1
|
Tsai IT, Sun CK. Stem Cell Therapy against Ischemic Heart Disease. Int J Mol Sci 2024; 25:3778. [PMID: 38612587 PMCID: PMC11011361 DOI: 10.3390/ijms25073778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Ischemic heart disease, which is one of the top killers worldwide, encompasses a series of heart problems stemming from a compromised coronary blood supply to the myocardium. The severity of the disease ranges from an unstable manifestation of ischemic symptoms, such as unstable angina, to myocardial death, that is, the immediate life-threatening condition of myocardial infarction. Even though patients may survive myocardial infarction, the resulting ischemia-reperfusion injury triggers a cascade of inflammatory reactions and oxidative stress that poses a significant threat to myocardial function following successful revascularization. Moreover, despite evidence suggesting the presence of cardiac stem cells, the fact that cardiomyocytes are terminally differentiated and cannot significantly regenerate after injury accounts for the subsequent progression to ischemic cardiomyopathy and ischemic heart failure, despite the current advancements in cardiac medicine. In the last two decades, researchers have realized the possibility of utilizing stem cell plasticity for therapeutic purposes. Indeed, stem cells of different origin, such as bone-marrow- and adipose-derived mesenchymal stem cells, circulation-derived progenitor cells, and induced pluripotent stem cells, have all been shown to play therapeutic roles in ischemic heart disease. In addition, the discovery of stem-cell-associated paracrine effects has triggered intense investigations into the actions of exosomes. Notwithstanding the seemingly promising outcomes from both experimental and clinical studies regarding the therapeutic use of stem cells against ischemic heart disease, positive results from fraud or false data interpretation need to be taken into consideration. The current review is aimed at overviewing the therapeutic application of stem cells in different categories of ischemic heart disease, including relevant experimental and clinical outcomes, as well as the proposed mechanisms underpinning such observations.
Collapse
Affiliation(s)
- I-Ting Tsai
- Department of Emergency Medicine, E-Da Hospital, I-Shou University, Kaohsiung City 82445, Taiwan;
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Cheuk-Kwan Sun
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan
- Department of Emergency Medicine, E-Da Dachang Hospital, I-Shou University, Kaohsiung City 80794, Taiwan
| |
Collapse
|
2
|
Maslovaric M, Fatic N, Delević E. State of the art of stem cell therapy for ischaemic cardiomyopathy. Part 1. ANGIOLOGII︠A︡ I SOSUDISTAI︠A︡ KHIRURGII︠A︡ = ANGIOLOGY AND VASCULAR SURGERY 2019; 25:39-52. [PMID: 31503246 DOI: 10.33529/angio2019324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Ischemic cardiomyopathy is becoming a leading cause of morbidity and mortality in the whole world. Stem cell-based therapy is emerging as a promising option for treatment of ischemic cardiomyopathy. Several stem cell types, including cardiac-derived stem cells, bone marrow-derived stem cells, mesenchymal stem cells, skeletal myoblasts, CD34+ and CD133+ stem cells have been used in clinical trials. Clinical effects mostly depend on transdifferentiation and paracrine factors. One important issue is that a low survival and residential rate of transferred stem cells blocks the effective advances in cardiac improvement. Many other factors associated with the efficacy of cell replacement therapy for ischemic cardiomyopathy mainly including the route of delivery, the type and number of stem cell infusion, the timing of injection, patient's physical conditions, the particular microenvironment onto which the cells are delivered, and clinical conditions remain to be addressed. Here we provide an overview of modern methods of stem cell delivery, types of stem cells and discuss the current state of their therapeutic potential.
Collapse
Affiliation(s)
- Milica Maslovaric
- Prona-Montenegrin Science Promotion Foundation, Podgorica, Montenegro
| | - Nikola Fatic
- Department of Vascular Surgery, Clinical Centre of Montenegro, Podgorica, Montenegro
| | - Emilija Delević
- Medical Faculty in Podgorica, University of Montenegro, Podgorica, Montenegro
| |
Collapse
|
3
|
Sarkar B, Nguyen PK, Gao W, Dondapati A, Siddiqui Z, Kumar VA. Angiogenic Self-Assembling Peptide Scaffolds for Functional Tissue Regeneration. Biomacromolecules 2018; 19:3597-3611. [PMID: 30132656 DOI: 10.1021/acs.biomac.8b01137] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Implantation of acellular biomimetic scaffolds with proangiogenic motifs may have exciting clinical utility for the treatment of ischemic pathologies such as myocardial infarction. Although direct delivery of angiogenic proteins is a possible treatment option, smaller synthetic peptide-based nanostructured alternatives are being investigated due to favorable factors, such as sustained efficacy and high-density epitope presentation of functional moieties. These peptides may be implanted in vivo at the site of ischemia, bypassing the first-pass metabolism and enabling long-term retention and sustained efficacy. Mimics of angiogenic proteins show tremendous potential for clinical use. We discuss possible approaches to integrate the functionality of such angiogenic peptide mimics into self-assembled peptide scaffolds for application in functional tissue regeneration.
Collapse
Affiliation(s)
| | | | | | | | | | - Vivek A Kumar
- Rutgers School of Dental Medicine , Newark , New Jersey 07101 , United States
| |
Collapse
|
4
|
Huang R, Lv H, Yao K, Ge L, Ye Z, Ding H, Zhang Y, Lu H, Huang Z, Zhang S, Zou Y, Ge J. Effects of different doses of granulocyte colony-stimulating factor mobilization therapy on ischemic cardiomyopathy. Sci Rep 2018; 8:5922. [PMID: 29651017 PMCID: PMC5897440 DOI: 10.1038/s41598-018-24020-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/21/2018] [Indexed: 01/14/2023] Open
Abstract
G-CSF mobilization might be beneficial to ICM, but the relationship between effect/safety and the dosage of G-CSF remains unclear. In this study, 24 pigs were used to build ICM models and were randomized into four groups. Four weeks later, different dosages of G-CSF were given daily by subcutaneous injection for 5 days. Another 4 weeks later, all the animals were sacrificed. Electrocardiography, coronary arteriography, left ventriculography, transthoracic echocardiography, cardiac MRI, and SPECT, histopathologic analysis, and immunohistochemistry techniques were used to evaluate left ventricular function and myocardial infarct size. Four weeks after G-CSF treatment, pigs in middle-dose G-CSF group exhibited obvious improvements of left ventricular remodeling and function. Moderate G-CSF mobilization ameliorated the regional contractility of ICM, preserved myocardial viability, and reduced myocardial infarct size. More neovascularization and fewer apoptotic myocardial cells were observed in the ischemic region of the heart in middle-dose group. Expression of vWF, VEGF and MCP-1 were up-regulated, and Akt1 was activated in high- and middle-dose groups. Moreover, CRP, TNF-α and S-100 were elevated after high-dose G-CSF mobilization. Middle-dose G-CSF mobilization therapy is an effective and safe treatment for ICM, and probably acts via a mechanism involving promoting neovascularization, inhibiting cardiac fibrosis and anti-apoptosis.
Collapse
Affiliation(s)
- Rongchong Huang
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, 116011, China
| | - Haichen Lv
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, 116011, China
| | - Kang Yao
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China
| | - Lei Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China
| | - Zhishuai Ye
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, 116011, China
| | - Huaiyu Ding
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, 116011, China
| | - Yiqi Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China
| | - Hao Lu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China
| | - Zheyong Huang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China
| | - Shuning Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China
| | - Yunzeng Zou
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China.,Institutes of Biomedical Science, Fudan University, 138 Dong'an Road, Shanghai, 200032, China
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai, 200032, China. .,Institutes of Biomedical Science, Fudan University, 138 Dong'an Road, Shanghai, 200032, China.
| |
Collapse
|
5
|
Pieper IL, Smith R, Bishop JC, Aldalati O, Chase AJ, Morgan G, Thornton CA. Isolation of Mesenchymal Stromal Cells From Peripheral Blood of ST Elevation Myocardial Infarction Patients. Artif Organs 2017; 41:654-666. [DOI: 10.1111/aor.12829] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 06/16/2016] [Accepted: 06/30/2016] [Indexed: 12/18/2022]
Affiliation(s)
| | - Rachel Smith
- Swansea University Medical School, Institute of Life Science
| | | | - Omar Aldalati
- Regional Cardiac Centre, Morriston Hospital; Swansea Wales UK
| | - Alex J. Chase
- Regional Cardiac Centre, Morriston Hospital; Swansea Wales UK
| | - Gareth Morgan
- Swansea University Medical School, Institute of Life Science
| | | |
Collapse
|
6
|
Choudhury T, Mozid A, Hamshere S, Yeo C, Pellaton C, Arnous S, Saunders N, Brookman P, Jain A, Locca D, Archbold A, Knight C, Wragg A, Davies C, Mills P, Parmar M, Rothman M, Choudry F, Jones DA, Agrawal S, Martin J, Mathur A. An exploratory randomized control study of combination cytokine and adult autologous bone marrow progenitor cell administration in patients with ischaemic cardiomyopathy: the REGENERATE-IHD clinical trial. Eur J Heart Fail 2016; 19:138-147. [PMID: 27790824 PMCID: PMC5248636 DOI: 10.1002/ejhf.676] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 07/21/2016] [Accepted: 07/22/2016] [Indexed: 12/28/2022] Open
Abstract
Aims The effect of combined cytokine and cell therapy in ischaemic cardiomyopathy is unknown. Meta‐analyses suggest improved cardiac function with cell therapy. The optimal cell delivery route remains unclear. We investigated whether granulocyte colony‐stimulating factor (G‐CSF) alone or in combination with intracoronary (i.c.) or intramyocardial (i.m.) injection of autologous bone marrow‐derived cells (BMCs) improves cardiac function. Methods and results Ninety patients with symptomatic ischaemic cardiomyopathy and no further treatment options were enrolled in the randomized, placebo‐controlled, single‐centre REGENERATE‐IHD study. Randomization was to one of three arms: peripheral, i.c., or i.m. In each arm, patients were randomized to active treatment or placebo. All patients, apart from the peripheral placebo group (saline only) received G‐CSF for 5 days. The i.c. and i.m. arms received either BMCs or serum (placebo). The primary endpoint was change in LVEF at 1 year assessed by cardiac magnetic resonance imaging/computed tomography. The i.m. BMC group showed a significant improvement in LVEF of 4.99% (95% confidence interval 0.33–9.6%; P = 0.038) at 1 year. This group also showed a reduction in NYHA class at 1 year and NT‐proBNP at 6 months. No other group showed a significant change in LVEF. This finding is supported by post‐hoc between‐group comparisons. Conclusion We have shown that G‐CSF combined with autologous i.m. BMCs has a beneficial effect on cardiac function and symptoms. However, this result should be considered preliminary in support of a clinical benefit of i.m. stem cell infusion in ‘no option’ patients and needs further exploration in a larger study.
Collapse
Affiliation(s)
- Tawfiq Choudhury
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Abdul Mozid
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Steve Hamshere
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Chia Yeo
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Cyril Pellaton
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Samer Arnous
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Natalie Saunders
- Stem Cell Laboratory, Barts Health NHS Trust and Blizard Institute, Queen Mary University of London, London, UK
| | - Pat Brookman
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Ajay Jain
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Didier Locca
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Andrew Archbold
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Charles Knight
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Andrew Wragg
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Ceri Davies
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Peter Mills
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | | | - Martin Rothman
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Fizzah Choudry
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Daniel A Jones
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Samir Agrawal
- Stem Cell Laboratory, Barts Health NHS Trust and Blizard Institute, Queen Mary University of London, London, UK
| | - John Martin
- British Heart Foundation Laboratories, Department of Medicine, University College London, London, UK
| | - Anthony Mathur
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| |
Collapse
|
7
|
Affiliation(s)
- Jens Kastrup
- The Heart Centre, Rigshospitalet University of Copenhagen, Denmark
| |
Collapse
|
8
|
Worel N, Buser A, Greinix HT, Hägglund H, Navarro W, Pulsipher MA, Nicoloso de Faveri G, Bengtsson M, Billen A, Espino G, Fechter M, Giudice V, Hölig K, Kanamori H, Kodera Y, Leitner G, Netelenbos T, Niederwieser D, van Walraven SM, Rocha V, Torosian T, Vergueiro C, Weisdorf D, Yabe H, Halter JP. Suitability Criteria for Adult Related Donors: A Consensus Statement from the Worldwide Network for Blood and Marrow Transplantation Standing Committee on Donor Issues. Biol Blood Marrow Transplant 2015; 21:2052-2060. [PMID: 26271194 DOI: 10.1016/j.bbmt.2015.08.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 08/05/2015] [Indexed: 12/20/2022]
Abstract
The number of allogeneic hematopoietic stem cell (HSC) transplants performed globally each year continues to increase. Advances in HLA typing, better supportive care, and administration of reduced-intensity conditioning regimens allow treatment of older patients with older sibling donors. Pretransplant donor assessment and testing are very important processes affecting the quality and safety of donation. For unrelated HSC donors detailed recommendations for health assessment have been published, allowing donation only if they are unrestrictedly healthy. Eligibility criteria for related donors are less strict and vary significantly between centers. In situations where a family donor does not meet the suitability criteria for unrelated donors, involved physicians often struggle with the decision whether the matched relative is suitable for donation or not. On behalf of the Worldwide Network for Blood and Marrow Transplantation Standing Committee on Donor Issues, we intended to develop a consensus document with recommendations for donor workup and final clearance of family donors who would not be able to serve as unrelated donors because of their age or pre-existing diseases. This article covers different topics intending to support decision-making, with the goal of minimizing medical risk to the donor and protection of the recipient from transmissible diseases.
Collapse
Affiliation(s)
- Nina Worel
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria.
| | - Andreas Buser
- Blood Transfusion Centre, Swiss Red Cross, Basel, Switzerland; Department of Hematology, University Hospital Basel, Basel, Switzerland
| | | | - Hans Hägglund
- Division of Hematology, Department of Medical Sciences Uppsala University, Uppsala, Sweden
| | | | - Michael A Pulsipher
- Division of Hematology and Hematological Malignancies, Huntsman Cancer Institute/University of Utah, Primary Children's Hospital, Salt Lake City, Utah
| | | | - Mats Bengtsson
- Tobias Registry of Swedish Bone Marrow Donors and Uppsala University, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | | | - German Espino
- Department of Internal Medicine, Hematology and Bone Marrow Transplantation Section, University Hospital Caja del Seguro Social, Panama City, Panama
| | - Mirjam Fechter
- Europdonor Foundation Leiden, Leiden University Medical Centre, Leiden, The Netherlands
| | - Valeria Giudice
- Department of Immunohematology and Transfusion Medicine, S. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Kristina Hölig
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Heiwa Kanamori
- Department of Hematology, Kanagawa Cancer Center, Kanagawa, Japan
| | - Yoshihisa Kodera
- Asia-Pacific Blood and Marrow Transplantation Group and Department of Promotion for Blood and Marrow Transplantation, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Gerda Leitner
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| | - Tanja Netelenbos
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Suzanna M van Walraven
- Europdonor Foundation Leiden, Leiden University Medical Centre, Leiden, The Netherlands; Ethics Working Group of the World Marrow Donor Association, Leiden, The Netherlands
| | - Vanderson Rocha
- Oxford University Hospitals NHS Trust, British Bone Marrow Donor Registry and Cord Blood Banks, NHS-BT, Oxford, United Kingdom
| | | | - Carmen Vergueiro
- FCM Santa Casa de São Paulo, Disciplina de Hematologia e Oncologia, São Paulo, Brasil
| | - Daniel Weisdorf
- Bone Marrow Transplant Program, University of Minnesota, Minneapolis, Minnesota
| | - Hiromasa Yabe
- Department of Cell Transplantation and Regenerative Medicine, Tokai University School of Medicine, Tokyo, Japan
| | - Jörg P Halter
- Department of Hematology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Kastrup J. Gene therapy and angiogenesis in patients with coronary artery disease. Expert Rev Cardiovasc Ther 2014; 8:1127-38. [DOI: 10.1586/erc.10.95] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
12
|
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
|
13
|
Incidence and clinical significance of cardiac biomarker elevation during stem cell mobilization, apheresis, and intramyocardial delivery: an analysis from ACT34-CMI. Am Heart J 2012; 164:689-697.e3. [PMID: 23137499 DOI: 10.1016/j.ahj.2012.06.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 06/20/2012] [Indexed: 12/29/2022]
Abstract
BACKGROUND Cell therapy is a promising therapeutic for a variety of cardiovascular conditions including refractory angina. Elevation of cardiac biomarkers during cell delivery has been frequently described, but the clinical implications have never been studied. METHODS ACT34-CMI was a randomized double-blind study assessing the use of intramyocardial delivery of autologous CD34(+) cells for the treatment of refractory angina. Patients (n = 167) underwent G-CSF-mediated (5 μg/[kg day] × 5 days) stem cell mobilization, apheresis, and intramyocardial injection of 1 × 10(5)/kg or 5 × 10(5)/kg CD34(+) cells or placebo. Troponin and creatinine kinase MB were assessed at baseline (n = 161), after cell mobilization and apheresis (n = 153 and 143, respectively), and post-intramyocardial injection (n = 155 and 141, respectively). Major adverse cardiac events (MACE) included death, myocardial infarction, acute congestive heart failure, urgent revascularization, or sustained ventricular arrhythmia. RESULTS Seven (4.3%) subjects had troponin above the upper limits of normal (ULN) at baseline. Thirty-four (22.2%) and 11 (7.2%) subjects had troponin levels > ULN or >3× ULN after cell mobilization and apheresis, whereas 72 (46.1%) and 39 (25.2%) subjects had troponin elevations > ULN or >3× ULN, respectively, after intramyocardial injections. Age, but no other preprocedural factors, was predictive of troponin elevation. Periprocedural troponin elevation was not associated with an increased risk of MACE during 1 year, especially in cell therapy-treated patients. CONCLUSIONS Troponin elevation is common during stem cell harvesting and intramyocardial administration, is usually asymptomatic, and does not appear to be associated with long-term MACE in subjects undergoing stem cell mobilization and intramyocardial injection.
Collapse
|
14
|
Konenkov VI, Pokushalov EA, Poveshchenko OV, Kim II, Romanov AB, Guleva NA, Bernvald VV, Soloviova AO, Yankayte EV, Poveshchenko AF, Karaskov AM. Phenotype of Peripheral Blood Cells Mobilized by Granulocyte Colony-Stimulating Factor in Patients with Chronic Heart Failure. Bull Exp Biol Med 2012; 153:124-8. [DOI: 10.1007/s10517-012-1659-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
15
|
Sanganalmath SK, Abdel-Latif A, Bolli R, Xuan YT, Dawn B. Hematopoietic cytokines for cardiac repair: mobilization of bone marrow cells and beyond. Basic Res Cardiol 2011; 106:709-33. [PMID: 21541807 PMCID: PMC4281455 DOI: 10.1007/s00395-011-0183-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 04/11/2011] [Accepted: 04/15/2011] [Indexed: 12/20/2022]
Abstract
Hematopoietic cytokines, traditionally known to influence cellular proliferation, differentiation, maturation, and lineage commitment in the bone marrow, include granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor, stem cell factor, Flt-3 ligand, and erythropoietin among others. Emerging evidence suggests that these cytokines also exert multifarious biological effects on diverse nonhematopoietic organs and tissues. Although the precise mechanisms remain unclear, numerous studies in animal models of myocardial infarction (MI) and heart failure indicate that hematopoietic cytokines confer potent cardiovascular benefits, possibly through mobilization and subsequent homing of bone marrow-derived cells into the infarcted heart with consequent induction of myocardial repair involving multifarious mechanisms. In addition, these cytokines are also known to exert direct cytoprotective effects. However, results from small-scale clinical trials of G-CSF therapy as a single agent after acute MI have been discordant and largely disappointing. It is likely that cardiac repair following cytokine therapy depends on a number of known and unknown variables, and further experimental and clinical studies are certainly warranted to accurately determine the true therapeutic potential of such therapy. In this review, we discuss the biological features of several key hematopoietic cytokines and present the basic and clinical evidence pertaining to cardiac repair with hematopoietic cytokine therapy.
Collapse
Affiliation(s)
- Santosh K. Sanganalmath
- Division of Cardiovascular Diseases, Cardiovascular Research Institute, University of Kansas Medical Center, 3901 Rainbow Blvd, Rm. 1001 Eaton, MS 3006, Kansas City, KS 66160, USA
| | - Ahmed Abdel-Latif
- Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY 40292, USA
| | - Yu-Ting Xuan
- Division of Cardiovascular Diseases, Cardiovascular Research Institute, University of Kansas Medical Center, 3901 Rainbow Blvd, Rm. 1001 Eaton, MS 3006, Kansas City, KS 66160, USA
| | - Buddhadeb Dawn
- Division of Cardiovascular Diseases, Cardiovascular Research Institute, University of Kansas Medical Center, 3901 Rainbow Blvd, Rm. 1001 Eaton, MS 3006, Kansas City, KS 66160, USA
| |
Collapse
|
16
|
Toyama T, Hoshizaki H, Kasama S, Yamashita E, Kawaguchi R, Adachi H, Oshima S, Hasegawa A, Kurabayashi M. Low-dose and long-term G-CSF treatment can improve severe myocardial ischemia in patients with severe coronary artery disease. J Nucl Cardiol 2011; 18:463-71. [PMID: 21328025 DOI: 10.1007/s12350-011-9350-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2010] [Accepted: 01/23/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND It has been reported that granulocyte colony-stimulating factor (G-CSF) can promote angiogenesis by mobilizing bone marrow stem cells to blood vessels. The purpose of this study is to clarify whether low-dose and long-term G-CSF treatment can improve severe myocardial ischemia. METHODS We studied 40 patients (M/F = 29/11, age = 68 ± 9 years) who had severe coronary artery disease (7 with and 5 without old myocardial infarction; 3VD/2VD/1VD = 17/17/6) and severe myocardial ischemia with no indication for revascularization. G-CSF (1.5 μg/kg) was injected for 14 consecutive days. All patients were evaluated using stress myocardial scintigraphy, the Canadian Cardiovascular Society (CCVS) score, and cardiopulmonary exercise testing before and after 3 months of treatment. On 17 SPECT segments, the total defect score (TDS) and delta TDS (TDS (stress) minus TDS (resting)) were evaluated to assess the severity of myocardial ischemia. RESULTS The changes in stress TDS, delta TDS, and regional wall motion score were significantly greater in the G-CSF group than the control group (P < .0001). The CCVS score improved significantly from baseline to the 3-month follow-up assessment in the G-CSF group (P < .0001). The increase of peak VO2 was significantly larger in the G-CSF group than the control group (P = .015). CONCLUSION Low-dose and long-term G-CSF treatment can improve severe ischemia in patients with severe coronary artery disease.
Collapse
Affiliation(s)
- Takuji Toyama
- Gunma Prefectural Cardiovascular Center, 3-12, Kameizumi-machi, Maebashi, Gunma, 371-0004, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Li J, Zou Y, Ge J, Zhang D, Guan A, Wu J, Li L. The effects of G-CSF on proliferation of mouse myocardial microvascular endothelial cells. Int J Mol Sci 2011; 12:1306-15. [PMID: 21541060 PMCID: PMC3083707 DOI: 10.3390/ijms12021306] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Revised: 02/07/2011] [Accepted: 02/18/2011] [Indexed: 11/21/2022] Open
Abstract
This paper explores the effect of granulocyte colony-stimulating factor (G-CSF) on mouse myocardial microvascular endothelial cell (CMECs) proliferation. CMECs were harvested from C57/BL6 mice. CMECs were cultured in medium containing G-CSF (0 ng/mL, 20 ng/mL, 40 ng/mL, 60 ng/mL) for five days. Proliferative activity of CMECs was examined by CCK-8 method. Hypoxia inducible factor-1 (HIF-1) and p53 expression levels was determined from the mRNA obtained by reverse transcription polymerase chain reaction (RT-PCR). Results showed that the purity quotient of the CMECs, which were cultured by the method of modified myocardial tissue explant culture, was higher than 95%. Compared with control untreated cells, the proliferative activity of CMECs and the expression level of HIF-1 mRNA in these cells were enhanced by G-CSF treatment, whereas the expression level of p53 mRNA was markedly reduced. It may be concluded that G-CSF could promote the proliferative activity of CMECs, which might be mediated by upregulation of HIF-1 and downregulation of p53.
Collapse
Affiliation(s)
- Jiming Li
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 150 Jimo Road, Shanghai 200120, China; E-Mails: (J.L.); (D.Z.)
| | - Yunzeng Zou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; E-Mails: (A.G.); (J.W.); (L.L.)
- Institutes of Biomedical Scienses, Fudan University, Shanghai 200032, China
- Authors to whom correspondence should be addressed; E-Mails: (Y.Z.); (J.G.); Tel.: +86-21-54237970; Fax: +86-21-54237969
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; E-Mails: (A.G.); (J.W.); (L.L.)
- Institutes of Biomedical Scienses, Fudan University, Shanghai 200032, China
- Authors to whom correspondence should be addressed; E-Mails: (Y.Z.); (J.G.); Tel.: +86-21-54237970; Fax: +86-21-54237969
| | - Daifu Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 150 Jimo Road, Shanghai 200120, China; E-Mails: (J.L.); (D.Z.)
| | - Aili Guan
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; E-Mails: (A.G.); (J.W.); (L.L.)
- Institutes of Biomedical Scienses, Fudan University, Shanghai 200032, China
| | - Jian Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; E-Mails: (A.G.); (J.W.); (L.L.)
- Institutes of Biomedical Scienses, Fudan University, Shanghai 200032, China
| | - Lei Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; E-Mails: (A.G.); (J.W.); (L.L.)
- Institutes of Biomedical Scienses, Fudan University, Shanghai 200032, China
| |
Collapse
|
18
|
Friis T, Haack-Sørensen M, Hansen SK, Hansen L, Bindslev L, Kastrup J. Comparison of mesenchymal stromal cells from young healthy donors and patients with severe chronic coronary artery disease. Scandinavian Journal of Clinical and Laboratory Investigation 2011; 71:193-202. [DOI: 10.3109/00365513.2010.550310] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
19
|
Shim W, Mehta A, Lim SY, Zhang G, Lim CH, Chua T, Wong P. G-CSF for stem cell therapy in acute myocardial infarction: friend or foe? Cardiovasc Res 2011; 89:20-30. [DOI: 10.1093/cvr/cvq301] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
20
|
Beohar N, Rapp J, Pandya S, Losordo DW. Rebuilding the damaged heart: the potential of cytokines and growth factors in the treatment of ischemic heart disease. J Am Coll Cardiol 2010; 56:1287-97. [PMID: 20888519 DOI: 10.1016/j.jacc.2010.05.039] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Revised: 04/21/2010] [Accepted: 05/10/2010] [Indexed: 12/15/2022]
Abstract
Cytokine therapy promises to provide a noninvasive treatment option for ischemic heart disease. Cytokines are thought to influence angiogenesis directly via effects on endothelial cells or indirectly through progenitor cell-based mechanisms or by activating the expression of other angiogenic agents. Several cytokines mobilize progenitor cells from the bone marrow or are involved in the homing of mobilized cells to ischemic tissue. The recruited cells contribute to myocardial regeneration both as a structural component of the regenerating tissue and by secreting angiogenic or antiapoptotic factors, including cytokines. To date, randomized, controlled clinical trials have not reproduced the efficacy observed in pre-clinical and small-scale clinical investigations. Nevertheless, the list of promising cytokines continues to grow, and combinations of cytokines, with or without concurrent progenitor cell therapy, warrant further investigation.
Collapse
|
21
|
Wang Y, Ripa RS, Jørgensen E, Hesse B, Mortensen S, Kastrup J. Mobilization of haematopoietic and non-haematopoietic cells by granulocyte-colony stimulating factor and vascular endothelial growth factor gene therapy in patients with stable severe coronary artery disease. SCAND CARDIOVASC J 2009; 41:397-404. [DOI: 10.1080/14017430701601651] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
22
|
Abstract
The ability to exploit angiogenesis and vascularization as a therapeutic strategy will be of enormous benefit to a wide range of medical and tissue-engineering applications. Angiogenic growth factor and cell-based therapies have thus far failed to produce a robust healing response in clinical trials for a variety of ischemic diseases, while engineered tissue substitutes are still size-limited by a lack of vascularization. The purpose of this review is to investigate current research advances in therapeutic vascularization strategies applied to ischemic disease states, tissue engineering and regenerative medicine. Recent advances are discussed that focus on better regulation of growth factor delivery and attempts to better mimic natural processes by delivering combinations of multiple growth factors, cells and bioactive materials in the right spatial and temporal setting. Some unconventional approaches and novel therapeutic targets that hold significant potential are also discussed.
Collapse
Affiliation(s)
- Edward A Phelps
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | | |
Collapse
|
23
|
Overgaard M, Ripa RS, Wang Y, Jørgensen E, Kastrup J. Timing of granulocyte-colony stimulating factor treatment after acute myocardial infarction and recovery of left ventricular function: results from the STEMMI trial. Int J Cardiol 2009; 140:351-5. [PMID: 19131132 DOI: 10.1016/j.ijcard.2008.11.120] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2008] [Revised: 11/12/2008] [Accepted: 11/17/2008] [Indexed: 11/18/2022]
Abstract
BACKGROUND Granulocyte-colony stimulating factor (G-CSF) therapy after ST-elevation myocardial infarction (STEMI) have not demonstrated impact on systolic recovery compared to placebo. However, recent studies suggest that timing of G-CSF therapy is crucial. METHODS Timing of G-CSF treatment was analyzed in the STEMMI MRI subpopulation including 54 patients with STEMI treated with primary percutaneous coronary intervention (PCI) <12 h after symptom onset. Patients were randomized to double blind treatment with G-CSF (10 microg/kg/day) or placebo. Treatment was initiated from 17 to 65 h (mean 30) after PCI. Left ventricular ejection fraction (LVEF) was evaluated with MRI. RESULTS Recovery of LVEF from baseline to 6 months was not associated with time from PCI to G-CSF. An identical improvement in LVEF was found in the placebo group and the G-CSF group (p=0.8). There was no correlation between time from PCI to G-CSF and maximum plasma concentration of CD34+ cells (r=-0.3, p=0.1). Similar results were found from data on recovery of the infarction size and change in the systolic wall thickening. CONCLUSIONS In the time window from 17 to 65 h after STEMI treated with PCI, the timing of G-CSF treatment does not seem to affect the recovery of LVEF. It remains to be determined if very early, or very late G-CSF treatment might be effective.
Collapse
Affiliation(s)
- Mikkel Overgaard
- Department of Cardiology, The Heart Centre, Rigshospitalet, Copenhagen University Hospital, Denmark
| | | | | | | | | |
Collapse
|
24
|
D'Souza A, Jaiyesimi I, Trainor L, Venuturumili P. Granulocyte colony-stimulating factor administration: adverse events. Transfus Med Rev 2008; 22:280-90. [PMID: 18848155 DOI: 10.1016/j.tmrv.2008.05.005] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Recombinant human granulocyte colony-stimulating factor (G-CSF) has been in clinical use for approximately 2 decades. In healthy donors, it has been used to mobilize peripheral blood progenitor cells for hematopoietic stem cell transplantation and granulocytes for apheresis collection. In patients, it has been used to decrease the duration of neutropenia after chemotherapy and to offset the neutropenia due to myelodysplasia, acquired immunodeficiency syndrome, and genetic disorders of granulocyte production. As the number of uses of G-CSF in clinical practice grows, more side effects of this generally safe pharmaceutical agent are being recognized. Our objective in this article is to provide an in-depth review of the reported adverse events associated with the use of G-CSF.
Collapse
Affiliation(s)
- Anita D'Souza
- Department of Internal Medicine, William Beaumont Hospital, Royal Oak, MI 48073, USA.
| | | | | | | |
Collapse
|
25
|
Kovacic JC, Macdonald P, Feneley MP, Muller DWM, Freund J, Dodds A, Milliken S, Tao H, Itescu S, Moore J, Ma D, Graham RM. Safety and efficacy of consecutive cycles of granulocyte-colony stimulating factor, and an intracoronary CD133+ cell infusion in patients with chronic refractory ischemic heart disease: the G-CSF in angina patients with IHD to stimulate neovascularization (GAIN I) trial. Am Heart J 2008; 156:954-63. [PMID: 19061712 DOI: 10.1016/j.ahj.2008.04.034] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2007] [Accepted: 04/14/2008] [Indexed: 01/12/2023]
Abstract
BACKGROUND Preclinical studies suggest granulocyte-colony stimulating factor (G-CSF) holds promise for treating ischemic heart disease; however; its clinical safety and efficacy in this setting remain unclear. We elected to evaluate the safety and efficacy of G-CSF administration in patients with refractory "no-option" ischemic heart disease. METHODS Twenty patients (18 males, 2 females, mean age 62.4 years) were enrolled and underwent baseline cardiac ischemia assessment (CA) (angina questionnaire, exercise stress test [EST], technetium Tc 99m sestamibi and dobutamine-stress echocardiographic imaging). Patients then received open-label G-CSF commencing at 10 microg/kg SC for 5 days, with an EST on days 4 and 6 (to facilitate myocardial cytokine generation and stem cell trafficking). After 3 months, CA and the same regimen of G-CSF+ESTs were repeated but, in addition, leukapheresis and a randomized double-blinded intracoronary infusion of CD133+ or unselected cells were performed. Final CA occurred 3 months thereafter. RESULTS There were no deaths, but only 16 patients were permitted to complete the study. Eight events fulfilled prespecified "adverse event" criteria, including 4 troponin I-positive events and 2 episodes of thrombocytopenia. Also, frequent minor troponin I-positive events (troponin I<0.9 microg/L) were observed, which did not meet adverse event criteria. The administration of consecutive cycles of G-CSF resulted in stepwise improvements in anginal frequency, EST performance, and Duke treadmill scores (all P<.005). However, from baseline to final follow-up, technetium Tc 99m sestamibi and dobutamine-stress echocardiographic results were unchanged. CONCLUSIONS Granulocyte-colony stimulating factor administration was associated with improvement in a range of subjective outcomes. However, adverse events were common, and objective measures of cardiac perfusion/ischemia were unchanged.
Collapse
Affiliation(s)
- Jason C Kovacic
- Victor Chang Cardiac Research Institute, St Vincent's Hospital, University of New South Wales, Sydney, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Haack-Sorensen M, Friis T, Bindslev L, Mortensen S, Johnsen HE, Kastrup J. Comparison of different culture conditions for human mesenchymal stromal cells for clinical stem cell therapy. Scandinavian Journal of Clinical and Laboratory Investigation 2008; 68:192-203. [PMID: 17852829 DOI: 10.1080/00365510701601681] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Mesenchymal stromal cells (MSCs) from adult bone marrow (BM) are considered potential candidates for therapeutic neovascularization in cardiovascular disease. When implementing results from animal trials in clinical treatment, it is essential to isolate and expand the MSCs under conditions following good manufacturing practice (GMP). The aims of the study were first to establish culture conditions following GMP quality demands for human MSC expansion and differentiation for use in clinical trials, and second to compare these MSCs with MSCs derived from culture in four media commonly used for MSC cultivation in animal studies simulating clinical stem cell therapy. MATERIAL AND METHODS Human mononuclear cells (MNCs) were isolated from BM aspirates by density gradient centrifugation and cultivated in a GMP-accepted medium (EMEA medium) or in one of four other media. RESULTS FACS analysis showed that the plastic-adherent MSCs cultured in EMEA medium or in the other four media were identically negative for the haematopoietic surface markers CD45 and CD34 and positive for CD105, CD73, CD90, CD166 and CD13, which in combined expression is characteristic of MSCs. MSC stimulation with vascular endothelial growth factor (VEGF) increased expression of the characteristic endothelial genes KDR and von Willebrand factor; the von Willebrand factor and CD31 at protein level as well as the capacity to develop capillary-like structures. CONCLUSIONS We established culture conditions with a GMP compliant medium for MSC cultivation, expansion and differentiation. The expanded and differentiated MSCs can be used in autologous mesenchymal stromal cell therapy in patients with ischaemic heart disease.
Collapse
Affiliation(s)
- M Haack-Sorensen
- Cardiology Stem Cell Laboratory, The Heart Centre, University Hospital Rigshospitalet, Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
27
|
Coats AJ. Most frequently cited and downloaded papers from volumes 99 and 100. Int J Cardiol 2008. [DOI: 10.1016/j.ijcard.2007.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
28
|
Abstract
Cellular transplantation has been employed for several years to deliver donor cardiomyocytes to normal and injured hearts. Recent reports of a variety of stem cells with apparent cardiomyogenic potential have raised the possibility of cell transplantation-based therapeutic interventions for heart disease. Here we review the preclinical studies demonstrating that intracardiac transplantation of skeletal myoblasts, cardiomyocytes and cardiomyogenic stem cells is feasible. In addition, recent clinical studies of skeletal myoblast and adult stem cell transplantation for heart disease are discussed.
Collapse
Affiliation(s)
- Michael Rubart
- Division of Pediatric Cardiology, Herman B Wells Center for Pediatric Research, Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | |
Collapse
|
29
|
Fan L, Chen L, Chen X, Fu F. A meta-analysis of stem cell mobilization by granulocyte colony-stimulating factor in the treatment of acute myocardial infarction. Cardiovasc Drugs Ther 2007; 22:45-54. [PMID: 18000749 DOI: 10.1007/s10557-007-6072-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2007] [Accepted: 10/18/2007] [Indexed: 11/25/2022]
Abstract
OBJECTIVE This project was aimed at evaluating the safety and efficacy of granulocyte colony-stimulating factor (G-CSF) as an adjunctive therapy to the standard therapy [percutaneous coronary interventions (PCI) and conventional medication] after acute myocardial infarction (AMI). METHODS A meta-analysis of randomized controlled trials (RCTs) of G-CSF as an adjunctive therapy to standard therapy versus standard therapy was performed. The endpoints were defined as (1) target-vessel restenosis, (2) cumulative cardiac events (CCEs) that were a combined endpoint of all-cause deaths, reinfarction, and target-vessel revascularization, and (3) the changes in left ventricular ejection fraction (LVEF) from baseline to follow-up. RESULTS 320 patients were involved in 6 RCTs, of whom 160 were randomized to the G-CSF group and 160 to the control group. The follow-up period was 6.17 +/- 3.49 months. There was no significant difference in the risk of target-vessel restenosis (P = 0.90) or CCEs (P = 0.59) between the two groups. When a pooled analysis of the changes in LVEF was performed with fixed-model effect, a significant heterogeneity was observed (P < 0.00001). The pooled analysis was thus conducted with random-model effect and did not show a significant improvement as compared to the control group (P = 0.34). A similar result was found in the sensitivity analysis based on five placebo-controlled trials involving 270 patients (P = 0.94). CONCLUSIONS G-CSF as an adjunctive therapy to standard therapy for patients with AMI may be safe. However, there is not much supporting evidence that this treatment could further improve LVEF. Since there are relatively few RCTs that meet the inclusion criteria and are heterogeneous in design, further research is required.
Collapse
Affiliation(s)
- Lin Fan
- Department of Cardiology, Union Hospital, Fujian Medical University & Fujian Provincial Institute of Coronary Disease, 29 Xinquan Road 350001, Fuzhou, Fujian, People's Republic of China
| | | | | | | |
Collapse
|
30
|
Miyake Y, Ohmori K, Yoshida J, Ishizawa M, Mizukawa M, Yukiiri K, Kohno M. Granulocyte colony-stimulating factor facilitates the angiogenesis induced by ultrasonic microbubble destruction. ULTRASOUND IN MEDICINE & BIOLOGY 2007; 33:1796-804. [PMID: 17686568 DOI: 10.1016/j.ultrasmedbio.2007.05.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2006] [Revised: 05/14/2007] [Accepted: 05/21/2007] [Indexed: 05/16/2023]
Abstract
Ultrasonic destruction of microbubbles (US/MB) in the microcirculation causes local inflammatory cell infiltration, which has been shown to induce angiogenesis. Granulocyte colony-stimulating factor (G-CSF), which mobilizes myelomonocytic cells from the bone marrow and enhances vascular endothelial growth factor (VEGF) release from these cells, has also been applied to therapeutic angiogenesis induction. In the present study, we sought to examine the potential of G-CSF pretreatment to enhance the angiogenic effect of US/MB. Ischemic hindlimbs in mice were treated with either a predetermined minimal effective dose (300 mug/kg) of G-CSF, US/MB alone or G-CSF pretreatment followed by US/MB at seven days after removal of the femoral artery. Ultrasonic destruction of microbubbles was performed as intermittent pulsed local insonation using a diagnostic ultrasound scanner at a peak negative pressure of 1.4 MPa after intravenous injection of perfluorocarbon microbubbles. At 21 days after the treatment, we quantified the surface vascularity using a grid method and the capillary density using an alkaline phosphatase stain. Relative to the capillary density in normal muscle, the capillary density in the treated limbs was restored to 74 +/- 13% by G-CSF alone and 90 +/- 20% by US/MB alone (p < 0.05 vs. both untreated and G-CSF alone), and further increased to 101 +/- 21% by G-CSF pretreatment. The collateral growth induced by the combination of G-CSF pretreatment and US/MB was 2.8- and 1.4-fold greater than the growth induced by G-CSF alone and US/MB alone, respectively (p < 0.05 for both). Thus, pretreatment with a single minimal effective dose of G-CSF can augment the angiogenic effect of US/MB.
Collapse
Affiliation(s)
- Yuichi Miyake
- Division of Cardiology, Department of Cardiorenal Cerebrovascular Medicine, Kagawa University School of Medicine, Kagawa, Japan
| | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
In sepsis and septic shock a series of immunological events are initiated that alter endothelial function in the macrocirculation and microcirculation. Endothelial swelling, deformation and apoptosis with detachment from the vasculature occur and endothelial cells (EC) appear in the circulation. Simultaneous to these pathological processes, reconstitution of the endothelial layer is initiated which can occur via migration and proliferation of surrounding mature ECs. However, terminally differentiated ECs have a low proliferative potential, hence their capacity to substitute damaged endothelium is limited. Therefore, adequate vascular repair requires additional support. Many studies have now convincingly demonstrated that vascular maintenance, repair, angiogenesis and neovascularization are partly mediated by recruitment of endothelial progenitor cells (EPCs) from the basal membrane. However, it seems that EPCs play a pivotal role not only in re-endothelialization after vascular damage, but also after severe inflammation. Recently, evidence was found that EPCs are increasingly mobilized during sepsis and that this mobilization is associated with clinical outcome. In septic patients the number of EPCs was significantly higher than in controls and was correlated with survival and the concentration of cytokines. In summary EPCs may exert an important function as an endogenous repair mechanism to maintain the integrity of the endothelial layer by replacing denuded parts of the microcirculation or by stimulation of EC proliferation. Therefore, EPC enumeration seems to be a valuable prognostic and diagnostic marker for the outcome in these patients and the induction of enhanced EPC mobilization a therapeutic option.
Collapse
Affiliation(s)
- G C Beck
- Klinik für Anästhesiologie und Operative Intensivmedizin, Universitätsklinikum Mannheim, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Deutschland.
| | | | | | | |
Collapse
|
32
|
Ripa RS, Wang Y, Goetze JP, Jørgensen E, Johnsen HE, Tägil K, Hesse B, Kastrup J. Circulating angiogenic cytokines and stem cells in patients with severe chronic ischemic heart disease — Indicators of myocardial ischemic burden? Int J Cardiol 2007; 120:181-7. [PMID: 17157396 DOI: 10.1016/j.ijcard.2006.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2006] [Revised: 09/11/2006] [Accepted: 09/20/2006] [Indexed: 11/28/2022]
Abstract
BACKGROUND Angiogenic growth factors and stem cell therapies have demonstrated varying results in patients with chronic coronary artery disease. A reason could be that these mechanisms are already up-regulated due to reduced blood supply to the myocardium. The objective of this study was to examine if plasma concentrations of circulating stem cells and angiogenic cytokines in patients with severe stable chronic coronary artery disease were correlated to the clinical severity of the disease. METHODS Fifty-four patients with severe coronary artery disease and reversible ischemia at stress myocardial perfusion scintigraphy were prospectively included. The severity of the disease was quantified by an exercise tolerance test, Canadian Cardiovascular Society angina classification, and Seattle Angina Pectoris Questionnaire. Fifteen persons without coronary artery disease served as control subjects. RESULTS Plasma concentration of VEGF-A, FGF-2, SDF-1, and circulating CD34+ and CD34-/CD45- cells were similar in the two groups, but early stem cell markers (CD105, CD73, CD166) and endothelial markers (CD31, CD144, VEGFR2) were significantly different between patients and control subjects (p<0.005-0.001). Diabetic patients had higher concentration of SDF-1 (2528 vs. 2150 pg/ml, p=0.004). We found significant correlations between both VEGF-A, FGF-2, and CD34+ to disease severity, including degree of reversible ischemia, angina stability score, and exertional dyspnoea. CONCLUSIONS Plasma concentrations of circulating stem cells and angiogenic cytokines have large inter-individual variations, which probably exclude them from being useful as indicators of myocardial ischemic burden.
Collapse
Affiliation(s)
- Rasmus Sejersten Ripa
- Cardiac Catheterization Laboratory, University Hospital Rigshospitalet, Copenhagen, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Ben-Shoshan J, George J. Endothelial progenitor cells as therapeutic vectors in cardiovascular disorders: from experimental models to human trials. Pharmacol Ther 2007; 115:25-36. [PMID: 17574679 DOI: 10.1016/j.pharmthera.2007.03.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2007] [Accepted: 03/27/2007] [Indexed: 11/19/2022]
Abstract
Cell-based therapy approaches for the restoration of blood flow in ischemic organs has recently received growing interest. A considerable number of reports have documented the presence of circulating, bone marrow-derived endothelial progenitor cells (EPC) in adult peripheral blood. These putative cells are thought to participate in postnatal growth of new blood vessels. Mounting evidence from animal studies point to potential therapeutic applications of EPCs in the treatment of a wide range of cardiovascular (CV) disorders, while preliminary results from the pilot clinical trials still remain equivocal. Here, we review the experimental data that has accumulated so far from animal and clinical studies regarding the potential importance of EPCs. In addition, we discuss the potential hurdles as well as future options of EPC-based therapy.
Collapse
Affiliation(s)
- Jeremy Ben-Shoshan
- Department of Cardiology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | | |
Collapse
|
34
|
Dunac A, Frelin C, Popolo-Blondeau M, Chatel M, Mahagne MH, Philip PJM. Neurological and functional recovery in human stroke are associated with peripheral blood CD34+ cell mobilization. J Neurol 2007; 254:327-32. [PMID: 17345048 DOI: 10.1007/s00415-006-0362-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2006] [Accepted: 08/16/2006] [Indexed: 01/28/2023]
Abstract
BACKGROUND A spontaneous mobilization of Peripheral Blood-Mononuclear CD34+ Cells (PB-MNC-CD34+) has recently been reported in human myocardial infarction and found to be related to improved heart function and survival. However, nothing is known regarding a possible relation between PB-MNC-CD34+ mobilization and neurological recovery in human acute cerebral ischemia. METHODS AND RESULTS PB-MNC-CD34+ were determined daily after an acute cerebral ischemic attack for 14 days in 25 patients with acute ischemic stroke and compared with controls. Results indicated that stroke was followed by large and bursting mobilizations of PB-MNC-CD34+. The amplitude of the mobilizations was similar to those observed in Granulocyte Colony Stimulating Factor (G-CSF) conditioned aplastic patients following myeloablative therapy before leukapheresis and autologous bone graft. The extent of PB-MNC-CD34+ mobilization in each patient was directly related to neurological and functional recoveries as assessed by NIH Stroke Scale, and modified Rankin Scale respectively. CONCLUSIONS The mobilization of PB-MNC-CD34+ cells might be predictive of neurological and functional recovery.
Collapse
Affiliation(s)
- Antoine Dunac
- Department of Neurology (Stroke Unit), University Hospital Pasteur, 30 avenue de la voie Romaine, Nice Cedex, BP69, 06002, France.
| | | | | | | | | | | |
Collapse
|
35
|
Sánchez PL, Villa A, Sanz R, Domínguez M, Ludwig I, Fernández ME, Fernández-Avilés F. Present and future of stem cells for cardiovascular therapy. Ann Med 2007; 39:412-27. [PMID: 17852037 DOI: 10.1080/07853890701513746] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In this review we summarize the available evidence regarding the application of stem cell therapy for human cardiovascular repair, going over the principal concepts that will help us understand the present and future of this therapy: first the different types of cells available in clinical practice, second the delivery approaches, and third highlighting the most important clinical studies and their efficacy and safety results. In addition, we also speculate on the value of current clinical data to gain an insight into the mechanism of stem cell-based cardiac repair and to design clinical trials in the future.
Collapse
Affiliation(s)
- Pedro L Sánchez
- Servicio de Cardiología, Hospital General Universitario Gregorio Marañón, Madrid, Spain.
| | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Patients with ischemic cardiomyopathy have a poor prognosis despite all pharmacological, interventional and surgical treatment modalities currently applied. Heart transplantation remains the ideal treatment for this group of patients but the scarcity of donors hinders its widespread application. The autologous transplantation of stem cells (SCs) for cardiac repair is emerging as a new therapy for patients with myocardial dysfunction early after an acute infarction or ischemic cardiomyopathy. The rationale of this novel method is the enhancement of the repair mechanisms achieved by tissue-specific and circulating stem/progenitor cells. SCs assist naturally occurring myocardial repair by contributing to increased myocardial perfusion and contractile performance especially in the setting of acute myocardial infarction (AMI), but also in patients with chronic ischemic heart failure and advanced, diffuse coronary artery disease. The exact mechanism of their action has not been fully elucidated. Few studies continue to suggest a formation of few new contractile tissue. The majority if investigators believe that these cells do not persist long in the myocardium but that they secrete vascular growth and other cardioprotective factors.
Collapse
|
37
|
Kovacic JC, Muller DWM, Graham RM. Actions and therapeutic potential of G-CSF and GM-CSF in cardiovascular disease. J Mol Cell Cardiol 2006; 42:19-33. [PMID: 17109881 DOI: 10.1016/j.yjmcc.2006.10.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2006] [Accepted: 10/02/2006] [Indexed: 01/16/2023]
Abstract
Despite their names, the cytokines granulocyte- and granulocyte-macrophage-colony stimulating factor (G-CSF and GM-CSF respectively) have actions far beyond simply stimulating the proliferation of neutrophil and monocyte lineage cells. A comprehensive body of evidence now exists demonstrating that G-CSF and GM-CSF effectively mobilize bone-marrow-derived progenitor cells into the peripheral circulation. These mobilized progenitor cells can be conveniently harvested for use in reconstituting bone marrow by transplantation after myelo-ablative treatment of hematological malignancies. In addition, much evidence has recently emerged to suggest that these cytokines may have multiple direct and indirect beneficial cardiovascular effects--including neovascularization of ischemic myocardium and reducing the extent of myocardial damage after infarction. Based on this knowledge and a strong safety record in hematological applications, a number of early clinical trials have evaluated the use of G-CSF or GM-CSF in patients with both acute and chronic myocardial ischemia. Although the interpretation of these trials is complicated by heterogeneity in study design, small patient numbers and methodological concerns related to appropriate selection and blinding of patients, the results of ongoing larger phase II/III trials should soon be available to determine if these agents will be useful additions to the cardiovascular armamentarium.
Collapse
|
38
|
Werneck-de-Castro JPS, Costa-E-Sousa RH, de Oliveira PF, Pinho-Ribeiro V, Mello DB, Peçanha R, Mattos E, Olivares EL, Maia ACV, Mill JG, Dos Santos Goldenberg RC, Campos-de-Carvalho AC. G-CSF does not improve systolic function in a rat model of acute myocardial infarction. Basic Res Cardiol 2006; 101:494-501. [PMID: 16783485 DOI: 10.1007/s00395-006-0605-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2006] [Revised: 05/29/2006] [Accepted: 05/17/2006] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Granulocyte colony-stimulating factor (G-CSF) has been reported to improve cardiac performance by increasing the number of bone marrow stem cell in the peripheral circulation. The aim of this study was to investigate the impact of G-CSF administration on cardiac function in a rat model of acute myocardial infarction. METHODS Recombinant human G-CSF (Filgrastim, 100 microg/kg, sc) twice a day during seven consecutive days (G-CSF group, n=13) or vehicle (control group, n=10) was administrated three hours after left anterior coronary artery ligation. Cardiac performance was evaluated 19-21 days after myocardial infarction by electro- and echocardiography, hemodynamic and treadmill exercise test. RESULTS Both infarcted groups exhibit impaired cardiac function compared to sham-operated rats. Moreover, all cardiac functional parameters were not statistically different between G-CSF and infarcted group at resting conditions as well as after treadmill exercise stress test. There was no sign of cardiac regeneration and infarct size was not different on histological analysis between groups. CONCLUSIONS These data clearly shows that G-CSF treatment was unable to prevent cardiac remodeling or to improve cardiovascular function in a rat model of acute myocardial infarction, by permanent LAD ligation, despite bone marrow stem cell mobilization.
Collapse
|
39
|
Lim SY, Kim YS, Ahn Y, Jeong MH, Rok LS, Kim JH, Kim KH, Park HW, Kim W, Cho JG, Park JC, Kang PM, Schwartz RS, Kang JC. The effects of granulocyte-colony stimulating factor in bare stent and sirolimus-eluting stent in pigs following myocardial infarction. Int J Cardiol 2006; 118:304-11. [PMID: 17052793 DOI: 10.1016/j.ijcard.2006.07.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2005] [Revised: 04/03/2006] [Accepted: 07/09/2006] [Indexed: 11/17/2022]
Abstract
OBJECTIVES The purpose of this study was to compare the effects of granulocyte-colony stimulating factor (G-CSF) on in-stent restenosis (ISR) in bare and sirolimus-eluting stents (SES) in a porcine myocardial infarction model. BACKGROUND Using G-CSF to mobilize stem cells has shown promise in infarcted heart. However, G-CSF may aggravate ISR and an aggressive strategy to prevent ISR is needed. METHODS Bare stents and SES were implanted in coronary arteries (Group I, bare stents; Group II, bare stents with G-CSF; Group III, SES; Group IV, SES with G-CSF, n=10 in each group) 72 h after experimental myocardial infarction (MI). G-CSF (10 microg/kg/day) was injected for 7 days from 24 h after stent implantation. RESULTS In coronary angiographic and histomorphometric analysis, percent area stenosis was significantly increased in Group II compared with that in Group I at 28 days (P<0.05). The ratio of inflammatory cells in the neointima was higher in Group II (P<0.05). No significant differences were observed between Group III and IV. In Group II, phosphorylated signal transducers and activators of transcription (STAT)-3, STAT-3, and vascular endothelial growth factor (VEGF) showed increased neointimal expression. In porcine aortic smooth muscle cells (PASMC), G-CSF increased the growth rate, migration, STAT-3 phosphorylation, and VEGF, which were suppressed by rapamycin and AG490, a STAT-3 inhibitor. CONCLUSIONS STAT-3 and VEGF are important in the development of enhanced ISR by G-CSF in bare stents. SES could be a good strategy to prevent the G-CSF-stimulated proliferation and migration of smooth muscle cells, which could be responsible for neointimal hyperplasia.
Collapse
Affiliation(s)
- Sang Yup Lim
- The Heart Center of Chonnam National University Hospital, Gwangju, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Honold J, Lehmann R, Heeschen C, Walter DH, Assmus B, Sasaki KI, Martin H, Haendeler J, Zeiher AM, Dimmeler S. Effects of Granulocyte Colony Stimulating Factor on Functional Activities of Endothelial Progenitor Cells in Patients With Chronic Ischemic Heart Disease. Arterioscler Thromb Vasc Biol 2006; 26:2238-43. [PMID: 16902165 DOI: 10.1161/01.atv.0000240248.55172.dd] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Bone marrow-derived circulating endothelial progenitor cells (EPCs) may contribute to regeneration of infarcted myocardium and enhance neovascularization. Granulocyte colony-stimulating factor (G-CSF) is well-established to mobilize hematopoietic stem cells (HSCs) and might, thereby, also increase the pool of endogenously circulating EPC. Therefore, we investigated the effects of G-CSF administration on mobilization and functional activities of blood-derived EPC in patients with chronic ischemic heart disease (CIHD). METHODS AND RESULTS Sixteen patients with CIHD received 10 microg/kg per day subcutaneous G-CSF injection for 5 days. Leukocyte counts, the number of HSCs and EPCs, and the migratory response to VEGF and SDF-1 were analyzed before and after G-CSF-therapy. At day 5 of G-CSF treatment, the number of circulating leukocytes, CD34+ CD45+ and CD34+ CD133+ cells was significantly increased. Likewise, G-CSF treatment augmented the numbers of colony forming units with endothelial cell morphology (EC-CFU). However, the functional activity of the EPC as assessed by the migratory response to VEGF and SDF-1 was significantly reduced after G-CSF treatment (P<0.01). Because G-CSF was previously shown to cleave the CXCR4 receptor, we determined the surface expression of the 6H8 epitope of the CXCR4 receptor by fluorescence-activated cell sorter (FACS) analysis. Consistent with the reduced migratory capacity, the surface expression of the functionally active CXCR4 receptor was significantly reduced. To test the functional activity of the cultivated EPCs in vivo, cells were intravenously infused in nude mice after hind limb ischemia. EPCs, which were cultivated before G-CSF administration, increased blood flow recovery and prevented limb necrosis. However, infusion of EPCs, which were isolated 5 days after G-CSF treatment from the same patient, showed a reduced capacity to augment blood flow recovery and to prevent necrosis by 27%. CONCLUSIONS G-CSF treatment effectively mobilizes HSCs and EPCs. However, the migratory response to SDF-1 and in vivo capacity of G-CSF-mobilized EPCs was significantly reduced.
Collapse
Affiliation(s)
- Joerg Honold
- Department of Cardiology and Molecular Cardiology, Internal Medicine III, J.W. Goethe University of Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Funabashi N, Komiyama N, Kato H, Umekita H, Asano M, Komuro I. Retrospective ECG-gated left ventriculography using multislice CT following left ventricular bolus injection and evaluation of its utility and motion artifact at every cardiac phase. Int J Cardiol 2006; 113:132-8. [PMID: 16298444 DOI: 10.1016/j.ijcard.2005.08.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2005] [Accepted: 08/20/2005] [Indexed: 10/25/2022]
Abstract
Following left ventricular bolus injection of contrast material, multislice CT scanning was performed. With retrospective ECG-gated reconstruction, we could acquire volume data for the heart at any cardiac phase and selectively depict only the left ventricle and aorta with maximum intensity projection. Temporal resolution of multislice CT was not sufficient to eliminate motion artifact except just before atrial contraction periods.
Collapse
|
42
|
Suzuki K, Nagashima K, Arai M, Uno Y, Misao Y, Takemura G, Nishigaki K, Minatoguchi S, Watanabe S, Tei C, Fujiwara H. Effect of granulocyte colony-stimulating factor treatment at a low dose but for a long duration in patients with coronary heart disease. Circ J 2006; 70:430-7. [PMID: 16565560 DOI: 10.1253/circj.70.430] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND In animal models, granulocyte colony-stimulating factor (G-CSF) improves post-infarct cardiac function. However, in pilot studies involving patients with angina and acute myocardial infarction (AMI), G-CSF at a high dose frequently induced coronary occlusion or restenosis, but those at a low dose showed no significant beneficial effect. We hypothesized that a low dose but long duration of G-CSF will have a beneficial effect without serious complications to patients with coronary heart disease. METHODS AND RESULTS Forty-six patients with angina or AMI were randomly assigned into G-CSF and non-G-CSF control groups, respectively. Recombinant G-CSF was subcutaneously injected once a day for 10 days. The leukocyte counts in the peripheral blood were controlled at approximately 30,000/microl. One month later, a Thallium-201 single photon emission computed tomography revealed the increased percentage uptake and the reduced extent and severity scores in the G-CSF angina group. In the G-CSF AMI group, the curve between the ejection fraction and peak creatine kinase shifted significantly upward, compared with that of the non-G-CSF AMI group. Serious complications were not observed during the 6 months of observation. CONCLUSIONS A low dose but long duration of G-CSF treatment may have a beneficial effect without any serious complications in patients with coronary heart disease.
Collapse
Affiliation(s)
- Koji Suzuki
- Department of Cardiology, Gifu University Graduate School of Medicine, Yanagido, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Deng Z, Yang C, Deng H, Yang A, Geng T, Chen X, Ma A, Liu Z. Effects of GM-CSF on the stem cells mobilization and plasma C-reactive protein levels in patients with acute myocardial infarction. Int J Cardiol 2006; 113:92-6. [PMID: 16891014 DOI: 10.1016/j.ijcard.2006.06.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2005] [Revised: 06/09/2006] [Accepted: 06/18/2006] [Indexed: 01/26/2023]
Abstract
BACKGROUND Stem cell mobilization with granulocyte colony-stimulating factor (G-CSF) has been proposed to improve cardiac function and prevent ventricular remodeling after acute myocardial infarction (AMI) in preclinical and clinical studies. It has been demonstrated that granulocyte-macrophage colony-stimulating factor (GM-CSF) can improve collateral flow in patients with coronary artery disease. In this study, we used GM-CSF to mobilize the bone marrow stem cells (BMSCs) in patients with AMI and assessed the safety, feasibility and efficacy of this treatment. METHODS Twenty patients with AMI were randomly divided into GM-CSF group (10 microg/kg body weight, for 7 days) and control group (saline). The absolute counts of CD34 positive cells in peripheral blood were enumerated with flow cytometry. Plasma levels of C-reactive protein (CRP), interleukin-6 (IL-6) and tumor necrotic factor-alpha (TNF-alpha) were measured on days 1, 3, 7, 10 and 14. Echocardiography (UCG) was done on day 7 and after 12 months. RESULTS Peripheral CD34 positive cells in GM-CSF patients obviously increased shortly after using GM-CSF and peaked on day 7 (p<0.01 versus controls). GM-CSF group had significantly higher mean level of plasma CRP than controls on day 10 (p<0.05). The levels of IL-6 and TNF-alpha in therapy patients were as same as in controls. Left ventricular ejection fraction (EF) at 12 months was significantly greater than that on day 7 in GM-CSF patients (p<0.05). The EF in controls had no obvious differences in follow-up. There were no statistically differences regarding the left ventricular end-systolic volume (LVESV), the left ventricular end-diastolic volume (LVEDV) and the resting wall thickening (WT) in the infarct zone in two groups in follow-up. CONCLUSIONS Our results demonstrate that GM-CSF can effectively mobilize the CD34 positive cells and at the same time may increase the levels of plasma CRP in patients with AMI. The remote effects of this drug need to be further defined.
Collapse
Affiliation(s)
- Zhengrong Deng
- Department of Cardiology, the First Hospital of Xi'an Jiaotong University, No. 1 Jiankang Road, Xi'an, Shaanxi, 710061, China.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Hüttmann A, Gutersohn A, Noppeney R, Neumann T, Erbel R, Dührsen U. Rapid succession of peripheral blood progenitor cell mobilization cycles in patients with chronic heart failure: effects on the hematopoietic system. Transfusion 2006; 46:1424-31. [PMID: 16934081 DOI: 10.1111/j.1537-2995.2006.00912.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND Circulating hematopoietic peripheral blood progenitor cells (PBPCs) may contribute to the regeneration of nonhematopoietic organs. An increase in circulating PBPC numbers may enhance this process. Therefore, an exploratory trial of repeated PBPC mobilization in patients with chronic heart failure was conducted. The safety and cardiovascular efficacy data have been described elsewhere. In the hematopoietic system, the trial offered an opportunity to study several new aspects of granulocyte-colony-stimulating factor (G-CSF) action. STUDY DESIGN AND METHODS Fourteen male patients with chronic heart failure were treated successively with G-CSF (four 10-day treatment periods interrupted by treatment-free intervals of equal length; daily dose adjustment to maintain a white blood cell [WBC] count of 45 x 10(9)-50 x 10(9)/L). RESULTS G-CSF induced a rapid increase in cells of all WBC lineages with return to levels equal to (neutrophilic, eosinophilic, and basophilic granulocytes) or lower than those before treatment (monocytes, lymphocytes) during the treatment-free intervals. Red cell counts remained unchanged, but platelet counts decreased followed by rebound thrombocytosis. The extent of CD34+ cell mobilization was highly variable. For each patient, the changes induced were identical through all cycles, but the G-CSF dose required in the first cycle was significantly higher than in subsequent cycles. In the cohort of patients, an inverse correlation was observed between the WBC level reached and the dose of G-CSF administered. CONCLUSIONS Rapid alternation between PBPC mobilization and recovery periods is feasible, with identical alterations in all treatment cycles. G-CSF responsiveness varies among patients and is increased by pretreatment with G-CSF.
Collapse
Affiliation(s)
- Andreas Hüttmann
- Hematology Clinic and the Cardiology Clinic, Center for Internal Medicine, University Hospital, Essen, Germany.
| | | | | | | | | | | |
Collapse
|
45
|
Lehrke S, Mazhari R, Durand DJ, Zheng M, Bedja D, Zimmet JM, Schuleri KH, Chi AS, Gabrielson KL, Hare JM. Aging impairs the beneficial effect of granulocyte colony-stimulating factor and stem cell factor on post-myocardial infarction remodeling. Circ Res 2006; 99:553-60. [PMID: 16873716 DOI: 10.1161/01.res.0000238375.88582.d8] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Granulocyte colony-stimulating factor (G-CSF) and stem cell factor (SCF) are potential new therapies to ameliorate post-myocardial infarction (post-MI) remodeling, as they enhance endogenous cardiac repair mechanisms and decrease cardiomyocyte apoptosis. Because both of these pathways undergo alterations with increasing age, we hypothesized that therapeutic efficacy of G-CSF and SCF is impaired in old versus young adult rats. MI was induced in 6- and 20-month-old rats by permanent ligation of the left coronary artery. In young animals, G-CSF/SCF therapy stabilized and reversed a decline in cardiac function, attenuated left ventricular dilation, decreased infarct size, and reduced cardiomyocyte hypertrophy. Remarkably, these effects on cardiac structure and function were absent in aged rodents. This could not be attributed to ineffective mobilization of bone marrow cells or decreased quantity of c-Kit(+) cells within the myocardium with aging. However, whereas the G-CSF/SCF cocktail reduced cardiac myocyte apoptosis in old as well as in young hearts, the degree of reduction was substantially less with age and the rate of cardiomyocyte apoptosis in old animals remained high despite cytokine treatment. These findings demonstrate that G-CSF/SCF lacks therapeutic efficacy in old animals by failing to offset periinfarct apoptosis and therefore raise important concerns regarding the efficacy of novel cytokine therapies in elderly individuals at greatest risk for adverse consequences of MI.
Collapse
Affiliation(s)
- Stephanie Lehrke
- Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Jørgensen E, Ripa RS, Helqvist S, Wang Y, Johnsen HE, Grande P, Kastrup J. In-stent neo-intimal hyperplasia after stem cell mobilization by granulocyte-colony stimulating factor. Int J Cardiol 2006; 111:174-7. [PMID: 16054717 DOI: 10.1016/j.ijcard.2005.06.045] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2005] [Revised: 06/08/2005] [Accepted: 06/11/2005] [Indexed: 01/24/2023]
Abstract
OBJECTIVE The influence of treatment with granulocyte-colony stimulating factor (G-CSF) on the development of in-stent intimal hyperplasia is not known. We aimed to study this phenomenon in patients who had stents implanted in the course of an acute ST-elevation infarction and successively were treated with G-CSF. METHOD We performed angiography and intracoronary ultrasound follow-up after 5 months in 41 consecutive patients in the STEMMI trial, which is a randomized double-blind placebo-controlled study on the effect of G-CSF injections on the myocardial function following acute myocardial infarction in patients treated with primary percutaneous coronary stent implantation. The intracoronary ultrasound images were analyzed by a blinded and independent core laboratory. RESULTS There were no differences in in-stent neo-intimal hyperplasia determined by intracoronary ultrasound between patients treated with G-CSF compared to patients treated with placebo. Neo-intimal hyperplasia per mm of stent was 1.87 (+/-1.41) and 1.89 (+/-1.39), respectively (p = 0.97). Angiographic in-segment restenosis (>50% diameter stenosis) was found in 28% of patients (24% in the G-CSF group and 33% in the placebo group; p = 0.55). CONCLUSION G-CSF treatment following coronary stent implantation in primary PCI treated AMI patients does not increase in-stent restenosis excessively and it does not seem warranted to limit further study of effects of G-CSF for that reason.
Collapse
|
47
|
Abstract
Many forms of pediatric and adult heart disease result from a deficiency in cardiomyocyte number. Through repopulation of the heart with new cardiomyocytes (that is, induction of regenerative cardiac growth), cardiac disease potentially can be reversed, provided that the newly formed myocytes structurally and functionally integrate in the preexisting myocardium. A number of approaches have been utilized to effect regenerative growth of the myocardium in experimental animals. These include interventions aimed at enhancing the ability of cardiomyocytes to proliferate in response to cardiac injury, as well as transplantation of cardiomyocytes or myogenic stem cells into diseased hearts. Here we review efforts to induce myocardial regeneration. We also provide a critical review of techniques currently used to assess cardiac regeneration and functional integration of de novo cardiomyocytes.
Collapse
Affiliation(s)
- Michael Rubart
- Herman B Wells Center for Pediatric Research and Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, Indiana 46202-5225, USA.
| | | |
Collapse
|
48
|
Hill JM, Bartunek J. The end of granulocyte colony-stimulating factor in acute myocardial infarction? Reaping the benefits beyond cytokine mobilization. Circulation 2006; 113:1926-8. [PMID: 16636185 DOI: 10.1161/circulationaha.106.623777] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
49
|
Hasegawa H, Takano H, Ohtsuka M, Ueda K, Niitsuma Y, Qin Y, Tadokoro H, Shiomi M, Komuro I. G-CSF prevents the progression of atherosclerosis and neointimal formation in rabbits. Biochem Biophys Res Commun 2006; 344:370-6. [PMID: 16600176 DOI: 10.1016/j.bbrc.2006.03.081] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2006] [Accepted: 03/14/2006] [Indexed: 12/22/2022]
Abstract
Granulocyte colony-stimulating factor (G-CSF) prevents left ventricular remodeling after myocardial infarction, but its effect on atherosclerosis is unknown. We examined two kinds of rabbit atherosclerosis models. Myocardial infarction-prone Watanabe heritable hyperlipidemic (WHHL-MI) rabbits were treated with G-CSF or saline for 7 days from 14 months old. The vascular injury models were created by inflating angioplasty balloon in the iliac artery of rabbits and were divided into G-CSF and saline group. G-CSF significantly reduced the stenosis score of coronary artery and lipid plaque area of thoracic aorta in WHHL-MI rabbits at 4 weeks after the treatment. In the vascular injury model, G-CSF significantly prevented an increase in neointima/media ratio at 4 weeks after the treatment. G-CSF accelerated the reendothelialization of denuded arteries, and the pretreatment with nitric oxide synthase inhibitor significantly inhibited it. These results suggest that G-CSF has a therapeutic potential for the progression of atherosclerosis.
Collapse
Affiliation(s)
- Hiroshi Hasegawa
- Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Hou XW, Son J, Wang Y, Ru YX, Lian Q, Majiti W, Amazouzi A, Zhou YL, Wang PX, Han ZC. Granulocyte Colony-Stimulating Factor Reduces Cardiomyocyte Apoptosis and Improves Cardiac Function in Adriamycin-Induced Cardiomyopathy in Rats. Cardiovasc Drugs Ther 2006; 20:85-91. [PMID: 16761189 DOI: 10.1007/s10557-006-7652-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Cardiomyocyte apoptosis reportedly participates in the occurrence and progression of dilated cardiomyopathy (DCM). Recent studies have shown that granulocyte colony-stimulating factor (G-CSF) enhances bone marrow cells migration to the damaged heart in the DCM model and improves the ultrastructure of the cardiomyocyte in adriamycin (ADR) induced DCM. However, its influence on cardiac pump function and cardiomyocyte apoptosis has not been studied. METHODS AND MATERIALS Wistar Rats were randomly grouped into control, ADR, ADR+PBS, ADR+G-CSF group (n = 10). ADR (2.5 mg/kg, 6 times for 2 weeks) was administered intraperitoneally in all rats except the control group. After 2 weeks, the rats in ADR+G-CSF group were injected with G-CSF (50 microg/kg/day for 8 days) subcutaneously. Cardiac function was evaluated by echocardiogram and cardiac catheterization after 4 weeks. Cardiomyocytes apoptosis and apoptosis-related protein Fas were detected by in situ terminal deoxynucleotidyl transferase assay (TUNEL method) and Western blot, respectively. RESULTS The ADR and ADR+PBS groups showed significant deteriorations of left ventricular functions and high cardiomyocyte apoptosis index, as well as high Fas expressions. Meanwhile, the ADR+G-CSF group showed significant improvement in LV function, inhibition of cardiomyocyte apoptosis compared with the ADR and ADR+Phosphate-Buffered Saline PBS group. The Fas protein expression was remarkably attenuated as well. CONCLUSION Our results suggest that administration of G-CSF inhibited cardiomyocyte apoptosis and Fas protein expression and contributes to improving cardiac pump function in vivo in ADR induced DCM rat model.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/administration & dosage
- Antibiotics, Antineoplastic/toxicity
- Apoptosis/drug effects
- Blotting, Western
- Body Weight/drug effects
- Cardiomyopathy, Dilated/chemically induced
- Cardiomyopathy, Dilated/drug therapy
- Cardiomyopathy, Dilated/physiopathology
- Disease Models, Animal
- Doxorubicin/administration & dosage
- Doxorubicin/toxicity
- Echocardiography
- Granulocyte Colony-Stimulating Factor/administration & dosage
- Granulocyte Colony-Stimulating Factor/pharmacology
- Granulocyte Colony-Stimulating Factor/therapeutic use
- Heart Ventricles/drug effects
- Heart Ventricles/physiopathology
- In Situ Nick-End Labeling
- Injections, Subcutaneous
- Male
- Microscopy, Electron
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/ultrastructure
- Rats
- Rats, Wistar
- Receptors, Tumor Necrosis Factor/metabolism
- Ventricular Pressure/drug effects
- fas Receptor
Collapse
Affiliation(s)
- Xu Wei Hou
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 30020, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|