 4 weeks. Autologous Treg cells were freshly isolated from 100 mL peripheral blood and purified by fluorescence-activated cell sorting. MSCs and Treg cells were then cotransplanted into the chronic ischemic myocardium of Yorkshire pigs by direct intramyocardial injection (1.2 × 10(8) MSCs plus an average of 1.5 million Treg cells in 25 injection sites). Animals were killed 6 weeks postinjection to study the fate of the cells and compare the effect of combined MSCs + Treg cells transplantation versus MSCs alone. RESULTS The coinjection of MSCs along with Tregs was safe and no deleterious side effects were observed. Six weeks after injection of the cell combination, spherical MSCs clusters with thin layer capsules were found in the injected areas. In animals treated with MSCs only, the MSC clusters were less organized and not encapsulated. Immunofluorescent staining showed CD25+ cells among the CD90+ (MSC marker) cells, suggesting that the injected Treg cells remained present locally, and survived. Factor VIII+ cells were also prevalent suggesting new angiogenesis. We found no evidence that coinjections were associated with the generation of cardiac myocytes. CONCLUSIONS The cotransplantation of Treg cells with MSCs dramatically increased the MSC survival rate, proliferation, and augmented their role in angiogenesis, which suggests a new way for future clinical application of cell-based therapy.
Collapse
Affiliation(s)
- Yifu Zhou
- Cardiothoracic Surgery Research Program, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md.
| | - Avneesh K Singh
- Cardiothoracic Surgery Research Program, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Robert F Hoyt
- Cardiothoracic Surgery Research Program, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Suna Wang
- Cardiothoracic Surgery Research Program, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Zuxi Yu
- Cardiothoracic Surgery Research Program, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Timothy Hunt
- Cardiothoracic Surgery Research Program, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Bogdan Kindzelski
- Cardiothoracic Surgery Research Program, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Philip C Corcoran
- Cardiothoracic Surgery Research Program, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Muhammad M Mohiuddin
- Cardiothoracic Surgery Research Program, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Keith A Horvath
- Cardiothoracic Surgery Research Program, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| |
Collapse
|
27
|
Hibbert B, Hayley B, Beanlands RS, Le May M, Davies R, So D, Marquis JF, Labinaz M, Froeschl M, O'Brien ER, Burwash IG, Wells GA, Pourdjabbar A, Simard T, Atkins H, Glover C. Granulocyte colony-stimulating factor therapy for stem cell mobilization following anterior wall myocardial infarction: the CAPITAL STEM MI randomized trial. CMAJ 2014; 186:E427-34. [PMID: 24934893 DOI: 10.1503/cmaj.140133] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Small studies have yielded divergent results for administration of granulocyte colony-stimulating factor (G-CSF) after acute myocardial infarction. Adequately powered studies involving patients with at least moderate left ventricular dysfunction are lacking. METHODS Patients with left ventricular ejection fraction less than 45% after anterior-wall myocardial infarction were treated with G-CSF (10 μg/kg daily for 4 days) or placebo. After initial randomization of 86 patients, 41 in the placebo group and 39 in the G-CSF group completed 6-month follow-up and underwent measurement of left ventricular ejection fraction by radionuclide angiography. RESULTS Baseline and 6-week mean ejection fraction was similar for the G-CSF and placebo groups: 34.8% (95% confidence interval [CI] 32.6%-37.0%) v. 36.4% (95% CI 33.5%-39.2%) at baseline and 39.8% (95% CI 36.2%-43.4%) v. 43.1% (95% CI 39.2%-47.0%) at 6 weeks. However, G-CSF therapy was associated with a lower ejection fraction at 6 months relative to placebo (40.8% [95% CI 37.4%-44.2%] v. 46.0% [95% CI 42.7%-44.3%]). Both groups had improved left ventricular function, but change in left ventricular ejection fraction was lower in patients treated with G-CSF than in those who received placebo (5.7 [95% CI 3.4-8.1] percentage points v. 9.2 [95% CI 6.3-12.1] percentage points). One or more of a composite of several major adverse cardiac events occurred in 8 patients (19%) within each group, with similar rates of target-vessel revascularization. INTERPRETATION In patients with moderate left ventricular dysfunction following anterior-wall infarction, G-CSF therapy was associated with a lower 6-month left ventricular ejection fraction but no increased risk of major adverse cardiac events. Future studies of G-CSF in patients with left ventricular dysfunction should be monitored closely for safety. TRIAL REGISTRATION ClinicalTrials.gov, no. NCT00394498.
Collapse
Affiliation(s)
- Benjamin Hibbert
- Division of Cardiology (Hibbert, Hayley, Beanlands, Le May, Davies, So, Marquis, Labinaz, Froeschl, O'Brien, Burwash, Wells, Pourdjabbar, Simard, Glover), Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ont.; Libin Cardiovascular Institute (O'Brien), Calgary, Alta.; Division of Hematology (Atkins), Department of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, Ont
| | - Bradley Hayley
- Division of Cardiology (Hibbert, Hayley, Beanlands, Le May, Davies, So, Marquis, Labinaz, Froeschl, O'Brien, Burwash, Wells, Pourdjabbar, Simard, Glover), Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ont.; Libin Cardiovascular Institute (O'Brien), Calgary, Alta.; Division of Hematology (Atkins), Department of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, Ont
| | - Robert S Beanlands
- Division of Cardiology (Hibbert, Hayley, Beanlands, Le May, Davies, So, Marquis, Labinaz, Froeschl, O'Brien, Burwash, Wells, Pourdjabbar, Simard, Glover), Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ont.; Libin Cardiovascular Institute (O'Brien), Calgary, Alta.; Division of Hematology (Atkins), Department of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, Ont
| | - Michel Le May
- Division of Cardiology (Hibbert, Hayley, Beanlands, Le May, Davies, So, Marquis, Labinaz, Froeschl, O'Brien, Burwash, Wells, Pourdjabbar, Simard, Glover), Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ont.; Libin Cardiovascular Institute (O'Brien), Calgary, Alta.; Division of Hematology (Atkins), Department of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, Ont
| | - Richard Davies
- Division of Cardiology (Hibbert, Hayley, Beanlands, Le May, Davies, So, Marquis, Labinaz, Froeschl, O'Brien, Burwash, Wells, Pourdjabbar, Simard, Glover), Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ont.; Libin Cardiovascular Institute (O'Brien), Calgary, Alta.; Division of Hematology (Atkins), Department of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, Ont
| | - Derek So
- Division of Cardiology (Hibbert, Hayley, Beanlands, Le May, Davies, So, Marquis, Labinaz, Froeschl, O'Brien, Burwash, Wells, Pourdjabbar, Simard, Glover), Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ont.; Libin Cardiovascular Institute (O'Brien), Calgary, Alta.; Division of Hematology (Atkins), Department of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, Ont
| | - Jean-François Marquis
- Division of Cardiology (Hibbert, Hayley, Beanlands, Le May, Davies, So, Marquis, Labinaz, Froeschl, O'Brien, Burwash, Wells, Pourdjabbar, Simard, Glover), Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ont.; Libin Cardiovascular Institute (O'Brien), Calgary, Alta.; Division of Hematology (Atkins), Department of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, Ont
| | - Marino Labinaz
- Division of Cardiology (Hibbert, Hayley, Beanlands, Le May, Davies, So, Marquis, Labinaz, Froeschl, O'Brien, Burwash, Wells, Pourdjabbar, Simard, Glover), Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ont.; Libin Cardiovascular Institute (O'Brien), Calgary, Alta.; Division of Hematology (Atkins), Department of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, Ont
| | - Michael Froeschl
- Division of Cardiology (Hibbert, Hayley, Beanlands, Le May, Davies, So, Marquis, Labinaz, Froeschl, O'Brien, Burwash, Wells, Pourdjabbar, Simard, Glover), Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ont.; Libin Cardiovascular Institute (O'Brien), Calgary, Alta.; Division of Hematology (Atkins), Department of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, Ont
| | - Edward R O'Brien
- Division of Cardiology (Hibbert, Hayley, Beanlands, Le May, Davies, So, Marquis, Labinaz, Froeschl, O'Brien, Burwash, Wells, Pourdjabbar, Simard, Glover), Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ont.; Libin Cardiovascular Institute (O'Brien), Calgary, Alta.; Division of Hematology (Atkins), Department of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, Ont
| | - Ian G Burwash
- Division of Cardiology (Hibbert, Hayley, Beanlands, Le May, Davies, So, Marquis, Labinaz, Froeschl, O'Brien, Burwash, Wells, Pourdjabbar, Simard, Glover), Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ont.; Libin Cardiovascular Institute (O'Brien), Calgary, Alta.; Division of Hematology (Atkins), Department of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, Ont
| | - George A Wells
- Division of Cardiology (Hibbert, Hayley, Beanlands, Le May, Davies, So, Marquis, Labinaz, Froeschl, O'Brien, Burwash, Wells, Pourdjabbar, Simard, Glover), Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ont.; Libin Cardiovascular Institute (O'Brien), Calgary, Alta.; Division of Hematology (Atkins), Department of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, Ont
| | - Ali Pourdjabbar
- Division of Cardiology (Hibbert, Hayley, Beanlands, Le May, Davies, So, Marquis, Labinaz, Froeschl, O'Brien, Burwash, Wells, Pourdjabbar, Simard, Glover), Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ont.; Libin Cardiovascular Institute (O'Brien), Calgary, Alta.; Division of Hematology (Atkins), Department of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, Ont
| | - Trevor Simard
- Division of Cardiology (Hibbert, Hayley, Beanlands, Le May, Davies, So, Marquis, Labinaz, Froeschl, O'Brien, Burwash, Wells, Pourdjabbar, Simard, Glover), Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ont.; Libin Cardiovascular Institute (O'Brien), Calgary, Alta.; Division of Hematology (Atkins), Department of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, Ont
| | - Harold Atkins
- Division of Cardiology (Hibbert, Hayley, Beanlands, Le May, Davies, So, Marquis, Labinaz, Froeschl, O'Brien, Burwash, Wells, Pourdjabbar, Simard, Glover), Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ont.; Libin Cardiovascular Institute (O'Brien), Calgary, Alta.; Division of Hematology (Atkins), Department of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, Ont
| | - Christopher Glover
- Division of Cardiology (Hibbert, Hayley, Beanlands, Le May, Davies, So, Marquis, Labinaz, Froeschl, O'Brien, Burwash, Wells, Pourdjabbar, Simard, Glover), Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ont.; Libin Cardiovascular Institute (O'Brien), Calgary, Alta.; Division of Hematology (Atkins), Department of Medicine, The Ottawa Hospital and University of Ottawa, Ottawa, Ont.
| |
Collapse
|
28
|
Zimmet H, Porapakkham P, Porapakkham P, Sata Y, Haas SJ, Itescu S, Forbes A, Krum H. Short- and long-term outcomes of intracoronary and endogenously mobilized bone marrow stem cells in the treatment of ST-segment elevation myocardial infarction: a meta-analysis of randomized control trials. Eur J Heart Fail 2014; 14:91-105. [DOI: 10.1093/eurjhf/hfr148] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Hendrik Zimmet
- Monash Centre of Cardiovascular Research & Education in Therapeutics, Department of Epidemiology & Preventive Medicine; School of Public Health & Preventive Medicine, Monash University; 99 Commercial Rd Melbourne VIC 3004 Australia
| | - Pramote Porapakkham
- Department of Cardiothoracic Surgery; Chest Disease Institute; Nonthaburi Thailand
| | | | - Yusuke Sata
- Department of Cardiovascular Dynamics; National Cerebral and CardioVascular Center Research Institute; Osaka Japan
| | - Steven Joseph Haas
- Monash Centre of Cardiovascular Research & Education in Therapeutics, Department of Epidemiology & Preventive Medicine; School of Public Health & Preventive Medicine, Monash University; 99 Commercial Rd Melbourne VIC 3004 Australia
| | - Silviu Itescu
- Department of Medicine; University of Melbourne, St. Vincent's Hospital; Melbourne Australia
| | - Andrew Forbes
- Monash Centre of Cardiovascular Research & Education in Therapeutics, Department of Epidemiology & Preventive Medicine; School of Public Health & Preventive Medicine, Monash University; 99 Commercial Rd Melbourne VIC 3004 Australia
| | - Henry Krum
- Monash Centre of Cardiovascular Research & Education in Therapeutics, Department of Epidemiology & Preventive Medicine; School of Public Health & Preventive Medicine, Monash University; 99 Commercial Rd Melbourne VIC 3004 Australia
| |
Collapse
|
29
|
Multicenter prospective nonrandomized controlled clinical trial to prove neurotherapeutic effects of granulocyte colony-stimulating factor for acute spinal cord injury: analyses of follow-up cases after at least 1 year. Spine (Phila Pa 1976) 2014; 39:213-9. [PMID: 24299727 DOI: 10.1097/brs.0000000000000121] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN An open-labeled multicenter prospective nonrandomized controlled clinical trial. OBJECTIVE To confirm the feasibility of using granulocyte colony-stimulating factor (G-CSF) for treatment of acute spinal cord injury (SCI). SUMMARY OF BACKGROUND DATA We previously reported that G-CSF promotes functional recovery after compression-induced SCI in mice. On the basis of these findings, we conducted a multicenter prospective controlled clinical trial to assess the feasibility of G-CSF therapy for patients with acute SCI. METHODS The trial ran from August 2009 to March 2011, and included 41 patients with SCI treated within 48 hours of onset. Informed consent was obtained from all patients. After providing consent, patients were divided into 2 groups. In the G-CSF group (17 patients), G-CSF (10 μg/kg/d) was intravenously administered for 5 consecutive days, and in the control group (24 patients), patients were similarly treated except for the G-CSF administration. We evaluated motor and sensory functions using the American Spinal Cord Injury Association score and American Spinal Cord Injury Association impairment scale at 1 week, 3 months, 6 months, and 1 year after onset. RESULTS Only 2 patients did not experience American Spinal Cord Injury Association impairment scale improvement in the G-CSF group. In contrast, 15 patients in the control group did not experience American Spinal Cord Injury Association impairment scale improvement. In the analysis of increased American Spinal Cord Injury Association motor score, a significant increase in G-CSF group was detected from 1 week after the administration compared with the control group. After that, some spontaneous increase of motor score was detected in control group, but the significant increase in G-CSF group was maintained until 1 year of follow-up. CONCLUSION Despite the limitation that patient selection was not randomized, the present results suggest the possibility that G-CSF administration has beneficial effects on neurological recovery in patients with acute SCI. LEVEL OF EVIDENCE 3.
Collapse
|
30
|
Granulocyte colony-stimulating factor attenuates left ventricular remodelling after acute anterior STEMI: results of the single-blind, randomized, placebo-controlled multicentre STem cEll Mobilization in Acute Myocardial Infarction (STEM-AMI) Trial. Eur J Heart Fail 2014; 12:1111-21. [DOI: 10.1093/eurjhf/hfq150] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
31
|
Achilli F, Malafronte C, Maggiolini S, Lenatti L, Squadroni L, Gibelli G, Capogrossi MC, Dadone V, Gentile F, Bassetti B, Di Gennaro F, Camisasca P, Calchera I, Valagussa L, Colombo GI, Pompilio G. G-CSF treatment for STEMI: final 3-year follow-up of the randomised placebo-controlled STEM-AMI trial. Heart 2014; 100:574-81. [DOI: 10.1136/heartjnl-2013-304955] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
32
|
Foresta C, De Toni L, Ferlin A, Di Mambro A. Clinical implication of endothelial progenitor cells. Expert Rev Mol Diagn 2014; 10:89-105. [DOI: 10.1586/erm.09.80] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
33
|
Gili M, Orsello A, Gallo S, Brizzi MF. Diabetes-associated macrovascular complications: cell-based therapy a new tool? Endocrine 2013; 44:557-75. [PMID: 23543434 DOI: 10.1007/s12020-013-9936-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 03/20/2013] [Indexed: 01/01/2023]
Abstract
Diabetes mellitus and its ongoing macrovascular complications represent one of the major health problems around the world. Rise in obesity and population ages correlate with the increased incidence of diabetes. This highlights the need for novel approaches to prevent and treat this pandemic. The discovery of a reservoir of stem/progenitors in bone marrow and in mesenchymal tissue has attracted interest of both biologists and clinicians. A number of preclinical and clinical trials were developed to explore their potential clinical impact, as target or vehicle, in different clinical settings, including diabetes complications. Currently, bone marrow, peripheral blood, mesenchymal, and adipose tissues have been used as stem/progenitor cell sources. However, evidences have been provided that both bone marrow and circulating progenitor cells are dysfunctional in diabetes. These observations along with the growing advantages in genetic manipulation have spurred researchers to exploit ex vivo manipulated cells to overcome these hurdles. In this article, we provide an overview of data relevant to stem-progenitors potential clinical application in revascularization and/or vascular repair. Moreover, the hurdles at using progenitor cells in diabetic patients will be also discussed.
Collapse
Affiliation(s)
- Maddalena Gili
- Department of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | | | | | | |
Collapse
|
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: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
35
|
Morris MW, Liechty KW. Cardiac Progenitor Cells in Myocardial Infarction Wound Healing: A Critical Review. Adv Wound Care (New Rochelle) 2013; 2:317-326. [PMID: 24527353 PMCID: PMC3751317 DOI: 10.1089/wound.2012.0390] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Indexed: 01/14/2023] Open
Abstract
SIGNIFICANCE Coronary artery disease is a major cause of morbidity and mortality as the loss of functional myocardium drives progressive ventricular remodeling and subsequent heart failure. Medical management has significantly improved outcomes for acute myocardial infarction (MI); however, improved strategies are needed to regenerate functional myocardium and prevent the progression to heart failure. Cytotherapy using cardiac progenitor cells (PCs) to regenerate functional myocardium holds tremendous potential; however, a better understanding of PC biology is needed. RECENT ADVANCES Reports of cardiac regeneration in lower animals have been reported in the last decade. However, just recently, two separate models of mammalian cardiac regeneration have been published and offer potential to better define PC biology, including PC recruitment, differentiation, proliferation, and integration. CRITICAL ISSUES Numerous clinical trials have been completed or are ongoing to evaluate possible cytotherapy options in the treatment of acute and chronic ischemic cardiac disease. To date, results have demonstrated improvements in cardiac function as a result of paracrine effects of cytotherapy, but regeneration of functional myocardium has yet to be observed. FUTURE DIRECTIONS Future translation of cardiac PC biology from these models is necessary to promote regenerative cardiac healing following MI and to prevent the progression to heart failure following the loss of functional myocardium. Knowledge gained from mammalian models of cardiac regeneration will allow for the development of therapeutic regimens in the treatment of heart failure.
Collapse
Affiliation(s)
- Michael W. Morris
- Department of Surgery, University of Mississippi Medical Center, Jackson, Mississippi
| | - Kenneth W. Liechty
- Department of Pediatric Surgery, Nemours Children's Hospital, Orlando, Florida
| |
Collapse
|
36
|
Moazzami K, Roohi A, Moazzami B. Granulocyte colony stimulating factor therapy for acute myocardial infarction. Cochrane Database Syst Rev 2013; 2013:CD008844. [PMID: 23728682 PMCID: PMC8454260 DOI: 10.1002/14651858.cd008844.pub2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Acute myocardial infarction (AMI) is the leading cause of death in developed countries, and current treatment modalities have failed to regenerate the dead myocardium resulting from the ischemic damage. Stem cells have the potential to regenerate the damaged myocardium. These cells can be mobilized from the bone marrow by factors such as granulocyte colony stimulating factor (G-CSF). OBJECTIVES To assess the effects of stem cell mobilization following granulocyte colony stimulating factor therapy in patients with acute myocardial infarction. SEARCH METHODS We searched CENTRAL (The Cochrane Library Issue 4, 2010), MEDLINE (1950 to November week 3, 2010), EMBASE (1980 to 2010 week 48), BIOSIS Previews (1969 to 30 November 2010), ISI Science Citation Index Expanded (1970 to 4 December 2010) and ISI Conference Proceedings Citation Index - Science (1990 to 4 December 2010). We also checked reference lists of articles. SELECTION CRITERIA We included randomized controlled trials including participants with a clinical diagnosis of AMI who were randomly allocated to the subcutaneous administration of G-CSF through a daily dose of 2.5, 5 or 10 microgram/kg for four to six days or placebo. No age or other restrictions were applied for the selection of patients. DATA COLLECTION AND ANALYSIS Two authors independently selected trials, assessed trials for eligibility and methodological quality, and extracted data regarding the clinical efficacy and adverse outcomes. Disagreements were resolved by the third author. MAIN RESULTS We included seven trials reported in 30 references in the review (354 participants). In all trials, G-CSF was compared with placebo preparations. Dosage of G-CSF varied among studies, ranging from 2.5 to 10 microgram/kg/day. Regarding overall risk of bias, data regarding the generation of randomization sequence and incomplete outcome data were at a low risk of bias; however, data regarding binding of personnel were not conclusive. The rate of mortality was not different between the two groups (RR 0.64, 95% CI 0.15 to 2.80, P = 0.55). Regarding safety, the limited amount of evidence is inadequate to reach any conclusions regarding the safety of G-CSF therapy. Moreover, the results did not show any beneficial effects of G-CSF in patients with AMI regarding left ventricular function parameters, including left ventricular ejection fraction (RR 3.41, 95% CI -0.61 to 7.44, P = 0.1), end systolic volume (RR -1.35, 95% CI -4.68 to 1.99, P = 0.43) and end diastolic volume (RR -4.08, 95% CI -8.28 to 0.12, P = 0.06). It should also be noted that the study was limited since the trials included lacked long enough follow up durations. AUTHORS' CONCLUSIONS Limited evidence from small trials suggested a lack of benefit of G-CSF therapy in patients with AMI. Since data of the risk of bias regarding blinding of personnel were not conclusive, larger RCTs with appropriate power calculations and longer follow up durations are required in order to address current uncertainties regarding the clinical efficacy and therapy-related adverse events of G-CSF treatment.
Collapse
Affiliation(s)
- Kasra Moazzami
- Cardiovascular ResearchCenter (CVRC), Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| | | | | |
Collapse
|
37
|
Ripa RS, Jørgensen E, Kastrup J. Clinical outcome after stem cell mobilization with granulocyte-colony-stimulating factor after acute ST-elevation myocardial infarction: 5-year results of the STEMMI trial. Scandinavian Journal of Clinical and Laboratory Investigation 2013; 73:125-9. [PMID: 23281844 DOI: 10.3109/00365513.2012.750010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Granulocyte-colony-stimulating factor (G-CSF) has been investigated in trials aiming to promote recovery of myocardial function after myocardial infarction. Long-term safety-data have never been reported. A few studies indicated an increased risk of in-stent re-stenosis. We aimed to investigate clinical events 5 years after inclusion into a randomized trial of G-CSF versus placebo. METHODS Seventy-eight patients were randomized, from 2003-2005, to G-CSF or placebo after myocardial infarction. Four patients withdrew consent prior to study treatment and were excluded leaving 36 and 38 in the placebo- and G-CSF groups. Information about all hospital admittances of included patients until 2010 was extracted from a national register. The only censoring event was immigration. The events were combined into four prespecified endpoints: Time to (1) first hospital admittance (all cause), (2) first cardiovascular-related hospital admittance, (3) first major cardiovascular event, and (4) death. RESULTS One patient (1%) was lost to follow-up. Four patients (4%) died in the follow-up period, three in the G-CSF group and one in the placebo group (p = 0.4). Hazard ratio for all cause hospital admittance was 0.7 (95% CI 0.38-1.29). The incidence of both new myocardial infarction (p = 1.0) and revascularization procedures (p = 0.4) were similar in the two groups. Survival analyses showed no differences in the occurrence of any of the four prespecified composite endpoints between the two groups (p = 0.6; 0.5; 0.8; 0.3). CONCLUSIONS We found no indication of increased risk of adverse events up to 5 years after G-CSF treatment. These results support the continued investigation of G-CSF for cardiac therapy.
Collapse
Affiliation(s)
- Rasmus S Ripa
- Department of Cardiology, Nuclear Medicine and PET, Copenhagen University Hospital, Rigshospitalet, Denmark.
| | | | | |
Collapse
|
38
|
Obana M, Miyamoto K, Murasawa S, Iwakura T, Hayama A, Yamashita T, Shiragaki M, Kumagai S, Miyawaki A, Takewaki K, Matsumiya G, Maeda M, Yoshiyama M, Nakayama H, Fujio Y. Therapeutic administration of IL-11 exhibits the postconditioning effects against ischemia-reperfusion injury via STAT3 in the heart. Am J Physiol Heart Circ Physiol 2012; 303:H569-77. [DOI: 10.1152/ajpheart.00060.2012] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Activation of cardiac STAT3 by IL-6 cytokine family contributes to cardioprotection. Previously, we demonstrated that IL-11, an IL-6 cytokine family, has the therapeutic potential to prevent adverse cardiac remodeling after myocardial infarction; however, it remains to be elucidated whether IL-11 exhibits postconditioning effects. To address the possibility that IL-11 treatment improves clinical outcome of recanalization therapy against acute myocardial infarction, we examined its postconditioning effects on ischemia/reperfusion (I/R) injury. C57BL/6 mice were exposed to ischemia (30 min) and reperfusion (24 h), and IL-11 was intravenously administered at the start of reperfusion. I/R injury mediated the activation of STAT3, which was enhanced by IL-11 administration. IL-11 treatment reduced I/R injury, analyzed by triphenyl tetrazolium chloride staining [PBS, 46.7 ± 14.4%; IL-11 (20 μg/kg), 28.6 ± 7.5% in the ratio of infarct to risk area]. Moreover, echocardiographic and hemodynamic analyses clarified that IL-11 treatment preserved cardiac function after I/R. Terminal deoxynucleotide transferase-mediated dUTP nick-end labeling staining revealed that IL-11 reduced the frequency of apoptotic cardiomyocytes after I/R. Interestingly, IL-11 reduced superoxide production assessed by in situ dihydroethidium fluorescence analysis, accompanied by the increased expression of metallothionein 1 and 2, reactive oxygen species (ROS) scavengers. Importantly, with the use of cardiac-specific STAT3 conditional knockout (STAT3 CKO) mice, it was revealed that cardiac-specific ablation of STAT3 abrogated IL-11-mediated attenuation of I/R injury. Finally, IL-11 failed to suppress the ROS production after I/R in STAT3 CKO mice. IL-11 administration exhibits the postconditioning effects through cardiac STAT3 activation, suggesting that IL-11 has the clinical therapeutic potential to prevent I/R injury in heart.
Collapse
Affiliation(s)
- Masanori Obana
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Kaori Miyamoto
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Shiho Murasawa
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Tomohiko Iwakura
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Akiko Hayama
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Tomomi Yamashita
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Momoko Shiragaki
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Shohei Kumagai
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Akimitsu Miyawaki
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Kana Takewaki
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Goro Matsumiya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Makiko Maeda
- Department of Clinical Pharmacogenomics, School of Pharmacy, Hyogo University of Health Sciences
| | - Minoru Yoshiyama
- Department of Internal Medicine and Cardiology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Hiroyuki Nakayama
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Yasushi Fujio
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| |
Collapse
|
39
|
Díez Villanueva P, Sanz-Ruiz R, Núñez García A, Fernández Santos ME, Sánchez PL, Fernández-Avilés F. Functional multipotency of stem cells: what do we need from them in the heart? Stem Cells Int 2012; 2012:817364. [PMID: 22966237 PMCID: PMC3433152 DOI: 10.1155/2012/817364] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 07/22/2012] [Accepted: 07/22/2012] [Indexed: 12/14/2022] Open
Abstract
After more than ten years of human research in the field of cardiac regenerative medicine, application of stem cells in different phases of ischemic heart disease has come to age. Randomized clinical trials have demonstrated that stem cell therapy can improve cardiac recovery after the acute phase of myocardial ischemia and in patients with chronic ischemic heart disease, and several efficacy phase III trials with clinical endpoints are on their way. Nevertheless, a complete knowledge on the mechanisms of action of stem cells still remains elusive. Of the three main mechanisms by which stem cells could exert their benefit, paracrine signaling from the administered cells and stimulation of endogenous repair are nowadays the most plausible ones. However, in this review we will define and discuss the concept of stem cell potency and differentiation, will examine the evidence available, and will depict future directions of research.
Collapse
Affiliation(s)
- Pablo Díez Villanueva
- Cardiology Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Ricardo Sanz-Ruiz
- Cardiology Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Alberto Núñez García
- Cardiology Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | | - Pedro L. Sánchez
- Cardiology Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | |
Collapse
|
40
|
Abstract
Therapeutic angiogenesis aims at treating ischemic diseases by generating new blood vessels from existing vasculature. It relies on delivery of exogenous factors to stimulate neovasculature formation. Current strategies using genes, proteins and cells have demonstrated efficacy in animal models. However, clinical translation of any of the three approaches has proved to be challenging for various reasons. Administration of angiogenic factors is generally considered safe, according to accumulated trials, and offers off-the-shelf availability. However, many hurdles must be overcome before therapeutic angiogenesis can become a true human therapy. This article will highlight protein-based therapeutic angiogenesis, concisely review recent progress and examine critical challenges. We will discuss growth factors that have been widely utilized in promoting angiogenesis and compare their targets and functions. Lastly, since bolus injection of free proteins usually result in poor outcomes, we will focus on controlled release of proteins.
Collapse
|
41
|
Nagai T, Komuro I. Gene and cytokine therapy for heart failure: molecular mechanisms in the improvement of cardiac function. Am J Physiol Heart Circ Physiol 2012; 303:H501-12. [PMID: 22777420 DOI: 10.1152/ajpheart.00130.2012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite significant advances in pharmacological and clinical treatment, heart failure (HF) remains a leading cause of morbidity and mortality worldwide. Many new therapeutic strategies, including cell transplantation, gene delivery, and cytokines or other small molecules, have been explored to treat HF. Recent advancement of our understanding of the molecules that regulate cardiac function uncover many of the therapeutic key molecules to treat HF. Furthermore, a theory of paracrine mechanism, which underlies the beneficial effects of cell therapy, leads us to search novel target molecules for genetic or pharmacological strategy. Gene therapy means delivery of genetic materials into cells to achieve therapeutic effects. Recently, gene transfer technology in the cardiovascular system has been improved and several therapeutic target genes have been started to examine in clinical research, and some of the promising results have been emerged. Among the various bioactive reagents, cytokines such as granulocyte colony-stimulating factor and erythropoietin have been well examined, and a number of clinical trials for acute myocardial infarction and chronic HF have been conducted. Although further research is needed in both preclinical and clinical areas in terms of molecular mechanisms, safety, and efficiency, both gene and cytokine therapy have a great possibility to open the new era of the treatment of HF.
Collapse
Affiliation(s)
- Toshio Nagai
- Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | | |
Collapse
|
42
|
Mullenix PS, Huddleston SJ, Stojadinovic A, Trachiotis GD, Alexander EP. A new heart: somatic stem cells and myocardial regeneration. J Surg Oncol 2012; 105:475-80. [PMID: 22441899 DOI: 10.1002/jso.23014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
End-stage heart failure is a global scourge. Current therapies merely delay its inexorable progression. Heart transplantation is resource-intensive and limited by organ availability. Bone marrow-derived and cardiac-specific stem cells have demonstrated potential for cardiac regeneration and repair, but the magnitude and durability of these promising findings are inconsistent. The purpose of this review is to (1) describe cells currently being investigated, (2) outline the status of current trials, and (3) discuss key objectives of future research.
Collapse
Affiliation(s)
- Philip S Mullenix
- Department of Surgery, Veterans Affairs Medical Center, Washington, District of Columbia, Washington, District of Columbia 20307, USA.
| | | | | | | | | |
Collapse
|
43
|
Cencioni C, Capogrossi MC, Napolitano M. The SDF-1/CXCR4 axis in stem cell preconditioning. Cardiovasc Res 2012; 94:400-7. [PMID: 22451511 DOI: 10.1093/cvr/cvs132] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We review the pivotal role of the stromal derived factor (SDF)-1 chemokine in tissue ischaemia and how it orchestrates the rapid revascularization of injured, ischaemic, and regenerating tissues via the CXC chemokine receptors CXCR4 and CXCR7. Furthermore, we discuss the effects of preconditioning (PC), which is a well-known protective phenomenon for tissue ischaemia. The positive effect of both hypoxic and acidic PC on progenitor cell therapeutic potential is reviewed, while stressing the role of the SDF-1/CXCR4 axis in this process.
Collapse
Affiliation(s)
- Chiara Cencioni
- Laboratorio di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino-IRCCS, Via Carlo Parea 4, 20138 Milan, Italy
| | | | | |
Collapse
|
44
|
Qujeq D, Abassi R, Faeizi F, Parsian H, Faraji AS, Taheri H, Tatar M, Elmi MM, Halalkhor S. Effect of granulocyte colony-stimulating factor administration on tissue regeneration due to carbon tetrachloride–induced liver damage in experimental model. Toxicol Ind Health 2012; 29:498-503. [DOI: 10.1177/0748233712440136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The mechanism by which granulocyte colony-stimulating factor (G-CSF) could lead to the protection from liver injury is not well known. Therefore, the resolution of this role needs further basic and clinical experimental investigation. Acute liver injury was induced in rats by single intraperitoneal injection of a 0.50-mL/kg dose of carbon tetrachloride (CCl4 ). Granulocyte colony-stimulating factor or vehicle of 150 µg/kg was given immediately after intoxicating the liver by CCl4 . The animals were divided into four groups of twelve each. Administration of G-CSF caused a decrease in the activity of liver enzymes, aminotransferases, compared with the untreated group.
Collapse
Affiliation(s)
- Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Babol University of Medical Sciences, Babol, Iran
- Department of Biochemistry and Biophysics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Roya Abassi
- Department of Biochemistry and Biophysics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Farideh Faeizi
- Department of Anatomical Sciences, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Hadi Parsian
- Department of Biochemistry and Biophysics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Alieh Sohan Faraji
- Department of Anatomical Sciences, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Hassan Taheri
- Department of Internal Medicine, Ayatollah Rouhani Hospital, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mohsen Tatar
- Department of Biochemistry and Biophysics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Marym M Elmi
- Cellular and Molecular Biology Research Center (CMBRC), Babol University of Medical Sciences, Babol, Iran
| | - Sohhrab Halalkhor
- Department of Biochemistry and Biophysics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
45
|
Neuroprotective therapy using granulocyte colony-stimulating factor for acute spinal cord injury: a phase I/IIa clinical trial. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2012; 21:2580-7. [PMID: 22391867 DOI: 10.1007/s00586-012-2213-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Revised: 01/10/2012] [Accepted: 02/17/2012] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Granulocyte colony-stimulating factor (G-CSF) is a cytokine that is clinically used to treat neutropenia. G-CSF also has non-hematopoietic functions and could potentially be used to treat neuronal injury. To confirm the safety and feasibility of G-CSF administration for acute spinal cord injury (SCI), we have initiated a phase I/IIa clinical trial of neuroprotective therapy using G-CSF. METHODS The trial included a total of 16 SCI patients within 48 h of onset. In the first step, G-CSF (5 μg/kg/day) was intravenously administered for 5 consecutive days to 5 patients. In the second step, G-CSF (10 μg/kg/day) was similarly administered to 11 patients. We evaluated motor and sensory functions of patients using the American Spinal Cord Injury Association (ASIA) score and ASIA impairment scale (AIS) grade. RESULTS In all 16 patients, neurological improvement was obtained after G-CSF administration. AIS grade increased by one step in 9 of 16 patients. A significant increase in ASIA motor scores was detected 1 day after injection (P < 0.01), and both light touch and pin prick scores improved 2 days after injection (P < 0.05) in the 10 μg group. No severe adverse effects were observed after G-CSF injection. CONCLUSION These results indicate that intravenous administration of G-CSF (10 μg/kg/day) for 5 days is essentially safe, and suggest that some neurological recovery may occur in most patients. We suggest that G-CSF administration could be therapeutic for patients with acute SCI.
Collapse
|
46
|
Sakuma T, Yamazaki M, Okawa A, Takahashi H, Kato K, Hashimoto M, Hayashi K, Furuya T, Fujiyoshi T, Kawabe J, Mannoji C, Kadota R, Hashimoto M, Takahashi K, Koda M. Neuroprotective therapy using granulocyte colony-stimulating factor for patients with worsening symptoms of compression myelopathy, Part 1: a phase I and IIa clinical trial. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2012; 21:482-9. [PMID: 21935680 PMCID: PMC3296845 DOI: 10.1007/s00586-011-2020-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 07/30/2011] [Accepted: 09/03/2011] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Based on the neuroprotective effects of granulocyte colony-stimulating factor (G-CSF) on experimental spinal cord injury, we initiated a clinical trial that evaluated the safety and efficacy of neuroprotective therapy using G-CSF for patients with worsening symptoms of compression myelopathy. METHODS We obtained informed consent from 15 patients, in whom the Japanese Orthopaedic Association (JOA) score for cervical myelopathy decreased two points or more during a recent 1-month period. G-CSF (5 or 10 μg/kg/day) was intravenously administered for five consecutive days. We evaluated motor and sensory functions of the patients and the presence of adverse events related to G-CSF therapy. RESULTS G-CSF administration suppressed the progression of myelopathy in all 15 patients. Neurological improvements in motor and sensory functions were obtained in all patients after the administration, although the degree of improvement differed among the patients. Nine patients in the 10-μg group (n=10) underwent surgical treatment at 1 month or later after G-CSF administration. In the 10-μg group, the mean JOA recovery rates 1 and 6 months after administration were 49.9±15.1 and 59.1±16.3%, respectively. On the day following the start of G-CSF therapy, the white blood cell count increased to more than 22,700 cells/mm3. It varied from 12,000 to 50,000 and returned to preadministration levels 3 days after completing G-CSF treatment. No serious adverse events occurred during or after treatment. CONCLUSION The results indicate that G-CSF administration at 10 μg/kg/day is safe for patients with worsening symptoms of compression myelopathy and may be effective for their neurological improvement.
Collapse
Affiliation(s)
- Tsuyoshi Sakuma
- Spine Section, Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| | - Masashi Yamazaki
- Spine Section, Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| | - Akihiko Okawa
- Spine Section, Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| | - Hiroshi Takahashi
- Spine Section, Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| | - Kei Kato
- Spine Section, Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| | - Mitsuhiro Hashimoto
- Spine Section, Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| | - Koichi Hayashi
- Spine Section, Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| | - Takeo Furuya
- Spine Section, Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| | - Takayuki Fujiyoshi
- Spine Section, Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| | - Junko Kawabe
- Spine Section, Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| | - Chikato Mannoji
- Spine Section, Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| | - Ryo Kadota
- Spine Section, Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| | - Masayuki Hashimoto
- Spine Section, Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| | - Kazuhisa Takahashi
- Spine Section, Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| | - Masao Koda
- Spine Section, Department of Orthopaedic Surgery, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677 Japan
| |
Collapse
|
47
|
Uchiyama R, Hasegawa H, Kameda Y, Ueda K, Kobayashi Y, Komuro I, Takano H. Role of regulatory T cells in atheroprotective effects of granulocyte colony-stimulating factor. J Mol Cell Cardiol 2012; 52:1038-47. [PMID: 22285481 DOI: 10.1016/j.yjmcc.2011.12.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 12/07/2011] [Accepted: 12/29/2011] [Indexed: 01/01/2023]
Abstract
We and others have previously reported that granulocyte colony-stimulating factor (G-CSF) prevents left ventricular remodeling and dysfunction after myocardial infarction in animal models and human. We have also reported that G-CSF inhibits the progression of atherosclerosis in animal models, but its precise mechanism is still elusive. So, we examined the effects of G-CSF on atherosclerosis in apolipoprotein E-deficient (ApoE(-/-)) mice. Twelve-week-old male ApoE(-/-) mice were subcutaneously administrated with 200 μg/kg of G-CSF or saline once a day for 5 consecutive days per a week for 4 weeks. Atherosclerotic lesion of aortic sinus was significantly reduced in the G-CSF-treated mice compared with the saline-treated mice (35% reduction, P<0.05). G-CSF significantly reduced the expression level of interferon-γ by 31% and increased the expression level of interleukin-10 by 20% in atherosclerotic lesions of aortic sinus. G-CSF increased the number of CD4(+)CD25(+) regulatory T cells in lymph nodes and spleen, and enhanced the suppressive function of regulatory T cells in vitro. G-CSF markedly increased the number of Foxp3-positive regulatory T cells in atherosclerotic lesions of aortic sinus. Administration of anti-CD25 antibody (PC61) that depletes regulatory T cells abrogated these atheroprotective effects of G-CSF. Moreover, in ApoE(-/-)/CD28(-/-) mice, that lack regulatory T cells, the protective effects of G-CSF on atherosclerosis were not recognized. These findings suggest that regulatory T cells play an important role in the atheroprotective effects of G-CSF.
Collapse
Affiliation(s)
- Raita Uchiyama
- Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | | | | | | | | | | | | |
Collapse
|
48
|
Yang C, Deng Z, Jiang X, Han K, Zhang T, Zhu W, Geng T, Chen X, Ma A. Long-term effects of primary early granulo-monocyte colony-stimulating factor treatment on the left ventricular function and remodeling in patients with acute myocardial infarction with five-year follow-up. Int J Cardiol 2012; 155:287-90. [PMID: 22222425 DOI: 10.1016/j.ijcard.2011.11.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 11/16/2011] [Accepted: 11/24/2011] [Indexed: 11/19/2022]
|
49
|
Hocher B, Sharkovska Y, Mark M, Klein T, Pfab T. The novel DPP-4 inhibitors linagliptin and BI 14361 reduce infarct size after myocardial ischemia/reperfusion in rats. Int J Cardiol 2012; 167:87-93. [PMID: 22217485 DOI: 10.1016/j.ijcard.2011.12.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 11/11/2011] [Accepted: 12/02/2011] [Indexed: 12/29/2022]
Abstract
BACKGROUND Dipeptidylpeptidase-4 inhibition is reported to have beneficial effects on myocardial ischemia. Mechanisms might include a reduced degradation of stromal cell-derived factor-1 alpha with subsequent increased recruitment of circulating stem cells and/or incretin receptor-dependent pathways. This study evaluated the novel xanthine-based dipeptidylpeptidase-4 inhibitors linagliptin (BI 1356) and BI 14361 in cardiac ischemia. METHODS Male Wistar rats were pretreated with linagliptin or BI 14361 and subjected to ligation of the left anterior descending coronary artery for 30 min. RESULTS Dipeptidylpeptidase-4 inhibition significantly reduced the infarct size after 7 days (-27.7%, p<0.05) and 8 weeks (-18.0%, p<0.05). There was a significantly improved maximum rate of left ventricular pressure decline (dP/dt min) in linagliptin-treated animals 8 weeks after ischemia/reperfusion. Apart from that, treatment did not improve cardiac function as determined by echocardiography and cardiac catheterization. Immunohistological staining revealed an increased number of cells positive for stromal cell-derived factor-1 alpha, CXCR-4 and CD34 within and around the infarcted area of BI 14361-treated animals. CONCLUSIONS Linagliptin and BI 14361 are able to reduce infarct size after myocardial ischemia. The immunohistological findings support the hypothesis that dipeptidylpeptidase-4 inhibition via reduced cleavage of stromal cell-derived factor-1 alpha might lead to an enhanced recruitment of CXCR-4+ circulating progenitor cells.
Collapse
Affiliation(s)
- Berthold Hocher
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany.
| | | | | | | | | |
Collapse
|
50
|
Progenitor cell mobilization and recruitment: SDF-1, CXCR4, α4-integrin, and c-kit. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 111:243-64. [PMID: 22917234 DOI: 10.1016/b978-0-12-398459-3.00011-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Progenitor cell retention and release are largely governed by the binding of stromal-cell-derived factor 1 (SDF-1) to CXC chemokine receptor 4 (CXCR4) and by α4-integrin signaling. Both of these pathways are dependent on c-kit activity: the mobilization of progenitor cells in response to either CXCR4 antagonism or α4-integrin blockade is impaired by the loss of c-kit kinase activity; and c-kit-kinase inactivation blocks the retention of CXCR4-positive progenitor cells in the bone marrow. SDF-1/CXCR4 and α4-integrin signaling are also crucial for the retention of progenitor cells in the ischemic region, which may explain, at least in part, why clinical trials of progenitor cell therapy have failed to display the efficacy observed in preclinical investigations. The lack of effectiveness is often attributed to poor retention of the transplanted cells and, to date, most of the trial protocols have mobilized cells with injections of granulocyte colony-stimulating factor (G-CSF), which activates extracellular proteases that irreversibly cleave cell-surface adhesion molecules, including α4-integrin and CXCR4. Thus, the retention of G-CSF-mobilized cells in the ischemic region may be impaired, and the mobilization of agents that reversibly disrupt SDF-1/CXCR4 binding, such as AMD3100, may improve patient response. Efforts to supplement SDF-1 levels in the ischemic region may also improve progenitor cell recruitment and the effectiveness of stem cell therapy.
Collapse
|