1
|
Patel AS, Ludwinski FE, Kerr A, Farkas S, Kapoor P, Bertolaccini L, Fernandes R, Jones PR, McLornan D, Livieratos L, Saha P, Smith A, Modarai B. A subpopulation of tissue remodeling monocytes stimulates revascularization of the ischemic limb. Sci Transl Med 2024; 16:eadf0555. [PMID: 38896604 DOI: 10.1126/scitranslmed.adf0555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 05/30/2024] [Indexed: 06/21/2024]
Abstract
Despite decades of effort aimed at developing clinically effective cell therapies, including mixed population mononuclear cells, to revascularize the ischemic limb, there remains a paucity of patient-based studies that inform the function and fate of candidate cell types. In this study, we showed that circulating proangiogenic/arteriogenic monocytes (PAMs) expressing the FcγIIIA receptor CD16 were elevated in patients with chronic limb-threatening ischemia (CLTI), and these amounts decreased after revascularization. Unlike CD16-negative monocytes, PAMs showed large vessel remodeling properties in vitro when cultured with endothelial cells and smooth muscle cells and promoted salvage of the ischemic limb in vivo in a mouse model of hindlimb ischemia. PAMs showed a propensity to migrate toward and bind to ischemic muscle and to secrete angiogenic/arteriogenic factors, vascular endothelial growth factor A (VEGF-A) and heparin-binding epidermal growth factor. We instigated a first-in-human single-arm cohort study in which autologous PAMs were injected into the ischemic limbs of five patients with CLTI. Greater than 25% of injected cells were retained in the leg for at least 72 hours, of which greater than 80% were viable, with evidence of enhanced large vessel remodeling in the injected muscle area. In summary, we identified up-regulation of a circulatory PAM subpopulation as an endogenous response to limb ischemia in CLTI and tested a potentially clinically relevant therapeutic strategy.
Collapse
Affiliation(s)
- Ashish S Patel
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, BHF Centre of Excellence, King's College London, London SE1 7EH, UK
- Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London SE1 7EH, UK
| | - Francesca E Ludwinski
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, BHF Centre of Excellence, King's College London, London SE1 7EH, UK
- Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London SE1 7EH, UK
| | - Alexander Kerr
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, BHF Centre of Excellence, King's College London, London SE1 7EH, UK
- Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London SE1 7EH, UK
| | - Simon Farkas
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, BHF Centre of Excellence, King's College London, London SE1 7EH, UK
- Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London SE1 7EH, UK
| | - Puja Kapoor
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, BHF Centre of Excellence, King's College London, London SE1 7EH, UK
- Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London SE1 7EH, UK
| | - Laura Bertolaccini
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, BHF Centre of Excellence, King's College London, London SE1 7EH, UK
- Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London SE1 7EH, UK
| | - Ramon Fernandes
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, BHF Centre of Excellence, King's College London, London SE1 7EH, UK
- Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London SE1 7EH, UK
| | - Paul R Jones
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, BHF Centre of Excellence, King's College London, London SE1 7EH, UK
- Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London SE1 7EH, UK
| | - Donal McLornan
- Department of Haematology, Guy's & St Thomas' NHS Foundation Trust, London SE1 7EH, UK
| | - Lefteris Livieratos
- Department of Biomedical Engineering, School of Biomedical Engineering & Imaging Sciences, King's College London, London SE1 7EH, UK
- Department of Nuclear Medicine, Guy's & St Thomas' NHS Foundation Trust, London SE1 7EH, UK
| | - Prakash Saha
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, BHF Centre of Excellence, King's College London, London SE1 7EH, UK
- Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London SE1 7EH, UK
| | - Alberto Smith
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, BHF Centre of Excellence, King's College London, London SE1 7EH, UK
- Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London SE1 7EH, UK
| | - Bijan Modarai
- Academic Department of Vascular Surgery, South Bank Section, School of Cardiovascular and Metabolic Medicine & Sciences, BHF Centre of Excellence, King's College London, London SE1 7EH, UK
- Biomedical Research Centre at Guy's & St Thomas' NHS Foundation Trust and King's College London, London SE1 7EH, UK
| |
Collapse
|
2
|
Rakshit P, Giri TK, Mukherjee K. Progresses and perspectives on natural polysaccharide based hydrogels for repair of infarcted myocardium. Int J Biol Macromol 2024; 269:132213. [PMID: 38729464 DOI: 10.1016/j.ijbiomac.2024.132213] [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: 01/31/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
Myocardial infarction (MI) is serious health threat and impairs the quality of life. It is a major causative factor of morbidity and mortality. MI leads to the necrosis of cardio-myocytes, cardiac remodelling and dysfunction, eventually leading to heart failure. The limitations of conventional therapeutic and surgical interventions and lack of heart donors have necessitated the evolution of alternate treatment approaches for MI. Polysaccharide hydrogel based repair of infarcted myocardium have surfaced as viable option for MI treatment. Polysaccharide hydrogels may be injectable hydrogels or cardiac patches. Injectable hydrogels can in situ deliver cells and bio-actives, facilitating in situ cardiac regeneration and repair. Polysaccharide hydrogel cardiac patches reduce cardiac wall stress, and inhibit ventricular expansion and promote angiogenesis. Herein, we discuss about MI pathophysiology and myocardial microenvironment and how polysaccharide hydrogels are designed to mimic and support the microenvironment for cardiac repair. We also put forward the versatility of the different polysaccharide hydrogels in mimicking diverse cardiac properties, and acting as a medium for delivery of cells, and therapeutics for promoting angiogenesis and cardiac repair. The objectives of this review is to summarize the factors leading to MI and to put forward how polysaccharide based hydrogels promote cardiac repair. This review is written to enable researchers understand the factors promoting MI so that they can undertake and design novel hydrogels for cardiac regeneration.
Collapse
Affiliation(s)
- Pallabita Rakshit
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India
| | - Tapan Kumar Giri
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India
| | - Kaushik Mukherjee
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, West Bengal, India.
| |
Collapse
|
3
|
Wang X, Wang Q, Meng L, Tian R, Guo H, Tan Z, Tan Y. Biodistribution-based Administration of cGMP-compliant Human Umbilical Cord Mesenchymal Stem Cells Affects the Therapeutic Effect of Wound Healing. Stem Cell Rev Rep 2024; 20:329-346. [PMID: 37889447 DOI: 10.1007/s12015-023-10644-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Although mesenchymal stem cells (MSCs) are used as therapeutic agents for skin injury therapy, few studies have reported the effects of dosing duration and delivery frequency on wound healing. In addition, before the clinical application of MSCs, it is important to assess whether their usage might influence tumor occurrence. METHODS We described the metabolic patterns of subcutaneous injection of hUC-MSCs using fluorescence tracing and qPCR methods and applied them to the development of drug delivery strategies for promoting wound healing. RESULTS (i) We developed cGMP-compliant hUC-MSC products with critical quality control points for wound healing; (ii) The products did not possess any tumorigenic or tumor-promoting/inhibiting ability in vivo; (iii) Fluorescence tracing and qPCR analyses showed that the subcutaneous application of hUC-MSCs did not result in safety-relevant biodistribution or ectopic migration; (iv) Reinjecting hUC-MSCs after significant consumption significantly improved reepithelialization and dermal regeneration. CONCLUSIONS Our findings provided a reference for controlling the quality of MSC products used for wound healing and highlighted the importance of delivery time and frequency for designing in vivo therapeutic studies.
Collapse
Affiliation(s)
- Xin Wang
- Qilu Cell Therapy Technology Co., Ltd, Gangyuan 6th Road, Licheng District, Ji'nan, Shandong, 250000, People's Republic of China
| | - Qiuhong Wang
- Qilu Cell Therapy Technology Co., Ltd, Gangyuan 6th Road, Licheng District, Ji'nan, Shandong, 250000, People's Republic of China
| | - Lingjiao Meng
- Qilu Cell Therapy Technology Co., Ltd, Gangyuan 6th Road, Licheng District, Ji'nan, Shandong, 250000, People's Republic of China
| | - Ruifeng Tian
- Qilu Cell Therapy Technology Co., Ltd, Gangyuan 6th Road, Licheng District, Ji'nan, Shandong, 250000, People's Republic of China
| | - Huizhen Guo
- Qilu Cell Therapy Technology Co., Ltd, Gangyuan 6th Road, Licheng District, Ji'nan, Shandong, 250000, People's Republic of China
| | - Zengqi Tan
- School of Medicine, Northwest University, Xi'an, China
| | - Yi Tan
- Qilu Cell Therapy Technology Co., Ltd, Gangyuan 6th Road, Licheng District, Ji'nan, Shandong, 250000, People's Republic of China.
- Shandong Yinfeng Life Science Research Institute, Ji'nan, People's Republic of China.
| |
Collapse
|
4
|
Yang Y, Johnson J, Troupes CD, Feldsott EA, Kraus L, Megill E, Bian Z, Asangwe N, Kino T, Eaton DM, Wang T, Wagner M, Ma L, Bryan C, Wallner M, Kubo H, Berretta RM, Khan M, Wang H, Kishore R, Houser SR, Mohsin S. miR-182/183-Rasa1 axis induced macrophage polarization and redox regulation promotes repair after ischemic cardiac injury. Redox Biol 2023; 67:102909. [PMID: 37801856 PMCID: PMC10570148 DOI: 10.1016/j.redox.2023.102909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 09/26/2023] [Indexed: 10/08/2023] Open
Abstract
Few therapies have produced significant improvement in cardiac structure and function after ischemic cardiac injury (ICI). Our possible explanation is activation of local inflammatory responses negatively impact the cardiac repair process following ischemic injury. Factors that can alter immune response, including significantly altered cytokine levels in plasma and polarization of macrophages and T cells towards a pro-reparative phenotype in the myocardium post-MI is a valid strategy for reducing infarct size and damage after myocardial injury. Our previous studies showed that cortical bone stem cells (CBSCs) possess reparative effects after ICI. In our current study, we have identified that the beneficial effects of CBSCs appear to be mediated by miRNA in their extracellular vesicles (CBSC-EV). Our studies showed that CBSC-EV treated animals demonstrated reduced scar size, attenuated structural remodeling, and improved cardiac function versus saline treated animals. These effects were linked to the alteration of immune response, with significantly altered cytokine levels in plasma, and polarization of macrophages and T cells towards a pro-reparative phenotype in the myocardium post-MI. Our detailed in vitro studies demonstrated that CBSC-EV are enriched in miR-182/183 that mediates the pro-reparative polarization and metabolic reprogramming in macrophages, including enhanced OXPHOS rate and reduced ROS, via Ras p21 protein activator 1 (RASA1) axis under Lipopolysaccharides (LPS) stimulation. In summary, CBSC-EV deliver unique molecular cargoes, such as enriched miR-182/183, that modulate the immune response after ICI by regulating macrophage polarization and metabolic reprogramming to enhance repair.
Collapse
Affiliation(s)
- Yijun Yang
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Jaslyn Johnson
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Constantine D Troupes
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Eric A Feldsott
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Lindsay Kraus
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Emily Megill
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Zilin Bian
- Tandon School of Engineering, New York University, NY, United States
| | - Ngefor Asangwe
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Tabito Kino
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Deborah M Eaton
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Tao Wang
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Marcus Wagner
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Lena Ma
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Christopher Bryan
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Markus Wallner
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States; Division of Cardiology, Medical University of Graz, 8036, Graz, Austria
| | - Hajime Kubo
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Remus M Berretta
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Mohsin Khan
- Center for Metabolic Disease Research (CMDR), Temple University Lewis Katz School of Medicine, PA, United States
| | - Hong Wang
- Center for Metabolic Disease Research (CMDR), Temple University Lewis Katz School of Medicine, PA, United States
| | - Raj Kishore
- Center for Translational Medicine, Temple University Lewis Katz School of Medicine, PA, United States
| | - Steven R Houser
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States
| | - Sadia Mohsin
- Cardiovascular Research Center (CVRC), Temple University Lewis Katz School of Medicine, PA, United States.
| |
Collapse
|
5
|
Yu B, Li H, Zhang Z, Chen P, Wang L, Fan X, Ning X, Pan Y, Zhou F, Hu X, Chang J, Ou C. Extracellular vesicles engineering by silicates-activated endothelial progenitor cells for myocardial infarction treatment in male mice. Nat Commun 2023; 14:2094. [PMID: 37055411 PMCID: PMC10102163 DOI: 10.1038/s41467-023-37832-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 04/03/2023] [Indexed: 04/15/2023] Open
Abstract
Extracellular vesicles have shown good potential in disease treatments including ischemic injury such as myocardial infarction. However, the efficient production of highly active extracellular vesicles is one of the critical limitations for their clinical applications. Here, we demonstrate a biomaterial-based approach to prepare high amounts of extracellular vesicles with high bioactivity from endothelial progenitor cells (EPCs) by stimulation with silicate ions derived from bioactive silicate ceramics. We further show that hydrogel microspheres containing engineered extracellular vesicles are highly effective in the treatment of myocardial infarction in male mice by significantly enhancing angiogenesis. This therapeutic effect is attributed to significantly enhanced revascularization by the high content of miR-126a-3p and angiogenic factors such as VEGF and SDF-1, CXCR4 and eNOS in engineered extracellular vesicles, which not only activate endothelial cells but also recruit EPCs from the circulatory system.
Collapse
Affiliation(s)
- Bin Yu
- The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510280, Guangzhou, China
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
| | - Hekai Li
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Southern Medical University, 510515, Guangzhou, China
| | - Zhaowenbin Zhang
- Wenzhou Institute, Zhejiang Engineering Research Center for Tissue Repair Materials, University of Chinese Academy of Sciences, 325000, Wenzhou, China
- State Key Laboratory of High-Performance Ceramics and Super fine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 200050, Shanghai, People's Republic of China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, China
| | - Peier Chen
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Southern Medical University, 510515, Guangzhou, China
| | - Ling Wang
- School of Biomedical Engineering, Biomaterials Research Center, Southern Medical University, 510515, Guangzhou, People's Republic of China
| | - Xianglin Fan
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Southern Medical University, 510515, Guangzhou, China
| | - Xiaodong Ning
- The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510280, Guangzhou, China
| | - Yuxuan Pan
- The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510280, Guangzhou, China
| | - Feiran Zhou
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Southern Medical University, 510515, Guangzhou, China
| | - Xinyi Hu
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Southern Medical University, 510515, Guangzhou, China
| | - Jiang Chang
- The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510280, Guangzhou, China.
- Wenzhou Institute, Zhejiang Engineering Research Center for Tissue Repair Materials, University of Chinese Academy of Sciences, 325000, Wenzhou, China.
- State Key Laboratory of High-Performance Ceramics and Super fine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 200050, Shanghai, People's Republic of China.
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, China.
| | - Caiwen Ou
- The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, 510280, Guangzhou, China.
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China.
- Department of Cardiology and Laboratory of Heart Center, Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Southern Medical University, 510515, Guangzhou, China.
| |
Collapse
|
6
|
Handley EL, Callanan A. Modulation of Tissue Microenvironment Following Myocardial Infarction. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Ella Louise Handley
- Institute for Bioengineering School of Engineering University of Edinburgh Edinburgh EH9 3DW UK
| | - Anthony Callanan
- Institute for Bioengineering School of Engineering University of Edinburgh Edinburgh EH9 3DW UK
| |
Collapse
|
7
|
Soltani S, Emadi R, Haghjooy Javanmard S, Kharaziha M, Rahmati A, Thakur VK, Lotfian S. Development of an Injectable Shear-Thinning Nanocomposite Hydrogel for Cardiac Tissue Engineering. Gels 2022; 8:121. [PMID: 35200502 PMCID: PMC8871917 DOI: 10.3390/gels8020121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/07/2022] [Accepted: 02/10/2022] [Indexed: 12/28/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) offer a promising therapeutic method for cardiac tissue regeneration. However, to monitor the fate of MSCs for tissue repair, a better stem cell delivery carrier is needed. Developing a unique injectable and shear-thinning dual cross-linked hybrid hydrogel for MSC delivery for cardiac tissue engineering is highly desirable. This hydrogel was synthesised using guest: host reaction based on alginate-cyclodextrin (Alg-CD) and adamantane-graphene oxide (Ad-GO). Here, the role of macromere concentration (10 and 12%) on the MSC function is discussed. Our hybrid hydrogels reveal a suitable oxygen pathway required for cell survival. However, this value is strongly dependent on the macromere concentrations, while the hydrogels with 12% macromere concentration (2DC12) significantly enhanced the oxygen permeability value (1.16-fold). Moreover, after two weeks of culture, rat MSCs (rMSCs) encapsulated in Alg-GO hydrogels expressed troponin T (TNT) and GATA4 markers. Noticeably, the 2DC12 hydrogels enhance rMSCs differentiation markers (1.30-times for TNT and 1.21-times for GATA4). Overall, our findings indicate that tuning the hydrogel compositions regulates the fate of encapsulated rMSCs within hydrogels. These outcomes may promote the advancement of new multifunctional platforms that consider the spatial and transient guidelines of undifferentiated cell destiny and capacity even after transplantation for heart tissue regeneration.
Collapse
Affiliation(s)
- Samaneh Soltani
- Biomaterials Research Group, Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran; (S.S.); (R.E.); (M.K.)
| | - Rahmatollah Emadi
- Biomaterials Research Group, Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran; (S.S.); (R.E.); (M.K.)
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran;
| | - Mahshid Kharaziha
- Biomaterials Research Group, Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran; (S.S.); (R.E.); (M.K.)
| | - Abbas Rahmati
- Department of Chemistry, University of Isfahan, Isfahan 81746-73441, Iran;
| | - Vijay Kumar Thakur
- Biorefining and Advanced Materials Research Center, Scotland’s Rural College (SRUC), Kings Buildings, Edinburgh EH9 3JG, UK
- School of Engineering, University of Petroleum & Energy Studies (UPES), Dehradun 248007, India
| | - Saeid Lotfian
- Faculty of Engineering, University of Strathclyde, Glasgow G4 0LZ, UK
| |
Collapse
|
8
|
Kang IS, Kwon K. Potential application of biomimetic exosomes in cardiovascular disease: focused on ischemic heart disease. BMB Rep 2022. [PMID: 34903320 PMCID: PMC8810547 DOI: 10.5483/bmbrep.2022.55.1.161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cardiovascular disease, especially ischemic heart disease, is a major cause of mortality worldwide. Cardiac repair is one of the most promising strategies to address advanced cardiovascular diseases. Despite moderate improvement in heart function via stem cell therapy, there is no evidence of significant improvement in mortality and morbidity beyond standard therapy. The most salutary effect of stem cell therapy are attributed to the paracrine effects and the stem cell-derived exosomes are known as a major contributor. Hence, exosomes are emerging as a promising therapeutic agent and potent biomarkers of cardiovascular disease. Furthermore, they play a role as cellular cargo and facilitate intercellular communication. However, the clinical use of exosomes is hindered by the absence of a standard operating procedures for exosome isolation and characterization, problems related to yield, and heterogeneity. In addition, the successful clinical application of exosomes requires strategies to optimize cargo, improve targeted delivery, and reduce the elimination of exosomes. In this review, we discuss the basic concept of exosomes and stem cell-derived exosomes in cardiovascular disease, and introduce current efforts to overcome the limitations and maximize the benefit of exosomes including engineered biomimetic exosomes.
Collapse
Affiliation(s)
- In Sook Kang
- Department of Internal Medicine, School of Medicine, Ewha Womans University, Seoul 07804, Korea
| | - Kihwan Kwon
- Department of Internal Medicine, School of Medicine, Ewha Womans University, Seoul 07804, Korea
| |
Collapse
|
9
|
Kamiyama Y, Naritomi Y, Moriya Y, Yamamoto S, Kitahashi T, Maekawa T, Yahata M, Hanada T, Uchiyama A, Noumaru A, Koga Y, Higuchi T, Ito M, Komatsu H, Miyoshi S, Kimura S, Umeda N, Fujita E, Tanaka N, Sugita T, Takayama S, Kurogi A, Yasuda S, Sato Y. Biodistribution studies for cell therapy products: Current status and issues. Regen Ther 2021; 18:202-216. [PMID: 34307798 PMCID: PMC8282960 DOI: 10.1016/j.reth.2021.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/02/2021] [Accepted: 06/16/2021] [Indexed: 01/01/2023] Open
Abstract
Information on the biodistribution (BD) of cell therapy products (CTPs) is essential for prediction and assessment of their efficacy and toxicity profiles in non-clinical and clinical studies. To conduct BD studies, it is necessary to understand regulatory requirements, implementation status, and analytical methods. This review aimed at surveying international and Japanese trends concerning the BD study for CTPs and the following subjects were investigated, which were considered particularly important: 1) comparison of guidelines to understand the regulatory status of BD studies in a global setting; 2) case studies of the BD study using databases to understand its current status in cell therapy; 3) case studies on quantitative polymerase chain reaction (qPCR) used primarily in non-clinical BD studies for CTPs; and 4) survey of imaging methods used for non-clinical and clinical BD studies. The results in this review will be a useful resource for implementing BD studies.
Collapse
Affiliation(s)
- Yoshiteru Kamiyama
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Yoichi Naritomi
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Yuu Moriya
- Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Syunsuke Yamamoto
- Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Tsukasa Kitahashi
- Bioscience & Engineering Laboratory, FUJIFILM Corp., 577 Ushijima, Kaisei-Machi, Ashigarakami-gun, Kanagawa, Japan
| | - Toshihiko Maekawa
- Bioscience & Engineering Laboratory, FUJIFILM Corp., 577 Ushijima, Kaisei-Machi, Ashigarakami-gun, Kanagawa, Japan
| | - Masahiro Yahata
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka, Japan
| | - Takeshi Hanada
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo.Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, Japan
| | - Asako Uchiyama
- Drug Safety Research Laboratories, Shin Nippon Biomedical Laboratories, Ltd., Kagoshima, Kagoshima, Japan
| | - Akari Noumaru
- Kumamoto Laboratories, LSIM Safety Institute Corporation, 1285 Kurisaki-machi, Uto, Kumamoto, Japan
| | - Yoshiyuki Koga
- Kumamoto Laboratories, LSIM Safety Institute Corporation, 1285 Kurisaki-machi, Uto, Kumamoto, Japan
| | - Tomoaki Higuchi
- Non-clinical Development, Axcelead Drug Discovery Partners, Inc., 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Masahiko Ito
- Tsukuba Research Institute, BoZo Research Center Inc., 8 Okubo, Tsukuba, Ibaraki, Japan
| | - Hiroyuki Komatsu
- Science BD Department, CMIC Pharma Science Co., Ltd., 1-1-1 Shibaura, Minato-ku, Tokyo, Japan
| | - Sosuke Miyoshi
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Sadaaki Kimura
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Nobuhiro Umeda
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Eriko Fujita
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki, Japan
| | - Naoko Tanaka
- Evaluation Center, Terumo Corporation, 1500 Inokuchi, Nakai-machi, Ashigarakami-gun, Kanagawa, Japan
| | - Taku Sugita
- Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan
| | - Satoru Takayama
- Cell Therapy Technology, Healthcare R&D Center, Asahi Kasei Corporation, 2-1 Samejima, Fuji-Shi, Shizuoka, Japan
| | - Akihiko Kurogi
- Regenerative Medicine Research & Planning Division, ROHTO Pharmaceutical Co., Ltd., Osaka, Japan
| | - Satoshi Yasuda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
| | - Yoji Sato
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan
| |
Collapse
|
10
|
Suppressing Pyroptosis Augments Post-Transplant Survival of Stem Cells and Cardiac Function Following Ischemic Injury. Int J Mol Sci 2021; 22:ijms22157946. [PMID: 34360711 PMCID: PMC8348609 DOI: 10.3390/ijms22157946] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/20/2021] [Accepted: 07/20/2021] [Indexed: 12/11/2022] Open
Abstract
The acute demise of stem cells following transplantation significantly compromises the efficacy of stem cell-based cell therapeutics for infarcted hearts. As the stem cells transplanted into the damaged heart are readily exposed to the hostile environment, it can be assumed that the acute death of the transplanted stem cells is also inflicted by the same environmental cues that caused massive death of the host cardiac cells. Pyroptosis, a highly inflammatory form of programmed cell death, has been added to the list of important cell death mechanisms in the damaged heart. However, unlike the well-established cell death mechanisms such as necrosis or apoptosis, the exact role and significance of pyroptosis in the acute death of transplanted stem cells have not been explored in depth. In the present study, we found that M1 macrophages mediate the pyroptosis in the ischemia/reperfusion (I/R) injured hearts and identified miRNA-762 as an important regulator of interleukin 1β production and subsequent pyroptosis. Delivery of exogenous miRNA-762 prior to transplantation significantly increased the post-transplant survival of stem cells and also significantly ameliorated cardiac fibrosis and heart functions following I/R injury. Our data strongly suggest that suppressing pyroptosis can be an effective adjuvant strategy to enhance the efficacy of stem cell-based therapeutics for diseased hearts.
Collapse
|
11
|
Moazzami K, Lima BB, Hammadah M, Ramadan R, Al Mheid I, Kim JH, Alkhoder A, Obideen M, Levantsevych O, Shah A, Liu C, Bremner JD, Kutner M, Sun YV, Waller EK, Hesaroieh IG, Raggi P, Vaccarino V, Quyyumi AA. Association Between Change in Circulating Progenitor Cells During Exercise Stress and Risk of Adverse Cardiovascular Events in Patients With Coronary Artery Disease. JAMA Cardiol 2021; 5:147-155. [PMID: 31799987 PMCID: PMC6902161 DOI: 10.1001/jamacardio.2019.4528] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Importance Stem and progenitor cells mobilize from the bone marrow in response to myocardial ischemia. However, the association between the change in circulating progenitor cell (CPC) counts and disease prognosis among patients with ischemia is unknown. Objective To investigate the association between the change in CPC counts during stress testing and the risk of adverse cardiovascular events in patients with stable coronary artery disease (CAD). Design, Setting, and Participants This prospective cohort study included a population-based sample of 454 patients with stable CAD who were recruited between June 1, 2011, and August 15, 2014, at Emory University-affiliated hospitals and followed up for 3 years. Data were analyzed from September 15, 2018, to October 15, 2018. Exposures Myocardial perfusion imaging with technetium Tc 99m sestamibi at rest and 30 to 60 minutes after conventional stress testing. Main Outcomes and Measures Circulating progenitor cells were enumerated with flow cytometry as CD34-expressing mononuclear cells (CD45med/CD34+), with additional quantification of subsets coexpressing the chemokine (C-X-C motif) receptor 4 (CD34+/CXCR4+). Changes in CPC counts were calculated as poststress minus resting CPC counts. Cox proportional hazards regression models were used to identify factors associated with the combined end point of cardiovascular death and myocardial infarction after adjusting for clinical covariates, including age, sex, race, smoking history, body mass index, and history of heart failure, hypertension, dyslipidemia, and diabetes. Results Of the 454 patients (mean [SD] age, 63 [9] years; 76% men) with stable CAD enrolled in the study, 142 (31.3%) had stress-induced ischemia and 312 (68.7%) did not, as measured by single-photon emission computed tomography. During stress testing, patients with stress-induced ischemia had a mean decrease of 20.2% (interquartile range [IQR], -45.3 to 5.5; P < .001) in their CD34+/CXCR4+ counts, and patients without stress-induced ischemia had a mean increase of 3.2% (IQR, -20.6 to 35.1; P < .001) in their CD34+/CXCR4+ counts. Twenty-four patients (5.2%) experienced adverse events. After adjustment, baseline CPC counts were associated with worse adverse outcomes, but this association was not present after stress-induced ischemia was included in the model. However, the change in CPC counts during exercise remained significantly associated with adverse events (hazard ratio, 2.59; 95% CI, 1.15-5.32, per 50% CD34+/CXCR4+ count decrease), even after adjustment for clinical variables and the presence of ischemia. The discrimination of risk factors associated with incident adverse events improved (increase in C statistic from 0.72 to 0.77; P = .003) with the addition of the change in CD34+/CXCR4+ counts to a model that included clinical characteristics, baseline CPC count, and ischemia. Conclusions and Relevance In this study of patients with CAD, a decrease in CPC counts during exercise is associated with a worse disease prognosis compared with the presence of stress-induced myocardial ischemia. Further studies are needed to evaluate whether strategies to improve CPC responses during exercise stress will be associated with improvements in the prognosis of patients with CAD.
Collapse
Affiliation(s)
- Kasra Moazzami
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia.,Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Bruno B Lima
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia.,Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Mohammad Hammadah
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia.,Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Ronnie Ramadan
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Ibhar Al Mheid
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Jeong Hwan Kim
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Ayman Alkhoder
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Malik Obideen
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Oleksiy Levantsevych
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Amit Shah
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia.,Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia.,Atlanta VA Medical Center, Decatur, Georgia
| | - Chang Liu
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - J Douglas Bremner
- Atlanta VA Medical Center, Decatur, Georgia.,Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia.,Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Michael Kutner
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Edmund K Waller
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Iraj Ghaini Hesaroieh
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Paolo Raggi
- Mazankowski Alberta Heart Institute, University of Alberta, Alberta, Canada
| | - Viola Vaccarino
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Arshed A Quyyumi
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
12
|
Chandy M, Wu JC. Molecular Imaging of Stem Cell Therapy in Ischemic Cardiomyopathy. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00065-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
13
|
Kim K, Bou-Ghannam S, Kameishi S, Oka M, Grainger DW, Okano T. Allogeneic mesenchymal stem cell sheet therapy: A new frontier in drug delivery systems. J Control Release 2020; 330:696-704. [PMID: 33347942 DOI: 10.1016/j.jconrel.2020.12.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022]
Abstract
The evolution of drug discovery exploded in the early 20th century with the advent of critical scientific advancements in organic chemistry, chemical analysis, and purification. Early drug generations focused largely on symptom control and pain management, effective targets for small-molecule drugs. Recently, the attention in drug discovery has shifted to pursuit of radical cures. Cell therapy presents the ideal attributes of a promising new drug, targeting specific tissues based on chemotactic cues and modulating secretion of instructive regenerative molecules in response to dynamic signaling from disease environments. To actuate the therapeutic potential of cell therapy toward worldwide clinical use, cell delivery methods that can effectively localize and engraft mesenchymal stem cells (MSCs) with high disease-site fidelity and enable dynamic MSC bioactive function are paramount. In this review, we discuss the evolution of cell therapies with a focus on stem cell advantages, as well as the limitations to these therapies. This review aims to introduce cell sheet technology as a breakthrough cell therapy with demonstrated therapeutic success across indications for heart, liver, and kidney tissue regeneration. Opportunities and anticipated clinical impacts of cell sheet technology using MSCs are discussed.
Collapse
Affiliation(s)
- Kyungsook Kim
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA.
| | - Sophia Bou-Ghannam
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, 36 South, Wasatch Drive, Salt Lake City, UT 84112, USA
| | - Sumako Kameishi
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA
| | - Masatoshi Oka
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA
| | - David W Grainger
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, 36 South, Wasatch Drive, Salt Lake City, UT 84112, USA
| | - Teruo Okano
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| |
Collapse
|
14
|
Hess A, Thackeray JT, Wollert KC, Bengel FM. Radionuclide Image-Guided Repair of the Heart. JACC Cardiovasc Imaging 2020; 13:2415-2429. [DOI: 10.1016/j.jcmg.2019.11.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/23/2019] [Accepted: 11/05/2019] [Indexed: 01/12/2023]
|
15
|
Zaw Thin M, Allan H, Bofinger R, Kostelec TD, Guillaume S, Connell JJ, Patrick PS, Hailes HC, Tabor AB, Lythgoe MF, Stuckey DJ, Kalber TL. Multi-modal imaging probe for assessing the efficiency of stem cell delivery to orthotopic breast tumours. NANOSCALE 2020; 12:16570-16585. [PMID: 32749427 PMCID: PMC7586303 DOI: 10.1039/d0nr03237a] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/09/2020] [Indexed: 05/05/2023]
Abstract
Stem cells have been utilised as anti-cancer agents due to their ability to home to and integrate within tumours. Methods to augment stem cell homing to tumours are being investigated with the goal of enhancing treatment efficacy. However, it is currently not possible to evaluate both cell localisation and cell viability after engraftment, hindering optimisation of therapy. In this study, luciferase-expressing human adipocyte-derived stem cells (ADSCs) were incubated with Indium-111 radiolabelled iron oxide nanoparticles to produce cells with tri-modal imaging capabilities. ADSCs were administered intravenously (IV) or intracardially (IC) to mice bearing orthotopic breast tumours. Cell fate was monitored using bioluminescence imaging (BLI) as a measure of cell viability, magnetic resonance imaging (MRI) for cell localisation and single photon emission computer tomography (SPECT) for cell quantification. Serial monitoring with multi-modal imaging showed the presence of viable ADSCs within tumours as early as 1-hour post IC injection and the percentage of ADSCs within tumours to be 2-fold higher after IC than IV. Finally, histological analysis was used to validate engraftment of ADSC within tumour tissue. These findings demonstrate that multi-modal imaging can be used to evaluate the efficiency of stem cell delivery to tumours and that IC cell administration is more effective for tumour targeting.
Collapse
Affiliation(s)
- May Zaw Thin
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK.
| | - Helen Allan
- Department of Chemistry, University College London, 20, Gordon Street, London, WC1H 0AJ, UK
| | - Robin Bofinger
- Department of Chemistry, University College London, 20, Gordon Street, London, WC1H 0AJ, UK
| | - Tomas D Kostelec
- Department of Chemistry, University College London, 20, Gordon Street, London, WC1H 0AJ, UK
| | - Simon Guillaume
- Department of Chemistry, University College London, 20, Gordon Street, London, WC1H 0AJ, UK
| | - John J Connell
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK.
| | - P Stephen Patrick
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK.
| | - Helen C Hailes
- Department of Chemistry, University College London, 20, Gordon Street, London, WC1H 0AJ, UK
| | - Alethea B Tabor
- Department of Chemistry, University College London, 20, Gordon Street, London, WC1H 0AJ, UK
| | - Mark F Lythgoe
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK.
| | - Daniel J Stuckey
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK.
| | - Tammy L Kalber
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, WC1E 6DD, UK.
| |
Collapse
|
16
|
Maghin E, Garbati P, Quarto R, Piccoli M, Bollini S. Young at Heart: Combining Strategies to Rejuvenate Endogenous Mechanisms of Cardiac Repair. Front Bioeng Biotechnol 2020; 8:447. [PMID: 32478060 PMCID: PMC7237726 DOI: 10.3389/fbioe.2020.00447] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/17/2020] [Indexed: 12/12/2022] Open
Abstract
True cardiac regeneration of the injured heart has been broadly described in lower vertebrates by active replacement of lost cardiomyocytes to functionally and structurally restore the myocardial tissue. On the contrary, following severe injury (i.e., myocardial infarction) the adult mammalian heart is endowed with an impaired reparative response by means of meager wound healing program and detrimental remodeling, which can lead over time to cardiomyopathy and heart failure. Lately, a growing body of basic, translational and clinical studies have supported the therapeutic use of stem cells to provide myocardial regeneration, with the working hypothesis that stem cells delivered to the cardiac tissue could result into new cardiovascular cells to replenish the lost ones. Nevertheless, multiple independent evidences have demonstrated that injected stem cells are more likely to modulate the cardiac tissue via beneficial paracrine effects, which can enhance cardiac repair and reinstate the embryonic program and cell cycle activity of endogenous cardiac stromal cells and resident cardiomyocytes. Therefore, increasing interest has been addressed to the therapeutic profiling of the stem cell-derived secretome (namely the total of cell-secreted soluble factors), with specific attention to cell-released extracellular vesicles, including exosomes, carrying cardioprotective and regenerative RNA molecules. In addition, the use of cardiac decellularized extracellular matrix has been recently suggested as promising biomaterial to develop novel therapeutic strategies for myocardial repair, as either source of molecular cues for regeneration, biological scaffold for cardiac tissue engineering or biomaterial platform for the functional release of factors. In this review, we will specifically address the translational relevance of these two approaches with ad hoc interest in their feasibility to rejuvenate endogenous mechanisms of cardiac repair up to functional regeneration.
Collapse
Affiliation(s)
- Edoardo Maghin
- Tissue Engineering Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy.,Department of Women's and Children Health, University of Padova, Padua, Italy
| | - Patrizia Garbati
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Rodolfo Quarto
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy.,UOC Cellular Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Martina Piccoli
- Tissue Engineering Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - Sveva Bollini
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| |
Collapse
|
17
|
Chan JL, Miller JG, Zhou Y, Robey PG, Stroncek DF, Arai AE, Sachdev V, Horvath KA. Intramyocardial Bone Marrow Stem Cells in Patients Undergoing Cardiac Surgical Revascularization. Ann Thorac Surg 2020; 109:1142-1149. [PMID: 31526779 PMCID: PMC8045460 DOI: 10.1016/j.athoracsur.2019.07.093] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 07/04/2019] [Accepted: 07/29/2019] [Indexed: 01/14/2023]
Abstract
BACKGROUND Bone marrow stromal or stem cells (BMSCs) remain a promising potential therapy for ischemic cardiomyopathy. The primary objective of this study was to evaluate the safety and feasibility of direct intramyocardial injection of autologous BMSCs in patients undergoing transmyocardial revascularization (TMR) or coronary artery bypass graft surgery (CABG). METHODS A phase I trial was conducted on adult patients who had ischemic heart disease with depressed left ventricular ejection fraction and who were scheduled to undergo TMR or CABG. Autologous BMSCs were expanded for 3 weeks before the scheduled surgery. After completion of surgical revascularization, BMSCs were directly injected into ischemic myocardium. Safety and feasibility of therapy were assessed. Cardiac functional status and changes in quality of life were evaluated at 1 year. RESULTS A total of 14 patients underwent simultaneous BMSC and surgical revascularization therapy (TMR+BMSCs = 10; CABG+BMSCs = 4). BMSCs were successfully expanded, and no significant complications occurred as a result of the procedure. Regional contractility in the cell-treated areas demonstrated improvement at 12 months compared with baseline (TMR+BMSCs Δ strain: -4.6% ± 2.1%; P = .02; CABG+MSCs Δ strain: -4.2% ± 6.0%; P = .30). Quality of life was enhanced, with substantial reduction in angina scores at 1 year after treatment (TMR+BMSCs: 1.3 ± 1.2; CABG+MSCs: 1.0 ± 1.4). CONCLUSIONS In this phase I trial, direct intramyocardial injection of autologous BMSCs in conjunction with TMR or CABG was technically feasible and could be performed safely. Preliminary results demonstrate improved cardiac function and quality of life in patients at 1 year after treatment.
Collapse
Affiliation(s)
- Joshua L Chan
- Cardiothoracic Surgery Research Program, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Justin G Miller
- Cardiothoracic Surgery Research Program, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Yifu Zhou
- Cardiothoracic Surgery Research Program, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Pamela G Robey
- NIH Bone Marrow Stromal Cell Transplantation Center, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - David F Stroncek
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Andrew E Arai
- Advanced Cardiovascular Imaging Group, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Vandana Sachdev
- Echocardiography Laboratory, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Keith A Horvath
- Cardiothoracic Surgery Research Program, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
18
|
Ashmore-Harris C, Fruhwirth GO. The clinical potential of gene editing as a tool to engineer cell-based therapeutics. Clin Transl Med 2020; 9:15. [PMID: 32034584 PMCID: PMC7007464 DOI: 10.1186/s40169-020-0268-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/29/2020] [Indexed: 12/13/2022] Open
Abstract
The clinical application of ex vivo gene edited cell therapies first began a decade ago with zinc finger nuclease editing of autologous CD4+ T-cells. Editing aimed to disrupt expression of the human immunodeficiency virus co-receptor gene CCR5, with the goal of yielding cells resistant to viral entry, prior to re-infusion into the patient. Since then the field has substantially evolved with the arrival of the new editing technologies transcription activator-like effector nucleases (TALENs) and clustered regularly interspaced short palindromic repeats (CRISPR), and the potential benefits of gene editing in the arenas of immuno-oncology and blood disorders were quickly recognised. As the breadth of cell therapies available clinically continues to rise there is growing interest in allogeneic and off-the-shelf approaches and multiplex editing strategies are increasingly employed. We review here the latest clinical trials utilising these editing technologies and consider the applications on the horizon.
Collapse
Affiliation(s)
- Candice Ashmore-Harris
- Imaging Therapy and Cancer Group, Dept of Imaging Chemistry & Biology, School of Biomedical Engineering & Imaging Sciences, St Thomas' Hospital, King's College London (KCL), London, SE1 7EH, UK
- Centre for Stem Cells & Regenerative Medicine, School of Basic and Medical Biosciences, Guy's Hospital, KCL, London, SE1 9RT, UK
| | - Gilbert O Fruhwirth
- Imaging Therapy and Cancer Group, Dept of Imaging Chemistry & Biology, School of Biomedical Engineering & Imaging Sciences, St Thomas' Hospital, King's College London (KCL), London, SE1 7EH, UK.
| |
Collapse
|
19
|
Langrzyk A, Nowak WN, Stępniewski J, Jaźwa A, Florczyk-Soluch U, Józkowicz A, Dulak J. Critical View on Mesenchymal Stromal Cells in Regenerative Medicine. Antioxid Redox Signal 2018; 29:169-190. [PMID: 28874054 DOI: 10.1089/ars.2017.7159] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE The belief in the potency of stem cells has resulted in the medical applications of numerous cell types for organ repair, often with the low adherence to methodological stringency. Such uncritical enthusiasm is mainly presented in the approaches employing so-called mesenchymal stem cells (MSC), for the treatment of numerous, unrelated conditions. However, it should be stressed that such broad clinical applications of MSC are mostly based on the belief that MSC can efficiently differentiate into multiple cell types, not only osteoblasts, chondrocytes and adipose cells. Recent Advances: Studies employing lineage tracing established more promising markers to characterize MSC identity and localization in vivo and confirmed the differences between MSC isolated from various organs. Furthermore, preclinical and clinical experiments proved that transdifferentiation of MSC is unlikely to contribute to repair of numerous tissues, including the heart. Therefore, the salvage hypotheses, like MSC fusion with cells in target organs or the paracrine mechanisms, were proposed to justify the widespread application of MSC and to explain transient, if any, effects. CRITICAL ISSUES The lack of standardization concerning the cells markers, their origin and particularly the absence of stringent functional characterization of MSC, leads to propagation of the worrying hype despite the lack of convincing therapeutic efficiency of MSC. FUTURE DIRECTIONS The adherence to rigorous methodological rules is necessary to prevent the application of procedures which can be dangerous for patients and scientific research on the medical application of stem cells. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
| | - Witold N Nowak
- 2 Cardiovascular Division, King's College London , London, United Kingdom .,3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Jacek Stępniewski
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Agnieszka Jaźwa
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Urszula Florczyk-Soluch
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Alicja Józkowicz
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Józef Dulak
- 1 Kardio-Med Silesia , Zabrze, Poland .,3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| |
Collapse
|
20
|
Hausburg F, Müller P, Voronina N, Steinhoff G, David R. Protocol for MicroRNA Transfer into Adult Bone Marrow-derived Hematopoietic Stem Cells to Enable Cell Engineering Combined with Magnetic Targeting. J Vis Exp 2018. [PMID: 29985305 DOI: 10.3791/57474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
While CD133+ hematopoietic stem cells (SCs) have been proven to provide high potential in the field of regenerative medicine, their low retention rates after injection into injured tissues as well as the observed massive cell death rates lead to very restricted therapeutic effects. To overcome these limitations, we sought to establish a non-viral based protocol for suitable cell engineering prior to their administration. The modification of human CD133+ expressing SCs using microRNA (miR) loaded magnetic polyplexes was addressed with respect to uptake efficiency and safety as well as the targeting potential of the cells. Relying on our protocol, we can achieve high miR uptake rates of 80-90% while the CD133+ stem cell properties remain unaffected. Moreover, these modified cells offer the option of magnetic targeting. We describe here a safe and highly efficient procedure for the modification of CD133+ SCs. We expect this approach to provide a standard technology for optimization of therapeutic stem cell effects and for monitoring of the administered cell product via magnetic resonance imaging (MRI).
Collapse
Affiliation(s)
- Frauke Hausburg
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, Rostock University Medical Center; Department Life, Light and Matter of the Interdisciplinary Faculty, Rostock University
| | - Paula Müller
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, Rostock University Medical Center; Department Life, Light and Matter of the Interdisciplinary Faculty, Rostock University
| | - Natalia Voronina
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, Rostock University Medical Center
| | - Gustav Steinhoff
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, Rostock University Medical Center; Department Life, Light and Matter of the Interdisciplinary Faculty, Rostock University
| | - Robert David
- Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), Department of Cardiac Surgery, Rostock University Medical Center; Department Life, Light and Matter of the Interdisciplinary Faculty, Rostock University;
| |
Collapse
|
21
|
Hammadah M, Samman Tahhan A, Mheid IA, Wilmot K, Ramadan R, Kindya BR, Kelli HM, O'Neal WT, Sandesara P, Sullivan S, Almuwaqqat Z, Obideen M, Abdelhadi N, Alkhoder A, Pimple PM, Levantsevych O, Mohammed KH, Weng L, Sperling LS, Shah AJ, Sun YV, Pearce BD, Kutner M, Ward L, Bremner JD, Kim J, Waller EK, Raggi P, Sheps D, Vaccarino V, Quyyumi AA. Myocardial Ischemia and Mobilization of Circulating Progenitor Cells. J Am Heart Assoc 2018; 7:e007504. [PMID: 31898922 PMCID: PMC5850188 DOI: 10.1161/jaha.117.007504] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background The response of progenitor cells (PCs) to transient myocardial ischemia in patients with coronary artery disease remains unknown. We aimed to investigate the PC response to exercise‐induced myocardial ischemia (ExMI) and compare it to flow mismatch during pharmacological stress testing. Methods and Results A total of 356 patients with stable coronary artery disease underwent 99mTc‐sestamibi myocardial perfusion imaging during exercise (69%) or pharmacological stress (31%). CD34+ and CD34+/chemokine (C‐X‐C motif) receptor 4 PCs were enumerated by flow cytometry. Change in PC count was compared between patients with and without myocardial ischemia using linear regression models. Vascular endothelial growth factor and stromal‐derived factor‐1α were quantified. Mean age was 63±9 years; 76% were men. The incidence of ExMI was 31% and 41% during exercise and pharmacological stress testing, respectively. Patients with ExMI had a significant decrease in CD34+/chemokine (C‐X‐C motif) receptor 4 (−18%, P=0.01) after stress that was inversely correlated with the magnitude of ischemia (r=−0.19, P=0.003). In contrast, patients without ExMI had an increase in CD34+/chemokine (C‐X‐C motif) receptor 4 (14.7%, P=0.02), and those undergoing pharmacological stress had no change. Plasma vascular endothelial growth factor levels increased (15%, P<0.001) in all patients undergoing exercise stress testing regardless of ischemia. However, the change in stromal‐derived factor‐1α level correlated inversely with the change in PC counts in those with ExMI (P=0.03), suggesting a greater decrease in PCs in those with a greater change in stromal‐derived factor‐1α level with exercise. Conclusions ExMI is associated with a significant decrease in circulating levels of CD34+/chemokine (C‐X‐C motif) receptor 4 PCs, likely attributable, at least in part, to stromal‐derived factor‐1α–mediated homing of PCs to the ischemic myocardium. The physiologic consequences of this uptake of PCs and their therapeutic implications need further investigation.
Collapse
Affiliation(s)
- Muhammad Hammadah
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Ayman Samman Tahhan
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Ibhar Al Mheid
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Kobina Wilmot
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Ronnie Ramadan
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Bryan R Kindya
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Heval M Kelli
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Wesley T O'Neal
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Pratik Sandesara
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Samaah Sullivan
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Zakaria Almuwaqqat
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Malik Obideen
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Naser Abdelhadi
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Ayman Alkhoder
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Pratik M Pimple
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Oleksiy Levantsevych
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Kareem H Mohammed
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Lei Weng
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Laurence S Sperling
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Amit J Shah
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA.,Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA.,Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Brad D Pearce
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Michael Kutner
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Laura Ward
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA
| | - J Douglas Bremner
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA
| | - Jinhee Kim
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Edmund K Waller
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Paolo Raggi
- Mazankowski Alberta Heart Institute University of Alberta, Edmonton, Alberta, Canada
| | - David Sheps
- Department of Epidemiology, University of Florida, Gainesville, FL
| | - Viola Vaccarino
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA.,Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Arshed A Quyyumi
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
22
|
Caobelli F, Wollenweber T, Bavendiek U, Kühn C, Schütze C, Geworski L, Thackeray JT, Bauersachs J, Haverich A, Bengel FM. Simultaneous dual-isotope solid-state detector SPECT for improved tracking of white blood cells in suspected endocarditis. Eur Heart J 2018; 38:436-443. [PMID: 27469371 DOI: 10.1093/eurheartj/ehw231] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 04/29/2016] [Indexed: 11/13/2022] Open
Abstract
Aims High-energy resolution and sensitivity of novel cadmium-zinc-telluride (CZT) detector equipped SPECT systems facilitate simultaneous imaging of multiple isotopes and may enhance the detection of molecular/cellular signals. This may refine the detection of endocarditis. This study was designed to determine the feasibility and diagnostic accuracy of simultaneous imaging of inflammation with 111In-labeled white blood cells (WBCs) and myocardial perfusion with 99mTc-sestamibi, for localization of WBCs relative to the valve plane in suspected endocarditis. Methods and results A dedicated cardiac CZT camera (Discovery 530c, GE Healthcare) was employed. Anthropomorphic thorax phantom studies were followed by clinical studies in 34 patients with suspected infection of native valves (n = 12) or implants (n = 22). Simultaneous 111In-WBC/99mTc perfusion imaging was performed, and compared with standard 111In-WBC planar scintigraphy and SPECT-CT. Phantom studies ruled out significant radioisotope crosstalk. Downscatter on 99mTc images was not observed for 111In activity as high as 2.5*99mTc activity. In patients, image quality was superior for CZT imaging vs. conventional SPECT-CT and planar scintigraphy (P < 0.01). Cadmium-zinc-telluride dual isotope imaging improved reader confidence for detection of inflammatory foci. Diagnostic accuracy based on surgery or Duke Criteria during follow-up was highest for CZT imaging (P < 0.001). Conclusion Novel CZT SPECT technology improves the accuracy of molecular/cellular cardiac imaging. Simultaneous multi-isotope imaging with 111In and 99mTc is feasible and aids in the workup of suspected endocarditis.
Collapse
Affiliation(s)
- Federico Caobelli
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Tim Wollenweber
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Udo Bavendiek
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Christian Kühn
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Christian Schütze
- Department of Radiation Protection and Medical Physics, Hannover Medical School, Hannover, Germany
| | - Lilli Geworski
- Department of Radiation Protection and Medical Physics, Hannover Medical School, Hannover, Germany
| | - James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Axel Haverich
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
23
|
Dorobantu M, Popa-Fotea NM, Popa M, Rusu I, Micheu MM. Pursuing meaningful end-points for stem cell therapy assessment in ischemic cardiac disease. World J Stem Cells 2017; 9:203-218. [PMID: 29321822 PMCID: PMC5746641 DOI: 10.4252/wjsc.v9.i12.203] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 11/08/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023] Open
Abstract
Despite optimal interventional and medical therapy, ischemic heart disease is still an important cause of morbidity and mortality worldwide. Although not included in standard of care rehabilitation, stem cell therapy (SCT) could be a solution for prompting cardiac regeneration. Multiple studies have been published from the beginning of SCT until now, but overall no unanimous conclusion could be drawn in part due to the lack of appropriate end-points. In order to appreciate the impact of SCT, multiple markers from different categories should be considered: Structural, biological, functional, physiological, but also major adverse cardiac events or quality of life. Imaging end-points are among the most used - especially left ventricle ejection fraction (LVEF) measured through different methods. Other imaging parameters are infarct size, myocardial viability and perfusion. The impact of SCT on all of the aforementioned end-points is controversial and debatable. 2D-echocardiography is widely exploited, but new approaches such as tissue Doppler, strain/strain rate or 3D-echocardiography are more accurate, especially since the latter one is comparable with the MRI gold standard estimation of LVEF. Apart from the objective parameters, there are also patient-centered evaluations to reveal the benefits of SCT, such as quality of life and performance status, the most valuable from the patient point of view. Emerging parameters investigating molecular pathways such as non-coding RNAs or inflammation cytokines have a high potential as prognostic factors. Due to the disadvantages of current techniques, new imaging methods with labelled cells tracked along their lifetime seem promising, but until now only pre-clinical trials have been conducted in humans. Overall, SCT is characterized by high heterogeneity not only in preparation, administration and type of cells, but also in quantification of therapy effects.
Collapse
Affiliation(s)
- Maria Dorobantu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Bucharest 014461, Romania
| | | | - Mihaela Popa
- Carol Davila, University of Medicine, "Carol Davila" University of Medicine and Pharmacy Bucharest, Bucharest 020022, Romania
| | - Iulia Rusu
- Carol Davila, University of Medicine, "Carol Davila" University of Medicine and Pharmacy Bucharest, Bucharest 020022, Romania
| | - Miruna Mihaela Micheu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Bucharest 014461, Romania.
| |
Collapse
|
24
|
Brooks A, Futrega K, Liang X, Hu X, Liu X, Crawford DHG, Doran MR, Roberts MS, Wang H. Concise Review: Quantitative Detection and Modeling the In Vivo Kinetics of Therapeutic Mesenchymal Stem/Stromal Cells. Stem Cells Transl Med 2017; 7:78-86. [PMID: 29210198 PMCID: PMC5746161 DOI: 10.1002/sctm.17-0209] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/02/2017] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) present a promising tool in cell‐based therapy for treatment of various diseases. Currently, optimization of treatment protocols in clinical studies is complicated by the variations in cell dosing, diverse methods used to deliver MSCs, and the variety of methods used for tracking MSCs in vivo. Most studies use a dose escalation approach, and attempt to correlate efficacy with total cell dose. Optimization could be accelerated through specific understanding of MSC distribution in vivo, long‐term viability, as well as their biological fate. While it is not possible to quantitatively detect MSCs in most targeted organs over long time periods after systemic administration in clinical trials, it is increasingly possible to apply pharmacokinetic modeling to predict their distribution and persistence. This Review outlines current understanding of the in vivo kinetics of exogenously administered MSCs, provides a critical analysis of the methods used for quantitative MSC detection in these studies, and discusses the application of pharmacokinetic modeling to these data. Finally, we provide insights on and perspectives for future development of effective therapeutic strategies using pharmacokinetic modeling to maximize MSC therapy and minimize potential side effects. Stem Cells Translational Medicine2018;7:78–86
Collapse
Affiliation(s)
- Anastasia Brooks
- Therapeutics Research Centre, The University of Queensland Diamantina Institute, Brisbane, Australia.,School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Kathryn Futrega
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Brisbane, Australia
| | - Xiaowen Liang
- Therapeutics Research Centre, The University of Queensland Diamantina Institute, Brisbane, Australia
| | - Xiaoling Hu
- Therapeutics Research Centre, The University of Queensland Diamantina Institute, Brisbane, Australia
| | - Xin Liu
- Therapeutics Research Centre, The University of Queensland Diamantina Institute, Brisbane, Australia
| | - Darrell H G Crawford
- School of Clinical Medicine, The University of Queensland, Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, Australia
| | - Michael R Doran
- Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia.,Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Brisbane, Australia.,Australian National Centre for the Public Awareness of Science, Australian National University, Canberra, Australia
| | - Michael S Roberts
- Therapeutics Research Centre, The University of Queensland Diamantina Institute, Brisbane, Australia.,School of Pharmacy and Medical Sciences, University of South Australia, Basil Hetzel Institute, Adelaide, Australia
| | - Haolu Wang
- Therapeutics Research Centre, The University of Queensland Diamantina Institute, Brisbane, Australia.,Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| |
Collapse
|
25
|
Seeto WJ, Tian Y, Winter RL, Caldwell FJ, Wooldridge AA, Lipke EA. Encapsulation of Equine Endothelial Colony Forming Cells in Highly Uniform, Injectable Hydrogel Microspheres for Local Cell Delivery. Tissue Eng Part C Methods 2017; 23:815-825. [DOI: 10.1089/ten.tec.2017.0233] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Wen J. Seeto
- Department of Chemical Engineering, Auburn University, Auburn, Alabama
| | - Yuan Tian
- Department of Chemical Engineering, Auburn University, Auburn, Alabama
| | - Randolph L. Winter
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Fred J. Caldwell
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Anne A. Wooldridge
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | | |
Collapse
|
26
|
Li X, Hacker M. Molecular imaging in stem cell-based therapies of cardiac diseases. Adv Drug Deliv Rev 2017; 120:71-88. [PMID: 28734900 DOI: 10.1016/j.addr.2017.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 07/06/2017] [Accepted: 07/16/2017] [Indexed: 12/26/2022]
Abstract
In the past 15years, despite that regenerative medicine has shown great potential for cardiovascular diseases, the outcome and safety of stem cell transplantation has shown controversial results in the published literature. Medical imaging might be useful for monitoring and quantifying transplanted cells within the heart and to serially characterize the effects of stem cell therapy of the myocardium. From the multiple available noninvasive imaging techniques, magnetic resonance imaging and nuclear imaging by positron (PET) or single photon emission computer tomography (SPECT) are the most used clinical approaches to follow the fate of transplanted stem cells in vivo. In this article, we provide a review on the role of different noninvasive imaging modalities and discuss their advantages and disadvantages. We focus on the different in-vivo labeling and reporter gene imaging strategies for stem cell tracking as well as the concept and reliability to use imaging parameters as noninvasive surrogate endpoints for the evaluation of the post-therapeutic outcome.
Collapse
Affiliation(s)
- Xiang Li
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Austria
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Austria.
| |
Collapse
|
27
|
Vo D, Nguyen PK. Multimodality molecular imaging in cardiac regenerative therapy. J Nucl Cardiol 2017; 24:1803-1809. [PMID: 28185234 DOI: 10.1007/s12350-017-0785-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 01/05/2017] [Indexed: 01/01/2023]
Abstract
Stem cell therapy holds great promise for the repair and regeneration of damaged myocardium. Disappointing results from recent large-scale randomized trials using adult stem cells, however, have led some to question the efficacy of this new therapeutic. Because most clinical stem cell trials have not incorporated molecular imaging to track cell fate, it may be premature to abandon this approach. Herein, we will review how multimodality imaging can be incorporated into cardiac regenerative therapy to facilitate the translation of stem cell therapy.
Collapse
Affiliation(s)
- Davis Vo
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, 300 Pasteur Drive, Stanford, CA, 94305, USA
- Cardiology Section, Department of Medicine, Veterans Affairs, 3801 Miranda Ave, Palo Alto, CA, 94304, USA
| | - Patricia K Nguyen
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, 300 Pasteur Drive, Stanford, CA, 94305, USA.
- Cardiology Section, Department of Medicine, Veterans Affairs, 3801 Miranda Ave, Palo Alto, CA, 94304, USA.
- Stanford University, 300 Pasteur Drive, Grant Building, S114, Stanford, CA, 94305-5208, USA.
| |
Collapse
|
28
|
Abstract
INTRODUCTION Over the past decade, it has become clear that long-term engraftment of any ex vivo expanded cell product transplanted into injured myocardium is modest and all therapeutic regeneration is mediated by stimulation of endogenous repair rather than differentiation of transplanted cells into working myocardium. Given that increasing the retention of transplanted cells boosts myocardial function, focus on the fundamental mechanisms limiting retention and survival of transplanted cells may enable strategies to help to restore normal cardiac function. Areas covered: This review outlines the challenges confronting cardiac engraftment of ex vivo expanded cells and explores means of enhancing cell-mediated repair of injured myocardium. Expert opinion: Stem cell therapy has already come a long way in terms of regenerating damaged hearts though the poor retention of transplanted cells limits the full potential of truly cardiotrophic cell products. Multifaceted strategies directed towards fundamental mechanisms limiting the long-term survival of transplanted cells will be needed to enhance transplanted cell retention and cell-mediated repair of damaged myocardium for cardiac cell therapy to reach its full potential.
Collapse
Affiliation(s)
| | - Darryl R Davis
- a University of Ottawa Heart Institute , Ottawa , ON , Canada
| |
Collapse
|
29
|
Traverse JH, Henry TD. Myocardial Injury as a New Target for Cell Therapy in Patients With Chronic Heart Failure: When Something Bad Is Actually Good? Circ Res 2017; 120:1857-1859. [PMID: 28596168 DOI: 10.1161/circresaha.117.311073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Jay H Traverse
- From the Minneapolis Heart Institute Foundation, Abbott Northwestern Hospital, MN (J.H.T.); Cardiovascular Division, The University of Minnesota Medical School, Minneapolis (J.H.T.); and Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.).
| | - Timothy D Henry
- From the Minneapolis Heart Institute Foundation, Abbott Northwestern Hospital, MN (J.H.T.); Cardiovascular Division, The University of Minnesota Medical School, Minneapolis (J.H.T.); and Cedars-Sinai Heart Institute, Los Angeles, CA (T.D.H.)
| |
Collapse
|
30
|
Luu B, Leistner DM, Herrmann E, Seeger FH, Honold J, Fichtlscherer S, Zeiher AM, Assmus B. Minute Myocardial Injury as Measured by High-Sensitive Troponin T Serum Levels Predicts the Response to Intracoronary Infusion of Bone Marrow-Derived Mononuclear Cells in Patients With Stable Chronic Post-Infarction Heart Failure: Insights From the TOPCARE-CHD Registry. Circ Res 2017; 120:1938-1946. [PMID: 28351842 DOI: 10.1161/circresaha.116.309938] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 03/16/2017] [Accepted: 03/27/2017] [Indexed: 12/13/2022]
Abstract
RATIONALE Cell-based therapies are a promising option in patients with chronic postinfarction heart failure (ischemic cardiomyopathy [ICM]). However, the responses after intracoronary infusion of autologous bone marrow-derived mononuclear cells (BMCs) are heterogeneous, which may be related to impaired cell retention in patients with ICM. Ischemic injury is associated with upregulation of prototypical chemoattractant cytokines mediating retention and homing of circulating cells. The development of ultrasensitive tests to measure high-sensitive troponin T (hs-TnT) serum levels revealed the presence of ongoing minute myocardial injury even in patients with stable ICM. OBJECTIVE To test the hypothesis that serum levels of hs-TnT correlate with cell retention and determine the response to intracoronary BMC application in patients with ICM. METHODS AND RESULTS About 157 patients with stable ICM and no substantial impairment of kidney function received intracoronary BMC administration. Immediately prior to cell application, hs-TnT levels to measure myocardial injury and NT-proBNP levels as marker of left ventricular wall stress were determined. Patients with elevated hs-TnT were older and had more severe heart failure. Importantly, only patients with elevated baseline hs-TnT≥15.19 pg/mL (upper tertile) demonstrated a significant (P=0.04) reduction in NT-proBNP serum levels (-250 [-1465; 33] pg/mL; relative reduction -24%) 4 months after BMC administration, whereas NT-proBNP levels remained unchanged in patients in the 2 lower hs-TnT tertiles. The absolute decrease in NT-proBNP at 4 months was inversely correlated with baseline hs-TnT (r=-0.27, P=0.001). Finally, retention of intracoronarily infused, 111Indium-labeled cells within the heart was closely associated with hs-TnT levels in patients with chronic ischemic heart failure (P=0.0008, n=10, triple measurements). CONCLUSIONS The extent of ongoing myocardial injury as measured by serum levels of hs-TnT predicts the reduction of NT-proBNP serum levels at 4 months after intracoronary BMC administration in patients with ICM, suggesting that the beneficial effects of BMC application on LV remodeling and wall stress are confined to patients with ongoing minute myocardial injury. CLINICAL TRIAL REGISTRATION URL: www.clinicaltrials.gov. Unique identifier: NCT00962364.
Collapse
Affiliation(s)
- Brigitte Luu
- From the Division of Cardiology, Department of Medicine III (B.L., D.M.L., F.H.S., J.H., S.F., A.M.Z., B.A.) and Institute of Biostatistics and Mathematical Modeling, Department of Medicine (E.H.), Goethe University Frankfurt, Germany; and German Center for Cardiovascular Research, DZHK, Partner Site Frankfurt Rhine-Main, Berlin, Germany (B.L., D.M.L., B.A., A.M.Z., B.A.)
| | - David M Leistner
- From the Division of Cardiology, Department of Medicine III (B.L., D.M.L., F.H.S., J.H., S.F., A.M.Z., B.A.) and Institute of Biostatistics and Mathematical Modeling, Department of Medicine (E.H.), Goethe University Frankfurt, Germany; and German Center for Cardiovascular Research, DZHK, Partner Site Frankfurt Rhine-Main, Berlin, Germany (B.L., D.M.L., B.A., A.M.Z., B.A.)
| | - Eva Herrmann
- From the Division of Cardiology, Department of Medicine III (B.L., D.M.L., F.H.S., J.H., S.F., A.M.Z., B.A.) and Institute of Biostatistics and Mathematical Modeling, Department of Medicine (E.H.), Goethe University Frankfurt, Germany; and German Center for Cardiovascular Research, DZHK, Partner Site Frankfurt Rhine-Main, Berlin, Germany (B.L., D.M.L., B.A., A.M.Z., B.A.)
| | - Florian H Seeger
- From the Division of Cardiology, Department of Medicine III (B.L., D.M.L., F.H.S., J.H., S.F., A.M.Z., B.A.) and Institute of Biostatistics and Mathematical Modeling, Department of Medicine (E.H.), Goethe University Frankfurt, Germany; and German Center for Cardiovascular Research, DZHK, Partner Site Frankfurt Rhine-Main, Berlin, Germany (B.L., D.M.L., B.A., A.M.Z., B.A.)
| | - Joerg Honold
- From the Division of Cardiology, Department of Medicine III (B.L., D.M.L., F.H.S., J.H., S.F., A.M.Z., B.A.) and Institute of Biostatistics and Mathematical Modeling, Department of Medicine (E.H.), Goethe University Frankfurt, Germany; and German Center for Cardiovascular Research, DZHK, Partner Site Frankfurt Rhine-Main, Berlin, Germany (B.L., D.M.L., B.A., A.M.Z., B.A.)
| | - Stephan Fichtlscherer
- From the Division of Cardiology, Department of Medicine III (B.L., D.M.L., F.H.S., J.H., S.F., A.M.Z., B.A.) and Institute of Biostatistics and Mathematical Modeling, Department of Medicine (E.H.), Goethe University Frankfurt, Germany; and German Center for Cardiovascular Research, DZHK, Partner Site Frankfurt Rhine-Main, Berlin, Germany (B.L., D.M.L., B.A., A.M.Z., B.A.)
| | - Andreas M Zeiher
- From the Division of Cardiology, Department of Medicine III (B.L., D.M.L., F.H.S., J.H., S.F., A.M.Z., B.A.) and Institute of Biostatistics and Mathematical Modeling, Department of Medicine (E.H.), Goethe University Frankfurt, Germany; and German Center for Cardiovascular Research, DZHK, Partner Site Frankfurt Rhine-Main, Berlin, Germany (B.L., D.M.L., B.A., A.M.Z., B.A.)
| | - Birgit Assmus
- From the Division of Cardiology, Department of Medicine III (B.L., D.M.L., F.H.S., J.H., S.F., A.M.Z., B.A.) and Institute of Biostatistics and Mathematical Modeling, Department of Medicine (E.H.), Goethe University Frankfurt, Germany; and German Center for Cardiovascular Research, DZHK, Partner Site Frankfurt Rhine-Main, Berlin, Germany (B.L., D.M.L., B.A., A.M.Z., B.A.).
| |
Collapse
|
31
|
MacAskill MG, Tavares AS, Wu J, Lucatelli C, Mountford JC, Baker AH, Newby DE, Hadoke PWF. PET Cell Tracking Using 18F-FLT is Not Limited by Local Reuptake of Free Radiotracer. Sci Rep 2017; 7:44233. [PMID: 28287126 PMCID: PMC5347009 DOI: 10.1038/srep44233] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 02/06/2017] [Indexed: 12/27/2022] Open
Abstract
Assessing the retention of cell therapies following implantation is vital and often achieved by labelling cells with 2'-[18F]-fluoro-2'-deoxy-D-glucose (18F-FDG). However, this approach is limited by local retention of cell-effluxed radiotracer. Here, in a preclinical model of critical limb ischemia, we assessed a novel method of cell tracking using 3'-deoxy-3'-L-[18F]-fluorothymidine (18F-FLT); a clinically available radiotracer which we hypothesise will result in minimal local radiotracer reuptake and allow a more accurate estimation of cell retention. Human endothelial cells (HUVECs) were incubated with 18F-FDG or 18F-FLT and cell characteristics were evaluated. Dynamic positron emission tomography (PET) images were acquired post-injection of free 18F-FDG/18F-FLT or 18F-FDG/18F-FLT-labelled HUVECs, following the surgical induction of mouse hind-limb ischemia. In vitro, radiotracer incorporation and efflux was similar with no effect on cell viability, function or proliferation under optimised conditions (5 MBq/mL, 60 min). Injection of free radiotracer demonstrated a faster clearance of 18F-FLT from the injection site vs. 18F-FDG (p ≤ 0.001), indicating local cellular uptake. Using 18F-FLT-labelling, estimation of HUVEC retention within the engraftment site 4 hr post-administration was 24.5 ± 3.2%. PET cell tracking using 18F-FLT labelling is an improved approach vs. 18F-FDG as it is not susceptible to local host cell reuptake, resulting in a more accurate estimation of cell retention.
Collapse
Affiliation(s)
- Mark G MacAskill
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Adriana S Tavares
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Junxi Wu
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | | | - Joanne C Mountford
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Andrew H Baker
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - David E Newby
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Patrick W F Hadoke
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
32
|
Der Sarkissian S, Lévesque T, Noiseux N. Optimizing stem cells for cardiac repair: Current status and new frontiers in regenerative cardiology. World J Stem Cells 2017; 9:9-25. [PMID: 28154736 PMCID: PMC5253186 DOI: 10.4252/wjsc.v9.i1.9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/20/2016] [Accepted: 10/24/2016] [Indexed: 02/06/2023] Open
Abstract
Cell therapy has the potential to improve healing of ischemic heart, repopulate injured myocardium and restore cardiac function. The tremendous hope and potential of stem cell therapy is well understood, yet recent trials involving cell therapy for cardiovascular diseases have yielded mixed results with inconsistent data thereby readdressing controversies and unresolved questions regarding stem cell efficacy for ischemic cardiac disease treatment. These controversies are believed to arise by the lack of uniformity of the clinical trial methodologies, uncertainty regarding the underlying reparative mechanisms of stem cells, questions concerning the most appropriate cell population to use, the proper delivery method and timing in relation to the moment of infarction, as well as the poor stem cell survival and engraftment especially in a diseased microenvironment which is collectively acknowledged as a major hindrance to any form of cell therapy. Indeed, the microenvironment of the failing heart exhibits pathological hypoxic, oxidative and inflammatory stressors impairing the survival of transplanted cells. Therefore, in order to observe any significant therapeutic benefit there is a need to increase resilience of stem cells to death in the transplant microenvironment while preserving or better yet improving their reparative functionality. Although stem cell differentiation into cardiomyocytes has been observed in some instance, the prevailing reparative benefits are afforded through paracrine mechanisms that promote angiogenesis, cell survival, transdifferentiate host cells and modulate immune responses. Therefore, to maximize their reparative functionality, ex vivo manipulation of stem cells through physical, genetic and pharmacological means have shown promise to enable cells to thrive in the post-ischemic transplant microenvironment. In the present work, we will overview the current status of stem cell therapy for ischemic heart disease, discuss the most recurring cell populations employed, the mechanisms by which stem cells deliver a therapeutic benefit and strategies that have been used to optimize and increase survival and functionality of stem cells including ex vivo preconditioning with drugs and a novel “pharmaco-optimizer” as well as genetic modifications.
Collapse
|
33
|
Eibel B, Markoski MM, Rodrigues CG, Dipp T, de Salles FB, Giusti II, Nardi NB, Plentz RDM, Kalil RAK. VEGF gene therapy cooperatively recruits molecules from the immune system and stimulates cell homing and angiogenesis in refractory angina. Cytokine 2016; 91:44-50. [PMID: 27997860 DOI: 10.1016/j.cyto.2016.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 10/26/2016] [Accepted: 12/06/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND New vessels are formed in response to stimuli from angiogenic factors, a process in which paracrine signaling is fundamental. OBJECTIVE To investigate the cooperative paracrine signaling profile in response to Vascular Endothelial Growth Factor (VEGF) gene therapy in patients with coronary artery disease (CAD) and refractory angina. METHOD A cohort study was conducted in which plasma was collected from patients who underwent gene therapy with a plasmid expressing VEGF 165 (10) and from surgical procedure controls (4). Blood samples were collected from both groups prior to baseline and on days 3, 9 and 27 after the interventions and subjected to systemic analysis of protein expression (Interleukin-6, IL-6; Tumor Necrosis Factor-α, TNF-α; Interleukin-10, IL-10; Stromal Derived Factor-1 α, SDF-1α; VEGF; Angiopoietin-1, ANGPT-1; and Endothelin-1, ET-1) using the enzyme-linked immunosorbent assay (ELISA). RESULTS Analysis showed an increase in proinflammatory IL-6 (p=0.02) and ET-1 (p=0.05) on day 3 after gene therapy and in VEGF (p=0.02) on day 9. A strong positive correlation was found between mobilization of endothelial progenitor cells and TNF-α on day 9 (r=0.71; p=0.03). Furthermore, a strong correlation between β-blockers, antiplatelets, and vasodilators with SDF-1α baseline in the group undergoing gene therapy was verified (r=0.74; p=0.004). CONCLUSION Analysis of cooperative paracrine signaling after VEGF gene therapy suggests that the immune system cell and angiogenic molecule expression as well as the endothelial progenitor cell mobilization are time-dependent, influenced by chronic inflammatory process and continuous pharmacological treatment.
Collapse
Affiliation(s)
- Bruna Eibel
- Laboratório de Cardiologia Molecular e Celular (Serviço de Medicina Experimental)/Secretaria de Cirurgia, Instituto de Cardiologia/Fundação Universitária de Cardiologia (IC/FUC), Brazil
| | - Melissa M Markoski
- Laboratório de Cardiologia Molecular e Celular (Serviço de Medicina Experimental)/Secretaria de Cirurgia, Instituto de Cardiologia/Fundação Universitária de Cardiologia (IC/FUC), Brazil
| | - Clarissa G Rodrigues
- Laboratório de Cardiologia Molecular e Celular (Serviço de Medicina Experimental)/Secretaria de Cirurgia, Instituto de Cardiologia/Fundação Universitária de Cardiologia (IC/FUC), Brazil
| | - Thiago Dipp
- Faculdade de Desenvolvimento do Rio Grande do Sul (FADERGS), Brazil
| | - Felipe B de Salles
- Instituto do Coração/Faculdade de Medicina da Universidade de São Paulo (INCOR/FMUSP), Brazil
| | - Imarilde I Giusti
- Laboratório de Cardiologia Molecular e Celular (Serviço de Medicina Experimental)/Secretaria de Cirurgia, Instituto de Cardiologia/Fundação Universitária de Cardiologia (IC/FUC), Brazil
| | | | - Rodrigo D M Plentz
- Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Brazil
| | - Renato A K Kalil
- Laboratório de Cardiologia Molecular e Celular (Serviço de Medicina Experimental)/Secretaria de Cirurgia, Instituto de Cardiologia/Fundação Universitária de Cardiologia (IC/FUC), Brazil; Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Brazil.
| |
Collapse
|
34
|
Curley CJ, Dolan EB, Cavanagh B, O'Sullivan J, Duffy GP, Murphy BP. An in vitro investigation to assess procedure parameters for injecting therapeutic hydrogels into the myocardium. J Biomed Mater Res B Appl Biomater 2016; 105:2618-2629. [PMID: 27764526 DOI: 10.1002/jbm.b.33802] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 08/22/2016] [Accepted: 10/02/2016] [Indexed: 12/12/2022]
Abstract
Localized delivery of stem cells is potentially a promising therapeutic strategy for regenerating damaged myocardium. Many studies focus on limiting the biologic component of cell loss, but few address the contribution of mechanical factors. This study investigates optimal parameters for retaining the largest volume of cell loaded hydrogels post intramyocardial injection, without compromising cell viability. In vitro, hydrogel was injected into porcine hearts using various needle designs. Hydrogel retention and distribution pattern was then determined. The two most promising needles were then investigated to understand the effect of needle geometry on stem cell viability. The needle to best impact cell viability was then used to investigate the effect of differing hydrogels on retention and distribution. Three-dimensional experimental modeling revealed needles with smaller diameter's to have greater poloxamer 407 hydrogel retention. No difference in retention existed among various needle designs of similar gauge, despite differences in bolus geometries. When hMSC's, embedded in fibrin hydrogel, were injected through helical and 26G bevel needles no difference in the percent of live cells was seen at 48 h. However, the helical group had almost half the metabolic activity of the 26G bevel group at both time points, and had a significant decline in the percent of live cells from 24 to 48 h. Varying gel type resulted in significantly more alginate being retained in the tissue in comparison to fibrin or poloxamer hydrogels. In conclusion, mechanical properties of injected hydrogels, and the diameter of the needle used, highly influences the volume of hydrogel retained. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 2618-2629, 2017.
Collapse
Affiliation(s)
- Clive J Curley
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland.,Advanced Materials and BioEngineering Research Centre (AMBER), TCD & RCSI, Dublin, Ireland
| | - Eimear B Dolan
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland.,Advanced Materials and BioEngineering Research Centre (AMBER), TCD & RCSI, Dublin, Ireland
| | - Brenton Cavanagh
- Cellular and Molecular Imaging Core, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Janice O'Sullivan
- Advanced Materials and BioEngineering Research Centre (AMBER), TCD & RCSI, Dublin, Ireland.,Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Garry P Duffy
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.,Advanced Materials and BioEngineering Research Centre (AMBER), TCD & RCSI, Dublin, Ireland.,Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Bruce P Murphy
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.,Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland.,Advanced Materials and BioEngineering Research Centre (AMBER), TCD & RCSI, Dublin, Ireland
| |
Collapse
|
35
|
Magnet-Bead Based MicroRNA Delivery System to Modify CD133 + Stem Cells. Stem Cells Int 2016; 2016:7152761. [PMID: 27795713 PMCID: PMC5067480 DOI: 10.1155/2016/7152761] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 08/24/2016] [Accepted: 08/30/2016] [Indexed: 12/13/2022] Open
Abstract
Aim. CD133+ stem cells bear huge potential for regenerative medicine. However, low retention in the injured tissue and massive cell death reduce beneficial effects. In order to address these issues, we intended to develop a nonviral system for appropriate cell engineering. Materials and Methods. Modification of human CD133+ stem cells with magnetic polyplexes carrying microRNA was studied in terms of efficiency, safety, and targeting potential. Results. High microRNA uptake rates (~80-90%) were achieved without affecting CD133+ stem cell properties. Modified cells can be magnetically guided. Conclusion. We developed a safe and efficient protocol for CD133+ stem cell modification. Our work may become a basis to improve stem cell therapeutical effects as well as their monitoring with magnetic resonance imaging.
Collapse
|
36
|
Nadlacki B, Suuronen EJ. Biomaterial strategies to improve the efficacy of bone marrow cell therapy for myocardial infarction. Expert Opin Biol Ther 2016; 16:1501-1516. [DOI: 10.1080/14712598.2016.1235149] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
37
|
Gervois P, Wolfs E, Ratajczak J, Dillen Y, Vangansewinkel T, Hilkens P, Bronckaers A, Lambrichts I, Struys T. Stem Cell-Based Therapies for Ischemic Stroke: Preclinical Results and the Potential of Imaging-Assisted Evaluation of Donor Cell Fate and Mechanisms of Brain Regeneration. Med Res Rev 2016; 36:1080-1126. [DOI: 10.1002/med.21400] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/27/2016] [Accepted: 06/17/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Pascal Gervois
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Esther Wolfs
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Jessica Ratajczak
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Yörg Dillen
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Tim Vangansewinkel
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Petra Hilkens
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Annelies Bronckaers
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Ivo Lambrichts
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| | - Tom Struys
- Morphology Research Group, Biomedical Research Institute, Hasselt University; Campus Diepenbeek; Bioville Diepenbeek Belgium
| |
Collapse
|
38
|
Abstract
Peripheral vascular disease (PVD) is a progressive atherosclerotic disease that leads to stenosis or occlusion of blood vessels supplying the lower extremities. Current diagnostic imaging techniques commonly focus on evaluation of anatomy or blood flow at the macrovascular level and do not permit assessment of the underlying pathophysiology associated with disease progression or treatment response. Molecular imaging with radionuclide-based approaches can offer novel insight into PVD by providing noninvasive assessment of biological processes such as angiogenesis and atherosclerosis. This article discusses emerging radionuclide-based imaging approaches that have potential clinical applications in the evaluation of PVD progression and treatment.
Collapse
Affiliation(s)
- Mitchel R Stacy
- Department of Internal Medicine, Yale University School of Medicine, PO Box 208017, Dana-3, New Haven, CT 06520, USA.
| | - Albert J Sinusas
- Department of Internal Medicine, Yale University School of Medicine, PO Box 208017, Dana-3, New Haven, CT 06520, USA; Department of Diagnostic Radiology, Yale University School of Medicine, PO Box 208042, New Haven, CT 06520, USA
| |
Collapse
|
39
|
In Vivo Tracking of Cell Therapies for Cardiac Diseases with Nuclear Medicine. Stem Cells Int 2016; 2016:3140120. [PMID: 26880951 PMCID: PMC4737458 DOI: 10.1155/2016/3140120] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 10/18/2015] [Accepted: 10/20/2015] [Indexed: 12/31/2022] Open
Abstract
Even though heart diseases are amongst the main causes of mortality and morbidity in the world, existing treatments are limited in restoring cardiac lesions. Cell transplantations, originally developed for the treatment of hematologic ailments, are presently being explored in preclinical and clinical trials for cardiac diseases. Nonetheless, little is known about the possible efficacy and mechanisms for these therapies and they are the center of continuous investigation. In this scenario, noninvasive imaging techniques lead to greater comprehension of cell therapies. Radiopharmaceutical cell labeling, firstly developed to track leukocytes, has been used successfully to evaluate the migration of cell therapies for myocardial diseases. A substantial rise in the amount of reports employing this methodology has taken place in the previous years. We will review the diverse radiopharmaceuticals, imaging modalities, and results of experimental and clinical studies published until now. Also, we report on current limitations and potential advances of radiopharmaceutical labeling for cell therapies in cardiac diseases.
Collapse
|
40
|
Lezaic L, Socan A, Peitl PK, Poglajen G, Sever M, Cukjati M, Cernelc P, Vrtovec B. Imaging and 1-day kinetics of intracoronary stem cell transplantation in patients with idiopathic dilated cardiomyopathy. Nucl Med Biol 2016; 43:410-4. [PMID: 27179747 DOI: 10.1016/j.nucmedbio.2015.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 11/18/2015] [Accepted: 12/29/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Stem cell transplantation is an emerging method of treatment for patients with cardiovascular disease. There are few studies completed or ongoing on stem cell therapy in patients with idiopathic dilated cardiomyopathy (IDCM). Information on stem cell homing and distribution in the myocardium after transplantation might provide important insight into effectiveness of transplantation procedure. AIM To assess early engraftment, retention and migration of intracoronarily transplanted stem cells in the myocardium of patients with advanced dilated cardiomyopathy of non-ischaemic origin using stem cell labeling with (99m)Tc-exametazime (HMPAO). MATERIALS, METHODS Thirty-five patients with IDCM and advanced heart failure were included in the study. Autologous hematopoietic (CD34+) stem cells were harvested by peripheral blood apheresis after bone marrow stimulation, labeled with (99m)Tc-HMPAO, tested for viability and injected into coronary vessel supplying areas of myocardium selected by myocardial perfusion scintigraphy as dysfunctional yet viable. Imaging was performed 1h and 18h after transplantation. RESULTS Myocardial stem cell retention ranged from 0 to 1.44% on early and 0-0.97% on delayed imaging. Significant efflux of stem cells occurred from site of delivery in this time period (p<0.001). Stem cell viability was not affected by labeling. CONCLUSION Stem cell labeling with (99m)Tc-HMPAO is a feasible method for stem cell tracking after transplantation in patients with IDCM.
Collapse
Affiliation(s)
- Luka Lezaic
- Department for Nuclear Medicine, UMC Ljubljana, Slovenia.
| | - Aljaz Socan
- Department for Nuclear Medicine, UMC Ljubljana, Slovenia
| | | | - Gregor Poglajen
- Advanced Heart Failure and Transplantation Center, UMC Ljubljana, Slovenia
| | - Matjaz Sever
- Department of Hematology, UMC Ljubljana, Slovenia
| | - Marko Cukjati
- National Blood Transfusion Institute, Ljubljana, Slovenia
| | | | - Bojan Vrtovec
- Advanced Heart Failure and Transplantation Center, UMC Ljubljana, Slovenia; Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
41
|
Cell Therapy in Ischemic Heart Disease: Interventions That Modulate Cardiac Regeneration. Stem Cells Int 2016; 2016:2171035. [PMID: 26880938 PMCID: PMC4736413 DOI: 10.1155/2016/2171035] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/26/2015] [Accepted: 11/10/2015] [Indexed: 12/15/2022] Open
Abstract
The incidence of severe ischemic heart disease caused by coronary obstruction has progressively increased. Alternative forms of treatment have been studied in an attempt to regenerate myocardial tissue, induce angiogenesis, and improve clinical conditions. In this context, cell therapy has emerged as a promising alternative using cells with regenerative potential, focusing on the release of paracrine and autocrine factors that contribute to cell survival, angiogenesis, and tissue remodeling. Evidence of the safety, feasibility, and potential effectiveness of cell therapy has emerged from several clinical trials using different lineages of adult stem cells. The clinical benefit, however, is not yet well established. In this review, we discuss the therapeutic potential of cell therapy in terms of regenerative and angiogenic capacity after myocardial ischemia. In addition, we addressed nonpharmacological interventions that may influence this therapeutic practice, such as diet and physical training. This review brings together current data on pharmacological and nonpharmacological approaches to improve cell homing and cardiac repair.
Collapse
|
42
|
How to Improve the Survival of Transplanted Mesenchymal Stem Cell in Ischemic Heart? Stem Cells Int 2015; 2016:9682757. [PMID: 26681958 PMCID: PMC4670674 DOI: 10.1155/2016/9682757] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/18/2015] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cell (MSC) is an intensely studied stem cell type applied for cardiac repair. For decades, the preclinical researches on animal model and clinical trials have suggested that MSC transplantation exerts therapeutic effect on ischemic heart disease. However, there remain major limitations to be overcome, one of which is the very low survival rate after transplantation in heart tissue. Various strategies have been tried to improve the MSC survival, and many of them showed promising results. In this review, we analyzed the studies in recent years to summarize the methods, effects, and mechanisms of the new strategies to address this question.
Collapse
|
43
|
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
|
44
|
Rose LC, Kadayakkara DK, Wang G, Bar-Shir A, Helfer BM, O'Hanlon CF, Kraitchman DL, Rodriguez RL, Bulte JWM. Fluorine-19 Labeling of Stromal Vascular Fraction Cells for Clinical Imaging Applications. Stem Cells Transl Med 2015; 4:1472-81. [PMID: 26511652 DOI: 10.5966/sctm.2015-0113] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 08/31/2015] [Indexed: 12/15/2022] Open
Abstract
UNLABELLED Stromal vascular fraction (SVF) cells are used clinically for various therapeutic targets. The location and persistence of engrafted SVF cells are important parameters for determining treatment failure versus success. We used the GID SVF-1 platform and a clinical protocol to harvest and label SVF cells with the fluorinated ((19)F) agent CS-1000 as part of a first-in-human phase I trial (clinicaltrials.gov identifier NCT02035085) to track SVF cells with magnetic resonance imaging during treatment of radiation-induced fibrosis in breast cancer patients. Flow cytometry revealed that SVF cells consisted of 25.0% ± 15.8% CD45+, 24.6% ± 12.5% CD34+, and 7.5% ± 3.3% CD31+ cells, with 2.1 ± 0.7 × 10⁵ cells per cubic centimeter of adipose tissue obtained. Fluorescent CS-1000 (CS-ATM DM Green) labeled 87.0% ± 13.5% of CD34+ progenitor cells compared with 47.8% ± 18.5% of hematopoietic CD45+ cells, with an average of 2.8 ± 2.0 × 10¹² ¹⁹F atoms per cell, determined using nuclear magnetic resonance spectroscopy. The vast majority (92.7% ± 5.0%) of CD31+ cells were also labeled, although most coexpressed CD34. Only 16% ± 22.3% of CD45-/CD31-/CD34- (triple-negative) cells were labeled with CS-ATM DM Green. After induction of cell death by either apoptosis or necrosis, >95% of ¹⁹F was released from the cells, indicating that fluorine retention can be used as a surrogate marker for cell survival. Labeled-SVF cells engrafted in a silicone breast phantom could be visualized with a clinical 3-Tesla magnetic resonance imaging scanner at a sensitivity of approximately 2 × 10⁶ cells at a depth of 5 mm. The current protocol can be used to image transplanted SVF cells at clinically relevant cell concentrations in patients. SIGNIFICANCE Stromal vascular fraction (SVF) cells harvested from adipose tissue offer great promise in regenerative medicine, but methods to track such cell therapies are needed to ensure correct administration and monitor survival. A clinical protocol was developed to harvest and label SVF cells with the fluorinated (¹⁹F) agent CS-1000, allowing cells to be tracked with (19)F magnetic resonance imaging (MRI). Flow cytometry evaluation revealed heterogeneous ¹⁹F uptake in SVF cells, confirming the need for careful characterization. The proposed protocol resulted in sufficient ¹⁹F uptake to allow imaging using a clinical MRI scanner with point-of-care processing.
Collapse
Affiliation(s)
- Laura C Rose
- Division of Magnetic Resonance Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Deepak K Kadayakkara
- Division of Magnetic Resonance Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guan Wang
- Department of Electrical and Computer Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amnon Bar-Shir
- Division of Magnetic Resonance Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | - Dara L Kraitchman
- Division of Magnetic Resonance Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Jeff W M Bulte
- Division of Magnetic Resonance Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA Department of Chemical & Biomolecular Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
45
|
Vajravelu BN, Hong KU, Al-Maqtari T, Cao P, Keith MCL, Wysoczynski M, Zhao J, Moore IV JB, Bolli R. C-Kit Promotes Growth and Migration of Human Cardiac Progenitor Cells via the PI3K-AKT and MEK-ERK Pathways. PLoS One 2015; 10:e0140798. [PMID: 26474484 PMCID: PMC4608800 DOI: 10.1371/journal.pone.0140798] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/29/2015] [Indexed: 01/01/2023] Open
Abstract
A recent phase I clinical trial (SCIPIO) has shown that autologous c-kit+ cardiac progenitor cells (CPCs) improve cardiac function and quality of life when transplanted into patients with ischemic heart disease. Although c-kit is widely used as a marker of resident CPCs, its role in the regulation of the cellular characteristics of CPCs remains unknown. We hypothesized that c-kit plays a role in the survival, growth, and migration of CPCs. To test this hypothesis, human CPCs were grown under stress conditions in the presence or absence of SCF, and the effects of SCF-mediated activation of c-kit on CPC survival/growth and migration were measured. SCF treatment led to a significant increase in cell survival and a reduction in cell death under serum depletion conditions. In addition, SCF significantly promoted CPC migration in vitro. Furthermore, the pro-survival and pro-migratory effects of SCF were augmented by c-kit overexpression and abrogated by c-kit inhibition with imatinib. Mechanistically, c-kit activation in CPCs led to activation of the PI3K and the MAPK pathways. With the use of specific inhibitors, we confirmed that the SCF/c-kit-dependent survival and chemotaxis of CPCs are dependent on both pathways. Taken together, our findings suggest that c-kit promotes the survival/growth and migration of human CPCs cultured ex vivo via the activation of PI3K and MAPK pathways. These results imply that the efficiency of CPC homing to the injury site as well as their survival after transplantation may be improved by modulating the activity of c-kit.
Collapse
Affiliation(s)
- Bathri N. Vajravelu
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Kyung U. Hong
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Tareq Al-Maqtari
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Pengxiao Cao
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Matthew C. L. Keith
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Marcin Wysoczynski
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - John Zhao
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Joseph B. Moore IV
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Roberto Bolli
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
- * E-mail:
| |
Collapse
|
46
|
Du W, Tao H, Zhao S, He ZX, Li Z. Translational applications of molecular imaging in cardiovascular disease and stem cell therapy. Biochimie 2015; 116:43-51. [PMID: 26134715 DOI: 10.1016/j.biochi.2015.06.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 06/25/2015] [Indexed: 12/21/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality and morbidity worldwide. Molecular imaging techniques provide valuable information at cellular and molecular level, as opposed to anatomical and structural layers acquired from traditional imaging modalities. More specifically, molecular imaging employs imaging probes which interact with specific molecular targets and therefore makes it possible to visualize biological processes in vivo. Molecular imaging technology is now progressing towards preclinical and clinical application that gives an integral and comprehensive guidance for the investigation of cardiovascular disease. In addition, cardiac stem cell therapy holds great promise for clinical translation. Undoubtedly, combining stem cell therapy with molecular imaging technology will bring a broad prospect for the study and treatment of cardiac disease. This review will focus on the progresses of molecular imaging strategies in cardiovascular disease and cardiac stem cell therapy. Furthermore, the perspective on the future role of molecular imaging in clinical translation and potential strategies in defining safety and efficacy of cardiac stem cell therapies will be discussed.
Collapse
Affiliation(s)
- Wei Du
- Collaborative Innovation Center for Biotherapy, Nankai University School of Medicine, Tianjin, China; Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University School of Medicine, Tianjin, China; The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Hongyan Tao
- Collaborative Innovation Center for Biotherapy, Nankai University School of Medicine, Tianjin, China; Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University School of Medicine, Tianjin, China
| | - Shihua Zhao
- Department of Radiology, Fuwai Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Zuo-Xiang He
- Department of Nuclear Imaging, Fuwai Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.
| | - Zongjin Li
- Collaborative Innovation Center for Biotherapy, Nankai University School of Medicine, Tianjin, China; Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University School of Medicine, Tianjin, China; The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China.
| |
Collapse
|
47
|
Lin YD, Chang MY, Cheng B, Liu YW, Lin LC, Chen JH, Hsieh PCH. Injection of Peptide nanogels preserves postinfarct diastolic function and prolongs efficacy of cell therapy in pigs. Tissue Eng Part A 2015; 21:1662-71. [PMID: 25686878 DOI: 10.1089/ten.tea.2014.0581] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Accumulating evidence suggests that the benefits of cell therapy for cardiac repair are modest and transient due to progressive harmful cardiac remodeling as well as loss of transplanted cells. We previously demonstrated that injection of peptide nanofibers (NFs) reduces ventricular remodeling and facilitates cell retention at 1 month after acute myocardial infarction (MI) in pigs. However, it remains unclear whether these benefits still persist as the material is being degraded. In this study, 2 mL of placebo or NFs, with or without 1×10(8) mononuclear cells (MNCs), was injected into the pig myocardium after MI (n≥5 in each group), and cardiac function was assessed by echocardiography, including myocardial deformation analyses and catheterization at 3 months post-MI. Our results reveal that MNC-only injection slightly improved cardiac systolic function at 1 month post-MI, but this benefit was lost at later time points (ejection fraction: 42.0±2.3 in MI+normal saline [NS] and 43.5±1.1 in MI+MNCs). In contrast, NF-only injection resulted in improved cardiac diastolic function and reduced pathological remodeling at 3 months post-MI. Furthermore, combined injection of MNCs/NFs provided a greater and longer term cardiac performance (52.1±1.2 in MI+MNCs/NFs, p<0.001 versus MI+NS and MI+MNCs) and 11.3-fold transplanted cell retention. We also found that about 30% NFs remained at 3 months after injection; however, endogenous myofibroblasts were recruited to the NF-injected microenvironment to replace the degraded NFs and preserved cardiac dimensions and mechanics. In conclusion, we demonstrated that injection of NFs contributes to preservation of ventricular mechanical integrity and sustains MNC efficacy at 3 months postinjection.
Collapse
Affiliation(s)
- Yi-Dong Lin
- 1 Institute of Biomedical Sciences , Academia Sinica, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Heart failure is a growing epidemic caused by cardiomyocyte depletion. Current therapies prolong survival by protecting remaining cardiomyocytes but are unable to overcome the fundamental problem of regenerating lost cardiomyocytes. Several strategies for promoting heart regeneration have emerged from decades of intensive study. Although some of these strategies remain confined to basic research, others are beginning to be tested in humans. We review strategies for cardiac regeneration and summarize progress of related clinical trials.
Collapse
Affiliation(s)
- Zhiqiang Lin
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA. Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
49
|
Abstract
The field of regenerative medicine has experienced considerable growth in recent years as the translation of pre-clinical biomaterials and cell- and gene-based therapies begin to reach clinical application. Until recently, the ability to monitor the serial responses to therapeutic treatments has been limited to post-mortem tissue analyses. With improvements in existing imaging modalities and the emergence of hybrid imaging systems, it is now possible to combine information related to structural remodeling with associated molecular events using non-invasive imaging. This review summarizes the established and emerging imaging modalities that are available for in vivo monitoring of clinical regenerative medicine therapies and discusses the strengths and limitations of each imaging modality.
Collapse
Affiliation(s)
- Mitchel R. Stacy
- Department of Internal Medicine, Yale University School of Medicine, P.O. Box 208017, Dana-3, New Haven, CT 06520 USA
| | - Albert J. Sinusas
- Departments of Internal Medicine & Diagnostic Radiology, Yale University School of Medicine, P.O. Box 208017, Dana-3, New Haven, CT 06520 USA
| |
Collapse
|
50
|
Abstract
Although cellular therapies hold great promise for the treatment of human disease, results from several initial clinical trials have not shown a level of efficacy required for their use as a first line therapy. Here we discuss how in vivo molecular imaging has helped identify barriers to clinical translation and potential strategies that may contribute to successful transplantation and improved outcomes, with a focus on cardiovascular and neurological diseases. We conclude with a perspective on the future role of molecular imaging in defining safety and efficacy for clinical implementation of stem cell therapies.
Collapse
|