1
|
Hosseinpour A, Kamalpour J, Dehdari Ebrahimi N, Mirhosseini SA, Sadeghi A, Kavousi S, Attar A. Comparative effectiveness of mesenchymal stem cell versus bone-marrow mononuclear cell transplantation in heart failure: a meta-analysis of randomized controlled trials. Stem Cell Res Ther 2024; 15:202. [PMID: 38971816 PMCID: PMC11227704 DOI: 10.1186/s13287-024-03829-7] [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: 06/04/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND There is no clear evidence on the comparative effectiveness of bone-marrow mononuclear cell (BMMNC) vs. mesenchymal stromal cell (MSC) stem cell therapy in patients with chronic heart failure (HF). METHODS Using a systematic approach, eligible randomized controlled trials (RCTs) of stem cell therapy (BMMNCs or MSCs) in patients with HF were retrieved to perform a meta-analysis on clinical outcomes (major adverse cardiovascular events (MACE), hospitalization for HF, and mortality) and echocardiographic indices (including left ventricular ejection fraction (LVEF)) were performed using the random-effects model. A risk ratio (RR) or mean difference (MD) with corresponding 95% confidence interval (CI) were pooled based on the type of the outcome and subgroup analysis was performed to evaluate the potential differences between the types of cells. RESULTS The analysis included a total of 36 RCTs (1549 HF patients receiving stem cells and 1252 patients in the control group). Transplantation of both types of cells in patients with HF resulted in a significant improvement in LVEF (BMMNCs: MD (95% CI) = 3.05 (1.11; 4.99) and MSCs: MD (95% CI) = 2.82 (1.19; 4.45), between-subgroup p = 0.86). Stem cell therapy did not lead to a significant change in the risk of MACE (MD (95% CI) = 0.83 (0.67; 1.06), BMMNCs: RR (95% CI) = 0.59 (0.31; 1.13) and MSCs: RR (95% CI) = 0.91 (0.70; 1.19), between-subgroup p = 0.12). There was a marginally decreased risk of all-cause death (MD (95% CI) = 0.82 (0.68; 0.99)) and rehospitalization (MD (95% CI) = 0.77 (0.61; 0.98)) with no difference among the cell types (p > 0.05). CONCLUSION Both types of stem cells are effective in improving LVEF in patients with heart failure without any noticeable difference between the cells. Transplantation of the stem cells could not decrease the risk of major adverse cardiovascular events compared with controls. Future trials should primarily focus on the impact of stem cell transplantation on clinical outcomes of HF patients to verify or refute the findings of this study.
Collapse
Affiliation(s)
- Alireza Hosseinpour
- Department of Cardiovascular Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jahangir Kamalpour
- Department of Cardiovascular Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Alireza Sadeghi
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahin Kavousi
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Armin Attar
- Department of Cardiovascular Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
2
|
In Search of the Holy Grail: Stem Cell Therapy as a Novel Treatment of Heart Failure with Preserved Ejection Fraction. Int J Mol Sci 2023; 24:ijms24054903. [PMID: 36902332 PMCID: PMC10003723 DOI: 10.3390/ijms24054903] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/20/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
Heart failure, a leading cause of hospitalizations and deaths, is a major clinical problem. In recent years, the increasing incidence of heart failure with preserved ejection fraction (HFpEF) has been observed. Despite extensive research, there is no efficient treatment for HFpEF available. However, a growing body of evidence suggests stem cell transplantation, due to its immunomodulatory effect, may decrease fibrosis and improve microcirculation and therefore, could be the first etiology-based therapy of the disease. In this review, we explain the complex pathogenesis of HFpEF, delineate the beneficial effects of stem cells in cardiovascular therapy, and summarize the current knowledge concerning cell therapy in diastolic dysfunction. Furthermore, we identify outstanding knowledge gaps that may indicate directions for future clinical studies.
Collapse
|
3
|
Bonora BM, Cappellari R, Grasso M, Mazzucato M, D'Anna M, Avogaro A, Fadini GP. Glycaemic Control Achieves Sustained Increases of Circulating Endothelial Progenitor Cells in Patients Hospitalized for Decompensated Diabetes: An Observational Study. Diabetes Ther 2022; 13:1327-1337. [PMID: 35676613 PMCID: PMC9240124 DOI: 10.1007/s13300-022-01273-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/12/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND AND AIM Diabetes reduces the levels of circulating endothelial progenitor cells (EPCs), which contribute to vascular homeostasis. In turn, low EPCs levels predict progression of chronic complications. Several studies have shown that hyperglycaemia exerts detrimental effects on EPCs. Improvement in glucose control with glucose-lowering medications is associated with an increase of EPCs, but only after a long time of good glycaemic control. In the present study, we examined the effect of a rapid glycaemic amelioration on EPC levels in subjects hospitalized for decompensated diabetes. METHODS We used flow cytometry to quantify EPCs (CD34+/CD133+KDR+) in patients hospitalized for/with decompensated diabetes at admission, at discharge, and 2 months after the discharge. During hospitalization, all patients received intensive insulin therapy. RESULTS Thirty-nine patients with type 1 or type 2 diabetes were enrolled. Average (± SEM) fasting glucose decreased from 409.2 ± 25.9 mg/dl at admission to 190.4 ± 12.0 mg/dl at discharge and to 169.0 ± 10.3 at 2 months (both p < 0.001). EPCs (per million blood cells) significantly increased from hospital admission (13.1 ± 1.4) to discharge (16.4 ± 1.1; p = 0.022) and remained stable after 2 months (15.5 ± 1.7; p = 0.023 versus baseline). EPCs increased significantly more in participants with newly-diagnosed diabetes than in those with pre-existing diabetes. The increase in EPCs was significant in type 1 but not in type 2 diabetes and in those without chronic complications. CONCLUSION In individuals hospitalized for decompensated diabetes, insulin therapy rapidly increases EPC levels for up to 2 months. EPC defect, reflecting impaired vascular repair capacity, may be reversible in the early diabetes stages.
Collapse
Affiliation(s)
- Benedetta Maria Bonora
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
- Venetian Institute of Molecular Medicine, 35128, Padua, Italy
| | | | - Marco Grasso
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
| | - Marta Mazzucato
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
| | - Marianna D'Anna
- Venetian Institute of Molecular Medicine, 35128, Padua, Italy
| | - Angelo Avogaro
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128, Padua, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128, Padua, Italy.
- Venetian Institute of Molecular Medicine, 35128, Padua, Italy.
| |
Collapse
|
4
|
Portillo Esquivel LE, Zhang B. Application of Cell, Tissue, and Biomaterial Delivery in Cardiac Regenerative Therapy. ACS Biomater Sci Eng 2021; 7:1000-1021. [PMID: 33591735 DOI: 10.1021/acsbiomaterials.0c01805] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases (CVD) are the leading cause of death around the world, being responsible for 31.8% of all deaths in 2017 (Roth, G. A. et al. The Lancet 2018, 392, 1736-1788). The leading cause of CVD is ischemic heart disease (IHD), which caused 8.1 million deaths in 2013 (Benjamin, E. J. et al. Circulation 2017, 135, e146-e603). IHD occurs when coronary arteries in the heart are narrowed or blocked, preventing the flow of oxygen and blood into the cardiac muscle, which could provoke acute myocardial infarction (AMI) and ultimately lead to heart failure and death. Cardiac regenerative therapy aims to repair and refunctionalize damaged heart tissue through the application of (1) intramyocardial cell delivery, (2) epicardial cardiac patch, and (3) acellular biomaterials. In this review, we aim to examine these current approaches and challenges in the cardiac regenerative therapy field.
Collapse
Affiliation(s)
| | - Boyang Zhang
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada.,School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontaria L8S 4L8, Canada
| |
Collapse
|
5
|
Pacheco-Herrero M, Soto-Rojas LO, Reyes-Sabater H, Garcés-Ramirez L, de la Cruz López F, Villanueva-Fierro I, Luna-Muñoz J. Current Status and Challenges of Stem Cell Treatment for Alzheimer's Disease. J Alzheimers Dis 2021; 84:917-935. [PMID: 34633316 PMCID: PMC8673502 DOI: 10.3233/jad-200863] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2021] [Indexed: 12/23/2022]
Abstract
Neurodegenerative diseases called tauopathies, such as Alzheimer's disease (AD), frontotemporal dementia, progressive supranuclear palsy, and Parkinson's disease, among others, are characterized by the pathological processing and accumulation of tau protein. AD is the most prevalent neurodegenerative disease and is characterized by two lesions: neurofibrillary tangles (NFTs) and neuritic plaques. The presence of NFTs in the hippocampus and neocortex in early and advanced stages, respectively, correlates with the patient's cognitive deterioration. So far, no drugs can prevent, decrease, or limit neuronal death due to abnormal pathological tau accumulation. Among potential non-pharmacological treatments, physical exercise has been shown to stimulate the development of stem cells (SCs) and may be useful in early stages. However, this does not prevent neuronal death from the massive accumulation of NFTs. In recent years, SCs therapies have emerged as a promising tool to repopulate areas involved in cognition in neurodegenerative diseases. Unfortunately, protocols for SCs therapy are still being developed and the mechanism of action of such therapy remains unclear. In this review, we show the advances and limitations of SCs therapy. Finally, we provide a critical analysis of its clinical use for AD.
Collapse
Affiliation(s)
- Mar Pacheco-Herrero
- Neuroscience Research Laboratory, Faculty of Health Sciences, Pontificia Universidad Católica Madre y Maestra, Dominican Republic
| | - Luis O. Soto-Rojas
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, State of Mexico, Mexico
| | - Heidy Reyes-Sabater
- Neuroscience Research Laboratory, Faculty of Health Sciences, Pontificia Universidad Católica Madre y Maestra, Dominican Republic
| | - Linda Garcés-Ramirez
- Escuela Nacional de Ciencias Biológicas, Depto de Fisiología, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Fidel de la Cruz López
- Escuela Nacional de Ciencias Biológicas, Depto de Fisiología, Instituto Politécnico Nacional, Mexico City, Mexico
| | | | - José Luna-Muñoz
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores Cuautitlán, UNAM, State of Mexico, Mexico
- Banco Nacional de Cerebros-UNPHU, Universidad Nacional Pedro Henríquez Ureña, Dominican Republic
| |
Collapse
|
6
|
Point of care, bone marrow mononuclear cell therapy in ischemic heart failure patients personalized for cell potency: 12-month feasibility results from CardiAMP heart failure roll-in cohort. Int J Cardiol 2020; 326:131-138. [PMID: 33091520 DOI: 10.1016/j.ijcard.2020.10.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 09/24/2020] [Accepted: 10/15/2020] [Indexed: 01/09/2023]
Abstract
AIM Heart failure following myocardial infarction (MI) is a potentially lethal problem with a staggering incidence. The CardiAMP Heart Failure trial represents the first attempt to personalize marrow-derived cell-based therapy to individuals with cell characteristics associated with beneficial responses in prior trials. Before the initiation of the randomized pivotal trial, an open-label "roll-in cohort" was completed to ensure the feasibility of the protocol's procedures. METHODS Patients with chronic post-MI heart failure (NYHA class II-III) receiving stable, guideline-directed medical therapy with a left ventricular ejection fraction between 20 and 40% were eligible. Two weeks prior to treatment, a ~ 5 mL bone marrow aspiration was performed to examine "cell potency". On treatment day, a 60 mL bone marrow aspiration, bone marrow mononuclear cell (BM MNC) enrichment and transendocardial injection of 200 million BM MNC's was performed in a single, point of care encounter. Patients were then followed to assess clinical outcomes. RESULTS The cell potency small volume bone marrow aspirate, the 60 mL bone marrow aspirate, and transendocardial injections were well tolerated in 10 patients enrolled. There were no serious adverse events related to bone marrow aspiration or cell delivery. Improvement in 6-min walk distance was observed at 6 months (+47.8 m, P = 0.01) and trended to improvement at 12 months (+46.4, P = 0.06). Similarly, trends to improved NYHA heart failure functional class, quality of life, left ventricular ejection fraction and recruitment of previously akinetic left ventricular wall segments were observed. CONCLUSION All CardiAMP HF protocol procedures were feasible and well tolerated. Favorable functional, echo and quality of life trends suggest this approach may offer promise for patients with post MI heart failure. The randomized CardiAMP Heart Failure pivotal trial is underway to confirm the efficacy of this approach. CLINICAL TRIAL REGISTRATION https://clinicaltrials.gov/ct2/show/NCT02438306.
Collapse
|
7
|
Abstract
Interleukin-15 is a pleotropic factor, capable of modulating metabolism, survival, proliferation, and differentiation in many different cell types. The rationale behind this study relates to previous work demonstrating that IL-15 is a major factor present in stem cell extracts, which protects cardiomyocytes subjected to hypoxic stress in vitro. The objective of this current study was to assess whether administration of IL-15 peptide will also show protective effects in vivo. The data indicate that administration of IL-15 reduces cell death, increases vascularity, decreases scar size, and significantly improves left ventricular ejection fraction in a mouse model of myocardial infarction.
Collapse
|
8
|
Parrotta EI, Lucchino V, Scaramuzzino L, Scalise S, Cuda G. Modeling Cardiac Disease Mechanisms Using Induced Pluripotent Stem Cell-Derived Cardiomyocytes: Progress, Promises and Challenges. Int J Mol Sci 2020; 21:E4354. [PMID: 32575374 PMCID: PMC7352327 DOI: 10.3390/ijms21124354] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are a class of disorders affecting the heart or blood vessels. Despite progress in clinical research and therapy, CVDs still represent the leading cause of mortality and morbidity worldwide. The hallmarks of cardiac diseases include heart dysfunction and cardiomyocyte death, inflammation, fibrosis, scar tissue, hyperplasia, hypertrophy, and abnormal ventricular remodeling. The loss of cardiomyocytes is an irreversible process that leads to fibrosis and scar formation, which, in turn, induce heart failure with progressive and dramatic consequences. Both genetic and environmental factors pathologically contribute to the development of CVDs, but the precise causes that trigger cardiac diseases and their progression are still largely unknown. The lack of reliable human model systems for such diseases has hampered the unraveling of the underlying molecular mechanisms and cellular processes involved in heart diseases at their initial stage and during their progression. Over the past decade, significant scientific advances in the field of stem cell biology have literally revolutionized the study of human disease in vitro. Remarkably, the possibility to generate disease-relevant cell types from induced pluripotent stem cells (iPSCs) has developed into an unprecedented and powerful opportunity to achieve the long-standing ambition to investigate human diseases at a cellular level, uncovering their molecular mechanisms, and finally to translate bench discoveries into potential new therapeutic strategies. This review provides an update on previous and current research in the field of iPSC-driven cardiovascular disease modeling, with the aim of underlining the potential of stem-cell biology-based approaches in the elucidation of the pathophysiology of these life-threatening diseases.
Collapse
|
9
|
Nasseri Maleki S, Aboutaleb N, Nazarinia D, Allahverdi Beik S, Qolamian A, Nobakht M. Conditioned medium obtained from human amniotic membrane-derived mesenchymal stem cell attenuates heart failure injury in rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:1253-1258. [PMID: 32128088 PMCID: PMC7038431 DOI: 10.22038/ijbms.2019.36617.8722] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 04/05/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVES Heart failure (HF) is one of the leading causes of death worldwide. Due to beneficial effects of stem cells, paracrine secretion of them has recently been used by researchers. The purpose of this study was to investigate the effects of intravenous injection (IV) of conditioned medium (CM) of human amniotic membrane-derived mesenchymal stem cell (MSC-CM) on HF. MATERIALS AND METHODS Male Wistar rats (n=35, 180 g) were randomly divided into five groups: sham, HF, HF+MSC-CM, HF+culture medium and HF+phosphate-buffered saline (PBS). To induce HF, isoproterenol (170 mg/kg/d) was injected subcutaneously for 4 consecutive days. After 28 days, induction of HF was evaluated by echocardiography. A day after echocardiography, 50 μg culture medium/5 ml PBS in HF+culture medium group, 50 μg MSC-CM/5 ml PBS in HF+MSC-CM group and 5 ml PBS in HF+PBS group were injected two times for 4 successive days. The echocardiography was performed 4 weeks after the last injection of isoproterenol. To evaluate the fibrosis, morphology, and cardiac function, Trichrome Masson's staining, Hematoxylin and Eosin staining and echocardiography were performed, respectively. RESULTS CM significantly increased fractional shortening and ejection fraction, and also significantly decreased apoptotic nuclear condensation. Moreover, significant decreased level of fibrosis and increased level of angiogenesis was observed in the treatment group (P<0.05). CONCLUSION Our results indicated that IV injection of CM has therapeutic effects on HF by reducing fibrosis and preventing the progression of failure due to its paracrine effects.
Collapse
Affiliation(s)
- Solmaz Nasseri Maleki
- Physiology Research Center and Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nahid Aboutaleb
- Physiology Research Center and Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Donya Nazarinia
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Allahverdi Beik
- Physiology Research Center and Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Asadollah Qolamian
- Physiology Research Center and Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maliheh Nobakht
- Department of Histology and Neuroscience, Anti-microbial Resistance Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Han YH, Kim KH, Abdi S, Kim TK. Stem cell therapy in pain medicine. Korean J Pain 2019; 32:245-255. [PMID: 31569916 PMCID: PMC6813895 DOI: 10.3344/kjp.2019.32.4.245] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 09/16/2019] [Accepted: 09/17/2019] [Indexed: 12/31/2022] Open
Abstract
Stem cells are attracting attention as a key element in future medicine, satisfying the desire to live a healthier life with the possibility that they can regenerate tissue damaged or degenerated by disease or aging. Stem cells are defined as undifferentiated cells that have the ability to replicate and differentiate themselves into various tissues cells. Stem cells, commonly encountered in clinical or preclinical stages, are largely classified into embryonic, adult, and induced pluripotent stem cells. Recently, stem cell transplantation has been frequently applied to the treatment of pain as an alternative or promising approach for the treatment of severe osteoarthritis, neuropathic pain, and intractable musculoskeletal pain which do not respond to conventional medicine. The main idea of applying stem cells to neuropathic pain is based on the ability of stem cells to release neurotrophic factors, along with providing a cellular source for replacing the injured neural cells, making them ideal candidates for modulating and possibly reversing intractable neuropathic pain. Even though various differentiation capacities of stem cells are reported, there is not enough knowledge and technique to control the differentiation into desired tissues in vivo. Even though the use of stem cells is still in the very early stages of clinical use and raises complicated ethical problems, the future of stem cells therapies is very bright with the help of accumulating evidence and technology.
Collapse
Affiliation(s)
- Yong Hee Han
- Department of Anesthesia and Pain Medicine, Pusan National University School of Medicine, Yangsan, Korea
| | - Kyung Hoon Kim
- Department of Anesthesia and Pain Medicine, Pusan National University School of Medicine, Yangsan, Korea
| | - Salahadin Abdi
- Division of Anesthesia and Critical Care, Department of Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tae Kyun Kim
- Department of Anesthesia and Pain Medicine, Pusan National University School of Medicine, Yangsan, Korea
| |
Collapse
|
11
|
Fanaroff AC, Morrow V, Krucoff MW, Seltzer JH, Perin EC, Taylor DA, Miller LW, Zeiher AM, Fernández-Avilés F, Losordo DW, Henry TD, Povsic TJ. A Path Forward for Regenerative Medicine. Circ Res 2019; 123:495-505. [PMID: 30355250 DOI: 10.1161/circresaha.118.313261] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Although clinical trials of cell-based approaches to cardiovascular disease have yielded some promising results, no cell-based therapy has achieved regulatory approval for a cardiovascular indication. To broadly assess the challenges to regulatory approval and identify strategies to facilitate this goal, the Cardiac Safety Research Consortium sponsored a session during the Texas Heart Institute International Symposium on Cardiovascular Regenerative Medicine in September 2017. This session convened leaders in cardiovascular regenerative medicine, including participants from academia, the pharmaceutical industry, the US Food and Drug Administration, and the Cardiac Safety Research Consortium, with particular focus on treatments closest to regulatory approval. A goal of the session was to identify barriers to regulatory approval and potential pathways to overcome them. Barriers identified include manufacturing and therapeutic complexity, difficulties identifying an optimal comparator group, limited industry capacity for funding pivotal clinical trials, and challenges to demonstrating efficacy on clinical end points required for regulatory decisions. Strategies to overcome these barriers include precompetitive development of a cell therapy registry network to enable dual-purposing of clinical data as part of pragmatic clinical trial design, development of standardized terminology for product activity and end points to facilitate this registry, use of innovative statistical methods and quality of life or functional end points to supplement outcomes such as death or heart failure hospitalization and reduce sample size, involvement of patients in determining the research agenda, and use of the Food and Drug Administration's new Regenerative Medicine Advanced Therapy designation to facilitate early discussion with regulatory authorities when planning development pathways.
Collapse
Affiliation(s)
- Alexander C Fanaroff
- From the Division of Cardiology (A.C.F., M.W.K., T.J.P.).,Duke Clinical Research Institute (A.C.F., V.M., M.W.K., T.J.P.)
| | - Valarie Morrow
- Duke Clinical Research Institute (A.C.F., V.M., M.W.K., T.J.P.)
| | - Mitchell W Krucoff
- From the Division of Cardiology (A.C.F., M.W.K., T.J.P.).,Duke Clinical Research Institute (A.C.F., V.M., M.W.K., T.J.P.)
| | - Jonathan H Seltzer
- Duke University School of Medicine, Durham, NC; ACI Clinical, Bala Cynwyd, PA (J.H.S.)
| | - Emerson C Perin
- Stem Cell Center and Regenerative Medicine Research, Texas Heart Institute, Houston (E.C.P., D.A.T., L.W.M.)
| | - Doris A Taylor
- Stem Cell Center and Regenerative Medicine Research, Texas Heart Institute, Houston (E.C.P., D.A.T., L.W.M.)
| | - Leslie W Miller
- Stem Cell Center and Regenerative Medicine Research, Texas Heart Institute, Houston (E.C.P., D.A.T., L.W.M.)
| | - Andreas M Zeiher
- Department of Cardiology, University of Frankfurt, Germany (A.M.Z.)
| | - Francisco Fernández-Avilés
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, CIBERCV, Madrid, Spain (F.F.-A.)
| | - Douglas W Losordo
- Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (D.W.L.).,Caladrius Biosciences Inc, Basking Ridge, NJ (D.W.L.)
| | - Timothy D Henry
- Cedars-Sinai Smidt Heart Institute, Los Angeles, CA (T.D.H.)
| | - Thomas J Povsic
- From the Division of Cardiology (A.C.F., M.W.K., T.J.P.).,Duke Clinical Research Institute (A.C.F., V.M., M.W.K., T.J.P.)
| |
Collapse
|
12
|
Mann I, Tseng CCS, Rodrigo SF, Koudstaal S, van Ramshorst J, Beeres SL, Dibbets-Schneider P, de Geus-Oei LF, Lamb HJ, Wolterbeek R, Zwaginga JJ, Fibbe WE, Westinga K, Bax JJ, Doevendans PA, Schalij MJ, Chamuleau SAJ, Atsma DE. Intramyocardial bone marrow cell injection does not lead to functional improvement in patients with chronic ischaemic heart failure without considerable ischaemia. Neth Heart J 2018; 27:81-92. [PMID: 30569306 PMCID: PMC6352621 DOI: 10.1007/s12471-018-1213-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background It has been suggested that bone marrow cell injection may have beneficial effects in patients with chronic ischaemic heart disease. However, previous trials have led to discrepant results of cell-based therapy in patients with chronic heart failure. The aim of this study was to evaluate the efficacy of intramyocardial injection of mononuclear bone marrow cells in patients with chronic ischaemic heart failure with limited stress-inducible myocardial ischaemia. Methods and results This multicentre, randomised, placebo-controlled trial included 39 patients with no-option chronic ischaemic heart failure with a follow-up of 12 months. A total of 19 patients were randomised to autologous intramyocardial bone marrow cell injection (cell group) and 20 patients received a placebo injection (placebo group). The primary endpoint was the group difference in change of left ventricular ejection fraction, as determined by single-photon emission tomography. On follow-up at 3 and 12 months, change of left ventricular ejection fraction in the cell group was comparable with change in the placebo group (P = 0.47 and P = 0.08, respectively). Also secondary endpoints, including left ventricle volumes, myocardial perfusion, functional and clinical parameters did not significantly change in the cell group as compared to placebo. Neither improvement was demonstrated in a subgroup of patients with stress-inducible ischaemia (P = 0.54 at 3‑month and P = 0.15 at 12-month follow-up). Conclusion Intramyocardial bone marrow cell injection does not improve cardiac function, nor functional and clinical parameters in patients with severe chronic ischaemic heart failure with limited stress-inducible ischaemia. Clinical Trial Registration: NTR2516 Electronic supplementary material The online version of this article (10.1007/s12471-018-1213-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- I Mann
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - C C S Tseng
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - S F Rodrigo
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - S Koudstaal
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J van Ramshorst
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - S L Beeres
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - P Dibbets-Schneider
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - L F de Geus-Oei
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - H J Lamb
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - R Wolterbeek
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - J J Zwaginga
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - W E Fibbe
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - K Westinga
- Department of Cell Therapy Facility, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J J Bax
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - P A Doevendans
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - M J Schalij
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - S A J Chamuleau
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - D E Atsma
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
13
|
Raval AN, Cook TD, Duckers HJ, Johnston PV, Traverse JH, Abraham WT, Altman PA, Pepine CJ. The CardiAMP Heart Failure trial: A randomized controlled pivotal trial of high-dose autologous bone marrow mononuclear cells using the CardiAMP cell therapy system in patients with post-myocardial infarction heart failure: Trial rationale and study design. Am Heart J 2018; 201:141-148. [PMID: 29803986 DOI: 10.1016/j.ahj.2018.03.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/24/2018] [Indexed: 01/10/2023]
Abstract
BACKGROUND Heart failure following myocardial infarction is a common, disabling, and deadly condition. Direct injection of autologous bone marrow mononuclear cells into the myocardium may result in improved functional recovery, relieve symptoms, and improve other cardiovascular outcomes. METHODS CardiAMP-HF is a randomized, double-blind, sham-controlled, pivotal trial designed to investigate the safety and efficacy of autologous bone marrow mononuclear cells treatment for patients with medically refractory and symptomatic ischemic cardiomyopathy. The primary end point is change in 6-minute walk distance adjusted for major adverse cardiovascular events at 12 months following treatment. Particularly novel aspects of this trial include a cell potency assay to screen subjects who have bone marrow cell characteristics that suggest a favorable response to treatment, a point-of-care treatment method, a high target dose of 200 million cells, and an efficient transcatheter intramyocardial delivery method that is associated with high cell retention. CONCLUSIONS This novel approach may lead to a new treatment for those with ischemic heart disease suffering from medically refractory heart failure.
Collapse
|
14
|
Chehelcheraghi F, Bayat M, Chien S. Effect of Mesenchymal Stem Cells and Chicken Embryo Extract on Flap Viability and Mast Cells in Rat Skin Flaps. J INVEST SURG 2018; 33:123-133. [DOI: 10.1080/08941939.2018.1479006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Farzaneh Chehelcheraghi
- Anatomical Sciences Department, School of Medicine, Lorestan University Medical of Sciences, Khoramabad, IR Iran
| | - Mohammad Bayat
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Price Institute of Surgical Research, University of Louisville, and Noveratech LLC of Louisville, Louisville, Kentucky, USA; Supported in part by NIH grant DK105692
| | - Sufan Chien
- Price Institute of Surgical Research, University of Louisville, and Noveratech LLC of Louisville, Louisville, Kentucky, USA; Supported in part by NIH grant DK105692
| |
Collapse
|
15
|
Platelet-Derived Growth Factor Receptor-Alpha Expressing Cardiac Progenitor Cells Can Be Derived from Previously Cryopreserved Human Heart Samples. Stem Cells Dev 2018; 27:184-198. [DOI: 10.1089/scd.2017.0082] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
16
|
Tan RP, Lee BS, Chan AH, Yuen SCG, Hung J, Wise SG, Ng MK. Non-invasive tracking of injected bone marrow mononuclear cells to injury and implanted biomaterials. Acta Biomater 2017; 53:378-388. [PMID: 28167301 DOI: 10.1016/j.actbio.2017.02.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/31/2017] [Accepted: 02/01/2017] [Indexed: 02/07/2023]
Abstract
Biomaterial scaffolds enhancing the engraftment of transplanted bone-marrow mononuclear cells (BM-MNC) have enormous potential for tissue regeneration applications. However, development of appropriate materials is challenging given the precise microenvironments required to support BM-MNC engraftment and function. In this study, we have developed a non-invasive, real-time tracking model of injected BM-MNC engraftment to wounds and implanted biomaterial scaffolds. BM-MNCs, encoded with firefly luciferase and enhanced GFP reporter genes, were tail vein injected into subcutaneously wounded mice. Luciferase-dependent cell bioluminescence curves revealed our injected BM-MNCs homed to and engrafted within subcutaneous wound sites over the course of 21days. Further immunohistochemical characterization showed that these engrafted cells drove functional changes by increasing the number of immune cells present at early time points and remodelling cell phenotypes at later time points. Using this model, we subcutaneously implanted electrospun polycaprolactone (PCL) and PCL/Collagen scaffolds, to determine differences in exogenous BM-MNC response to these materials. Following BM-MNC injection, immunohistochemical analysis revealed a high exogenous BM-MNC density around the periphery of PCL scaffolds consistent with a classical foreign body response. In contrast, transplanted BM-MNCs engrafted throughout PCL/Collagen scaffolds indicating an improved biological response. Importantly, these differences were closely correlated with the real-time bioluminescence curves, with PCL/Collagen scaffolds exhibiting a∼2-fold increase in maximum bioluminescence compared with PCL scaffolds. Collectively, these results demonstrate a new longitudinal cell tracking model that can non-invasively determine transplanted BM-MNC homing and engraftment to biomaterials, providing a valuable tool to inform the design scaffolds that help augment current BM-MNC tissue engineering strategies. STATEMENT OF SIGNIFICANCE Tracking the dynamic behaviour of transplanted bone-marrow mononuclear cells (BM-MNCs) is a long-standing research goal. Conventional methods involving contrast and tracer agents interfere with cellular function while also yielding false signals. The use of bioluminescence addresses these shortcomings while allowing for real-time non-invasive tracking in vivo. Given the failures of transplanted BM-MNCs to engraft into injured tissue, biomaterial scaffolds capable of attracting and enhancing BM-MNC engraftment at sites of injury are highly sought in numerous tissue engineering applications. To this end, the results from this study demonstrate a new longitudinal tracking model that can non-invasively determine exogenous BM-MNC homing and engraftment to biomaterials, providing a valuable tool to inform the design of scaffolds with implications for countless tissue engineering applications.
Collapse
|
17
|
Bao L, Meng Q, Li Y, Deng S, Yu Z, Liu Z, Zhang L, Fan H. C-Kit Positive Cardiac Stem Cells and Bone Marrow-Derived Mesenchymal Stem Cells Synergistically Enhance Angiogenesis and Improve Cardiac Function After Myocardial Infarction in a Paracrine Manner. J Card Fail 2017; 23:403-415. [PMID: 28284757 DOI: 10.1016/j.cardfail.2017.03.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 02/16/2017] [Accepted: 03/06/2017] [Indexed: 11/17/2022]
Abstract
BACKGROUND Stem cell transplantation offers a promising treatment for heart failure. Recent studies show that both c-kit positive cardiac stem cells (CSCs) and bone marrow-derived mesenchymal stem cells (BM-MSCs) are good candidates for stem cell therapy to treat heart failure; however, the exact mechanism of stem cell therapy in improving cardiac function of ischemic cardiomyopathy is not fully known. Our objective was to test our hypothesis that CSCs and/or BM-MSCs repair the damaged heart by boosting post-myocardial infarction (MI) angiogenesis in a paracrine manner. METHODS AND RESULTS We isolated and purified CSCs and BM-MSCs from rats. Intramyocardial injections of CSCs and/or BM-MSCs were performed at 28 days after MI. We applied cardiac ultrasound and histological analysis to evaluate the effect of cell therapy on cardiac function and cardiac remodeling. In vivo donor cell transplantation experiments showed that CSCs and/or BM-MSCs improved cardiac function after MI and reduced infarct size. However, in vivo cell tracking experiments showed that minimal donor cells remained in the myocardium after cell transplantation. Our further in vitro and in vivo experiments showed that transplantation of CSCs enhanced the expression of pro-angiogenic factors and boosted post-MI angiogenesis in the myocardium in a paracrine manner, which in part contributed to the effect of CSCs on cardiac recovery after MI. CSCs and BM-MSCs synergistically inhibited CSC/BM-MSC apoptosis and enhanced their proliferation in a paracrine manner. This resulted in a larger number of transplanted cells remaining in the post-MI myocardium after coinjection of CSCs and BM-MSCs, and therefore the accumulation of more pro-angiogenic factors in the heart tissue compared to transplantation of CSCs or MSCs alone. Consequently, transplantation of both CSCs and BM-MSCs was superior to transplantation of either CSCs or BM-MSCs alone to boost post-MI angiogenesis and improve cardiac function after MI. CONCLUSION C-kit+ CSC and/or BM-MSC transplantation can improve cardiac function after MI in a paracrine manner. Coinjection of both CSCs and BM-MSCs improves cardiac function more significantly than CSC or BM-MSC transplantation alone in a paracrine manner by improving the engraftment of donor cells and boosting the expression of multiple pro-angiogenic factors.
Collapse
Affiliation(s)
- Luer Bao
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Qingshu Meng
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yuan Li
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Shengqiong Deng
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Zuoren Yu
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Zhongmin Liu
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Lin Zhang
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | - Huimin Fan
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| |
Collapse
|
18
|
Le TYL, Thavapalachandran S, Kizana E, Chong JJ. New Developments in Cardiac Regeneration. Heart Lung Circ 2016; 26:316-322. [PMID: 27916592 DOI: 10.1016/j.hlc.2016.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 11/07/2016] [Indexed: 01/13/2023]
Abstract
Numerous pharmacological and device therapies have improved adverse cardiac remodelling and mortality in heart failure. However, none are able to regenerate damaged cardiac tissue. Stem cell based therapies using multipotent (adult) stem cells and pluripotent stem cells are new approaches that could potentially achieve the elusive goal of true cardiac regeneration. Over the past two decades, various stem cell based approaches have been shown to improve left ventricular function in pre-clinical animal models. Promising results rapidly led to clinical trials, initially using bone marrow-derived mononuclear cells, then mesenchymal stromal cell populations and, more recently, progenitor cells from the adult heart itself. These have been shown to be safe and have advanced our understanding of potential suitable recipients, cell delivery routes, and possible mechanisms of action. However, efficacy in these trials has been inconsistent. Human pluripotent stem cells (hPSCs) are another potential source of stem cells for cardiac regeneration. They could theoretically provide an unlimited source of cardiomyocytes or cardiac progenitors. Pre-clinical studies in both small and large animal models have shown robust engraftment and improvements in cardiac function. The first clinical trial using hPSC-derived cardiac derivatives has now commenced and others are imminent. In this brief review article, we summarise recent developments in stem cell therapies aimed at cardiac regeneration, including discussion of types of cell and non-cell-based strategies being explored.
Collapse
Affiliation(s)
- Thi Yen Loan Le
- Centre for Heart Research, Westmead Institute for Medical Research, Sydney, NSW, Australia; Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia
| | - Sujitha Thavapalachandran
- Centre for Heart Research, Westmead Institute for Medical Research, Sydney, NSW, Australia; Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Eddy Kizana
- Centre for Heart Research, Westmead Institute for Medical Research, Sydney, NSW, Australia; Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - James Jh Chong
- Centre for Heart Research, Westmead Institute for Medical Research, Sydney, NSW, Australia; Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
19
|
Sheng L, Mao X, Yu Q, Yu D. Effect of the PI3K/AKT signaling pathway on hypoxia-induced proliferation and differentiation of bone marrow-derived mesenchymal stem cells. Exp Ther Med 2016; 13:55-62. [PMID: 28123468 PMCID: PMC5245145 DOI: 10.3892/etm.2016.3917] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/09/2016] [Indexed: 12/15/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cell (BM-MSC) transplantation has been demonstrated to be an effective way of augmenting angiogenesis of ischemic tissue. The low oxygen conditions in ischemic tissue directly affect the biological behavior of engrafted cells. However, to date, the mechanism through which hypoxia regulates self-renewal, differentiation and paracrine function of BM-MSCs remains unclear. Clarification of this mechanism would be beneficial to the use of stem cell-based therapy. The PI3K/AKT pathway has been extensively investigated for its role in cell proliferation, cell transformation, paracrine function and angiogenesis. The present study aimed to analyze the role of PI3K/AKT pathway in hypoxia-induced proliferation of BM-MSCs and their differentiation into endothelial cells in vitro by the application of LY294002, a PI3K/AKT pathway inhibitor, with cells cultured in normoxia serving as a control. The results showed that rat BM-MSCs at passage 3 and 4 displayed only few phenotypical differences in the expression of surface antigens as detected by flow cytometry. When compared with the cells treated in normoxia, the proliferation of BM-MSCs in hypoxia was promoted, a greater number of cells expressed CD31 and a higher expression of vascular endothelial growth factor was observed after culture in hypoxic conditions. However, by inhibiting with LY294002, these changes induced by hypoxia were partly inhibited. In conclusion, the present study showed that the PI3K/AKT pathway served an important role in hypoxia-enhanced in vitro proliferation of BM-MSCs and their differentiation into endothelial cells and paracrine vascular endothelial growth factor.
Collapse
Affiliation(s)
- Lingling Sheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, P.R. China
| | - Xiyuan Mao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, P.R. China
| | - Qingxiong Yu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, P.R. China
| | - Dong Yu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, P.R. China
| |
Collapse
|
20
|
Dynamic Tracking of Injected Mesenchymal Stem Cells after Myocardial Infarction in Rats: A Serial 7T MRI Study. Stem Cells Int 2016; 2016:4656539. [PMID: 27656215 PMCID: PMC5021478 DOI: 10.1155/2016/4656539] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 07/25/2016] [Indexed: 11/26/2022] Open
Abstract
Purpose. To track the fate of micron-sized particles of iron oxide (MPIO) labeled mesenchymal stem cells (MSCs) in vivo in a rat myocardial infarction model using 7T magnetic resonance imaging (MRI) scanner. Materials and Methods. Male MSCs (2 × 106/50 μL) dual-labeled with MPIO and CM-DiI were injected into the infarct periphery 7 days after myocardial infarction (MI). The control group received cell-free media injection. The temporal stem cell location, signal intensity, and cardiac function were dynamically assessed using a 7T MRI at 24 h before transplantation (baseline), 3 days, 2 weeks, and 4 weeks after transplantation, respectively. Results. MR hypointensities caused by MPIOs were observed on T2⁎-weighted images at all time points after MSCs injection. Cine-MRI showed that MSCs moderated progressive left ventricular remodeling. Double staining for iron and CD68 revealed that most of the iron-positive cells were CD68-positive macrophages. Real-time PCR for rat SRY gene showed the number of survival MSCs considerably decreased after transplantation. MSC-treated hearts had significantly increased capillary density in peri-infarct region and lower cardiomyocytes apoptosis and fibrosis formation. Conclusions. Iron particles are not a reliable marker for in vivo tracking the long-term fate of MSCs engraftment. Despite of poor cell retention, MSCs moderate left ventricular remodeling after MI.
Collapse
|
21
|
Fadini GP, Ciciliot S, Albiero M. Concise Review: Perspectives and Clinical Implications of Bone Marrow and Circulating Stem Cell Defects in Diabetes. Stem Cells 2016; 35:106-116. [PMID: 27401837 DOI: 10.1002/stem.2445] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 05/27/2016] [Accepted: 05/28/2016] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus is a complex systemic disease characterized by severe morbidity and excess mortality. The burden of its multiorgan complications relies on an imbalance between hyperglycemic cell damage and defective endogenous reparative mechanisms. Inflammation and abnormalities in several hematopoietic components are typically found in diabetes. The discovery that diabetes reduces circulating stem/progenitor cells and impairs their function has opened an entire new field of study where diabetology comes into contact with hematology and regenerative medicine. It is being progressively recognized that such rare circulating cell populations mirror finely regulated processes involved in hematopoiesis, immunosurveillance, and peripheral tissue homeostasis. From a clinical perspective, pauperization of circulating stem cells predicts adverse outcomes and death. Furthermore, studies in murine models and humans have identified the bone marrow (BM) as a previously neglected site of diabetic end-organ damage, characterized by microangiopathy, neuropathy, fat deposition, and inflammation. As a result, diabetes impairs the mobilization of BM stem/progenitor cells, a defect known as mobilopathy or myelokathexis, with negative consequences for physiologic hematopoiesis, immune regulation, and tissue regeneration. A better understanding of the molecular and cellular processes that govern the BM stem cell niche, cell mobilization, and kinetics in peripheral tissues may uncover new therapeutic strategies for patients with diabetes. This concise review summarizes the current knowledge on the interplay between the BM, circulating stem cells, and diabetes, and sets the stages for future developments in the field. Stem Cells 2017;35:106-116.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, and Venetian Institute of Molecular Medicine, Padova, 35128, Italy
| | - Stefano Ciciliot
- Department of Medicine, University of Padova, and Venetian Institute of Molecular Medicine, Padova, 35128, Italy
| | - Mattia Albiero
- Department of Medicine, University of Padova, and Venetian Institute of Molecular Medicine, Padova, 35128, Italy
| |
Collapse
|
22
|
Xu JY, Lee YK, Ran X, Liao SY, Yang J, Au KW, Lai WH, Esteban MA, Tse HF. Generation of Induced Cardiospheres via Reprogramming of Skin Fibroblasts for Myocardial Regeneration. Stem Cells 2016; 34:2693-2706. [PMID: 27333945 DOI: 10.1002/stem.2438] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/15/2016] [Accepted: 05/28/2016] [Indexed: 11/06/2022]
Abstract
Recent pre-clinical and clinical studies have suggested that endogenous cardiospheres (eCS) are potentially safe and effective for cardiac regeneration following myocardial infarction (MI). Nevertheless the preparation of autologous eCS requires invasive myocardial biopsy with limited yield. We describe a novel approach to generate induced cardiospheres (iCS) from adult skin fibroblasts via somatic reprogramming. After infection with Sox2, Klf4, and Oct4, iCS were generated from mouse adult skin fibroblasts treated with Gsk3β inhibitor-(2'Z,3'E)- 6-Bromoindirubin-3'-oxime and Oncostatin M. They resembled eCS, but contained a higher percentage of cells expressing Mesp1, Isl1, and Nkx2.5. They were differentiated into functional cardiomyocytes in vitro with similar electrophysiological properties, calcium transient and contractile function to eCS and mouse embryonic stem cell-derived cardiomyocytes. Transplantation of iCS (1 × 106 cells) into mouse myocardium following MI had similar effects to transplantation of eCS but significantly better than saline or fibroblast in improving left ventricular ejection fraction, increasing anterior/septal ventricular wall thickness and capillary density in the infarcted region 4 weeks after transplantation. No tumor formation was observed. iCS generated from adult skin fibroblasts by somatic reprogramming and a cocktail of Gsk3β inhibitor-6-Bromoindirubin-3'-oxime and Oncostatin M may represent a novel source for cell therapy in MI. Stem Cells 2016;34:2693-2706.
Collapse
Affiliation(s)
- Jian-Yong Xu
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, SAR, China.,Shenzhen Institutes of Research and Innovation, the University of Hong Kong, Hong Kong, SAR, China
| | - Yee-Ki Lee
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, SAR, China
| | - Xinru Ran
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, SAR, China
| | - Song-Yan Liao
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, SAR, China
| | - Jiayin Yang
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, SAR, China.,Shenzhen Institutes of Research and Innovation, the University of Hong Kong, Hong Kong, SAR, China
| | - Ka-Wing Au
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, SAR, China
| | - Wing-Hon Lai
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, SAR, China
| | - Miguel A Esteban
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, SAR, China.,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, China.,Laboratory of Chromatin and Human Disease, Key Laboratory of Regenerative Biology of the Chinese Academy of Sciences and Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, SAR, China.,Shenzhen Institutes of Research and Innovation, the University of Hong Kong, Hong Kong, SAR, China.,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, China.,Research Center of Heart, Brain, Hormone and Healthy Aging, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| |
Collapse
|
23
|
Hodgkinson CP, Bareja A, Gomez JA, Dzau VJ. Emerging Concepts in Paracrine Mechanisms in Regenerative Cardiovascular Medicine and Biology. Circ Res 2016; 118:95-107. [PMID: 26837742 DOI: 10.1161/circresaha.115.305373] [Citation(s) in RCA: 201] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In the past decade, substantial evidence supports the paradigm that stem cells exert their reparative and regenerative effects, in large part, through the release of biologically active molecules acting in a paracrine fashion on resident cells. The data suggest the existence of a tissue microenvironment where stem cell factors influence cell survival, inflammation, angiogenesis, repair, and regeneration in a temporal and spatial manner.
Collapse
Affiliation(s)
- Conrad P Hodgkinson
- From the Department of Medicine, Mandel Center for Hypertension Research and Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC
| | - Akshay Bareja
- From the Department of Medicine, Mandel Center for Hypertension Research and Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC
| | - José A Gomez
- From the Department of Medicine, Mandel Center for Hypertension Research and Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC
| | - Victor J Dzau
- From the Department of Medicine, Mandel Center for Hypertension Research and Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC.
| |
Collapse
|
24
|
Mann I, Rodrigo SF, van Ramshorst J, Beeres SL, Dibbets-Schneider P, de Roos A, Wolterbeek R, Zwaginga JJ, Fibbe WE, Bax JJ, Schalij MJ, Atsma DE. Repeated Intramyocardial Bone Marrow Cell Injection in Previously Responding Patients With Refractory Angina Again Improves Myocardial Perfusion, Anginal Complaints, and Quality of Life. Circ Cardiovasc Interv 2016; 8:CIRCINTERVENTIONS.115.002740. [PMID: 26259770 DOI: 10.1161/circinterventions.115.002740] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Intramyocardial bone marrow cell injection is associated with improvements in myocardial perfusion and anginal symptoms in patients with refractory angina pectoris. This study evaluates the effect of repeated intramyocardial bone marrow cell injection in patients with residual or recurrent myocardial ischemia. METHODS AND RESULTS Twenty-three patients (17 men; 69±9 years) who had improved myocardial perfusion after the first injection but had residual or recurrent angina and ischemia on single-photon emission computed tomographic myocardial perfusion imaging were included. Patients again received intramyocardial injection of 100×10(6) autologous bone marrow mononuclear cells, 4.6±2.5 years after their first injection. No periprocedural complications occurred. Myocardial perfusion assessed using single-photon emission computed tomographic myocardial perfusion imaging improved from a summed stress score of 27.3±5.8 at baseline to 24.5±4.4 at 3 months (P=0.002) and 25.4±4.9 at 12 months of follow-up (P=0.002). Perfusion improvement after 3 months was comparable with the effect of the first injection (P=0.379). Anginal complaints improved ≤12 months after cell injection in Canadian Cardiovascular Society score (mean change at 3, 6, and 12 months: 0.6±0.9%, 0.5±0.9%, and 0.6±0.9%, respectively; Pslope=0.007, first versus repeated; P=0.188) and in quality of life score as measured by Seattle Angina Questionnaire (mean change at 3, 6, and 12 months: 7±14%, 8±14%, and 7±15%, respectively; Pslope=0.020, first versus repeated; P=0.126). CONCLUSIONS Repeated bone marrow cell injection in previously responding patients with refractory angina is associated with improvements in myocardial perfusion, anginal complaints, and quality of life score ≤12 months of follow-up. CLINICAL TRIAL REGISTRATION URL: http://www.trialregister.nl. Unique identifier: NTR2664.
Collapse
Affiliation(s)
- Imke Mann
- From the Departments of Cardiology (I.M., S.F.R., J.v.R., S.L.B., J.J.B., M.J.S., D.E.A.), Nuclear Medicine (P.D.-S.), Radiology (A.d.R.), Medical Statistics and Bioinformatics (R.W.), and Hematology (J.J.Z., W.E.F.), Leiden University Medical Center, Leiden, The Netherlands
| | - Sander F Rodrigo
- From the Departments of Cardiology (I.M., S.F.R., J.v.R., S.L.B., J.J.B., M.J.S., D.E.A.), Nuclear Medicine (P.D.-S.), Radiology (A.d.R.), Medical Statistics and Bioinformatics (R.W.), and Hematology (J.J.Z., W.E.F.), Leiden University Medical Center, Leiden, The Netherlands
| | - Jan van Ramshorst
- From the Departments of Cardiology (I.M., S.F.R., J.v.R., S.L.B., J.J.B., M.J.S., D.E.A.), Nuclear Medicine (P.D.-S.), Radiology (A.d.R.), Medical Statistics and Bioinformatics (R.W.), and Hematology (J.J.Z., W.E.F.), Leiden University Medical Center, Leiden, The Netherlands
| | - Saskia L Beeres
- From the Departments of Cardiology (I.M., S.F.R., J.v.R., S.L.B., J.J.B., M.J.S., D.E.A.), Nuclear Medicine (P.D.-S.), Radiology (A.d.R.), Medical Statistics and Bioinformatics (R.W.), and Hematology (J.J.Z., W.E.F.), Leiden University Medical Center, Leiden, The Netherlands
| | - Petra Dibbets-Schneider
- From the Departments of Cardiology (I.M., S.F.R., J.v.R., S.L.B., J.J.B., M.J.S., D.E.A.), Nuclear Medicine (P.D.-S.), Radiology (A.d.R.), Medical Statistics and Bioinformatics (R.W.), and Hematology (J.J.Z., W.E.F.), Leiden University Medical Center, Leiden, The Netherlands
| | - Albert de Roos
- From the Departments of Cardiology (I.M., S.F.R., J.v.R., S.L.B., J.J.B., M.J.S., D.E.A.), Nuclear Medicine (P.D.-S.), Radiology (A.d.R.), Medical Statistics and Bioinformatics (R.W.), and Hematology (J.J.Z., W.E.F.), Leiden University Medical Center, Leiden, The Netherlands
| | - Ron Wolterbeek
- From the Departments of Cardiology (I.M., S.F.R., J.v.R., S.L.B., J.J.B., M.J.S., D.E.A.), Nuclear Medicine (P.D.-S.), Radiology (A.d.R.), Medical Statistics and Bioinformatics (R.W.), and Hematology (J.J.Z., W.E.F.), Leiden University Medical Center, Leiden, The Netherlands
| | - Jaap J Zwaginga
- From the Departments of Cardiology (I.M., S.F.R., J.v.R., S.L.B., J.J.B., M.J.S., D.E.A.), Nuclear Medicine (P.D.-S.), Radiology (A.d.R.), Medical Statistics and Bioinformatics (R.W.), and Hematology (J.J.Z., W.E.F.), Leiden University Medical Center, Leiden, The Netherlands
| | - Willem E Fibbe
- From the Departments of Cardiology (I.M., S.F.R., J.v.R., S.L.B., J.J.B., M.J.S., D.E.A.), Nuclear Medicine (P.D.-S.), Radiology (A.d.R.), Medical Statistics and Bioinformatics (R.W.), and Hematology (J.J.Z., W.E.F.), Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen J Bax
- From the Departments of Cardiology (I.M., S.F.R., J.v.R., S.L.B., J.J.B., M.J.S., D.E.A.), Nuclear Medicine (P.D.-S.), Radiology (A.d.R.), Medical Statistics and Bioinformatics (R.W.), and Hematology (J.J.Z., W.E.F.), Leiden University Medical Center, Leiden, The Netherlands
| | - Martin J Schalij
- From the Departments of Cardiology (I.M., S.F.R., J.v.R., S.L.B., J.J.B., M.J.S., D.E.A.), Nuclear Medicine (P.D.-S.), Radiology (A.d.R.), Medical Statistics and Bioinformatics (R.W.), and Hematology (J.J.Z., W.E.F.), Leiden University Medical Center, Leiden, The Netherlands
| | - Douwe E Atsma
- From the Departments of Cardiology (I.M., S.F.R., J.v.R., S.L.B., J.J.B., M.J.S., D.E.A.), Nuclear Medicine (P.D.-S.), Radiology (A.d.R.), Medical Statistics and Bioinformatics (R.W.), and Hematology (J.J.Z., W.E.F.), Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
25
|
Ho YT, Poinard B, Kah JCY. Nanoparticle drug delivery systems and their use in cardiac tissue therapy. Nanomedicine (Lond) 2016; 11:693-714. [DOI: 10.2217/nnm.16.6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cardiovascular diseases make up one of the main causes of death today, with myocardial infarction and ischemic heart disease contributing a large share of the deaths reported. With mainstream clinical therapy focusing on palliative medicine following myocardial infarction, the structural changes that occur in the diseased heart will eventually lead to end-stage heart failure. Heart transplantation remains the only gold standard of cure but a shortage in donor organs pose a major problem that led to clinicians and researchers looking into alternative strategies for cardiac repair. This review will examine some alternative methods of treatment using chemokines and drugs carried by nanoparticles as drug delivering agents for the purposes of treating myocardial infarction through the promotion of revascularization. We will also provide an overview of existing studies involving such nanoparticulate drug delivery systems, their reported efficacy and the challenges facing their translation into ubiquitous clinical use.
Collapse
Affiliation(s)
- Yan Teck Ho
- Department of Biomedical Engineering, National University of Singapore, 9 Engineering Drive 1, Block EA #07–25, Singapore 117575
- NUS Graduate School of Integrative Sciences & Engineering, National University of Singapore, 28 Medical Drive, Singapore 117456
| | - Barbara Poinard
- Department of Biomedical Engineering, National University of Singapore, 9 Engineering Drive 1, Block EA #07–25, Singapore 117575
- NUS Graduate School of Integrative Sciences & Engineering, National University of Singapore, 28 Medical Drive, Singapore 117456
| | - James Chen Yong Kah
- Department of Biomedical Engineering, National University of Singapore, 9 Engineering Drive 1, Block EA #07–25, Singapore 117575
- NUS Graduate School of Integrative Sciences & Engineering, National University of Singapore, 28 Medical Drive, Singapore 117456
| |
Collapse
|
26
|
The Evolution of the Stem Cell Theory for Heart Failure. EBioMedicine 2015; 2:1871-9. [PMID: 26844266 PMCID: PMC4703721 DOI: 10.1016/j.ebiom.2015.11.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 10/16/2015] [Accepted: 11/04/2015] [Indexed: 12/22/2022] Open
Abstract
Various stem cell-based approaches for cardiac repair have achieved encouraging results in animal experiments, often leading to their rapid proceeding to clinical testing. However, freewheeling evolutionary developments of the stem cell theory might lead to dystopian scenarios where heterogeneous sources of therapeutic cells could promote mixed clinical outcomes in un-stratified patient populations. This review focuses on the lessons that should be learnt from the first generation of stem cell-based strategies and emphasizes the absolute requirement to better understand the basic mechanisms of stem cell biology and cardiogenesis. We will also discuss about the unexpected “big bang” in the stem cell theory, “blasting” the therapeutic cells to their unchallenged ability to release paracrine factors such as extracellular membrane vesicles. Paradoxically, the natural evolution of the stem cell theory for cardiac regeneration may end with the development of cell-free strategies with multiple cellular targets including cardiomyocytes but also other infiltrating or resident cardiac cells. Varied sources of therapeutic cells and low repair ability of the failing heart contribute to mixed results in clinical trials. Consensus is still lacking concerning the appropriate type of therapeutic stem cells. A clear understanding of cardiac development and adult cardiogenesis might increase the efficiency of regenerative therapies. Delivery of stem cell-derived paracrine factor alone to the damaged heart may be sufficient to activate repair mechanisms.
Collapse
|
27
|
Takagi G, Miyamoto M, Fukushima Y, Yasutake M, Tara S, Takagi I, Seki N, Kumita S, Shimizu W. Imaging Angiogenesis Using 99mTc-Macroaggregated Albumin Scintigraphy in Patients with Peripheral Artery Disease. J Nucl Med 2015; 57:192-7. [PMID: 26541773 DOI: 10.2967/jnumed.115.160937] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 10/19/2015] [Indexed: 01/09/2023] Open
Abstract
UNLABELLED One problem of vascular angiogenesis therapy is the lack of reliable methods for evaluating blood flow in the microcirculation. We aimed to assess whether (99m)Tc-macroaggregated albumin perfusion scintigraphy ((99m)Tc-MAA) predicts quantitated blood flow after therapeutic angiogenesis in patients with peripheral artery disease. METHODS Forty-six patients with peripheral artery disease were treated with bone marrow mononuclear cell implantation (BMCI). Before and 4 wk after BMCI, blood flow was evaluated via transcutaneous oxygen tension (TcPO2), ankle-brachial index, intravenous (99m)Tc-tetrofosmin perfusion scintigraphy ((99m)Tc-TF), and intraaortic (99m)Tc-MAA. RESULTS Four weeks after BMCI, TcPO2 improved significantly (20.4 ± 14.4 to 36.0 ± 20.0 mm Hg, P < 0.01), but ankle-brachial index did not (0.65 ± 0.30 to 0.76 ± 0.24, P = 0.07). Improvement in (99m)Tc-TF count (0.60 ± 0.23 to 0.77 ± 0.29 count ratio/pixel, P < 0.01) and (99m)Tc-MAA count (5.21 ± 3.56 to 10.33 ± 7.18 count ratio/pixel, P = 0.02) was observed in the foot region but not the lower limb region, using both methods. When these data were normalized by subtracting the pixel count of the untreated side, the improvements in (99m)Tc-TF count (-0.04 ± 0.26 to 0.08 ± 0.32 count ratio/pixel, P = 0.04) and (99m)Tc-MAA count (1.49 ± 3.64 to 5.59 ± 4.84 count ratio/pixel, P = 0.03) in the foot remained significant. (99m)Tc-MAA indicated that the newly developed arteries were approximately 25 μm in diameter. CONCLUSION BMCI induced angiogenesis in the foot, which was detected using (99m)Tc-TF and (99m)Tc-MAA. (99m)Tc-MAA is a useful method to quantitate blood flow, estimate vascular size, and evaluate flow distribution after therapeutic angiogenesis.
Collapse
Affiliation(s)
- Gen Takagi
- Department of Cardiovascular Medicine, Nippon Medical School, Tokyo, Japan
| | - Masaaki Miyamoto
- Department of Cardiovascular Medicine, Nippon Medical School, Tokyo, Japan
| | | | - Masahiro Yasutake
- Department of Cardiovascular Medicine, Nippon Medical School, Tokyo, Japan
| | - Shuhei Tara
- Department of Cardiovascular Medicine, Nippon Medical School, Tokyo, Japan
| | - Ikuyo Takagi
- Department of Cardiovascular Medicine, Nippon Medical School, Tokyo, Japan
| | - Naoki Seki
- FUJIFILM RI Pharma Co., Ltd., Tokyo, Japan
| | | | - Wataru Shimizu
- Department of Cardiovascular Medicine, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
28
|
Gaffey AC, Chen MH, Venkataraman CM, Trubelja A, Rodell CB, Dinh PV, Hung G, MacArthur JW, Soopan RV, Burdick JA, Atluri P. Injectable shear-thinning hydrogels used to deliver endothelial progenitor cells, enhance cell engraftment, and improve ischemic myocardium. J Thorac Cardiovasc Surg 2015; 150:1268-76. [PMID: 26293548 PMCID: PMC4637242 DOI: 10.1016/j.jtcvs.2015.07.035] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 06/30/2015] [Accepted: 07/12/2015] [Indexed: 01/26/2023]
Abstract
OBJECTIVES The clinical translation of cell-based therapies for ischemic heart disease has been limited because of low cell retention (<1%) within, and poor targeting to, ischemic myocardium. To address these issues, we developed an injectable hyaluronic acid (HA) shear-thinning hydrogel (STG) and endothelial progenitor cell (EPC) construct (STG-EPC). The STG assembles as a result of interactions of adamantine- and β-cyclodextrin-modified HA. It is shear-thinning to permit delivery via a syringe, and self-heals upon injection within the ischemic myocardium. This directed therapy to the ischemic myocardial border zone enables direct cell delivery to address adverse remodeling after myocardial infarction. We hypothesize that this system will enhance vasculogenesis to improve myocardial stabilization in the context of a clinically translatable therapy. METHODS Endothelial progenitor cells (DiLDL(+) VEGFR2(+) CD34(+)) were harvested from adult male rats, cultured, and suspended in the STG. In vitro viability was quantified using a live-dead stain of EPCs. The STG-EPC constructs were injected at the border zone of ischemic rat myocardium after acute myocardial infarction (left anterior descending coronary artery ligation). The migration of the enhanced green fluorescent proteins from the construct to ischemic myocardium was analyzed using fluorescent microscopy. Vasculogenesis, myocardial remodeling, and hemodynamic function were analyzed in 4 groups: control (phosphate buffered saline injection); intramyocardial injection of EPCs alone; injection of the STG alone; and treatment with the STG-EPC construct. Hemodynamics and ventricular geometry were quantified using echocardiography and Doppler flow analysis. RESULTS Endothelial progenitor cells demonstrated viability within the STG. A marked increase in EPC engraftment was observed 1-week postinjection within the treated myocardium with gel delivery, compared with EPC injection alone (17.2 ± 0.8 cells per high power field (HPF) vs 3.5 cells ± 1.3 cells per HPF, P = .0002). A statistically significant increase in vasculogenesis was noted with the STG-EPC construct (15.3 ± 5.8 vessels per HPF), compared with the control (P < .0001), EPC (P < .0001), and STG (P < .0001) groups. Statistically significant improvements in ventricular function, scar fraction, and geometry were noted after STG-EPC treatment compared with the control. CONCLUSIONS A novel injectable shear-thinning HA hydrogel seeded with EPCs enhanced cell retention and vasculogenesis after delivery to ischemic myocardium. This therapy limited adverse myocardial remodeling while preserving contractility.
Collapse
Affiliation(s)
- Ann C Gaffey
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | - Minna H Chen
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa
| | - Chantel M Venkataraman
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | - Alen Trubelja
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | | | - Patrick V Dinh
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | - George Hung
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | - John W MacArthur
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | - Renganaden V Soopan
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pa
| | - Pavan Atluri
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pa.
| |
Collapse
|
29
|
Nemoto M, Koyama H, Nishiyama A, Shigematsu K, Miyata T, Watanabe T. Adequate Selection of a Therapeutic Site Enables Efficient Development of Collateral Vessels in Angiogenic Treatment With Bone Marrow Mononuclear Cells. J Am Heart Assoc 2015; 4:e002287. [PMID: 26370447 PMCID: PMC4599510 DOI: 10.1161/jaha.115.002287] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background Induction of angiogenic mechanisms to promote development of collateral vessels is considered promising for the treatment of peripheral arterial diseases. Collateral vessels generally develop from preexisting arteriolar connections, bypassing the diseased artery. We speculated that induction of angiogenic mechanisms should be directed to such arteriolar connections to achieve efficient collateral development. The aim of this study was to verify this hypothesis using autologous transplantation of bone marrow mononuclear cells in the rabbit model of chronic limb ischemia. Methods and Results The left femoral artery was excised to induce limb ischemia in male rabbits. In this model, arteriolar connections in the left coccygeofemoral muscle tend to develop into collateral vessels, although this transformation is insufficient to alleviate the limb ischemia. In contrast, arteriolar connections in the closely located adductor muscle do not readily develop into collateral vessels. At 21 days after ischemia initiation, a sufficient number of automononuclear cells were selectively injected in the left coccygeofemoral muscle (coccygeo group) or left adductor muscle (adductor group). Evaluation of calf blood pressure ratios, blood flow in the left internal iliac artery, and angiographic scores at day 28 after injection revealed that collateral development and improvement of limb ischemia were significantly more efficient in the coccygeo group than in the adductor group. Morphometric analysis of the coccygeofemoral muscle at day 14 showed similar results. Conclusions Specific delivery of mononuclear cells to the coccygeofemoral but not the adductor muscle effectively improves collateral circulation in the rabbit model of limb ischemia and suggests that adequate site selection can facilitate therapeutic angiogenesis.
Collapse
Affiliation(s)
- Masaru Nemoto
- Department of Vascular Surgery, Graduate School of Medicine, University of Tokyo, Japan (M.N., H.K., A.N., K.S., T.W.)
| | - Hiroyuki Koyama
- Department of Vascular Surgery, Graduate School of Medicine, University of Tokyo, Japan (M.N., H.K., A.N., K.S., T.W.) Translational Research Center, The University of Tokyo Hospital, Tokyo, Japan (H.K.) Department of Vascular Surgery, Saitama Medical Center, Saitama Medical University, Saitama, Japan (H.K.)
| | - Ayako Nishiyama
- Department of Vascular Surgery, Graduate School of Medicine, University of Tokyo, Japan (M.N., H.K., A.N., K.S., T.W.)
| | - Kunihiro Shigematsu
- Department of Vascular Surgery, Graduate School of Medicine, University of Tokyo, Japan (M.N., H.K., A.N., K.S., T.W.)
| | - Tetsuro Miyata
- Vascular Center, Sanno Hospital and Sanno Medical Center, Tokyo, Japan (T.M.)
| | - Toshiaki Watanabe
- Department of Vascular Surgery, Graduate School of Medicine, University of Tokyo, Japan (M.N., H.K., A.N., K.S., T.W.)
| |
Collapse
|
30
|
Granton J, Langleben D, Kutryk MB, Camack N, Galipeau J, Courtman DW, Stewart DJ. Endothelial NO-Synthase Gene-Enhanced Progenitor Cell Therapy for Pulmonary Arterial Hypertension: The PHACeT Trial. Circ Res 2015. [PMID: 26195220 DOI: 10.1161/circresaha.114.305951] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
RATIONALE Pulmonary arterial hypertension (PAH) remains a progressive and eventually lethal disease characterized by increased pulmonary vascular resistance because of loss of functional lung microvasculature, primarily at the distal (intracinar) arteriolar level. Cell-based therapies offer the potential to repair and regenerate the lung microcirculation and have shown promise in preclinical evaluation in experimental models of PAH. OBJECTIVE The Pulmonary Hypertension and Angiogenic Cell Therapy (PHACeT) trial was a phase 1, dose-escalating clinical study of the tolerability of culture-derived endothelial progenitor cells, transiently transfected with endothelial nitric oxide synthase, in patients with PAH refractory to PAH-specific therapies. METHODS AND RESULTS Seven to 50 million endothelial nitric oxide synthase-transfected endothelial progenitor cells, divided into 3 doses on consecutive days, were delivered into the right atrium via a multiport pulmonary artery catheter during continuous hemodynamic monitoring in an intensive care unit setting. Seven patients (5 women) received treatment from December 2006 to March 2010. Cell infusion was well tolerated, with no evidence of short-term hemodynamic deterioration; rather, there was a trend toward improvement in total pulmonary resistance during the 3-day delivery period. However, there was 1 serious adverse event (death) which occurred immediately after discharge in a patient with severe, end stage disease. Although there were no sustained hemodynamic improvements at 3 months, 6-minute walk distance was significantly increased at 1, 3, and 6 months. CONCLUSION Delivery of endothelial progenitor cells overexpressing endothelial nitric oxide synthase was tolerated hemodynamically in patients with PAH. Furthermore, there was evidence of short-term hemodynamic improvement, associated with long-term benefits in functional and quality of life assessments. However, future studies are needed to further establish the efficacy of this therapy. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT00469027.
Collapse
Affiliation(s)
- John Granton
- From the Division of Respirology, Pulmonary Hypertension Program, University Health Network (J. Granton) and Division of Cardiology, Keenan Research Center for Biomedical Science at the Li Ka Shing Knowledge Institute, St. Michael's Hospital (M.B.K.), Department of Medicine, University of Toronto, Toronto, Ontario, Canada (J. Granton, M.B.K.); Center for Pulmonary Vascular Disease, Division of Cardiology, and Lady Davis Research Institute, Jewish General Hospital, Department of Medicine, McGill University, Montreal, Quebec, Canada (D.L.); Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (N.C., D.W.C., D.J.S.); Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (D.W.C., D.J.S.); Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, GA (J. Galipeau); and Northern Therapeutics, Montreal, Quebec, Canada (D.W.C., D.J.S.)
| | - David Langleben
- From the Division of Respirology, Pulmonary Hypertension Program, University Health Network (J. Granton) and Division of Cardiology, Keenan Research Center for Biomedical Science at the Li Ka Shing Knowledge Institute, St. Michael's Hospital (M.B.K.), Department of Medicine, University of Toronto, Toronto, Ontario, Canada (J. Granton, M.B.K.); Center for Pulmonary Vascular Disease, Division of Cardiology, and Lady Davis Research Institute, Jewish General Hospital, Department of Medicine, McGill University, Montreal, Quebec, Canada (D.L.); Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (N.C., D.W.C., D.J.S.); Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (D.W.C., D.J.S.); Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, GA (J. Galipeau); and Northern Therapeutics, Montreal, Quebec, Canada (D.W.C., D.J.S.)
| | - Michael B Kutryk
- From the Division of Respirology, Pulmonary Hypertension Program, University Health Network (J. Granton) and Division of Cardiology, Keenan Research Center for Biomedical Science at the Li Ka Shing Knowledge Institute, St. Michael's Hospital (M.B.K.), Department of Medicine, University of Toronto, Toronto, Ontario, Canada (J. Granton, M.B.K.); Center for Pulmonary Vascular Disease, Division of Cardiology, and Lady Davis Research Institute, Jewish General Hospital, Department of Medicine, McGill University, Montreal, Quebec, Canada (D.L.); Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (N.C., D.W.C., D.J.S.); Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (D.W.C., D.J.S.); Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, GA (J. Galipeau); and Northern Therapeutics, Montreal, Quebec, Canada (D.W.C., D.J.S.)
| | - Nancy Camack
- From the Division of Respirology, Pulmonary Hypertension Program, University Health Network (J. Granton) and Division of Cardiology, Keenan Research Center for Biomedical Science at the Li Ka Shing Knowledge Institute, St. Michael's Hospital (M.B.K.), Department of Medicine, University of Toronto, Toronto, Ontario, Canada (J. Granton, M.B.K.); Center for Pulmonary Vascular Disease, Division of Cardiology, and Lady Davis Research Institute, Jewish General Hospital, Department of Medicine, McGill University, Montreal, Quebec, Canada (D.L.); Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (N.C., D.W.C., D.J.S.); Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (D.W.C., D.J.S.); Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, GA (J. Galipeau); and Northern Therapeutics, Montreal, Quebec, Canada (D.W.C., D.J.S.)
| | - Jacques Galipeau
- From the Division of Respirology, Pulmonary Hypertension Program, University Health Network (J. Granton) and Division of Cardiology, Keenan Research Center for Biomedical Science at the Li Ka Shing Knowledge Institute, St. Michael's Hospital (M.B.K.), Department of Medicine, University of Toronto, Toronto, Ontario, Canada (J. Granton, M.B.K.); Center for Pulmonary Vascular Disease, Division of Cardiology, and Lady Davis Research Institute, Jewish General Hospital, Department of Medicine, McGill University, Montreal, Quebec, Canada (D.L.); Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (N.C., D.W.C., D.J.S.); Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (D.W.C., D.J.S.); Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, GA (J. Galipeau); and Northern Therapeutics, Montreal, Quebec, Canada (D.W.C., D.J.S.)
| | - David W Courtman
- From the Division of Respirology, Pulmonary Hypertension Program, University Health Network (J. Granton) and Division of Cardiology, Keenan Research Center for Biomedical Science at the Li Ka Shing Knowledge Institute, St. Michael's Hospital (M.B.K.), Department of Medicine, University of Toronto, Toronto, Ontario, Canada (J. Granton, M.B.K.); Center for Pulmonary Vascular Disease, Division of Cardiology, and Lady Davis Research Institute, Jewish General Hospital, Department of Medicine, McGill University, Montreal, Quebec, Canada (D.L.); Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (N.C., D.W.C., D.J.S.); Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (D.W.C., D.J.S.); Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, GA (J. Galipeau); and Northern Therapeutics, Montreal, Quebec, Canada (D.W.C., D.J.S.)
| | - Duncan J Stewart
- From the Division of Respirology, Pulmonary Hypertension Program, University Health Network (J. Granton) and Division of Cardiology, Keenan Research Center for Biomedical Science at the Li Ka Shing Knowledge Institute, St. Michael's Hospital (M.B.K.), Department of Medicine, University of Toronto, Toronto, Ontario, Canada (J. Granton, M.B.K.); Center for Pulmonary Vascular Disease, Division of Cardiology, and Lady Davis Research Institute, Jewish General Hospital, Department of Medicine, McGill University, Montreal, Quebec, Canada (D.L.); Ottawa Hospital Research Institute, Ottawa, Ontario, Canada (N.C., D.W.C., D.J.S.); Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada (D.W.C., D.J.S.); Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, GA (J. Galipeau); and Northern Therapeutics, Montreal, Quebec, Canada (D.W.C., D.J.S.).
| |
Collapse
|
31
|
Stempien-Otero A, Helterline D, Plummer T, Farris S, Prouse A, Polissar N, Stanford D, Mokadam NA. Mechanisms of bone marrow-derived cell therapy in ischemic cardiomyopathy with left ventricular assist device bridge to transplant. J Am Coll Cardiol 2015; 65:1424-34. [PMID: 25857908 DOI: 10.1016/j.jacc.2015.01.042] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 12/22/2014] [Accepted: 01/27/2015] [Indexed: 11/15/2022]
Abstract
BACKGROUND Clinical trials report improvements in function and perfusion with direct injection of bone marrow cells into the hearts of patients with ischemic cardiomyopathy. Preclinical data suggest these cells improve vascular density, which would be expected to decrease fibrosis and inflammation. OBJECTIVES The goal of this study was to test the hypothesis that bone marrow stem cells (CD34+) will improve histological measurements of vascularity, fibrosis, and inflammation in human subjects undergoing left ventricular assist device (LVAD) placement as a bridge to cardiac transplantation. METHODS Subjects with ischemic cardiomyopathy who were scheduled for placement of an LVAD as a bridge to transplantation underwent bone marrow aspiration the day before surgery; the bone marrow was processed into cell fractions (bone marrow mononuclear cells, CD34+, and CD34-). At LVAD implantation, all fractions and a saline control were injected epicardially into predetermined areas and each injection site marked. At the time of transplantation, injected areas were collected. Data were analyzed by paired Student t test comparing the effect of cell fractions injected within each subject. RESULTS Six subjects completed the study. There were no statistically significant differences in complications with the procedure versus control subjects. Histological analysis indicated that myocardium injected with CD34+ cells had decreased density of endothelial cells compared to saline-injected myocardium. There were no significant differences in fibrosis or inflammation between groups; however, density of activated fibroblasts was decreased in both CD34+ and CD34- injected areas. CONCLUSIONS Tissue analysis does not support the hypothesis that bone marrow-derived CD34+ cells promote increased vascular tissue in humans with ischemic cardiomyopathy via direct injection.
Collapse
Affiliation(s)
- April Stempien-Otero
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington.
| | - Deri Helterline
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Tabitha Plummer
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Stephen Farris
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Andrew Prouse
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Nayak Polissar
- The Mountain-Whisper-Light Statistics, Seattle, Washington
| | - Derek Stanford
- The Mountain-Whisper-Light Statistics, Seattle, Washington
| | - Nahush A Mokadam
- Department of Surgery, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
32
|
Effect of Morphofunctional Properties of Mobilized Progenitor Cells of Patients with Chronic Heart Failure on the Efficiency of Autologous Intramyocardial Cell Transplantation. Bull Exp Biol Med 2014; 157:695-700. [DOI: 10.1007/s10517-014-2645-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Indexed: 10/24/2022]
|
33
|
Atluri P, Miller JS, Emery RJ, Hung G, Trubelja A, Cohen JE, Lloyd K, Han J, Gaffey AC, MacArthur JW, Chen CS, Woo YJ. Tissue-engineered, hydrogel-based endothelial progenitor cell therapy robustly revascularizes ischemic myocardium and preserves ventricular function. J Thorac Cardiovasc Surg 2014; 148:1090-7; discussion 1097-8. [PMID: 25129603 DOI: 10.1016/j.jtcvs.2014.06.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 06/10/2014] [Accepted: 06/11/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVES Cell-based angiogenic therapy for ischemic heart failure has had limited clinical impact, likely related to low cell retention (<1%) and dispersion. We developed a novel, tissue-engineered, hydrogel-based cell-delivery strategy to overcome these limitations and provide prolonged regional retention of myocardial endothelial progenitor cells at high cell dosage. METHODS Endothelial progenitor cells were isolated from Wistar rats and encapsulated in fibrin gels. In vitro viability was quantified using a fluorescent live-dead stain of transgenic enhanced green fluorescent protein(+) endothelial progenitor cells. Endothelial progenitor cell-laden constructs were implanted onto ischemic rat myocardium in a model of acute myocardial infarction (left anterior descending ligation) for 4 weeks. Intramyocardial cell injection (2 × 10(6) endothelial progenitor cells), empty fibrin, and isolated left anterior descending ligation groups served as controls. Hemodynamics were quantified using echocardiography, Doppler flow analysis, and intraventricular pressure-volume analysis. Vasculogenesis and ventricular geometry were quantified. Endothelial progenitor cell migration was analyzed by using endothelial progenitor cells from transgenic enhanced green fluorescent protein(+) rodents. RESULTS Endothelial progenitor cells demonstrated an overall 88.7% viability for all matrix and cell conditions investigated after 48 hours. Histologic assessment of 1-week implants demonstrated significant migration of transgenic enhanced green fluorescent protein(+) endothelial progenitor cells from the fibrin matrix to the infarcted myocardium compared with intramyocardial cell injection (28 ± 12.3 cells/high power field vs 2.4 ± 2.1 cells/high power field, P = .0001). We also observed a marked increase in vasculogenesis at the implant site. Significant improvements in ventricular hemodynamics and geometry were present after endothelial progenitor cell-hydrogel therapy compared with control. CONCLUSIONS We present a tissue-engineered, hydrogel-based endothelial progenitor cell-mediated therapy to enhance cell delivery, cell retention, vasculogenesis, and preservation of myocardial structure and function.
Collapse
Affiliation(s)
- Pavan Atluri
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | | | - Robert J Emery
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - George Hung
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Alen Trubelja
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Jeffrey E Cohen
- Department of Cardiothoracic Surgery, Stanford University, Stanford, Calif
| | - Kelsey Lloyd
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Jason Han
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Ann C Gaffey
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - John W MacArthur
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | | | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, Calif.
| |
Collapse
|
34
|
Bone marrow-derived mesenchymal stromal cells improve vascular regeneration and reduce leukocyte-endothelium activation in critical ischemic murine skin in a dose-dependent manner. Cytotherapy 2014; 16:1345-60. [PMID: 24972742 DOI: 10.1016/j.jcyt.2014.05.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 04/26/2014] [Accepted: 05/08/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND AIMS Stem cells participate in vascular regeneration following critical ischemia. However, their angiogenic and remodeling properties, as well as their role in ischemia-related endothelial leukocyte activation, need to be further elucidated. Herein, we investigated the effect of bone marrow-derived mesenchymal stromal cells (BM-MSCs) in a critically ischemic murine skin flap model. METHODS Groups received either 1 × 10(5), 5 × 10(5), or 1 × 10(6) BM-MSCs or cell-free conditioned medium (CM). Controls received sodium chloride. Intravital fluorescence microscopy was performed for morphological and quantitative assessment of micro-hemodynamic parameters over 12 days. RESULTS Tortuosity and diameter of conduit-arterioles were pronounced in the MSC groups (P < 0.01), whereas vasodilation was shifted to the end arteriolar level in the CM group (P < 0.01). These effects were accompanied by angiopoietin-2 expression. Functional capillary density and red blood cell velocity were enhanced in all treatment groups (P < 0.01). Although a significant reduction of rolling and sticking leukocytes was observed in the MSC groups with a reduction of diameter in postcapillary venules (P < 0.01), animals receiving CM exhibited a leukocyte-endothelium interaction similar to controls. This correlated with leukocyte common antigen expression in tissue sections (P < 0.01) and p38 mitogen-activated protein kinase expression from tissue samples. Cytokine analysis from BM-MSC culture medium revealed a 50% reduction of pro-inflammatory cytokines (interleukin [IL]-1β, IL-6, IL-12, tumor necrosis factor-α, interferon-γ) and chemokines (keratinocyte chemoattractant, granulocyte colony-stimulating factor) under hypoxic conditions. DISCUSSION We demonstrated positive effects of BM-MSCs on vascular regeneration and modulation of endothelial leukocyte adhesion in critical ischemic skin. The improvements after MSC application were dose-dependent and superior to the use of CM alone.
Collapse
|
35
|
Kinney MA, Hookway TA, Wang Y, McDevitt TC. Engineering three-dimensional stem cell morphogenesis for the development of tissue models and scalable regenerative therapeutics. Ann Biomed Eng 2014; 42:352-67. [PMID: 24297495 PMCID: PMC3939035 DOI: 10.1007/s10439-013-0953-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 11/21/2013] [Indexed: 12/11/2022]
Abstract
The physiochemical stem cell microenvironment regulates the delicate balance between self-renewal and differentiation. The three-dimensional assembly of stem cells facilitates cellular interactions that promote morphogenesis, analogous to the multicellular, heterotypic tissue organization that accompanies embryogenesis. Therefore, expansion and differentiation of stem cells as multicellular aggregates provides a controlled platform for studying the biological and engineering principles underlying spatiotemporal morphogenesis and tissue patterning. Moreover, three-dimensional stem cell cultures are amenable to translational screening applications and therapies, which underscores the broad utility of scalable suspension cultures across laboratory and clinical scales. In this review, we discuss stem cell morphogenesis in the context of fundamental biophysical principles, including the three-dimensional modulation of adhesions, mechanics, and molecular transport and highlight the opportunities to employ stem cell spheroids for tissue modeling, bioprocessing, and regenerative therapies.
Collapse
Affiliation(s)
- Melissa A. Kinney
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology/Emory University, Atlanta, GA, USA
| | - Tracy A. Hookway
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology/Emory University, Atlanta, GA, USA
| | - Yun Wang
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology/Emory University, Atlanta, GA, USA
| | - Todd C. McDevitt
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology/Emory University, Atlanta, GA, USA
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
36
|
Intramyocardial autologous bone marrow cell transplantation for ischemic heart disease: a systematic review and meta-analysis of randomized controlled trials. Atherosclerosis 2014; 233:485-492. [PMID: 24530783 DOI: 10.1016/j.atherosclerosis.2014.01.027] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 12/20/2013] [Accepted: 01/13/2014] [Indexed: 12/16/2022]
Abstract
OBJECTIVE This study was undertaken to evaluate the efficacy of intramyocardial bone marrow cell (BMC) transplant therapy for ischemic heart disease (IHD). METHODS The PubMed, Embase, and Cochrane Library databases through October 2013 were searched for randomized clinical trials (RCTs) of intramyocardial BMCs to treat IHD. The primary endpoint was change in left ventricular ejection fraction (LVEF). Secondary endpoints were changes in left ventricular end-systolic volume (LVESV) and left ventricular end-diastolic volume (LVEDV). Weighted mean differences for the changes were estimated with a random-effects model. RESULTS Eleven RCTs with 492 participants were included. Intramyocardial BMC transplantation increased LVEF (4.91%; 95% confidence interval [CI] 2.84%-6.99%; P<0.00001), reduced LVESV (10.66 mL; 95% CI, -18.92 mL to -2.41 mL; P=0.01), and showed a trend toward decreased LVEDV (-7.82 mL; 95% CI, -16.36 mL-0.71 mL; P=0.07). Patients suitable for revascularization with coronary artery bypass grafting had greater improvement in LVEF (7.60%; 95% CI, 4.74%-10.46%, P<0.00001) than those unsuitable for revascularization (3.76%; 95% CI, 2.20%-5.32%; P<0.00001). LVEDV reduction was also more significant in revascularizable IHD (-16.51 mL; 95% CI, -22.05 mL to -10.07 mL; P<0.00001) than non-revascularizable IHD (-0.89 mL; 95% CI, -8.44 mL-6.66 mL; P=0.82). CONCLUSION Intramyocardial BMC injection contributes to improvement in left ventricular dysfunction and reduction in left ventricular volume. Patients with revascularizable IHD may benefit more from this therapy.
Collapse
|
37
|
Liao SY, Tse HF. Multipotent (adult) and pluripotent stem cells for heart regeneration: what are the pros and cons? Stem Cell Res Ther 2013; 4:151. [PMID: 24476362 PMCID: PMC4056686 DOI: 10.1186/scrt381] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Heart failure after myocardial infarction is the leading cause of mortality and morbidity worldwide. Existing medical and interventional therapies can only reduce the loss of cardiomyocytes during myocardial infarction but are unable to replenish the permanent loss of cardiomyocytes after the insult, which contributes to progressive pathological left ventricular remodeling and progressive heart failure. As a result, cell-based therapies using multipotent (adult) stem cells and pluripotent stem cells (embryonic stem cells or induced pluripotent stem cells) have been explored as potential therapeutic approaches to restore cardiac function in heart failure. Nevertheless, the optimal cell type with the best therapeutic efficacy and safety for heart regeneration is still unknown. In this review, the potential pros and cons of different types of multipotent (adult) stem cells and pluripotent stem cells that have been investigated in preclinical and clinical studies are reviewed, and the future perspective of stem cell-based therapy for heart regeneration is discussed.
Collapse
|
38
|
Silvestre JS, Smadja DM, Lévy BI. Postischemic revascularization: from cellular and molecular mechanisms to clinical applications. Physiol Rev 2013; 93:1743-802. [PMID: 24137021 DOI: 10.1152/physrev.00006.2013] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
After the onset of ischemia, cardiac or skeletal muscle undergoes a continuum of molecular, cellular, and extracellular responses that determine the function and the remodeling of the ischemic tissue. Hypoxia-related pathways, immunoinflammatory balance, circulating or local vascular progenitor cells, as well as changes in hemodynamical forces within vascular wall trigger all the processes regulating vascular homeostasis, including vasculogenesis, angiogenesis, arteriogenesis, and collateral growth, which act in concert to establish a functional vascular network in ischemic zones. In patients with ischemic diseases, most of the cellular (mainly those involving bone marrow-derived cells and local stem/progenitor cells) and molecular mechanisms involved in the activation of vessel growth and vascular remodeling are markedly impaired by the deleterious microenvironment characterized by fibrosis, inflammation, hypoperfusion, and inhibition of endogenous angiogenic and regenerative programs. Furthermore, cardiovascular risk factors, including diabetes, hypercholesterolemia, hypertension, diabetes, and aging, constitute a deleterious macroenvironment that participates to the abrogation of postischemic revascularization and tissue regeneration observed in these patient populations. Thus stimulation of vessel growth and/or remodeling has emerged as a new therapeutic option in patients with ischemic diseases. Many strategies of therapeutic revascularization, based on the administration of growth factors or stem/progenitor cells from diverse sources, have been proposed and are currently tested in patients with peripheral arterial disease or cardiac diseases. This review provides an overview from our current knowledge regarding molecular and cellular mechanisms involved in postischemic revascularization, as well as advances in the clinical application of such strategies of therapeutic revascularization.
Collapse
|
39
|
Atluri P, Trubelja A, Fairman AS, Hsiao P, MacArthur JW, Cohen JE, Shudo Y, Frederick JR, Woo YJ. Normalization of postinfarct biomechanics using a novel tissue-engineered angiogenic construct. Circulation 2013; 128:S95-104. [PMID: 24030426 DOI: 10.1161/circulationaha.112.000368] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Cell-mediated angiogenic therapy for ischemic heart disease has had disappointing results. The lack of clinical translatability may be secondary to cell death and systemic dispersion with cell injection. We propose a novel tissue-engineered therapy, whereby extracellular matrix scaffold seeded with endothelial progenitor cells (EPCs) can overcome these limitations using an environment in which the cells can thrive, enabling an insult-free myocardial cell delivery to normalize myocardial biomechanics. METHODS AND RESULTS EPCs were isolated from the long bones of Wistar rat bone marrow. The cells were cultured for 7 days in media or seeded at a density of 5 × 10(6) cells/cm(2) on a collagen/vitronectin matrix. Seeded EPCs underwent ex vivo modification with stromal cell-derived factor-1α (100 ng/mL) to potentiate angiogenic properties and enhance paracrine qualities before construct formation. Scanning electron microscopy and confocal imaging confirmed EPC-matrix adhesion. In vitro vasculogenic potential was assessed by quantifying EPC cell migration and vascular differentiation. There was a marked increase in vasculogenesis in vitro as measured by angiogenesis assay (8 versus 0 vessels/hpf; P=0.004). The construct was then implanted onto ischemic myocardium in a rat model of acute myocardial infarction. Confocal microscopy demonstrated a significant migration of EPCs from the construct to the myocardium, suggesting a direct angiogenic effect. Myocardial biomechanical properties were uniaxially quantified by elastic modulus at 5% to 20% strain. Myocardial elasticity normalized after implant of our tissue-engineered construct (239 kPa versus normal=193, P=0.1; versus infarct=304 kPa, P=0.01). CONCLUSIONS We demonstrate restoration and normalization of post-myocardial infarction ventricular biomechanics after therapy with an angiogenic tissue-engineered EPC construct.
Collapse
Affiliation(s)
- Pavan Atluri
- Division of Cardiovascular Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kito T, Shibata R, Ishii M, Suzuki H, Himeno T, Kataoka Y, Yamamura Y, Yamamoto T, Nishio N, Ito S, Numaguchi Y, Tanigawa T, Yamashita JK, Ouchi N, Honda H, Isobe K, Murohara T. iPS cell sheets created by a novel magnetite tissue engineering method for reparative angiogenesis. Sci Rep 2013; 3:1418. [PMID: 23475393 PMCID: PMC3593218 DOI: 10.1038/srep01418] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 02/25/2013] [Indexed: 11/16/2022] Open
Abstract
Angiogenic cell therapy represents a novel strategy for ischemic diseases, but some patients show poor responses. We investigated the therapeutic potential of an induced pluripotent stem (iPS) cell sheet created by a novel magnetite tissue engineering technology (Mag-TE) for reparative angiogenesis. Mouse iPS cell-derived Flk-1+ cells were incubated with magnetic nanoparticle-containing liposomes (MCLs). MCL-labeled Flk-1+ cells were mixed with diluted extracellular matrix (ECM) precursor and a magnet was placed on the reverse side. Magnetized Flk-1+ cells formed multi-layered cell sheets according to magnetic force. Implantation of the Flk-1+ cell sheet accelerated revascularization of ischemic hindlimbs relative to the contralateral limbs in nude mice as measured by laser Doppler blood flow and capillary density analyses. The Flk-1+ cell sheet also increased the expressions of VEGF and bFGF in ischemic tissue. iPS cell-derived Flk-1+ cell sheets created by this novel Mag-TE method represent a promising new modality for therapeutic angiogenesis.
Collapse
Affiliation(s)
- Tetsutaro Kito
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Kandala J, Upadhyay GA, Pokushalov E, Wu S, Drachman DE, Singh JP. Meta-analysis of stem cell therapy in chronic ischemic cardiomyopathy. Am J Cardiol 2013; 112:217-25. [PMID: 23623290 DOI: 10.1016/j.amjcard.2013.03.021] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 03/06/2013] [Accepted: 03/06/2013] [Indexed: 12/18/2022]
Abstract
Studies investigating bone marrow stem cell therapy (BMSCT) in patients with chronic ischemic cardiomyopathy have yielded mixed results. A meta-analysis of randomized controlled trials of BMSCT in patients with chronic ischemic cardiomyopathy was undertaken to assess its efficacy and the best route of administration. The MEDLINE, Embase, Cumulative Index to Nursing & Allied Health Literature, and Cochrane Library databases were searched through April 2012 for randomized controlled trials that investigated the impact of BMSCT and its route of administration on left ventricular (LV) function in patients with chronic ischemic cardiomyopathy and systolic dysfunction. Of the 226 reports identified, 10 randomized controlled trials including 519 patients (average LV ejection fraction [LVEF] at baseline 32 ± 7%) were included in the analysis. On the basis of a random-effects model, BMSCT improved the LVEF at 6 months by 4.48% (95% confidence interval [CI] 2.43% to 6.53%, p = 0.0001). A greater improvement in the LVEF was seen with intramyocardial injection compared with intracoronary infusion (5.13% [95% CI 3.17% to 7.10%], p <0.00001, vs 2.32% [95% CI -2.06% to 6.70%], p = 0.30). Overall, there were reductions in LV end-systolic volume of -20.64 ml (95% CI -33.21 to -8.07, p = 0.001) and LV end-diastolic volume of -16.71 ml (95% CI -31.36 to -2.06, p = 0.03). In conclusion, stem cell therapy may improve LVEF and favorably remodel the heart in patients with chronic ischemic cardiomyopathy. On the basis of current limited data, intramyocardial injection may be superior to intracoronary infusion in patients with LV systolic dysfunction.
Collapse
|
42
|
Lai WH, Ho JCY, Chan YC, Ng JHL, Au KW, Wong LY, Siu CW, Tse HF. Attenuation of hind-limb ischemia in mice with endothelial-like cells derived from different sources of human stem cells. PLoS One 2013; 8:e57876. [PMID: 23472116 PMCID: PMC3589485 DOI: 10.1371/journal.pone.0057876] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 01/29/2013] [Indexed: 01/23/2023] Open
Abstract
Functional endothelial-like cells (EC) have been successfully derived from different cell sources and potentially used for treatment of cardiovascular diseases; however, their relative therapeutic efficacy remains unclear. We differentiated functional EC from human bone marrow mononuclear cells (BM-EC), human embryonic stem cells (hESC-EC) and human induced pluripotent stem cells (hiPSC-EC), and compared their in-vitro tube formation, migration and cytokine expression profiles, and in-vivo capacity to attenuate hind-limb ischemia in mice. Successful differentiation of BM-EC was only achieved in 1/6 patient with severe coronary artery disease. Nevertheless, BM-EC, hESC-EC and hiPSC-EC exhibited typical cobblestone morphology, had the ability of uptaking DiI-labeled acetylated low-density-lipoprotein, and binding of Ulex europaeus lectin. In-vitro functional assay demonstrated that hiPSC-EC and hESC-EC had similar capacity for tube formation and migration as human umbilical cord endothelial cells (HUVEC) and BM-EC (P>0.05). While increased expression of major angiogenic factors including epidermal growth factor, hepatocyte growth factor, vascular endothelial growth factor, placental growth factor and stromal derived factor-1 were observed in all EC cultures during hypoxia compared with normoxia (P<0.05), the magnitudes of cytokine up-regulation upon hypoxic were more dramatic in hiPSC-EC and hESC-EC (P<0.05). Compared with medium, transplanting BM-EC (n = 6), HUVEC (n = 6), hESC-EC (n = 8) or hiPSC-EC (n = 8) significantly attenuated severe hind-limb ischemia in mice via enhancement of neovascularization. In conclusion, functional EC can be generated from hECS and hiPSC with similar therapeutic efficacy for attenuation of severe hind-limb ischemia. Differentiation of functional BM-EC was more difficult to achieve in patients with cardiovascular diseases, and hESC-EC or iPSC-EC are readily available as "off-the-shelf" format for the treatment of tissue ischemia.
Collapse
Affiliation(s)
- Wing-Hon Lai
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, HKSAR, China
| | - Jenny C. Y. Ho
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, HKSAR, China
- Research Center of Heart, Brain, Hormone and Healthy Aging, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong, HKSAR, China
| | - Yau-Chi Chan
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, HKSAR, China
| | - Joyce H. L. Ng
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, HKSAR, China
| | - Ka-Wing Au
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, HKSAR, China
| | - Lai-Yung Wong
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, HKSAR, China
| | - Chung-Wah Siu
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, HKSAR, China
- Research Center of Heart, Brain, Hormone and Healthy Aging, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong, HKSAR, China
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, Queen Mary Hospital, the University of Hong Kong, Hong Kong, HKSAR, China
- Research Center of Heart, Brain, Hormone and Healthy Aging, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong, HKSAR, China
| |
Collapse
|
43
|
Giordano C, Kuraitis D, Beanlands RSB, Suuronen EJ, Ruel M. Cell-based vasculogenic studies in preclinical models of chronic myocardial ischaemia and hibernation. Expert Opin Biol Ther 2012; 13:411-28. [PMID: 23256710 DOI: 10.1517/14712598.2013.748739] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Coronary artery disease commonly leads to myocardial ischaemia and hibernation. Relevant preclinical models of these conditions are essential to evaluate new therapeutic options such as cell-based vasculogenic therapies. AREAS COVERED In this article, the authors first review basic concepts of myocardial ischaemia/hibernation and relevant techniques to assess myocardial viability. Then, preclinical models of chronic myocardial ischaemia and hibernation, induced by devices such as ameroid constrictors, Delrin stenosis, hydraulic occluders, and coils/stents are described. Lastly, the authors discuss cell-based vasculogenic therapy, and summarise studies conducted in large animal models of chronic myocardial ischaemia and hibernation. EXPERT OPINION Approximately one-third of patients with viable myocardium do not undergo revascularisation; however, this population is at high risk for cardiac events and would surely benefit from effective cell-based therapy. Because of the modest benefits in clinical studies, preclinical models accurately representing clinical myocardial ischemia/hibernation are necessary to better understand and appropriately direct regenerative therapy research.
Collapse
Affiliation(s)
- Céline Giordano
- University of Ottawa Heart Institute, Division of Cardiac Surgery, 40 Ruskin Street, Suite 3403, Ottawa, Ontario, K1Y 4W7, Canada
| | | | | | | | | |
Collapse
|
44
|
Brenes RA, Bear M, Jadlowiec C, Goodwin M, Hashim P, Protack CD, Ziegler KR, Li X, Model LS, Lv W, Collins MJ, Dardik A. Cell-based interventions for therapeutic angiogenesis: review of potential cell sources. Vascular 2012; 20:360-8. [PMID: 23086985 DOI: 10.1258/vasc.2011.201205] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Alternative therapies are currently being developed to treat patients with chronic limb ischemia who are unable to be revascularized in order to avoid amputation. Cell-based therapy using mononuclear cells is gaining attention as many clinical trials are currently underway. We review cell differentiation along with the different potential cell sources for use in therapeutic angiogenesis.
Collapse
Affiliation(s)
- Robert A Brenes
- Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT 06520-8089, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Liu Y, Lai WH, Liao SY, Siu CW, Yang YZ, Tse HF. Lack of cardiac nerve sprouting after intramyocardial transplantation of bone marrow-derived stem cells in a swine model of chronic ischemic myocardium. J Cardiovasc Transl Res 2012; 5:359-64. [PMID: 22302631 PMCID: PMC3349852 DOI: 10.1007/s12265-012-9350-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2011] [Accepted: 01/21/2012] [Indexed: 02/02/2023]
Abstract
Previous experimental studies suggested that mesenchymal stem cell transplantation causes cardiac nerve sprouting; however, whether bone marrow (BM)-derived mononuclear cells (MNC) and endothelial progenitor cells (EPC) can also lead to cardiac nerve sprouting and alter gap junction expression remains unclear. We investigated the effect of electroanatomical mapping-guided direct intramyocardial transplantation of BM-MNC (n = 8) and CD31+EPC (n = 8) compared with saline control (n = 8) on cardiac nerve sprouting and gap junction expression in a swine model of chronic ischemic myocardium. At 12 weeks after transplantation, the distribution and density of cardiac nerve sprouting were determined by staining of tyrosine hydroxylase (TH) and growth associated protein 43(GAP-43) and expression of connexin 43 in the targeted ischemic and remote normal myocardium. After 12 weeks, no animal developed sudden death after the transplantation. There were no significant differences in the number of cells with positive staining of TH and GAP-43 in the ischemic and normal myocardium between three groups. Furthermore, expression of connexin 43 was also similar in the ischemic and normal myocardia in each group of animals (P > 0.05). The results of this study demonstrated that intramyocardial BM-derived MNC or EPC transplantation in a large animal model of chronic myocardial ischemia was not associated with increased cardiac nerve sprouting over the ischemic myocardium.
Collapse
Affiliation(s)
- Yuan Liu
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, HKSAR, China
| | | | | | | | | | | |
Collapse
|
46
|
Chimenti I, Forte E, Angelini F, Messina E, Giacomello A. Biochemistry and biology: heart-to-heart to investigate cardiac progenitor cells. Biochim Biophys Acta Gen Subj 2012; 1830:2459-69. [PMID: 22921810 DOI: 10.1016/j.bbagen.2012.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 07/10/2012] [Accepted: 08/07/2012] [Indexed: 12/12/2022]
Abstract
BACKGROUND Cardiac regenerative medicine is a rapidly evolving field, with promising future developments for effective personalized treatments. Several stem/progenitor cells are candidates for cardiac cell therapy, and emerging evidence suggests how multiple metabolic and biochemical pathways strictly regulate their fate and renewal. SCOPE OF REVIEW In this review, we will explore a selection of areas of common interest for biology and biochemistry concerning stem/progenitor cells, and in particular cardiac progenitor cells. Numerous regulatory mechanisms have been identified that link stem cell signaling and functions to the modulation of metabolic pathways, and vice versa. Pharmacological treatments and culture requirements may be exploited to modulate stem cell pluripotency and self-renewal, possibly boosting their regenerative potential for cell therapy. MAJOR CONCLUSIONS Mitochondria and their many related metabolites and messengers, such as oxygen, ROS, calcium and glucose, have a crucial role in regulating stem cell fate and the balance of their functions, together with many metabolic enzymes. Furthermore, protein biochemistry and proteomics can provide precious clues on the definition of different progenitor cell populations, their physiology and their autocrine/paracrine regulatory/signaling networks. GENERAL SIGNIFICANCE Interdisciplinary approaches between biology and biochemistry can provide productive insights on stem/progenitor cells, allowing the development of novel strategies and protocols for effective cardiac cell therapy clinical translation. This article is part of a Special Issue entitled Biochemistry of Stem Cells.
Collapse
Affiliation(s)
- Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnology, Sapienza University, Italy
| | | | | | | | | |
Collapse
|
47
|
Khokhlova ON, Ilyushkina IA, Fatkhudinov TK, Slashcheva GA, Baikova YP, Bol'shakova GB, Bukharova TB, Turobov VI, Glinkina VV, Murashev AN, Gol'dshtein DV. Improvement of cardiac contractile function in rats with postinfarction cardiosclerosis after transplantation of mononuclear and multipotent stroma bone marrow cells. Bull Exp Biol Med 2012; 153:545-549. [PMID: 22977867 DOI: 10.1007/s10517-012-1763-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We compared the efficiency of autologous mononuclear cells and multipotent stromal cells of the bone marrow after their non-selective intracoronary transplantation on day 30 after acute coronary infarction in rats. Improvement of hemodynamic parameters of myocardial contractility (rates of left ventricular pressure rise and drop) in comparison with the initial values and deceleration of postinfarction prolongation of QRS and QT intervals were observed in rats of the experimental group in contrast to controls in 4 weeks after transplantation. These functional changes were more intensive after transplantation of multipotent stromal cells and were accompanied by more pronounced morphological signs of reverse myocardial remodeling: thickening of the scarred left ventricular wall, shrinkage of the scar, and decrease in left ventricular dilatation index.
Collapse
Affiliation(s)
- O N Khokhlova
- Laboratory of Biological Testing, Branch of M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino, Russia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Liu Y, Tse HF. The proarrhythmic risk of cell therapy for cardiovascular diseases. Expert Rev Cardiovasc Ther 2012; 9:1593-601. [PMID: 22103878 DOI: 10.1586/erc.11.171] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Stem cell therapy is an emerging therapeutic approach for the treatment of cardiovascular diseases. Experimental studies have demonstrated that different types of stem cells, including bone marrow-derived cells, mesenchymal stem cells, skeletal myoblasts, and cardiac progenitor cells and embryonic stem cells, can improve cardiac function after myocardial injuries. Nevertheless, the potential proarrhythmic risk after stem cell transplantation remains a major concern. Several mechanisms, including the immaturity of electrical phenotypes of the transplanted cardiomyocytes, poor cell-cell coupling and cardiac nerve sprouting, may contribute to arrhythmogenic risk after stem cell transplantation. This review summarizes the potential theoretical arrhythmogenic mechanisms associated with different types of stem cells for the treatment of cardiovascular diseases. Nevertheless, current experimental and clinical data on the proarrhythmic risk for different types of stem cell transplantation are limited, and await further experimental and clinical investigation.
Collapse
Affiliation(s)
- Yuan Liu
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, HKSAR, China
| | | |
Collapse
|
49
|
Angeli FS, Zhang Y, Sievers R, Jun K, Yim S, Boyle A, Yeghiazarians Y. Injection of human bone marrow and mononuclear cell extract into infarcted mouse hearts results in functional improvement. Open Cardiovasc Med J 2012; 6:38-43. [PMID: 22550548 PMCID: PMC3339429 DOI: 10.2174/1874192401206010038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Revised: 01/23/2012] [Accepted: 02/13/2012] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND We have previously shown that mouse whole bone marrow cell (BMC) extract results in improvement of cardiac function and decreases scar size in a mouse model of myocardial infarction (MI), in the absence of intact cells. It is not clear if these results are translatable to extracts from human BMC (hBMC) or mononuclear cells (hMNC), which would have significant clinical implications. METHODS Male C57BL/6J (10-12 weeks old) mice were included in this study. MI was created by permanent ligation of the left anterior descending artery. Animals were randomized into three groups to receive ultrasound-guided myocardial injections with either hBMCs extract (n=6), hMNCs extract (n=8) or control with 0.5% bovine serum albumin (BSA) (n=7). Cardiac function was assessed by echocardiography at baseline, 2 and 28 days post-MI. Infarct size and vascularity was assessed at 28 days post-MI. RESULTS hBMC and hMNC extract preserve cardiac function and result in smaller scar size post-MI when compared with the control group. CONCLUSIONS The current study for the first time reports that hBMC and hMNC extracts improve cardiac function post-MI in a mouse MI model. Further studies are necessary to fully address the potential clinical benefits of these therapies.
Collapse
Affiliation(s)
- Franca S Angeli
- Division of Cardiology, Department of Medicine, University of California, San Francisco, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Transplantation of bone marrow-derived mononuclear cells improves mechanical hyperalgesia, cold allodynia and nerve function in diabetic neuropathy. PLoS One 2011; 6:e27458. [PMID: 22125614 PMCID: PMC3220696 DOI: 10.1371/journal.pone.0027458] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 10/17/2011] [Indexed: 01/19/2023] Open
Abstract
Relief from painful diabetic neuropathy is an important clinical issue. We have previously shown that the transplantation of cultured endothelial progenitor cells or mesenchymal stem cells ameliorated diabetic neuropathy in rats. In this study, we investigated whether transplantation of freshly isolated bone marrow-derived mononuclear cells (BM-MNCs) alleviates neuropathic pain in the early stage of streptozotocin-induced diabetic rats. Two weeks after STZ injection, BM-MNCs or vehicle saline were injected into the unilateral hind limb muscles. Mechanical hyperalgesia and cold allodynia in SD rats were measured as the number of foot withdrawals to von Frey hair stimulation and acetone application, respectively. Two weeks after the BM-MNC transplantation, sciatic motor nerve conduction velocity (MNCV), sensory nerve conduction velocity (SNCV), sciatic nerve blood flow (SNBF), mRNA expressions and histology were assessed. The BM-MNC transplantation significantly ameliorated mechanical hyperalgesia and cold allodynia in the BM-MNC-injected side. Furthermore, the slowed MNCV/SNCV and decreased SNBF in diabetic rats were improved in the BM-MNC-injected side. BM-MNC transplantation improved the decreased mRNA expression of NT-3 and number of microvessels in the hind limb muscles. There was no distinct effect of BM-MNC transplantation on the intraepidermal nerve fiber density. These results suggest that autologous transplantation of BM-MNCs could be a novel strategy for the treatment of painful diabetic neuropathy.
Collapse
|